You are on page 1of 1306

Niedermeyer’s

Electroencephalography
Basic Principles, Clinical Applications, and Related Fields

SIXTH EDITION
Niedermeyer’s
Electroencephalography
Basic Principles, Clinical Applications, and Related Fields

SIXTH EDITION

Donald L. Schomer, MA, MD


Professor of Neurology
Harvard Medical School
Director, Laboratory for Clinical Neurophysiology
Chief, Comprehensive Epilepsy Program
Beth Israel Deaconess Medical Center
Boston, Massachusetts

Fernando H. Lopes da Silva, MD, PhD


Emeritus Professor
Swammerdam Institute for Life Sciences
Center of Neurosciences
Faculty of Science
University of Amsterdam
Amsterdam, The Netherlands
Acquisitions Editor: Frances Destefano
Product Manager: Tom Gibbons
Vendor Manager: Bridgett Dougherty
Senior Manufacturing Manager: Benjamin Rivera
Marketing Manager: Brian Freiland
Design Coordinator: Stephen Druding
Production Service: MPS, a Macmillan Company

© 2011 by LIPPINCOTT WILLIAMS & WILKINS, a WOLTERS KLUWER business.

Two Commerce Square


2001 Market Street
Philadelphia, PA 19103 USA
LWW.com

Printed in China

All rights reserved. This book is protected by copyright. No part of this book may be reproduced in
any form by any means, including photocopying, or utilized by any information storage and retrieval
system without written permission from the copyright owner, except for brief quotations embodied
in critical articles and reviews. Materials appearing in this book prepared by individuals as part of
their official duties as U.S. government employees are not covered by the above-mentioned copyright.

Library of Congress Cataloging-in-Publication Data

Niedermeyer’s electroencephalography: basic principles, clinical applications, and related fields/


[edited by] Donald L. Schomer, Fernando H. Lopes da Silva.—6th ed.
p. ; cm.
Electroencephalography
Rev. ed. of: Electroencephalography/[edited by] Ernst Niedermeyer, Fernando H. Lopes da Silva.
5th ed. c2005.
Includes bibliographical references and index.
ISBN-13: 978-0-7817-8942-4 (hardback: alk. paper)
ISBN-10: 0-7817-8942-7 (hardback: alk. paper) 1.
Electroencephalography. I. Niedermeyer, Ernst, 1920– II. Schomer, Donald L. III. Lopes da Silva,
F. H., 1935– IV. Title: Electroencephalography.
[DNLM: 1. Electroencephalography. 2. Central Nervous System Diseases—diagnosis. WL 150]
RC386.6.E43N54 2011
616.8’047547—dc22
2010037480

Care has been taken to confirm the accuracy of the information presented and to describe generally
accepted practices. However, the authors, editors, and publisher are not responsible for errors or
omissions or for any consequences from application of the information in this book and make no
warranty, expressed or implied, with respect to the currency, completeness, or accuracy of the
contents of the publication. Application of the information in a particular situation remains the
professional responsibility of the practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug selection and
dosage set forth in this text are in accordance with current recommendations and practice at the time
of publication. However, in view of ongoing research, changes in government regulations, and the
constant flow of information relating to drug therapy and drug reactions, the reader is urged to check
the package insert for each drug for any change in indications and dosage and for added warnings and
precautions. This is particularly important when the recommended agent is a new or infrequently
employed drug.
Some drugs and medical devices presented in the publication have Food and Drug Administration
(FDA) clearance for limited use in restricted research settings. It is the responsibility of the health care
provider to ascertain the FDA status of each drug or device planned for use in their clinical practice.
To purchase additional copies of this book, call our customer service department at (800)
638-3030 or fax orders to (301) 223-2320. International customers should call (301) 223-2300.
Visit Lippincott Williams & Wilkins on the Internet: at LWW.com. Lippincott Williams &
Wilkins customer service representatives are available from 8:30 am to 6 pm, EST.
10 9 8 7 6 5 4 3 2 1
Dedicated to Ernst Niedermeyer, M.D.
for all of the help and inspiration offered to us and
to countless clinical neurophysiologists worldwide
over his truly remarkable career.
Preface to the First Edition

The history of clinical electroencephalography (EEG) has just aspects of lesions and tissue changes, neurology would be
passed the 50-year mark. The age of the pioneers was followed shallow and barren without awareness of the constant fluctua-
by the stage of expansion. What began in a few prestigious cen- tion of functional states in the CNS. Another important sti-
ters gradually became a tool of all academic medical institu- mulus has been the establishment of standards of quality for
tions, and eventually of all major hospitals. In more recent electroencephalographers and EEG laboratories.
years, EEG even invaded the private offices of practicing neurol- In these times of challenge, a review of the state of affairs in
ogists and other specialists interested in central nervous system EEG seems to be appropriate. Such thoughts have prompted the
(CNS) disease. genesis of this large one-volume textbook, which, by its mere
From this perspective, the history of clinical EEG looks like size, sets itself apart from the group of smaller introductory
the “via triumphalis” of a buoyantly dynamic new subspecialty. textbooks and from the huge multivolume handbook. The one-
The elucidation of the electrophysiologic process underlying volume character of the book symbolizes the spirit of unity that
epileptic seizure disorders and a variety of other CNS dysfunc- should reign among clinical electroencephalographers, basic
tions was indeed a unique achievement made possible by the science researchers, and the workers in the field of compu -
new method. The original intention of the founder of clinical terized data analysis.
EEG, Hans Berger, had been the exploration of mental and psy- One author can hardly undertake such a task alone. For this
chological processes, and even in this domain the yield has been reason, we have reached out across the ocean for collaboration.
substantial. Moreover, electroencephalographers have not con- It became clear, however, that a two-man effort would not
fined themselves to the spontaneous wave patterns of the brain; suffice to cover the entire field in the relatively short working
forms of EEG data analysis with the aid of computers were period of 2 years. We solicited for assistance and found a
introduced in order to demonstrate evoked and event-related wonderful group of contributors in various special areas. Above
potentials and to investigate the wealth of frequencies that con- all, coverage of the fields of neurophysiology and neurophar-
stitute the EEG. In the search for the sources of EEG generation, macology has depended most heavily on the assistance of
the brain tissue became the target of exploration. prestigious specialists.
Depth electrodes became one of the most important tools of Attempts at synthesis have not been the goal of this book.
experimental neurophysiologists, who also investigated the sin- Instead the reader will find a more individualistic approach
gle neuron using microelectrodes. The implantation of depth from which the personal basic philosophy of each author can be
electrodes in the human brain has aided in the evaluation of derived. No effort has been made to achieve strict standardiza-
chronic epileptics considered candidates for seizure surgery. tion of symbols and terminology; as an example, frequencies
This impressive progress, however, has been counterbal- are described in various terms (10 cps, 10/sec, 10 Hz, etc.).
anced by signs of pessimism, fatigue, and resignation. A certain There is also some overlap between certain chapters; we feel
malaise has inched its way into the hearts of thousands of elec- that the reader will benefit from the presentation of a given
troencephalographers who have started to feel the grip of stag- topic as seen from two somewhat different viewpoints.
nation. Generation of EEG potentials has proved to be A piece of technical information might be worthwhile.
extremely complex and difficult to understand; the feeling of Unless the filter setting is specifically indicated in the illus-
doing pragmatically useful work with an ill-understood trations of EEG tracings, a time constant between 0.15 and
method has been depressing to many workers in the field. The 0.5 seconds was used (above 0.4 seconds when slow frequencies
pragmatists have further suffered from the limitations of EEG played a major role). The use of “muscle filters” was avoided.
as a method of localization of cerebral lesions. These feelings We have tried to combine didactic and academic elements in
have been nourished by the phenomenal achievements of non- this book. Hence, trainees as well as seasoned professionals in
invasive radionuclear and radiologic scanning methods; this the field will, we hope, find what they are searching for. This
progress of new methods in the field of structural diagnosis has dual approach does mean that some parts of the book require
been a matter of concern for many electroencephalographers. greater sophistication of the reader than do others.
A more real danger, perhaps, is presented by the poorly trained Acknowledgments for invaluable help in this undertaking
colleagues who are tarnishing the image of EEG. come from the depth of our hearts. Mr. Braxton Dallam
In reality, however, these challenges present a stimulus for Mitchell, President of Urban & Schwarzenberg in Baltimore,
the electrophysiologic field. The function-oriented aspects of Maryland, deserves the honor of having been the initiator of
neurologic sciences will always be of paramount significance. this book. His encouragement has been very much appreciated.
The loss of function-oriented neurology would foreshadow the Detlev Moos has coordinated the production of this book with
death of neurology. With all due respect for the structural care and efficiency; Suzanne Lohmeyer has copyedited and

vi
Preface vii

indexed it well; and Nan Tyler, Carola Sautter, and Victoria physiology, Dr. A Earl Walker (presently of Albuquerque, New
Doherty at the publisher’s Baltimore office assisted with their Mexico), deserve special gratitude.
experience. In the Johns Hopkins Hospital in Baltimore, Gratitude is expressed (by F.L.S.) to professor Dr. W. Storm
Maryland, the technical staff of the laboratory deserves great von Leeuwen, Dr. G. Wieneke, and Dr. K. Van Hulten (Utrecht
praise: Mrs. Judy Nastalski, R. EEG T. and chief technologist, University Hospital), Mr. N.J.I. Mars (Twente University of
Mr. Eric DeShields, Mrs. Debbie Reichenbach, R. EEG T., Technology), and Mr. A. Van Rotterdam (Institute of Medical
Miss Sharon Vaughan, Mrs. Kathleen Daniecki, R. EEG T., Physics, Utrecht) for their advice and encouragement. The high
Mrs. Cindy Haywood, and Miss Kim Rimel. How deeply the professional competence of Mrs. Ada Van Schaik and Mr. Nico
electroencephalographer depends on the quality of the record- Haagen (Institute of Medical Physics, Utrecht) in their fields of
ings and the dedication of the technologists! Truly invaluable secretarial work and artwork was of invaluable help.
was the secretarial assistance of Mrs. Catherine Bonolis. The Heartfelt thanks are also expressed to the contributing authors
operation of the laboratory was further aided by the experience of the book; they have naturally become a part of this undertak-
of Mrs. Marie Simpson. Important contributions came from ing. The response of these splendid coworkers was exemplary.
Mr. Joe Dieter, who is responsible for the pictorial artwork, Joseph J. Tecce and Lynn Cattanach, the authors of the article on
Mr. Ron Garret (lettering of tracings), and Mr. Zuhair Kareem contingent negative variation, substituted for a colleague who
and Mrs. Lillian Reich, the staff of Medical Photography. had to step down from his obligations at the last minute. They
Assistance and advice in the clinical EEG field was given made possible the almost impossible when they declared their
most freely by Dr. Gisela Freund, visiting assistant professor at willingness to join the team of coworkers. To them, and to all the
the John Hopkins Hospital EEG Laboratory (1980/81), of the contributing authors of this volume, our deepest thanks.
Department of Clinical Neurophysiology, Free University
Berlin (Klinikum Westend). E.N.’s principal teacher in the field Ernst Niedermeyer
of EEG, Dr. John R. Knott (presently of Boston, Massachusetts), Fernando H. Lopes da Silva
and the great master of neurosurgery, epileptology, and neuro- Spring 1981
Preface to the Sixth Edition

We would like to express our most sincere thanks to Dr. Ernst this classic volume continue for the years to come and evolve
Niedermeyer for his leadership and direction over the past several with the field as we have seen Dr. Niedermeyer do so success-
years as we have moved forth to produce this sixth edition of the fully over the last 25 years as its senior editor.
textbook that he had first introduced back in 1982. Additionally, This edition has several new features, reflective of the changes
we would like to honor him as he is so richly deserving by renam- that have occurred in our field over the past 5 years since the fifth
ing this book Niedermeyer’s Electroencephalography: Basic edition. More and more, the field of digital recording has
Principles, Clinical Applications, and Related Fields. expanded; however, in order to understand some of the short-
Dr. Niedermeyer was born in Vienna, Austria, in 1920. He comings and pitfalls of digital EEG, people need to still address
attended medical school in Austria but, during his third year in the issues of basic analog recording principles. With an increased
medical school, he was witness to the rise of Nazism in use of digital recording, laboratories have collected new and dif-
Germany and Austria and was inducted into the German army ferent “technical artifacts.” We present here an attempt to start a
just prior to the onset of World War II. He was stationed in database for such artifacts in a hope that future editions will
Berlin and was then literally drafted into the medical service continue to expand upon this and offer a fairly complete library
even though he had not yet graduated from medical school. He for beginners interested in our field. As noted in the fifth edition,
said that he was basically treating wounded soldiers coming epilepsy monitoring units (EMUs) have continued to mush-
back from the eastern front during the first several years of the room. Similar growth has occurred in the use of EEG monitor-
German invasion of the Soviet Union. He remained there until ing in newborn, cardiac, trauma, and postoperative intensive
Nazi officials learned that his maternal grandmother was Jewish care units. With the significant advances in wireless communica-
which, according to German law, made him an “undesirable.” tion and easy access to the Internet, such recordings can also be
However, because of his medical background, he was not viewed and transmitted locally virtually instantaneously and can
interned but rather was sent to Normandy. He mentioned that allow well-trained clinical neurophysiologists to see and opine
he was, as were many of the soldiers in Normandy, well aware of about patients’ conditions on a very time-relevant basis.
the coming invasion by the Allied forces and he looked upon Hopefully, as future generations may show, this ability will sig-
the day of the invasion as one of his “happiest.” He said he was nificantly influence our patients’ outcomes. Similarly, the field of
wounded during the invasion and was taken as a POW by the intraoperative clinical neurophysiology for spinal cord function,
American forces taking part in the invasion. He was sent back as cranial nerve function, and cranial vascular therapies has con-
a POW to Kansas where he remained and served out the rest of tinued to evolve along with the wireless and Internet communi-
the war as the prison camp doctor. He returned to Austria fol- cations. This has allowed for close monitoring of neurologic
lowing cessation of fighting and finished his medical school and function during critical periods of operations, again with a time
stayed on in Austria, interested in and learning neurology/psy- course that allows for corrective actions to be taken on a mean-
chiatry. He was introduced into clinical neurophysiology ingful time frame.
through some of the historical works in neurophysiology that We have reorganized the text regarding normal EEG and
had been conducted in Europe prior to World War II, including epilepsy to more closely follow the normal aging patterns. We
the work of Hans Berger. He became a clinical neurophysiology have continued to present some of the more classical chapters
addict and was offered a training position at the University of that have evolved over years on evoked potentials and routine
Iowa with Dr. John Knott. He took that position and stayed electroencephalography, including EEG in common neuro-
with Dr. Knott for a number of years but was then offered a logic, metabolic, and heredito-degenerative diseases. We have
position to work with Dr. A. Earl Walker at the Johns Hopkins added and updated chapters and text regarding automated and
Hospital, Baltimore. He continued to do his clinical and basic specialized mathematics-based analysis techniques to try to
science research at Hopkins, where his career blossomed and he keep up with this rapidly expanding field. We will continue to
became well known in the field. He has been an inspiration to update these techniques in future editions. We have included a
many trainees and colleagues, both at Johns Hopkins and chapter on linking clinical neurophysiology to other investiga-
worldwide. Many of our colleagues have come to know him tive techniques such as functional MRI. We have updated the
through his kindness and his willingness to personally take an chapter on magnetoencephalography to, again, reflect the sig-
interest in their research and to make recommendations and nificant changes that have taken place, both from a technical
suggestions about directions for further study. His knowledge and a clinical perspective, in this field. The last several chapters
of the field is legendary and his personality is truly “old world” of this textbook attempt to present an overview of clinical neu-
in the best sense of the term. It is our most sincere desire that rophysiology research that intersects with many other aspects of

viii
Preface ix

the realm of brain sciences such as consciousness and cognitive have offered tremendous help and support in the technical
processing. aspects of getting the chapters organized and together and for
We would like to encourage the readers of these words and Tom Gibbons from Lippincott and all those who have helped
this text to please let us know if they are aware of other areas of again in getting the textbook prepared for publication. Again, in
interest that could or should be involved in future editions of the words of Dr. Niedermeyer, “many, many thanks.”
this textbook that reflect either oversights on our part or are
harbingers of future development. Finally, we thank all of the Donald L. Schomer
authors and coauthors of the chapters of this book for their Fernando H. Lopes da Silva
tremendous effort in keeping this textbook relevant to our field.
Special thanks go to Fran Destefano and Franny Murphy who

ix
Contributors

Florin Amzica, Ph.D. Sudhansu Chokroverty, M.D., F.R.C.P.


Associate Professor of Stomatology Professor of Neuroscience
Montreal University Seton Hall University
Montreal, Canada South Orange, NJ
Clinical Professor of Neurology
Mary Repole Andriola, M.D.
Robert Wood Johnson Medical School
Professor of Neurology and Pediatrics
New Brunswick, NJ
Director, Divisions of Child Neurology, Clinical Neurophysiology
Professor and Co-chair of Neurology
and Pediatric Epilepsy
Program Director, Sleep Medicine and Clinical Neurophysiology
SUNY at Stony Brook
New Jersey Neuroscience Institute at JFK Medical Center
Stony Brook, NY
Edison, NJ
Helen Barkan, M.D., Ph.D.
Nathan E. Crone, M.D.
Assistant Professor of Neurology
Associate Professor of Neurology
Upstate Medical University Hospital
Johns Hopkins University
SUNY Upstate Medical University
Attending
Syracuse, NY
Johns Hopkins Hospital
Gerhard Bauer, M.D. Baltimore, MD
Professor of Neurology
Edgar dePeralta, M.D.
Medical University Innsbruck
Fellow in Neurology and Clinical Neurophysiology
Innsbruck, Austria
State University of New York Upstate
Richard Bauer, M.D., M.Sc. Syracuse, NY
Oberarzt, Department of Neurosurgery
Frank W. Drislane, M.D.
Medical University Innsbruck
Professor of Neurology
Innsbruck, Austria
Harvard Medical School
Robert L. Beach, M.D., Ph.D. Neurologist
Professor of Neurology Comprehensive Epilepsy Center
Upstate Medical University Beth Israel Deaconess Medical Center
Director, Epilepsy Program and EEG Laboratories Boston, MA
Upstate Medical University Hospital
Barbara Ann Dworetzky, M.D.
Syracuse, NY
Assistant Professor of Neurology
Steve Bild, R. EEG/ EP T., C.N.I.M. Harvard Medical School
Manager of Clinical Services, EEG/Evoked Potentials Lab Chief, Division of Epilepsy, EEG, and Sleep Neurology
Rush University Medical Center Brigham and Women’s Hospital
Chicago, IL Boston, MA
Gastone G. Celesia, M.D. Günter Edlinger, M.Sc., Ph.D.
Professor of Neurology (Retired) CEO
Loyola University of Chicago g.tec Medical Engineering GmbH
Chicago, IL Schiedlberg, Austria
Bernard S. Chang, M.D., M.M.Sc. Jonathan Charles Edwards, M.D.
Assistant Professor of Neurology Associate Professor of Neurosciences
Harvard Medical School Director, Comprehensive Epilepsy Center and Clinical
Comprehensive Epilepsy Center Neurophysiology Laboratories
Beth Israel Deaconess Medical Center Medical University of South Carolina
Boston, MA Charleston, SC
Keith H. Chiappa, M.D. Christian E. Elger, M.D., F.R.C.P.
Associate Professor of Neurology Professor of Epileptology
Harvard Medical School University of Bonn
Director, Electroencephalography Laboratory Head, Department of Epileptology
Massachusetts General Hospital University of Bonn Medical Centre
Boston, MA Bonn, Germany

x
Contributors xi

Ronald G. Emerson, M.D. Richard A. Hrachovy, M.D.


Professor of Clinical Neurology Professor of Neurology
Columbia University College of Physicians and Surgeons Baylor College of Medicine
Attending Neurologist Medical Director of Neurophysiology
New York Presbyterian Hospital St. Luke’s Episcopal Hospital
Columbia University Medical Center Houston, TX
New York, NY
Aatif M. Husain, M.D.
Charles M. Epstein, M.D. Department of Neurology
Professor of Neurology Duke University Medical Center and Neurodiagnostic Center
Emory University School of Medicine Veterans Affairs Medical Center
Emory Healthcare Durham, NC
Atlanta, GA
Kai Kaila, Ph.D.
Bruce J. Fisch, M.D. Professor of Biological and Environmental Sciences
Professor of Neurology Neuroscience Center
University of New Mexico University of Helsinki
Director, Neurodiagnostic Laboratories and MEG Center Helsinki, Finland
University of New Mexico Hospital
Stiliyan Kalitzin, M.D.
Albuquerque, NM
Image Sciences Institute
John N. Gaitanis, M.D. University Medical Center Utrecht
Assistant Professor of Neurology and Pediatrics, Clinical Utrecht, The Netherlands
Warren Alpert School of Medicine at Brown University
Anton Kamp
Director of Pediatric Epilepsy
Biological Centre
Rhode Island Hospital/Hasbro Children’s Hospital
University of Amsterdam
Providence, RI
Amsterdam, The Netherlands
Ali Gorji, M.D.
Andres M. Kanner, M.D.
Institute for Physiology
Professor of Neurological Sciences and Psychiatry
Münster University
Rush Medical College at Rush University
Münster, Germany
Director, Laboratories of EEG and Video-EEG-Telemetry
Jean Gotman, Ph.D. Associate Director, Epilepsy Section
Professor Rush University Medical Center
Montreal Neurological Institute Chicago, IL
McGill University
Peter W. Kaplan, M.B., F.R.C.P.
Montreal, Québec, Canada
Department of Neurology
Riitta Hari, M.D., Ph.D. Johns Hopkins Bayview Medical Center
Academy Professor Baltimore, MD
Brain Research Unit, Low Temperature Laboratory
Andrew D. Krystal, M.D., M.S.
Aalto School of Science and Technology
Professor of Psychiatry and Behavioral Sciences
Espoo, Finland
Director, Quantitative EEG Laboratory
Consultant
Director, Insomnia and Sleep Research Laboratory
Department of Clinical Neurophysiology
Duke University School of Medicine
HUSLAB, Helsinki University Central Hospital
Durham, NC
Helsinki, Finland
Ekrem Kutluay, M.D.
Adam L. Hartman, M.D.
Associate Professor of Neurology
Assistant Professor of Neurology and Pediatrics
Medical College of Wisconsin
Johns Hopkins University School of Medicine
Milwaukee, WI
Attending Physician
Johns Hopkins Hospital Emma Laureta, M.D.
Baltimore, MD Assistant Professor of Neurology
Albert Einstein College of Medicine
Bin He
Attending
Department of Biomedical Engineering and Center for
Montefiore Medical Center
Neuroengineering
New York, NY
University of Minnesota
Minneapolis, MN Alan D. Legatt, M.D., Ph.D.
Professor of Neurology
Susan T. Herman, M.D.
Albert Einstein College of Medicine
Assistant Professor of Neurology
Director of Intraoperative Neurophysiology
Harvard Medical School
Montefiore Medical Center
Beth Israel Deaconess Medical Center
New York, NY
Boston, MA
xii Contributors

Ronald P. Lesser, M.D. Ernst Niedermeyer, M.D.


Professor of Neurology and Neurosurgery Professor Emeritus of Neurology and Neurological Surgery
Johns Hopkins University Johns Hopkins University School of Medicine
Johns Hopkins Medical Institutions Baltimore, MD
Baltimore, MD
Douglas R. Nordli, Jr., M.D.
Fernando H. Lopes da Silva, M.D., Ph.D. Professor of Pediatrics
Emeritus Professor Northwestern University Feinberg School of Medicine
Swammerdam Institute for Life Sciences, Center of Neurosciences Lorna S. and James P. Langdon Chair of Pediatric Epilepsy
Faculty of Science Children’s Memorial Hospital
University of Amsterdam Chicago, IL
Amsterdam, The Netherlands
Marc R. Nuwer, M.D., Ph.D.
François Mauguière, M.D. Professor of Neurology
Head, Department of Functional Neurology and Epileptology UCLA School of Medicine
Lyon University and Lyon Federative Institute of Neuroscience Department Head
Neurological Hospital Clinical Neurophysiology
Lyon, France Ronald Reagan UCLA Medical Center
Los Angeles, CA
Douglas Maus, M.D., Ph.D.
Assistant Professor of Neurology Trudy D. Pang, M.D., M.M.Sc.
SUNY Downstate Medical Center Instructor in Neurology
State University of New York at Brooklyn Harvard University
New York, NY Staff Neurologist
Beth Israel Deaconess Medical Center
Christoph M. Michel, M.D.
Boston, MA
Functional Brain Mapping Laboratory
Neurology Clinic and Department of Basic Neuroscience Alvaro Pascual-Leone, M.D., Ph.D.
University Hospital Professor of Neurology
University of Geneva Harvard Medical School
Geneva, Switzerland Director, Department of Neurology
Berenson-Allen Center for Noninvasive Brain Stimulation
Eli M. Mizrahi, M.D.
Beth Israel Deaconess Medical Center
Chair, Department of Neurology
Boston, MA
Professor of Neurology and Pediatrics
Peter Kellaway Section of Neurophysiology, Department of Neal S. Peachey, M.D., Ph.D.
Neurology Professor of Ophthalmology
Section of Pediatric Neurology, Department of Pediatrics Cleveland Clinic Lerner College of Medicine
Baylor College of Medicine Associate Chief of Staff for Research
Houston, TX Cleveland VA Medical Center
Cleveland, OH
Solomon L. Moshé, M.D.
Professor of Neurology Pediatrics, and Neuroscience Gert Pfurtscheller, Ph.D.
Albert Einstein College of Medicine Emeritus Professor
Director, Pediatric Neurology and Clinical Neurophysiology Laboratory of Brain–Computer Interfaces (BCI-Lab)
Montefiore Medical Center Institute for Knowledge Discovery
New York, NY Graz University of Technology
Graz, Austria
Janet M. Mullington, Ph.D.
Associate Professor of Neurology Rodney A. Radtke, M.D.
Harvard University Professor of Neurology
Beth Israel Deaconess Medical Center Duke University
Boston, MA Medical Director
Duke Hospital Sleep Disorder Center
Christa Neuper, Ph.D.
Duke University Hospital
Professor
Durham, NC
Institute of Psychology
University of Graz James J. Riviello, Jr., M.D.
Head of Institute George Peterkin Endowed Chair in Pediatrics
Laboratory of Brain–Computer Interfaces (BCI-Lab) Professor of Pediatrics and Neurology
Institute for Knowledge Discovery Baylor College of Medicine
Graz University of Technology Director, Epilepsy and Neurophysiology Program
Graz, Austria Director, Neurocritical Service, Section of Neurology
Chief of Neurophysiology
Texas Children’s Hospital
Houston, TX
Contributors xiii

Alexander Rotenberg, M.D., Ph.D. William O. Tatum IV, D.O.


Assistant Professor of Neurology Professor of Neurology
Harvard Medical School Mayo School of Medicine
Attending Staff Physician, Neurology Director, Epilepsy Monitoring Unit
Children’s Hospital Mayo Clinic and Hospital
Boston, MA Jacksonville, FL
Erik Rumpl, M.D. Barbara Tettenborn, M.D., Ph.D.
Professor of Neurology Chair, Department of Neurology
Landeskrankenhaus Kantonsspital St. Gallen
Klagenfurt, Austria St. Gallen, Switzerland
Johannes Gutenberg University
Donald L. Schomer, M.A., M.D.
Mainz, Germany
Professor of Neurology
Harvard Medical School Ab van Rotterdam, Ph.D.
Director, Laboratory for Clinical Neurophysiology Senior Researcher (Retired)
Chief, Comprehensive Epilepsy Program Radiobiological Laboratory
Beth Israel Deaconess Medical Center Radiobiological Institute TNO
Boston, MA Rijswijk, The Netherlands
Margitta Seeck, M.D. Sampsa Vanhatalo, M.D., Ph.D.
Professor Department of Clinical Neurophysiology
Director of the EEG and Epilepsy Unit Children’s Castle
University Hospital of Geneva Helsinki University Central Hospital
Geneva, Switzerland Helsinki, Finland
Megan Selvitelli, M.D. Juha Voipio, Ph.D.
Maine Medical Partners Neurology Professor of Biological and Environmental Sciences
Scarborough, Maine University of Helsinki
Helsinki, Finland
Erw in-Josef Speckmann, M.D.
Professor Emeritus Thoru Yamada, M.D.
Institute of Physiology (Neurophysiology) Professor of Neurology
University of Münster Roy J. and Lucille A. Carver College of Medicine
Münster, Germany University of Iowa
Director, Division of Clinical Electrophysiology
Cornelis Jan Stam, M.D.
University of Iowa Hospitals and Clinics
Department of Clinical Neurophysiology
Iowa City, IA
VU University Medical Center
Amsterdam, The Netherlands Malcolm Yeh, M.D.
Associate Clinical Professor of Neurology
Travis R. Stoub, Ph.D.
Division of Clinical Electrophysiology
Assistant Professor of Neurological Sciences
Roy J. and Lucille A. Carver College of Medicine
Affiliated Scientist
University of Iowa
Rush University Medical Center
Staff Neurologist
Chicago, IL
University of Iowa Hospitals and Clinics
Takeo Takahashi, M.D. Iowa City, IA
Yaotome Clinic
Sendai, Japan
Contents

Preface to the First Edition vi 19 Cerebrovascular Diseases and EEG . . . . . . . . . . . . . . . . . . . . . . 351


Barbara Tettenborn, Ernst Niedermeyer, and Donald L. Schomer
Preface to the Sixth Edition viii
Contributors x 20 Dementia and EEG . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 375
Cornelis Jan Stam

Part I Basic Principles 21 Metabolic Disorders and EEG . . . . . . . . . . . . . . . . . . . . . . . . . . 395


Trudy Pang, Megan Selvitelli, Donald L. Schomer, and Ernst Niedermeyer
1 Historical Aspects of EEG . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1 22 EEG and Craniocerebral Trauma . . . . . . . . . . . . . . . . . . . . . . . . 411
Ernst Niedermeyer and Donald L. Schomer Erik Rumpl
2 Neurophysiologic Basis of EEG and DC Potentials . . . . . . . . . . 17 23 Anoxia, Coma, and Brain Death . . . . . . . . . . . . . . . . . . . . . . . . 435
Erwin-Josef Speckmann, Christian E. Elger, and Ali Gorji Peter W. Kaplan and Gerhard Bauer
3 Cellular Substrates of Brain Rhythms . . . . . . . . . . . . . . . . . . . . . 33 24 The EEG in Patients with Migraine and Other
Florin Amzica and Fernando H. Lopes da Silva
Forms of Headache . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457
4 Dynamics of EEGs as Signals of Neuronal Populations: Ernst Niedermeyer and Donald L. Schomer
Models and Theoretical Considerations . . . . . . . . . . . . . . . . . . . 65
Fernando H. Lopes da Silva
Part IV Clinical EEG in Epilepsy
5 Biophysical Aspects of EEG and Magnetoencephalogram and Related Disorders
Generation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 91
Fernando H. Lopes da Silva With an Appendix by Ab Van Rotterdam 25 Seizures and Epilepsies in the Preterm and Term Neonate ... 465
Emma Laureta, Eli M. Mizrahi, and Solomon L. Moshé
6 Analog Signal Recording Principles . . . . . . . . . . . . . . . . . . . . . . 111
Charles M. Epstein 26 Seizures and Epilepsy in Infants to Adolescents . . . . . . . . . . . . 479
Douglas R. Nordli, Jr., James J. Riviello, Jr., and Ernst Niedermeyer
7 Digital EEG . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Douglas Maus, Charles M. Epstein, and Susan T. Herman 27 Epilepsy in Adults and the Elderly . . . . . . . . . . . . . . . . . . . . . . . 541
Bernard S. Chang, Donald L. Schomer, and Ernst Niedermeyer
8 Polarity and Field Determinations . . . . . . . . . . . . . . . . . . . . . . . 143
Bruce J. Fisch 28 Convulsive Status Epilepticus . . . . . . . . . . . . . . . . . . . . . . . . . . . 563
Frank W. Drislane, Susan T. Herman, and Peter W. Kaplan

Part II Normal EEG 29 Nonconvulsive Status Epilepticus . . . . . . . . . . . . . . . . . . . . . . . 595


Frank W. Drislane, Peter W. Kaplan, and Susan T. Herman
9 Normal EEG and Sleep: Preterm and Term Neonates . . . . . . . 153 30 Anticipating Seizures Based on EEG . . . . . . . . . . . . . . . . . . . . . 645
Eli M. Mizrahi, Solomon L. Moshé, and Richard A. Hrachovy
Fernando H. Lopes da Silva and Stiliyan Kalitzin
10 Normal EEG and Sleep: Infants to Adolescents . . . . . . . . . . . . 163 31 Nonepileptic Attacks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 659
James J. Riviello, Jr., Douglas R. Nordli, Jr., and Ernst Niedermeyer
Megan Selvitelli, Trudy Pang, Donald L. Schomer, and Ernst Niedermeyer
11 Normal EEG and Sleep: Adults and Elderly . . . . . . . . . . . . . . . 183
Bernard S. Chang, Donald L. Schomer, and Ernst Niedermeyer
Part V Complementary and Special Techniques
12 Activation Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
Takeo Takahashi and Keith H. Chiappa 32 Nasopharyngeal, Anterotemporal,
13 Artifacts of Recording . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239 and Sphenoidal Electrodes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 669
Andres M. Kanner, Travis Stoub, and Steve Bild
Barbara Dworetzky, Susan Herman, and William O. Tatum IV
33 Intracranial Monitoring: Depth, Subdural,
Part III Clinical EEG: General and Foramen Ovale Electrodes . . . . . . . . . . . . . . . . . . . . . . . . . . 677
Margitta Seeck, Donald L. Schomer, and Ernst Niedermeyer
14 Patterns of Unclear Significance . . . . . . . . . . . . . . . . . . . . . . . . . 267 34 Electrocorticography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 715
Jonathan Charles Edwards and Ekrem Kutluay Marc R. Nuwer
15 EEG of Degenerative Disorders of the Central 35 Principles and Techniques for Long-Term EEG Recording
Nervous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281 (EMU, ICU, Ambulatory) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 725
John Gaitanis Jean Gotman, Marc Nuwer, and Ronald G. Emerson
16 The EEG in Congenital Malformations of Cortical 36 Infraslow EEG Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 741
Development, Neurocutaneous Disorders, Cerebral Palsy, Sampsa Vanhatalo, Juha Voipio, and Kai Kaila
Autism/Mental Retardation, and ADHD/Learning 37 High-Frequency EEG Activity . . . . . . . . . . . . . . . . . . . . . . . . . . 749
Disabilities of Childhood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299 Jean Gotman and Nathan E. Crone
Mary Repole Andriola
38 Intraoperative Evoked Potential Monitoring . . . . . . . . . . . . . . 767
17 Brain Tumors and Other Space-Occupying Lesions . . . . . . . . 321 Alan D. Legatt
Adam L. Hartman and Ronald P. Lesser
39 Monitoring EEG during Carotid Surgery . . . . . . . . . . . . . . . . . 787
18 The EEG in Inflammatory CNS Conditions . . . . . . . . . . . . . . . 331 Thoru Yamada and Malcolm Yeh
Robert L. Beach, Helen Barkan, and Edgar DePeralta

xiv
Contents xv

40 Polygraphy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809 50 Neurocognitive Processes and the EEG/MEG . . . . . . . . . . . . . 1083


Anton Kamp, Gert Pfurtscheller, Günter Edlinger, Fernando H. Lopes da Silva
and Fernando H. Lopes da Silva

41 Polysomnography: Technical and Clinical Aspects . . . . . . . . . 817 Part VIII EEG and Neurocognitive Functions
Sudhansu Chokroverty, Rodney Radtke, and Janet Mullington
51 Psychiatric Disorders and EEG . . . . . . . . . . . . . . . . . . . . . . . . . 1113
42 Magnetoencephalography: Methods and Applications . . . . . . 865 Andrew D. Krystal
Riitta Hari
52 Technical Aspects of Transcranial Magnetic
Part VI EEG: Neuropharmacology and Anesthesia and Electrical Stimulation of the Brain . . . . . . . . . . . . . . . . . . 1129
Alan D. Legatt, Alvaro Pascual-Leone, and Alexander Rotenberg
43 EEG, Drug Effects, and Central Nervous System Poisoning . . 901 53 Transcranial Magnetic Stimulation (TMS):
Gerhard Bauer and Richard Bauer
Clinical Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1139
Alexander Rotenberg and Alvaro Pascual-Leone
Part VII Evoked Potentials and Event-Related EEG
Phenomena Part IX Computer-Assisted EEG Analysis
44 Event-Related Potentials: General Aspects of 54 EEG Analysis: Theory and Practice . . . . . . . . . . . . . . . . . . . . . 1147
Methodology and Quantification . . . . . . . . . . . . . . . . . . . . . . . 923 Fernando H. Lopes da Silva
Fernando H. Lopes da Silva
55 EEG Mapping and Source Imaging . . . . . . . . . . . . . . . . . . . . . 1179
45 EEG Event-Related Desynchronization (ERD) Christoph M. Michel and Bin He
and Event-Related Synchronization (ERS) . . . . . . . . . . . . . . . . 935 56 Computer-Assisted EEG Pattern Recognition
Gert Pfurtscheller and Fernando H. Lopes da Silva
and Diagnostic Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1203
46 Visual Evoked Potentials and Electroretinograms . . . . . . . . . . 949 Fernando H. Lopes da Silva
Gastone G. Celesia and Neal S. Peachey
57 EEG-Based Brain–Computer Interfaces . . . . . . . . . . . . . . . . . 1227
47 Brainstem Auditory Evoked Potentials (BAEPs) Gert Pfurtscheller and Christa Neuper
and Other Auditory Evoked Potentials . . . . . . . . . . . . . . . . . . . 975 58 Multimodal Monitoring of EEG and Evoked Potentials . . . . 1237
Gastone G. Celesia Gert Pfurtscheller
48 Somatosensory-Evoked Potentials: Normal Responses,
Abnormal Waveforms, and Clinical Applications
in Neurologic Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1003 Index 1243
François Mauguière

49 Evoked Potentials in Children and Infants . . . . . . . . . . . . . . . 1057


Aatif M. Husain
Part I Basic Principles
CHAPTER

Historical Aspects of EEG


ERNST NIEDERMEYER AND DONALD L. SCHOMER 1
DISCOVERY OF ELECTRICAL PHENOMENA electricity) and placed the emphasis on physics—on his “pile,”
the first battery (around 1800). This bimetallic pile was a gen-
Thales from Miletos has been credited with the discovery of erator capable of producing a steady flow of electricity. Volta’s
static electricity produced by friction (rubbing fur or glass with view more or less prevailed in this hotly debated argument. The
silk). He was one of the pre-Socratic “natural philosophers” of laws governing flowing electricity were soon discovered by
Greece (around 620–550 BC) and considered water the origin Georg Ohm in 1827.
of all things. Thus, friction was recognized as the generator of a Nevertheless, Galvani’s belief in “animal electricity” was not
phenomenon that derived its name from the Greek work “elec- lost with other discarded false ideas. There still remained the
tron,” which stands for amber. This discovery fell into a dor- nagging question of an active electrical contribution of animal
mant stage for more than two millennia. muscle tissue.
Around 1600, William Gilbert began to study the electrical
properties of various substances, and Otto von Guericke
(1602–1686) invented the friction machine to create electrical BEGINNINGS OF ELECTROPHYSIOLOGY
fields. This machine eventually found its way into doctors’
offices and even university hospitals. Its electrical field would The introduction of the galvanometer has been associated
make a patient’s hair stand up, creating a strong impression on chiefly with the name of Nobili in Florence; this instrument was
a psychologically gullible patient. These friction machines now refined in 1858 by William Thompson (Lord Kelvin) in
ornament high school laboratories and technical museums. In England (O’Leary and Goldring, 1976). These galvanometers
the 17th and 18th centuries, the friction machine taught invalu- would faithfully demonstrate continuous electrical currents
able lessons on attraction and repulsion of charged bodies, on and their variations in intensity but failed in the detection of
conductors and nonconductors, and on the rather questionable instantaneous electrical phenomena.
dualism of positive and negative electricity. Carlo Matteucci (1811–1868) in Bologna and Emil Du Bois-
A new and very important piece of electrical equipment Reymond (1818–1896) in Berlin became the major proponents of
entered the scene in 1746 when the Leyden jar was introduced an electrophysiologically based physiology of the nervous system.
by Pieter van Musschenbroek (following the earlier work of (The French name of Du Bois-Reymond indicates the Huguenot
Ewald von Kleist). This invention resulted in the storage of elec- origin of this Prussian investigator.) Du Bois-Reymond coined the
tricity, and its upshot, the condenser or capacitor, turned into term negative variation for a phenomenon occurring during mus-
an indispensable part of modern electronics. Benjamin cle contraction when the galvanometer indicated an unexpected
Franklin’s bold experiment caught electrical discharges of a decrease in current intensity (O’Leary and Goldring, 1976). This
thunderstorm in a Leyden jar. term was later resurrected in earliest electroencephalogram (EEG)
What the friction machine could generate, the Leyden jar research (Caton, 1875) and with the discovery of the “contingent
could store. Its sudden discharge was used in many experiments negative variation” (Walter, 1964).
(O’Leary and Goldring, 1976). Hermann von Helmholtz (1821–1894) accurately measured
The role of static electricity in medicine appeared to be for- the velocity of nerve conduction, which had been vastly overes-
gotten for about 150 years and became resurrected with the timated up to that time. The electrodes used in physiologic
introduction of the defibrillating cardioversion by William B. research were improved and made nonpolarizable (Du Bois-
Kouwenhoven and his coworkers in the 1950s and 1960s; this Reymond). The concept of “action current” was introduced by
approach may hold promises for cerebral applications L. Hermann (1834–1919) and thus clarified Du Bois-Reymond’s
(Niedermeyer, 2003a). negative variations found during muscle contraction. Julius
A serious scientific controversy developed in Italy between Bernstein (1839–1917) proposed a membrane theory of nerve
Luigi Galvani (1737–1798), a professor at the University of tissue, which ultimately was elucidated as late as 1939 and the
Bologna, and Alessandro Volta (1745–1832) in the wake of following years by A. L. Hodgkin and A. F. Huxley in England.
Galvani’s discovery of frog leg contractions within an electrical Against this background of strongly evolving electrophysiology
circuit and especially in the presence of a thunderstorm (1780). of the nervous system, the first observation of EEG-like electri-
Volta doubted the biologic nature of the contraction (animal cal brain activity took place.

1
2 Part I ■ Basic Principles

CATON: THE FIRST ATTEMPT AT THE Caton also described a few more interesting observations.
ELECTRICAL ACTIVITY OF THE BRAIN He noted that the external surface of the gray matter was posi-
tive in relation to deep structures of the cerebrum. He also
Richard Caton (1842–1926) (Fig. 1.1) was a physician practic- noted that the electric currents of the cerebrum appeared to
ing in Liverpool who became deeply interested in electrophysi- have a relation to underlying function: “When any part of the
ologic phenomena and eventually received a grant from the grey matter is in a state of functional activity, its electric current
British Medical Association to explore electrical phenomena of usually exhibits negative variation.” Thus, Caton has also been
the exposed cerebral hemispheres of rabbits and monkeys. credited with pioneer work on evoked potential. Furthermore,
According to Brazier (1961), Caton presented his findings to the the difference in polarity found between cortical surface and
association on August 24, 1875, and a very short report of 20 deeper areas could be interpreted as the discovery of the “steady
lines subsequently appeared in the British Medical Journal. A potential” (“DC potential”), but it might be wise to refrain from
more detailed report was presented in the same journal in 1877 such statements that cannot be fully supported by the evidence.
on experiments of more than 40 rabbits, cats, and monkeys, the With regard to the fluctuations, Geddes (1987) pointed out that
rabbit having been principally employed. Caton’s galvanometer had a very limited frequency response
Caton used a galvanometer. A beam of light was thrown on range from 0 to 6 Hz.
the mirror of the galvanometer and reflected on a large scale Caton found some measure of success and recognition with
placed on the wall. With this type of visualization, Caton found this work and held the chair of physiology at the University
that “feeble currents of varying direction pass through the mul- College of Liverpool from 1884 to 1891, when he resigned from
tiplier when the electrodes are placed on two points of the this post. Later he became dean of the medical faculty and, in
external surface, or one electrode on the grey matter, and one 1907, mayor (Lord Mayor) of Liverpool. The electrical activity of
on the surface of the skull.” This sentence is regarded as indicat- the brain did not occupy a predominant position in his further
ing the birth of the electrophysiologram because one can endeavors. Even though Caton became an EEG research dropout,
assume that EEG phenomena made the needle move from one his bold work will always remain a milestone in the history of the
direction to the other. (The suffix “gram” naturally is out of electrical activity of the brain. (More information on Caton’s life
place since “graphein” means “to write” and there was no writ- and work is found in Mary Brazier’s (1961) fine account.)
ten recording.) Even though artifacts could have played a major
role, Caton deserves credit for the discovery of the fluctuating EASTERN EUROPEAN STUDIES OF
potentials that constitute the EEG. ELECTRICAL BRAIN ACTIVITY
The time was ripe for further studies of electrical phenomena of
the cerebrum. Concurrent with Caton’s epochal work of 1875,
physiologists of Eastern Europe began to demonstrate their
independent observations and discoveries concerning the brain
and its electrical activity. Another discovery of the 1870s had
an incomparably greater impact on the neuroscientific world
than Caton’s demonstration of electrical activity of the brain.
The capability of the human cerebral cortex to be electrically
stimulated was discovered by G. Fritsch (1838–1927) and Julius
Eduard Hitzig (1838–1907) in a joint study in 1870. According
to O’Leary and Goldring (1976), an unusual observation had
prompted Fritsch in his work: he had observed contralateral
muscle contractions during dressing of an open brain wound in
the Prussian–Danish War of 1864. The work of Fritsch and
Hitzig was furthered by D. Ferrier and G. F. Yeo in 1880, who
performed electrical stimulations of the cerebrum in apes and
also in a patient who was operated on for a brain tumor. The
repercussions of the stimulation studies were considerable since
many investigators of that time held the view that the entire
cerebrum is a homogeneous organ that harbors mental functions.
The response of the cortex to electrical stimulation probably
was a special incentive for the study of its spontaneous electri-
cal phenomena. This incentive was particularly strong in
Eastern Europe, that is, in laboratories of Russian and Polish
Figure 1.1 Richard Caton at the time of his work on the electrical universities. (In spite of the important historical ethnic and
activity of the brain. (From Brazier MAB. A History of the Electrical national differences, the fact cannot be ignored that most of
Activity of the Brain. The First Half-Century. London: Pitman; 1961, with Poland was part of the Czarist Russian Empire throughout the
permission from Macmillan.) 19th century.)
Chapter 1 ■ Historical Aspects of EEG 3

Vasili Yakovlevich Danilevsky (1852–1939) was only 25 years the most important advances was Willem Einthoven’s introduc-
old when he finished his thesis entitled “Investigations into the tion of the string galvanometer in 1903, a very sensitive instru-
Physiology of the Brain” (Danilevsky, 1877), written at the ment that required photographic recording and became the
University of Kharkov. This work was based on electrical stim- standard instrumentation for electrocardiography at the turn of
ulation as well as on spontaneous electrical activity in the the century.
brains of animals. Thus, Danilevsky walked in Caton’s foot- Kaufman’s work and life are portrayed in Brazier’s (1961)
steps; in 1891, he gave full credit to Caton’s priority. Mary historical account. Kaufman expressed the view that an epilep-
Brazier (1961) comments on the disappointment of Danilevsky tic attack would have to be associated with abnormal electrical
who saw his high hopes unfulfilled as far as the spontaneous discharges, and he studied the effects of cortical electrical stim-
electrical activity of the brain was concerned; he had expected ulation. With World War I, he took the name of Rostoutsev and
better correlation with psychic and emotional processes. He worked mainly at the University of Baku.
remained deeply involved in brain physiology and published an Pravdich-Neminsky began recording electrical brain activity
extensive textbook of human physiology in 1915. He was not of animals in 1912 with the string galvanometer. As Brazier
the only EEG researcher with shattered hopes in the field of (1961) has pointed out, his recordings, published in 1912, were
psychophysiology. the first pictorial demonstration of EEG and appeared 2 years
The life and work of Adolf Beck (1863–1939) have been earlier than Cybulski’s tracings. Pravdich-Neminsky recorded
described in great detail by Brazier (1961). Beck worked in the EEG from the brain, the dura, or the intact skull of the dog.
Kraków as well as in Lwow (the Polish province of Galicia, at He described a 12 to 14/sec rhythm under normal conditions
that time a part of the Austrian–Hungarian monarchy). With and marked slowing under asphyxia. Furthermore, he coined
nonpolarizable electrodes, Beck investigated the spontaneous the term electrocerebrogram (Fig. 1.2).
electrical activity of the brain in rabbits and dogs. He observed The achievements of the Eastern European neuroscientists
the disappearance of rhythmical oscillations when the eyes were during those 50 years preceding the outbreak of World War I fill
stimulated with light and thus became a forebear of Berger’s us with awe and clearly demonstrate their special talent for elec-
discovery of alpha blocking. His work became widely known trophysiologic neurophysiology. Limiting discussion to EEG
due to its publication in the Centralblatt. history can show only the tip of the iceberg. To assess the true
To present a chronologic account of the events, let us leave strength of their neuroscientific institutions, one must mention
Eastern Europe for a moment, but not Vienna. In 1883, Ernst investigators in somewhat related electrophysiologic areas. Ivan
Fleischl von Marxow (1846–1891) deposited a sealed letter at Michailovich Sechenov (1829–1905) appears to be founder of
the Imperial Academy of Sciences in Vienna that contained this powerful school of eminent neurophysiologists. He studied
observations on cerebral electrical activity recorded over the the electrical activity of the spinal cord and oblongata in the
visual cortex in various species of animals. He did not observe frog and was a predecessor of Pavlovian thought. Nikolai
oscillatory activity. He claimed priority when Beck in 1890 pub- Yevgenevich Wedensky followed Sechenov as the chair and pro-
lished his data but was not aware of earlier work done by Caton fessor of physiology at St. Petersburg (known for the concept of
and Danilevsky. The oddity of this episode is underscored by Wedensky inhibition). Vladimir Efimovich Larionov, also
more recent historical accounts (Brazier, 1961; O’Leary and working in St. Petersburg, conducted beautiful studies of the
Goldring, 1976) that indicate that Fleischl von Marxow’s work auditory cortex in the dog.
was not of first-rate quality. This does not detract from the ren- The greatest Russian neuroscientist was also the most eminent
aissance-man versatility of this Austrian physiologist, who was clinical neurologist of his country: Vladimir Mikhailovich
well versed in linguistics (even Sanskrit), swimming, hunting, Bechterev (also “Bekhterev”) (1857–1927). He occupied the chair
and mountain climbing. of psychiatry in St. Petersburg, which included the field of clini-
Exciting new studies were being conducted in Eastern cal neurology. He was a disciple of Du Bois-Reymond, Paul Emil
European universities. Napoleon Cybulski (1854–1919), who Flechsig, and Wilhelm Wundt, and also worked at Charcot’s clinic
was Beck’s teacher in Kraków and an internationally renowned in Paris. The influence of Wundt prompted Bechterev’s associa-
leader in general physiology, presented experimental electroen- tive reflexology (I treasure his work “Allgemeine Grundlagen der
cephalographic studies in graphical form by using a gal- Reflexologie des Menschen,” Deuticke, Leipzig, 1926, even
vanometer with a photographic attachment. He provided EEG though I can hardly agree with his “objective study of personal-
evidence of an epileptic seizure in the dog caused by electrical ity”). He combined his clinical work and private practice with
stimulation. tireless psychophysiologic methods. A photograph with one of
Two Russian physiologists made further studies along these his two assistants shows Bechterev wearing a thick winter coat in
lines: Pavel Yurevich Kaufman (1877–1951) and Vladimir his institute in Leningrad, giving testimony to the icy cold in the
Vladimirovich Pravdich-Neminsky (1879–1952). Prior to the unheated laboratory. “Functional anatomy of the brain, experi-
discussion of their work, a few words must be said about tech- mental psychology and clinical neurology were the three fields in
nological developments. The d’Arsonval galvanometer featured which Bekhterev carved out a place for himself” (Yakovlev, 1953).
a mirror mounted on a movable coil; light focused on the mir- The Soviet regime had to choose between Bechterev and
ror was deflected when a current passed the coil. The capillary Ivan Petrovich Pavlov (1849–1936), a physiologist who had won
electrometer was introduced by G. Lippmann and H. J. Marey the Nobel Prize in 1904 for his early work on conditioned
(for further details, see O’Leary and Goldring (1976)). One of reflexes. Pavlov was the choice, and the magic of conditioned
4 Part I ■ Basic Principles

Figure 1.2 The first photographs to be published of electroencephalograms. In the upper record Neminsky shows (in the
third trace) the brain potentials of a curarized dog with the pulsations from an artery in the brain recorded above them.
In the lower record the sciatic nerve is being stimulated from time to time, and the decrease in activity noted by Neminsky
can be seen. The record reads from right to left, line I being a time marker in one fifth of a second, line III the galvanometer
string, and line V the signal for stimulation. (From Pravdich-Neminsky VV. Ein Versuch der Registrierung der elektrischen
Gehirnerscheinungen. Zbl. Physiol. 1913;27:951–960, with permission from Dr. Mary Brazier and Macmillan.)

reflex overshadowed all Soviet neurophysiology by highest The Western neurophysiologists followed attentively the
decree (even though Pavlov himself was highly critical of the work of their neuroanatomical confreres and the great contro-
regime). The Pavlovian concept was closer to the ideology of versy between network theories (the nerve cells forming a felt-
dialectic materialism, and this maxim with all its intolerant like net) and the neuron theory (the neuron representing a
dogmatism outlasted Pavlov’s death by two decades. This unit). The net theory was supported by Joseph von Gerlach and
ideopolitically governed form of neuroscience stifled all by Camillo Golgi, the discoverer of the silver chromate staining
progress of customary neurophysiology. The world leadership method, but the neuron theory with its great principal propo-
in EEG and related fields quickly crumbled, accompanied by a nent Santiago Ramón y Cajal (1852–1934) proved to be victo-
terrifying decline of a dogmatically governed neurophysiology. rious in this struggle despite further attempts of new
generations of “reticularist” believers in a continuous network
DEVELOPMENTS IN WESTERN of nerve cells.
AND CENTRAL EUROPE In a similar manner, cerebral localizationists struggled
against antilocalizationists. In Germany, Friedrich Leopold
Electroencephalographic research was in a dormant state in Goltz (1834–1902) removed the cerebral hemispheres in the
Western and Central Europe while it was flourishing in Eastern “dog without cerebrum” living in a state of extreme lethargy
European countries. This is quite amazing because neurophys- and mental inertia (Goltz, 1888). H. Rothmann (1923) per-
iology in general was healthy and well outside Russia and her formed similar investigations. This type of research was aimed
neighbors, but the ancestral lineage from Galvani to Du Bois- at the working of the cerebral hemispheres as a whole and de-
Reymond and Caton broke off and the neurophysiological emphasized the aspects of cortical localization. As it was
field was watered by rivulets of different orientation. Thus, the pointed out previously, the cerebral stimulation studies of
work of Fleischl von Marxow lies like an erratic patch in the Fritsch, Hitzig, Ferrier, and Yeo initiated a new era of interest in
vast field. cortical localization.
Chapter 1 ■ Historical Aspects of EEG 5

In this period—the last third of the 19th and the dawning of


the 20th century—the work of Charles Scott Sherrington
(1857–1952), performed in Liverpool and Oxford, became
most influential in the development of a modern Western type
of reflexology. The Integrative Action of the Nervous System
(Sherrington, 1906) was based on a series of lectures held at Yale
University in 1904. The scope of this work reaches from reflex-
ology to decerebrate rigidity, from motor cortex to sensory
function, while the issue of mental function is being skirted
with the modesty of a truly great neuroscientist. Inhibition is
one of the great Sherringtonian discoveries. Even Ramón y
Cajal’s net of independent synaptically connected neurons
stood solely in the service of neural excitation. (Incidentally, the
term synapse was introduced by Sherrington.)
It is unfortunate that this greatest master of neurophysiology
stood miles away from electrophysiologic thought. His work
was based chiefly on ablation techniques. He may hardly ever
have given a thought to EEG methods even though he lived a
full active life. His disciples—to name only Edward Liddell and
Derek Ernest Denny-Brown—held similar views, while in
Cambridge electrically oriented neurophysiology found its
greatest proponent in Edgar Douglas Adrian (1889–1977),
whom we discuss later in his relationship to Hans Berger’s
work.

HANS BERGER AND THE HUMAN Figure 1.3 Hans Berger. (From Kolle K. Hans Berger. In: Kolle K, ed.
Grosse NervenŠrzte. Vol. 1. Stuttgart: Thieme; 1956: 1–6.)
ELECTROENCEPHALOGRAM
Hans Berger (1873–1941) (Fig. 1.3), the discoverer of the
human EEG, was a neuropsychiatrist. What neuropsychiatry capillary electrometer after Lippmann, but the results were dis-
really meant in those years is poorly understood nowadays. appointing. Naturally, Berger was aware of the scanty pertinent
Neurology and psychiatry formed one specialty, one discipline, literature from Caton to Cybulski and Pravdieh-Neminsky. His
in Germany, Austria, and a considerable number of other coun- studies of the human EEG started in 1920; the introduction to
tries. Neuropsychiatric departments at university hospitals and Gloor’s authoritative translation of Berger’s work contains a
other institutions consisted of neurologic and psychiatric plethora of interesting details (Gloor, 1969).
floors; medical specialty training meant rotation from one dis- Every electroencephalographer should be familiar with
cipline to the other, and a the head of one department was sup- Berger’s work, an undertaking that has been greatly facilitated
posed to be a master of both disciplines. Pure neurology was by Gloor’s English translation. It is true that the original
just beginning to emerge as a special discipline in the German- German text is cumbersome and does not make easy reading,
speaking countries (with the work of Wilhelm Erb which is probably a reason for the very slow acceptance of
(1840–1921), Max Nonne (1861–1959), and the incomparable Berger’s work. Those 14 reports bear the same title: “On the
Otfrid Foerster who, like Hans Berger, lived from 1873 to 1941). Electroencephalogram of Man.” Surely, a more attractive title
Berger was not a leader, neither in neurology nor in psychi- would have helped somewhat. Berger’s humanistic educational
atry. Without his pioneering EEG work, his name would have background becomes obvious in the rejection of the term elec-
been forgotten. Biographic sketches (especially Kolle, 1956) trocerebrogram of Pravdich-Neminsky for strictly linguistic
portray Berger as an extremely meticulous and conscientious reasons: the “ugly” mixture of Greek (“electro,” “gram”) and
person, somewhat aloof in his contact with his patients, a very Latin (“cerebro”) fragments. What Berger proposed in German
strict and authoritarian department head, and an “anima can- was the term Elektrenkephalogram (sic) since the root
dida” (a pure soul), a hard-working professor without any enkephalo from the Greek is linguistically more correct than
interest in faculty schemes and diatribes. He hardly ever encephalo.
attended the annual meetings of the German neuropsychiatric Before Berger’s work as such is brought into focus, we must
society. His electroencephalographic work was carried out in a discuss his electrophysiologic instrumentation. He used a string
small and very primitive laboratory. His first scientific interest galvanometer starting in 1910—first with the Einthoven type,
aimed at the cerebral circulation; plethysmographic methods later with the smaller Edelmann model, and after 1924 with
were used in patients with skull defects. From 1902 to 1910, he the larger Edelmann model. In 1926, Berger started to use the
studied the electrical activity of the cerebrum in the dog with a more powerful Siemens double-coil galvanometer (attaining a
6 Part I ■ Basic Principles

sensitivity of 130 V/cm; Grass, 1984). With this instrument depression, which remained undiagnosed. He ended his life by
and the use of nonpolarizable pad electrodes, Berger recorded suicide on June 1, 1941 at the age of 68. External factors may
the human EEG tracings shown in his first report of 1929. have contributed to his depression: in addition to his forced
The records were made on photographic paper with record- retirement at 65 (a few additional “years of honor” were granted
ings from 1 to 3 minutes’ duration. Berger used a bipolar record- to most retiring directors of university institutions), he also felt
ing technique with fronto-occipital leads for his one-channel challenged by a group of independent EEG workers at the
EEG tracings along with simultaneous electrocardiogram Institute of Brain Research at Berlin-Buch. This group was led
(ECG) recording and a time marker. In 1932, he received an by A. E. Kornmüller and produced excellent experimental EEG
oscillograph from Siemens but was unable to obtain further work, which is discussed later. Kornmüller might have had the
amplifiers with oscillographs in order to obtain multichannel better connections to the government institutions in Berlin,
recordings. and the highly sensitive and often insecure Berger was afraid
Studies of the human EEG began in 1924. Appointments that his discovery was being taken away from him by his more
were made for several patients with large skull bone defects aggressive colleagues in Berlin-Buch.
(there was no scarcity of such patients in post-World War I Berger was a very complex person and investigator. It was
Germany). On July 6, 1924, the small Edelmann string gal- pointed out previously that he did not excel clinically, neither
vanometer showed oscillations presumably coming from the as psychiatrist nor as neurologist (even though he was very
underlying brain. In 1925, Berger recognized that skull defects interested in cerebral localization and particularly in the local-
were not necessarily advantageous to obtaining a recording ization of brain tumors). Berger also developed very unscien-
because of thickening of dura, postoperative adhesions, etc., tific ideas about the nature of the EEG, even though he was a
and he found that recordings could be made just as well (or meticulous scientist in his EEG work. The driving force in all
even better) through the intact skull and scalp. Between 1926 his research work was the quest for the nature of the all-pow-
and 1929 Berger obtained good records with alpha waves; the erful force of mental energy (“psychische Energie”). An early
double-coil galvanometer was crucial for these observations. personal experience convinced him that such a mental
The data were often uncertain and, in 1928, Berger was beset energy—even capable of transmitting thoughts and emotions
with doubts concerning the authenticity of his observations from person to person—does exist. According to Berger’s con-
(according to his diary entries, very impressively demonstrated cept, influenced by the Danish physiologist Alfred Lehmann in
by Jung, 1963). 1901, mental energy is thought to be a partial product of meta-
The first report of 1929 features the alpha rhythm and the bolic energies (warmth and electricity being the other two
alpha blocking response (naturally along with a description of products). This concept gives the EEG waves the eerie charac-
the smaller beta waves). Chlorinated silver needle electrodes, ter of messengers within the mental activities, even as messen-
platinum wires, and zinc-plated steel needles were used in those gers from person to person.
years (Fig. 1.4). The bold first report of 1929 produced no Friedrich Rueckert (1788–1866), a great German poet of the
“waves” until a confirming report came from Adrian in Romantic period, was Berger’s maternal grandfather. Behind
Cambridge (Adrian and Matthews, 1934). Throughout the the strict directorial facade of Berger was the gentle soul of a
1930s, Berger’s reports on the human EEG contained veritable highly vulnerable man. It was the psychophysiologist Berger
gems: studies of fluctuation of consciousness, first EEG record- who searched for the correlate of mental energy and, on this
ings of sleep (the first recording of spindles), the effect of voyage, he found the human EEG. It was one of those
hypoxia on the human brain, a variety of diffuse and localized “Columbus syndromes”: that a discovery is made as a by-
brain disorders, and even an inkling of epileptic discharges. product of a search for a different goal. Jung (1963) noted that
Eventually, Berger was invited to an international congress of Berger pursued his goal with the extremely powerful energy of
psychologists in Paris in 1937 and to Bologna where the bicen- a dilettante who had found a concept. Specialists, like the excel-
tennial of Galvani’s birthday was celebrated (also 1937). His lent physiologist (and electrophysiologist) Wilhelm
relationship to the Nazi regime was not good, and Berger was Biedermann, in Jena were convinced that Berger’s dilettantism
most unceremoniously made a professor emeritus at earliest would lead nowhere, but it was the dilettante and not the sea-
convenience, in 1938. This was indeed a hard blow to his plans soned specialist who emerged victorious. Even though the EEG
for further electroencephalographic studies and, in the wake of is not exactly what Berger assumed, his contribution was the
a flu-like disease, he evidently developed a severe endogenous greatest in the history of electroencephalography.

Figure 1.4 The first recorded electroencephalogram of a human.


The lower line is a 10 cycles/ sec sine wave for use as a time marker.
The upper line is the recording from Berger’s young son made in
1925. (From Berger H. Über das Elektrenkephalogramm des
Menschen. 1st report. Arch Psychiat Nervenkr. 1929;87:527–570,
with permission from Dr. Mary Brazier and Macmillan.)
Chapter 1 ■ Historical Aspects of EEG 7

BERGER’S CONTEMPORARIES of personal gratitude. Decades earlier, Vogt successfully treated


an ailing Krupp—with hypnosis. Indeed, Vogt also used to be
The Berliner Group a master of this method!) Hugo Spatz replaced Vogt and con-
The Institute of Brain Research (Hirnforschungs-Institut) in siderably changed the goals of research. Toennies stayed with
Berlin-Bush harbored a group of ambitious and energetic H. S. Gasser at the Rockefeller Institute in New York, where he
investigators in various neurosciences. Oskar Vogt constructed the first cathode follower to record from high-
(1870–1959), one of the great neuroanatomists and neu- resistance electrodes. This was the birth of microelectrode
ropathologists of his time and a remarkably independent recording, which developed into an enormously powerful sci-
thinker with a wide intellectual horizon, was the director of the entific tool in the 1950s and 1960s.
institute. In 1936 he lost his “directorship for lifetime” when the Kornmüller’s work declined after World War II, when he
Nazi government became aware of Vogt’s activities at a similar appeared to be obsessed with a totally unproven theory of glia as
institute in Moscow and his reluctance to get rid of Jewish the generator of slow brain potentials. Jung, however, developed
coworkers (Hassler, 1959). into one of the greatest electroneurophysiologists of his time.
The Berliner Institute (a section of the Kaiser-Wilhelm
Institutes) was composed of a variety of departments. In this Developments in Great Britain
context, the Department of Physiology under M. H. Fischer and Edgar Douglas Adrian (Baron of Cambridge and, as such, Lord
the Department of Electrophysiology under A. E. Kornmüller Adrian) (1889–1977) was not only one of the greatest electrophys-
must be singled out. These departments enjoyed the collabora- iologic neurophysiologists of the 20th century; his name is also
tion of an outstanding physicist and electronic engineer, J. F. intimately associated with the discovery of the EEG because of his
Toennies (1902–1970), a personal friend of Oskar Vogt (both confirmation of Berger’s observations (Adrian and Matthews,
coming from the town of Husum in Holstein). 1934). He showed his colleagues his own beautiful alpha rhythm
Toennies built the first ink-writing biologic amplifier for the and the blocking effect due to eye opening, but, alas, his great
recording of brain potentials. While in New York as a fellow of electronic wizard, Brian Matthews, apparently had a low-voltage
the Rockefeller Foundation in 1932, he designed the differential EEG with no alpha rhythm. Due to some strange quirk, Adrian’s
amplifier—the still all-important principle of EEG amplifica- recording from the head ganglion of a water beetle happened to
tion—but he shares this achievement with Brian Matthews, be indistinguishable from Adrian’s alpha rhythm and was blocked
Adrian’s ingenious coworker whose work is discussed in the in the same manner, namely by light falling on the beetle’s eyes.
next section. The Adrian-water beetle similarity and the Adrian-Matthews EEG
The collaboration with Toennies gave the Berlin group a dissimilarity must have been terribly confusing to the onlookers
much better tool for EEG research in comparison with Berger’s (also see Adrian, 1936).
instrumentation. Kornmüller quickly recognized the impor- Adrian was already a neurophysiologist of great prestige
tance of recordings from a greater number of electrodes. His when he confirmed Berger’s data. He had been credited with
EEG studies in the humans placed particular emphasis on the the demonstration of single sensory nerve fiber potential and
differences between given regions of the cerebrum the analysis of unit activity, which resulted in the Adrian–Bronk
(“Hirnrindenfelder”) (Kornmüller, 1932, 1933, 1935, 1937). His law (Adrian and Bronk, 1929). Incidentally, his collaborator
studies of the clinical significance of EEG (Kornmüller, 1944) Detlef Bronk became president of Johns Hopkins University in
appear to be somewhat pale when compared with the impor- later years. Prior to Matthews, Keith Lucas had been Adrian’s
tance of his earliest experimental EEG work carried out with brilliant electronic engineer and experimental coworker.
Fischer and also with H. Löwenbach, who later came to the W. Grey Walter became the pioneer of clinical electroen-
United States, where he became one of the earliest EEG pioneers. cephalography in England, and his discovery of foci of slow
In those early studies, the EEG was obtained from the cortex of activity (delta waves, named by Walter) generated enormous
animals following poisoning with convulsive substances. This is clinical interest in the new method. Grey Walter, however, was a
the first EEG work focusing on epileptic manifestations and the Ph.D. basic scientist. The fact that he was principally an aca-
first demonstration of epileptiform spikes (Fischer, 1933; demic scholar, not a clinician, could have laid the foundation for
Fischer and Löwenbach, 1934a,b; Kornmüller, 1935). the aversion of England’s great neurologists toward the method
Oskar Vogt (his wife Cécile also being known as a great neu- of EEG, which in the following years was either ignored or left to
ropathologist) developed a concept of strict cerebral compart- Ph.D. electroencephalographers in the laboratory. We will find
mentalization in sharply separated areas. He showed indeed Grey Walter again in later decades; let it be said that he was one
extremely impressive boundaries between healthy and dis- of the most brilliant minds in all neurosciences—an independ-
eased areas in the hippocampus (Vogt and Vogt, 1937) and ent thinker, a powerful writer, and quite often a man nearly con-
conceived the cortex as divided into about 200 regions with sumed by the flame of his own brilliance. He founded a small
precise demarcation from field to field. This concept became a but very effective school in Bristol at the Burden Institute.
powerful leitmotiv for Vogt’s coworkers, and Kornmüller’s EEG
work clearly shows the marks of his authoritarian boss. Developments in France and Belgium
Richard Jung joined this group in 1937 when Vogt was Thus far, France seems to be unduly neglected in this historical
already fired. (The tycoon Alfred Krupp provided the Vogts overview. It had its own proud neurophysiologic schools in the
with a privately built institute in the Black Forest in his expression 19th century, and the names of François Magendie (1783–1855)
8 Part I ■ Basic Principles

and, above all, Claude Bernard (1813–1878) belong in the pan- America Enters the Scene
theon of neuroscience. Around 1935, the center of gravity in the still modest bulk of
A fine school of early electroencephalographers developed in EEG work started to shift from Europe to North America.
Paris in the 1930s. A. Fessard at the Collège de France must be Fascinating new reports came from the United States. European
singled out as the most towering figure. Together with G. investigators started to travel across the Atlantic, and even Hans
Durup, he also confirmed the results of Berger. Durup and Berger was about to accept an invitation to the United States in
Fessard even used EEG in the study of conditioned reflexes. 1939, when the beginning of World War II thwarted his plans.
Clinical electroencephalography started in France under the In the pre-Berger development of experimental EEG studies,
aegis of A. Baudouin and G. Fischgold. Fischgold had come America had not played any role. Schwab (1951) reports that, in
from Rumania, developed into a leading clinical electroen- 1918, a medical student of Harvard Medical School, Donald
cephalographer, and—what an unusual combination—became McPherson, worked under the eminent physiologist Alexander
a leader in neuroradiology. Baudouin was the key figure in Forbes. When McPherson placed two electrodes on the exposed
the invitation of Berger to Paris in 1937. brain of a cat and ran the output into a string galvanometer, he
Neighboring Belgium was the home of a giant in electro- saw rhythmical 10/sec EEG activity. This finding was rejected as
physiologic neurophysiology: Frederic Bremer (1892–1982) from an artifact by Forbes. Was Forbes completely unaware of the
the Université Libre of Brussels. Bremer quickly recognized the work from Caton to Pravdich-Neminsky?
usefulness of EEG methods in the experimental investigation of The rise of American EEG work to international fame is cus-
the brain. He recognized the influence of afferent signals on the tomarily associated with the work of Hallowell Davis, Frederic
state of vigilance and compared his feline preparation called A. Gibbs, and Erna Gibbs at Harvard and also with Herbert
“cerveau isolé” (with midbrain transection) with the “encéphale Jasper’s work at Brown University in Providence, Rhode Island.
isolé” resulting from transection at the boundary between the According to O’Leary and Goldring (1976), A. J. Derbyshire, a
medulla oblongata and cervical cord. The former preparation graduate student of Hallowell Davis, brought Berger’s paper of
would produce permanent coma; the latter would cause a vari- 1929 to Davis’s attention. Derbyshire, Pauline Davis, and H. N.
able state of vigilance, with waking and sleeping demonstrated Simpson then tried in vain to demonstrate their own alpha
on the EEG recording. In other words, trigeminal-sensory, audi- rhythms. There were finally shouts of joy when Hallowell Davis
tory, visual, and probably also olfactory influences would help himself was found to have a good alpha rhythm. Otherwise, the
to keep the (artificially ventilated) encéphale isolé preparation in first human EEG study in America would have been a negative
a waking–sleeping rhythm (Bremer, 1935). one. This work was done in 1934, just before human EEG stud-
The greatness of this investigator must be reemphasized, ies started to mushroom in the United States.
especially in today’s era of short memory. Whoever reads his The EEG, however, had been used for animal experiments
study entitled “Cerebral and Cerebellar Potentials” (Bremer, for some years in the United States, starting with Bartley and
1958) will roughly understand the dimensions of this Newman (1930, 1931) and Bartley (1932), who produced EEG
neurophysiologist. tracings in the dog. Howard Bartley did his work at Washington
University in St. Louis, a place that had already developed into
Developments in Other European Countries
a hotbed of neurophysiology due to the magnificent work of
Italy was one of the first countries where the EEG found fertile Herbert S. Gasser, Joseph Erlanger, and George Bishop—a
soil. Mario Gozzano, for many years professor of clinical neu- group that made excellent use of Braun’s cathode ray oscillo-
rology in Bologna (later in Rome), published his experiences scope (oscillograph) in the study of peripheral nerve potentials.
with the new method as early as 1935. Gozzano personifies the This outstanding group was joined later by James L. O’Leary, a
(not too common) example of a leading neurologist assuming prominent neurophysiologist, electroencephalographer, and
leadership in clinical electroencephalography (Mazza et al., neurologic clinician. Early experimental EEG work was done by
2002). All too often, eminent clinical neurologists spurned the Davis and Saul (1931), Travis and Dorsey (1932), Travis and
new method. Herren (1931), Bishop and Bartley (1932), Bartley (1932), and
A. Gemelli came from the diametrically opposite area of Gerard et al. (1933) (after Grass, 1984). The work of Ralph W.
neurosciences. This great scholar was a monk, psychologist, Gerard (1900–1974) is linked with the introduction of a con-
philosopher, polyhistor, and president of the Catholic centric needle electrode for the stereotaxic exploration of the
University in Milan. In 1937, he reported his first studies of the brain in experimental animals. Gerard joined forces (in 1934)
human EEG. Gemelli hence represents the psychological wing with Franklin Offner, one of the leading electronic engineers in
of EEG research, which subsequently spawned a number of the development of EEG and related equipment.
outstanding Ph.D. electroencephalographers. (Others would American EEG work in the human started, as it was pointed
come from the ranks of experimental neurophysiologists.) out before, at Harvard in Boston (Hallowell and Pauline Davis,
The Austrian psychologist Hubert Rohracher falls into the Frederic and Erna Gibbs, and William G. Lennox), at Brown in
Gemelli category. He held the chair of psychology for many Providence (Herbert H. Jasper), but also at the University of
years at the University of Vienna, but, in his early academic Iowa in Iowa City where Lee Travis worked, an experimental
work, he fell under the spell of the alpha rhythm and even made psychologist who became the founder of a powerful school
a “pilgrimage” to Hans Berger in Jena (in the 1930s). His early (Herbert Jasper, Donald Lindsley, John R. Knott, and Charles
EEG studies can be dated back to 1938. Henry).
Chapter 1 ■ Historical Aspects of EEG 9

The great international breakthrough in clinical electroen- few examples of paroxysmal EEG discharges in a case of pre-
cephalography came in 1934 with studies of epileptic patients. sumed petit mal attacks and also during a focal motor seizure
Frederic Gibbs had come from Johns Hopkins University in in a patient with general paresis. These observations were just
Baltimore to join the Harvard group. He sought out William G. mentioned in passing and the opportunity of a major break-
Lennox, who had already become a widely known epileptologist. through was missed.
It might be interesting to point out that Lennox had started As to the other great pioneers of electroencephalography in
studies of the cerebral circulation by measuring the O2 and CO2 North America, Hallowell and Pauline Davis produced fine
content of the jugular veins (Lennox, 1930, 1931; Lennox and work on the normal EEG and its variants. They were also
Gibbs, 1932). E. L. Gibbs was Erna L. Gibbs, originally the tech- among the earliest investigators of the human sleep EEG. In the
nical coworker of Lennox but who became the wife of Frederic domain of sleep, A. L. Loomis and his coworkers E. N. Harvey
Gibbs and one of the world’s first EEG technicians and the coau- and G. A. Hobart were the first who methodically studied the
thor of numerous papers. She had come to Boston as an immi- human sleep EEG patterns and the stages of sleep. This research
grant from Germany. The pre-EEG work of Lennox and Gibbs was done off the academic track in Tuxedo Park, New Jersey
on the cerebral blood flow was a milestone in this field. (One of (Loomis et al., 1935, their first study). The Davises eventually
the great present-day masters of cerebral blood flow, Louis turned to audiology and moved to St. Louis. At Brown
Sokoloff from the National Institutes of Health, expressed to me University in Providence, Rhode Island, Jasper studied the EEG
in a personal communication his profound admiration for this of behavior disorders in children before he found his niche in
pioneering work.) EEG simply exerted a greater degree of fasci- basic and clinical epileptology at McGill University in Montreal
nation to W. G. Lennox than did cerebral blood flow. in his epochal collaboration with Wilder Penfield (discussed
Twelve children with petit mal epilepsy were the clinical sub- later). Lee Travis gradually disappeared from the scene but his
jects for the petit mal epilepsy study of Gibbs and Davis (1935) foremost disciples, John R. Knott and Charles E. Henry (Ph.D.
and Gibbs et al. (1935, 1937). This work remains evergreen in electroencephalographers with strong clinical inclinations)
the entire EEG literature; hardly any EEG finding has left such were bound to assume a very important role in America’s EEG
an indelible impression as the association of petit mal absences work. Their ultimate skill and supreme dedication turned them
and 3/sec spike-wave complexes. (Of course, it was found out into the “conscience of EEG,” steering developments into the
later that spike waves could occur without petit mal.) While right direction and correcting the course when there was dan-
Berger was gripped by the rhythms, Frederic Gibbs came under ger of going astray. D. Lindsley became one of the pioneers in
the fascination of paroxysmal patterns such as spike waves. the investigation of maturational EEG aspects; at the University
Shortly afterward, the EEG patterns of grand mal and psy- of California at Los Angeles, he directed excellent neurophysio-
chomotor seizures were reported by the same team (Gibbs, logic EEG research.
Lennox, and Gibbs), but the stretches of fast spikes (in grand This was the first wave of American EEG pioneers and their
mal) and the rhythmical activity in 4 or 6/sec frequency (in psy- immediate disciples and followers. It is impossible for the his-
chomotor seizures) were no match in popularity for the 3/sec torian to do justice to the second wave, which started before the
spike waves of petit mal. decade of the 1930s was over. There was Robert Schwab at
The technical quality of the EEG tracings shown in these Harvard and at the Massachusetts General Hospital in Boston,
studies left much to be desired. Dr. and Mrs. Gibbs traveled to in whom the mastery of EEG was combined with great clinical
Germany in the summer of 1935, paid a visit to Hans Berger, neurologic talents (especially in the field of myasthenia gravis,
spent some time at the Berlin-Buch Institute, and studied the parkinsonism, and epilepsy). Across the Charles River, at the
“polyneurograph” instrument of Jan F. Toennies; they also saw Massachusetts Institute of Technology, there was Warren
the instrumentation of Matthews in England. Frederic Gibbs McCulloch, a fiery genius like Grey Walter in Bristol and a pro-
then contracted Albert Grass (then at the Massachusetts found thinker. His scope would range light years beyond the
Institute of Technology) to build a three-channel preamplifier. limits of EEG and neurophysiology. (One must read his
In 1935, the Grass Model I went into use; it had three channels Embodiments of Mind to fathom his greatness, even though one
and an ink writer that recorded on rolls of paper (the folded may be inclined to disagree in many points.) He and Grey
paper not yet being in use). Walter lived in the world of brain machines, but there was
The Gibbs–Gibbs–Lennox era of the 1930s proved to be per- still a niche for a psyche (when one tries to read between the
haps the most exciting period in the history of EEG. In those lines). Earlier at Northwestern University in Evanston, Illinois,
years, EEG found the domain of greatest effectiveness: the outside Chicago, McCulloch had been involved with Dusser de
realm of the epileptic seizure disorders. Epileptology can Barenne in “neuronographic” work, an import from Utrecht,
be divided historically into two periods: before and after the Netherlands; this work was based on topical strychnine poison-
advent of EEG. Insights into the nature of the epileptic mecha- ing of the cortex and exploration of transmitted spiking to other
nisms deepened, not in a subtle manner but with a huge leap. regions.
What Fischer had started in 1931 with his experimental studies Clinical EEG research already started to conquer certain
on picrotoxin and its effect on the cortical EEG in animals, the fields outside epileptology. Grey Walter’s discovery of the delta
Gibbses and Lennox applied to human epileptology, and a wide focus (Walter, 1936) located over hemispheric brain tumors
door was flung open for the work of future decades. It is true had opened the search for further relationships between brain
that Berger in his seventh report (Berger, 1933) had shown a lesions and focal EEG correlates; metabolic disturbances and
10 Part I ■ Basic Principles

especially hypoglycemia were explored with EEG. (The work of (or Canadian) EEG laboratories started to become more
H. Hoagland and his coworkers dates back to 1937.) sophisticated than that of their European counterparts because
With the end of the 1930s, North America found itself in a of the inclusion of sleep.
leading position in the domain of EEG, whereas progress in Frederic Gibbs enjoyed enormous international prestige at
Europe was quite limited. that time as the world’s leader in clinical electroencephalogra-
phy. Nevertheless, his position at Harvard was much less presti-
World War II and the 1940s gious; he held the academic rank of an instructor (below the
During World War II, from 1939 to 1945, research and clinical professorial ranks), even though a visit to his laboratory was the
EEG activities were not flourishing, particularly in Europe. goal of European colleagues (who could afford the trip). This
There were some neurologic units where the EEG was used in disproportion drastically shows the negative attitude toward
the localization of traumatic brain lesions and epileptogenic EEG in neurologic departments (not universally, of course).
foci. After World War II, the gap between North America and Robert Schwab did not fare much better at Harvard in spite of
Europe was bigger than ever before, and European EEG his fine clinical neurologic talents.
research found itself at a low point. Toward the end of the 1940s, Herbert H. Jasper turned into
After the war, new activities started in England and France, a strong competitor of Frederic Gibbs. Jasper had moved to the
while the situation in Germany looked desperate. Neurologic Institute of McGill University in Montreal, joining
W. Grey Walter with his associates V. J. Dovey and H. Shipton forces with Wilder Penfield, a neurosurgeon with a profound
(a brilliant electronic engineer who later moved to Iowa City neuroscientific background. We discuss the rise of the Montreal
and then St. Louis) at the Burden Institute in Bristol discovered group in section Developments in the 1950s.
the paroxysmal response to flickering light at critical frequen- Two new developments started in the late 1940s. The EEG
cies between 10 and 20/sec. Further work on epileptic photo- technique started to become invasive and, with the use of spe-
sensitivity immediately shifted from Bristol to Marseille, cial depth electrodes, the exploration of deep intracerebral
France, where a young and incredibly talented Henri Gastaut regions began. This is discussed in section Developments in the
used this method, in combination with intravenous dosages of 1950s. Automatic frequency analysis also started in the 1940s,
pentylenetetrazol, to determine the “seuil épileptique,” that is, but this development reached loftier heights in the 1960s.
the individual threshold for paroxysmal responses (Gastaut, A discussion of the 1940s would be incomplete without a
1949; Gastaut et al., 1948). brief glance at the work of the neurophysiologists. A large seg-
In 1947, the American EEG Society was founded and the ment of neurophysiologic work was dominated by the use of
First International EEG Congress was held in London; a second EEG. One of the most fascinating results of these researchers
one followed in 1949 in Paris (in association with clinical neu- was the demonstration of thalamocortical relationships, thus
rology and other neurologic disciplines). EEG activities in far explored solely with anatomical methods (e.g., the study of
Germany were still minimal; Japan, however, gained attention the thalamus by A. Earl Walker in 1938, which propelled this
by the work of K. Motokawa, a researcher of EEG rhythms. young neuroscientist to great fame for decades to come). The
Switzerland started to develop its own profile; the neurophysi- work of Morison and Dempsey (1942) on the recruiting
ologist Marcel Monnier was instrumental in this regard. W. R. response had great impact on the neuroscientific world with the
Hess, however, a Nobelist, had gained great prestige by the func- demonstration of cortical responses to relatively slow stimula-
tional mapping of thalamus and hypothalamus with regard to tion of the intralaminar structures of the thalamus in the cat.
autonomic responses to electrical stimulation. This work emphasized the role of the thalamus in the cortical
The American scene was bustling with activities. Frederic A. electrogenesis and broke the ground for the concept of a “cen-
Gibbs with his coworkers Erna Gibbs and B. Fuster from trencephalic epilepsy,” a concept promoted by Penfield and
Uruguay produced another epochal study on the interictal Jasper in Montreal (somewhat naively understood as a concept
anterior temporal spike or sharp wave discharge in the inter- of the thalamic origin of primary generalized epilepsy).
seizure interval in patients with psychomotor seizures. This was Even greater was the impact of the work of Horace W.
an important step in the elucidation of temporal lobe epilepsy, Magoun, who had studied the effects of descending and mostly
a work with far-reaching consequences for the entire develop- inhibitory influences of the brainstem reticular formation dur-
ment of EEG laboratories and their routine work. It was found ing his work at Northwestern University. Together with G.
(Gibbs et al., 1948) that the anterior temporal discharges were Moruzzi (a fine neurophysiologist from Pisa, Italy, and investi-
often limited to the state of sleep. This observation meant that gator of basic epileptic mechanisms), Magoun subsequently
a tracing without a sleep portion could be insufficient, uninfor- studied the ascending system of the brainstem reticular forma-
mative, and even misleading. Thus, EEG laboratories would tion (chiefly in the midbrain level) and the effect of high-
include sleep in most (if not all) of their EEG evaluations. This frequency electrical stimulation, consisting of EEG
required pasted electrodes (rather than rubber bands or caps), desynchronization and behavioral arousal, on cortical function.
a much longer recording time, and a much smaller numerical Magoun, who had moved to the University of California at Los
output of recordings per technician (incidentally, an evolving Angeles, subsequently investigated the effects of acute lesions
profession, which is discussed later). made in the midbrain-level reticular formation in cats. These
Transatlantic communication was poor at that time, and cats remained in a comatose state with EEG slowing in spite
it was at this point when the routine work in American of electrical brainstem stimulation because of the destruction
Chapter 1 ■ Historical Aspects of EEG 11

of the all-important ascending portion of the brainstem reticu- effects. At that time, local anesthesia was still widely used in
lar formation (Lindsley et al., 1949). It is no exaggeration when neurosurgery. Jasper was chiefly a neuroscientist and not
one describes the effect of these studies on the world of neuro- merely an electroencephalographer. The book entitled Epilepsy
science as a “bombshell.” For the ensuing 10 to 15 years, the and the Functional Anatomy of the Human Brain (Penfield and
association of consciousness with reticular formation and Jasper, 1954) was a result of this fruitful collaboration.
Magoun’s name was so strong that it even had considerable Very controversial, however, was a concept of the primary
influence on the Pavlovian dogmatism of the Eastern Bloc generalized form of epilepsy characterized by generalized syn-
countries. Nowadays, however, even talented young neuroscien- chronous paroxysmal EEG discharges and exemplified by the
tists react to Magoun’s name with a blank expression—sic transit 3/sec spike waves of petit mal absences. Penfield and Jasper
gloria mundi! listed these epilepsies as “centrencephalic” with the concept of
The reason for discussing this experimental work in a histor- “center of the encephalon” (i.e., “thalamic midline structures”)
ical overview is to demonstrate the incredibly powerful role of serving as the starting point of the bilateral discharges. Henri
EEG in the neurophysiology of the 1940s. This was a high Gastaut from Marseille would follow the lead and so did many
watermark. Subsequently, experimental EEG work started to others, but Frederic Gibbs and a host of other electroen-
concentrate on single neurons while the “macro-EEG” gradu- cephalographers and neuroscientists became detractors of the
ally declined. centrencephalic concept. It wasn’t until the late 1960s that it
became clear that the centrencephalic concept stood on a very
Developments in the 1950s shaky ground and was ripe for being dismantled. Montreal’s
This is the last decade presented for historical analysis. Our story own Pierre Gloor helped to do this in a cautious and diplomatic
is gradually approaching the present, and a historical outline manner; others buried the centrencephalic concept more
must shy away from events that occurred over the last 40+ years. bluntly.
It does not behoove the historian to place living and active col- Frederic Gibbs had moved to the University of Illinois
leagues into the focus of discussion (with few exceptions). School of Medicine in Chicago (where full professorship was
The 1950s was the decade when EEG became a household given to him instantly after Harvard had denied him any pro-
word. During the early stretch of the decade, almost every uni- motion for more than a decade). Chicago—especially the
versity (teaching) hospital had at least one EEG machine. At the University of Chicago but also Northwestern University and the
end of the decade, EEG apparatuses had found their way into a University of Illinois—had become a world leader in neurologic
large number of other hospitals and even into private practice. sciences over the past 20 years. Percival Bailey, Paul Bucy, Roy
At university hospitals, central as well as departmental EEG lab- Grinker, A. Earl Walker, Gerhardt Von Bonin, C. J. Herrick,
oratories emerged. The latter were usually limited to children or Frederic Gibbs, and many others give testimony to the glory of
adults, and pediatric EEG units evolved (while specialized neona- neurologic science in Chicago at the middle of that century. In
tologic EEG units followed suit about 10 years later). Some psy- the field of EEG, the Chicago group under the Gibbses and the
chiatric departments took particular pride in their clinical and Montreal group under Jasper and Gloor were strong rivals
research-oriented EEG work. It is absolutely true that psychiatry throughout the 1950s, especially with respect to leadership in
was always “nice” to the electroencephalographer. Psychiatry’s epileptologic electroencephalography. One of the greatest mas-
domain was in need of organic or neurophysiologic substrata of ters from the Chicago school, A. Earl Walker, came to Johns
disorders and dysfunctions of psychiatric–psychological nature. Hopkins in Baltimore in 1947, introducing depth EEG, electro-
What could the electroencephalographer give in return? It was corticography, epilepsy surgery, and a scientifically oriented
very little, but the psychiatrists did not seem to mind. On the epileptology to his new place. Walker, who in 1972 had moved
other hand, there was so much to give to neurology. At that to the University of New Mexico and died in 1995, will always
time, it had become clear that the majority of diseases affecting be remembered as one of the great scholars of neurosurgery
the central nervous system (CNS) had more or less impressive and epileptology.
EEG correlates, but the majority of neurologists remained The 1950s saw a strong comeback of the Europeans. Henri
either reserved or hostile to EEG. Neurosurgeons were inter- Gastaut’s intellectual brilliancy was hard to match. In Marseille,
ested as long as EEG could contribute to the determination of disciples of great stature flocked around him, especially Robert
focal cerebral lesions (and before EEG became overpowered by Naquet, Joseph Roger, and Annette Beaumanoir, to mention
noninvasive neuroimaging techniques). Some epilepsy-ori- only the earliest nucleus of this group. At the great world cen-
ented neurosurgeons like W. Penfield or A. Earl Walker ters of neurology, Salpêtrière Hospital in Paris and National
remained interested in EEG and its use in the depth of the cere- Hospital, Queen Square, London, Antoine Remond and
brum or on the cortex. William Cobb, respectively, represented the EEG, but unfortu-
The epileptologic EEG work of Herbert Jasper in collabora- nately too much in the shadow of the leading neurologists.
tion with Wilder Penfield reached new heights, and Montreal Remond later turned into a protagonist of computerization of
reigned supreme as the place for neurosurgical treatment of EEG data.
focal epilepsies. Penfield was far more than a neurosurgeon. His The star neurologists of both Queen Square and Salpêtrière
operations for the removal of epileptogenic foci and, in a later lived in the world of classical neurology, which gave them so
phase, large portions of affected lobes were associated with elec- much satisfaction and happiness that one could hardly expect
trical stimulation and a systematic study of the behavioral their openness for the world of brain potentials. The Queen
12 Part I ■ Basic Principles

Square guard appeared to be more detached from EEG than Signals in a Large Irregular Background,” George D. Dawson
their Parisian confrères, perhaps due to the fact that Gastaut, from the National Hospital, Queen Square, in London demon-
the man from Marseille, came from the neurologic ranks to strated evoked potentials to electrical stimulation of the ulnar
achieve instant stardom with his EEG achievements. Probably nerve (Dawson, 1951). This required advanced analog technol-
no other famous neurologist has expressed his opinion about ogy. Thus, Dawson became the father of evoked potential stud-
EEG more scathingly than Francis M. R. Walshe has done. He is ies, which developed into a major outcropping of
the brilliant Queen Square star who apparently knew every- electroencephalography, eventually constituting a field of its
thing about neurology except EEG. To Sir Francis, the electrical own. The ingenious superimposition method of Dawson was
activity of the brain was, “a bloodless dance of action potentials eventually superseded by the advent of computerized averaging
… hurrying to and fro of its molecules” (after Critchley, 1990). methods in the 1960s.
Let us assume, for everyone’s benefit, that Sir Francis had meant Computational techniques of wave analysis started early in
it to be a joke. the history of EEG. First attempts were made by Hans Berger
Fine schools of EEG developed in the Netherlands with O. (1932); he was assisted by the physicist Dietsch (1932), who
Magnus (son of a Nobel Prize winning physiologist) in applied Fourier analysis to short EEG sections. Further work in
Wassenaar and Storm van Leeuwen in Utrecht. In Switzerland, this field was produced by Grass and Gibbs (1938) and Knott
Rudolf Hess (also son of a Nobelist physiologist) created an and Gibbs (1939). At the Massachusetts Institute of Technology
important school of electroencephalographers at the Zurich near Boston, Guillemin applied Fourier analysis to communica-
University Hospital. Giuseppe Pampiglione, from Italy, initiated tion theory, and one of his students was Albert Grass who
pediatric electroencephalography at the Hospital for Sick “could not wait to get the Gibbs interested” (Grass, 1984). The
Children in London, concurrently with William Lennox’s work 1950s saw the early generation of automatic frequency analyz-
at the Boston Children’s Hospital. ers approaching and eventually saw the end of these magnifi-
These European centers of EEG activities had rather an cent but mostly unused machines.
international flavor with strong North American influence. Eventually, the EEG branched out into the world of single
This cannot be said about the evolving field of clinical EEG in neurons, and the microelectrode technique was introduced in
West Germany, which was dominated by its prestigious leader the early 1950s. Microelectrodes can be made of metal such as
Richard Jung in Freiburg. Jung’s greatness pertained to experi- tungsten with tips of 1- to 3- m diameter; glass electrodes
mental neurophysiology; he also had great interest in the clini- filled with electrolytes such as KCI have tips of 0.5 m or even
cal fields and even in philosophy. This renaissance man smaller. Because of their characteristics, microelectrodes reach
designed the outline for EEG training and the routine of the very high impedance values (1–60 M ), which render conven-
EEG laboratory—unfortunately not without shortcomings, tional EEG recording techniques unsuitable. The introduction
which hamstrung the further development of clinical EEG in of the cathode follower by Toennies created the technical pre-
West Germany. requisite for single-cell recordings.
The 1950s also saw EEG sprouting into related fields. Extracellular microelectrode recording was used on a larger
Depth electroencephalography with implanted intracerebral scale in the early 1950s (Jung et al., 1952; Li et al., 1952;
electrodes was used in the humans for the first time by Moruzzi, 1952). About 10 years later, extracellular microelec-
Meyers and Hayne (1948) and Knott et al. (1950) at the trode studies were even done intraoperatively in humans. Far
University of Iowa, Iowa City, and also by Hayne et al. more revolutionary was the introduction of the extremely labo-
(1949a) in Chicago. These short recordings served the study rious intracellular microelectrode technology (Brock et al.,
of EEG activity in the human basal ganglia and thalamus 1952, in the spinal cord; Phillips, 1961, in the cortex). This tech-
with regard to basal ganglia dyskinesias and epilepsy. In the nique opened the gates to a new world of biochemical
following years, deep structures were also explored in processes. These insights taught lessons in humility to the elec-
patients with psychiatric disorders until doubt was cast upon trophysiologic neurophysiologist. There was no doubt that the
the ethical basis of this invasive approach (see Chapter 33, chemical changes were of primary significance, while the elec-
“Depth Electroencephalography”). In the 1960s, depth EEG trical phenomena were more or less by-products.
would find its true field in epileptic patients considered can- We cannot leave the 1950s without mentioning epochal
didates for epilepsy surgery. developments in the field of sleep research. At the University of
The origin of intraoperative electrocorticography dates back Chicago, N. Kleitman stood out as one of the world’s leading
to Foerster and Altenburger (1935). How was it possible that investigators of the organization of sleep. This institution pro-
Otfried Foerster, perhaps the greatest clinical neurologist ever duced the first study of rapid eye movement (REM) sleep
and an amazing self-taught master of neurosurgery, failed to (Aserinsky and Kleitman, 1953), but it must be pointed out that
recognize the future potential of EEG? Did his mind work Blake and Gerard (1937) described a “null stage” in the EEG of
mainly in the world of Sherringtonian concepts? Most of the nocturnal sleep, thus indicating the desynchronization of EEG
work in electrocorticography remains associated with the in REM sleep but without observation of the accompanying
Montreal Neurologic Institute and the names of Penfield and ocular, muscular, and other autonomic changes. William C.
Jasper (also see Chapter 34, “Electrocorticography”). Dement continued the work of Kleitman and, following his
The related fields of EEG started to bloom in the 1950s. In a move from Chicago to Stanford, became a world leader in the
study entitled “A Summation Technique for Detecting Small study of nocturnal sleep.
Chapter 1 ■ Historical Aspects of EEG 13

Sleep research gradually became based on polygraphic somatosensory evoked potentials is associated with the names
recording, and its share in the overall EEG research declined. of Roger and Joan Cracco.
This development led to a constantly widening gap between The 1960s and 1970s witnessed a regrettable alienation of
EEG and nocturnal sleep research (in the 1960s and the follow- EEG and epileptology, which had existed before in an almost
ing decades). perfect marriage. A sizable number of epileptologists lost inter-
est in EEG. Was it early enthusiasm about the introduction of
The Rest of the Story antiepileptic serum levels? Was the path to mastery of EEG
The last 30 years of the history of EEG and related fields can be becoming too laborious? This situation changed in the 1980s
gleaned directly from this book. The events of the 1960s, 1970s, due to the rapidly increasing emphasis on EEG and related
and 1980s are just too close for us to see with the eyes of the his- techniques in the presurgical workup of patients considered
torian. Nevertheless, modern trends are briefly discussed in this candidates for seizure surgery.
final section. The 1970s and 1980s saw brilliant structural neuroimaging
The development of clinical and experimental EEG work techniques emerging: computed tomography and magnetic res-
reached a high point around 1960 after 30 years of steady onance imaging. This seemed to knock out electroencephalog-
progress. There is no doubt that the 1960s slowed down the raphy from the contributors to focal CNS diagnosis. Such a
smooth progress. The interest of electroencephalographers in knockout blow, however, was also more apparent than real. The
academic institutions tended to shift from the tracing, with all EEG, by its very nature, never was a structure-oriented test.
its waves and patterns, to automatic data analysis. Whether the patient has a hemispheric brain tumor, a vascular
Computerization was the direction—tendencies reaching back lesion, or a traumatic contusion, EEG can always demonstrate
to Berger’s coworker Dietsch (1932), but flourishing in the the degree of dysfunctional changes around the lesion (or sec-
1960s and 1970s. This development had many positive aspects. ondary diffuse cerebral dysfunction). The sad story is that the
The names of Barlow, Brazier, Remond, Lopes da Silva, neurologist of our day seems to lose interest in the realm of
Bickford, Saltzberg, Dumermuth, Matousek, D. O. Walter, function and dysfunction. This development must be halted
Cooper, Künkel, Lehmann, Gasser, Burch, Hjorth, Schenk, (and eventually will be).
Matejcek, and Low should be mentioned in this context. In par- Topical EEG diagnosis, however, has made a comeback of its
ticular, Cooley and Tukey (1965) have been credited with the own in the form of computerized brain mapping. This fascinat-
introduction of the fast Fourier transforms as the basis of ing recent development is associated chiefly with the name of
power spectral analysis. Frank Duffy. Again, it is the old EEG in new clothes: it can be
This work led us into a “brave new world” of EEG comput- understood only by an expert of the conventional EEG.
erization and, as early as in 1967, we were told that custom- Starting in the late 1960s, a completely new development
ary EEG reading would soon be a thing of the past, replaced took place in the EEG exploration of the full-term and the pre-
by a fully automatic EEG interpretation. The fears of many mature newborn. The historical aspects of this development are
clinical electroencephalographers were unfounded; nobody found in Chapters 11 and 49.
became jobless, because such an automatization of EEG read- This historical overview is not complete without a few words
ing was fictional. It was found that EEG is far too complex for about the technicians (technologists) doing the EEG laboratory
such automation. Its interpretation requires the wonderful routine work. They had to place electrodes with greatest accu-
computer that is located between the ears—the educated and racy and to obtain a readable tracing, even under the most
experienced brain. One simply must consider that the meth- adverse conditions. They were considered the electroencephalo-
ods of those years—frequency or time domain—were limited grapher’s attendants for a long time, even though their work
to an analysis of frequencies. Automatic spike detection had required considerable sophistication.
barely reached its earliest stage. John R. Knott and Charles E. Henry invested incredible energy
The electroencephalographer needed to be aware that all into the founding of the American Society of EEG (later
types of data computerization were nothing but the EEG in Electroneurodiagnostic) Technologists, which came into being in
disguise—“an analog of an analog.” Computerized frequency 1962. Soon afterward, the first group of technicians underwent a
analysis was here to stay and to prove to be of enormous value stiff examination that made them registered EEG technologists.
not only in psychophysiologic research but also in the assess- There have been similar developments in many other countries.
ment of neuropharmacologic effects. This evolution has been helpful in giving EEG technologists the
In the 1970s, the evoked potential technique progressed dignity they deserve, but this process is far from being completed.
greatly. The introduction of the pattern changer in the visual With every record we read, we must be thankful for the work of
evoked potential technique made this method highly reliable; our technical staff and invest some of our energies in their con-
the names of H. Speckrejse and R. Spehlmann ought to be men- tinuous education and training, for the sake of better technical
tioned in this context. In the field of auditory evoked potentials, EEG quality and thus for the sake of our patients.
the location of primary cortical discharge was elusive for many This historical overview remains a fragmented account
years, and the late vertex potential of limited clinical value. The because much remains untold in this story. However, it is hoped
introduction of the far-field technique for the demonstration of that the historical perspective this chapter brings to the reader
the brainstem auditory evoked potentials (Jewell), however, will foster in our ranks insights that may help us avoid the mis-
proved to be extremely valuable. Analogous work in the field of takes of the past.
14 Part I ■ Basic Principles

EPILOGUE: THOUGHTS ABOUT REFERENCES


PRESENT AND FUTURE Major Historical Works
Clinical Electroencephalogram Brazier MAB. A History of the Electrical Activity of the Brain. The First
Half-Century. London: Pitman; 1961.
The role of clinical electroencephalography has been diminish- Critchley M. The Ventricle of Memory. New York: Raven Press; 1990.
ing throughout the past 30 years except for epileptology, which Gloor P. Hans Berger on the Electroencephalogram of Man. Amsterdam:
has been using the tool of EEG video monitoring to its full Elsevier; 1969.
extent. This produces an enormous accumulation of data and Grass AM. The electroencephalographic heritage. Am J EEG Technol.
needs thorough analysis and interpretation—a highly time- 1984;24:133–173.
consuming task that is often “farmed out” or subcontracted to Hassler R. Cécile und Oskar Vogt. In: Kolle K, ed. Grosse NervenŠrzte.
outside readers of sometimes unacceptable EEG expertize. Vol. 2. Stuttgart: Thieme; 1959:45–64.
Presence, number or absence of spikes, and spike-related dis- Jung R. Hans Berger und die Entdeckung des EEG nach seinen
Tagebüchern und Protokollen. In: Werner R, ed. Jenenser EEG-
charges are not the only criteria; full justice must be done to all
Symposium: 30 Jahre Elektroenzephalographie. Berlin: VEB Verlag
EEG abnormalities of nonparoxysmal character, and this task is Volk und Gesundheit; 1963:20–53.
a lot more difficult. Such expensive long-term recordings should Kolle K. Hans Berger. In: Kolle K, ed. Grosse NervenŠrzte. Vol. 1.
be either artfully interpreted or not done at all. Stuttgart: Thieme; 1956:1–6.
Acutely requested EEG on an emergency basis nowadays Mazza S, Pavone A, Niedermeyer E. Mario Gozzano: the work of an
deals mainly with the question of “rule out status epilepticus” EEG pioneer. Clin Electroencephalogr. 2002;33:155–159.
(Varelas et al., 2003). With presently widespread confusions con- Niedermeyer E. Benjamin Franklin and static electricity.
cerning the limitations of nonconvulsive status epilepticus (see Considerations of past, present, and future. Am J End Technol.
Chapters 28 and 29), the answer needs profound neurologic– 2003a;43:26–29.
epileptologic understanding. Cases of acute cerebral anoxia O’Leary JL, Goldring S. Science and Epilepsy. New York: Raven Press;
(postcardiopulmonary arrest) may cause both convulsive as 1976.
Schwab RS. Electroencephalography. Philadelphia: WB Saunders; 1951.
well as nonconvulsive pictures with massive paroxysmal EEG
Upton M. Electronics for Everyone. New York: New American Library of
abnormalities but do not represent true status epilepticus World Literature (Signet Key Books); 1960.
(see Chapter 23). Werner R. Hans Berger zum Gedächtnis. In: Werner R, ed. Jenenser
Outside the confines of epileptic conditions, the EEG is full EEG-Symposium: 30 Jahre Elektroenzephalographie. Berlin: VEB
of information for the vast majority of neurologic diseases. It is Verlag Volk und Gesundheit; 1963:13–19.
most regrettable that this rich source of information has been Work of Specially Mentioned EEG Pioneers
badly underused in most neurologic teaching institutions and Adrian ED. The Berger rhythm in the monkey’s brain. J Physiol.
hospitals in general (Niedermeyer, 2003b). 1936;87:83P–84P.
Adrian ED, Bronk DW. The frequency of discharge in reflex and volun-
The Basis of Electroencephalogram tary contractions. J Physiol. 1929;67:119–151.
The cellular basis of EEG activity has been the topic of intensive Adrian ED, Matthews BHC. The interpretation of potential waves in
studies of extracellular current flow and voltage-dependent the cortex. J. Physiol. 1934;81:440–471.
Aserinsky W, Kleitman N. Regularly occurring episodes of eye motility
intrinsic oscillations (see Buzsaki et al., 2003). Ongoing work
and concomitant phenomena during sleep. Science. 1953;118:
on cortical and subcortical generators is found in Chapter 3
273–274.
(also see Ebersole, 2003). This shows that the genesis of EEG is Bartley SH. Analysis of cortical response to stimulation of the optic
still a widely discussed issue. nerve. Am J Physiol. 1932;101:4P.
Bartley SH, Newman EB. Recording cerebral action currents. Science.
Electroencephalogram and Neurocognition
1930;71:587.
This area has become the perhaps most fascinating aspect of Bartley SH, Newman EB. Studies on the dog’s cortex. Am J Physiol.
modern EEG interest (see Chapter 51). It is true that most of 1931;99:1–8.
this type of research has been done with other tools such as Berger H. Über das Elektrenkephalogramm des Menschen. 1st report.
functional magnetic resonance imaging, positron emission Arch Psychiat Nervenkr. 1929;87:527–570.
tomography (PET) scanning, and single-photon emission com- Berger H. Über das Elektrenkephalogramm des Menschen. 4th report.
puted tomography (SPECT) (methods demonstrating regional Arch Psychiat Nervenkr. 1932;97:6–26.
blood flow and metabolic needs). With the extension of EEG Berger H. Über das Elektrenkephalogramm des Menschen. 7th report.
Arch Psychiat Nervenkr. 1933;100:301–320.
into the ultrafast frequency ranges (Chapter 37), a powerful
Bishop GH, Bartley SH. Electrical study of the cerebral cortex as com-
upswing of EEG-oriented neurocognitive research in animals pared to the action potential of excised nerve. Proc Soc Exp Biol
and humans can be expected. Neurocognition has assumed the (New York). 1932;29:698–699.
position of the “Holy Grail” of all neuroscience, and EEG stands Blake K, Gerard RW. Brain potentials during sleep. Am J Physiol.
a great chance to become a principal contributor. After years of 1937;119:692–703.
frantic efforts, it will be found that neuroscience can illuminate Bremer F. Cerveau isolé et physiologie du sommeil. C R Soc Biol
the “brain-mind barrier” only to a certain degree. For the time (Paris). 1935;118:1235–1241.
being, however, let us not disturb the glowing enthusiasm with Bremer F. Cerebral and cerebellar potentials. Physiol Rev. 1958;38:
Cassandra calls. 357–388.
Chapter 1 ■ Historical Aspects of EEG 15

Brock LG, Coombs JS, Eccles JC. The recordings of potentials from Hayne R, Meyers R, Knott JR. Characteristics of electrical activity of
motor neurons with an intracellular electrode. J Physiol. 1952;117: human corpus striatum and neighboring structures. J Neurophysiol.
431–460. 1949b;12:185–195.
Buzsaki G, Traub R, Pedley TA. The cellular basis of EEG activity. In: Hoagland H, Rubin MA, Cameron DF. The electroencephalogram of
Ebersole JS, Pedley TA, eds. Current Practice of Clinical schizophrenics during insulin hypoglycemia and recovery. Am J
Electroencephalography. 3rd ed. Philadelphia: Lippincott Williams Physiol. 1937;120:559–570.
and Wilkins; 2003:1–11. Jung R, Baumgarten RV, Baumgartner G. Mikroableitungen von
Caton R. The electric currents of the brain. Br Med J. 1875;2:278. einzelnen Nervzellen im optischen Cortex der Katze. Die lich tak-
Cooley JW, Tukey JW. An algorithm for the machine calculation of tiviertcn B-Neurone. Arch Psychiat Z Ges Neurol. 1952;189:
complex Fourier series. Math Comp. 1965;19:267–301. 521–539.
Danilevsky VD. Investigations into the Physiology of the Brain. Doctoral Knott JR, Gibbs FA. A Fourier transform of the electroencephalogram
thesis. University Charkov (cited in Brazier, 1961), 1877. from one to eighteen years. Psychol Bull. 1939;36:512–513.
Davis H, Saul LV. Action currents in the auditory tracts of the midbrain Knott JR, Gibbs FA, Henry CE. Fourier transforms of electroen-
of the cat. Science. 1931;86:448–450. cephalogram during sleep. J Exp Psychol. 1942;31:465–477.
Dawson GD. A summation technique for the detection of small signals in Knott JR, Hayne RA, Meyers HR. Physiology of sleepwave characteris-
a large irregular background. J Physiol (London). 1951;115:2P. tics and temporal relations of human electroencephalograms
Dietsch G. Fourier-Analyse von Elektrenkephalogrammen des recorded from the thalamus, the corpus striatum and the surface of
Menschen. Pflugers Arch Ges Physiol. 1932;230:106–112. the scalp. Arch Neurol Psychiatry (Chicago). 1950;63:526–527.
Ebersole JS. Cortical generators and EEG voltage fields. In: Ebersole JS, Kornmüller AE. Architektonische Lokalisation bioelektriseher
Pedley TA, eds. Current Practice of Clinical Electroencephalography. Erscheinungen auf der Grosshirnrinde. 1. Mitteilung:
3rd ed. Philadelphia: Lippincott Williams and Wilkins; 2003:12–31. Untersuchungen am Kaninchen bei Augenbelichtung. J Psychol
Fischer MH. Elektrobiologische Auswirkungen von Krampfgiften am Neurol. 1932;44:447–459.
Zentralnervensystem. Med Klin. 1933;29:15–19. Kornmüller AE. Die Ableitung bioelektischer Effekte architektonischer
Fischer MH, Löwenbach H. Aktionsströme des Zentralnervensystems Rindenfelder vom uneröffneten Schadel. J Psychol Neurol.
unter der Einwirkung von Krampfgiften. 1. Mitteilung Strychnin und 1933;45:172–184.
Pikrotoxin. Arch Exp Pathol Pharmakol. 1934a;174:357–382. Kornmüller AE. Der Mechanismus des epileptischen Anfalles auf
Fischer MH, Löwenbach H. Aktionsströme des Zentralnervensystems Grund bioelektrischer Untersuchungen am Zentralnervensystem.
unter der Einwirkung von Krampfgiften. 2. Mitteilung: Cardiazol, Fortschr Neurol Psychiatry. 1935;7:391–400, 414–432.
Coffein und andere. Arch Exp Pathol Pharmakol. 1934b;174: Kornmüller AE. Die Bioelektrischer Erseheinungen der Hirnrindenfelder.
502–516. Leipzig: Thieme; 1937.
Foerster O, Altenburger H. Elektrobiologische Vorgänge an der Kornmüller AE. Klinische Elektrenkephalographie. Munich: Lehmann;
mensehlichen Hirnrinde. Dtsch Z Nervenheilk. 1935;135:277–288. 1944.
Gastaut H. Effets des stimulations physiques sur l’E.E.G. de l’homme. Kornmüller AE. Die Elemente der Nervšsen TŠtigkeit. Stuttgart:
Electroencephalogr Clin Neurophysiol Suppl. 1949;2:69–82. Thieme; 1947.
Gastaut H, Roger J, Corriol JH, et al. Les formes expérimentales Lennox WG. The oxygen and carbon dioxide content of blood from
d l’épilepsie humaine. L’épilepsie induite par la stimulation the internal jugular and other veins. Arch Intern Med.
lumineuse intermittente ou épilepsie photogénique. Rev Neurol 1930;46:630–636.
(Paris). 1948;80:161–183. Lennox WG. The cerebral circulation. Arch Neurol Psychiatry
Geddes LA. What did Caton see? Electroencephalogr Clin Neurophysiol. (Chicago). 1931;26:719–724.
1987;67:2–6. Lennox WG, Gibbs EL. The blood flow in the brain and the leg of man,
Gerard RW, Marshall WH, Saul LJ. Cerebral action potentials. Proc Soc and the changes induced by alteration of blood gases. J Clin Invest.
Exp Biol (New York). 1933;30:1123–1125. 1932;1:1155–1177.
Gibbs EL, Gibbs FA. Diagnostic and localizing value of electroen- Li CL, Jasper HH, McLennan H. Décharge d’unités cellulaires en rela-
cephalographic studies in sleep. Publ Assoc Res Nerv Ment Dis. tion avec les oscillations électriques de l’écorce cérébrale. Rev
1947;26:366–376. Neurol (Paris). 1952;87:149–151.
Gibbs EL, Fuster B, Gibbs FA. Peculiar low temporal localization of Liberson WT. Recherches sur les électroencéphalogramme tran-
sleep-induced seizure discharges of psychomotor epilepsy. Arch scraniens de l’homme. Travail Hum. 1937;5:431–463.
Neurol Psychiatry (Chicago). 1948;60:95–97. Lindsley DB, Bowden JW, Magoun HW. The effect of subcortical
Gibbs FA, Davis H. Changes in the human electroencephalogram asso- lesions upon the electroencephalogram. Am Psychol. 1949;4:
ciated with loss of consciousness. Am J Physiol. 1935;113:49–50. 233–234.
Gibbs FA, Davis H, Lennox WG. The electroencephalogram in epilepsy Loomis AL, Harvey EN, Hobart GA. Potential rhythms of the cerebral
and in conditions of impaired consciousness. Arch Neurol cortex during sleep. Science. 1935;82:198–200.
Psychiatry (Chicago). 1935;34:1133–1148. Meyers HR, Hayne R. Electrical potentials of the corpus striatum and
Gibbs FA, Gibbs EL, Lennox WG. Epilepsy paroxysmal cerebral dys- cortex in Parkinsonism and hemiballism. Trans Am Neurol Assoc.
rhythmia. Brain. 1937;60:377–388. 1948;73:10–14.
Goltz FL. Ueber die Verrichtungen des Groszhirns. Pflueger’s Arch Ges Morison RS, Dempsey EW. A study of thalamo-cortical relations. Am J
Physiol. 1888;42:419–467. Physiol. 1942;135:281–292.
Grass AM, Gibbs FA. A Fourier transform of the electroencephalo- Moruzzi G. L’attività dei neuroni corticali durante il sonne e durante la
gram. J Neurophysiol. 1938;1:521–526. reazione elettroencefalografica di risveglio. Ricerca Sci. 1952;22:
Hayne R, Belinson L, Gibbs FA. Electrical activity of subcortical 1165–1173.
areas in epilepsy. Electroencephalogr Clin Neurophysiol. Moruzzi G, Magoun HW. Brain stem reticular format and activation of
1949a;1:437–445. the EEG. Electroencephalogr Clin Neurophysiol. 1949;1:455–473.
16 Part I ■ Basic Principles

Niedermeyer E. The clinical relevance of EEG interpretation. Clin Varelas PN, Spanaki MV, Hacein-Bey L, et al. Emergent EEG.
Electroencephalogr. 2003b;34:93–98. Neurology. 2003;61:702–704.
Penfield W, Jasper HH. Epilepsy and the Functional Anatomy of the Vogt C, Vogt O. Sitz und Wesen der Krankheiten im Lichte der topistis-
Human Brain. Boston: Little, Brown; 1954. chen Hirnforschung und des Variierens der Tiere. Leipzig: Barth;
Phillips CG. Some properties of pyramidal neurones of the motor cor- 1937.
tex. In: Wolstenholme GEW, O’Conner M, eds. The Nature of Sleep. Walter WG. The location of brain tumors by electroencephalogram.
Boston: Little, Brown; 1961:4–24. Proc R Soc Med. 1936;30:579–598.
Rothmann H. Zusammenfassender Bericht über den Rothmann’schen Walter WG. Slow potential waves in the human brain associated with
groszhirnlosen Hund nach klinischer und anatomischer expectancy, attention and decision. Arch Psychiat Nervenkr.
Untersuchung. Z Ges Neurol Psychiat. 1923;87:247–313. 1964;206:309–322.
Sherrington CS. Integrative Action of the Nervous System. New Haven, Walter WG, Dovey VJ, Shipton H. Analysis of electrical responses
CT: Yale University Press; 1906. of the human cortex to photic stimulation. Nature. 1946;
Travis LE, Dorsey JM. Action current studies of simultaneously active 158:540–541.
disparate fields of the central nervous system of the rat. Arch Neurol Walter WG, Cooper R, Aldridge VJ, et al. Contingent negative varia-
Psychiat. 1932;28:331–338. tion. An electric sign of sensorimotor association and expectancy in
Travis LE, Herren RY. The relation of electrical changes in the brain to the human brain. Nature. 1964;203:380–384.
reflex activity. J Comp Psychol. 1931;12:23–29. Yakovlev PI. Vladimir Mikhailovich Bekhterev (1857–1927). In:
Travis LE, Knott JR. Brain potential studies. I. Perseveration time to Haymaker W, ed. The Founders of Neurology. Springfield, IL:
light. J Psychol. 1936;3:97–100. Charles C Thomas; 1953:244–247.
CHAPTER
Neurophysiologic Basis of
EEG and DC Potentials
ERWIN-JOSEF SPECKMANN, CHRISTIAN E. ELGER, AND ALI GORJI
2
T
he clinical electroencephalographer correlates central numbers. Thus, nerve cells are usually covered with several
nervous system (CNS) functions as well as dysfunctions thousand synapses (2). The glia cells are imbedded between
and diseases with certain patterns of the electroen- nerve cell somata, dendrites, and axons. They usually have sev-
cephalograhy (EEG) on an empirical basis. Obviously, this eral processes that make contact with somata and processes of
method has been found valuable in clinical practice. Therefore, nerve cells; they may also make contact with vessels. This histo-
why should the clinical electroencephalographer study the basic logic arrangement results in a cerebral extracellular space con-
elementary processes underlying the EEG? There is little doubt sisting of very narrow intercellular clefts (3).
that the range of EEG interpretations can be much widened and
misinterpretations avoided when the underlying elementary NEURONAL MEMBRANE POTENTIALS:
processes are also considered. This is true especially for convul-
INTRACELLULAR RECORDINGS
sive disorders and cerebral metabolic disturbances. For exam-
ple, an isoelectric EEG can be caused by selective pCO2 increase Essential potentials that can be demonstrated with intracellular
while the brain is sufficiently supplied with O2. On the other recordings are characterized briefly. When the membrane of the
hand, in the presence of practically normal pCO2 levels, cere- nerve cell body is penetrated by a microelectrode, a potential of
bral hypoxia may be the cause. It will be pointed out below that about 60 to 70 mV with negative polarity in the intracellular
the prognosis may be quite different in these two cases. space can be recorded. This membrane potential is subject to
various fluctuations that are elicited chiefly by synaptic activi-
POTENTIAL GENERATION AT NEURONAL ties. Their mechanisms are shown in greater detail in Figure 2.2.
AND GLIAL ELEMENTS: MEMBRANE As can be derived from this schematic illustration, the neuron
POTENTIALS AND FIELD POTENTIALS
The basic mechanisms that give rise to potentials recorded out-
side the CNS elements will be described. Such extracellular
potentials are generally known as field potentials (1).
In the course of this presentation, the morphology of gen-
erator structures is discussed briefly. Then, the electrical activ-
ity demonstrable with intracellular recordings from neurons
and glia cells is described. On the basis of this information,
the principles of the generation of extracellular field poten-
tials are outlined and the various types of field potentials are
characterized.

GENERATOR STRUCTURES
The CNS essentially consists of nerve cells and glia cells. The
arrangement of neurons usually shows a specific type of lami-
nar character. Glia cells are located between neurons. As shown
in Figure 2.1, several processes emerge from the nucleus-con-
taining cellular soma (body) of the nerve cell. These processes
can be divided into two types according to their function. Most
of the processes are dendrites that branch off into numerous
small ramifications. Every cell also has an axon that may split
up into multiple collaterals. Such an axon provides contact with
other nerve cells or with other target organs. In the case of
interneuronal connections, the contact consists of synapses that Figure 2.1 Schematic drawing of morphology and histology of neu-
cover the dendrites, the soma, and the axon hillock in large ronal and glial elements.

17
18 Part I ■ Basic Principles

contribute primarily to the generation of the extracellular field


potentials in question (1,5,7,8). For this reason, the ionic
mechanisms of these potentials are discussed in greater detail.
The individual events of this process are presented with a mag-
nified time base (Fig. 2.3). With the elicitation of an EPSP, a net
inflow of cations occurs across the subsynaptic membrane. As
shown in Figure 2.2B, a potential gradient develops along the
neuronal membrane in the intra- and extracellular spaces.
Because of this potential gradient, cations move along the
nerve cell membrane through the extracellular space in the
direction of the subsynaptic region. An inversely directed flow
takes place in the intracellular space. With the generation of an
IPSP, there is an outflow of cations from the nerve cell and/or
an inflow of anions into the nerve cell. These changes first
increase the membrane potential at the subsynaptic membrane
in comparison with the surrounding segments of the mem-
brane. For this reason, a potential gradient develops along the
nerve cell membrane, as in the case of the EPSP genesis. This
potential gradient causes, in the extracellular space, a flow of
cations from the subsynaptic region to the surrounding por-
tions of the membrane. An inverse process develops in the
intracellular space (5).
The ion fluxes in the extracellular space are of paramount
significance in the generation of field potentials. Therefore,
these processes are further discussed in the following chapters.

GLIAL MEMBRANE POTENTIALS:


INTRACELLULAR RECORDINGS
Figure 2.2 Membrane potential (MP) changes and current flows dur-
In addition to the neurons, glial cells may also play a role in the
ing synaptic activation. A: The MP of the postsynaptic neuron and the
generation of extracellular field potentials (9,10). Therefore, the
MP of the presynaptic fibers are recorded by means of intracellular
bioelectric properties of glial cells are summarized.
microelectrodes. Action potentials in the excitatory and inhibitory
If a glia cell is penetrated with a microelectrode, a membrane
presynaptic fiber lead to excitatory postsynaptic potential (EPSP) and
potential can be recorded with a polarity similar to that of the
inhibitory postsynaptic potential (IPSP), respectively, in the postsynap-
nerve cells. The size of this membrane potential approximates
tic neuron. Two EPSPs sum up to a superthreshold potential, triggering
the potassium equilibrium potential and hence somewhat
an action potential in the postsynaptic neuron. B: During EPSP and
exceeds the membrane potential of nerve cells. In contrast to
IPSP, ionic current flows occur through as well as along the neuronal
neurons, glial cells fail to show any action potentials, and there
membrane, as shown by arrows. The density of and signs indicate
are also no postsynaptic potentials. Thus, in contrast to neu-
the polarization of the subsynaptic (dark area) as well as that of the
rons, glial cells do not show characteristic potentials that distin-
postsynaptic membrane during synaptic activation.
guish them unmistakably from other cells. The glial membrane
potential, however, is also not constant. An augmentation of the
extracellular potassium concentration (potassium activity)
from which the soma membrane potential is recorded has causes depolarization of glial cells (Fig. 2.4A). Concentration
synaptic connections. The corresponding presynaptic struc- changes of other ions cause only negligible alterations of the
tures are also explored with microelectrodes. If an action poten- glial cell membrane potential. The glial cell is hence compara-
tial travels along the fiber, which ends in an excitatory synapse, ble to a potassium electrode (9,11).
an excitatory postsynaptic potential (EPSP) occurs in the The dependency of the glial membrane potential on the
following neuron (Fig. 2.2A). If two action potentials travel extracellular potassium concentration is the reason for a
along the same fiber with a short interval, there will be a sum- functional linkage with adjacent neuronal structures.
mation of EPSP triggering an action potential on the postsy- Neuronal activity is associated with outflow of potassium
naptic neuron after reaching the membrane threshold. If an ions. As shown schematically in Figure 2.4B, repetitive firing
action potential travels along a fiber ending in an inhibitory of neurons gives rise to increased extracellular potassium
synapse, then hyperpolarization will occur, representing an concentration and hence to glial cell depolarization (12,13).
inhibitory postsynaptic potential (IPSP) (4–6). If the potassium concentration does not affect the entire glial
Because of the time course of the various membrane poten- cell membrane and remains increased only locally, then
tial fluctuations, the postsynaptic potentials are thought to potential gradients build up along the glial cell, giving rise to
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 19

Figure 2.3 Basic mechanisms underlying


generation of potentials (electroencephalogra-
phy [EEG]) and of magnetic fields (magnetoen-
cephalography [MEG]) in the extracellular space
of central nervous system. The description is
based on the assumption that an extended neu-
ronal process, for example, a dendrite, is locally
depolarized by activation of an excitatory synapse.

intra- and extracellular current flows similar to the ones extension may develop on the basis of the aforementioned
described in reference to neuronal synaptic transmissions mechanisms (1,10,14,15). In view of the above-described
(Fig. 2.2). Glial cells frequently have widespread processes functional interconnections, it is quite likely that in the gen-
and furthermore may have close connections with each other. esis of extracellular field potentials an amplifying effect can
For this reason, potential fields of considerable spatial be attributed to the glial cells.

Figure 2.4 Membrane potential (MP) changes of glia cells induced by an increase in the extracellular K concentration
(arrows in the schematic drawings). A: Potassium is applied extracellularly to the glia cell. B: The potassium concentra-
tion is increased due to an activation of a neighboring neuron. (From original tracings from Kuffler SW, Nicholls JG, Orkand
RK. Physiologic properties of glial cells in the central nervous system of amphibia. J Neurophysiol. 1966;29:768–787.)
20 Part I ■ Basic Principles

FIELD POTENTIALS
It has been shown in the preceding section that primary trans-
membranous currents generate secondary ionic currents along
the cell membranes in the intra- and extracellular spaces. The
portion of these currents that flows through the extracellular
space is directly responsible for the generation of field poten-
tials (Fig. 2.3). Particular significance must be ascribed to the
synaptic processes as causing events for the field potentials in
question, especially for their time course. In accordance with
these statements, the generation of extracellular field potentials
will be discussed as exemplified by extracellular fields accompa-
nying synaptic activity (5,8,15,16). The discussion of these
events will again make use of a very protracted time axis
(Fig. 2.3). The explanation of the events is given in reference to
the schematic view in Figure 2.5. This figure shows a widely
stretched neuronal element, with one end segment lying close
to the surface of a central nervous structure. At both ends of
this neuronal unit, the microelectrodes ME1 and ME2 are
inserted. At the same time, the extracellular electrodes E1 and E2
are located at the surface and at the deeper end of the neuronal
element. The potentials picked up from the intra- and extracel-
lular electrodes are shown in the vicinity of each electrode. The
potential recorded from the surface of the nervous structure is
accentuated by thicker lines. Figure 2.5 shows active excitatory
and inhibitory synapses, either close to the surface or located in
the depth. As described elsewhere, the activation of an excita-
tory synapse leads to a net inward flow of cations. If this state-
ment is applied to Figure 2.5A1, then it becomes evident that Figure 2.5 Membrane potential (MP) changes and field potentials
the upper end of the neuronal element will be depolarized in (FPs) elicited by the activation of excitatory and inhibitory synapses
comparison with other segments of the same cell. Accordingly, in the central nervous system. The elementary processes are explained
the synaptic current flow causes an EPSP at the microelectrode by means of a neuronal element (hatched area), the one end of which
ME1. This local depolarization then gives rise to further intra- contracts the surface of a structure in the central nervous system. The
and extracellular ionic currents along the nerve cell membrane. MP of the neuron element is recorded at both ends by the microelec-
Because of the intracellular movements of positive charges, trodes ME1 and ME2. The extracellular field is picked up at the surface
depolarization in the area of microelectrode ME2 also takes of the neuronal structure by the electrode E1, as well as in the vicin-
place. This depolarization, however, is less steep and of smaller ity of ME2 by the electrode E2. Active excitatory and inhibitory
amplitude. At the superficially located extracellular electrode synapses are marked by open triangles and black triangles (S), respec-
E2, the inflow of positive charges into the neuronal element tively. A1: The inward current at S generates an EPSP that appears in
causes a negative field potential. The extracellular electrode E2 the region of ME1, as well as in that of ME2. Because S is located
is, metaphorically speaking, approached by positive charges so superficially, the FP generated, due to the direction of the extracellu-
that a positive field potential will develop in this area. The point lar current flow (arrows), is of negative polarity at the surface (E1)
of reversal of the field potentials is localized between electrodes and of positive polarity in the deeper recording (E2). A2: The activa-
E1 and E2. The exact position of the point of reversal depends tion of a deep excitatory synapse elicits a current flow with inverse
on the distribution of extracellular impedances. direction as compared with A1. Therefore, the extracellular FP con-
Current flows of reversed direction (in reference to the sists of a positive deflection at the surface and a negative one at the
recording electrodes) will occur if the active excitatory synapse depth. B1: The outward current at S generates an IPSP in the region
is located at the deeper end of the neuronal element (Fig. of ME2, as well as in that of ME1. Due to the direction of the extracel-
2.5A2). In this case, positive charges approach the superficially lular current flow, the FP generated consists of a positive fluctuation
located electrode (E1) (again speaking metaphorically) and in the depth (E2) and a negative one in the surface recording (E1).
remove themselves from the deeply located electrode (E2). This B2: The current flow during the activation of a superficial inhibitory
arrangement of the active synaptic structures causes a positive synapse is inverse as compared with B1. Therefore, the FP recorded
field potential at the surface and a negative one at the deep elec- from the surface consists of a positive fluctuation. Differences in the
trode. The current flows accompanying the activation of time course of the various potentials are caused by the electrical prop-
inhibitory synapses located in deeper and in more superficial erties of the tissue.
areas, respectively, are shown in Figure 2.5B. As can be derived
from this illustration, the activation of a deep inhibitory
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 21

synapse (Fig. 2.5B1) produces a current flow that is largely sim-


ilar to the one generated by the activation of a superficial exci-
tatory synapse (Fig. 2.5A1). In the same manner, there are also
similar current flows in the extracellular space when a superfi-
cial inhibitory synapse (Fig. 2.5B2) or a deeply located excita-
tory synapse (Fig. 2.5A2) is activated. Accordingly, a negative
field potential will develop at the surface of a central nervous
structure (in the schematic view of Fig. 2.5) whenever a super-
ficial excitatory or a more deeply located inhibitory synapse is
activated. The corresponding principle applies to generation of
the superficial field potentials of positive polarity.
Due to their higher densities especially the currents flowing
through the intracellular spaces give rise to magnetic fields sur-
rounding the elements. These magnetic fields can be picked up
from the scalp as magnetoencephalography (MEG; Fig. 2.3).

Types of Field Potentials


The field potentials, whose generation has been described, can
be subdivided into different types. If field potentials are
recorded against an inactive reference point with an upper fre-
quency limit of about 100 Hz, then two types of field potentials
can be distinguished, depending on the time constant of the
amplifying recording device. In the case of a time constant of
1 second or less, the extracellular field potentials correspond
with that which is commonly known as the EEG. If the record-
ing is carried out with an infinite time constant, that is, with
direct current (DC) amplifier, then slower potentials can also be
picked up. Potentials recorded with this technique are generally
known as DC potentials (1,8,15,17). Thus, DC potentials com-
prise slow as well as fast field potentials. The fast components
correspond with the potential fluctuations of the EEG. Due to
different time constants, however, the faster potential compo-
nents may differ from each other as far as their time course is
concerned when recordings are done either with conventional
EEG amplifiers or with DC amplifiers.
Thus far, technical problems have made it difficult to carry
out DC recordings from the scalp. Except for special areas of
application, DC recordings are usually performed in animal
experiments. DC potentials directly reflect the state of activity
of central nervous cells and therefore contribute to the explana- Figure 2.6 Recording of neuronal membrane potentials using voltage-
tion of the mechanisms of genesis of cerebral field potentials sensitive dyes, and principles and schematic example of application.
(14,18). For this reason, DC potentials will be discussed jointly A: (1) A dye is incorporated into the double lipid membrane of nerve
with EEG waves. cells and illuminated by light with dye-specific wave length; simultane-
For the sake of comparison, Figure 2.6 shows the EEG and ously the membrane potential is recorded with an intracellular micro-
the DC potentials during convulsive activity, hypercapnia, and electrode against a reference electrode in the extracellular space.
asphyxia. As shown in this illustration, a tonic–clonic convul- (2) Changes of the membrane potential (MP) starting from the resting
sion is associated with a negative DC shift (8,13–15,19–21). level passing a decrease (depolarization) with action potentials superim-
Furthermore, it can be seen that the hypercapnia-induced dis- posed and a subsequent increase (hyperpolarization) and eventually
appearance of the EEG is associated with a monophasic positive returning to resting level. (3) In correspondence to the different MP levels
DC shift. In the case of EEG extinction due to primary fluorescence and absorption of the dye change. With fluorescent dyes a
asphyxia, however, there are characteristic patterns of DC fluc- depolarization is associated with a decrease and a hyperpolarization with
tuation. Hence, similar findings in the conventional EEG may an increase of fluorescence (symbols). B: (1) Two neurons in a popula-
be associated with different DC shifts. tion; one stays in the resting state and the other changes its MP as in
A2. (2) By the aid of a microscope and a connected array of diodes
Original Remarks Made by Ernst Niedermeyer (squares) the different MP changes of both neurons can be detected via
What does the term DC shift mean? What is DC? Speaking from the different optical behaviors. (Speckmann E-J. Das Gehirn meiner Kunst.
experience, many electroencephalographers have no clear Kreativität und das selbstbewußteGehirn. Münster: Daedalus; 2008.)
22 Part I ■ Basic Principles

concept regarding DC potentials or DC shifts. One cannot


blame them because, for strange reasons, “DC” has two mean-
ings in this context:
1. DC means direct current (and this is, of course, common-
place): a current without oscillations; a current derived from
a battery source; a current maintained in one direction
through a circuit. A more imperfect DC is produced by a rec-
tifier, used to change alternating current (AC) into DC. For
multilingual readers, DC is courant continu in French,
Gleichstrom in German, and corrente continuo in Italian.
Electroneurophysiologically, DC shifts are ultraslow poten-
tials, about as slow as 0.1 to 0.2/sec. This, however, is not true
DC. Such slow activity is just a bit more “DC-like” since it
does not show the faster “AC-like” activity. One simply has to
live with this kind of misnomer.
2. DC also means direct coupling (and this is much less
known). What coupling? The coupling between the stages of
EEG amplification. Conventional EEG machines have stages
coupled by capacitors. Now one has to remember that capac-
itors (a) reject DC and (b) determine the time constant.
Even a very long time constant (several seconds duration)
may not suffice for the recording of DC potentials. Direct
coupling is a capacitor-free coupling between the stages of Figure 2.7 Simultaneous detection of membrane potentials (MP) of all
amplification and provides the optimal condition for DC neurons in a population (voltage-sensitive dye) and conventional
recording. This is technically quite difficult in clinical con- recording of the local field potential (FP) at the same time. Living brain
ventional EEG recording but easier under experimental neu- tissue (0.5 mm thick) from the temporal lobe of a patient who under-
rophysiologic conditions in animals.
went epilepsy surgery. A: Recording of the local FP (“local EEG”).
Hence, be aware of the dual significance of the term DC. (1) Resting state; (2 to 4) epileptic discharges of different intensities.
Epileptic discharges appeared spontaneously, that is, they were not
Validation of Field Potentials induced experimentally. B: MP changes indicated by the intensity of flu-
As to Their Functional Significance orescence of the dye (black, decrease of the MP, depolarization).
Correlating field potentials with other signals accompanying Resting state (1); similar epileptic potentials in the FP (2 and 3) are
neuronal activity may contribute to a validation of them. There associated with different extents of neuronal depolarizations and simi-
is a variety of methods other than electroencephalography for lar extents of neuronal depolarizations with different epileptic poten-
detecting brain activity. Thus, single photon emission tomogra- tials in the FP (3 and 4). (Speckmann E-J. Das Gehirn meiner Kunst.
phy (SPECT), positron emission tomography (PET), functional Kreativität und das selbstbewußteGehirn. Münster: Daedalus; 2008.)
magnetic resonance imaging (fMRI), and intrinsic optical imag-
ing (IOI) are based on metabolic changes associated with
increases of local neuronal activity. The latter methods are “very Figure 2.7. The living brain slices are stained with fluorescence
indirect” (22). EEG including evoked potentials (EP) and MEG (or absorption) dyes (Fig. 2.7A1). With depolarization and
represent “more direct ones” since they measure the field effects hyperpolarization the fluorescence is decreased and increased,
of the proper neuronal activity and therewith of the information respectively (Fig. 2.7A2 and A3). The changes in fluorescence are
processing brain activity. For the analysis of neuronal network measured via a microscope by an array of detectors and there-
functions, the immediate and simultaneous recording of mem- with the actual membrane potentials of the neurons are
brane potentials of all neurons in a population by application of observed (Fig. 2.7B1 and B2).
voltage-sensitive dyes is the “only direct” method available yet The method of simultaneous measurement of neuronal
(23–28). All these methods have advantages and disadvantages. membrane potentials of all neurons in a population is success-
Thus, the functional imaging using voltage-sensitive dyes cannot fully applied in living human brain slices (0.5 mm thick) in
be applied in patients for several reasons, for example, prerequi- vitro obtained from neurosurgical interventions (tumor and
site of direct access to the brain structure to be investigated, pho- epilepsy surgery) (25–27,29).
totoxicity, and pharmacologic side effects of the dyes. But, this A comparison of the field potentials, that is, the local EEG,
method is helpful to analyze the principles of functional mean- and of the neuronal membrane potentials detected by the aid
ing of field potentials in living human brain slices in vitro, espe- of voltage-sensitive dyes is given in Figure 2.8. The tissue is a slice
cially with spontaneously occurring epileptic discharges. preparation from the temporal neocortex resected from a patient
A principle and schematic example of recording neuronal suffering from pharmacoresistant complex partial seizures. Most
membrane potentials using voltage-sensitive dyes is displayed in of these living human brain slices show spontaneous epileptic
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 23

Figure 2.9 Principles of wave generation. The excitatory synapses of


two afferent fibers contact the superficial dendritic arborization of two
longitudinal neuronal elements. The afferent fiber activity is recorded by
means of the intracellular electrodes E1 and E2, and the membrane
potentials (MPs) of the dendritic elements are recorded by the electrodes
Figure 2.8 EEG (time constant: 1 second; upper frequency limit: E3 and E4. The field potential at the surface of the neuronal structure
100 Hz) and DC/ EEG recordings (DC recording: upper frequency limit, (cortex) is led by the electrode E5. Synchronized groups of action poten-
100 Hz) during a generalized seizure induced by pentylenetetrazol tials in the afferent fibers (E1, E2) generate wavelike EPSPs in the den-
(A), during hypercapnia (B), and during asphyxia (C). Original record- dritic areas (E3, E4) and corresponding field potentials in the EEG and
ings were obtained from cats and rats. Note the different time scales. DC/ EEG recording (E5). Tonic activity in the afferent fibers results in a
long-lasting EPSP with small fluctuations. During this period the EEG
(5b) shows only a reduction in amplitude, whereas the DC/ EEG record-
EEG potentials, that is, epileptic discharges not induced by exper- ing (5a) reflects the depolarization of the neuronal elements as well.
imental procedures. One can derive the following from the
recordings:
microelectrodes E1 and E2 are located in the ascending fibers and
1. During epileptic discharges only a certain portion of the neu-
the microelectrodes E3 and E4 are in the superficial dendrites of
rons in the population is active simultaneously, that is, a com-
the postsynaptic neurons. To pick up the extracellular field
plete synchronization is missing (Fig. 2.7, numbers 2 to 4).
potentials, the electrode E5 lies on the surface of the central
2. Similar epileptic potentials in the EEG (Fig. 2.7, numbers
nervous structure.
2 and 3) are associated with different extents of neuronal
As shown in tracings 1 and 2, action potentials occur synchro-
depolarizations and similar extents of neuronal depolar-
nously in the afferent fibers. There are grouped discharges that
izations with different epileptic potentials in the EEG
are temporarily supplanted by tonic activity. The ascending
(numbers 3 and 4).
action potentials elicit individual EPSP in the upper dendrites of
the neurons; these EPSPs are subsequently summated into major
WAVE GENERATION depolarizations in accordance with the discharge frequency. As
shown in tracings 3 and 4, amplitude and duration of the depo-
In the preceding sections, the generation of single field poten- larizations depend on the discharge pattern of the afferent fibers.
tials was described. In this section, the principles of the genera- The synaptic activity at the superficial structures gives rise to
tion of wavelike potential fluctuations are outlined. This is extracellular current flows resulting in superficial field potentials.
followed by the discussion of the laminar distribution of such With the use of DC recording techniques, the superficial field
potentials in the cerebral cortex. potentials reflect the potential fluctuations of the dendritic mem-
To present the generation of wavelike potential fluctuations brane. If, however, the superficial field potentials are recorded
on the surface of a central nervous structure, a simple model as with a time constant of 1 second or less, then only the fast fluctu-
shown in Figure 2.9 is used. This model consists of two extended ations of the superficial field potentials are demonstrable.
pyramidal neurons of vertical orientation. Terminals of afferent Thus far, the principles of genesis of EEG and DC waves
fibers make contact with the superficial dendrites of both neu- have been shown in the schematic view of Figure 2.9.
rons via excitatory synapses. The bioelectrical activity of these Accordingly, the generation of physiologic EEG waves may be
structures is recorded with intracellular microelectrodes. The explained as follows. If a grouped and synchronous influx
24 Part I ■ Basic Principles

takes place in afferent fiber systems toward the superficial


generator structures, then EEG waves evolve that are of high
amplitude and distinctly separated from each other. In case
of a periodic sequence of the afferent bursts, the recording of
the field potentials shows sinusoidal potential fluctuations.
This mechanism has been presumed by several groups of
investigators as the principle of the generation of the alpha
rhythm and slower periodic EEG waves. According to these
workers, thalamocortical feedback loops are believed to play
a significant role in the generation of the alpha rhythm
(see Chapters 3 and 4 for detailed descriptions of the genera-
tion of rhythmic activities) (1,30).
If the afferent influx of impulses occurs at a high frequency
for a longer period and/or synchronously, then negative field
potentials with small fluctuations will result from the extracel-
lular current flows. Accordingly, the EEG recording will pick
up only waves of smaller amplitude and mostly higher fre-
quency. In the DC recording, however, the prolonged depolar-
ization of the superficial structures caused by the afferent
high-frequency influx will express itself by a negative DC
potential shift (31,32). There is a close correlation between the Figure 2.10 Surface (1) and laminar recordings (2 to 6) of EEG waves
amplitude of the negative DC shift and average discharge fre- of the cortex. The schematic drawing symbolizes conical neuronal ele-
quency in the afferent fiber systems. This mechanism may ments densely packed with synapses. (Drawings from original tracings
apply principally to the generation of beta activity and other obtained in experiments in the rat’s motor cortex during pentobarbital
EEG waves of higher frequencies (see also cortical sources of anesthesia.)
beta and gamma rhythms in Chapter 3 and Fig. 3.16). A
decrease of the amplitudes of the EEG waves can also occur
when the afferent activity is diminished. In this case, however, CORTICAL FIELD POTENTIALS
the depression of EEG waves is accompanied by a positive DC DURING EPILEPTIFORM ACTIVITY
shift (see Fig. 2.17) (33).
The principles of generation of individual and wavelike In the following subsections, the generation of cortical field
field potentials at the surface of central nervous structures potentials during convulsive activity is discussed. The first sub-
such as the cerebral cortex have been described. If the wave- section deals with focal activity, and the second discusses gener-
like potential fluctuations are recorded not only from the alized tonic–clonic convulsive activity. For methodical reasons,
cortical surface, but also from different cortical layers, then it we refer to data derived from experimental work in animals.
can be shown that potential fluctuations in the latter record-
ings may differ considerably from those at the surface. Focal Activity
These differences imply polarity, frequency, and amplitude If a convulsive substance such as penicillin is applied to the sur-
(1,34,35). Such a recording from the cortex of the rat is shown face of the cerebral cortex, steep negative potentials of high
in Figure 2.10. According to this illustration, field potentials amplitude can be picked up from the area of application after a
reverse their polarity between electrode 1 (on the surface) and short latency period. These discharges repeat themselves in
electrode 2 (located 300 m beneath the cortical surface). Two stereotyped form and periodicity (Fig. 2.11A) (13,36,37). If the
and sometimes more of such phase reversals may be observed membrane potential of a cortical neuron is simultaneously
in deeper recording sites depending on the experimental con- recorded with a microelectrode while a second microelectrode
ditions (phase reversals of alpha oscillations are shown in picks up the corresponding field potentials, then potential fluc-
Chapter 3, Figs. 3.9 and 3.10). The vertical distribution type of tuations occur as shown in Figure 2.11B. It can be derived from
field potential will be discussed in greater detail in connection this illustration that the monotonously recurrent negative field
with the generation of cortical field potentials during convul- potentials are associated with equally stereotyped membrane
sive activity. potential fluctuations. These oscillations of the membrane
In the course of the discussion of cerebral field potentials, it commence with a steep depolarization that, having exceeded
was pointed out that particular significance must be attributed to the membrane threshold, triggers a series of action potentials.
synaptic activity. A view of the laminar distribution of neurons in This is followed by a plateau that, after 80 to 100 msec, changes
the cortex and the dense coverage of these unitary structures with into a steep repolarization, and frequently also into a hyperpo-
synapses makes it clear that different patterns of potentials must larization. These membrane potential fluctuations have proved
necessarily occur in different layers when populations of synapses to be characteristic in the epileptiform activity of individual
are activated in a different manner. This should be clarified by the neurons. They are generally known as paroxysmal depolariza-
schematic drawing in Figure 2.10. tion shifts (PDSs) (13,38).
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 25

There are, however, field potentials with predominantly posi-


tive components in the deeper contacts 4 to 6. If penicillin is
applied to the surface without penicillinase, then negative
field potential will also develop in deeper cortical layers. If it
is assumed that the negative field potentials mirror the direct
epileptiform activity of neuronal structures (Fig. 2.11), then it
must also be assumed that deeper cortical elements are
involved in convulsive activity shown in of Figure 2.12B in
contrast with Figure 2.12A. This is further supported by the
observation that neuronal activity descending to the spinal
cord and producing characteristic spinal field potentials
occurs only under the experimental conditions shown in
Figure 2.12B. If one compares the recordings in Figure 2.12A
and B, it becomes clear that, with a monotonous epileptiform
Figure 2.11 EEG (A) and membrane potential (MP) A changes of a potential at the cortical surface, the intracortical potential dis-
pyramidal tract neuron and extracellular field potential (FP) recorded in tribution and the occurrence of descending activity may differ
the vicinity of the impaled neuron (B) during focal interictal activity considerably (34,39–44).
elicited by application of penicillin to the cortical surface (hatched area If penicillin is applied to deeper cortical laminae
in A). Drawings of original tracings from experiments in the rat. The (Fig. 2.12C), then negative field potentials will be confined to
sweep speed in B is five times that in A. The recording sites are shown that region. These potentials are consistently accompanied by
in the schematic drawings. descending activity to the spinal cord. Under these conditions,
there is frequently nothing but a positive potential fluctuation
of minor amplitude at the cortical surface (34,40).
Investigation of potential distribution within the cerebral In summary, it can be derived from the described experi-
cortex after the local application of penicillin yields a variety mental models that, in focal convulsive activity limited to the
of findings. An appropriate model is shown in Figure 2.12. In cortex, the surface potential does not necessarily reflect the bio-
this experiment, recordings of interictal field potentials were electrical events in deeper cortical layers.
carried out from the cortical surface, from inside the cortex,
and from the spinal cord. The spinal field potentials permit Generalized Tonic–Clonic Activity
the observation of electrical activity descending from the cor- Here, possible mechanisms involved in the generation of cortical
tex to the spinal cord. In Figure 2.12A, negative field poten- field potentials during tonic–clonic convulsive activity are
tials are recorded from the cortical surface and from the two described. Again, data are based on experimental observations in
upper intracortical contacts after the application of penicillin animals. Tonic–clonic convulsive activity was triggered by
together with penicillin-metabolizing enzyme penicillinase. repeated injections of pentylenetetrazol (also see Refs. 13 and 37).

Figure 2.12 Cortical field potentials recorded at the


surface (1) and from within the cortex (2 to 6) and
spinal field potentials (7) during interictal activity.
The interictal activity was elicited by penicillin.
A,B: Potential distribution after surface application of
the drug. In A, the spread of penicillin is limited by the
use of penicillinase. C: Potential distribution after intra-
cortical application of penicillin at recording point 4.
The areas directly involved in the epileptiform activity
as indicated by negative field potentials are marked by
hatching in the schematic drawings. Spinal field poten-
tials are linked to the occurrence of negative field
potentials in lamina V (B and C, 4). Distance between
the intracortical electrodes, 300 m. (From original trac-
ings from Elger CE, Speckmann EJ, Caspers H, et al.
Focal interictal epileptiform discharges in the cortex of
the rat: laminar restriction and its consequences for
activity descending to the spinal cord. In: Klee MR, Lux
HD, Speckmann EJ, eds. Physiology and Pharmacology
of Epileptogenic Phenomena. New York, NY: Raven
Press; 1981.)
26 Part I ■ Basic Principles

Figure 2.13 Simultaneous recordings of EEG and DC/ EEG (A) and of
DC/ EEG and membrane potential (MP) of a pyramidal tract neuron (B)
during generalized tonic–clonic seizures elicited by pentylenetetrazol.
(Drawings after original tracings from experiments in the cat’s motor
cortex. The sweep speed in B is 10 times that in A.) Figure 2.14 Single potential fluctuations at the cortical surface
(DC/ EEG) and concomitant membrane potential (MP) of a pyramidal
tract cell (PTC) and field potentials (FP) in the PTC layer during gener-
Figure 2.13A shows a tonic–clonic convulsion recorded with alized tonic–clonic seizures. The seizure activity was induced by
a conventional EEG amplifier, as well as with a DC amplifier. pentylenetetrazol. A: The negative potential (1), the positive–negative
There is a negative DC shift from the baseline during a convul- fluctuation (2), and the positive potential (3) in the DC/ EEG recording
sive seizure. This negative DC shift gradually recedes during the coincide with monophasic negative FP and stereotyped paroxysmal
termination of the convulsions and frequently changes into a depolarization shift in the neuron. The negative DC shift occurring dur-
transient positive after shift (13–15,18,19,21,41). ing the seizure is indicated by a dashed line in the upper row.
When the membrane potential of a pyramidal tract neuron Monophasic negative potentials in the DC/ EEG recording occur with
of lamina V is recorded during a convulsive seizure, it can be small and monophasic positive fluctuations along with a marked DC
shown that under these conditions typical PDSs become mani- displacement. B: The relations between DC/ EEG potentials and MP of
fest (Fig. 2.13B). If these PDSs are correlated with the potential PTC as described for A1 and A3 also hold true for trains of potentials
fluctuations in the DC recording, it can be noticed that the (1 and 2). (From original tracings from Speckmann EJ, Caspers H,
PDSs in pyramidal tract neurons are coupled at the beginning Jansen RWC. Laminar distribution of cortical field potentials in relation
of the convulsive seizure with superficial negative potential to neuronal field activities during seizure discharges. In: Brazier MAB,
fluctuations and at the end of the convulsive seizure with sur- Petsche H. eds. Architectonics of the Cerebral Cortex, IBRO Monograph
face-positive potential fluctuations (Fig. 2.13B) (1,18,45). Series. Vol 3. New York, NY: Raven Press; 1978:191–209.)
In addition to the field potentials of the cortical surface and
the membrane potentials of the pyramidal tract cells, field
potentials were also recorded in the fifth lamina. Under these when the negative DC shift at the cortical surface reaches and
conditions, it can be shown that every PDS is associated with a exceeds a critical value (1,18,45,46).
negative monophasic field potential in the depth (Fig. 2.14A). These data are interpreted with flow charts in Figure 2.15.
These stereotyped potential fluctuations in deep cortical layers The amplitude of the negative DC shift at the cortical surface
correspond with field potentials at the cortical surface with depends greatly on the amount of the afferent influx of
either monophasic negative or positive (Fig. 2.14A1 and A3) or impulses to the generator structures in the superficial cortical
with polyphasic (Fig. 2.14A2) configurations. This statement laminae. This predominantly asynchronous afferent influx is
does not merely apply to individual ictal potentials but is also symbolized by the width of hatched arrows in Figure 2.15.
true for prolonged trains of potentials during the convulsion. Accordingly, the afferent influx in Figure 2.15A is smaller than
As Figure 2.14B shows, paroxysmal depolarizations of pyrami- that in Figure 2.15B. Therefore, there is a smaller DC shift in
dal tract cells may be accompanied by a sequence of either neg- Figure 2.15A and a prominent one in Figure 2.15B. In the case
ative or positive potentials on the cortical surface. If one of Figure 2.15A, a synchronized inflow of impulses from sub-
correlates these various field potentials on the cortical surface cortical structures is assumed to reach the cortex (widened
with the slow DC shifts occurring during the convulsion (also afferent fiber in schematic view). As a consequence, pyramidal
see Fig. 2.14A), then it can be demonstrated that the surface- tract cells will be stimulated to generate a PDS, and structures
negative field potentials are associated primarily with a slight close to the surface will be depolarized through the mediation
DC shift and that surface-positive field potentials will appear of interneurons. Accordingly, in such a constellation of
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 27

Figure 2.15 Flow charts of neuronal processes possibly responsible for the generation of DC/ EEG waves of opposite polar-
ity during a generalized tonic–clonic seizure. (Hatched arrows) Symbols for continuous asynchronous input to the cortex;
(heavy lines) symbols for phasic volleys giving rise to single convulsive discharges; (PTC) pyramidal tract cell; (IN)
interneuron; (MP) membrane potential; (UA) extracellularly recorded unit activity. A: During a moderate asynchronous
input to the cortex (small hatched arrow), a burst of UA triggers a paroxysmal depolarization shift in a PTC.
Simultaneously, it leads to a depolarization of superficial neuronal structures and therewith to a negative fluctuation in the
DC/ EEG recording at the cortical surface. B: With an increased asynchronous input to the cortex (wide hatched arrow),
the DC potential shifts to a more negative level than in A (1). When in these conditions a phasic volley reaches the cor-
tex, paroxysmal depolarization shifts are also triggered in PTC, whereas the enhanced asynchronous UA is interrupted
mainly due to inactivation. The latter process results in a disfacilitation of the upper neuronal structures and therewith to
a positive fluctuation of the superficial DC/ EEG potential (2). (From original tracings from Speckmann EJ, Caspers H,
Jansen RWC. Laminar distribution of cortical fluid potentials in relation to neuronal field activities during seizure dis-
charges. In: Brazier MAB, Petsche H, eds. Architectonics of the Cerebral Cortex. IBRO Monograph Series. Vol 3. New York,
NY: Raven Press; 1978:191–209.)

excitatory processes, the paroxysmal depolarization in the characteristic physiologic feature of SD is a negative DC poten-
depth will be coupled with a surface-negative field potential. tial shift with maximal amplitude of 5 to 30 mV and duration
With augmentation of the already existing afferent inflow of of 30 to 90 seconds (Fig. 2.16A) (47). SD recordings by extracel-
impulses, the interneurons involved will necessarily exhibit a lular microelectrodes inserted into the gray matter of the neo-
heightened level of excitation (Fig. 2.15B). If an additional cortex demonstrate that SD can be biphasic or triphasic with
highly synchronized afferent influx of impulses takes place the main negative component. SD begins with a first small pos-
under these conditions, then further PDSs will be triggered in itive component that is sometimes not observed. The main fea-
the pyramidal tract cells, but, in the interneurons, the previ- ture of the SD is a negative wave of high amplitude, which is
ously existing high-frequency activity will be temporarily inter- then followed by a positive-going wave of smaller amplitude,
rupted, chiefly due to inactivation. This causes a decline of the but longer duration. The white matter beneath the neocortical
excitatory inflow of impulses to the superficial cortical struc- gray substance becomes positive, while the gray matter itself
tures. This disfacilitation gives rise to a positive field potential undergoes the negative wave. Apical dendrites were preferen-
at the cortical surface. In this manner, a massive afferent inflow tially involved in the generation of the negative DC deflection.
of impulses provides the basis for a correlation of positive When recordings were made in hippocampal CA1 region,
epicortical field potentials with stereotyped paroxysmal depo- SD invariably started earlier and ended later in the layer of the
larizations and monophasic negative field potentials in the dendritic trees than among the cell body. SD flows in the oppo-
depth (18,45). site direction compared with the current underlying
tonic–clonic seizure discharges, which is inward in the neuron
SPREADING DEPRESSION soma layers (48,49).
A brief period of excitation heralds SD, which is immediately
In this section, the generation and propagation of cortical field followed by prolonged neuronal activity depression. This is
potentials during spreading depression (SD) is discussed. SD is replaced by a sustained increase in the neuronal activity; SD
a strong and rapid depolarization of neuronal tissues and prop- also produces transient suppression of synaptic transmission
agates slowly (3 to 5 mm/min) as a wave in brain tissue (47). that is replaced by a sustained increase in the efficacy of synaptic
The SD phenomenon is exclusive to the CNS and appears to transmission. SD significantly enhanced the amplitude of field
influence both the neuronal and the glial cells. The most EPSP following a transient suppressive period and increased the
28 Part I ■ Basic Principles

from a depolarization of both presynaptic and postsynaptic


processes. In addition, presynaptic depolarization evokes
Ca2 -dependent synaptic transmission and glutamate release.
The latter in turn enhances depolarization of postsynaptic ele-
ments and causes further increases in [K ]o. In addition to potas-
sium and proton release from the cells, sodium, calcium, and
chloride enter together with water causing cells to swell and the
volume of the extracellular compartment to be decreased. SD is
accompanied by an increase of glucose utilization and O2 con-
sumption. Recovery of SD depends on energy metabolism (51,52).
SD can be initiated by different stimuli. The most common
model of SD initiation is KCl application to the neuronal
tissues. Other methods of SD induction include mechanical,
electrical, and thermal stimulations, application of the excita-
tory amino acids such as glutamate and aspartate, the Na –K
ATPase inhibitor ouabain, and metabolic inhibitors such as
NaCN (53). The ionic processes responsible for the generation
of SD are complex. Several ionic currents are involved; both
high [K ] o and low[Ca2 ] o can support either SD or epilepti-
form discharges depending on the state of the various mem-
brane ion channels. Insight into the interplay of these different
factors can best be obtained by the study of computer models
solidly based on biophysical data. Thus, the model of Kager
et al. (2008) revealed how the ignition of SD can result from the
shift in the balance between slowly inactivating (persistent)
inward and outward currents in apical dendrites of pyramidal
Figure 2.16 Propagation of negative DC potential shift in the neocor- cells in favor of the former. The effects of hypoxia and anoxia,
tex and its effect on neuronal excitability. A: Recording of slow spread through changes in Na/K pump and glial buffering, can also be
of negative DC potential wave after injection of KCl in human neocorti- put in evidence with these models (54,55).
cal tissue. Voltage variations were recorded simultaneously by four Processes similar to SD in animal and human neocortex
electrodes (DC1 to DC4), which are set apart by 1 mm. KCl was injected observed in experimental studies are thought to take place in a
in layer VI. Injecting and recording electrodes arranged as shown. number of pathologic conditions in humans. SD belongs to the
B: Suppression of evoked potentials (EP) during spreading of negative domain of the pathophysiology of the brain, and there are rea-
DC potential fluctuation. The DC potential was recorded from layer III sons to believe that it is involved in different clinical disorders,
of a human neocortical slice. EP was elicited by a single electrical including migraine, cerebrovascular diseases, head injury,
stimu lation (ST) applied to afferent fibers. Note the transient suppres- spinal cord disorders, epilepsy, and transient global amnesia
sion of EP during propagation of spreading depression. (Modified from (51). SD can alter the neurochemistry of cortical and subcorti-
Gorji A. Spreading depression: a review of the clinical relevance. Brain cal structures, modulating oxygen distribution, cell survival in
Res Brain Res Rev. 2001;38:33.) these structures, and behavior. Direct alterations of electrical
activity of cortical neurons by the locally spreading wave can
lead to clinical symptoms (e.g., the aura phase of migraine).
The most common symptoms in the aura phase are visual, aris-
induction of long-term potentiation in the neocortex (50). The ing from dysfunction of occipital lobe neurons. The positive
prodromal excitation as well as late hyperexcitability is mani- (excitatory) neurologic symptoms, for example, flashing lights,
fested in appearance of a burst of multiple action potentials and are usually followed by negative (suppressive) ones, for exam-
intense synaptic noise representing synchronized firing of neu- ple, scotoma or hemianopia, in this phase. Subdural recordings
rons. The widely spread synchronization seems best explained in patients demonstrated that SD is critically involved in vari-
by electrical continuity that could be provided by gap junctions. ous disorders associated with acute neuronal injury including
Patent gap junctions may provide a pathway not only for electric traumatic and spontaneous intracerebral hemorrhage (56).
currents and ions, but also for intracellular “second” messengers
and other active ingredients in cytosol (48). CORTICAL FIELD POTENTIALS DURING
SD is associated with transient, but major, localized redistrib- GAS TENSION CHANGES IN TISSUE
utions of ions between the extra- and intracellular spaces. The
[K ] o rapidly rises and can reach levels as high as 40 to This section deals with the alterations of epicortical field poten-
80 mmol/L. This causes neuronal excitation followed by depolar- tials and concomitant changes of the membrane potentials
ization and a period of electrical silence during which the DC caused by deviations of the gas tension in brain tissue. Such
potential at the brain surface falls. Increases in [K ]o could result changes of the gas tension may occur when, for instance, the
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 29

pulmonary and circulatory function is disturbed or when the With isolated increment of the CO 2 tension in the cerebral
local cerebral blood flow is inadequate. tissue by means of the apnea technique, the amplitude of the
First, the alterations of epicortical field potentials during conventional EEG decreases progressively. This amplitude
selective hypercapnia are discussed; then, those associated with reduction affects first the waves of higher frequency and then
primary asphyxia are considered. It is shown that EEG changes those of lower frequency. Prior to the extinction of normal
may be similar under both conditions. The cortical DC poten- EEG activity, there is once again a phase characterized by
tial, however, shows typical shifts that permit inferences con- high-frequency EEG activity in the range of 50 to 70 Hz
cerning the cause of the accompanying EEG changes. The (1,57). The extinction of the EEG is associated with a shift of
discussion of the effects of gas tension alterations on the bio- the DC potential in a positive direction. If the CO 2 tension is
electrical activity of the CNS is based, again, on data derived then lowered again by reventilation, the EEG waves return in
from experimental work in animals. the original spectral composition after a short latency. At the
same time, the positive DC shift resolves (Fig. 2.17).
Hypercapnia Experiments in animals have shown that, with reduction of
If the CO2 tension in the brain tissue is increased in a selective the pCO2, the EEG returns to normal activity even though the
manner, typical reactions of the cortical field potentials as well hypercapnia-induced suppression lasted for 1 hour or more.
as of the membrane potential and the postsynaptic potentials of In these cases, a positive DC deflection of monophasic charac-
individual neurons are found. These findings are shown in a ter was found to occur during the whole period of apnea
summarized schematic view in Figure 2.17. (33,57,58).
The animal experiments on which Figure 2.17 is based were Under the aforementioned conditions, the recording of the
carried out with the use of the so-called apnea technique. With membrane potential of a cortical nerve cell shows a hyperpolar-
this technique, interference of the effects of hypercapnia with ization while the CO2 tension is increased. Extensive experi-
simultaneous effects of hypoxia could be avoided. According to mental studies in animals have demonstrated that such a
this technique, the experimental animal is ventilated for at least hyperpolarization is caused primarily by a reduction of the
a half hour with pure oxygen. Thereafter, artificial ventilation is EPSP (Fig. 2.17; also see Ref. 58). Consideration of field poten-
discontinued while the trachea of the animal remains con- tials, membrane potentials, and EPSP shows that epicortical DC
nected with the O2 reservoir. Under these conditions, the CO2 potentials reflect neuronal hyperpolarization. The disappear-
tension progressively rises in the tissue for about 15 minutes ance of the EEG waves is presumed to be caused mainly by the
without a concomitant fall of the oxygen tension below the reduction of postsynaptic activity.
baseline level.
Asphyxia
Primary asphyxia exemplified by respiratory arrest after air ven-
tilation is associated with combined CNS effects of hypercapnia
and hypoxia. The effects of gas tension changes on the field
potentials and on the membrane potential of individual neu-
rons are schematically shown in Figure 2.18. In the correspon-
ding animal experiments, the artificial ventilation with air was
either temporarily (Fig. 2.18A) or persistently (Fig. 2.18B)
interrupted.
With such an interruption of artificial ventilation with
air, the conventional EEG waves disappear within less than
1 minute. This process is accompanied by a negative DC poten-
tial shift from the baseline, which has been characterized as ini-
tial negativity (1 in Fig. 2.18). While the EEG shows an
isoelectric line in the further course of asphyxia, additional
potential shifts are detectable with DC recording technique.
The initial negativity is followed by a positive DC shift termed
Figure 2.17 Effects of an isolated hypercapnia on epicortical field intermediate positivity (2 in Fig. 2.18). If reventilation is per-
potentials (EEG, DC/ EEG) and on membrane potential (MP). With formed in this phase of asphyxia, an additional positive DC
increasing pCO2, the EEG disappears even if the pO2 is above normal shift is observed, appropriately termed reactive positivity (3 in
levels. The disappearance of the EEG is associated with a positive DC Fig. 2.18A). According to the analysis of the experimental work,
shift and a hyperpolarization of most of the neurons. Simultaneously, the intermediate and the reactive types of positivity are due to
the amplitudes of stimulus (St) evoked EPSP are markedly reduced. an increase of CO2 tension in the brain tissue. With the resolu-
(From original tracings from Speckmann EJ, Caspers, H. The effect of tion of the reactive positivity, a restitution of the fast field
O2 and CO2 tensions in the nervous tissue on neuronal activity and DC potentials occurs that is also demonstrable with the conven-
potentials. In: Remond A, ed-in-chief. Handbook of Electroence- tional EEG. A comparison of the DC shifts and the alterations
phalography and Clinical Neurophysiology. Vol 2. Part C. Amsterdam: of the membrane potentials shows a parallelism of both events
Elsevier; 1974:71–89.) (1,14,15,33,57,58).
30 Part I ■ Basic Principles

5. Hubbard JI, Llinas R, Quastel DMJ. Electrophysiological Analysis of


Synaptic Transmission/Monographs of the Physiological Society.
London: Edward Arnold; 1969.
6. Shepherd GM. The Synaptic Organization of the Brain. London:
Oxford University Press; 1974.
7. Creutzfeldt O, Houchin J. Neuronal basis of EEG waves. In:
Remond A, ed-in-chief. Handbook of Electroencephalography and
Clinical Neurophysiology. Vol 2. Part C. Amsterdam: Elsevier;
1974:5–55.
8. Speckmann EJ, Caspers H, Elger CE. Neuronal mechanisms
underlying the generation of field potentials. In: Elbert T,
Rockstroh B, Lutzenberger W, et al., eds. Self-Regulation of the
Brain and Behavior. New York, NY: Springer; 1984:9–25.
9. Kuffler SW, Nicholls JG. The physiology of neuroglial cells. Erg
Physiol. 1966;57:1–90.
10. Somjen GG, Trachtenberg M. Neuroglia as generator of extracellu-
Figure 2.18 Alterations of EEG, DC/ EEG, and neuronal membrane lar current. In: Speckmann EJ, Caspers H, eds. Origin of Cerebral
potential (MP) during primary asphyxia. A: The abolition and the reap- Field Potentials. Stuttgart: Thieme; 1979:21–32.
pearance of EEG during a transient asphyxia goes in parallel with typi- 11. Kuffler SW, Nicholls JG, Orkand RK. Physiological properties of
cal DC shifts: (1) initial negativity, (2) intermediate positivity, and (3) glial cells in the central nervous system of amphibia. J Neuro-
physiol. 1966;29:768–787.
reactive positivity. These DC fluctuations are accompanied by corre-
12. Orkand RK, Nicholls JG, Kuffler SW. Effect of nerve impulses on
sponding reactions of the MP. B: With continuing asphyxia, the EEG
the membrane potential of glial cells in the central nervous system
remains abolished and the intermediate positivity (2) turns over into a of amphibia. J Neurophysiol. 1966;29:788–806.
terminal negativity (4). The latter DC negativity corresponds to a break- 13. Speckmann E-J. Experimentelle Epilepsieforschung. Darmstadt:
down of neuronal membrane potential. (From original tracings from Wissenschaftliche Buchgesellschaft; 1986.
Speckmann EJ, Caspers H. The effect of O2 and CO2 tensions in the 14. Caspers H, Speckmann E-J, Lehmenkühler A. Electrogenesis of
nervous tissue on neuronal activity and DC potentials. In: Remond A, cortical DC potentials. In: Kornhuber HH, Deecke L, eds.
ed-in-chief. Ha ndbook of Electroen cepha logra phy a nd Clinica l Motivation Factor and Sensory Processes of the Brain: Electrical
Neurophysiology. Vol 2. Part C. Amsterdam: Elsevier; 1974:71–89.) Potentials, Behaviour and Clinical Use. Progress in Brain Research.
Vol 54. New York, NY: Elsevier; 1980:3–15.
15. Caspers H, Speckmann E-J, Lehmenkühler A. Electrogenesis of
slow potentials of the brain. In: Elbert T, Rockstroh B,
If the interruption of the artificial ventilation is continued Lutzenberger W, et al., eds. Self-Regulation of the Brain and
for a longer period of time, then the intermediate positivity Behavior. New York, NY: Springer; 1984:26–41.
converts into the so-called terminal negativity (4 in Fig. 2.18B). 16. Rall W. Core conductor theory and cable properties of neurons. In:
This negative DC shift correlates with the breakdown of the Kandel ER, ed. Handbook of Physiology/The Nervous System. Vol 1.
neuronal membrane potential. The terminal effects are due to a Part 1. Bethesda, MD: American Physiological Society;
critical lack of oxygen. The terminal negativity may be 1977:39–97.
reversible for a substantial period of time under certain experi- 17. Caspers H, ed. DC potentials recorded directly from the cortex. In:
mental conditions if the artificial ventilation is resumed and the Remond A, ed-in-chief. Handbook of Electroencephalography
reduction of the cerebral circulation is counteracted with circu- and Clinical Neurophysiology. Vol 10. Part A. Amsterdam: Elsevier;
1974.
lation support measures (58).
18. Speckmann E-J, Caspers H. Cortical field potentials in relation to
In summary, a comparison of EEG and DC potentials in
neuronal activities in seizure conditions. In: Speckmann E-J,
selective hypercapnia and primary asphyxia shows that the Caspers H, eds. Origin of Cerebral Field Potentials. Stuttgart:
recording of cortical field potentials with DC amplifiers pro- Thieme; 1979:205–213.
vides a more accurate picture of the actual functional state of 19. Caspers H, Speckmann E-J. DC potential shifts in paroxysmal
nerve cells. states. In: Jasper HH, Ward AA Jr, Pope A, eds. Basic Mechanisms of
the Epilepsies. Boston, MA: Little, Brown & Company; 1969:
375–388.
REFERENCES 20. Gumnit RJ, Matsumoto H, Vasconetto C. DC activity in the depth
of an experimental epileptic focus. Electroencephalogr Clin
1. Speckmann E-J, Caspers H, eds. Origin of Cerebral Field Potentials. Neurophysiol. 1970;28:333–339.
Stuttgart: Thieme; 1979. 21. Speckmann E-J, Elger CE. The neurophysiological basis of epilep-
2. Palay SL, Chan-Palay V. General morphology of neurons and neu- tic activity: a condensed overview. In: Degen R, Niedermeyer E,
roglia. In: Kandel ER, ed. Handbook of Physiology/The Nervous eds. Epilepsy, Sleep, and Sleep Deprivation. Amsterdam: Elsevier;
System. Vol 1. Part 1. Bethesda, MD: American Physiological 1984:23–34.
Society; 1977:5–37. 22. Speckmann E-J. Das Gehirn meiner Kunst. Kreativität und das
3. De Robertis EDP, Carrea R, eds. Biology of Neuroglia/Progress in selbstbewußteGehirn. Münster: Daedalus; 2008.
Brain Research. Vol 15. New York, NY: Elsevier; 1964. 23. Cohen LB, Salzberg BM. Optical measurement of membrane
4. Eccles JC. The Physiology of Synapses. Berlin: Springer; 1964. potential. Rev Physiol Biochem Pharmacol. 1978;83:35–83.
Chapter 2 ■ Neurophysiologic Basis of EEG and DC Potentials 31

24. Ebner TJ, Chen G. Use of voltage-sensitive dyes and optical record- 42. Petsche H, Pockberger H, Rappelsberger P. Current source density
ings in the central nervous system. Prog Neurobiol. 1985;46: 463–506. studies of epileptic phenomena and the morphology of the rabbit’s
25. Köhling R, Höhling J-M, Straub H, et al. Optical monitoring of neu- striate cortex. In: Klee MR, Lux HD, Speckmann E-J, eds.
ronal activity during spontaneous sharp waves in chronically epilep- Physiology and Pharmacology of Epileptogenic Phenomena. New
tic human neocortical tissue. J Neurophysiol. 2000;84: 2161–2165. York, NY: Raven Press; 1981.
26. Köhling R, Reinel J, Vahrenhold J, et al. Spatio-temporal patterns 43. Speckmann E-J, Elger CE, eds. Epilepsy and Motor System.
of neuronal activity: analysis of optical imaging data using geo- Baltimore: Urban & Schwarzenberg; 1983.
metric shape matching. J Neurosci Methods. 2002;114:17–23. 44. Wieser HG. Electroclinical Features of the Psychomotor Seizure. A
27. Straub H, Kuhnt U, Höhling J-M, et al. Stimulus induced patterns Stereoencephalographic Study of Ictal Symptoms and
of bioelectric activity in human neocortical tissue recorded by a Chronotopographical Seizure Patterns Including Clinical Effects of
voltage sensitive dye. Neuroscience. 2003;121:587–604. Intracerebral Stimulation. Stuttgart: Gustav Fischer; 1983.
28. Grinvald A, Hildesheim R. VSDI: a new era in functional imaging 45. Speckmann E-J, Caspers H, Janzen RWC. Laminar distribution of
of cortical dynamics. Nat Rev Neurosci. 2004;5:874–885. cortical field potentials in relation to neuronal activities during
29. Gorji A, Straub H, Speckmann E-J. Epilepsy surgery: perioperative seizure discharges. In: Brazier MAB, Petsche H, eds. Architectonics
investigations of intractable epilepsy. Anat Embryol (Berlin). of the Cerebral Cortex. IBRO Monograph Series. Vol 3. New York,
2005;210:525–537. NY: Raven Press; 1978:191–209.
30. Andersen P, Andersson SA. Physiological Basis of the Alpha Rhythm. 46. Speckmann E-J, Caspers H, Janzen RWC. Relations between corti-
New York, NY: Meredith; 1968. cal DC shifts and membrane potential changes of cortical neurons
31. Caspers H. Relations of steady potential shifts in the cortex to the associated with seizure activity. In: Petsche H, Brazier MAB, eds.
wakefulness–sleep spectrum. In: Brazier MAB, ed. Brain Function. Synchronization of EEG Activity in Epilepsies. New York, NY:
Berkeley, CA: University of California Press; 1963:177–213. Springer; 1972:93–111.
32. Goldring S. DC shifts released by direct and afferent stimulation. 47. Leao AAP. Spreading depression of activity in the cerebral cortex.
In: Remond A, ed-in-chief. Handbook of Electroencephalography J Neurophysiol. 1944;7:359–390.
and Clinical Neurophysiology. Vol 10. Part A. Amsterdam: Elsevier; 48. Somjen GG. Mechanisms of spreading depression and hypoxic
1974:12–24. spreading depression-like depolarization. Physiol Rev.
33. Caspers H, Speckmann E-J. Cortical DC shifts associated 2001;81:1065–1096.
with changes of gas tensions in blood and tissue. In: Remond A, 49. Gorji A, Scheller D, Straub H, et al. Spreading depression in
ed-in-chief. Handbook of Electroencephalography and Clinical human neocortical slices. Brain Res. 2001;906:74–83.
Neurophysiology. Vol 10. Part A. Amsterdam: Elsevier; 1974:41–65. 50. Berger M, Speckmann EJ, Pape HC, et al. Spreading depression
34. Elger CE, Speckmann E-J. Penicillin-induced epileptic foci in enhances human neocortical excitability in vitro. Cephalalgia.
the motor cortex: vertical inhibition. Electroencephalogr Clin 2008;28:558–562.
Neurophysiol. 1983;56:604–622. 51. Gorji A. Spreading depression: a review of the clinical relevance.
35. Petsche H, Muller-Paschinger IB, Pockberger H, et al. Depth profiles Brain Res Brain Res Rev. 2001;38:33–60.
of electrocortical activities and cortical architectonics. In: Brazier 52. Smith JM, Bradley DP, James MF, et al. Physiological studies of
MAB, Petsche H, eds. Architectonics of the Cerebral Cortex. IBRO cortical spreading depression. Biol Rev Camb Philos Soc.
Monograph Series. Vol 3. New York, NY: Raven Press; 1978:257–280. 2006;81:457–481.
36. Klee MR, Lux HD, Speckmann E-J, eds. Physiology and 53. Bures J, Buresova O, Krivanek J. The Mechanisms and Applications
Pharmacology of Epileptogenic Phenomena. New York, NY: Raven of Leao’s Spreading Depression of Electroencephalographic Activity.
Press; 1982. New York, NY: Academic Press; 1974.
37. Purpura DP, Penry JK, Tower DB, et al., eds. Experimental Models 54. Kager H, Wadman WJ, Somjen GG. Conditions for the triggering
of Epilepsy. New York, NY: Raven Press; 1972. of spreading depression studied with computer simulations.
38. Jasper HH, Ward AA, Pope A, eds. Basic Mechanisms of the J Neurophysiol. 2002;88:2700–2712.
Epilepsies. Boston, MA: Little, Brown & Company; 1969. 55. Somjen GG, Kager H, Wadman WJ. Computer simulations of neu-
39. Elger CE, Speckmann E-J. Focal interictal epileptiform discharges ron–glia interactions mediated by ion flux. J Comput Neurosci.
(FIED) in the epicortical EEG and their relations to spinal field 2008;25(2):349–365.
potentials in the rat. Electroencephalogr Clin Neurophysiol. 56. Strong AJ, Fabricius M, Boutelle MG, et al. Spreading and synchro-
1980;48:447–460. nous depressions of cortical activity in acutely injured human
40. Elger CE, Speckmann E-J, Caspers H, et al. Focal interictal epilep- brain. Stroke. 2002;33:2738–2743.
tiform discharges in the cortex of the rat: laminar restriction and 57. Caspers H, Speckmann E-J, Lehmenkühler A. Effects of CO2 on
its consequences for activity descending to the spinal cord. In: cortical field potentials in relation to neuronal activity. In:
Klee MR, Lux HD, Speckmann E-J, eds. Physiology and Speckmann E-J, Caspers H, eds. Origin of Cerebral Field Potentials.
Pharmacology of Epileptogenic Phenomena. New York, NY: Raven Stuttgart: Thieme; 1979:151–163.
Press; 1981. 58. Speckmann E-J, Caspers H. The effect of O2 and CO2 tensions in the
41. Gumnit R. DC shifts accompanying seizure activity. In: Remond A, nervous tissue on neuronal activity and DC potentials. In: Remond
ed-in-chief. Handbook of Electroencephalography and Clinical A, ed-in-chief. Handbook of Electroencephalography and Clinical
Neurophysiology. Vol 10. Part A. Amsterdam: Elsevier; 1974:66–77. Neurophysiology. Vol 2. Part C. Amsterdam: Elsevier; 1974:71–89.
CHAPTER

Cellular Substrates of Brain Rhythms*


FLORIN AMZICA AND FERNANDO H. LOPES DA SILVA 3
Q
uasiperiodically recurring waves, henceforth termed 2. The study of the intrinsic properties of cells is mostly carried
oscillations, are, besides evoked potentials, one of the out in vitro or in cultures, preparations that are as far as one can
essential components of the electroencephalogram imagine from the complex reality of the whole brain, both in
(EEG). The curiosity of understanding the cellular counterparts terms of network linkages and physiologic state. At best, these
of EEG waves started almost simultaneously with the discovery preparations would correspond to a deeply comatose brain.
and description of different EEG patterns during the early 1930s 3. The study of the network interactions, however multisite
and 1940s as an effort to relate the global graphic aspect of the they might be, is still based on recordings from spatially dis-
EEG with its more intimate cellular counterparts. In this chap- crete and limited locations. The assumption of continuity
ter, we will discuss how various patterns of oscillatory activity should therefore bear reserve.
develop in cellular brain networks and are further reflected at 4. Anesthesia is often an unavoidable companion of animal
the EEG level. The EEG is essentially an electric phenomenon; studies, and the elimination of the pharmacologic effect is
therefore, we will focus on the ionic currents active during vari- time and again more complicated than a mere subtraction.
ous oscillating patterns but we will also identify the (minimal)
anatomical structures necessary and sufficient for the genesis of
the aforementioned activities. This approach has unavoidably SLOW DELTA RHYTHMS
oversimplifying virtues, while the EEG signal is a complex one.
Grey Walter (1) was the first to assign the term “delta waves” to
This reality will be recreated through the coalescence of various
particular types of slow waves recorded in the EEG of humans.
oscillations that coexist during a given behavioral context.
Although Walter introduced the term delta waves in correspon-
From its very beginning, the EEG was characterized through
dence to pathologic potentials due to cerebral tumors, with
its oscillatory nature. Thus the dominant frequency of an oscil-
time delta activities became more related to sleep and anesthe-
lation became its main descriptor. This trend was further
sia. The IFSECN (2) defines delta waves as waves with a dura-
enforced with the advent of spectral analysis (e.g., Fourier
tion of more than 0.25 seconds (which implies a frequency
transforms) and is still largely used nowadays. Although this
band between 0 and 4 Hz). Thus the delta term is associated
might enclose certain advantages, it also contains numerous
with frequency bands rather than with phenomena generating
pitfalls that prevent the reaching of correct conclusions. This
specific electrographic patterns. There have been various stud-
situation is generated by the fact that a given spectral compo-
ies aiming at disclosing the relationship between cellular activ-
nent might ambiguously result either from the presence in the
ities and EEG (3) and the sources of delta activities (4,5). In the
EEG of oscillations or from the contribution of particular
following it will be emphasized that the 0- to 4-Hz frequency
waveforms, not necessarily rhythmic, to the EEG spectrum.
range reflects more than one phenomenon and that definitions
In the following we will organize this chapter according to
based exclusively on frequency bands may conceal the underly-
the frequency bands traditionally encountered in the EEG
ing mechanism. Studies in the last two decades have unveiled
praxis, however, emphasizing the possible sources of haziness
the electrophysiologic substrates of several distinct activities in
related to the interpretation of EEG signals. Furthermore, as a
the frequency range below 4 Hz during sleep and anesthesia.
section devoted to the understanding of the cellular mecha-
Their interaction within corticothalamic networks yields a
nisms underlying the genesis of the oscillatory behavior of the
complex pattern whose reflection at the EEG level takes the
EEG, the intracellular data constitutes a very strong, although
shape of polymorphic waves.
not absolute, source of knowledge. The reader should, however,
It should also be stressed from the beginning that delta activ-
keep in mind that, when extrapolating from cellular data to
ities cover two EEG phenomena: waves and oscillations, with no
EEG, one should not overlook essential details, such as
clear separations between the two, as far as spectral analysis is
1. Cellular recordings are almost exclusively performed in ani- concerned. The term oscillation designates a repeated variation
mals, whose philogenetical development, behavioral, and of a parameter (e.g., current, voltage) between two extreme val-
structural peculiarities are often neglected. ues, with an optional requirement of regularity of the variation.

*This chapter was initiated and written in the previous editions by the late Dr. Mircea Steriade (1924–2006). The present authors wish to pay tribute to his outstand-
ing work that has much advanced our understanding of neuronal mechanisms underlying EEG rhythms. They hope to continue and preserve the spirit of his
contributions.

33
34 Part I ■ Basic Principles

However, most of the biologic phenomena resulting from large delta waves (as obtained by lesions of the subcortical white mat-
population interactions, such as the EEG, are rarely associated ter, the thalamus, or the mesencephalic reticular [RE] forma-
with clocklike rhythmicity. At the other limit of the definition is tion) have shown a relationship between the firing probability
the wave, as a single variation of a parameter between two and the surface-positive (depth-negative) delta waves, whereas
extreme values. From the above definitions it appears that oscil- the depth-positive waves were associated with a diminution in
lations are made of waves. discharge rates (15). These field–unit relationships led to the
As for now, there are at least two cellular sources of delta assumption that the depth-positive component of delta waves
activities: one originating in the thalamus and the other one in reflects the inhibition of pyramidal-shaped neurons by local-
the cortex. circuit cells, implying maximal firing of inhibitory interneurons
Thalamic delta oscillations have been found in a series of in during the depth-positive delta waves. However, this has not
vitro studies. They revealed that a clocklike oscillation within the been found. It was then suggested that, far from resulting exclu-
delta frequency range is generated by the interplay of two intrin- sively from inhibitory postsynaptic potentials (IPSPs), EEG
sic currents of thalamocortical cells. Whereas most of brain delta waves are rather generated by summation of long-lasting
oscillations are generated by interactions within networks of afterhyperpolarizations produced by a variety of potassium
neurons, but also glial cells, the thalamic delta oscillation is an currents in deeply lying pyramidal neurons (16,17).
intrinsic oscillation depending on two inward currents of thala- The discovery of a novel slow-wave sleep- or anesthesia-related
mocortical cells (7–9). It was shown that thalamocortical neu- oscillation (around, but generally less than 1 Hz) (18), together
rons recorded in vitro display rhythmic bursts of high-frequency with its thorough investigation at the cellular level (see below), set
spikes with an interburst frequency of 1 to 2 Hz. This oscillation the basis for revisiting the cellular basis of delta rhythms. The fre-
results from the interplay between two membrane currents: the quency of the slow oscillation depends on the depth or type of
transient calcium current (It) underlying the low-threshold anesthesia, or the sleep stage: it is mainly between 0.3 and 0.6 Hz
spike (LTS) and a hyperpolarization-activated cation current under urethane anesthesia, between 0.6 and 0.9 Hz under keta-
(Ih). Ih is a noninactivating inward (anomalous) rectifier carried mine-xylazine anesthesia, and between 0.7 and ~1 Hz during nat-
by sodium and potassium. The model proposed for the genesis ural sleep. This oscillation, termed slow oscillation by its authors,
of this clocklike delta oscillation is depicted in Figure 3.1A: when was first described in intracellular recordings from neocortical
the membrane potential hyperpolarizes (more negative than neurons in anesthetized animals and in EEG recordings from
–70 mV), the Ih becomes activated and produces a depolarizing humans (18). It was subsequently found during natural slow-
sag, which in turn will activate the It (deinactivated by the hyper- wave sleep of animals (19) and humans (20–24).
polarized membrane). The ensuing LTS deactivates the Ih but The slow oscillation is generated within the cortex because it
also generates a hyperpolarizing overshoot that will reactivate survives after thalamectomy (25), is absent from the thalamus
the Ih, thus restarting a new oscillatory cycle. of decorticated animals (26), and is present in cortical slices
This oscillation was also found in vivo, in the thalamocorti- (27). At the intracellular level, cortical neurons throughout lay-
cal neurons of the cat, after decortications (10–12). ers II to VI (with physiologically identified thalamic and/or cal-
Thalamocortical neurons from a variety of sensory, motor, asso- losal inputs, and with projections to the thalamus and/or
ciational, and intralaminar thalamic nuclei were able to display homotopic points of the contralateral hemisphere) displayed a
a clocklike delta rhythm either induced by imposed hyperpolar- spontaneous oscillation recurring with periods of 1–1.5 to
izing current pulses (Fig. 3.1B) or spontaneously (Fig. 3.1C). 5 seconds, depending on the anesthetic, and consisting of pro-
Interestingly, the deafferentation achieved through decortica- longed depolarizing and hyperpolarizing components (Fig.
tion, by removing the powerful depolarizing impingement from 3.2A). The long-lasting depolarizations of the slow oscillation
corticothalamic neurons and setting thalamic cells at a more consisted of excitatory postsynaptic potentials (EPSPs), fast
hyperpolarized membrane potential (around –70 mV), was prepotentials (FPPs), and fast IPSPs, reflecting the action of
instrumental in permitting the delta oscillation to emerge. synaptically coupled GABAergic local-circuit cortical cells (18).
However, in intact preparations, with functional corticothalamic Data also demonstrated the contribution of both NMDA-
loops, the regular thalamic delta oscillation was absent or largely mediated synaptic excitatory events and a voltage-dependent
prevented by the ongoing cortical activity (13). At the cellular persistent sodium current, INa(p), to the depolarizing component
level, the occurrence of the Ih was blocked during the depolariz- of the slow oscillation. On the other hand, the long-lasting
ing phases of the slow oscillation ( 1 Hz; see below), due to the hyperpolarization, interrupting the depolarizing events, is asso-
interference of synaptic currents and the ensuing decreased ciated with network disfacilitation in the cortex. This is sup-
membrane resistance with the intrinsically generated Ih. These ported by the fact that the membrane input resistance is highest
more recent findings raised concerns as to the emergence of this during the long-lasting hyperpolarizing component of the slow
intrinsically generated thalamic rhythm at the level of the EEG. oscillation (28). The disfacilitation is probably achieved by a
The existence of cortical delta oscillations was suggested on progressive depletion of the extracellular calcium ions during
the basis that delta waves, at 1 to 2 Hz, survive in the EEG of the depolarizing phase of the slow oscillation (29) (Fig. 3.2B–C).
athalamic cats (14). Whether such procedures generate a phys- All major cellular classes in the cerebral cortex, as identified
iologic or a pathologic pattern remains an open question; by electrophysiologic characteristics and intracellular staining,
nonetheless, they create a deafferentation of the cortex. display the slow oscillation: regular spiking and intrinsically
Extracellular recordings of cortical activity during pathologic bursting cells, as well as local-circuit inhibitory basket cells
Figure 3.1 Delta oscillations in thalamocortical cells result from the interplay between two intrinsic currents, Ih and It. A: Proposed model for interac-
tion between these intrinsic currents. Activation of the low-threshold calcium current, It, depolarizes the membrane toward threshold for a burst of
sodium-dependent fast action potentials. The depolarization inactivates the portion of Ih that was active immediately before the calcium spike.
Repolarization of the membrane due to It inactivation is followed by a hyperpolarizing overshoot, due to the reduced depolarizing effect of Ih. The hyper-
polarization in turn deinactivates It and activates Ih, which depolarizes the membrane toward threshold for another calcium spike. (Modified from
McCormick DA, Pape HC. Properties of a hyperpolarization-activated cation current and its role in rhythmic oscillation in thalamic relay neurones.
J Physiol. 1990;431:291–318.) B: Delta oscillation of cat ventrolateral thalamocortical cell triggered by hyperpolarizing current pulses (0.7 nA in 1, 1 nA
in 2, 1.1 nA in 3, and 1.2 nA in 4). Note increasing number of cycles at a frequency of 1.6 Hz. C: Latero-posterior (LP) thalamocortical cell after decor-
tication of areas projecting to LP nucleus. The cell oscillated spontaneously at 1.7 Hz. A 0.5-nA depolarizing current (between arrows) prevented the
oscillation, and its removal set the cell back in the oscillatory mode. Three cycles after removal of depolarizing current in 1 are expanded in 2 to show
high-frequency bursts crowning the low-threshold calcium spike. In this and following figures, polarity of EEG and field potentials is as for intracellular
recordings (positivity up). (Modified from Steriade M, Curró Dossi R, Nuñez A. Network modulation of a slow intrinsic oscillation of cat thalamocorti-
cal neurons implicated in sleep delta waves: cortically induced synchronization and brainstem cholinergic suppression. J Neurosci. 1991;11:3200–3217.)

35
36 Part I ■ Basic Principles

Figure 3.2 A: Pyramidal cell from the somatosensory cortex (area 3b) during the slow ( 1 Hz) oscillation. The left panel
shows intracellular recordings and simultaneously recorded EEG in the vicinity (~ 1 mm) of the cell. The EEG was recorded
by means of coaxial electrodes located on the surface and at a depth of approximately 0.6 mm. The cell oscillated at 0.9 Hz
with depolarizing phases corresponding to depth-EEG negative (surface-positive) potentials. The right panel shows the cor-
responding cell stained with Neurobiotin (calibration bar in mm). (Modified from Contreras D, Steriade M. Cellular basis
of EEG slow rhythms: a study of dynamic corticothalamic relationships. J Neurosci. 1995;15:604–622.) B: Fluctuations dur-
ing the slow oscillation. Relationships between intracellular membrane potential, extracellular calcium [Ca] out, and field
potential. Periodic neuronal depolarizations triggering action potentials were interrupted by periods (300–500 msec) of
hyperpolarization and silenced synaptic activity. [Ca]out dropped by about 0.25 mM during the depolarizing phase reach-
ing a minimum just before the onset of the hyperpolarization. Then, [Ca] out rose back until the beginning of the next cycle.
C: Thirty cycles were averaged (spikes from the neuronal signal were clipped) after being extracted around the onset of
the neuronal depolarization. The vertical dotted lines tentatively indicate the boundaries of the two phases of the slow
oscillation (B–C modified from Massimini M, Amzica F. Extracellular calcium fluctuations and intracellular potentials in
the cortex during the slow sleep oscillation. J Neurophysiol. 2001;85:1346–1350.).
Chapter 3 ■ Cellular Substrates of Brain Rhythms 37

Figure 3.3 Effect of lidocaine inactivation on synchrony between intracellular activities and FPs. A: Double intracellular
recording in the anterior and posterior parts of the suprasylvian gyrus (see scheme). Synchrony was present between all
three recording sites. Intracortical injection of lidocaine (40 L, 20% ; bottom) inactivated the electrical activity close to
the cannula in the middle part of the gyms, disrupted the synchrony between pools of neurons, and between individual
neurons. B: Sequential correlation analyses of intracellular and field potential data. Two-minute periods were analyzed
with the sequential correlation method. (Modified from Amzica F, Steriade M. Disconnection of intracortical synaptic link-
ages disrupts synchronization of a slow oscillation. J Neurosci. 1995;15:4658–4677.)

(18,30,31). All these neuronal types exhibit similar relation- Such cortical coherence is expected to equally involve the
ships with the EEG components of the slow oscillation: during thalamus. Indeed, RE thalamic cells exhibit similar and time-
the depth-positive EEG wave cortical neurons are hyperpolar- locked variations of their membrane potential, with prolonged
ized, whereas during the sharp depth-negative EEG deflection depolarizations interrupted by prolonged hyperpolarizations
cortical neurons are depolarized (Fig. 3.2). The spectacular (34) in reflection of the excitatory cortico-RE projections. The
coherence between all types of cortical neurons and EEG wave- depolarizing component in RE neurons is transmitted to thala-
forms raised the question of the underlying synchronizing mocortical neurons, via GABAergic projections, where it trig-
mechanisms. Dual intracellular recordings in vivo revealed that gers rhythmic IPSPs, leading to rebound spike bursts (Fig. 3.4),
intracellular potentials were highly synchronized among neu- which in turn will generate phasic excitations in the cortex,
rons (31,32). This synchronization, displaying time lags as a shaping the steady depolarization of cortical neurons.
function of the distance between cortical areas (33), mainly The better comprehension of the mechanisms determining
relies on the integrity of intracortical synaptic linkages. the pacing of the slow oscillations and the switch between
Although the coherence was impaired after local blockage of depolarizing and hyperpolarizing phases came from experi-
axonal transmission through local injections of lidocaine (Fig. ments considering the possible dialogue between neurons and
3.3), the slow oscillation survived at both locations and con- glial cells. Dual simultaneous intracellular recordings from neu-
served a rudiment of synchronization suggesting that, besides rons and adjacent glial cells were performed during sleeplike
direct intracortical linkages, other projections, possibly cortico- patterns produced by ketamine-xylazine anesthesia, exploring
thalamo-cortical, as well as networks of gap junctions (see the possibility that glia may not only passively reflect, but also
below) might contribute to the synchronization of the slow influence the state of neuronal networks (35–37). The behavior
oscillation. of simultaneously recorded neurons and glia is illustrated in
38 Part I ■ Basic Principles

Figure 3.4 The slow oscillation (~ 0.9 Hz) in dual simultaneous intracellular recordings from a regular-spiking cell
in cortical area 4 and a thalamocortical cell in the ventrolateral nucleus. Cat under ketamine-xylazine anesthesia.
Arrow in A points to a low-threshold spike burst. An expanded cycle is shown in B. Notes: (a) Depth-positive
(upward) EEG waves are associated with hyperpolarization of cortical and thalamic cells, whereas the sharp depth-
negatives are associated with depolarization and action potentials in cortical cell, while the thalamic neuron displays
a rebound spike burst with a delay of 150–200 msec; (b) brief sequence of EEG spindles after depth-negative (surface-
positive) sharp deflection; and (c) fast depolarizing waves (40–50 Hz) in cortical neuron during the sustained depo-
larization. (Unpublished data by M Steriade and D Contreras.)

Figure 3.5. During spontaneously occurring slow oscillations, glial cells might control the pace of the oscillation through
the onset of the glial depolarization did, in the vast majority of changes in the concentration of the extracellular potassium (38).
the cases, follow the onset of the neuronal depolarization with The overall synchronization of the slow oscillation in the
an average time lag of around 90 msec (Fig. 3.5). This time lag cortex is also assisted by glial cells, which are imbedded in a gap
is, however, longer than the one obtained from pairs of neurons junction-based network, through the phenomenon of spatial
(around 10 ms in Ref. 33). The glial depolarization reflects with buffering (41–43). Spatial buffering evens local increases of
virtually no delay the potassium uptake (38). As potassium is extracellular potassium by transferring it from the extracellular
also continuously recaptured by neurons through Na /K -ATP milieu to neighboring glial cells, then through gap junctions,
pumps, the actual onset of the glial depolarization may thus and along the potassium concentration gradient, at more dis-
mark the moment where the increase of extracellular potassium tant sites with lower concentrations of potassium, where it is
concentration exceeds the capacity of the neuronal pumps. This again expelled in the extracellular space. This pathway is pre-
behavior suggests that glial cells are essential for maintaining ferred to the direct one through the extracellular space because
extracellular potassium within controlled values. of the tortuosity of the latter (reviewed in Ref. 44). The rela-
Toward the end of the depolarizing phase, the glial membrane tionship between intracellular glial potentials and extracellular
begins to repolarize before the neuronal membranes (Fig. 3.5) potassium (38) shows little accumulation of potassium in the
(37). Interestingly, this observation may expose the mechanism extracellular space ( 1.1 mM/cycle), and this causes a minimal
ruling the onset of the hyperpolarizing phase of the slow oscilla- imbalance between the extra- and intracellular potassium con-
tion. It was initially proposed that this hyperpolarization relies centrations. Given that normally glial cells deal with low
either on active GABA inhibition or on the activation of slow amounts of potassium but have a high propensity to uptake it
calcium-dependent potassium currents. In the first case, the (43,45,48), it is likely that spatial buffering would occur at a
release of GABA by neurons would either depolarize glial cells reduced spatial scale, thus contributing to an uniform distribu-
through direct action on GABAA receptors (39,40) or indirectly tion of the potassium around the membrane of neurons (38).
through the potassium output resulting from the neuronal acti- The local spatial buffering during slow oscillations might
vation of neuronal GABAB receptors. The alternative implication play two roles: (i) it contributes to the steady depolarization of
of calcium-dependent potassium currents should be discarded neurons during the depolarizing phase of the slow oscillation
because of the fact that the extracellular potassium starts to drop in a Nernst-related manner and (ii) it modulates the neuronal
before the onset of the hyperpolarizing phase. In addition, the excitability. The latter mechanism could favor the synaptic
glial membrane potential returns to control value at the end of interaction within cortical networks at the onset of the depo-
each of the recurring oscillatory cycles, leading to the assumption larizing phase of the slow oscillation, but it could equally
Chapter 3 ■ Cellular Substrates of Brain Rhythms 39

Figure 3.5 Simultaneous recording


of a neuron–glia pair in the cortex of
the cat during slow oscillatory pat-
terns. A: Relationship between neu-
ronal and glial depolarizations on
the one hand, and between neu-
ronal hyperpolarization and glial
repolarizations on the other hand
(note enlargement within the box).
The glial impalement occurred dur-
ing this period of the recording and
is depicted as a brisk drop of the
potential from the extracellular
(0 mV) to the intracellular (–90 mV)
level. B: Histogram of membrane
potential variations in the neuron
(in gray) and glial cell (in black). C:
Typical time relationship between
the onset and offset moments of the
two phases of a slow oscillatory
cycle. Note that the onset of the
depolarizing phase starts in the neu-
ron, but that the end of this period is
anticipated in the glia. (Modified
from Amzica F, Massimini M. Glial
and neuronal interactions during
slow wave and paroxysmal activities
in the neocortex. Cereb Cortex.
2002;12:1101–1113.)

induce a gradual disfacilitation as the extracellular potassium decades (see the excellent review in Ref. 51) during which
reaches higher values toward the top of the glial depolariza- researchers struggled with the inherent shape variability intro-
tion. The amplitude of the potassium variations during the duced by various bipolar electrode configurations, reference sys-
slow sleep oscillation ranges between 1 and 2 mM, which, tems, and filter settings. These technical difficulties, together
when added to the physiologic values of resting concentration with the relative vagueness of the definition, often led to discor-
(~3 mM), may assist cortical neurons in oscillating between dant conclusions and to difficult interpretations. The cellular
hyper- and hypoexcitability (47–49). This hypothesis was con- mechanisms of the K-complex were only recently described (52)
firmed after testing the excitability of the neurons during the and disclosed that the K-complex is a recurrent event with the
various phases of the slow oscillation cycle with cortical elec- rhythmicity of the slow oscillation (less than but close to 1 Hz)
tric stimuli (37). The results demonstrate that the neuronal and that, through its shape, it is one of the main contributors to
excitability is maximal toward the beginning of the depolariz- the delta spectrum (Fig. 3.6) (21,35).
ing cycle and almost zero toward the end of the depolarizing The rhythmicity of K-complexes during the onset of sleep is
phase of the slow oscillation. less obvious due to the lesser organization of the slow oscilla-
tion. This corroborates with the fact that bipolar recordings are
THE SLOW OSCILLATION often the only ones allowing the detection of K-complexes in
AND THE K-COMPLEX the initial part of slow-wave sleep. However, with the deepening
of sleep, the slow oscillation becomes more regular and faster
One of the functional correlates of the slow cortical oscillation is (the frequency approaches 1 Hz). During late stages of sleep
the fact that, at the EEG level, each sequence of most of the K-complexes may be confounded with delta waves.
depolarizing–hyperpolarizing episodes within an oscillatory The frequency shift (usually from 0.6 to 0.9 Hz) of the slow
cycle corresponds to a waveform called K-complex. Although oscillation, as well as the shape modification of the K-complex
the phenomenon was first described in 1938 as a landmark of (Fig. 3.7), accompanying the deepening of sleep can be
slow-wave sleep (50), its study remained at the EEG level for explained by taking into account the intrinsic properties of the
40 Part I ■ Basic Principles

Figure 3.6 K-complex in human sleep. Scalp monopolar recordings with respect to the contralateral ear (see figurine).
A: Short episode from a stage 3 period of sleep. The two arrows point to a K-complex (KC) followed by a spindle sequence
( ) and to a KC in isolation. The two K-complexes are embedded into a slow oscillation at about 0.6 Hz. Note the syn-
chrony of K-complexes in all recorded sites and the diminution of their amplitudes in the occipital area. B: Average of
50 K-complexes aligned on the positive peak (in this figure positivity is downwards) of the upper channel
(vertical dotted line). C: Power spectrum of the C3-lead for a period of 80 seconds of stable stage 3 activity
containing the period depicted in A. The three frequency bands (S, , and ; further illustrated in D) are rep-
resented in the power spectrum. Moreover, the slow (S) activity displays a high peak, distinct from the onset of delta
( ) activity (see inset). D: Frequency decomposition of the C3-lead (upper trace) into three frequency bands:
slow (S, 0–1 Hz), delta ( , 1–4 Hz), and sigma ( , 12–15 Hz). It is shown that the K-complex results from a
combination of slow and delta waves. (Modified from Amzica F, Steriade M. The K-complex: its slow ( 1-Hz) rhythmicity
and relation to delta waves. Neurology. 1997;49:952–959.)

neuronal membrane. At sleep onset, neurons in the midbrain Due to the controversial criteria for shape recognition, the
reticular formation and mesopontine cholinergic nuclei dimi- study of K-complexes has relied mostly on evoked K-complexes
nish their firing rate (53,54), thus removing a steady excitatory because stimulation generally produces stereotyped responses.
drive from thalamocortical neurons and allowing the Although of certain experimental value, the evoked K-
membrane potential of cortical neurons to reach more hyper- complexes have set a bias in the understanding of the underly-
polarized levels. The progressive hyperpolarization of thalamo- ing mechanisms because it was forgotten that the K-complex is
cortical neurons is a constant correlate of sleep deepening (55) mainly a spontaneous phenomenon (50). The comparison at
and induces a gradual reduction of the duration of the depolar- the cellular level between spontaneous and evoked K-
izing phase of the slow cortical oscillation together with the complexes shows, in agreement with the original observation of
acceleration of the rhythm. The former action contributes Davis et al. (56), that the latter appears as a secondary discharge.
mostly to the shape of the field potentials, while the second Both spontaneous and evoked K-complexes display similar
determines the oscillatory frequency. depth profiles and current source density (CSD) distributions
Chapter 3 ■ Cellular Substrates of Brain Rhythms 41

Figure 3.7 Rhythmic KCs in human EEG during natural slow-wave sleep. A: Four leads recorded from the
two hemispheres during stage 2 sleep show quasirhythmic (around 0.5 Hz) KCs. The expanded insets display
a simple K-complex (KC, left) and a K-complex followed by a spindle (right). Stage 3–4 EEG (below) is char-
acterized by a more regular oscillation of the K-complexes (0.7 Hz). Asterisks mark the most obvious
K-complexes in order to suggest their rhythmicity. B: Averaged K-complexes (n 200) from stages 2 (left)
and 3–4 (right). The gray surface around the averaged K-complexes covers the standard deviation. The low-
est trace (dotted line) results from the filtering of the average K-complex in the delta band (1–4 Hz). C: Power
spectra of 2-minute epochs containing the ones displayed above. Note a principal peak at 0.5 Hz for the stage
2 episode (left), surrounded by other lower peaks as evidence for distributed rhythmicity. At variance, deep
sleep shows a dominant peak at 0.7 Hz. The two FFT graphs are scaled with the same ordinate. (Modified
from Amzica F, Steriade M. The K-complex: its slow ( 1-Hz) rhythmicity and relation to delta waves.
Neurology. 1997;49:952–959.)
42 Part I ■ Basic Principles

(52), further supporting their common origin and generating lowest end of the theta frequency range that presented larger
mechanisms. power spectrum during rapid eye movement (REM) sleep than
The complex electrographic pattern of slow-wave sleep during other states and suggested that this oscillation may be
results from the coalescence of the slow oscillation with sleep considered the counterpart of the hippocampal theta of mam-
spindles (see below) and, possibly, thalamic delta oscillations malian REM sleep. Tesche and Karhu (67) recorded MEG sig-
(57). The weight of each of these major components is dynam- nals in human subjects and reconstructed from these signals the
ically modulated during sleep by synaptic coupling, local- corresponding hippocampal sources of theta rhythmic activity.
circuit configurations, and the general behavioral state of the They reported that during the presentation of a memory task
network. The discussion of the functional significance of the the duration of theta bursts increased with memory load and
K-complex is determined by the role of the slow sleep oscilla- suggested a relation between “stimulus-locked hippocampal
tion of which the K-complex is the electrographic reflection. theta” and the processing of working memory. Using a whole-
Through its rhythmic occurrence at a frequency that is lower head 275- channel MEG system in normal subjects, Cornwell et
than the one of other sleep rhythms (spindles and delta), and al. (68) reported hippocampal and parahippocampal theta
due to its wide synchronization at the cortical level, the sharp oscillations during a spatial navigation task (virtual reality
onset of the K-complex provides a synchronous input to thala- Morris water maze). They carried out source analyses by means
mic neurons, thus triggering and/or grouping sequences of of which they were able to show larger theta activity in the left
spindles or delta oscillations. anterior hippocampus and parahippocampal cortices during
goal-directed navigation relative to aimless movements. Thus,
THETA RHYTHMS similar to other species (see below), the human theta may rep-
resent a dynamic state emerging from hippocampal networks
Theta waves are usually defined as occurring within the 4- to engaged in spatial navigation and in memory processes.
7-Hz frequency range. The normal theta activity should not be
confused with pathologic theta waves, described as a slowing Limbic Theta Rhythms in Animals
down of alpha activity, expressed during cerebral blood flow Stewart and Fox (69), working on urethane-anesthetized pri-
reduction (58) or metabolic encephalopathies (59). Rhythmic mates, recorded hippocampal theta activity similar to that of rats
activities in the theta frequency range are conspicuous in limbic but at a slightly higher frequency and appearing as relatively
areas in various animal species as extensively reviewed by short bursts. Arnolds et al. (63) made a comparative study of
Buzsáki (60). hippocampal theta rhythms in cat, dog, and human and showed
that all three displayed the same type of qualitative changes in
Limbic Theta Rhythms in Humans spectral parameters (peak frequency, peak amplitude, and
The presence of theta rhythms was first denied in humans rhythmicity) in relation to behavioral tasks involving motor
(61,62), but they were later recorded in the hippocampus of activities. Particularly in cat, remarkable relationships between
epileptic patients with indwelling electrodes (63–65) or by way theta spectral properties and vestibular stimulation (e.g., body
of mesio-temporal corticography with foramen ovale elec- acceleration) and/or eye movements were put in evidence. Theta
trodes (66), and also using magnetoencephalography (MEG) in rhythms appear conspicuously in rodents such as rabbits (first
normal subjects (67,68). In all these studies human hippocam- description of the rhythm was made by Green and Arduini (70))
pal theta rhythm was studied under specific behavioral condi- and rats in relation with sensory processing and different types
tions. In the study of Arnolds et al. (69) the hippocampal of movements (71) and have been recorded in several structures
activity was recorded while the subject was freely moving and of the cortical limbic system (72,73). Depending on species and
performing a cognitive task; the frequency and rhythmicity of conditions, these rhythmic may extend from 3–4 to 10 Hz,
the hippocampal theta component while the subject was writ- which is somehow larger than the conventional range of the
ing was consistently higher than while sitting or walking. In a EEG theta rhythm (4 to 7 Hz). This is the reason why these lim-
word association task the amplitude, frequency, and rhythmic- bic theta oscillations are also named “rhythmic slow activity
ity showed a significant rise during the period of silent mental (RSA)” in animal neurophysiology to avoid confusion with the
activity immediately following a verbal cue to which the subject classic human EEG theta frequency band. The two designations
was requested to give a verbal answer. Ekstrom et al. (65) found are commonly used in the literature.
evidence for movement-related theta oscillations in human In the rat, theta oscillations are most regular in frequency
hippocampus and suggested that both cortical and hippocam- and present the largest amplitude in the stratum lacunosum-
pal oscillations play a role in attention and sensorimotor inte- moleculare of the hippocampal CA1 region but are also found
gration. Kahana et al. (64) investigated hippocampal EEG in the dentate gyrus and the CA3 region. In addition to the hip-
activity using subdural recordings from epileptic patients dur- pocampal formation, theta oscillations have been observed in
ing spatial navigation in computer-generated mazes and found the subiculum, entorhinal cortex, perirhinal cortex, amygdala,
that theta rhythmic activity occurred more frequently when the and cingulate cortex (74). Although these areas are capable of
subject was confronted with solving complex mazes. In addi- displaying theta oscillations that can be recorded extracellularly,
tion, theta oscillations were more evident during recall trials they are not able to generate theta activity on their own, that is,
than during learning trials. Bódizs et al. (66) recorded in the isolated from the rest of the brain, unless manipulated by intra-
human hippocampus a rhythmic activity with frequency at the cellular current injection or by the addition of pharmacologic
Chapter 3 ■ Cellular Substrates of Brain Rhythms 43

substances. In this respect it is interesting to mention that neu- frequency, can thus cause rhythmic IPSPs on their target pyrami-
rons of several limbic structures have intrinsic membrane prop- dal cells. Consequently somatic outward currents at the level of
erties that might facilitate their entrainment in theta the CA1 pyramidal layer can contribute to the theta extracellular
oscillations as shown in vitro. This was demonstrated for some field (81,90). Further cholinergic agonists applied to hippocampal
types of CA1 interneurons depolarized near spike threshold by slices cause rhythmic depolarizations of lacunosum-moleculare
current injection that generated rhythmic firing at about 7 Hz interneurons at the theta frequency, which are blocked by atropine
(75) and also for neurons of the perirhinal cortex that can dis- (91). This indicates that muscarinic induction of theta-frequency
play oscillations around 8 to 9 Hz when depolarized at thresh- membrane potential oscillations in lacunosum moleculare
old levels (76). These intrinsic properties could facilitate the interneurons may contribute to the generation of rhythmic inhi-
entrainment of these neurons in theta frequency oscillations. bition that paces intrinsically generated theta activity in CA1
pyramidal cells. In addition, the CA3 can also contribute to the
Theta Rhythm Generation generation of theta field potentials, since the recurrent network of
Since the early years of this research line several studies were CA3 pyramidal cells, and possibly hilar mossy cells, may form an
carried out with the aim of finding a system that may act as intrahippocampal oscillator that may contribute to the atropine-
theta “pacemaker.” The search for a theta pacemaker has led to sensitive component (73).
controversial results: first it was claimed that the medial A classic view (92) is that hippocampal CA1 extracellular
septo/diagonal band-hippocampal cholinergic system, driven field during theta can be accounted for by a two-dipole model.
by the brainstem reticular core, is the “pacemaker” of theta According to this view the CA1 pyramidal cells receive rhyth-
(77,78). The medial septum provides cholinergic and mic inputs in the theta frequency range from other sources that
GABAergic input to the hippocampus. While cholinergic neu- consist of atropine-sensitive and atropine-resistant inputs. The
rons innervate both principal cells and interneurons, former would drive the somata and the latter the distal den-
GABAergic projection neurons from the medial septum make drites. The atropine-sensitive theta rhythm would be mainly
synapses on hippocampal interneurons, including parvalbu- caused by Cl--mediated IPSPs on pyramidal cells (93,94).
min-positive basket cells (79). Further intrinsic voltage-dependent membrane potential oscil-
More recent investigations revealed that hippocampal lations may modulate the response to a theta frequency driving,
interneurons with long-range projections also innervate the although hippocampal pyramidal neurons do not oscillate in
cells of origin in the medial septum. Further, the supramammil- isolation (60). They show, however, in vitro, resonant properties
lary nucleus of the hypothalamus strongly connected to the in the theta frequency range that are determined by the inter-
medial septum, and to extended networks of the brainstem and play between two main ionic currents with different kinetics,
diencephalon, has also been proposed to play a role in pace- the Ih and Im currents. The former is a hyperpolarization-
making and modulating hippocampal theta (80). activated cation current and the latter is a muscarinic K cur-
The fact that theta rhythms survive after the blockade of rent. In contrast, neurons of the entorhinal cortex layer II show
cholinergic muscarinic inputs from the septal area using subthreshold oscillations at the theta frequency that appear to
atropine brought to the fore the existence of an additional non- result from the interplay of the Ih and the depolarization-
cholinergic source of theta activity in the entorhinal cortex (81). activated persistent Na current (60).
Indeed, a dipole of theta activity was found also in the entorhi- More recent studies (88), however, show that hippocampal
nal cortex with two amplitude maxima, one superficial in layers theta is associated with multiple current sinks and sources with
I–II and the other in layer III (82–85). We may conclude that a wide range of phase relationships between the firing of
the classic view of a septal/diagonal band pacemaker impinging pyramidal cells and of interneurons to the theta recorded
rhythmic activity on the pyramidal cells of the hippocampus, extracellularly from different hippocampal layers.
within the theta frequency band, is too simplistic.
Many investigations analyzed the characteristics of the two Intrahippocampal Inhibitory System
types of hippocampal theta: one atropine-sensitive and the other and Theta Generation
atropine-resistant (86). Removal of the entorhinal cortex makes The phase relationship between spike firing and the polarity of
hippocampal theta atropine-sensitive, so that it may be con- the extracellular field depends on the relative strengths of the
cluded that the atropine-resistant theta originates from the dendritic depolarization and somatic inhibition. The insightful
entorhinal inputs to the hippocampus (81,87). These inputs are review of Somogyi and Klausberger (95) summarizes the com-
glutamatergic and it was shown that NMDA synapses on the dis- plex interrelationship between the firing of different popula-
tal apical dendrites of CA1 pyramidal neurons are important tions of inhibitory interneurons and pyramidal cells during
sources of theta-generating inputs (73). Which pathways are theta oscillations (Fig. 3.8). During theta oscillations several
responsible for the atropine-sensitive theta component is still types of interneurons exhibited considerable variability in the
unclear despite many investigations using sophisticated methods relation between firing probability and the mean theta phase
such as high-density 96-site silicon probes enabling recordings recorded extracellularly. Figure 3.8 shows the relationships of
simultaneously from the dentate gyrus, CA3 and CA1 areas (88). the firing of different kinds of interneurons not only with theta
Cholinergic inputs can cause excitation of interneurons that waves, but also with high-frequency ripples and oscillations in
may be responsible for theta rhythmic discharges of hippocampal the gamma frequency range. Here we limit ourselves to a
interneurons (79,89). These interneurons, discharging at the theta description of the former. Axo-axonic cells displayed the highest
44 Part I ■ Basic Principles

Figure 3.8 Spatiotemporal interaction between pyramidal cells and several classes of interneurons during network oscillations,
shown as a schematic summary of the main synaptic connections of pyramidal cells (P), parvalbumine (PV)-positive-expressing
basket, axo-axonic, bistratified, oriens-lacunosum moleculare (O-LM), and three classes of cholecystokinin (CCK)-expressing
interneurons. The firing probability histograms show that interneurons innervating different domains of pyramidal cells fire with
distinct temporal patterns during theta and ripple oscillations, and their spike timing is coupled to field gamma oscillations to dif-
fering degrees. The same somatic and dendritic domains receive differentially timed input from several types of GABAergic
interneuron. ACh, acetylcholine. (Modified from Klausberger T, Somogyi P. Neuronal diversity and temporal dynamics: the unity
of hippocampal circuit operations. Science. 2008;321:53–57.)

firing rate around the positive peak of extracellular theta field constitute an important mechanism for the occurrence of
recorded at the level of the pyramidal layer, that is, when the epileptogenic activity in the hippocampus (97). Fuentealba et al.
average pyramidal cells are most hyperpolarized. Parvalbumin (98) also described more recently a subpopulation of
immunopositive basket cells fired during the descending phase GABAergic interneurons expressing enkephalin that innervate
of the theta oscillations were recorded at the same level. parvalbumin-positive interneurons and thus may contribute to
Bistratified cells innervating strati radiatum and oriens were the organization of rhythmic activities in CA1 area.
fired during the trough of the extracellular theta in the pyram- The peak of the intracellular theta recorded in the dendrites
idal cell layer. The oriens-lacunosum moleculare cells terminat- of pyramidal cells slightly lags the peak of the extracellular theta
ing on the most distal dendrites in conjunction with the in the pyramidal cell layer (99).
entorhinal input were also fired during the trough of the theta An important new finding (100) that further emphasizes the
field potential. role of interneurons in the generation of hippocampal theta
In addition it should be noted that some inhibitory interneu- was obtained using transgenic mice where the fast synaptic
rons make synapses with other inhibitory interneurons such as GABAergic inhibition was removed from parvalbumin-positive
described by Banks et al. (96) who demonstrated an interaction interneurons. In CA1, these interneurons comprise bistratified
between two groups of CA1 interneurons defined according to and possibly oriens-lacunosum moleculare cells that target
the kinetics of the corresponding IPSCs: GABAA generating dendrites, and basket cells that target somata and axo-axonic
IPSPs with slow kinetics and GABAA with fast kinetics that may cells (101). Each of the perisomatic inhibitory parvalbumin-
contribute to theta and gamma rhythms, respectively. Most positive cells innervates more than 1000 pyramidal cells (102)
interesting is the fact that incoming stimuli in stratum lacuno- and causes IPSPs in the pyramidal cell layer. Through synaptic
sum moleculare do not only inhibit pyramidal cells, by activat- GABAA receptors, parvalbumin-positive basket cells recipro-
ing “GABA-slow” IPSCs in these cells, but simultaneously inhibit cally inhibit each other and receive inhibition from the medial
“GABA-fast” interneurons. The authors hypothesize that this septum and other local interneurons (103). Wulff et al. (100)
inhibitory–inhibitory interaction contributes to nested found that this interneuron-specific ablation of the inhibitory
theta/gamma rhythms in the hippocampus. In Chapter 4, we septohippocampal projection can severely impair theta
describe that an alteration of this inhibitory interaction may rhythm in the hippocampus. This study implies that the
Chapter 3 ■ Cellular Substrates of Brain Rhythms 45

intrahippocampal systems of inhibition of parvalbumin- Alpha Waves: Cortical Dipolar Sources


positive cells contribute to theta generation. and Thalamic Influences
From all these data it emerges that to understand theta gen- In a series of experimental studies performed on dogs, Lopes da
eration in the hippocampus and associated areas it is necessary Silva and colleagues established that alpha rhythms are the
to integrate single neuronal data with knowledge of local net- undertaking of the cerebral cortex, mostly in the visual areas,
works, their local synaptic wiring, and the corresponding although they can also be recorded in the visual thalamus (lat-
dynamical properties. In other words, it is necessary to make eral geniculate and pulvinar nuclei) (113). In the visual cortex,
explicit computational models (104) including networks of dif- alpha waves are generated by an equivalent dipole layer cen-
ferent types of interneurons as discussed in Chapter 4, as well as tered at the level of the somata and basal dendrites of pyrami-
pyramidal neurons and granule cells; the latter should be simu- dal neurons in layers IV and V (Fig. 3.9A) (114). Furthermore,
lated using compartmental models to account for the spatial the coherence of alpha waves within the visual cortex was only
distribution of specific inputs to dendrites and somata. partially dependent on thalamic sources measured in the same
animal (Fig. 3.9B) (114,115) leading to the conclusion that hor-
ALPHA RHYTHMS izontal intracortical linkages are essential to the spread of alpha
activities, with only moderate implication of the thalamus.
This section summarizes the few elements presently known The cortical generator of alpha rhythms was confirmed and
about the mechanism(s) and origin(s) of alpha rhythm occur- extended by more recent studies performed in awake monkeys
ring in the frequency range of 8 to 13 Hz, mainly around 10 Hz. using fine microelectrode arrays implanted across the visual
Its initial description (105) also marked the beginning of EEG. cortical areas V2, V4, and the inferior temporal cortex by
A more recent and exhaustive review of different EEG patterns Bollimunta et al. (116) (Fig. 3.10). CSD analysis was combined
of alpha rhythms in the waking adult can be found in with CSD–multiple unit activity coherence to identify intracor-
Niedermeyer (106). Besides the classic alpha rhythm of the tical alpha current generators. In V2 and V4, alpha current gen-
visual cortex, there are rhythmic activities in the same fre- erators were found in all layers, with the infragranular neurons
quency range that can be recorded from the somatosensory cor- (layer V) acting as primary local generator. In contrast, in the
tex (called the mu rhythm) and the temporal cortex (called the inferior temporal cortex, alpha current generators were found
tau rhythm). Occipital alpha waves usually occur during only in supragranular and infragranular layers, with the supra-
reduced visual attention, while the mu rhythms of the somato- granular generator acting as primary local generator. Granger
motor cortex occur as the subject is in a state of muscular relax- causality analysis revealed that the influence of the infragranu-
ation. In this way, the presence of dominant activity within the lar on the supragranular cells has both a direct component and
alpha frequency range has been interpreted, since Adrian and a component mediated by the granular layer cells. Most inter-
Matthews (107), as indicating an “idling state” of the brain esting, the analysis of the coherence between CSD sinks and
although paradoxical findings with alpha enhancement by local multiunit activity in infragranular and granular layers
attention tasks (108,109) were reported. These observations are showed that these cells are depolarized and tend to generate
now well understood since a number of observations, both of action potentials during the local current sinks; however, at the
visual alpha and somatomotor mu rhythms, have shown that supragranular layer the coherence between the CSD sinks and
enhancement and attenuation of these rhythmic activities can the multiunit activity was poor, suggesting that the cells of this
be recorded simultaneously at different sites and successively layer are inhibited due to inhibitory inputs from layer V
depending on a given task. This can be explained taking into interneurons that innervate superficial pyramidal cells.
consideration the interaction between different thalamocortical
modules as explained in Chapter 45 (event-related alpha desyn-
chronization [ERD]/event-related alpha synchronization [ERS] Alpha Waves: Synaptic Processes Studied In Vitro
phenomena). In this respect, the experimental findings of Rihs Furthermore the nature of the synaptic processes underlying
et al. (110,111) showing enhancement of occipital alpha associ- alpha frequency oscillations in the cortex was studied in in vitro
ated with active suppression of unattended positions during a preparations, although it is always a matter of discussion
visual spatial orienting task provides further evidence for the whether in vitro preparations submitted to pharmacologic
facilitative role of alpha-power decreases (ERD) versus the manipulations and displaying this kind of oscillations provide
inhibitory role of alpha-power enhancement (ERS) of atten- reliable models of the situation in the intact brain, where the
tional processes. With few exceptions, the study of the cellular neurons are in different physiologic conditions. Nonetheless,
behavior underlying alpha oscillations was prevented mainly by Flint and Connors (117) were able to obtain oscillations in the
the fact that alpha rhythms appear only during wakefulness and alpha frequency range when slices of rat somatosensory cortex
no valid anesthesia model was established. The fact that the were activated by low extracellular magnesium in vitro. These
alpha band overlaps with another EEG phenomenon, the sleep oscillations were NMDA-dependent since they were reversibly
spindles (or sigma rhythms, see next section), led to a legitimate abolished by the NMDA receptor antagonist D-2-amino-
although failed attempt to study it under barbiturate anesthesia 5-phosphonovaleric acid (AP-5) but were relatively unaffected
(112). Thus, most of our present knowledge is based on scalp by the non-NMDA receptor antagonist 6,7-dinitroquinoxaline-
recordings in humans and laminar profiles of cortical field 2,3-dione (DNQX). The duration of oscillatory events was
potentials in animals. increased by blocking GABAA receptors with bicuculline or by
46 Part I ■ Basic Principles

Figure 3.9 Alpha rhythm in dog. A: (Left) Photomicrograph of a section of the marginal gyrus (visual cortex). The elec-
trode bundle consisted of seven wires with a bare tip of 0.15 mm. (Middle) An epoch of alpha activity recorded simulta-
neously from six sites corresponding to those at left, against a common frontal cranial reference (negativity upward). Note
that there is a polarity change between the most superficial sites (E25, 24) and the deepest sites (E22, 21, 19). (Right) Phase
shift between the most superficial site (E25) and deeper lying sites computed by using spectral analysis based on the aver-
age of a number of epochs within the frequency band 11.2–13.1 Hz. The mean values of the phase shift and the correspon-
ding standard deviations are indicated. (Modified from Lopes da Silva FH, Storm Van Leeuwen W. The cortical source of
the alpha rhythm. Neurosci Lett. 1977;6:237–241.) B: Schematic view of the relationship between thalamic and cortical
alpha activity. The lines indicate the amount of influence that a thalamic signal has on the coherence between a pair of
cortical signals (shaded area) recorded during alpha activity. This was measured by partializing the intercortical coherence
on the thalamic signals from the lateral geniculate nucleus (LGN) and pulvinar (PUL) or other cortical signals. Despite the
fact that PUL has an influence on corticocortical domains of alpha activity, the intercortical factors play a significant role
in the establishment of cortical domains of alpha activity. (Modified from Lopes da Silva FH, Vos JE. Mooibroek J, et al.
Relative contributions of intracortical and thalamocortical processes in the generation of alpha rhythms, revealed by par-
tial coherence analysis. Electroencephalogr Clin Neurophysiol. 1980;50:449–456.)
Chapter 3 ■ Cellular Substrates of Brain Rhythms 47

Figure 3.10 A: Multielectrode with 14 equally spaced (200 m) contacts. B: A short segment (200 msec) of alpha
rhythm. C: CSD displayed as a color-coded plot, sinks in red and sources in blue. D: Spectra profiles shown for three sites.
E: Coherence between CSD and multiunit activity (MUA) showing high coherence for the granular and infragranular lay-
ers, and in contrast with the superficial layer. (Modified from Bollimunta A, Chen Y, Schroeder CE, et al. Neuronal mech-
anisms of cortical alpha oscillations in awake-behaving macaques. J Neurosci. 2008;28:9976–9988.)(See color insert.)

activating metabotropic glutamate receptors with trans- vitro conditions may differ from those that are responsible for
1-aminocyclopentane-1,3-dicarboxylic acid (trans-ACPD). alpha rhythm generation in the intact brain. A comprehensive
A similar kind of study in slices of the cat lateral geniculate review of the basic findings with respect to the generation and
nucleus maintained in vitro (118) showed that activation of the organization of alpha rhythms, both in vivo and in vitro, can be
metabotropic glutamate receptor, mGluR1a, which is postsy- found in Ref. 119.
naptic to corticothalamic fibers induces synchronized oscilla- Remarkably, interesting additional findings in the last few
tions at alpha and theta frequencies in a subset of years have enriched our understanding of how alpha rhythms
thalamocortical neurons that can be synchronized by gap junc- are generated and have posed some new challenges for further
tions. The frequency of the oscillations, whether alpha (8 to 13 research. Lörincz et al. (120) confirmed that a sparse network of
Hz) or theta (2 to 7 Hz), depended on the concentration of the high threshold bursting thalamocortical cells coupled through
agonist trans-ACPD. These oscillations are driven by a subset gap junctions is capable of generating alpha rhythmic activity
(~25%) of lateral geniculate thalamocortical neurons display- in cat slices of the lateral geniculate nucleus and ventrobasal
ing high threshold burst activity occurring at membrane poten- complex (somatosensory thalamus). They also found that this
tials more depolarized than –55 mV, which depends on activity can be induced in this preparation by administering a
Ni2 -sensitive Ca2 channels. In these in vitro conditions the cholinergic agonist and can be abolished by muscarinic M3
alpha/theta oscillations do not appear to require chemical receptor antagonists. However, this was evident in only about
synapses because they can be sustained by coupling through 26% of the thalamocortical cells.
gap junctions between thalamocortical neurons. These in An important issue is that of the remaining majority of thal-
vitro results may indicate that activation of mGluRs of corti- amocortical cells. These authors suggest that the remaining
cothalamic synapses is an important mechanism for the gener- cells are probably under phasic inhibition from local interneu-
ation of EEG alpha and theta activity but, as stated above, one rons activated by the bursting thalamocortical cells. It still
should keep in mind that the mechanisms underlying oscilla- remains unclear how these different neuronal populations
tions in the alpha frequency range elicited in this kind of in behave in the intact brain where thalamocortical cells receive
48 Part I ■ Basic Principles

several important inputs from the cortex, RE nucleus of the travel along the dorsal thalamic relay pathway en route to the cor-
thalamus, and several brainstem sources that use various neu- tex, which probably is responsible for the genesis of the dipole
rotransmitters, particularly serotonin, noradrenalin, and hista- that allows them to surface in the EEG. Interestingly, no spindles
mine that are mainly released during the waking state but not are recorded in the EEG of kittens during the first 6 to 7 days of
during REM sleep (121). An important input to these thalamic life, although they are already present in thalamic recordings 3 to
nuclei is the cholinergic input that arises from brainstem and 4 hours after birth (129), suggesting that no viable dipole is gen-
basal forebrain nuclear groups. It has been shown that cholin- erated in the thalamus. The brainstem and basal forebrain pro-
ergic inputs are excitatory to thalamocortical cells causing a jections exert activating effects on the above-mentioned circuit
direct depolarization associated with a decrease of a K cur- with the result of inhibiting spindles.
rent; at the same time there is a decrease of the inhibitory input The RE nucleus has been pointed out as the pacemaker for
arising from the thalamic RE neurons, since the latter are spindle oscillations because spindles were abolished in the dor-
hyperpolarized by cholinergic activation of a K conductance, sal thalamus after disconnection from the RE (130) but were
as was shown in vitro in guinea pig (122) and in vivo in cat preserved in the rostral part of the RE nucleus severed from the
(123,124). In this way these cholinergic inputs favor the block- dorsal thalamus (131). Further support is provided by the fact
age of rhythmic spindles, a phenomenon that is generated in that in cat’s anterior nucleus, which does not receive inputs from
the thalamus and shares the same frequency range with alpha the RE nucleus (132,133), spindles are absent (134). Similarly,
oscillations (see below). It is possible that the major effect of spindles are absent in the cingular cortex (74) and habenular
cholinergic inputs on thalamic oscillations in vivo may be to nuclei, other structures that are devoid of RE inputs. Modeling
induce their blockage through muscarinic inhibition of RE studies of isolated RE neurons, with minimal or more realistic
thalamic neurons, although more subtle effects appear to exist ionic models of RE cells (135–137), further strengthened the
in some thalamic subpopulations, but the relevance of these idea that the deafferented RE nucleus is a spindle pacemaker.
effects for alpha oscillatory activity is not yet clear. These recent Spindle oscillations, in order to produce a coherent pattern
findings provide new insights but it is evident that more at the level of the target structures, need to be synchronized at
research in in vivo conditions is necessary in order to achieve a the very site of their genesis. Besides the axonal projections
deeper understanding of the mechanisms responsible for the allowing obvious connectivity between RE neurons, two other
generation of alpha oscillations of different kinds and localization. mechanisms have been proposed. The first relies on the dendro-
dendritic GABAergic contacts between RE neurons (138–140)
SPINDLE (SIGMA) RHYTHMS and the second on the functional interconnection of RE neu-
rons through gap junctions (141).
Classically, spindles have been regarded as one of the first signs A fundamental question concerns the triggering factor of a
of EEG synchronization during the early stage of quiescent spindle. Steriade and colleagues (131) have assumed that any
sleep. This type of oscillation is defined by the presence of two excitatory drive stimulating RE cells would start the process.
distinct rhythms: waxing and waning spindle waves at 7 to The decreased activity of brainstem cholinergic neurons at the
14 Hz within sequences lasting for 1 to 2 seconds, and the peri- onset of sleep (53) contributes to the overall hyperpolarization
odic recurrence of spindle sequences with a slow rhythm, gen- of thalamocortical cells, thus bringing the membrane potential
erally 0.2 to 0.5 Hz (Fig. 3.11A). Spindles are generated within in the range where bursting discharges can occur. Such clusters
the thalamus because they survive in the thalamus after decor- of high-frequency action potentials would excite the dendrites
tications and high brainstem transection (125). More radical of RE neurons and would trigger the dendrodendritic ava-
procedures, including complete removal of the striatum and lanche leading to the final synchronization of the whole RE
rhinencephalon, do not affect thalamic spindles either (14). nucleus. It should be emphasized that thalamic-RE loops may
Sleep spindles have been mostly studied in cats (in vivo) and assist the onset of a spindle; however, its synchronization can-
ferrets (in vitro). Humans and cats have similar sleep cycles, not rely on the dorsal thalamic circuitry because there is scarce,
EEG patterns, and ultrastructural organization of their thala- if any, communication between thalamocortical neurons and
mus. Both intrinsic properties of various thalamic neurons dorsal thalamic nuclei (142,143).
(126) and network linkages (for review, see Ref. 127) contribute Once a spindle has been started through one of the mecha-
to the patterning of spindles. nisms mentioned above, the pacing of the actual oscillations
As indicated above, the thalamic circuits comprise two main within the spindle sequence depends on the RE–thalamic dia-
structures: (i) the dorsal thalamus, made of several nuclei, each of logue. This was investigated in ferret thalamic visual slices con-
them containing both relay (thalamocortical) and local-circuit taining geniculate and perigeniculate (RE) nuclei (144–146) and
(inter-) neurons and (ii) the RE nucleus of the thalamus (see with multisite recordings within the thalamus (147,148). Results
scheme in Fig. 3.11A). The latter is a thin sheet of GABAergic show that the bursting of RE neurons imposes powerful
neurons that covers the rostral, lateral, and ventral surfaces of the GABAergic IPSP in thalamocortical neurons. The end of this inhi-
thalamus (128). It mainly receives inputs from the cerebral cor- bition triggers a rebound LTS, crowned by a high-frequency burst
tex and dorsal thalamus, but also from the rostral sector of the of action potentials, which in turn will again excite the target RE
brainstem and basal forebrain. The RE–thalamic–cortical–RE cells. This is also the moment where intrinsic properties of thala-
loop constitutes a resonating circuit that reinforces the spindle mic, both thalamocortical and RE, neurons play an important
oscillation. Moreover, spindles generated within the RE nucleus role. The amount of hyperpolarization of the thalamocortical cells
Chapter 3 ■ Cellular Substrates of Brain Rhythms 49

Figure 3.11 Spindle oscillations in reticular


thalamic (RE), thalamocortical (Th-Cx, ventro-
lateral nucleus), and cortical (Cx, motor area)
neurons. A: Circuit of three neuronal types and
two rhythms (7–14 Hz and 0.1–0.2 Hz) of spin-
dle oscillations in cortical EEG. B: Intracellular
recordings in cats under barbiturate anesthesia.
(Modified from Steriade M, Deschênes M.
Intrathalamic and brainstem-thalamic networks
involved in resting and alert states. In:
Bentivoglio M, Spreafico R, eds. Cellula r
Thalamic Mechanisms. Amsterdam: Elsevier;
1988:37–62.)

determines the degree of deinactivation of the LTS (149). Another In an intact sleeping brain, the RE–thalamic network under-
intrinsic property of thalamic cells is the voltage-dependent, non- goes periodic, synchronized excitatory inputs from the cortex
inactivating or very slowly inactivating persistent sodium current, such as K-complexes (see above), which then shape and modu-
INa(p) (149), which modulates postinhibitory rebound depolariza- late the duration of spindles (153). Corticothalamic stimuli
tions. The blockage of the INa(p) by intracellular injection of qua- produce shorter and waning spindles that lack the initial wax-
ternary derivatives of local anesthetics transformed spindles of ing component, probably because the volleys entrained from
thalamocortical cells into a single, long-lasting period of hyperpo- the very beginning large populations of thalamic cells.
larization (because unopposed by INa(p)) without any rhythmic Moreover, the synchronization of spindles in the thalamus was
rebounds within the frequency range of spindles (150). Finally, widespread, with virtual zero time lag, in intact preparations,
the calcium-dependent potassium current, IK(Ca) , or other but dropped drastically after decortications (147,148) (Fig. 3.12).
voltage-dependent potassium currents (151,152) may prolong the This result stands in some contrast to a propagating pattern of
long-lasting IPSPs generated in thalamocortical neurons by RE spindles in thalamic slices (154) likely due to the absence of
neurons and, thus, assist in the production of the LTS and in corticothalamic inputs of this preparation and to the overall
inducing rebound bursts in thalamocortical targets. diminished synaptic activity of the slice.
Figure 3.12 Control of spatiotemporal coherence of thalamic spindles by the cerebral cortex in cat. Top panels: Disruption of the spatiotemporal coher-
ence of thalamic oscillation after removal of the neocortex. Spatiotemporal maps of electrical activity across the thalamus were constructed by plotting
time (time runs from top to bottom in each column; arrow indicates 1 second), space [from left to right, the width of each column represents 8 mm in
the anteroposterior axis of the thalamus), and local field potential (LFP) voltage (from blue to yellow, color represents the amplitude of the negative
deflection of thalamic LFPs; the color scale ranged in ten steps from the baseline [blue] to 100 V [yellow]). Time was divided into frames, each rep-
resenting a snapshot of 4 msec of thalamic activity. A total of 40 seconds is represented (9880 frames). Each frame consisted of eight color spots, each
corresponding to the LFP of one electrode from anterior to posterior (left to right in each column). Middle panels (experiments): Decay of correlation
with distance. Cross-correlations were computed for all possible pairs of thalamic sites, and the value at time zero from each correlation was repre-
sented as a function of the intersite distance for six different consecutive epochs of 20 seconds. Spatial correlation was calculated for thalamic record-
ings in the intact brain (left) and after removal of cortex (right). Bottom panel (model): Decay of correlation with distance (in units of sites). Similar
computation of cross-correlations as in the above panel from experiments, in the presence of cortex (left) and after decortication (right). (Modified from
Contreras D, Destexhe A, Sejnowski TJ, et al. Control of spatiotemporal coherence of a thalamic oscillation by corticothalamic feedback. Science.
1996;274:771–774. and unpublished data by A Destexhe, D Contreras, TJ Sejnowski and M Steriade.) (See color insert.)
50
Chapter 3 ■ Cellular Substrates of Brain Rhythms 51

The main functional correlate of sleep spindles is the block- a conditioned response to a visual stimulus in monkey (163),
age of incoming stimuli on their way to the cortex. This could during tasks requiring fine finger movements and focused atten-
result from the fact that thalamocortical neurons alternate tion in monkey motor cortical cells (164), and during behavioral
between IPSPs and LTS bursts. Both events preclude the relay- immobility associated with an enhanced level of vigilance while
ing of afferent synaptic signals either due to the shunting nature a cat was watching a visible but unseizable mouse (165). Other
of the former or due to the all-or-none feature of the latter. The studies have described stimulus-dependent oscillations at 25 to
functional blockage of sensory transmission at the thalamic 45 Hz of the focal EEG and/or neuronal firing in the olfactory
level promotes the cortical deafferentation, further contribut- system (166) and visual cortex (167–171).
ing to the process of falling asleep. The underlying mechanism responsible for cortical gamma
A final observation concerns the overlapping frequency oscillations was further unraveled by the finding that synchro-
range of spindles and alpha waves. It should be emphasized that nized gamma frequency oscillations are enhanced with arousal
both behavioral context and mechanisms are dissimilar. Alpha and attention (172) or by electrical stimulation of the mesen-
waves generally occur during relaxed wakefulness, and in some cephalic RE formation (173), and are mediated by acetylcholine
studies they are considered to regulate afferent and efferent sig- muscarinic receptors. In addition, the spectral power of local field
nals (155). At difference, spindle oscillations are occurring dur- potentials in the gamma frequency range in the visual cortex of
ing unconsciousness, thus making the idea of an the cat is closely correlated with hemodynamic signals (174).
alpha-to-spindle continuum obsolete. Nonetheless the same The main functional implication of these rhythms in the cor-
basic neural circuitries seem to be involved in both kinds of tex was proposed to rely on the degree of spatial and temporal
oscillatory activities—sleep spindles and alpha rhythms—but synchronization, thus allowing, at a given moment, the aggrega-
working according to different functional modes. tion of various cortical areas with the purpose of creating global
and coherent properties of patterns, a prerequisite for scene seg-
FASTER (BETA AND GAMMA) RHYTHMS mentation and figure-ground distinction (175). The issue of this
“feature binding” has, however, been toned down by the fact that
At the opposite side of the EEG spectrum of slow sleep rhythms such fast rhythms are coherent also within thalamocortical and
( 15 Hz) are fast oscillations (generally 20 to 50 Hz) that are cortical networks during states such as sleep and deep anesthe-
proper to vigilance states associated with wakefulness, but also sia reputed for the absence of any conscious behavior
paradoxical (REM) sleep. The brain substrate of EEG activation (19,176,177). In these studies it was established that thalamocor-
upon arousal has begun to be understood since the pioneering tical cells do spontaneously oscillate within the beta–gamma fre-
work of Moruzzi and Magoun (156). They reported an “activa- quency range and this occurs coherently with field potentials
tion” response to the stimulation of brainstem structures con- recorded from the related cortical areas (Fig. 3.13).
sisting of the suppression of spindles and slower EEG rhythms. Most likely there exist different kinds of beta–gamma
The cellular mechanisms of this suppressing mechanism have rhythms with different properties and behavioral/cognitive
recently been analyzed: the blockage of spindles occurs at the associations. Since the report by Gray and Singer (169) that the
very site of their genesis (the RE nucleus) through the action of firing probability of neurons of the visual cortex of the cat, in
acetylcholine serotonin and norepinephrine. Although acetyl- response to the presentation of appropriate visual stimuli, oscil-
choline hyperpolarizes RE neurons (124,157), while the latter lates with a frequency in the gamma frequency range, along
two depolarize them (158), the combined action of these neu- with the observation that no evidence for similar oscillations
rotransmitters is far from being understood. The intrinsically were found in the thalamus, these gamma oscillations are con-
generated thalamic delta oscillation is blocked through the sidered to be generated intracortically. This observation led to
depolarizing action of both acetylcholine (10) and the assumption that these gamma oscillations may reflect a gen-
monoamines (159) on thalamocortical cells. Finally, slow corti- eral mechanism that is capable of binding together activities of
cal delta activities are prevented by cholinergic actions of the spatially separate cortical areas (179). In general we may state
nucleus basalis neurons (30,160). that synchronization of neuronal networks by way of common
oscillations in the gamma frequency range may enable fast
Beta–Gamma Rhythms: Functional Implications routing and processing of information in the cortex (180).
The disappearance of slow sleep waveforms during the activat-
ing response is also accompanied by the appearance of peculiar Genesis of Beta–Gamma Rhythms
fast rhythms characterizing wakefulness (161). This study The genesis of fast (beta–gamma) activities lies at the crossroads
reported, in addition to the ocular syndrome of arousal, a clear- of intrinsic and network properties. Cortical neurons generate
cut enhancement in amplitude of the spontaneous EEG rhythms upon depolarization intrinsic oscillations in a range around 40
and their regular acceleration to 40 to 45 Hz instead of the till Hz (181,182). This is equally valid for a subclass of rostral
then observed flattening of the cortical EEG. Since then, several intralaminar thalamic neurons (183) (Fig. 3.14), a property that
papers have mentioned the presence of 20 to 40 Hz waves in var- is in line with their implication in fast oscillations during acti-
ious cortical areas, during different conditions of increased vated states associated with neuronal depolarizations, waking,
alertness. For instance, fast rhythms were observed in a canine and REM sleep. Intralaminar nuclei are particularly fit for
subject in the occipital cortex while the dog paid intense atten- wide range synchronization due to their widespread projections
tion to a visual stimulus (162), during accurate performance of to the cerebral cortex (125) including the visual areas (184).
52 Part I ■ Basic Principles

Figure 3.13 Episodes of tonic activation are


associated with coherent fast rhythms (40 Hz)
in cortical EEG and intracellularly recorded
thalamocortical neuron. Cat under ketamine-
xylazine anesthesia. A: Four traces represent
simultaneous recordings of surface and depth
EEG from motor cortical area 4, extracellular
discharges of neuron from the rostrolateral part
of the thalamic reticular (RE) nucleus, and
intracellular activity of thalamocortical neuron
from ventrolateral (VL) nucleus. EEG, RE, and
VL cells displayed a slow oscillation (0.7–
0.8 Hz) during which the sharp depth-negative
(excitatory) EEG waves led to IPSPs in VL cell,
presumably generated by spike bursts in a cor-
tically driven GABAergic RE neuron. Part
marked by horizontal bar, taken from a short-
lasting period of spontaneous EEG activation, is
expanded in B (arrow), with EEG waves and
field potentials from the RE nucleus filtered
between 30 and 50 Hz; part marked by horizon-
tal bar in this panel is further expanded in C to
illustrate relations between action potentials of
VL cell and depth-negative waves in cortical
EEG at a frequency of 40 Hz. D: Cross-
correlations (CROSS) between action potentials
and depth-EEG shows clear-cut relation, with
opposition of phase, between intracellularly
recorded VL neuron and EEG waves. (Modified
from Steriade M, Contreras D, Amzica F, et al.
Synchronization of fast (30–40 Hz) sponta-
neous oscillations in intrathalamic and thalam-
ocortical networks. J Neurosci. 1996;16:
2788–2808.)

Despite these intrinsic properties that make neurons prone somatosensory slices with slightly reduced inhibition display
to generate fast oscillations, their imbedding in complex cir- activities around 37 Hz generated by networks of intrinsically
cuits, especially in the cortex, is required for the short- and pyramidal-shaped (excitatory) cells.
long-range synchronization, in order to make the rhythm The complexity of the cortical generators of fast
emerge at the EEG level. As examples of different kinds of (beta–gamma) rhythmic activities is illustrated further by the
circuits that may engage fast rhythmic activities we may men- observation that, under anesthesia, they do not show field rever-
tion the following two findings: Freeman (180) postulated that sal at any depth of the cortex (19). Volume conduction was ruled
the generation of 40- to 80-Hz activity in the olfactory bulb out because the negative field potentials of the fast oscillations
depends on feedback inhibitory circuits involving local-circuit were systematically associated, at all depths, with neuronal firing
GABAergic neurons acting on output elements, the mitral cells, and were not observable in the underlying white matter. This
while Chagnac-Amitai and Connors (185) showed that rat fact can be interpreted as being due to the existence of several
Chapter 3 ■ Cellular Substrates of Brain Rhythms 53

Figure 3.14 Fast oscillatory patterns of neuron recorded from the dorsolateral part of the centrolat-
eral (CL) intralaminar thalamic nucleus, characterized by exceedingly high-frequency (800 to
1000 Hz) spike bursts recurring rhythmically at 50 to 60 Hz. Intracellular recording in cat under bar-
biturate anesthesia. A: Activities triggered by depolarizing current pulses ( 1.2 nA, 50 msec) at dif-
ferent levels of membrane potential (as indicated at left). Two examples are illustrated for each
membrane potential level. At bottom, presence of low-threshold spike (LTS) leading to high-
frequency (800 to 1000 Hz) spike burst at –78 mV and its absence at –84 mV. Note fast-recurring (50
to 60 Hz) spike doublets or spike triplets at relatively depolarized levels (–64 and –58 mV), a feature
that is not seen in other types of thalamocortical neurons. B: Oscillatory patterns similar to those
elicited by current injection occurred spontaneously at similar membrane potentials (from top to bot-
tom, –58, –64, –68, and –78 mV). (Modified from Steriade M, Curró Dossi R, Contreras D.
Electrophysiologic properties of intralaminar thalamocortical cells discharging rhythmic (approxi-
mately 40 Hz) spike-bursts at approximately 1000 Hz during waking and rapid eye movement sleep.
Neuroscience. 1993;56:1–9.)

intracortical distributed microsinks and microsources as shown increased levels of alertness, thus closely following the onset of
by current-source-density analyses (176). Thus, the absence of activity in cholinergic aggregates of the brainstem and basal
potential reversal was due to the lower intensity of vertical cur- forebrain. The cholinergic activation induces a twofold increase
rents, compared with that of transmembrane currents. of cortical EEG waves around 40 Hz, a potentiating effect that
Despite their presence during sleep and under anesthesia, is mediated by muscarinic receptors as it is blocked by scopo-
fast (beta–gamma) oscillations are mainly associated with lamine (30,183). Beyond the depolarizing effect exerted by
54 Part I ■ Basic Principles

acetylcholine on thalamocortical and cortical neurons, this a remarkable quantitative indicator of the pharmacologic effect
neurotransmitter hyperpolarizes a subpopulation of cortical of benzodiazepines. In addition to the classic beta and gamma
glial cells maintained in culture (186). When tested in vivo, oscillations, episodes of oscillations at high frequency (100 to
acetylcholine hyperpolarized most of the glial cells (Fig. 3.15), 600 Hz) and short duration (~50 to 100 ms) have been
in parallel with an overall decrease in the extracellular potas- described in the brain of rodents and human, both under nor-
sium concentration (187) (Fig. 3.15; see also next section). This mal and epileptic conditions. These oscillatory bursts are called
effect was also accompanied by an increased cerebral blood ripples (see Fig. 3.8) or fast ripples depending on frequency and
flow, leading to the interpretation that the glial hyperpolariza- are discussed in Chapter 37.
tion is due to a boosted transport of potassium across capillary
membranes. This is further supported by the fact that EEG acti- Fast (Beta–Gamma) Rhythms in Human EEG
vation, when recorded with DC amplifiers, generally displays a While the mechanisms of generation of gamma and beta
persistent positive DC shift (187). In addition, brain activation rhythms in cortical slices, in vitro, present clear different prop-
was accompanied in glial cells by a reduced membrane capaci- erties, the situation in vivo in humans is even more complex.
tance suggesting that the interglial syncytium shuts down dur- On the one hand, beta EEG or MEG activities recorded over the
ing wakefulness, preventing nonspecific synchronization Rolandic cortical area (about 15 and 30 Hz) display a close tem-
through spatial buffering mechanisms (as mentioned previ- poral relationship with peripheral EMG activity during isomet-
ously). This feature may thus leave the synchronization of ric contractions, as shown using MEG recordings (190–192).
beta–gamma activities strictly confined to synaptic networks, The MEG signals in the beta frequency range lead the EMG sig-
achieving a more specific aggregation of the neuronal popula- nals in time, and the time lag increases with increasing
tions involved. brain–muscle distance. This implies that this beta rhythmic
Recent studies in cortical slices revealed particularly interest- activity is associated with rhythmic firing of neurons of the
ing properties with respect to the generation of gamma and motor cortex that generate the commands to drive spinal
beta rhythmic activities. Roopun et al. (188) showed that motoneurons (193) (see also Chapter 42). On the other hand,
gamma (30 to 70 Hz) rhythms could be generated in the super- however, there are other conditions where beta rhythms are not
ficial somatosensory cortical layers II/III, while beta rhythms associated with a state of active neuronal firing in the sense
(20 to 30 Hz) were generated in deep layer V mainly associated described above. This is the case of an increase of beta activity
with the activity of fast-spiking interneurons when kainate was after a finger movement (postmovement beta rebound at 16 to
applied to the slices. It is interesting to note that in layer IV both 21 Hz) or foot movement (19 to 26 Hz) (194) when the mus-
activities may coexist (Fig. 3.16). A remarkable difference in the cles relax. The combination of EEG recording with the applica-
physiologic mechanisms underlying these two types of cortical tion of transcranial magnetic stimulation showed that during
oscillations is that the beta rhythms of the deep layers are this beta rebound the corticospinal excitability was diminished
reduced by carbenoxolone, which blocks gap junction conduc- (195). The dynamics of changes of beta rhythms under differ-
tances, but are not affected by the blockade of synaptic trans- ent behavioral conditions is further detailed in Chapter 45.
mission. In contrast, gamma rhythms in superficial layers are Related to the beta rebound after a movement it was shown that
reduced by the blockade of chemical synaptic transmission. a surge of gamma activity (above 30 Hz) may immediately pre-
The fact that gamma and beta oscillations rely on different cede a finger movement (196), which is probably directly asso-
mechanisms is further emphasized in experiments with trans- ciated with the activation of motor cortical neurons.
versal cuts within the somatosensory cortical slices that com-
pletely separate layers I to II from layers V to VI. The gamma DC EEG POTENTIALS
rhythmic activity of the superficial layers and the beta rhythmic
activity of the deep layers survived the cut, showing that they Although the chapter is devoted to oscillations in the brain, this
can be maintained separately. The two kinds of rhythmic activ- section will briefly raise the issue of less conventional, very slow
ities also respond differently to pharmacologic agents. While EEG signals, which are overwhelmingly left out of the clinical
the blockage of GABAA receptors in these slices abolished focus (see, however, Refs. 197 and 198). Interestingly, the first
gamma rhythms, it enhanced beta rhythms. This means that EEG machines used by Berger for his first EEG recordings were
gamma oscillations depend on the discharge of pyramidal cells true DC amplifiers. This changed with the introduction of
coupled to GABA-mediated local inhibitory circuits. We should increasingly performing filters offering the possibility to limit
emphasize, however, that demonstrations of this kind of prop- the intrusion of artifacts in clinical recordings. Filters also pre-
erties in vitro are not a guarantee that precisely the same mech- cluded the expression of neurophysiologic relevant informa-
anisms are also essential in vivo. Nonetheless in vitro studies as tion, which, in time, limited the knowledge of brain
this one yield a proof-of-principle that such mechanisms can be mechanisms. The consecutive conventional theories in elec-
operational in the cortex. troencephalography considered that most of the electrical
It was reported in the rat in vivo that benzodiazepines, which activity recorded on the scalp, including very slow waves, is gen-
enhance GABAA receptor-mediated synaptic transmission, also erated by neuronal networks with particular emphasis on corti-
increased beta oscillations in the EEG. In this sense, Mandema cal dipoles. Several studies have also demonstrated the
and Danhof (189) reviewed data showing that changes in the involvement of glial cells in generating slow local field poten-
EEG spectral amplitude within the beta frequency band provide tials during spreading depression (199,200), seizures
Chapter 3 ■ Cellular Substrates of Brain Rhythms 55

Figure 3.15 Comparison between electrically elicited and spontaneously occurring activation in a pair of simultane-
ously impaled neuron and glia in a cat under ketamine-xylazine anesthesia. A: Sequence of slow ( 1 Hz) oscillation,
brainstem cholinergic activation, and return to slow oscillation. Depolarizing phases of the slow oscillation are indi-
cated with a black arrow head (up state), while hyperpolarizing phases are marked with an empty arrow head (down
state). After activation, the sustained neuronal depolarization is accompanied by glial hyperpolarization and the DC
field potential assumes rather positive values. B: Histograms of neuronal (panel 1) and glial (panel 2) Vm during the
slow oscillation (in gray) and during activation (black trace). Histograms were calculated over the respective under-
lined epochs in A. They illustrate the incidence of different values of Vm , sampled at 20 kHz, over periods indicated
by bar below traces. Note the bimodal histograms during sleep (up states correspond to the black arrow head, down
states to the empty arrow head) and the unimodal histogram after activation, as well as the opposite evolution of glial
and neuronal Vm (glia hyperpolarizes and neuron depolarizes). C1: Similar pattern of activity in the same double intra-
cellular recording during a period of spontaneous activation of the EEG. Periods within squares are expanded at right
(panels 2 and 3). D: Histograms from equivalent time periods before and after activation. (Modified from Seigneur J,
Kroeger D, Nita DA, et al. Cholinergic action on cortical glial cells in vivo. Cereb Cortex. 2006;16:655–668.)
56 Part I ■ Basic Principles

Figure 3.16 2 rhythms are generated in deep layers of cortex. A: Spectrograms of field rhythms generated
in somatosensory cortex slices by 400 nM kainate. Superficial layers generated gamma frequency (30 to
50 Hz) signals (layer II/ III). Deep layers concurrently generated 2 frequency signals (layer V, 20 to 30 Hz).
Layer IV recordings show both frequency bands coexisting. Below each spectrogram are representative field
potential traces (scale bars: 0.2 mV, 100 ms). B: Surgical separation of deep from superficial layers at the layer
IV/ V border abolished neither rhythm. Cartoon illustrates the electrode position for the power spectra taken
from 60-second epochs of field potential data in the superficial layers (a–c, black lines) and deep layers
(d–e, red lines). (Modified from Roopun AK, Middleton SJ, Cunningham MO, et al. A 2-frequency (20–30 Hz)
oscillation in nonsynaptic networks of somatosensory cortex. Proc Natl Acad Sci USA. 2006;103:
15646–15650.)(See color insert.)

(199–203), and sleep (36). Besides these concepts, pioneering mechanisms generating ventilation-related DC shifts that
studies in the early 1950s to 1970s have proposed that some of emphasized an almost exclusive role of cortical neurons and their
the slow potential shifts are generated at the interface between dendritic tree (201,213). Recent data (212) demonstrate that such
cerebrospinal fluid and blood as a function of the partial pres- ventilation-induced DC shifts occurred in the absence of any
sure of CO2 (PCO2) (204–208). Such potentials were suggested parallel change in the neuronal or glial membrane potential. No
to originate across the blood–brain barrier (209–212). The extracellular, spatial polarity reversal of the DC shift responses
merit, but also the difficulty, of these studies consisted in intro- was seen across or within the neocortex and the underlying white
ducing among the variables that condition brain phenomena matter. This finding provides further support for the view that
extraneuronal parameters. the DC shifts are not caused by cortical current dipoles generated
The basic aspects related to EEG potentials situated at the in response to neuronal and/or glial activity. And finally, the
infraslow limit of the spectrum (DC to 0.01 Hz, although the breakdown of the blood–brain barrier produced itself a DC shift
upper limit is only informative) rely on the electrical scheme as mark of the polarizing potential of the barrier. Once the
proposed by Voipio and colleagues (211) (Fig. 3.17A). It proved blood–brain barrier was disrupted, ventilation-induced DC
to be particularly useful to understand how large DC shifts that shifts were abolished (Fig. 3.18). These examples clearly state
can be recorded on the scalp in response to hypo/hypercapnia that, other than neurons and glia, a series of other parenchymal
(Fig. 3.17C,D) are not generated by networks of neurons and/or elements (blood flow and capillary epithelial cells) participate in
glia. This is in stark contrast with the prevailing view about the the genesis of potentials that translate into EEG signals.
Chapter 3 ■ Cellular Substrates of Brain Rhythms 57

Figure 3.17 DC shift potentials generated in the EEG by variations of the systemic CO2. A left: Schematic
drawing of the human head divided into four compartments: brain, blood, the blood–brain or blood–CSF bar-
rier (black double line), and all other tissues. EBB, the electromotive force of the voltage source across the
brain–blood interface; RBB, its internal resistance. This voltage source generates a volume current (lines with
arrowheads) that flows first through RB (the distributed resistance that couples brain potential to the
surrounding extracortical tissue layers) and RS (the distributed resistance of the layers between brain surface
and skin surface pooled together) and gives rise to the voltage drop VDC that can be measured on scalp.
Current returns back to the brain–blood interface through RT1 (resistance of wider tissue pathways below the
level of cranial fossae), RT2 (access resistance to blood), and RT3 (resistance of blood). A right: Simplified
equivalent circuit of the scheme depicted at left. IBB denotes the current that is driven in the circuit by the
brain–blood potential difference (VBB). (Modified from Voipio J, Tallgren P, Heinonen E, et al. Millivolt-scale
DC shifts in the human scalp EEG: evidence for a nonneuronal generator. J Neurophysiol.
2003;89:2208–2214.) B: Scheme with the position of the scalp electrodes with respect to the brain of the cat.
DC shifts induced by hyperventilation (C) and hypoventilation (D) in a cat under ketamine-xylazine anesthe-
sia. Below the EEG signals, variations of the CO2 concentration in the expired air. Hyperventilation induced
positive DC shifts, while hypoventilation was associated with negative DC shifts. (Modified from Nita DA,
Vanhatalo S, Lafortune FD, et al. Nonneuronal origin of CO2-related DC EEG shifts: an in vivo study in the
cat. J Neurophysiol. 2004;92:1011–1022.)
58 Part I ■ Basic Principles

Figure 3.18 DC shifts induced by the disruption of the blood–brain barrier. Scalp DC EEG recording in a cat
under ketamine-xylazine anesthesia. After a control period, a volume of 3 ml saline (SAL) was slowly injected
into the right carotid artery with no effect. Then an identical volume of sodium dehydrocholate (DHC; 17.5% )
was injected in order to break the blood–brain barrier. This induced a clear positive DC shift, with right lat-
eralization. During the period with open blood–brain barrier, the amplitude of the responses to hypo/ hyper-
ventilation (hv/ HV) maneuvers was drastically diminished. The asterisk during the first hypoventilation
maneuver indicates that initially for a period of about 1 minute the respirator was stopped. (Modified from
Nita DA, Vanhatalo S, Lafortune FD, et al. Nonneuronal origin of CO2-related DC EEG shifts: an in vivo study
in the cat. J Neurophysiol. 2004;92:1011–1022.)

REFERENCES 10. Steriade M, Curró Dossi R, Nuñez A. Network modulation of a


slow intrinsic oscillation of cat thalamocortical neurons impli-
1. Walter G. The location of cerebral tumors by electro- cated in sleep delta waves: cortically induced synchronization
encephalography. Lancet. 1936;8:305–308. and brainstem cholinergic suppression. J Neurosci. 1991;
2. IFSECN. A glossary of terms most commonly used by clinical elec- 11:3200–3217.
troencephalographers. Electroencephalogr Clin Neurophysiol. 11. Curró Dossi R, Nuñez A, Steriade M. Electrophysiology of a slow
1974;37:538–548. (0.5–4 Hz) intrinsic oscillation of cat thalamocortical neurones in
3. Creutzfeldt OD, Watanabe S, Lux HD. Relations between EEG phe- vivo. J Physiol. 1992;447:215–234.
nomena and potentials of single cortical cells. II. Spontaneous and 12. Nuñez A, Curró Dossi R, Contreras D, et al. Intracellular evidence
convulsoid activity. Electroencephalogr. Clin Neurophysiol. for incompatibility between spindle and delta oscillations in thal-
1966;20:19–37. amocortical neurons of cat. Neuroscience. 1992;48:75–85.
4. Kellaway P, Gol A, Proler M. Electrical activity of the isolated cerebral 13. Nita DA, Steriade M, Amzica F. Hyperpolarisation rectification in
hemisphere and isolated thalamus. Exp Neurol. 1966;14:281–304. cat lateral geniculate neurons modulated by intact corticothalamic
5. Rappelsberger P, Pockberger H, Petsche H. The contribution of the projections. J Physiol. 2003;552:325–332.
cortical layers to the generation of the EEG: field potential and 14. Villablanca J. Role of the thalamus in sleep control: Sleep-
current source density analyses in the rabbit’s visual cortex. wakefulness studies in chronic diencephalic and athalamic cats. In:
Electroencephalogr. Clin Neurophysiol. 1982;53:254–269. Petre-Quadens O, Schlag J, eds. Basic Sleep Mechanisms. New York:
6. Leresche N, Jassik-Gerschenfeld D, Haby M, et al. Pacemaker-like Academic; 1974:51–81.
and other types of spontaneous membrane potential oscillations 15. Ball GJ, Gloor P, Schaul N. The cortical electromicrophysiology of
of thalamocortical cells. Neurosci. Lett. 1990;113:72–77. pathological delta waves in the electroencephalogram of cats.
7. McCormick DA, Pape HC. Properties of a hyperpolarization- Electroencephalogr. Clin Neurophysiol. 1977;43:346–361.
activated cation current and its role in rhythmic oscillation in 16. Steriade M, Buzsáki G. Parallel activation of thalamic and cortical
thalamic relay neurones. J Physiol. 1990;431:291–318. neurons by brainstem and basal forebrain cholinergic systems. In:
8. Leresche N, Lightowler S, Soltesz I, et al. Low-frequency oscillatory Steriade M, Biesold D, eds. Brain Cholinergic Systems. New York:
activities intrinsic to rat and cat thalamocortical cells. J Physiol. Oxford University Press; 1990:3–62.
1991;441:155–174. 17. Steriade M, Gloor P, Llinás RR, et al. Report of IFCN committee
9. Soltesz I, Lightowler S, Leresche N, et al. Two inward currents and on basic mechanisms. Basic mechanisms of cerebral rhythmic
the transformation of low-frequency oscillations of rat and cat activities. Electroencephalogr Clin Neurophysiol. 1990;76:
thalamocortical cells. J Physiol. 1991;441:175–197. 481–508.
Chapter 3 ■ Cellular Substrates of Brain Rhythms 59

18. Steriade M, Nuñez A, Amzica F. A novel slow (< 1 Hz) oscillation 39. Bowman CL, Kimelberg HK. Excitatory amino acids directly
of neocortical neurons in vivo: depolarizing and hyperpolarizing depolarize rat brain astrocytes in primary culture. Nature.
components. J Neurosci. 1993;13:3252–3265. 1984;311:656–659.
19. Steriade M, Amzica F, Contreras D. Synchronization of fast (30–40 40. Kettenmann H, Schachner M. Pharmacological properties of
Hz) spontaneous cortical rhythms during brain activation. J gamma-aminobutyric acid-, glutamate-, and aspartate-induced
Neurosci. 1996;16:392–417. depolarizations in cultured astrocytes. J Neurosci. 1985;5:
20. Achermann P, Borbély AA. Low-frequency (<1 Hz) oscillations in the 3295–3301.
human sleep electroencephalogram. Neuroscience. 1997;81:213–222. 41. Orkand RK, Nicholls JG, Kuffler SW. Effect of nerve impulses on
21. Amzica F, Steriade M. The K-complex: its slow (<1-Hz) rhythmic- the membrane potential of glial cells in the central nervous system
ity and relation to delta waves. Neurology. 1997;49:952–959. of amphibia. J Neurophysiol. 1966;29:788–806.
22. Simon NR, Manshanden I, Lopes da Silva FH. A MEG study of 42. Kettenmann H, Ransom BR. Electrical coupling between astro-
sleep. Brain Res. 2000;860:64–76. cytes and between oligodendrocytes studied in mammalian cell
23. Mölle M, Marshall L, Gais S, et al. Grouping of spindle activity cultures. Glia. 1988;1:64–73.
during slow oscillations in human non-rapid eye movement sleep. 43. Walz W. Role of glial cells in the regulation of the brain ion
J Neurosci. 2002;22:10941–10947. microenvironment. Prog Neurobiol. 1989;33:309–333.
24. Massimini M, Rosanova M, Mariotti M. EEG slow (approximately 44. Nicholson C. Extracellular space as the pathway for neuron-glial
1 Hz) waves are associated with nonstationarity of thalamo- cell interaction. In: Kettenmann H, Ransom BR, eds. Neuroglia.
cortical sensory processing in the sleeping human. J Neurophysiol. New York: Oxford University Press; 1995;387–397.
2003;89:1205–1213. 45. Ballanyi K, Grafe P, ten Bruggencate G. Ion activities and potas-
25. Steriade M, Nuñez A, Amzica F. Intracellular analysis of relations sium uptake mechanisms of glial cells in guinea-pig olfactory cor-
between the slow (< 1 Hz) neocortical oscillation and other tex slices. J Physiol. 1987;382:159–174.
sleep rhythms of the electroencephalogram. J Neurosci. 46. Kettenmann H. K+ and Cl- uptake by cultured oligodendrocytes.
1993;13:3266–3283. Can J Physiol Pharmacol. 1987;65:1033–1037.
26. Timofeev I, Steriade M. Low-frequency rhythms in the thalamus 47. Kocsis JD, Malenka RC, Waxman SG. Effects of extracellular potas-
of intact-cortex and decorticated cats. J Neurophysiol. 1996;76: sium concentration on the excitability of the parallel fibres of the
4152–4168. rat cerebellum. J Physiol. 1983;334:225–244.
27. Sanchez-Vives MV, McCormick DA. Cellular and network mecha- 48. Rausche G, Igelmund P, Heinemann U. Effects of changes in extra-
nisms of rhythmic recurrent activity in neocortex. Nat Neurosci. cellular potassium, magnesium and calcium concentration on
2000;3:1027–1034. synaptic transmission in area CA1 and the dentate gyrus of rat
28. Contreras D, Timofeev I, Steriade M. Mechanisms of long-lasting hippocampal slices. Pflugers Arch. 1990;415:588–593.
hyperpolarizations underlying slow sleep oscillations in cat corti- 49. Hille B. Ion Channels of Excitable Membranes. Sunderland:
cothalamic networks. J Physiol. 1996;494(Pt 1):251–264. Sinauer; 2001.
29. Massimini M, Amzica F. Extracellular calcium fluctuations and 50. Loomis AL, Harvey EN, Hobart G. Distribution of disturbance
intracellular potentials in the cortex during the slow sleep oscilla- patterns in the human electroencephalogram, with special refer-
tion. J Neurophysiol. 2001;85:1346–1350. ence to sleep. J Neurophysiol. 1938;1:413–440.
30. Steriade M, Amzica F, Nuñez A. Cholinergic and noradrenergic 51. Colrain IM. The K-complex: a 7-decade history. Sleep.
modulation of the slow (approximately 0.3 Hz) oscillation in neo- 2005;28:255–273.
cortical cells. J Neurophysiol. 1993;70:1385-1400. 52. Amzica F, Steriade M. Cellular substrates and laminar profile of
31. Contreras D, Steriade M. Cellular basis of EEG slow rhythms: a sleep K-complex. Neuroscience. 1998;82:671–686.
study of dynamic corticothalamic relationships. J Neurosci. 53. Steriade M, Oakson G, Ropert N. Firing rates and patterns of mid-
1995;15:604–622. brain reticular neurons during steady and transitional states of the
32. Amzica F, Steriade M. Disconnection of intracortical synaptic sleep-waking cycle. Exp Brain Res. 1982;46:37–51.
linkages disrupts synchronization of a slow oscillation. J Neurosci. 54. Steriade M, Datta S, Paré D, et al. Neuronal activities in brain-stem
1995;15:4658–4677. cholinergic nuclei related to tonic activation processes in thalam-
33. Amzica F, Steriade M. Short- and long-range neuronal synchro- ocortical systems. J Neurosci. 1990;10:2541–2559.
nization of the slow (< 1 Hz) cortical oscillation. J Neurophysiol. 55. Hirsch JC, Fourment A, Marc ME. Sleep-related variations of
1995;73:20–38. membrane potential in the lateral geniculate body relay neurons of
34. Steriade M, Contreras D, Curró Dossi R, et al. The slow (< 1 Hz) the cat. Brain Res. 1983;259:308–312.
oscillation in reticular thalamic and thalamocortical neurons: sce- 56. Davis H, Davis PA, Loomis AL, et al. Electrical reactions of the
nario of sleep rhythm generation in interacting thalamic and neo- human brain to auditory stimulation during sleep. J Neurophysiol.
cortical networks. J Neurosci. 1993;13:3284–3299. 1939;6:500–514.
35. Amzica F, Steriade M. Electrophysiological correlates of sleep delta 57. Steriade M, Amzica F. Coalescence of sleep rhythms and their
waves. Electroencephalogr Clin Neurophysiol. 1998;107:69–83. chronology in corticothalamic networks. Sleep Res Online.
36. Amzica F, Neckelmann D. Membrane capacitance of cortical neu- 1998;1:1–10.
rons and glia during sleep oscillations and spike-wave seizures. J 58. Ingvar DH, Sjölund B, Ardo A. Correlation between dominant
Neurophysiol. 1999;82:2731–2746. EEG frequency, cerebral oxygen uptake and blood flow.
37. Amzica F, Massimini M. Glial and neuronal interactions during Electroencephalogr Clin Neurophysiol. 1976;41:268–276.
slow wave and paroxysmal activities in the neocortex. Cereb 59. Saunders MG, Westmoreland BF. The EEG in evaluation of disor-
Cortex. 2002;12:1101–1113. ders affecting the brain diffusely. In: Current Practice of Clinical
38. Amzica F, Massimini M, Manfridi A. Spatial buffering during slow Electroencephalography, Klass DW, Daly DD, eds. New York: Raven
and paroxysmal sleep oscillations in cortical networks of glial cells Press; 1979;343–379.
in vivo. J Neurosci. 2002;22:1042–1053. 60. Buzsáki G. Rhythms of the Brain. Oxford University Press; 2006.
60 Part I ■ Basic Principles

61. Brazier MAB. Studies of the EEG activity of limbic structures in 83. Alonso A, Garcia-Austt E. Neuronal sources of theta rhythm in
man. Electroencephalogr. Clin Neurophysiol. 1968;25:309–318. the entorhinal cortex of the rat. I. Laminar distribution of theta
62. Halgren E, Babb TL, Crandall PH. Human hippocampal forma- field potentials. Exp Brain Res. 1987;67:493–501.
tion EEG desynchronizes during attentiveness and movement. 84. Alonso A, Garcia-Austt E. Neuronal sources of theta rhythm in
Electroencephalogr Clin Neurophysiol. 1978;44:778–781. the entorhinal cortex of the rat. II. Phase relations between unit
63. Arnolds DE, Lopes da Silva FH, Aitink JW, et al. The spectral prop- discharges and theta field potentials. Exp Brain Res.
erties of hippocampal EEG related to behaviour in man. 1987;67:502–509.
Electroencephalogr. Clin Neurophysiol. 1980;50:324–328. 85. Boeijinga PH, Lopes da Silva FH. Differential distribution of beta
64. Kahana MJ, Sekuler R, Caplan JB, et al. Human theta oscillations and theta EEG activity in the entorhinal cortex of the cat. Brain
exhibit task dependence during virtual maze navigation. Nature. Res. 1988;448:272–286.
1999;399:781–784. 86. Vanderwolf CH, Buzsáki G, Cain DP, et al. Neocortical and hip-
65. Ekstrom AD, Caplan JB, Ho E, et al. Human hippocampal theta pocampal electrical activity following decapitation in the rat.
activity during virtual navigation. Hippocampus. 2005;15: Brain Res. 1988;451:340–344.
881–889. 87. Vanderwolf CH, Leung LS. Hippocampal rhythmical slow activ-
66. Bódizs R, Kántor S, Szabó G, et al. Rhythmic hippocampal slow ity: a brief history and effects entorhinal lesions and phencyci-
oscillation characterizes REM sleep in humans. Hippocampus. dine. In: Seifert W, ed. The Neurobiology of the Hippocampus.
2001;11:747–753. London: Academic Press; 1983:275–302.
67. Tesche CD, Karhu J. Theta oscillations index human hippocampal 88. Montgomery SM, Betancur MI, Buzsáki G. Behavior-dependent
activation during a working memory task. Proc Natl Acad Sci USA. coordination of multiple theta dipoles in the hippocampus. J
2000;97:919–924. Neurosci. 2009;29:1381–1394.
68. Cornwell BR, Johnson LL, Holroyd T, et al. Human hippocampal 89. Stewart M, Fox SE. Do septal neurons pace the hippocampal
and parahippocampal theta during goal-directed spatial naviga- theta rhythm? Trends Neurosci. 1990;13:163–168.
tion predicts performance on a virtual Morris water maze. J 90. Leung LW. Model of gradual phase shift of theta rhythm in the
Neurosci. 2008;28:5983–5990. rat. J Neurophysiol. 1984;52:1051–1065.
69. Stewart M, Fox SE. Hippocampal theta activity in monkeys. Brain 91. Chapman CA, Lacaille JC. Cholinergic induction of theta-fre-
Res. 1991;538:59–63. quency oscillations in hippocampal inhibitory interneurons
70. Green JD, Arduini AA. Hippocampal electrical activity in arousal. and pacing of pyramidal cell firing. J Neurosci. 1999;19:
J Neurophysiol. 1954;17:533–557. 8637–8645.
71. Buzsáki G. The hippocampo-neocortical dialogue. Cereb Cortex. 92. Leung LS. Generation of theta and gamma rhythms in the hip-
1996;6:81–92. pocampus. Neurosci. Biobehav Rev. 1998;22:275–290.
72. Witter MP, Naber PA, van HT, et al. Cortico-hippocampal com- 93. Buzsáki G, Czopf J, Kondákor I, et al. Laminar distribution of
munication by way of parallel parahippocampal-subicular path- hippocampal rhythmic slow activity (RSA) in the behaving rat:
ways. Hippocampus. 2000;10:398–410. current-source density analysis, effects of urethane and atropine.
73. Buzsáki G. Theta oscillations in the hippocampus. Neuron. Brain Res. 1986;365:125–137.
2002;33:325–340. 94. Soltesz I, Deschênes M. Low- and high-frequency membrane
74. Leung LW, Borst JG. Electrical activity of the cingulate cortex. I. potential oscillations during theta activity in CA1 and CA3
Generating mechanisms and relations to behavior. Brain Res. pyramidal neurons of the rat hippocampus under ketamine-
1987;407:68–80. xylazine anesthesia. J Neurophysiol. 1993;70:97–116.
75. Chapman CA, Lacaille JC. Intrinsic theta-frequency membrane 95. Somogyi P, Klausberger T. Defined types of cortical interneurone
potential oscillations in hippocampal CA1 interneurons of stra- structure space and spike timing in the hippocampus. J Physiol.
tum lacunosum-moleculare. J Neurophysiol. 1999;81:1296–1307. 2005;562:9–26.
76. Bilkey DK, Heinemann U. Intrinsic theta-frequency membrane 96. Banks MI, White JA, Pearce RA. Interactions between
potential oscillations in layer III/V perirhinal cortex neurons of distinct GABA(A) circuits in hippocampus. Neuron. 2000;25:
the rat. Hippocampus. 1999;9:510–518. 449–457.
77. Petsche H, Stumpf C, Gogolak G. The significance of the rabbit’s 97. Wendling F, Bartolomei F, Bellanger JJ, et al. Epileptic fast activ-
septum as a relay station between the midbrain and the hippocam- ity can be explained by a model of impaired GABAergic dendritic
pus. The control of hippocampus arousal activity by septum cells. inhibition. Eur J Neurosci. 2002;15:1499–1508.
Electroencephalogr. Clin Neurophysiol. 1962;14:202–211. 98. Fuentealba P, Tomioka R, Dalezios Y, et al. Rhythmically active
78. Petsche H, Gogolak G, van Zwieten PA. Rhythmicity of septal cell dis- enkephalin-expressing GABAergic cells in the CA1 area of the
charges at various levels of reticular excitation. Electroencephalogr. hippocampus project to the subiculum and preferentially inner-
Clin Neurophysiol. 1965;19:25–33. vate interneurons. J Neurosci. 2008;28:10017–10022.
79. Freund TF, Antal M. GABA-containing neurons in the septum 99. Kamondi A, Acsady L, Wang XJ, et al. Theta oscillations in
control inhibitory interneurons in the hippocampus. Nature. somata and dendrites of hippocampal pyramidal cells in vivo:
1988;336:170–173. activity-dependent phase-precession of action potentials.
80. Vertes RP, Kocsis B. Brainstem-diencephalo-septohippocampal Hippocampus. 1998;8:244–261.
systems controlling the theta rhythm of the hippocampus. 100. Wulff P, Ponomarenko AA, Bartos M, et al. Hippocampal theta
Neuroscience. 1997;81:893–926. rhythm and its coupling with gamma oscillations require fast
81. Buzsáki G, Leung LW, Vanderwolf CH. Cellular bases of hip- inhibition onto parvalbumin-positive interneurons. Proc Natl
pocampal EEG in the behaving rat. Brain Res. 1983;287: Acad Sci USA. 2009;106:3561–3566.
139–171. 101. Klausberger T, Somogyi P. Neuronal diversity and temporal
82. Mitchell SJ, Ranck JB Jr. Generation of theta rhythm in medial dynamics: the unity of hippocampal circuit operations. Science.
entorhinal cortex of freely moving rats. Brain Res. 1980;189:49–66. 2008;321:53–57.
Chapter 3 ■ Cellular Substrates of Brain Rhythms 61

102. Sik A, Penttonen M, Ylinen A, et al. Hippocampal CA1 interneu- 122. McCormick DA, Prince DA. Actions of acetylcholine in the
rons: an in vivo intracellular labeling study. J Neurosci. guinea-pig and cat medial and lateral geniculate nuclei, in vitro.
1995;15:6651–6665. J Physiol. 1987;392:147–165.
103. Toth K, Freund TF, Miles R. Disinhibition of rat hippocampal 123. Hu B, Steriade M, Deschênes M. The effects of brainstem peri-
pyramidal cells by GABAergic afferents from the septum. J brachial stimulation on neurons of the lateral geniculate nucleus.
Physiol. 1997500(Pt 2):463–474. Neuroscience. 1989;31:13–24.
104. Rotstein HG, Pervouchine DD, Acker CD, et al. Slow and fast 124. Hu B, Steriade M, Deschênes M. The effects of brainstem peri-
inhibition and an H-current interact to create a theta rhythm in brachial stimulation on perigeniculate neurons: the blockage of
a model of CA1 interneuron network. J Neurophysiol. spindle waves. Neuroscience. 1989;31:1–12.
2005;94:1509–1518. 125. Morison RS, Bassett DL. Electrical activity of the thalamus and
105. Berger H. Über das Elektroenkephalogramm des Menschen. 1st basal ganglia in decorticate cats. J Neurophysiol. 1945;8:09–314.
report. Arch Psychiat Nervenkr. 1929;87:527–570. 126. Steriade M, Llinás RR. The functional states of the thalamus and
106. Niedermeyer E. The normal EEG of the waking adult. In: the associated neuronal interplay. Physiol Rev. 1988;68:649–742.
Niedermeyer E,Lopes da Silva FH, eds. Electroencephalography: 127. Steriade M. Cellular substrates of brain rhythms. In:
Basic Principles, Clinical Applications, and Related Fields. Niedermeyer E, Lopes da Silva FH, eds. Electroencephalography:
Baltimore: Lippincott, Williams & Wilkins; 2005:167–192. Basic Principles, Clinical Applications and Related Fields.
107. Adrian ED, Mathews BHC. The interpretation of potential waves Baltimore: Lippincott Williams & Wilkins; 2004:31–84.
in the cortex. J Physiol. 1934;81:440–471. 128. Jones EG. The Thalamus. New York: Plenum; 1985.
108. Creutzfeld O, Grünvald G, Simonova O, et al. Changes of the 129. Domich L, Oakson G, Deschênes M, et al. Thalamic and cortical
basic rhythms of the EEG during the performance of mental and spindles during early ontogenesis in kittens. Brain Res.
visuomotor tasks. In: Evans CR, Mulholland TB, eds. Attention in 1987;428:140–142.
Neurophysiology. London: Butterworth; 1969;148–168. 130. Steriade M, Deschênes M, Domich L, et al. Abolition of spindle
109. Ray WJ, Cole HW. EEG alpha activity reflects attentional oscillations in thalamic neurons disconnected from nucleus
demands, and beta activity reflects emotional and cognitive reticularis thalami. J Neurophysiol. 1985;54:1473–1497.
processes. Science. 1985;228:750–752. 131. Steriade M, Domich L, Oakson G, et al. The deafferented reticu-
110. Rihs TA, Michel CM, Thut G. Mechanisms of selective inhibition lar thalamic nucleus generates spindle rhythmicity. J
in visual spatial attention are indexed by alpha-band EEG syn- Neurophysiol. 1987;57:260–273.
chronization. Eur J Neurosci. 2007;25:603–610. 132. Steriade M, Parent A, Hada J. Thalamic projections of nucleus
111. Rihs TA, Michel CM, Thut G. A bias for posterior alpha-band reticularis thalami of cat: a study using retrograde transport of
power suppression versus enhancement during shifting versus horseradish peroxidase and fluorescent tracers. J Comp Neurol.
maintenance of spatial attention. Neuroimage. 2009;44:190–199. 1984;229:531–547.
112. Andersen P, Andersson SA. Physiological Basis of the Alpha 133. Velayos JL, Jimenez-Castellanos J Jr, Reinoso-Suarez F.
Rhythm. New York: Appleton-Century-Crofts; 1968. Topographical organization of the projections from the reticular
113. Lopes da Silva FH, Van Lierop THMT, Schrijer CFM, et al. thalamic nucleus to the intralaminar and medial thalamic nuclei
Organization of thalamic and cortical alpha rhythm: spectra and in the cat. J Comp Neurol. 1989;279:457–469.
coherences. Electroencephalogr Clin Neurophysiol. 134. Paré D, Steriade M, Deschênes M, et al. Physiological character-
1973;35:627–639. istics of anterior thalamic nuclei, a group devoid of inputs from
114. Lopes da Silva FH, Storm Van Leeuwen W. The cortical alpha reticular thalamic nucleus. J Neurophysiol. 1987;57:1669–1685.
rhythm in dog: depth and surface profile of phase. In: Brazier 135. Wang XJ, Rinzel J. Spindle rhythmicity in the reticularis thalami
MAB, Petsche H, eds. Architecture of the Cerebral Cortex, IBRO nucleus: synchronization among mutually inhibitory neurons.
monograph series, Vol. 3. New York: Raven Press; 1978;319–333. Neuroscience. 1993;53:899–904.
115. Lopes da Silva FH, Vos JE. Mooibroek J, et al. Relative contribu- 136. Destexhe A, Contreras D, Sejnowski TJ, et al. A model of spindle
tions of intracortical and thalamo-cortical processes in the gen- rhythmicity in the isolated thalamic reticular nucleus. J
eration of alpha rhythms, revealed by partial coherence analysis. Neurophysiol. 1994;72:803–818.
Electroencephalogr Clin Neurophysiol. 1980;50:449–456. 137. Golomb D, Wang XJ, Rinzel J. Synchronization properties of
116. Bollimunta A, Chen Y, Schroeder CE, et al. Neuronal mechanisms spindle oscillations in a thalamic reticular nucleus model. J
of cortical alpha oscillations in awake-behaving macaques. J Neurophysiol. 1994;72:1109–1126.
Neurosci. 2008;28:9976–9988. 138. Deschênes M, Paradis M, Roy JP, et al. Electrophysiology of neu-
117. Flint AC, Connors BW. Two types of network oscillations in neo- rons of lateral thalamic nuclei in cat: resting properties and burst
cortex mediated by distinct glutamate receptor subtypes and discharges. J Neurophysiol. 1984;51:1196–1219.
neuronal populations. J Neurophysiol. 199675:951–957. 139. Yen CT, Conley M, Hendry SH, et al. The morphology of physi-
118. Hughes SW, Lörincz M, Cope DW, et al. Synchronized oscilla- ologically identified GABAergic neurons in the somatic sensory
tions at alpha and theta frequencies in the lateral geniculate part of the thalamic reticular nucleus in the cat. J Neurosci.
nucleus. Neuron. 2004;42:253–268. 1985;5:2254–2268.
119. Hughes SW, Crunelli V. Thalamic mechanisms of EEG alpha 140. Williamson AM, Ohara PT, Ralston DD, et al. Analysis of gamma-
rhythms and their pathological implications. Neuroscientist. aminobutyric acidergic synaptic contacts in the thalamic reticular
2005;11:357–372. nucleus of the monkey. J Comp Neurol. 1994;349:182–192.
120. Lörincz ML, Crunelli V, Hughes SW. Cellular dynamics of 141. Landisman CE, Long MA, Beierlein M, et al. Electrical synapses
cholinergically induced alpha (8-13 Hz) rhythms in sensory thal- in the thalamic reticular nucleus. J Neurosci. 2002;22:1002–1009.
amic nuclei in vitro. J Neurosci. 2008;28:660–671. 142. Yen CT, Jones EG. Intracellular staining of physiologically identi-
121. Steriade M. Neuronal Substrates of Sleep and Epilepsy. Cambridge: fied neurons and axons in the somatosensory thalamus of the cat.
University Press; 2003. Brain Res. 1983;280:148–154.
62 Part I ■ Basic Principles

143. Steriade M, Deschênes M. The thalamus as a neuronal oscillator. 164. Murthy VN, Fetz EE. Coherent 25- to 35-Hz oscillations in the
Brain Res. 1984;320:1–63. sensorimotor cortex of awake behaving monkeys. Proc Natl Acad
144. von Krosigk M, Bal T, McCormick DA. Cellular mechanisms of a Sci USA. 1992;89:5670–5674.
synchronized oscillation in the thalamus. Science. 1993;261: 165. Bouyer JJ, Montaron MF, Vahnee JM, et al. Anatomical localization
361–364. of cortical beta rhythms in cat. Neuroscience. 1987;22:863–869.
145. Bal T, von KM, McCormick DA. Role of the ferret perigeniculate 166. Freeman WJ. Mass Action in the Nervous System. New York:
nucleus in the generation of synchronized oscillations in vitro. Academic Press; 1975.
J Physiol. 1995;483(Pt 3):665–685. 167. Eckhorn R, Bauer R, Jordan W, et al. Coherent oscillations: a mech-
146. Bal T, von KM, McCormick DA. Synaptic and membrane mech- anism of feature linking in the visual cortex? Multiple electrode
anisms underlying synchronized oscillations in the ferret lateral and correlation analyses in the cat. Biol Cybern. 1988;60:121–130.
geniculate nucleus in vitro. J Physiol. 1995;483(Pt 3):641–663. 168. Gray CM, Konig P, Engel AK, et al. Oscillatory responses in cat
147. Contreras D, Destexhe A, Sejnowski TJ, et al. Control of spa- visual cortex exhibit inter-columnar synchronization which
tiotemporal coherence of a thalamic oscillation by corticothala- reflects global stimulus properties. Nature. 1989;338:334–337.
mic feedback. Science. 1996;274:771–774. 169. Gray CM, Singer W. Stimulus-specific neuronal oscillations in
148. Contreras D, Destexhe A, Sejnowski TJ, et al. Spatiotemporal orientation columns of cat visual cortex. Proc Natl Acad Sci USA.
patterns of spindle oscillations in cortex and thalamus. J Neurosci. 1989;86:1698–1702.
1997;17:1179–1196. 170. Engel AK, Konig P, Gray CM, et al. Stimulus-dependent neuronal
149. Jahnsen H, Llinás R. Electrophysiological properties of guinea-pig oscillations in cat visual cortex: inter-columnar interaction as
thalamic neurones: an in vitro study. J Physiol. 1984;349:205–226. determined by cross-correlation analysis. Eur J Neurosci.
150. Mulle C, Steriade M, Deschênes M. The effects of QX314 on thal- 1990;2:588–606.
amic neurons. Brain Res. 1985;333:350–354. 171. Gray CM, Engel AK, Konig P, et al. Stimulus-dependent neuronal
151. McCormick DA. Functional properties of a slowly inactivating oscillations in cat visual cortex: receptive field properties and fea-
potassium current in guinea pig dorsal lateral geniculate relay ture dependence. Eur J Neurosci. 1990;2:607–619.
neurons. J Neurophysiol. 1991;66:1176–1189. 172. Rodriguez R, Kallenbach U, Singer W, et al. Short- and long-term
152. Budde T, Mager R, Pape HC. Different types of potassium out- effects of cholinergic modulation on gamma oscillations and
ward current in relay neurons acutely isolated from the rat lateral response synchronization in the visual cortex. J Neurosci.
geniculate nucleus. Eur J Neurosci. 1992;4:708–722. 2004;24:10369–10378.
153. Contreras D, Steriade M. Spindle oscillation in cats: the role of 173. Munk MH, Roelfsema PR, Konig P, et al. Role of reticular activa-
corticothalamic feedback in a thalamically generated rhythm. tion in the modulation of intracortical synchronization. Science.
J Physiol. 1996;490(Pt 1):159–179. 1996;272:271–274.
154. Kim U, Bal T, McCormick DA. Spindle waves are propagating 174. Niessing J, Ebisch B, Schmidt KE, et al. Hemodynamic signals
synchronized oscillations in the ferret LGNd in vitro. correlate tightly with synchronized gamma oscillations. Science.
J Neurophysiol. 1995;74:1301–1323. 2005;309:948–951.
155. Sanford AJ. A periodic basis for perception and action. In: 175. von der Malsburg C, Schneider W. A neural cocktail-party
Colquhoun WP, ed. Biological Rhythms and Human Perception. processor. Biol Cybern. 1986;54:29–40.
New York: Academic Press; 1971;179–209. 176. Steriade M, Amzica F. Intracortical and corticothalamic
156. Moruzzi G, Magoun HW. Brain stem reticular formation and coherency of fast spontaneous oscillations. Proc Natl Acad Sci
activation of the EEG. Electroenceph Clin Neurophysiol. 1949;1: USA. 1996;93:2533–2538.
445–473. 177. Steriade, M., Contreras, D., Amzica, F., and Timofeev, I.
157. McCormick DA, Prince DA. Acetylcholine induces burst firing in Synchronization of fast (30–40 Hz) spontaneous oscillations in
thalamic reticular neurones by activating a potassium conduc- intrathalamic and thalamocortical networks. J. Neurosci.
tance. Nature. 1986;319:402–405. 1996;16:2788–2808.
158. McCormick DA, Wang Z. Serotonin and noradrenaline excite 178. Roelfsema PR, Engel AK, Konig P, et al. Visuomotor integration
GABAergic neurones of the guinea-pig and cat nucleus reticu- is associated with zero time-lag synchronization among cortical
laris thalami. J Physiol. 1991;442:235–255. areas. Nature. 1997;385:157–161.
159. McCormick DA, Pape HC. Noradrenergic and serotonergic mod- 179. Fries P, Nikolic D, Singer W. The gamma cycle. Trends Neurosci.
ulation of a hyperpolarization-activated cation current in thala- 2007;30:309–316.
mic relay neurones. J Physiol. 1990;431:319–342. 180. Llinás RR, Grace AA, Yarom Y. In vitro neurons in mammalian
160. Buzsáki G, Bickford RG, Ponomareff G, et al. Nucleus basalis and cortical layer 4 exhibit intrinsic oscillatory activity in the 10- to
thalamic control of neocortical activity in the freely moving rat. 50-Hz frequency range. Proc Natl Acad Sci USA. 1991;88:
J Neurosci. 1988;8:4007–4026. 897–901.
161. Bremer F, Stoupel N, Van Reeth PC. Nouvelles recherches sur la 181. Nuñez A, Amzica F, Steriade M. Voltage-dependent fast
facilitation et l’inhibition des potentiels évoqués corticaux dans (20–40 Hz) oscillations in long-axoned neocortical neurons.
l’éveil réticulaire. Arch Ital Biol. 1960;98:229–247. Neuroscience. 1992a;51:7–10.
162. Lopes da Silva FH, van Rotterdam A, Storm van Leeuwen W, et al. 182. Steriade M, Curró Dossi R, Paré D, et al. Fast oscillations (20–40
Dynamic characteristics of visual evoked potentials in the dog. II. Hz) in thalamocortical systems and their potentiation by meso-
Beta frequency selectivity in evoked potentials and background pontine cholinergic nuclei in the cat. Proc Natl Acad Sci USA.
activity. Electroencephalogr Clin Neurophysiol. 1970;29:260–268. 1991;88:4396–4400.
163. Freeman WJ, van Dijk BW. Spatial patterns of visual cortical fast 183. Cunningham ET, LeVay S. Laminar and synaptic organization of
EEG during conditioned reflex in a rhesus monkey. Brain Res. the projection from the thalamic nucleus centralis to primary
1987;422:267–276. visual cortex in the cat. J Comp Neurol. 1986;254:66–77.
Chapter 3 ■ Cellular Substrates of Brain Rhythms 63

184. Chagnac-Amitai Y, Connors BW. Synchronized excitation and 199. Somjen GG, Trachtenberg M. Neuroglial as generator of extracel-
inhibition driven by intrinsically bursting neurons in neocortex. lular current. In: Speckmann EJ, Caspers H, eds. Origin of
J Neurophysiol. 1989;62:1149–1162. Cerebral Field Potentials. Stuttgart: Thieme; 1979:21–32.
185. Hösli L, Hösli E, Della BG, et al. Action of acetylcholine, mus- 200. Caspers H, Speckmann E-J, Lehmenkuler A. DC potentials of the
carine, nicotine and antagonists on the membrane potential of cerebral cortex: Seizure activity and changes in gas pressures. Rev
astrocytes in cultured rat brainstem and spinal cord. Neurosci Physiol Biochem Pharmacol. 1987;106:127–178.
Lett. 1988;92:165–170. 201. Heinemann U, Walz W. Contributions of potassium currents and
186. Seigneur J, Kroeger D, Nita DA, et al. Cholinergic action on cor- glia to slow potential shifts (SPSs). In: Laming PR, Sykova E,
tical glial cells in vivo. Cereb Cortex. 2006;16:655–668. Reichenbach A, et al., eds. Glial Cells: Their Role in Behaviour.
187. Roopun AK, Middleton SJ, Cunningham MO, et al. A beta2- Cambridge: Cambridge University Press; 1998:197–209.
frequency (20–30 Hz) oscillation in nonsynaptic networks of 202. Amzica F, Steriade M. Neuronal and glial membrane potentials
somatosensory cortex. Proc Natl Acad Sci USA. 2006;103: during sleep and paroxysmal oscillations in the neocortex. J
15646–15650. Neurosci. 2000;20:6648–6665.
188. Mandema JW, Danhof M. Electroencephalogram effect measures 203. Tschirgi RD, Taylor JL. Slowly changing bioelectric potentials
and relationships between pharmacokinetics and pharmacody- associated with the blood-brain barrier. Am J Physiol.
namics of centrally acting drugs. Clin Pharmacokinet. 1992;23: 1958;195:7–22.
191–215. 204. Held D, Fencl V, Pappenheimer JR. Electric potential of cere-
189. Salenius S, Salmelin R, Neuper C, et al. Human cortical 40 Hz brospinal fluid. J Neurophysiol. 1964;27:942–959.
rhythm is closely related to EMG rhythmicity. Neurosci Lett. 205. Kjällquist Å, Siesjö BK. Regulation of CSF pH - influence of the
1996;213:75–78. CSF/plasma potential. Scand J Clin Lab Invest Suppl. 1968;102:I:C.
190. Salenius S, Portin K, Kajola M, et al. Cortical control of human 206. Sorensen SC, Severinghaus JW. Effect of cerebral acidosis on the
motoneuron firing during isometric contraction. J Neurophysiol. CSF-blood potential difference. Am J Physiol. 1970;219:68–71.
1997;77:3401–3405. 207. Hornbein TF, Sorensen SC. dc Potential difference between dif-
191. Gross J, Tass PA, Salenius S, et al. Cortico-muscular synchroniza- ferent cerebrospinal fluid sites and blood in dogs. Am J Physiol.
tion during isometric muscle contraction in humans as revealed 1972;223:415–418.
by magnetoencephalography. J Physiol. 2000;527(Pt 3):623–631. 208. Woody CD, Marshall WH, Besson JM, et al. Brain potential shift
192. Hari R. Action-perception connection and the cortical mu with respiratory acidosis in the cat and monkey. Am J Physiol.
rhythm. Prog Brain Res. 2006;159:253–260. 1970;218:275–283.
193. Pfurtscheller G, Neuper C, Andrew C, et al. Foot and hand area 209. Revest PA, Jones HC, Abbott NJ. The transendothelial DC poten-
mu rhythms. Int J Psychophysiol. 1997;26:121–135. tial of rat blood-brain barrier vessels in situ. Adv Exp Med Biol.
194. Chen R, Yaseen Z, Cohen LG, et al. Time course of corticospinal 1993;331:71–74.
excitability in reaction time and self-paced movements. Ann 210. Voipio J, Tallgren P, Heinonen E, et al. Millivolt-scale DC shifts in
Neurol. 1998;44:317–325. the human scalp EEG: evidence for a nonneuronal generator.
195. Pfurtscheller G, Neuper C, Kalcher J. 40-Hz oscillations during J Neurophysiol. 2003;89:2208–2214.
motor behavior in man. Neurosci Lett. 1993;164:179–182. 211. Nita DA, Vanhatalo S, Lafortune FD, et al. Nonneuronal origin of
196. Vanhatalo S, Tallgren P, Andersson S, et al. DC-EEG discloses CO 2-related DC EEG shifts: an in vivo study in the cat.
prominent, very slow activity patterns during sleep in preterm J Neurophysiol. 2004;92:1011–1022.
infants. Clin Neurophysiol. 2002;113:1822–1825. 212. Speckmann E-J, Elger CE. Introduction to the neurophysiologi-
197. Vanhatalo S, Holmes MD, Tallgren P, et al. Very slow EEG cal basis of the EEG and DC potentials. In: Niedermeyer E,Lopes
responses lateralize temporal lobe seizures: an evaluation of non- da Silva R, eds. Electroencephalography: Basic Principles, Clinical
invasive DC-EEG. Neurology. 2003;60:1098–1104. Applications, and Related Fields. Baltimore: Lippincott, Williams
198. Somjen GG. Electrogenesis of sustained potentials. Prog & Wilkins; 2005:17–30.
Neurobiol. 1973;1:201–237.
CHAPTER
Dynamics of EEGs as Signals of
Neuronal Populations: Models and
Theoretical Considerations
4
FERNANDO H. LOPES DA SILVA

MODELS OF EEG GENERATION— tions, whether by way of chemical synapses or electrical cou-
INTRODUCTION plings; and (d) the intrinsic ionic and synaptic currents.
There are both inhibitory and excitatory feedback loops. The
In Chapters 2 and 3 the neurophysiological basis of the elec- terms negative feedback and positive feedback are often used in a
troencephalogram (EEG) was discussed with special emphasis rather loose way to characterize the interactions between popula-
on the phenomena at the cellular level. In Chapter 5, the bio- tions of neurons. However, the sign of feedback is defined in an
physical aspects of the generation of EEG phenomena are con- exact way by comparing the overall gain modulus of the system
sidered mainly in terms of volume conduction, but the with feedback (closed loop gain: |Yo(j)|) and without feedback
dynamic properties of EEG patterns are only briefly men- (open loop gain: |Y1(j)|). The feedback is said to be negative if
tioned. These dynamic properties, however, are essential for |Yo(j)| |Y1(j)| and positive in the opposite case (2). The exact
understanding EEG phenomena. In this chapter, EEG signals sign of feedback in the interaction between neuron populations is
are considered as the result of the dynamic behavior of neu- in most cases not known. It therefore seems preferable to use the
ronal populations as revealed by model studies. According to terms excitatory and inhibitory feedback to define the main synap-
this perspective, it is necessary to integrate experimental and tic type of interaction between neuron populations. This will be
theoretical results. The latter must be obtained using models of considered from the reference point of the “main” population,
neuronal networks and corresponding computer simulations. which usually is the population that receives the input from
The mathematical treatment of such models has been avoided another source and/or sends its output to another structure.
here; the interested reader is referred to a number of other In this chapter, models of the generation of characteristic
publications that are cited for their thorough mathematical types of EEGs, including the spatial spread of rhythmic activity
treatment of these areas. over the cortex, and nonlinear dynamics as a new approach for
The fundamental assumption on which the following dis- understanding EEG phenomena are presented.
cussion is based is that EEG signals reflect the dynamics of elec- The processes of generation of EEG signals in a large number
trical activity in populations of neurons. A property of such of neurons forming a neural mass are complex. Therefore, it is
populations that is of utmost importance for the generation of useful to use both analytic and synthetic approaches to under-
EEG signals is the capacity of the neurons to work in synchrony. stand such processes. It is essential to use both approaches in a
This depends on the connectivity between the neurons that dialectic way. The experimental data, obtained through histolog-
form a network. The classic terminology of Freeman (1) pro- ical and physiological analytic methods, must be put together just
vides a useful systematization. Populations of neurons with as pieces of a puzzle are assembled. Very often the relationship
mutual interactions are called KI sets; the interactions may be between the different pieces is still unclear. It is in this context
excitatory (KIe) or inhibitory (KIi). Groups of two interacting that a synthetic approach may be most helpful. The point is then
populations of neurons are called KII sets; they are formed by to put together the available data in such a way as to form the
an interaction of a KIe set with a KIt set. Further, KIII sets most likely pattern. In this way, a model of a neurophysiological
formed by the interaction between two KII sets can be defined. system can be constructed. Such a model can help to advance
A number of such KIII sets constitute a neural mass that may knowledge of the system in two respects. First, it provides the
occupy a few square millimeters of cortical surface or a few possibility for testing the influence of different types of inputs or
cubic millimeters of nuclear volume in the brainstem or spinal of changing some of the properties of the constituting elements
cord. Typically, a neural mass consists of approximately 104 to upon the output of the system. Second, it allows the formulation
107 neurons. An example of a KIII set is given in a symbolic way of hypotheses concerning new elementary properties, relation-
in Figure 4.1. An essential feature of such a basic module is the ships, and overall behavior; it may thus predict new properties of
existence of multiple feedback loops. Such a set of neurons can the system, raise new questions, and suggest new experiments to
produce oscillatory phenomena as revealed in EEG signals. The explore these hypotheses. In this way, the model can constitute a
main parameters that characterize the dynamic behavior of the bridge between experimentation and theory. As Harmon (3)
set are (a) the synaptic time constants; (b) the length constants, stated long ago, it is in suggesting functional relationships
which define the distance of the interactions between different between the activity of single neurons and the behavior of groups
neurons; (c) the gain factors, that is, the strength of interac- of neurons that theoretical neurophysiology may be most potent.
65
66 Part I ■ Basic Principles

the generation of the main properties of EEG/MEG


(electro/magnetoencephalographic) signals, we pay special atten-
tion to macroscopic models. It should be emphasized that the
brain is a very complex system, and even one cortical column
consists of a large number of elements, neurons, ionic channels,
synapses, neurotransmitter systems, and glial cells that require
enormous amount of parameters and state variables for a proper
description. The dynamics of EEG/MEG signals, however, can be
accounted for within the framework of systems operating within
spaces with much lower dimensions. This is the realm of macro-
scopic models, namely neural mass models (NMMs) that use
mean field concepts, where the properties of subpopulations of
neurons are lumped together based on the fact that many neu-
rons within a given neuronal system share basic properties.
Nonetheless we also consider, albeit in abbreviated form, some
results of studies using microscopic models that are also relevant
for understanding some characteristics of EEG/MEG signals. In
this respect the models developed by Edelman and collaborators
(4) are good examples of models that combine detailed proper-
ties of neurons and synapses within large-scale systems. Although
these models have been introduced mainly to study functional
connectivity at different scales of complexity with respect to neu-
rocognitive processes, their capacity to generate brain rhythms
and EEG-like phenomena should also be stressed. It is to be
expected that the applications of this new generation of large-
scale computer models of complex neuronal networks will grow
fast in the coming years, since these kinds of models can now be
realized by virtue of the current availability of powerful com-
puter systems. An ambitious project along this research line is the
Blue Brain project that has the aim of creating a synthetic mam-
malian brain by reverse-engineering, using IBM’s Blue Gene
supercomputer, in order to investigate the brain’s architectural
and functional principles underlying cognitive processes (5).
Figure 4.1 Lumped circuit diagram of the sets of neurons in the olfac-
Another new trend is the development of Topological Models
tory bulb (OB), the anterior olfactory nucleus (AON), and the prepyri-
of neuronal networks with their patterns of connectivity that
form cortex (PC). PON, primary olfactory nerve; LOT, lateral olfactory
can be studied using the mathematical theoretical approach of
tract; EC, external capsule; R, receptors of the nasal mucosa; gl,
“small-world” graphs. These are mathematical graphs that con-
glomeruli; P, periglomerular neuron; M, mitral neuron; G, granule cell;
sist of nodes that can be reached from every other by a small
E, superficial pyramidal cells of AON; I, inhibitory neuron; A, superficial
number of steps, which have been applied in a wide variety of
pyramidal neuron; B, short local interneuron; C, deep pyramidal neuron;
fields, from gene to social networks. Watts and Strogatz (6)
, excitation; , inhibition; L, latency. (Adapted from Freeman WJ.
classified such graphs according to two independent structural
Analytic techniques used in the search for the physiological basis of the
features, namely the clustering coefficient and average node-to-
EEG. In: Gevins A, Remond A, eds. Handbook of Electroencephalography
node path length. This approach is in essence an analytical tool
and Clinical Neurophysiology, vol. 1: Methods of Analysis of Brain
that can extract interesting parameters to characterize func-
Electrical and Magnetic Signals. Amsterdam: Elsevier; 1986.)
tional connectivity within networks of cortical neurons and
conditions determining neuronal synchrony (7,177) and to ana-
With these general objectives in mind, this chapter considers a lyze fMRI and MEG/EEG data (8), but it has not addressed
few models of the generation of EEG phenomena that have been explicitly the question of the generation of EEG/MEG activities.
elaborated in the past decades. We focus here on models of EEG
rhythmic activities both at the macroscopic and the microscopic
level. The former are simplified models in which detailed mech- MACROSCOPIC MODELS
anisms are kept to a minimum and the main properties of sub- OF EEG GENERATION
populations of neurons are lumped together. These models can,
however, be easily adapted to include more specific neuronal Basic Principles: The Wilson–Cowan Approach
properties and more complex connectivity. Taking into consider- At the macroscopic level we describe NMMs that are based on
ation that in this chapter we are particularly interested in the mean-field approximation approach. Under this heading we
dynamic models of neuronal networks that are responsible for consider two classes of models: (i) models that account for
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 67

time-dependent properties of neural populations and (ii) models average membrane potential. The latter was interpreted as the
that include, in addition, space-dependent properties. In gen- local EEG signal, which in this case consisted of a predominant
eral terms this approach was introduced in a systematic way in alpha rhythm. Later the analysis was extended to take into account
the Neurosciences by Wilson and Cowan (9) who proposed a a first approximation of the nonlinear behavior (11). In the sec-
theoretical framework that emphasizes not the properties of tion of alpha rhythms we describe these models in more detail.
individual cells but rather those of neuronal populations. A More recently these kinds of models were extended to account
basic assumption is that cortical neurons form a dense network for EEG frequency components other than the alpha rhythms.
consisting of, at least, two subpopulations that are intercon- In this context the model study of Jansen and Rit (12) marks an
nected either directly or by way of interneurons. The basic vari- important advance. These authors introduced a lumped-parame-
able is the proportion of neurons of each subpopulation that ter model of a cortical column consisting of a population of
are active at a given time, such that the relevant variable is not pyramidal cells interconnected with populations of excitatory and
the single spike but the average spike frequency. The dynamics inhibitory interneurons, which displays five kinds of simulated
of the neural mass depends on the interaction of excitatory and EEG outputs when fed with random noise. These different EEG
inhibitory neuronal subpopulations. Accordingly the model outputs are the following: low amplitude noise for high values of
requires the use of two variables E(t) and I(t), describing the the inhibitory feedback, slow periodic activity similar to that
activity of the excitatory and inhibitory subpopulations, respec- obtained in comatose patients, typical alpha rhythmic activity,
tively, to characterize the state of the whole population. noisy alpha activity, and low-amplitude high-frequency activity
Based on these assumptions the general Wilson and Cowan similar to beta activity for high values of the excitatory feedback. In
equations (9) governing the dynamics of a localized population order to explore how different cortical columns interact, Jansen
of neurons were derived: and Rit investigated the behavior of two identical single-column
t models and showed that they may produce in-phase oscillations.
E(t r) c1 E(t¿)dt¿d #Se Another case where NMMs of interacting neuronal popula-
tions helped to clarify the origin of a specific EEG/MEG phenom-
t r
t
enon was studied by Suffczynski et al. (13). This phenomenon is
the so-called focal ERD–surround ERS, described in detail in
e a (t t¿) [c1E(t¿) c2I(t¿) P(t¿) ] dt¿f Chapter 45, where a given event (e.g., finger movement) can cause
q at the same time a decrease (event-related desynchronization or
and ERD) and an increase (event-related synchronization or ERS) of
t EEG/MEG power within the alpha frequency range (mu rhythm)
I(t¿)dt¿d #Si
depending on the site over the scalp. The analysis of coupled
I(t r¿) c1
NMMs representing thalamocortical systems responsible for the
t r¿ hand and for the foot movements, respectively, was able to reveal
t
how this reciprocal “ERD–ERS” phenomenon takes place.
e a (t t¿) [c3E(t¿) c4I(t¿) Q(t¿) ] dt¿f . Based on the results obtained with NMMs generating rhyth-
q mic activities within the alpha frequency range, several
researchers extended these models to account for different EEG
This formalism describes the activities of the excitatory E(t) frequency bands besides the alpha rhythm. In this respect,
and inhibitory I(t) subpopulations; c1 and c2, and c3 and c4, are David and Friston (14) described a lumped model that gener-
the connectivity constants representing the average number of ates various rhythmic activities ranging from delta to gamma,
excitatory and inhibitory synapses per cell for each subpopula- according to the kinetics of the populations included in the
tion, is the decay time constant, Se(x) and Si (x) are the model. These authors also investigated the dynamics of the
response functions that give the expected proportion of cells in interaction between different cortical areas by coupling two
a subpopulation that would respond to a given level of excita- lumped models. Depending on coupling strength, direction of
tion and are represented by sigmoid functions; P(t) and Q(t) coupling (unidirectional or reciprocal coupling), and propaga-
are the external inputs to the excitatory and the inhibitory sub- tion delays, they showed that the oscillation frequency depends
populations, respectively; r is the refractory period. on the delay and that a reciprocal coupling is characterized by
oscillatory signals in phase or in antiphase. This latter result
Neural Mass Models of EEG Rhythms
reproduces the experimental finding of zero-lag synchroniza-
Based on the theoretical formalism introduced by Wilson and tion among remote cortical areas as found experimentally by
Cowan (9), Lopes da Silva et al. (10) described an NMM with the Roelfsema et al. (15) and Chawla et al. (16).
aim of simulating the conditions under which alpha rhythms With the aim of investigating how different cortical regions
occur. The model consisted of lumped subpopulations of inter- can display complex EEG frequency spectra and several pat-
connected excitatory and inhibitory neurons with properties of terns of functional connectivity, Zavaglia et al. (17,18) also
thalamocortical cells and a special subpopulation of interneurons. developed interconnected lumped models. They showed that
The mathematical analysis was initially carried out for a linearized three populations, consisting of four subpopulations (pyrami-
approximation of the neuronal network in order to express the dal neurons, excitatory, slow, and fast inhibitory interneurons),
transfer function of the network and the power spectrum of the arranged in parallel are sufficient to account for complex EEG
68 Part I ■ Basic Principles

spectra with dominant frequency ranging form theta to alpha Neural Mass Models w ith Spatial-Temporal
and gamma. This model is akin to the comprehensive NMM Features—Continuous Neural Field Models
introduced by Wendling et al. (19) to account for the generation In addition to the lumped NMMs described above, where spatial
of different kinds of neural activities and the corresponding aspects are not explicitly simulated, there are also models that
EEG signals, including epileptic activity as described in more include spatial properties and consist of spatiotemporal chains of
detail in the section dedicated to models of epileptic phenom- different subpopulations of neurons. An example is the model of
ena generated by neural populations. This model illustrates very van Rotterdam et al. (20) that consists of a series of pyramidal
clearly the main features of the currently used NMMs (Fig. 4.2). cells and interneurons interconnected by means of recurrent col-

Figure 4.2 (a): Neural mass model consisting of a subpopulation of principal cells (pyramidal cells) that project to and receive feedback from sub-
populations of local interneurons. Feedback to pyramidal cells is either excitatory (recurrent excitation) or inhibitory: dendritic-projecting interneu-
rons with slow synaptic kinetics—IPSP (slow)—shown in (c)—and somatic-projecting interneurons (gray hexagon) with faster synaptic
kinetics—IPSP (fast)—shown in (c). (b): Correspon ding block diagram representing for each subpopulation, the average pulse density of afferent
action potentials that is transformed into an average inhibitory or excitatory postsynaptic membrane potential using a linear dynamic transfer func-
tion with impulse response h EXC(t), h SDI(t), and h FSI(t) while this potential is converted into an average pulse density of action potentials fired by
the neurons by way of a static nonlinear function (asymmetric sigmoid curve, S(v)). Interactions between main cells and interneurons are repre-
sented by seven connectivity constants (C1 to C7), which account for the average number of synaptic contacts. One of the model outputs represents
the summated average postsynaptic potentials on pyramidal cells. It simulates an EEG signal. The three main parameters of the model respectively
correspond to the average excitatory synaptic gain (EXC), to the average slow inhibitory synaptic gain (SDI), and to the average fast inhibitory synap-
tic gain (FSI). (Adapted from Wendling F, Hernandez A, Bellanger JJ, et al. Interictal to ictal transition in human temporal lobe epilepsy: insights
from a computational model of intracerebral EEG. J Clin Neurophysiol. 2005;22(5):343–356.)
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 69

laterals and inhibitory fibers with connectivity functions Neural Mass Models and Hemodynamic Signals
assumed to be homogenous while the strength of the connectiv- NMMs are also being applied to make a bridge between the
ity was an exponentially decreasing function of distance. This activity of neuronal networks as reflected in EEG signals and
model was constructed to study the factors that determine the hemodynamic signals as measured by way of functional MRI, in
propagation of alpha waves along a cortical sheet. Assuming a order to understand better the working of cortical networks
continuous chain of neurons the model’s output, u(x,t), is the underlying cognitive processes. This constitutes a central feature
space-averaged membrane potential density of the main cells. To of dynamic causal modeling (DCM) (30) that has been applied
study propagation properties in the neuronal chain an expression not only to fMRI signals but also to electrophysiological signals
has been derived as a linear inhomogenous partial differential such as event-related potentials (ERPs) (31).
equation by means of which one can determine group velocity of Valdés-Sosa et al. (32) proposed an approach where the
alpha waves and dispersion properties. Alternatively the neuronal summed postsynaptic potentials generated by an NMM of a cor-
chain may be conceived as a two-dimensional system with a spe- tical area are used in the forward mode to estimate the primary
cific transfer function relating a random input p(x,t) to the out- current density distribution of a cortical area (Fig. 4.3). This is
put u(x,t). The model results were able to emulate experimental then transformed into the signal recorded at EEG sensors by
data, namely the propagation velocity that was found experimen- applying a linear spatial convolution with the EEG lead field, that
tally in the canine cortex to be about 0.3 m/sec (21) based on is, the function that relates any source in the brain to the corre-
parameters taken from physiology and histology, as explained sponding measurements at the EEG sensors. Simultaneously the
further in more detail in this chapter. summed postsynaptic potentials generated by the NMM are trans-
A series of other models, akin to that of van Rotterdam et al. formed into a local Vasomotor feed forward signal that is then fur-
(20), where the cortical neuronal populations are modeled as ther transformed by way of a temporal convolution with the
forming a continuum have been made, including models incor- hemodynamic response function (HRF) into a simulated BOLD
porating stochastic features called continuous neural field models. signal. In this way the information obtained from the EEG signal
In this chapter we focus on those aspects that are specifically rele- and from the corresponding fMRI BOLD signal recorded from the
vant for EEG/MEG generation, while the more general aspects of same region of interest can be fused (Fig. 4.3). We should note that
cortical processing of information with respect to neurocognitive although this approach opens interesting novel perspectives and
functions lie outside the scope of this text. With respect to these
continuous neural field models, in general, the reader may be
referred to comprehensive reviews of Harrison et al. (22), Deco et
al. (23), and Jirsa (24). Examples of continuous neural field models Forward Models for fusion of EEG/MEG & BOLD
signals based on a common Neural Mass Model
are those of Jirsa (24,25) and of the Australian group (26–29). Jirsa
(24) gives a very thorough account of how a set of state equations Neural Mass Model
with parameter values based on physiology can account for the generates an ensemble
of postsynaptic potentials
dynamics of cortical neuronal populations at different spatial
scales: microscopic, mesoscopic, and macroscopic. At microscopic
level, fast synaptic feedbacks appear to be crucial to the stability of
the excited cortex. At mesoscopic scale the model shows how fields Transfer function Transfer function
of synchrony can be established, and at macroscopic scale it shows Estimated cortical Vasomotor feedforward
how traveling waves may be generated in the cortex. The last two primary current density signal
levels are particularly relevant with respect to the dynamics of
EEG/MEG generation. With respect to these models the activity
state of a single neuron can be described by its membrane poten-
tial, V, the capacitive current, I, or the time since the last action
potential, T. Each of these variables can be represented by a EEG/MEG BOLD signal
dimension in the phase space of the neuron’s activity state.
Accordingly this phase space would be three-dimensional and the
state of a neuron would correspond to a point v = {V,I,T} or par- Figure 4.3 The output of a neural mass model in the form of summed
ticle in phase space. Using a general formalism the activity state of postsynaptic potentials can be used to estimate the primary current
a population of neurons at any time, t, can be described in phase density distribution of a cortical area that is transformed into the
space by a density p(v,t). As the state of each neuron evolves the EEG/ MEG signal. Simultaneously the summed postsynaptic potentials
ensemble density, p(v,t) evolves until it reaches some steady state. generated are transformed into a local Vasomotor feed-forward signal
The evolution of the ensemble density dynamics can be described that is then further transformed by way of a temporal convolution with
using the formalism of the Fokker–Planck equation. The latter the hemodynamic response function into a simulated BOLD signal. In
describes in general terms the time evolution of the probability this way EEG/ MEG information can be fused with the corresponding
density function of the position of a particle in a given multidi- fMRI BOLD signal recorded from the same region of interest. (Inspired
mensional space. The interested reader may find a thorough treat- from Valdes-Sosa PA, Sanchez-Bornot JM, Sotero RC, et al. Model
ment of the applications of this formalism in the study of driven EEG/ fMRI fusion of brain oscillations. Hum Brain Mapp.
continuous field models by Deco et al. (23) and Harrison et al. (22). 2009;30(9):2701–2721.)
70 Part I ■ Basic Principles

merits being further elaborated, it is based on concepts that are still the prepyriform cortex (PC). Excitation of the M cells by pri-
insufficiently established. Namely more research is needed to bet- mary olfactory nerve (PON) inputs leads to excitation of the
ter understand how neural activities, generated by neural popula- granule (G) neurons and to feedback inhibition of the M neu-
tions with complex geometrical configurations, are transformed rons. In consequence, the G neurons are disexcited and the M
into cortical current distributions on the one hand, and how they neurons are disinhibited. In this way, the M neurons may
are associated with BOLD signals on the other. become excited again, and the whole process may continue in
an oscillatory mode. The basic oscillation is at 40 Hz in cat and
60 to 80 Hz in rabbit (61). A similar type of inhibitory feedback
MODELS OF SPECIFIC EEG ACTIVITIES loop occurs in the PC, and the EEG of this cortical structure
Here we describe a number of models that account for the gen- resembles that of the olfactory bulb (OB) in frequency content.
eration of specific types of EEG activities, namely the following: In this model and similar ones, the action potentials are con-
verted into graded currents at the somadendritic synapses caus-
• Models describing the generation of the EEG of olfactory ing the postsynaptic potentials. In a first approximation, these
areas of the brain as proposed extensively by Freeman (1) and postsynaptic potentials may be considered to sum at the soma.
later further elaborated (33,34) that may be considered para- At the trigger zone, the axon hillock, these potentials are trans-
digmatic of neural mass modeling approaches; formed into spike trains depending on a threshold.
• Models of alpha rhythms of the thalamus and cortex, since The postsynaptic potentials may be considered as the wave
these are one of the most conspicuous features of the human (W) mode of operation of the neural set; the action potentials
EEG, as proposed by Lopes da Silva et al. (10,11) and elabo- represent the pulse (P) mode. This is, of course, a simplification
rated in more detail by van Rotterdam et al. (20), and of processes taking place at the membrane, but at the level of gen-
extended later by Jansen and Rit (12), Nunez (35), Wright and eralization at which NMMs of the EEG are constructed, these are
Liley (36), Stam et al. (37), Valdes et al. (38), Robinson et al. the main features that must be considered. The sensitivity of the
(39), Suffczynski et al. (13), David and Friston (14), and threshold process in the bulb and cortex is controlled by centrifu-
Zavaglia et al. (17,18); gal influences that originate in the forebrain and brainstem,
• Detailed models of membrane and synaptic properties of among others from the locus coeruleus (62). The conversion
thalamic cells and circuits responsible for the generation of between the postsynaptic potential (W mode) and the pulse rate
7- to 14-Hz spindle rhythmicity that occurs at the onset and (P mode) may be given in terms of the sigmoid curves shown in
in the light stages of sleep, based on the experimental findings Figure 4.4 for a single neuron and for the population or ensem-
of Steriade et al. (40–48); ble. The wave–pulse conversion can be considered as a static sig-
• Models of epileptiform EEG, namely of absence seizures, that moidal nonlinearity at the single neuron level. However, over
display complex nonlinear dynamics, namely bistable behav- small signal ranges this relation may be assumed to be linearized,
ior, where abrupt transitions occur between two states—nor- and therefore it may be represented by a simple coefficient. To
mal and paroxysmal (49,50)—and of epileptic seizures of the simulate the local dynamics of the EEG, delays due to synaptic
limbic cortex (19,51), as well as detailed models of the gener- and conduction processes may be accounted for by a stage of
ation of epileptiform transients by Traub (52), Traub and integration. Along these lines, the dynamics of these structures
Wong (53), Traub et al. (54,55), and Traub and Miles (56); may be represented by a mathematical model consisting of cou-
• Models of beta and gamma rhythms developed by Traub et al. pled differential equations that incorporate static nonlinear func-
(57) with realistic simulations of synapses (55,57–60). We do tions (for a mathematical treatment see Refs. 1, 34, and 63). The
not include here models of hippocampal theta rhythms since main input to the populations of the OB comes from the olfac-
the basic physiology of these rhythmic activities has already tory mucosa and is modulated by the respiration rate. A study of
been described in Chapter 3. the behavior of the KII set described above shows that this set has
multiple stable states. Two are of particular interest because they
Model of Olfactory Area EEG represent the most common situations. The first is a quasiequi-
The main neuronal populations of the olfactory brain struc- librium state that is maintained by a steady input of low intensity;
tures that are of importance for an understanding of the gener- the second is the so-called carrier state (34), characterized by a
ation of the olfactory EEG can be summarized as follows: in the limit cycle with quasisinusoidal EEG activity at 40 Hz (cat) or 60
bulb, the mitral and tufted cell (M) population, which is excita- to 80 Hz (rabbit) that occurs during inspiration. This nonlinear
tory, and the granule cell (G) population, which is inhibitory; in oscillation represents a limit cycle because the feedback gain is
the PC, the surface pyramidal population, which is excitatory displaced to the excitatory side of the resting value with an
(E), and the basket cell population, which is inhibitory (I). The increase in pulse input (either of sensory origin or depending on
interaction between the M and the granule neuron populations arousal level) that may drive the KII set out of the equilibrium
forms an inhibitory feedback loop. The interaction between the state. Certain drugs such as carbachol or picrotoxin can induce a
surface pyramidal and the basket cell populations in the PC also limit cycle state of long duration (34). A later section shall con-
forms an inhibitory feedback loop (Fig. 4.1). sider the relevance of nonlinear dynamics such as the theory of
As can be seen in Figure 4.1, there are also mutual excitatory deterministic chaos for understanding EEG generation. Here it is
interactions between M neurons in the bulb and between sufficient to observe that the EEG signals simulated by the set of
pyramidal A neurons in the PC, as well as mutual inhibitory nonlinear coupled differential equations that describe the KII set
interactions between granule cells in the bulb and basket cells in approximate the experimentally recorded EEGs.
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 71

rhythmic activities is the question of whether such rhythms are


caused by single cells with pacemaker properties. A significant
advance in this discussion was achieved when it was demon-
strated by Jahnsen and Llinás (70,71) that some types of thalamic
neurons display oscillatory behavior in vitro, even after blockage
of synaptic transmission. Most neurons studied in this way tend
to generate oscillations in the frequency range of 6 to 10 Hz.
These intrinsic oscillatory properties of some neurons are most
likely of importance in shaping the rhythmic behavior of the net-
works to which they belong. However, these properties may not
be sufficient to account for the network rhythmic behavior. One
main argument supports this statement: the disconnection of
cortically projecting thalamic cells from their inputs, which arise
in the reticular (RE) nucleus of the thalamus, abolishes spindles
in thalamocortical relay (TCR) neurons (72). However, single
TCR neurons may preserve their intrinsic membrane oscillatory
properties in vitro (70,71). Interestingly, the capacity of both
TCR–RE circuits and the isolated RE nucleus, in vivo, to generate
spindle oscillations contrasts with the fact that the RE nucleus
isolated in vitro does not show spontaneous oscillations (73–78).
Figure 4.4 Conversion operations between the wave mode (W) and This is likely because the input conditions of the RE neurons in
the pulse mode (P) for a neuron and an ensemble of neurons. At the vitro are different from the situation in vivo because the neurons
neuronal level both operations are time-varying (t); the P-W conversion lack neuromodulation influences, including cholinergic, nora-
is nonlinear; the W-P conversion is linear between threshold and catho- drenergic, and serotoninergic inputs. In modeling studies, the
dal block. At the ensemble level, the P-W conversion is restricted to a influence of neuromodulators on RE neurons membrane poten-
small linear range and W-P conversion is static and nonlinear. The tial was simulated (68), namely noradrenaline and serotonin, by
derivative of this curve defines the gain; this may change ( o) depend- blocking leak K currents and thus depolarizing the RE neurons
ing on arousal level. The P-W conversion can vary to represent synap- to the same level as observed in vivo. These studies showed that
tic changes occurring with learning. (Adapted from Freeman WJ. waxing–waning oscillations could occur in the RE network, sim-
Dynamics of image formation by nerve cell assemblies. In: Basar E, ilar to those seen in vivo. This finding provides clear evidence that
Flohr H, Haken H, et al., eds. Synergetics of the Brain. Berlin: Springer- the oscillatory behavior of these neuronal populations of RE neu-
Verlag; 1983:102–121.) rons depends on specific levels of the neuronal membrane poten-
tial that is affected by several inputs, both at short- and
long-range. Among the latter, the neuromodulatory inputs aris-
Alpha Rhythm Models: Spatial ing from the raphe nucleus (serotoninergic), the locus coeruleus
and Temporal Characteristics (noradrenergic), as well as glutamatergic projections form fore-
A current view in neurophysiology is that the origin of certain brain areas are particularly important. Among the former it is
rhythmic electrical activity seen in the EEG, such as that char- interesting to note that gap junctions have been shown to exist
acteristic of some thalamic nuclei and cortical areas after between RE nucleus (79) and thus may also participate in the
administration of a barbiturate (64,65) and possibly also that modulation of their oscillatory properties.
occurring spontaneously, such as sleep spindles and alpha To understand how oscillations may be generated in thala-
rhythms (66,67) is to be found in populations of neurons char- mic nuclei, we must consider the basic structure of these neu-
acterized by the interaction between excitatory and inhibitory ronal networks. In the thalamic relay nuclei where this type of
neuronal populations. Different types of models have been rhythmic activity can be recorded, namely the relay nuclei of
developed to account for the generation of EEG rhythmic activ- the thalamus, three main types of neurons can be distinguished
ities within the alpha frequency range. Some of these models (see also Chapter 3 for a discussion of the physiology of these
operate at the microscopic level of the single neuron or the cellular elements): the TCR neurons, whose axons project to the
small local network (68,69), others at the macroscopic level cortex; the RE nucleus neurons that contribute to the inhibitory
with the aim of accounting for global properties of EEG signals feedback control of the former; and the local, intrinsic, neu-
recorded from the scalp (not limited to alpha rhythms) (35). rons. The RE forms a thin sheet of neurons that surround the
Recently attempts have been made to combine the local and the anterior and lateral side of the thalamus and interact synapti-
global levels within a synthetic theoretical framework, among cally with the TCR neurons. The axons of the RE cells have as
others by Nunez who developed a thoughtful “marriage of local main target the TCR cells. Their dendrites make synaptic con-
and global theories” (Chapter 11 in Ref. 35). A particular inter- tacts with the axons of the TCR cells. Thus the TCR and the RE
esting discussion of this issue is the paper of Wright and Liley neurons are interconnected by means of a feedback loop as
(36) followed by a wide-open peer commentary. schematically shown in Figure 4.5. Both the RE and the local-
At the cellular level, an important issue that has been repea- circuit neurons contain aminobutyric acid (GABA) as trans-
tedly discussed in studies of the mechanisms underlying EEG mitter. This implies that the feedback of the RE on the TCR
72 Part I ■ Basic Principles

due to microphysiological investigations in thalamic slices, in


vitro, and in isolated brain preparations by the groups of Llinás
and collaborators (70,71). For a comprehensive account of
these membrane properties, the reader is referred to the origi-
nal publications and to the excellent reviews by Steriade and
Llinás (80), Steriade et al. (66,67), McCormick (81),
McCormick and Bal (82), Destexhe et al. (69), Destexhe and
Sejnowski (83), (84), Lörincz et al. (85), and to Chapter 3.
A basic question is whether the different types of oscillations
can be found in vitro in the absence of external inputs, since
this would give support to the idea that these thalamic cells may
support pacemaker rhythmic activity. Jahnsen and Llinás (70)
provided an initial answer to this question by showing that the
oscillatory activity at about 10 Hz may be observed in the
absence of external drive in vitro. However, we must also con-
sider what is the contribution of the feedback circuits between
TCR and RE neurons to the generation of EEG oscillations
in vivo.
In this context the most direct experimental evidence per-
tains to the generation of spindle oscillations occurring during
the early stages of sleep. Spindles appear in the EEG of most
mammals as oscillations with a frequency in the range from 7
Figure 4.5 Simplified scheme of a thalamic neural network where the
to 14 Hz, which form bursts lasting for 1.5 to 2 sec that recur
main features are presented. The thalamocortical relay (TCR), the retic-
periodically with an irregular periodicity of about 5 to 10 sec.
ular thalamic (RE), and the local-circuit interneurons (IN) are indicated,
This type of spindle can also be seen in animals under barbitu-
along with projections from a cholinergic population (ACh). The synap-
rate anesthesia. It was shown by Steriade et al. (72) that no thal-
tic triad formed by the dendrites of TCR and IN neurons with the spe-
amic nucleus is capable of generating spindle oscillations after
cific afferent (spec. aff.) fibers is also indicated. Three types of synapse
disconnection from the RE nucleus. Therefore, the existence of
are shown: excitatory, aminobutyric acid (GABA)ergic (inhibitory), and
feedback circuits including the RE nucleus is necessary for spin-
cholinergic. The effect of the latter on RE neurons is assumed to be an
dling to occur under normal conditions in vivo. Nonetheless
increase in potassium conductance (gk), whereas on the TCR neurons
there are other mechanisms that may be responsible for alpha
the effect is the opposite. (From Lopes da Silva FH. Neural mechanisms
rhythmic generation at least in brain slices in vitro but have not
underlying brain waves: from neural membranes to networks.
been incorporated in explicit models as described by Lörincz et
Electroencephalogr Clin Neurophysiol. 1991;79:81–93, with permission.)
al. (85) and in Chapter 3.
In conclusion, under the normal conditions of an intact
brain, both synaptic interactions and input signals play an
neurons is GABAergic and thus inhibitory. The RE neurons essential role in setting the conditions necessary for oscillatory
receive fast GABAA-mediated inhibitory postsynaptic potentials behavior to occur. The frequency of this oscillation depends on
(IPSPs) from neighboring RE neurons; in this way these neu- the intrinsic membrane properties on the membrane potential
rons form a chain of cells interconnected by inhibitory of the individual neurons and on the strength of the synaptic
synapses. In addition they receive fast glutamatergic (AMPA interactions.
type) excitatory postsynaptic potential (EPSPs) from TCR neu-
rons. In turn the TCR neurons receive both fast GABAA and Simplified Distributed Network Model
slow GABAB IPSPs from RE neurons (74,75). In a thalamic A simplified model has been proposed (10,11) that incorpo-
nucleus, a number of feedback circuits between TCR and RE rates the limited physiological and histological data available at
neurons can be distinguished. that time; these assumptions were sufficient to explain the gen-
In the context of this discussion, it is important to note that eration of the alpha rhythm, that is, the EEG stochastic signal
the TCR neurons can work essentially according to two distinct that lies in the frequency range of 8 to 13 Hz. This appears
modes: either (a) as relay cells that simply depolarize and can mainly at eye closure and can be recorded best from the poste-
produce single spikes in response to an adequate input volley or rior regions of the scalp in humans. A similar type of activity
(b) as oscillatory cells producing bursts of high-frequency has also been recorded at the visual cortex and some thalamic
spikes that are repeated in a rhythmic fashion. The resting nuclei in dogs (86) and cats (87,88). The model in its most sim-
membrane potential of a TCR neuron determines whether such ple form consists of a set of simulated neurons, thalamocortical
a neuron will be active in the relay or in the oscillatory mode. relay (TCR) cells, and interneurons (INs). The latter may be
To understand this question, it is necessary to take into account assumed to be the RE cells as shown in Figure 4.5, although the
the intrinsic membrane properties of these cells, namely their IN cells shown in this figure may have a complementary effect.
main ionic conductances and the corresponding dynamics. It For simplicity in the model, the cells providing feedback are
should be noted that these properties became known mainly denoted, in the following, as INs. In contrast with the lumped
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 73

models, some examples of which have been given for the olfac- tory period then followed. A basic characteristic of this model
tory system, this model is a distributed one in the sense that was that the input received by each TCR neuron was in the
each neuron is modeled individually; it occupies a specific posi- form of a series of pulses (action potentials) that had a
tion within a matrix of neurons. Such a distributed model Poisson distribution; the inputs to the individual TCR neu-
offers some special possibilities for theoretical studies that are rons were uncorrelated. The model’s output signal was the
not easily available in lumped models. sum of the membrane potential fluctuations of all TCR neu-
Without entering into great detail about the alpha rhythm rons. This output signal simulated the general spectral prop-
distributed model, it is of interest to point out some of its erties of an alpha rhythm. This result is illustrated in Figures
characteristics. First, the structure of the distributed model 4.6 through 4.8.
initially used (10) consisted of 144 TCR neurons and 36 INs; In the distributed model, it is also easy to investigate the
the 32 TCR neurons that surrounded one IN were responsible result of changing the range of excitatory and inhibitory influ-
for its excitation, while each IN was responsible for the inhi- ence of, respectively, TCRs and INs. In the case of 4.7, there
bition of the 12 TCRs around it. The excitation and inhibition were 12 TCR neurons that were inhibited by one IN. This is the
were represented by time functions that were an approxima- common situation; the effects of reducing this number to four
tion of the waveforms of EPSPs and IPSPs, respectively. The and of increasing it to 21 are shown in Figures 4.7 and 4.8.
matrix of neurons should be viewed as lying at the surface of Reduction of the inhibitory area of influence of an IN results
a torus. Furthermore, each neuron fired whenever the mem- in a decrease of the spectral peak amplitude (alpha frequency)
brane potential exceeded a simple threshold; a short refrac- and an increase in bandwidth; an increase in the inhibitory
area has the opposite effects. These are some of the phenom-
ena that can be studied using a distributed neuronal model of
rhythmic activity. However, this type of model has one main
disadvantage: it is difficult to treat analytically. Therefore, it
was necessary to develop a lumped model that would take into
account the main characteristics of the distributed model. A
lumped model of the same type as those of Freeman, previ-
ously described, permits a more general treatment of the acti-
vity of neuronal populations.

The Neural Mass (Lumped) Alpha Rhythm Model


The development of the lumped model in analytical terms
included the step of translating the properties of the discrete

Figure 4.6 Results obtained with the distributed model of alpha Figure 4.7 Output signals of the distributed alpha model: Vm (t) repre-
rhythm. Upper panel: Input density of the input to all TCR neurons. sents the summed membrane potentials of all TCR neurons and E(t) rep-
Middle panel: Wave mode summed membrane potentials of all TCR resents the impulse density of the same neurons. The power spectrum
neurons. Lower panel: Pulse mode: impulse density of all TCR neurons. of Vm (t) is shown on the left below. In this case, a smaller inhibitory area
Right: The amplitude distributions and the power spectra of the three than in the case of Figure 4.8 has been simulated. Here one interneuron
signals are shown. Note the apparent waxing and waning of the simu- (IN) inhibits only four TCR neurons. (Adapted from Lopes da Silva FH,
lated alpha rhythm and the dominant alpha rhythm with a second har- van Rotterdam A, Barts P, et al. Models of neuronal populations: the
monic. (Adapted from Lopes da Silva FH, Hoeks A, Smits H, et al. basic mechanisms of rhythmicity. In: Corner MA, Swaab DF, eds.
Model of brain rhythmic activity. Kybernetik. 1974;15:27–37.) Perspectives of Brain Research, vol. 45. Prog Brain Res. 1976:281–308.)
74 Part I ■ Basic Principles

Figure 4.8 Similar to Figure 4.7 but for a model with a larger Figure 4.9 Block diagram of the lumped model for rhythmic activity
inhibitory area. Here 1 IN inhibits 21 TCR neurons. Note that the inten- with a possibility of modulation of inhibitory feedback and excitatory
sity of the alpha peak is larger and the bandwidth is smaller in compar- feedback. The cartel consists of four populations. M, main cells with
ison with the model shown in Figure 4.7. (Adapted from Lopes da Silva input and output impulse density, respectively, P(t) and E(t), and mem-
FH, van Rotterdam A, Barts P, et al. Models of neuronal populations: brane potential, Vm (t); spike generation nonlinearity sigmoid function.
the basic mechanisms of rhythmicity. In: Corner MA, Swaab DF, eds. INIF, inhibitory population with membrane potential Vi1(t) and modulat-
Perspectives of Brain Research. vol. 45. Prog Brain Res. 1976;281–308.) ing inputs: excitatory by means of M1(t) and inhibitory by means of
M2(t), spike generation with sigmoid. INFF, excitatory population driven
by the main input P(t) inhibiting the inhibitory population INIF; INEF,
excitatory population feeding back on to the main population, spike gen-
matrix of neurons into a system such as that depicted in eration sigmoid. The interconnectivity constants giving the number of
Figure 4.9. Essentially, the neural mass (here thalamic nucleus), cells of one population projecting to one cell of the other population are
the EEG of which is of interest, forms a KII set, which is consti- C1 and C2 for the inhibitory feedback and C4 and C5 for the excitatory
tuted by the interaction of an excitatory (M) and an inhibitory feedback. C3 represents the number of cells of the INFF population
(INIF) population (KIe and KIi sets). In addition, the scheme of excited by one input fiber (feed-forward). (Adapted from Lopes da Silva
Figure 4.9 shows a second KIe (INEF) set that provides excita- FH, van Rotterdam A, Barts P, et al. Models of neuronal populations: the
tory feedback and an additional KIe set that may receive a direct basic mechanisms of rhythmicity. In: Corner MA, Swaab DF, eds.
input (P(t)) from a source situated outside the neural mass and Perspectives of Brain Research. vol. 45. Prog. Brain Res. 1976:281–308.)
in this way lead to feed-forward inhibition. To obtain an ana-
lytic simplification of the distributed model, it is useful to con-
sider the general formalism of Wilson and Cowan (9) as
described in the introduction to this chapter.
Zetterberg (89) has made use of this formalism to simplify
the distributed model of alpha rhythm. Considering the simpli-
fied block diagram of Figure 4.10, where the EPSPs and IPSPs
are represented by he(t) and hi(t), respectively, and the sigmoid
wave pulse transfer functions are represented by fE(V) and fi (V),
expressions for the transfer function of the simulated neuronal
set can be obtained (for the mathematical treatment, see Ref. 11).
One of the outputs of such a neuronal set represents the EEG
signal of the population, assuming that the latter is determined
by the somadendritic membrane potential of the main cells.
The corresponding spectrum is shown in Figure 4.11 for differ- Figure 4.10 Block diagram of the lumped model for rhythmic activity of
ent values of K, which represents a measure of the gain factor of simplified alpha rhythm model. The thalamocortical relay (TCR) neurons
the neuronal set. The spectrum R(f) indeed shows a peak are represented by two linear systems having impulse responses simulat-
around 10 Hz, as was expected. The bandwidth and the peak ing an EPSP (h c(t)) and an IPSP (h i(t)), respectively, together with the
frequency both depend on the coefficient K. Up to a point, a larger static nonlinearity (fE(V)) representing the spike-generating process. The
K will cause an increase in the frequency of the rhythmic activ- interneurons (IN cells) are represented by one linear system (h e(t)) and a
ity, but too large a K will cause instability. K is also proportional nonlinearity (fI(V)). C1 represents the number of INs to which one TCR
to the derivatives 1/ael and 1/ail of the sigmoid functions in their neuron projects; C2 represents the number of TCRs to which one IN proj-
working point, which in turn is determined by the mean input ects. (From Lopes da Silva FH, Hoeks A, Smits H, et al. Model of brain
pulse density P(t). The peak frequency will increase and the rhythmic activity. Kybernetik. 1974;15:27–37, with permission.)
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 75

Figure 4.12 An epoch of alpha rhythm (A) experimentally recorded


from the visual cortex of the dog (technique as used in Ref. 86). The
Figure 4.11 Power spectra of the output of the lumped alpha rhythm power spectrum (B) and the bicoherence (C) were obtained by ensem-
model shown in Figure 4.10 for different values of the feedback gain ble averaging. Note the peak in the power spectrum at about 10 Hz and
(K), which is mainly determined by the coupling constants representing the smaller peak at the second harmonic frequency, which are related
the synaptic interactions within the neuronal set. As the synaptic inter- to each other as the bicoherence shows. A: Experimentally recorded
actions become stronger and K increases, the spectrum acquires a clear alpha rhythm (arbitrary scale). B: Power spectrum (arbitrary scale). C:
selectivity at the alpha frequency. (Adapted from Lopes da Silva FH, Bicoherence limits 99% at 0.119, 95% at 0.096, five lines from 0.18
Hocks A, Smits H, et al. Model of brain rhythmic activity. Kybernetik. until 0.26. (Adapted from Lopes da Silva FH, van Rotterdam A, Barts P,
1974;15:27–37.) et al. Models of neuronal populations: the basic mechanisms of rhyth-
micity. In: Corner MA, Swaab DF, eds. Perspectives of Brain Research.
vol. 45. Prog. Brain Res. 1976:281–308.)
bandwidth will decrease with an increase of the coupling
constants, C2 and C1, that is, with an increase in synaptic inter-
actions within the neuronal cartel. terms of phase differences in rhythmic components at different
The linear analysis of the preceding section gives only a first locations on the cortical surface.
approximation to the spectral characteristics of the alpha One hypothesis is that the propagation of alpha activity
rhythm, both experimentally measured and simulated. The spec- depends on spatial properties of the cortical neuronal net-
trum of the experimentally measured alpha rhythm also presents works; the strength of inhibition is a function of distance
a clear component at the second harmonic of the peak frequency depending on the length of interneuronal connections. This
(Fig. 4.12). It can be demonstrated by means of bispectral analy- hypothesis has been realized by way of a “spatially distributed
sis that this component is indeed a second harmonic of the peak model” of neurons and interneurons (20), which is an exten-
frequency. This fact implies that static nonlinear properties of the sion of the lumped parameter model described by Lopes da
network have also to be taken into account. Silva et al. (10). In this spatially distributed model, it is assumed
that two populations of neurons (main cells and local interneu-
Continuous Neural Field Model of Alpha Rhythm rons) are arranged in a one-dimensional row and are intercon-
The spatial properties of alpha rhythms are less well under- nected by means of recurrent collaterals and inhibitory fibers.
stood, although it has been found experimentally that, in the Such a neuronal chain is assumed to have the following proper-
visual cortex of the dog, small cortical areas appear to act as ties: (a) the transfer function of input-output spike densities are
“epicenters” from which alpha rhythm activity “spreads” in dif- second-order linear functions with a longer time constant for
ferent directions (21,179). The spreading has been measured in inhibition than for excitation; (b) the interconnectivity properties
76 Part I ■ Basic Principles

Figure 4.13 Transfer properties of the spatially distributed model or


neuronal chain. Left: The time courses of the simulated IPSP and EPSP Figure 4.14 Schematic drawing of a section of the spatially distributed
are shown with the corresponding equations. Right: The spatial connec- model (neuronal chain). For explanation see text. (Adapted with per-
tivity functions are shown. Ce(x) and Di(x) describe the homogeneous mission from van Rotterdam A, Lopes da Silva FH, Van den Ende J, et
interconnectivity functions for the excitatory and the inhibitory connec- al. A model of the spatial-temporal characteristics of the alpha rhythm.
tions, which depend only on distance x. The values of the characteris- Bull Math Biol. 1982;44(2):283–305.)
tic lengths correspond to l/ c and l/ d. The numerical values of c and d
in the figure are only indicative. (Adapted from van Rotterdam A, Lopes
da Silva FH, Van den Ende J, et al. A model of the spatial-temporal char- model may be written as a linear inhomogeneous partial differ-
acteristics of the alpha rhythm. Bull Math Biol. 1982;44(2):283–305.) ential equation, which is a rather complicated type of wave
equation. By assuming a wave packet existing over a restricted
length somewhere in the neuronal chain at some instant of
are homogeneous in space, that is, the strength of inhibition is time, one can try to determine group velocity and dispersion
only a function of distance between the interconnected neu- properties. Alternatively, the neuronal chain may be conceived
rons; and (c) the strength of interconnectivity decreases expo- as a two-dimensional linear system with a specified transfer
nentially as a function of distance, as shown in Figure 4.13. function relating the input p(x,t) to the output u(x,t). The
To simulate alpha activity as a function of both space and response to an arbitrary input can then be obtained in the time
time, the lumped model must be extended in such a way that it domain by means of a two-dimensional convolution or in the
can account for spatial properties. This can be done as follows:
the spatially distributed model consists of a chain of neurons
and interneurons interconnected by means of spatially distrib-
uted excitatory collaterals and inhibitory fibers from the
interneurons. The interconnections are described by Ce(k,l),
denoting the number of excitatory synapses from the kth main
cell projecting on the lth interneuron, and Ci(k,l), denoting the
number of inhibitory synapses from the kth interneuron on the
lth main cell. Figure 4.14 shows a schematic drawing of such a
neuronal chain, where T k is the kth main cell and Ik is the kth
local interneuron. The main cells receive input along afferent
fibers from outside the chain. Figure 4.15 shows the correspon-
ding block diagram. The time-dependent behavior of the mem-
brane potential U(k,t) of the kth cell is determined by the
transfer functions he(t) and hi(t) of the lumped model. Nle and
Nli denote the static nonlinearities that relate membrane poten-
tials to impulse densities of main cells and interneurons,
respectively. These static nonlinearities are assumed to have the
same characteristics as fe(V e) and fi(V i) in the lumped model
and hence can be approximated as linear gain factors in the Figure 4.15 Block diagram of a section of the neural chain model
neuronal chain. Finally, P(k,t) denotes the input impulse den- showing the main excitatory and inhibitory interconnections as
sity on the main cell k. explained in text. (Adapted with permission from van Rotterdam A,
To study the propagation properties in the neuronal chain, Lopes da Silva FH, Van den Ende J, et al. A model of the spatial-tempo-
various approaches can be used. In the first place, the expres- ral characteristics of the alpha rhythm. Bull Ma th Biol.
sion describing the spatial and temporal characteristics of the 1982;44(2):283–305.)
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 77

two-dimensional frequency domain by multiplication of the One of these systems consists of tangential intracortical col-
spectra of the transfer function and the input function (see laterals of the pyramidal cells. These may spread over dis-
mathematical treatment of van Rotterdam et al. (20)). tances of about 3 mm; they form the “pyramid collateral
It is reasonable to assume that, in order to simulate alpha spread module” (95). Another system consists of long sur-
rhythm conditions, the neuronal chain must be excited ran- face parallel terminal axons that may extend over distances
domly in space as well as in time. Due to the linear character of of about 6 to 8 mm; this forms the “surface parallel intracor-
the model, however, a simple local harmonic excitation of the tical system” (95). These systems of overlapping intracortical
chain already reveals the propagation properties of the neu- connections will lead to the establishment of cooperative
ronal chain. action between large populations of cortical neurons and
If the neuronal chain is locally excited at x = 0 with a har- thus to the appearance of dynamic patterns over the cortex
monic component of frequency 0, the model response with (96). In addition to the corticocortical systems described
the same time characteristics has a local component at x = 0 and above, one must also take into consideration the role of local
two components that spread in two directions along the chain. interneurons; Tombol (97) described, among others, basket
The amplitudes of these components decay exponentially pro- cells in the cat cortex with vertical axons branching horizon-
portional to exp[ Re{ki( 0)}|x|] and the phase shifts that tally over 500 to 1,000 m, with terminals surrounding and
reflect the propagation properties of the harmonic component making contact with pyramidal cell somata; these basket
equal exp[ Im{ki( 0)}|x|] radians when measured at a distance cells are probably inhibitory interneurons. The collaterals
x in relation to x = 0. Both phase shift and decay are thus belonging to the pyramid collateral spread module system of
dependent on the frequency of excitation. Szentagothai, which is most likely excitatory, and the axons
The results of the evaluation of the roots of the equation of the inhibitory interneurons can be identified as the exci-
describing the output of the chain as a function of space and tatory collaterals and the inhibitory fibers modeled in the
time (20) show that the rhythmic components existing in the model neuronal chain; because this system of interconnec-
chain in the alpha band are less damped than the components tions ranges over distance in the order of magnitude of hun-
outside the alpha band. Furthermore, it follows that the damp- dreds of micrometers, the estimated characteristic lengths
ing in the alpha band is mainly determined by the interconnec- computed from the experimentally measured alpha rhythm
tivity function with the largest characteristic length. frequency and phase velocity with the help of the neuronal
The model just described schematically has characteristics chain model are in agreement with neuroanatomical data.
that are in qualitative agreement with experimental data on
Finally, note that the neuronal chain can be easily general-
alpha rhythmic activity obtained from the visual cortex of dog
ized to a neuronal network extending in two or three dimen-
(21). This can be examined in relation first to coherence analy-
sions. A more general approach that accounts for the
sis and second to experimental phase measurements:
generation of alpha rhythms and its spread in the cortex was
1. Coherence analysis of EEG signals recorded from chronically proposed more recently by Liley et al. (98–100), who consid-
implanted subdural electrodes lying directly on the marginal ered the cortex as an excitable spatial continuum of recipro-
gyrus revealed a higher correlation between EEG compo- cally connected excitatory and inhibitory neurons interacting
nents in the alpha band than between components outside by way of short-range (intracortical) and long-range (cortico-
that band (90,91). These experimental data support the con- cortical) connections.
clusion from the model study that alpha rhythms are the An essential feature of this general model is that IPSPs are
least damped in the chain. described by third-order differential equations, since according
2. Phase shifts at the alpha frequency (~12 Hz) measured on to these authors lower orders were not able to support wide-
the marginal gyrus in four dogs reflected phase velocities spread activity in the alpha band. This theoretical approach
that were about 0.3 m/sec (21). Using the model, the charac- showed that the strength and the form of synaptic interactions
teristic length corresponding to this phase velocity and fre- between inhibitory interneurons constitute the most relevant
quency was computed. The order of magnitude of the determinants of the frequency and damping of alpha band
characteristic lengths 1/c and 1/d of the connectivity func- oscillations. This feature derives from the assumption that local
tions Ce(x) and Di(x) (Fig. 4.13) was estimated and found to inhibitory–inhibitory loop delays are longer than the corre-
be within the range of 160 to 330 m. The characteristic sponding local and long-range excitatory–excitatory loop
lengths correspond to the distance at which the influence of delays. This theory has been presented in the form of a set of
a neuron on a neighbor is reduced to 37% of the starting nonlinear differential equations that can be solved by numeri-
value and not to the whole length of the interconnecting cal methods, but most important predictions of the theory can
fibers. The figures that were calculated using the model are be obtained by a simplified set of linear equations. In this way
in the same range as the dimensions of the basic cortical the input to the cortex can be assumed to be indistinguishable
space module, which is considered to be a vertical cylinder of from band-limited white noise, while the cortex operates as a
200/300 m diameter (92–95). Different systems of intra- noise filter that passes preferentially frequencies around 8 to 13
cortical fibers, which involve much larger distances than the Hz. The filter can be described by a transfer function, the prop-
diameter of a column (at least distances one order of magni- erties of which are determined by both the physiological state
tude larger), assure the interconnectivity of several columns. and the anatomical structure of the cortex.
78 Part I ■ Basic Principles

MODELS OF OSCILLATIONS AT THE SINGLE


CELL AND LOCAL THALAMIC NETWORK
LEVELS
In the last decades, important advances were obtained with
respect to the physiology of single cells of the thalamus and to
their patterns of interaction at the level of local networks,
including the corresponding intrinsic membrane and synaptic
properties. Interesting models incorporating these properties
were proposed, among others by Wang (44), Wang et al. (48),
Golomb et al. (42,43), Destexhe et al. (69), and have been
reviewed extensively by Destexhe and Sejnowski (83). These
models are mainly based on the experimental results of Steriade
and colleagues (101) and also of McCormick and collaborators
(74,75). The latter results were obtained in the ferret slice
preparation that facilitates carrying out controlled experi-
ments, although under circumstances that differ from the con-
ditions in the intact brain. Nevertheless, in these slices it was
possible to show (74,75) the occurrence of spindles with main
frequency between 5 and 9 Hz, during which the RE neurons
tend to fire synchronously with almost every cycle of local
rhythmic spindle. In contrast, TCR neurons tend to fire bursts
also phase-locked to the local rhythm, but not at every cycle
since they may skip one, two, or even more cycles. The modula-
tion of this activity by blocking GABAA receptors results in a
decrease of the main frequency to 2 to 4 Hz, whereas the rhyth-
mic pattern is hardly changed by blocking GABAB receptors. In
addition, the spindle bursts appear to propagate in the slice, as
traveling fronts, at a velocity of about 1 mm/sec.
As indicated above, the RE and the TCR neurons can exhibit Figure 4.16 Synaptic architecture of the model of Golomb et al. (43).
oscillations of the membrane potential, which depend strongly A: Reticular neurons (RE) receive thalamocortical (TC) excitation via
on the state of the potassium leak conductance of the cells (gkl). glutamatergic (AMPA) synapses and intra-RE inhibition via GABAA and
In the model of Golomb et al. (43) both RE and TCR neurons GABAB synapses from other RE cells. B: This model has a one-dimen-
are at rest, that is, they do not display oscillatory behavior, for sional architecture. The RE-to-TC coupling strength decays with dis-
too small or too large values of gkl and they oscillate at interme- tance. The typical decay length RT is called the synaptic footprint length.
diary levels. In this model the TCR neurons elicit EPSPs on RE The same applies to the TC-to-RE coupling (TR) and to the RE-to-RE
neurons by way of glutamatergic (AMPA) synapses, whereas RE coupling (RR). C: A typical footprint shape of the synaptic coupling with
neurons elicit both GABAA- and GABAB-mediated IPSPs on distance; the decay length (either TR or RT or RR) is the distance for
TCR neurons. Due to the existence of a low-voltage activated which the synaptic coupling reaches 1/ e of its maximal value. (Adapted
Ca current (T current) the TCR neurons can elicit a rebound from Golomb D, Wang XJ, Rinzel J. Propagation of spindle waves in a
burst after a hyperpolarization due to the GABAergic IPSP, thalamic slice model. J Neurophysiol. 1996;75:750–769.)
whether or not associated with a Ca-dependent K current
(afterhyperpolarization [AHP] current). In the model simula-
tions if the neurons were not in intrinsic oscillatory mode, the
network would be quiescent. However, the introduction of a oscillations, whereas an up-regulation of Ih elicited refractori-
few oscillating TCR neurons was sufficient to recruit the other ness of the neurons, leading to extinction of the oscillations. In
neurons into a rhythmic spindle. The oscillation was critically this model, the responses of the neuronal network to different
dependent on the existence of a cationic current activated by stimulus patterns were not reported, although this is most
hyperpolarization, the Ih or “sag” current, in these neurons (44). important to understand the behavior of the network of the
Using this model it was possible to investigate the patterns of intact brain under physiological conditions.
propagation of the oscillations throughout the network. The
neurons in the network were connected by synaptic couplings, MODELS OF EPILEPTIFORM ACTIVITY
the strength of which decays with spatial distance, as shown in
Figure 4.16. An oscillation started typically at a focal site and Here we consider those models by means of which one attempts
propagated as a wavefront that recruited successively more neu- to explain how EEG phenomena characteristic of epileptiform
rons into the oscillatory mode. The Ih current modulated the seizures may emerge from the normal ongoing activity. Most
occurrence of these oscillations: a reduction of Ih led to sustained theoretical studies have addressed in general terms the question
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 79

of how oscillations may take place in a network of neurons,


assuming that such oscillations are akin to seizure activity. A
general way to investigate this problem using a model of such a
network is to analyze the trajectories in the phase plane whose
coordinates are the potential and its first derivative. A nonlinear
system that goes into stable oscillation even after the input
stimulus has ceased will produce a closed trajectory in the phase
plane; such a closed trajectory is called a limit cycle. Working
from the model initially described by Lopes da Silva et al.
(10,11), Zetterberg et al. (102) have shown that a slightly mod-
ified model may generate limit cycle oscillations that can simu-
late some forms of epileptogenic activity recorded from the
Figure 4.17 Simulation of a bursting neuron showing an epileptiform
cortex. Freeman noted that his KII model was not initially
discharge. Each simulated neuron had a number of compartments and
designed to simulate epileptiform activity. However, by using a
ion conductances, as explained in the text. The intracellular burst
different nonlinear gain curve, Babloyantz and Kaczmarek
model is shown above a recorded burst. The network contains 100 neu-
(103) have generated limit cycle oscillations that are thought to
rons interconnected in a random synaptic network. Vertical calibration,
resemble epileptiform activity. In such models, however, a more
25 mV by 25 msec (model) and 40 msec (experimental). (Adapted from
or less precise fit between simulated and recorded EEG activity
Traub RD, Wong RKS. Cellular mechanism of neuronal synchronization
was not attempted. More recently, models that attempt a precise
in epilepsy. Science. 1982;216:745–747.)
waveform fit have been introduced. Such a model was proposed
by Traub (52) and Traub and Wong (104) and is based on
experimental data at the cellular level concerning the mem-
brane mechanisms responsible for epileptogenesis (53) together experimentally recorded epileptiform transients was obtained,
with the properties of synaptic interactions in hippocampal as shown in Figure 4.17. A second problem analyzed was how
neurons. sustained afterdischarges can be produced in such a model. It
We should note that in recent years the interest for NMMs was found that for the occurrence of repetitive afterdischarges,
with respect to epileptic phenomena has steadily increased, as some form of pacing inhibitory process was needed. This was
illustrated below, most likely due to the fact that epileptic phe- accomplished by introducing two additions in the model: (a) a
nomena involve brain systems with complex dynamics that are slow voltage-dependent K conductance (M current) and (b)
difficult to comprehend exclusively on the basis of knowledge at an axonal refractoriness that causes intermittent conduction
the cellular level. from one to the next depolarized cell, located at the axonal ini-
tial segment, at branch points, or at presynaptic terminals.
Cellular and Network Models Furthermore, a number of loops allowing reentrant paths
of Epileptiform Activity within the neuronal population are of importance for the main-
Observations at the cellular level obtained in the hippocampal tenance of a series of afterdischarges. Using this kind of models,
slice preparation have yielded a number of new facts concern- in a series of studies Traub and collaborators (57,106) investi-
ing the origin of an epileptogenic focus. It has been shown that gated the role of a number of factors in inducing epileptiform
pyramidal cells of the hippocampus (cornus ammonis or CA3) oscillations similar to those observed in brain slices, in vitro,
have active membrane regions, the so-called soma and den- namely synaptic mediated activation of N-methyl- D-aspartate
dritic hot spots that have voltage-dependent ion conductances (NMDA) dendritic synapses and intrinsic voltage-dependent
(g) for Na , K , and Ca2 and a calcium-activated K conduc- conductances. The possible importance of the firing of ectopic
tance; furthermore, a voltage-dependent inactivation of gK and presynaptic action potentials in eliciting the synchronization of
a partial inactivation of gCa by the cytosolic accumulation of neurons during epileptiform seizure-like activity was suggested
Ca2 ions has been assumed (52). Incorporating all these (57). Another factor that may contribute to epileptiform activity
properties, Traub (52) has made a multicompartmental model has been put forward on the basis of models and in vitro physi-
by means of which spontaneous bursts in CA3 cells can be gen- ological studies of Traub’s group (55): gap junctions located
erated, as shown in Figure 4.17. Using these elementary cellular between the axons of principal hippocampal neurons would
components (105), Traub et al. (54) constructed a network of play a role in the generation of very fast (about 200 Hz) oscilla-
CA3 neurons interconnected by excitatory synaptic connec- tions. These model simulations have allowed making the predic-
tions. The generation of epileptiform transients induced by the tion that action potentials can cross from axon to axon via gap
administration of antagonists of the neurotransmitter GABA junctions. Axonal network oscillations would, in turn, induce
was the first problem considered. The conditions for the pro- oscillatory activity in larger neuronal networks, by a variety of
duction of a chain reaction of excitation that spreads over the mechanisms, and could underlie in vivo ripples (to ~200 Hz in
whole network were found using the simulations. The most hippocampal EEG) to drive gamma (30 to 70 Hz) oscillations
important of these conditions was that the excitatory intercon- that appear in the presence of carbachol and to initiate certain
nectivity should be sufficiently dense and the synaptic interac- types of ictal discharges. A comprehensive review of the work of
tions of sufficient strength. A good fit between simulated and Traub and collaborators can be found in Traub et al. (107).
80 Part I ■ Basic Principles

Along the same line high-frequency ripples may be determined further, that the transition between interictal and fast ictal
by high-frequency inhibitory activity, the breakdown of which activity is explained, in the model, by the impairment of den-
may enable seizure activity to propagate from an initial focal dritic inhibition (Fig. 4.18).
area (108,109). Also using NMMs Kramer et al. (119–121) constructed
In addition, model studies have also addressed the possibil- models of human cortical networks that can generate seizure-
ity that changes in extracellular ionic concentrations (increase like activity, as the result of increased connectivity between
of K and/or decrease of Ca2 ) may be important for the gen- excitatory and inhibitory cell populations or of decreased con-
eration of epileptiform seizure activity. Somjen and collabora- nectivity within either excitatory or inhibitory cell populations.
tors (110–112) used computer models to better understand These studies also allowed to test the possibility of controlling
how transmembrane ionic and water movements may affect seizures (120) by means of feedback controllers of the model
neuronal excitability and observed that the model produced dynamics.
epileptiform afterdischarges if during stimulation interstitial In contrast with NMMs where the topology of the network
K concentration was sufficiently enhanced so that the persist- is not taken into account or is much simplified, topological mod-
ent Na (slowly inactivating) current was activated. In order for els there have been developed, that is, graphical representation
firing of a neuron to be maintained after the end of a stimulus, models where topological details of the neuronal networks are
the [K o]/[K i] ratio had to be sufficiently large to maintain explicitly included. These models permit to simulate the effects
the neuronal membrane depolarized so that the inward cur- of histological changes that may occur in an epileptogenic tis-
rents could persist, which activated a series of action potentials. sue, such as the death of certain cellular types and the sprout-
In the course of time the persistent Na current was inactivated ing of new axonal or dendritic connections. Thus Lytton et al.
and the ion pumps, associated with the buffering of K ions by (122) developed a network model of the dentate gyrus of the
glia cells, restored the normal levels of K extracellular concen- hippocampus including granule cells, inhibitory aspiny
tration. These model studies strongly suggest that the elevation interneurons, and excitatory mossy cells. According to this
of extracellular K is not the initial stimulus for a seizure, but it model mossy fibers sprouting and disinhibition were necessary
may cause the transition from interictal to ictal discharges for repetitive granule cell firing, alike to EEG seizure activity, to
(113). The initial stimulus starting the seizure may require occur. More elaborate topological models of similar kind were
synaptic interactions among a population of interconnected developed by Santhakumar et al. (123) and Dyhrfjeld-Johnsen
neurons. et al. (124). The former showed that if sprouting of mossy fibers
In contrast with the models described above, which are exceeds a certain value, seizure activity can occur regardless of
directed to the simulation of epileptiform activity as seen at the the number of hilar cells lost. The latter model study, including
cellular level, especially in in vitro preparations, other models more than one million cells, simulated the topology of the den-
are based on a neural mass approach using mean-field concepts. tate gyrus. This can display globally and locally connected
A paramount example of these is the model study of Wendling (“small world”) architecture. The effect of changes of network
et al. (19) that focuses on high-frequency EEG activities topology during epileptogenesis was modeled on the one hand
(gamma band) that constitute one of the most characteristic by removing long-distance projecting hilar cells which causes a
EEG patterns in focal seizures of human epilepsy, especially reduction in the connectivity and, on the other, by sprouting of
those that have an epileptogenic zone in the hippocampus and granule cell axons which has the opposite effect of increasing
associated limbic cortical areas (114,115). This model starts local connectivity. The model simulations demonstrated that
from the hypothesis that networks of inhibitory interneurons the survival of even a small fraction of hilar cells was enough to
with different properties are essential for the generation of sustain hyperexcitability in the network. This is consistent with
gamma frequency oscillations (116). As already pointed out experimental data since in animal models of hippocampal scle-
above, this lumped model includes different kinds of neuronal rosis the loss of hilar cells is never 100%; in our study (125) it
populations, in particular two kinds of GABAergic neurons: was about 66% to 74% in rats with a progressive form of epilep-
GABAA fast and GABAA slow populations, where the former tic seizures. Also in brain tissue extracted from patients with
cause IPSPs with short time constants on the soma of pyrami- hippocampal sclerosis about 20% of mossy cells have been esti-
dal neurons, while the latter cause IPSPs with slow time con- mated to survive (126), which corresponds closely with the
stants on the dendrites of these neurons. Furthermore as finding in the model of Dyhrfjeld-Johnsen et al. (124) that
demonstrated experimentally by Banks et al. (117), both classes maximal epileptiform activity was seen at a level of about 80%
interact: GABAA slow cells inhibit not only pyramidal cells but sclerosis.
also GABAA fast interneurons. The model is based on the In general we may state that a variety of changes at the cellu-
important experimental observation that a nonuniform alter- lar level may lead to the occurrence of epileptiform seizure
ation of GABAergic inhibition underlies the generation of activity. In fact there is no unique process that can account for
epileptiform activity in hippocampus and associated areas of the generation of epileptiform activity (127). A generalized
the limbic brain, namely a reduced dendritic inhibition and concept is that seizures occur due to a shift from a dynamical
increased somatic inhibition (118). The simulations show that unbalance between excitatory and inhibitory processes with a
strikingly realistic activities are produced by the model when predominance of the former in strongly coupled networks. Van
compared to real EEG signals recorded with intracerebral elec- Drongelen et al. (128) showed, however, in a detailed computa-
trodes in patients with temporal lobe epilepsy. They show, tional model that seizure-like activity may occur when the
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 81

Figure 4.18 A: A real depth-EEG signal


recorded in human hippocampus at the begin-
ning of a partial temporal lobe seizure with
intracerebral electrodes (SEEG). Four phases (a-1
to a-4) are distinguished according to the pseudo-
stationary nature of the activities reflected by the
signal: normal background activity (a-1), dis-
charge of rhythmic spikes (a-2), low-voltage
rapid discharge (a-3), and high-amplitude quasi-
sinusoidal slowing down activity (a-4). The nor-
malized power spectral density (PSD) shows that
part of the activity corresponding to phase a-3
belongs to the gamma band (55 to 60 Hz). B: A
candidate path on the activity map, for a value of
excitation (A = 5) and different values of slow
(B) and fast (G) inhibition, showing the transi-
tions between different types of activity observed
in the real signal (type 1 → type 3 → type 4 →
type 6). C: Slow and fast inhibitory synaptic gain
profiles defined by the candidate path and used
in the model to simulate a timeseries signal with
transitions in dynamics. D: Simulated EEG signal
obtained with the model when the synaptic gains
vary as a function of time and PSDs computed on
the four periods of 10 seconds (b-1 to b-4).
(Adapted from Wendling F, Bartolomei F,
Bellanger JJ, et al. Epileptic fast activity can be
explained by a model of impaired GABAergic
dendritic inhibition. Eur J Neurosci.
2002;15(9):1499–1508.)

excitatory synapses are weakened. The model predictions were respect the single neuron model of a thalamic relay neuron of
tested experimentally and the authors showed that a pharmaco- Wang (44), which shows both 10- and 3-Hz bursting dynamical
logical reduction of excitatory synaptic transmission could lead modes, is particularly instructive. This model demonstrates
to repetitive network bursting. clearly how distinct oscillatory modes depend on different bal-
ances of sets of ion currents. In this model the emergence of
Models of Thalamic Spike-and-Wave Seizures 3-Hz bursts, which resemble the 3-Hz oscillations seen in gen-
Thalamic neurons can display different modes of activity eralized epilepsies, depends on the value of the hyperpolarization-
including rhythmic bursting that are akin to spike-and-wave activated cationic current, Ih the so-called sag current, although
(SW) discharges characteristic of generalized epilepsies. In this the T-type Ca current also plays an important role. According
82 Part I ■ Basic Principles

to this model the 10-Hz oscillatory mode is maintained, at the both kinds of activity without specific adjustments of parame-
level of a single neuron, by the dynamics of the low-voltage- ters being expressly made. Indeed the essence of epilepsy is that
activated Ca current, IT. Under normal conditions the neuronal a patient displays (long) periods of normal EEG activity (i.e.,
membrane does not hyperpolarize sufficiently to activate the Ih nonepileptiform) intermingled with epileptiform paroxysmal
current. activity only occasionally. Thus, it is essential to understand the
Indeed if the neuronal membrane would be deeply hyperpo- mechanisms responsible for transitions from normal activity to
larized, it would stay in such a condition in case only IT would paroxysmal SW discharges. This aspect of the dynamical
be active, because the latter current is not sufficient to overcome process responsible for epilepsy was addressed in a computa-
a strong hyperpolarization. However, the presence of Ih allows tional model of absence epilepsy (50). In this model the behav-
the neuron to escape this strong hyperpolarized state. Thus the ior of populations of interacting neurons integrating neuronal
transition between the 10- and the 3-Hz modes depends on the and network properties was simulated, as a more extended and
level of hyperpolarization. This single neuron model is com- elaborated version of a previous model of the alpha rhythm
pletely deterministic and it may display “strange attractors” typ- (132). This approach facilitated investigating system dynamics
ical of a chaotic dynamic state. at the macroscopic level, that is, at the level where electric brain
A model of a thalamic neuronal network that is closer to the signals such as local field potentials or EEG are recorded. The
reality of epileptic discharges than the single neuron model model may exhibit two qualitatively different types of behavior,
described above was developed to account for SW seizures at such as seen in experimental animals with genetically deter-
the level of a neuronal population. The basic physiological phe- mined absence-like seizure (WAG/Rij rats and GAERS ani-
nomenon simulated was the finding that during SW discharges mals). The output signal may display a waxing and waning
the RE neurons discharge with long spike bursts riding on a “spindle-like” oscillation having a spectrum with a peak at
depolarization, whereas the TCR neurons are either entrained approximately 11 Hz or a high-amplitude “seizure-like” oscilla-
into the seizure oscillation or are quiescent (129). The spatial tion at a frequency around 9 Hz, as seen in these rats during
model of these neuronal populations predicts that in a center of absence seizures (Fig. 4.19). The former behavior corresponds
seizure activity there will be intense RE activity and TCR quies- to the normal ongoing activity, while the latter corresponds to
cence and that this focus will be surrounded by an area where the paroxysmal epileptiform activity. Thus, for a set of neuronal
the TCR neurons will be less hyperpolarized such that low- parameters, the model is in a “bistable regime” where it may
threshold oscillatory spikes will occur. This surrounding area generate both normal and paroxysmal oscillations and sponta-
will be the forefront of a wave of propagating seizure activity. neous transitions between these two types of behavior. This
The model shows that the complex nonlinear dynamics of the model facilitated making a number of important predictions
neuronal network depends critically on the low-voltage-acti- that can be tested experimentally, both in animals and in
vated (LVA) Ca-current IT. In this context it is interesting to
note that in the genetic model of absence epilepsy GAERS
(genetic absence epilepsy rats of Strasbourg), the presence of a
significantly enhanced LVA current in thalamic neurons was
demonstrated by Tsakiridou et al. (130). It is also noteworthy
that drugs, like ethosuximide, that depress IT should suppress
seizure initiation. It should be noted, however, that more recent
studies showed that ethosuximide acts also on the noninactivat-
ing Na current and on a Ca2 -activated K current (131). The
level of hyperpolarization of TCR cells, which appears to be a
crucial determinant of the dynamic behavior of the neuronal
population, depends on synaptic mechanisms. The model of
Destexhe et al. (69) goes further than previous models and
shows that the transition from 9–11 Hz to 3 Hz oscillations
appears to depend on GABAB synapses. Relatively weak RE
activity would mainly elicit GABAA responses, whereas stronger
activity would recruit GABAB synapses and this would cause the
quiescent mode of the TCR neurons. Indeed, GABAB presynap-
tic inhibition is conspicuous between RE neurons, such that an
enhancement of the latter would result in a decrease of intra-RE Figure 4.19 Upper panel: 20 seconds of a simulation with the occur-
inhibition and, thus in an increase of inhibition of TCR neu- rence of a spontaneous paroxysmal episode. Lower panels: Average
rons (quiescent state). Thus GABAergic drugs would aggravate power spectra of signals simulated by the model and recorded from a
the tendency for seizure oscillations to occur. WAG/ Rij rat with absence seizures. Left-hand side: The spectra of nor-
The models discussed above have given insight into some mal activity. Right-hand side: The spectra of paroxysmal activity.
basic neuronal mechanisms of SW discharges but do not (Adapted from Suffczynski P, Kalitzin S, Lopes da Silva FH. Dynamics
address specifically the most essential issue of this type of of non-convulsive epileptic phenomena modeled by a bistable neuronal
epileptic activity—that a given thalamocortical loop can display network. Neuroscience. 2004;126(2):467–484.)
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 83

humans: (a) The transitions between the normal and the parox- nectivity function among chains of cortical neurons; although
ysmal states can emerge spontaneously and are not induced these values are in the same order of magnitude, many factors
necessarily by any parameter changes. (b) Probabilities of tran- may account for the difference between them, such as the dif-
sitions between normal neuronal activity and paroxysmal oscil- ferences in anatomical connectivity between human and
lations depend on a number of model parameters; therefore, canine’s cortex.
these probabilities can be controlled in a nonunique way; in
more general terms these probabilities may be described by MODELS OF EEG GENERATION INVOLVING
gamma distributions, of which the exponential distribution is a CORTICAL AND SUBCORTICAL AREAS
special case. Interestingly, exponential shape of histograms of
durations of ictal events has also been found in other types of The models presented in the previous section describe only a
human epilepsy. (c) Paroxysmal oscillations can be annihilated rather small number of EEG phenomena. There are many EEG
by a well-timed pulse, such that, under given conditions, this patterns that have not yet been modeled in terms of sets of neu-
kind of seizures may be aborted. The model facilitates investi- ronal networks although some NMMs have been shown to gen-
gating these conditions such that the possibility of aborting erate EEG-like oscillations at different frequencies. Moreover,
seizures may be tested experimentally. Another model of most models have been limited to simulations of the behavior
absence seizures gives special attention to the time delays of of local neural networks. This chapter has already considered a
thalamocortical and corticothalamic pathways on the frequency model that involved a KIII set including networks of different
of SW discharges (133). Although these time delays likely con- areas. However, several EEG phenomena have started to be ana-
tribute to the frequency of the seizure oscillations, it is difficult lyzed in these terms. For example, sleep EEG patterns are
to deduce from this model what is the role of these delays in this known in some detail in terms of cellular processes in a num-
process, since this model does not include other important cel- ber of brainstem and cortical regions (136–138; see also
lular elements that may also affect the oscillation frequency, Chapter 3), and a model describing many essential features of
such as GABA synaptic processes with different time constants slow-wave sleep and activated states in the thalamocortical sys-
and calcium T-channels with particular kinetics. Further it does tem as well as the transition between them was described (139).
not account for nonstationary changes in apparent time delays Specific models that address the question of how pathological
between cortex and thalamus that have been shown experimen- slow oscillations (140) may be generated have not yet been
tally to occur during one episode of SW discharge (134). For a developed. It is to be expected that efforts in this direction will
comprehensive review of computer modeling in epilepsy the lead to a better understanding of such EEG phenomena by
reader is referred to Lytton (135). combining physiological data at the cellular level with mathe-
matical modeling.
LARGE-SCALE THALAMOCORTICAL MODELS Another important theoretical aspect of the generation of
EEG phenomena is the question of relating the temporal and
As stated in the introductory remarks brain rhythms and EEG- spatial aspects of such phenomena. Most models are concerned
like phenomena have also been investigated in large-scale mod- with local networks, but not with the question of the coupling
els including detailed neuronal properties by the group of between relatively distant sets of neurons (e.g., between the
Edelman et al. In the context of the present discussion it is par- thalamus and the cortex). However, a model aimed at analyzing
ticularly interesting to note that in a large-scale thalamocortical such a problem concerning the spatial organization of alpha
model rhythmic activities can emerge characterized by frequen- rhythms has been introduced by Lopes da Silva et al. (90,91). A
cies typical of EEG/MEG rhythms, such as delta, alpha, beta, number of experimental studies of alpha rhythm in the dog
and gamma, although these activities were not explicitly built in have revealed that intracortical coherences are in general larger
the neuronal elements (4). than any thalamocortical coherence measured in the same ani-
In this model it was shown that rhythms at specific fre- mal. This led investigators to question the amount of influence
quencies occur in different cortical areas, for example, a beta of thalamic sources on the large intracortical coherences. This
rhythm at about 20 Hz in areas corresponding to motor and amounts to choosing between two models: either thalamocor-
somatomotor cortex, while the microcircuitry of different tical relations are determined by point-to-point projections or
cortical areas was the same. Therefore these authors conclude each thalamic population may project to several cortical areas.
that these differences in frequency of oscillations should be Using partial coherence analysis (91), it has been shown that a
determined by differences in white-matter anatomical con- model based on point-to-point projections cannot be accepted
nectivity (the latter was modeled based on diffusion tensor to explain the organization of thalamocortical alpha rhythms.
imaging data of the human brain) between and within corti- Furthermore, it was found that there are widespread influences
cal areas. Multiple propagating waves were also generated by of populations of the dorsal thalamus on intracortical coher-
the model within limited cortical regions with typical propa- ences. Notwithstanding these influences, the residual intracor-
gation velocities around 0.1 m/sec. We noted above that the tical coherences were so large that other factors must be
propagation speed of alpha waves on the surface of the dog’s assumed to explain this intracortical coherent activity; these
cortex was in the range of 0.3 m/sec also within limited corti- factors may have as an anatomical substrate inter- and intra-
cal areas, and the model of van Rotterdam et al. (20) revealed cortical systems of fibers responsible for the spatial distribution
that this propagation is mainly determined by the intercon- of alpha rhythms within the cortex. Indeed, if such systems did
84 Part I ■ Basic Principles

not exist, investigators would be left with a patchy nonoverlap- Beta rhythms with a different frequency (~28 Hz) have been
ping assembly of cortical modules with little intracortical shown to be rather localized to occipital areas in dogs engaged
coherence; it would be almost impossible in such a case to in a visual discrimination task (146). Particularly in the visual
record an EEG at a relatively large distance from the cortex. cortex, oscillations in the gamma frequency range ( 30 Hz) at
A similar problem has been mathematically elaborated by the single neuronal level (147) have been recorded (reviewed in
Nunez (141), who developed a model, also based on the exis- Ref. 148). In this way, synchronization occurs between neurons
tence of interconnections between different cortical modules, that represent related features of an object that should be inte-
to account for the global wave pattern of the EEG at the scalp. grated for the generation of a coherent visual percept.
In this way, Nunez takes into account that traveling EEG waves Furthermore, it was shown that neurons in other areas of the
may coalesce to form standing waves because, he assumes, the cerebral cortex may also synchronize their activity with those of
cortex forms a closed surface. In his view, there is a fixed rela- the visual cortex, remarkably without time lags; this synchrony
tionship between the nondimensional temporal and spatial fre- was particularly strong between areas subserving related func-
quency of EEG given by the dispersion relation. This relation tions (15).
relates the temporal frequency with the characteristic velocity A large number of studies have demonstrated the existence
of propagation of activity in associative cortical fibers and the of high-frequency rhythmic activities in the human EEG under
so-called wave number. different circumstances, particularly in relation with motor and
Nunez (142) discusses the theory proposed by Lopes da Silva cognitive functions (149–152). A review of clinically relevant
et al. (91) and van Rotterdam et al. (20), which he calls the beta rhythms is given in Chapter 37 and of the influence of
“local theory” of the electrocorticogram, in connection with his drugs on these EEG activities in Chapter 43. Model studies of
“global theory,” which predicts traveling and standing waves these EEG fast rhythms are being developed. The demonstra-
with wavelengths up to the cortical circumference having phase tion of these high-frequency activities in animals, particularly
velocities in the 10 m/sec range. According to the local theory, in the hippocampus and associated areas, has inspired a num-
alpha activity spreads away from multiple epicenters with a pre- ber of model studies that try to account for the generation of
ferred wave number range for alpha activity and with phase these activities, integrating experimental observations and
velocities of about 0.3 m/sec. It is likely that a combined computer simulations. Such a model to account for the occur-
local/global theory is needed. Indeed, as Nunez points out rence of fast oscillations in a neuronal network was proposed by
(35,142), the simultaneous existence of both short- and long- Traub et al. (57) and Jefferys et al. (58,59). The model consists
wavelength phenomena in the same frequency range can occur. of a chain of interconnected excitatory pyramidal and
Some EEG phenomena may be dominated by short-range inhibitory interneurons. According to this model, when the
intracortical interactions, while others are dominated by long- excitatory drive on the interneurons reaches a level sufficient to
range corticocortical interactions. The human scalp EEG results induce pairs of action potentials at short intervals (“spike dou-
probably from both types of activity. In the combined local- blets”) the network generates oscillations in the gamma fre-
global theory of Nunez (35), the input to the cortex consists of quency range on a millisecond time scale from one end of the
the modulating afferent input from subcortical areas and inputs chain to the other. It should be noted, however, that neuronal
from other cortical regions, whether distant or neighboring, oscillations in a similar frequency range have been found in
through corticocortical fibers. In this model, it is assumed that other areas of the brain, mainly in olfactory bulb and cortex (1,
the local networks include inhibitory and excitatory feedback 33,153,154), but also in the hippocampus and other limbic
circuits, whereas the global corticocortical interactions are exci- areas (for review, see Ref. 155). These rhythmic activities have
tatory. Nunez and collaborators (143,144) proposed a theoreti- been modeled by means of cortical neuronal networks with
cal framework supporting experimental measures of dynamic recurrent inhibitory connections, and it was shown that such
properties of human EEG. According to this model both glob- models, with the relevant physiological parameters are capable
ally coherent and locally dominated behavior can occur within of accounting for the 20- to 50-Hz gamma rhythms (see discus-
the alpha band, depending on narrow band frequency, spatial sion on this topic in Refs. 58, 59, 155, and 156).
measurement scale, and brain state. At the same time, alpha and Based on experimental observations in brain slice prepara-
theta phase locking between cortical regions during certain tions of the hippocampus or the neocortex, in vitro, where
behavioral states can occur. oscillations are induced by electrical stimulation and/or by
exogenous drug application, a series of models was created as
MODELS OF CORTICAL BETA/ GAMMA reviewed by Whittington et al. (157). These models demon-
RHYTHMS strate that fast gamma rhythms can be generated by the mech-
anism of mutual inhibition in populations of interconnected
In the cortex fast rhythms have been identified, both in the EEG inhibitory interneurons, which can be studied independently by
and at the level of single neuronal activity, that are usually pharmacological manipulations. Despite the artificial condi-
called beta or gamma rhythms, depending on frequency range. tions of these experiments, these physiological data have led to
It has been found experimentally that fast frequency rhythms interesting theoretical generalizations (60), as for instance the
(33–45 Hz) occurring in the frontoparietal areas may occupy a possible role of electrical coupling via gap junctions in the gen-
restricted cortical area in the awake cat (145). These thalamo- eration of very fast oscillations both under normal and epilep-
cortical beta rhythms are characteristic of attentive immobility. tic conditions (158,159).
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 85

Recently, a generalized model that accounts for changes in freedom can display unpredictable behavior over long time
rhythmic activities with behavioral state, from the slower alpha scales; in other words, they can display what mathematicians
to the faster gamma and beta as arousal increases, was proposed call chaotic behavior (163). Such systems can have multiple sta-
(160). This model is especially interesting because the same set ble states governed by “equilibrium,” “limit cycle,” or “chaotic”
of underlying intrinsic and network mechanisms appears to be (“strange”) attractors. Whether the system will find itself in one
responsible for transitions between the different types of or another stable state depends on the input conditions and on
rhythms. The effects of neuromodulation on ionic conduc- the system’s parameters.
tances were investigated in a cortical circuit model, consisting These mathematical notions can be illustrated using the
of synaptically coupled excitatory and inhibitory neurons that example of the KIII set defined by Freeman (34). Such a set of
support rhythmic activity in the alpha, beta, and gamma neurons (Fig. 4.1) may have a least four equilibrium states.
ranges. It was shown that the effects of neuromodulation on There is a rest state, in which the nonlinear pulse-wave function
ionic conductances are, by themselves, sufficient to induce tran- is set to zero and corresponds to the state during deep anesthe-
sitions between synchronous gamma and beta rhythms and sia. Another is a state in which a near sinusoidal limit cycle
asynchronous alpha rhythms. occurs, such as is produced in the olfactory bulb and cortex by
It should be noted that despite extensive search, no evidence drugs and by hyperthermia (1). Still another state is character-
was found by Menon et al. (161) for globally correlated activity ized by recurrent limit cycle behavior in which the olfactory
in the human EEG, either in narrow (i.e., 35–45 Hz) or in broad bulb depends on the level of the normal respiratory rhythm. If
(i.e., 20–50 Hz) frequency bands. Instead, spatially and tempo- slightly structurally changed, a KIII set can be led to another
rally intermittent synchronization was observed between pairs state in which chaotic activity is generated. In such a case, the
of electrodes in 1-cm 2 regions with high variability. The distri- output consists of long stretches, or bursts, with unpredictable
bution of correlation coefficients differed substantially from amplitudes and phases that resemble the fluctuations of the
background levels at interelectrode distances of 1 and 1.4 cm EEG at rest. A similar behavior can be obtained by giving a low-
but not 2 cm or more. These findings suggest that the surface level random noise input to the KII set. This fact illustrates a
diameters of domains of spatially correlated activity within the general question with which one is faced when modeling the
gamma band in human EEG are limited to less than 2 cm. Thus, EEG: Is the ongoing EEG a manifestation of a chaotic attractor
if such gamma band spatial patterns exist in the human brain, or of an equilibrium state under random perturbation? The
no existing technology would be capable of measuring them at mathematical techniques with which to characterize chaotic
the scalp, and subdural electrode arrays for cortical surface behavior of nonlinear dynamic systems and to differentiate it
recording would have to have spacings under 5 mm. This means from random activity are described in detail in a number of
that on the cortex there are well-characterized spatial domains specialized publications (163–167). Furthermore, the problems
of rhythmic activities that do not spread all over the cortical involved in applying these mathematical tools of nonlinear
surface. dynamical analysis to EEG signals are discussed in a number of
publications (168–174).
RELEVANCE OF NONLINEAR DYNAMIC In general, one can state that it has been difficult to find
SYSTEMS FOR EEG GENERATION unambiguous evidence for the existence of nonlinear determin-
ism in EEG signals except for those recorded during epileptic
The models of neuronal networks presented above possess non- seizures (175). The latter observations have led to the concept
linear wave-pulse functions. In a number of studies of such net- that a given neuronal network may present essentially different
works, a simplification has been introduced; it was assumed modes of activity, that is, it may display different dynamics,
that this nonlinear characteristic could be approximated by a depending on specific conditions. For some values of control
linearized version, at least under those conditions in which the parameters, a neuronal network may change its dynamical
input signals are limited to a small range of amplitude. mode of activity; for example, instead of a random noisy state,
However, such simplification is not necessary, and it may lead a limit cycle or even a chaotic attractor may occur. Under these
one to ignore essential nonlinear properties of the system, such circumstances we say that a bifurcation has taken place. Thus a
as the generation of higher harmonics (Fig. 4.12). Indeed, it is bifurcation represents a qualitative change and depends on a set
possible to account for the behavior of neuronal networks of control parameters that define the operating regimen of the
assuming that these networks can be described by a number of system. We hypothesized (127) that the main difference
coupled nonlinear differential equations as a function of time between a normal and an epileptic brain is that the operating
and space. In general, such a set of nonlinear differential equa- regimen of a neuronal network in the epileptic brain is much
tions may be considered to provide the basis for understanding closer to a bifurcation point than in the normal brain, leading
the generation of all types of EEG activity. to a low-dimensional chaotic mode of behavior. This means
A number of investigators, such as Freeman (34,178) and that compared to the normal brain, where the operating regi-
Babloyantz (162), started to explore the possibilities given by men is situated far from such a bifurcation point, in an epilep-
the modern mathematical field of research of nonlinear tic neuronal network the distance between operating and
dynamic systems in order to obtain a better understanding of bifurcation points is so small that the system may easily switch
EEG phenomena. A relevant aspect for this discussion is that all from a stable equilibrium to a chaotic attractor, even in
nonlinear dynamic systems with more than two degrees of response to a very weak stimulus. The latter may pass undetected.
86 Part I ■ Basic Principles

Accordingly, in the case of epilepsy the basic question is which 10. Lopes da Silva FH, Hoeks A, Smits H, et al. Model of brain rhyth-
factors are responsible for the change in the operating point of mic activity. Kybernetik. 1974;15:27–37.
the involved neuronal networks (176). It should be added that 11. Lopes da Silva FH, van Rotterdam A, Barts P, et al. Models of neu-
this nonlinear dynamical behavior, which we formulate here at ronal populations: the basic mechanisms of rhythmicity. In:
Corner MA, Swaab DF, eds. Perspectives of Brain Research, vol. 45.
the macroscopic level, has also been identified at the neuronal
Prog Brain Res. 1976:281–308.
level, since near the transition from subthreshold to bursting
12. Jansen BH, Rit VG. Electroencephalogram and visual evoked
oscillations in thalamic neurons an apparently chaotic oscilla- potential generation in a mathematical model of coupled cortical
tion was observed. This was made evident in the model studies columns. Biol Cybern. 1995;73:357–366.
of Wang (44,48). 13. Suffczynski P, Kalitizin S, Pfurtscheller G, et al. Computational model
of thalamocortical networks: dynamical control of alpha rhythms in
relation to focal attention. Int J Psychophysiol. 2001;43(1):25–40.
CONCLUSION 14. David O, Friston KJ. A neural mass model for MEG/EEG: coupling
and neuronal dynamics. NeuroImage. 2003;20:1743–1755.
The complexity of EEG phenomena calls for the use of mathe-
15. Roelfsema PR, Engel AK, Koenig P, et al. Visuomotor integration is
matical models and computer simulations in order to under- associated with zero timelag synchronization among cortical areas.
stand the underlying processes of generation. In the last few Nature. 1997;385:157–161.
years, a number of problems in electroencephalography have 16. Chawla D, Friston KJ, Lumer ED. Zero-lag synchronous dynamics
been tackled in this way. Such theoretical studies are justified if in triplets of interconnected cortical areas. Neural Netw.
they are combined with experimental investigations. In this 2001;14:727–735.
way, one may not only obtain new insights about the generation 17. Zavaglia M, Astolfi L, Babiloni F, et al. A neural mass model for the
of EEG patterns, but also formulate hypotheses to be tested simulation of cortical activity estimated from high resolution EEG
under experimental conditions. during cognitive or motor tasks. J Neurosci Methods. 2006;
In the course of the last decades, a shift of attention from 157(2):317–329.
18. Zavaglia M, Astolfi L, Babiloni F, et al. The effect of connectivity on
models describing the behavior of local neuronal networks in
EEG rhythms, power spectral density and coherence among cou-
the temporal domain toward models that take into account the
pled neural populations: analysis with a neural mass model. IEEE
spatial properties of complex networks has occurred. Trans Biomed Eng. 2008;55(1):69–77.
Furthermore, an evolution can be seen in the techniques used. 19. Wendling F, Bartolomei F, Bellanger JJ, et al. Epileptic fast activity
In the early phase, the approach was based mainly on the use of can be explained by a model of impaired GABAergic dendritic
linear systems analysis, but more recently new mathematical inhibition. Eur J Neurosci. 2002;15(9):1499–1508.
tools for the analysis of nonlinear dynamic systems have been 20. van Rotterdam A, Lopes da Silva FH, Van den Ende J, et al. A model
introduced in this domain of theoretical neurophysiology. of the spatial-temporal characteristics of the alpha rhythm. Bull
Moreover, the availability of powerful computer tools opens Math Biol. 1982;44(2):283–305.
new possibilities for modeling both complex membrane phe- 21. Lopes da Silva FH, Storm van Leeuwen W. The cortical alpha
nomena and network properties. The analysis of the dynamics rhythm in dog: the depth and surface profile of phase. In: Brazier
MAB, Petsche H, eds. Architectonics of the Cerebral Cortex. New
of such networks is a challenge for the theoretical neurophysi-
York: Raven Press; 1978:319–333.
ologist interested in understanding EEG phenomena.
22. Harrison LM, David O, Friston KJ. Stochastic models of neuronal
dynamics. Phil Trans Biol Sci. 2005;360(1457):1075–1091.
23. Deco G, Jirsa VK, Robinson PA, et al. The dynamic brain: from
REFERENCES
spiking neurons to neural masses and cortical fields. PLoS Comput.
1. Freeman WJ. Mass Action in the Nervous System. New York: Biol. 2008;4(8):e1000092. doi:10.1371/journal.pcbi.1000092.
Academic Press; 1975. 24. Jirsa VK. Neural field dynamics with local and global connectivity
2. Hammond PH. Feedback Theory and Its Applications. London: and time delay. Philos Transact. Series A, Math Phys Eng Sci.
English University Press; 1958. 2009;367(1891):1131–1143.
3. Harmon LD. Problems in neural modeling. In: Reiss RF, ed. Neural 25. Jirsa VK, Haken H. Field theory of electromagnetic brain activity.
Theory and Modeling, Proceedings of the 1962 OGAI Symposium. Phys Rev Lett. 1996;77:960–963.
Stanford, CA: Stanford University Press; 1964. 26. Wright JJ, Robinson PA, Rennie CJ, et al. Toward an integrated
4. Izhikevich EM, Edelman GM. Large-scale model of mammalian continuum model of cerebral dynamics: the cerebral rhythms,
thalamocortical systems. Proc Natl Acad Sci. 2008;105(9): synchronous oscillation and cortical stability. Biosystems.
3593–3598. 2001;63(1–3):71–88.
5. Markram H. The blue brain project. Nat Rev Neurosci. 2006;7(2): 27. Wright JJ, Rennie CJ, Lees GJ, et al. Simulated electrocortical activity
153–160. at microscopic, mesoscopic, and global scales. Neuropsychopharma-
6. Watts DJ, Strogatz SH. Collective dynamics of “small-world” net- cology. 2003;28(suppl 1):S80–SA93.
works. Nature. 1998;393(6684):440–442. 28. Rennie CJ, Robinson PA, Wright JJ. Unified neurophysical model
7. Yu S, Huang D, Singer W, et al. A small world of neuronal syn- of EEG spectra and evoked potentials. Biol Cybern. 2002;86(6):
chrony. Cereb Cortex. 2008;18(12):2891–2901. 457–471.
8. Stam CJ, Reijneveld JC. Graph theoretical analysis of complex net- 29. Robinson PA, Whitehouse RW, Rennie CJ. Nonuniform corti-
works in the brain. Nonlinear Biomed Phys. 2007;1(1):3. cothalamic continuum model of electroencephalographic spectra
9. Wilson HR, Cowan JD. Excitatory and inhibitory interaction in with application to split-alpha peaks. Phys Rev E Stat Nonlin Soft
localized populations of model neurons. Biophys J. 1972;12:1–23. Matter Phys. 2003;68(2 pt 1):021922.
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 87

30. Friston KJ, Harrison L, Penny W. Dynamic causal modeling. 52. Traub RD. Simulation of intrinsic bursting in CA3 hippocampal
NeuroImage. 2003;19:1273–1302. neurons. Neuroscience. 1982;7:1233–1242.
31. Stephan KE, Harrison LM, Kiebel SJ, et al. Dynamic causal models 53. Traub RD, Wong RKS. Synaptic mechanisms underlying interictal
of neural system dynamics: current state and future extensions. spike initiation in a hippocampal network. Neurology. 1983;33:
J Biosci. 2007;32(1):129–144. 258–266.
32. Valdes-Sosa PA, Sanchez-Bornot JM, Sotero RC, et al. Model 54. Traub RD, Knowles WD, Miles R, et al. Synchronized afterdis-
driven EEG/fMRI fusion of brain oscillations. Hum Brain Mapp. charges in the hippocampus: simulation studies of the cellular
2009;30(9):2701–2721. mechanism. Neuroscience. 1984;12:1191–1200.
33. Freeman WJ. Nonlinear dynamics of paleocortex manifested in the 55. Traub RD, Draguhn A, Whittington MA, et al. Axonal gap junc-
olfactory EEG. Biol Cybern. 1979;35:21–34. tions between principal neurons: a novel source of network oscil-
34. Freeman WJ. Analytic techniques used in the search for the physi- lations, and perhaps epileptogenesis. Rev Neurosci. 2002;13
ological basis of the EEG. In: Gevins A, Remond A, eds. Handbook (1):1–30.
of Electroencephalography and Clinical Neurophysiology, vol. 1: 56. Traub RD, Miles R. Neuronal Networks of the Hippocampus.
Methods of Analysis of Brain Electrical and Magnetic Signals. Cambridge: Cambridge University Press; 1991.
Amsterdam: Elsevier; 1986. 57. Traub RD, Whitington MA, Colling SB, et al. Analysis of gamma
35. Nunez PL, ed. Neocortical Dynamics and Human EEG Rhythms. rhythms in the rat hippocampus in vitro and in vivo. J Physiol
New York: Oxford University Press; 1995. (Lond.) 1996;493:471–484.
36. Wright JJ, Liley DTJ. Dynamics of the brain at global and micro- 58. Jefferys JG, Traub RD, Whittington MA. Neuronal networks for
scopic scales: neural networks and the EEG. Behav Brain Sci. induced “40 Hz” rhythms. Trends Neurosci. 1996;19:202–208.
1996;19:285–320. 59. Jefferys JG, Traub RD, Whittington MA. Reply. Trends Neurosci.
37. Stam CJ, Pijn JP, Suffczynski P, et al. Dynamics of the human alpha 1996;19:469–470.
rhythm: evidence for non-linearity? Clin Neurophysiol. 1999; 60. Whittington M, Traub RD. Inhibitory interneurons and network
110(10):1801–1813. oscillations in vitro. Trends Neurosci. 2003;26(12):676–682.
38. Valdes PA, Jimenez JC, Riera J, et al. Nonlinear EEG analysis based 61. Freeman WJ. Linear analysis of the dynamics of neural masses.
on a neural mass model. Biol Cybern. 1999;81(5–6):415–424. Annu Rev Biophys Bioeng. 1972;1:225–256.
39. Robinson PA, Loxley PN, O’Connor SC, et al. Modal analysis of 62. Gray CM, Freeman WJ, Skinner JE. Associative changes in the spa-
corticothalamic dynamics, electroencephalographic spectra, and tial amplitude patterns of rabbit olfactory EEG are norepinephrine
evoked potentials. Phys Rev E Stat Nonlin Soft Matter Phys. dependent. Neurosci Abstr. 1984;10(36,2):121.
2001;63(4 pt 1):041909. 63. Freeman WJ. A proposed name for aperiodic brain activity: sto-
40. Golomb D, Rinzel J. Dynamics of globally coupled inhibitory neu- chastic chaos. Neural Netw. 2000;13(1):11–13.
rons with heterogeneity. Physiol Rev E. 1993;48:4810–4814. 64. Andersen P, Andersson SA. Physiological Basis of Alpha Rhythm.
41. Golomb D, Rinzel J. Clustering in globally coupled inhibitory neu- New York: Appleton-Century-Crofts; 1968.
rons. Physica D. 1994;72:259–282. 65. Andersen P, Andersson SA, Lomo T. Nature of thalamocortical
42. Golomb D, Wang XJ, Rinzel J. Synchronization properties of spin- relations during spontaneous barbiturate spindle activity. J
dle oscillations in a thalamic reticular nucleus model. Physiol. 1967;192:283–307.
J Neurophysiol. 1994;72:1109–1126. 66. Steriade M, Jones EG, Llinás RR. Thalamic Oscillations and
43. Golomb D, Wang XJ, Rinzel J. Propagation of spindle waves in a Signaling. Neuroscience Institute Publications. New York: John
thalamic slice model. J Neurophysiol. 1996;75:750–769. Wiley & Sons; 1990.
44. Wang XJ. Multiple dynamical modes of thalamic relay neurons: 67. Steriade M, Gloor P, Llinás RR, et al. Report of the IFCN commit-
rhythmic bursting and intermittent phase-locking. Neuroscience. tee on basic mechanisms. Electroencephalogr Clin Neurophysiol.
1994;59:21–31. 1990;76:481–509.
45. Wang XJ, Rinzel J. Alternating and synchronous rhythms in recip- 68. Destexhe A, Contreras D, Sejnowski TJ, et al. Modeling the control
rocally inhibitory model neurons. Neural Comp. 1992;4:84–97. of reticular thalamic oscillations by neuromodulators. Neuroreport.
46. Wang XJ, Rinzel J. Spindle rhythmicity in the reticularis thalami 1994;5:2217–2220.
nucleus: synchronization among mutually inhibitory neurons. 69. Destexhe A, Bal T, McCormick DA, et al. Ionic mechanisms under-
Neuroscience. 1993;53:899–904. lying synchronized oscillations and propagating waves in a model
47. Wang XJ, Rinzel J, Rogawski MA. A model of the T-type calcium of ferret thalamic slices. J Neurophysiol. 1996;76:2049–2070.
current and the low threshold spike in thalamic neurons. 70. Jahnsen H, Llinás R. Electrophysiological properties of guinea-pig
J Neurophysiol. 1991;66:839–850. thalamic neurones: an in vitro study. J Physiol (Lond.) 1984;349:
48. Wang XJ, Golomb D, Rinzel J. Emergent spindle oscillations and 205–226.
intermittent burst firing in a thalamic model: specific neuronal 71. Jahnsen H, Llinás R. Ionic basis for the electro-responsiveness and
mechanisms. Proc Natl Acad Sci USA. 1995;92:5577–5581. oscillatory properties of guinea-pig thalamic neurons in vitro.
49. Suffczynski P, Kalitzin S, Pfurtscheller G, et al. Computational J Physiol (Lond.) 1984;349:227–247.
model of thalamo-cortical networks: dynamical control of alpha 72. Steriade M, Deschènes M, Domich L, et al. Abolition of spindle
rhythms in relation to focal attention. Int J Psychophysiol. 2001; oscillations in thalamic neurons disconnected from nucleus retic-
43(1):25–40. ularis thalami. J Neurophysiol. 1985;54:1473–1497.
50. Suffczynski P, Kalitzin S, Lopes da Silva FH. Dynamics of non-con- 73. Avanzini G, de Curtis M, Panzica F, et al. Intrinsic properties of
vulsive epileptic phenomena modeled by a bistable neuronal net- nucleus reticularis thalami neurons of the rat studied in vitro.
work. Neuroscience. 2004;126(2):467–484. J Physiol. 1989;416:111–122.
51. Wendling F, Hernandez A, Bellanger JJ, et al. Interictal to ictal transi- 74. Bal T, von Krosigk M, McCormick DA. Synaptic and membrane
tion in human temporal lobe epilepsy: insights from a computational mechanisms underlying synchronized oscillations in the ferret
model of intracerebral EEG. J Clin Neurophysiol. 2005;22(5):343–56. LGNd in vitro. J Physiol (Lond.) 1995;483:641–663.
88 Part I ■ Basic Principles

75. Bal T, von Krosigk M, McCormick DA. Role of the ferret perigenic- 95. Szentagothai J. The local neuronal apparatus of the cerebral
ulate nucleus in the generation of synchronized oscillations in cortex. In: Buser P, Rougeaul-Buser A, eds. Cerebral Correlates of
vitro. J Physiol (Lond.) 1995;483:665–685. Conscious Experience, Inserm Symposium no. 6. Amsterdam:
76. Huguenard JR, Prince DA. Intrathalamic rhythmicity studied in Elsevier; 1978:131–138.
vitro: nominal T-current modulations causes robust anti-oscillatory 96. Katchalsky AK, Rowland V, Blumenthal R. Dynamic patterns of
effects. J Neurosci. 1994;14:5485–5502. brain cell assemblies. Neurosci Res Prog Bull. 1974;12(1):1–187.
77. Von Krosigk M, Bal T, McCormick DA. Cellular mechanisms of 97. Tombol T. Comparative data on the Golgi architecture of
a synchronized oscillation in the thalamus. Science. 1993;261: interneurons of different conical areas in cat and rabbit. In:
361–364. Brazier MAB, Petsche H, eds. Architectonics of the Cerebral Cortex.
78. Warren RA, Agmon A, Jones EG. Oscillatory synaptic interactions New York: Raven Press; 1978:59–76.
between ventroposterior and reticular neurons in mouse thalamus 98. Liley DTJ. A continuum model of the mammalian alpha rhythm.
in vitro. J Neurophysiol. 1994;72:1993–2003. In: Lopes da Silva F, Cadusch JC, Borges de Almeida L, eds.
79. Landisman CE, Long MA, Beierlein M, et al. Electrical synapses in Spatiotemporal Models in Biological and Artificial Systems.
the thalamic reticular nucleus. J Neurosci. 2002;22:1002–1009. Amsterdam: IOS Press; 1997:89–96.
80. Steriade M, Llinás RR. The functional states of the thalamus and 99. Liley DTJ, Cadusch PC, Wright JJ. A continuum theory of elec-
the associated neuronal interplay. Physiol Rev. 1988;68:649–742. tro-cortical activity. Neurocomputing. 1999;26–27:795–800.
81. McCormick DA. Neurotransmitter actions in the thalamus and 100. Liley DTJ, Cadusch PJ, Dafilis MP. A spatially continuous mean
cerebral cortex and their role in neuromodulation of thalamocor- field theory of electrocortical activity. Network Comp Neural Sys.
tical activity. Prog Neurobiol. 1992;39:337–388. 2002;13:67–113.
82. McCormick DA, Bal T. Sensory gating mechanisms of the thala- 101. Contreras D, Steriade M. Spindle oscillation in cats: the role of
mus. Curr Opin Neurobiol. 1994;4:550–556. corticothalamic feedback in a thalamically generated rhythm. J
83. Destexhe A, Sejnowski TJ. Interactions between membrane con- Physiol (Lond.) 1996;490:159–179.
ductances underlying thalamocortical slow-wave oscillations. 102. Zetterberg LH, Kristiansson L, Mossberg K. Performance of a
Physiol Rev. 2003;83(4):1401–1453. model for a local neuron population. Biol Cybern. 1978;31:15–26.
84. Hughes SW, Crunelli V. Thalamic mechanisms of EEG alpha 103. Babloyantz A, Kaczmarek LK. Self-organization in biological sys-
rhythms and their pathological implications. Neuroscientist. 2005; tems with multiple cellular contacts. Bull Math Biol. 1979;41:
11(4):357–372. 193–201.
85. Lörincz ML, Crunelli V, Hughes SW. Cellular dynamics of cholin- 104. Traub RD, Wong RKS. Synchronized burst discharge in disinhib-
ergically induced alpha (8–13 Hz) rhythms in sensory thalamic ited hippocampal slice. II. Model of cellular mechanism. J
nuclei in vitro. J Neurosci. 2008;28(3):660–671. Neurophysiol. 1983;49:442–458.
86. Lopes da Silva FH, van Lierop THMT, Schrijer CF, et al. Organization 105. Traub RD, Wong RKS. Cellular mechanism of neuronal synchro-
of the thalamic and cortical alpha rhythms: spectra and coherences. nization in epilepsy. Science. 1982;216:745–747.
Electroencephalogr Clin Neurophysiol. 1973;35:627–639. 106. Traub RD, Jefferys JG, Whittington MA. Functionally relevant
87. Chatile M, Milleret C, Buser P, et al. A 10 Hz “alpha-like” rhythm and functionally disruptive (epileptic) synchronized oscillations
in the visual cortex of the waking cat. Electroencephalogr Clin in brain slices. Adv Neurol. 1999;79:709–724.
Neurophysiol. 1992;83:217–230. 107. Traub RD, Bibbig A, LeBeau FE, et al. Cellular mechanisms of
88. Lanoir J. Étude électrocorticographique de la veille et du sommeil neuronal population oscillations in the hippocampus in vitro.
chez le chat, organisation du cycle nycthéméral, rôle du thalamus. Annu Rev Neurosci. 2004;27:247–278.
Thesis Doctor of Science, Centre Régional de recherche et de doc- 108. Trevelyan AJ, Sussillo D, Watson BO, et al. Modular propagation
umentation Pédagogiques, Marseille, France, 1972. of epileptiform activity: evidence for an inhibitory veto in neo-
89. Zetterberg LH. Stochastic activity in a population of neurons—A cortex. J Neurosci. 2006;26(48):12447–12455.
system analysis approach. Rep Inst Med Physics, TNO, Utrecht 109. Trevelyan AJ, Sussillo D, Yuste R. Feedforward inhibition con-
1973;1:53. tributes to the control of epileptiform propagation speed. J
90. Lopes da Silva FH, Vos JE, Mooibroek J, et al. Partial coherence Neurosci. 2007;27(13):3383–3387.
analysis of thalamic and cortical alpha rhythms in dog—a contri- 110. Kager H, Wadman WJ, Somjen GG. Simulated seizures and spread-
bution towards a general model of the cortical organization of ing depression in a neuron model incorporating interstitial space
rhythmic activity. In: Pfurtscheller G, Buser P, Lopes da Silva FH, and ion concentrations. J Neurophysiol. 2000;84(1):495–512.
eds. Rhythmic EEG Activities and Cortical Functioning. Amsterdam: 111. Kager H, Wadman WJ, Somjen GG. Conditions for the triggering
Elsevier; 1980:33–59. of spreading depression studied with computer simulations. J
91. Lopes da Silva FH, Vos JE, Mooibroek J, et al. Relative contribu- Neurophysiol. 2002;88(5):2700–2712.
tions of intracortical and thalamocortical processes in the genera- 112. Kager H, Wadman WJ, Somjen GG. Seizure-like afterdischarges
tion of alpha rhythms, revealed by partial coherence analysis. simulated in a model neuron. J Comput Neurosci. 2007;22(2):
Electroencephalogr Clin Neurophysiol. 1980;50:449–456. 105–128.
92. Braitenberg V. On the Texture of Brains. New York: Springer-Verlag; 113. Borck C, Jefferys JGR. Seizure-like events in disinhibited ventral
1977. slices of adult rat hippocampus. J Neurophysiol. 1999;82:
93. Chow KL, Leimann AL. Aspects of the structural and functional 2130–2142.
organization of the neocortex. Neurosci Res Prog Bull. 1970;8(2): 114. Allen PJ, Fish DR, Smith SJ. Very high-frequency rhythmic activ-
157–183. ity during SEEG suppression in frontal lobe epilepsy.
94. Szentagothai J. Specificity versus (quasi) randomness in cortical Electroencephalogr Clin Neurophysiol. 1992;82(2):155–162.
connectivity. In: Brazier MAB, Petsche H, eds. Architectonics of the 115. Bragin A, Engel J Jr, Wilson CL, et al. Hippocampal and entorhinal
Cortex. New York: Raven Press; 1978:77–97. cortex high-frequency oscillations (100–500 Hz) in human epilep-
Chapter 4 ■ Dynamics of EEGs as Signals of Neuronal Populations: Models and Theoretical Considerations 89

tic brain and in kainic acid-treated rats with chronic seizures. 133. Roberts JA, Robinson PA. Modeling absence seizure dynamics: impli-
Epilepsia. 1999;40(2): 127–137. cations for basic mechanisms and measurement of thalamocortical
116. White JA, Banks MI, Pearce RA, et al. Networks of interneurons and corticothalamic latencies. J Theor Biol. 2008;253:180–201.
with fast and slow gamma-aminobutyric acid type A (GABA-A) 134. Meeren HK, Pijn JP, Van Luijtelaar EL, et al. Cortical focus drives
kinetics provide substrate for mixed gamma-theta rhythm. Proc widespread corticothalamic networks during spontaneous
Natl Acad Sci USA. 2000;97:8128–8133. absence seizures in rats. J Neurosci. 2002;22(4):1480–1495.
117. Banks MI, White JA, Pearce RA. Interactions between distinct 135. Lytton WW. Computer modelling of epilepsy. Nat Rev Neurosci.
GABA(A) circuits in hippocampus. Neuron. 2000;25(2): 2008;9:626–637.
449–457. 136. Steriade M. Mechanisms underlying cortical activation: neuronal
118. Cossart R, Dinocourt C, Hirsch JC, et al. Dendritic but not organization and properties of the midbrain reticular core and
somatic GABAergic inhibition is decreased in experimental intralaminar thalamic nuclei. In: Pompeiano O, Ajmone-Marsan
epilepsy. Nat Neurosci. 2001;4(1):52–62. C, eds. Brain Mechanisms and Perceptual Awareness. New York:
119. Kramer MA, Kirsch HE, Szeri AJ. Pathological pattern formation Raven Press; 1981:327–377.
and cortical propagation of epileptic seizures. J R Soc Interface. 137. Steriade M, Nunez A, Amzica F. A novel slow ( 1 Hz) oscillation
2005;2:113–127. of neocortical neurons in vivo: depolarizing and hyperpolarizing
120. Kramer MA, Lopour BA, Kirsch HE, et al. Bifurcation control of components. J Neurosci. 1993;13:3252–3265.
a seizing human cortex. Phys Rev E Stat Nonlin Soft Matter Phys. 138. Steriade M, Nunez A, Amzica F. Intracellular analysis of relations
2006;73(4 pt 1):041928. between the slow ( 1 Hz) neocortical oscillation and other sleep
121. Kramer MA, Szeri AJ, Sleigh JW, et al. Mechanisms of seizure rhythms of the electroencephalogram. Neuroscience. 1993;13:
propagation in a cortical model. J Comput Neurosci. 2007;22(1): 3266–3283.
63–80. 139. Bazhenov M. Timofeev I, Steriade M, et al. Model of thalamocor-
122. Lytton WW, Hellman KM, Sutula TP. Computer models of hip- tical slow-wave sleep oscillations and transitions to activated
pocampal circuit changes of the kindling model of epilepsy. Artif states. J Neurosci. 2002;22(19):8691–8704.
Intell Med. 1998;13(1–2):81–97. 140. Ball GJ, Gloor R, Schaul N. The cortical electromicrophysiology
123. Santhakumar V, Aradi I, Soltesz I. Role of mossy fiber sprouting of pathological delta waves in the electroencephalogram of cats.
and mossy cell loss in hyperexcitability: a network model of the Electroencephalogr Clin Neurophysiol. 1977;43:346–361.
dentate gyrus incorporating cell types and axonal topography. J 141. Nunez PL. Electric Field of the Brain. New York: Oxford
Neurophysiol. 2005;93(1):437–453. University Press; 1981.
124. Dyhrfjeld-Johnsen J, Santhakumar V, Morgan RJ, et al. 142. Nunez PL. Generation of human EEG by a combination of long
Topological determinants of epileptogenesis in large-scale struc- and short range neocortical interactions. Brain Topogr. 1989;1:
tural and functional models of the dentate gyrus derived from 199–215.
experimental data. J Neurophysiol. 2007;97:1566–1587. 143. Nunez PL. Toward a quantitative description of large-scale neo-
125. Gorter JA, van Vliet EA, Aronica E, et al. Progression of sponta- cortical dynamic function and EEG. Behav Brain Sci. 2000;23(3):
neous seizures after status epilepticus is associated with mossy 371–398; discussion 399–437.
fiber sprouting and extensive bilateral loss of hilar parvalbumin 144. Nunez PL, Wingeier BM, Silberstein RB. Spatial-temporal struc-
and somatostatin-immunoreactive neurons. Eur J Neurosci. tures of human alpha rhythms: theory, microcurrent sources,
2001;13:657–669. multiscale measurements, and global binding of local networks.
126. Blumcke I, Suter B, Behle K, et al. Loss of hilar mossy cells in Hum Brain Mapp. 2001;13:125–164.
Ammon’s horn sclerosis. Epilepsia. 2000;41(suppl. 6):S174–S180. 145. Montaron MF, Bouyer JJ, Rougeul A, et al. Ventral mesencephalic
127. Lopes da Silva F, Blanes W, Kalitzin SN, et al. Dynamical diseases tegmentum (VMT) controls electrocortical beta rhythms and
of brain systems: different routes to epileptic seizures. IEEE Trans associated alternative behavior in the cat. Behav Brain Res.
Biomed Eng. 2003;50:540–549. 1982;6:129–145.
128. Van Drongelen W, Lee HC, Hereld M, et al. Emergent epilepti- 146. Lopes da Silva FH, van Rotterdam A, Storm van Leeuwen W, et
form activity in neural networks with weak excitatory synapses. al. Dynamic characteristics of visual EP’s in the dog. II. Beta fre-
IEEE Trans Neural Syst Rehabil Eng. 2005;13(2):236–241. quency selectivity in evoked potentials and background activity.
129. Lytton WW, Confreras D, Destexhe A, et al. Dynamic interactions Electroencephalogr Clin Neurophysiol. 1970;29: 260–268.
determine partial thalamic quiescence in a computer network 147. Gray C, Konig P, Engel A, et al. Oscillatory responses in cat visual
model of spoke-and-wave seizures. J Neurophysiol. 1997;77: cortex exhibit intercolumnar synchronization which reflects
1676–1696. global stimulus properties. Nature. 1988;338:334–337.
130. Tsakiridou E, Bertollini L, de Curtis M, et al. Selective increase in 148. Singer W, Gray CH. Visual feature integration and the temporal
T-type calcium conductance of reticular thalamic neurons in a correlation hypothesis. Annu Rev Neurosci. 1995;18:555–586.
rat model of absence epilepsy. J Neurosci. 1995;15:3310–3317. 149. Freeman WJ, Burke BC, Holmes MD. Aperiodic phase resetting
131. Leresche N, Parri HR, Erdemli G, et al. On the action of the in scalp EEG of beta-gamma oscillations by state transitions at
antiabsence drug ethosuximide in the rat and cat thalamus. J alpha-theta rates. Hum Brain Mapp. 2003;19(4):248–272.
Neurosci. 1998;18(13):4842–4853. 150. Howard MW, Rizzuto DS, Caplan JB, et al. Gamma oscillations
132. Suffczynski P, Pijn JPM, Pfurtscheller G, et al. Event-related dynam- correlate with working memory load in humans. Cerebral Cortex.
ics of alpha band rhythms: a neuronal network model of focal 2003;13:1369–1374.
ERD/surround ERS. In: Pfurtscheller G, Lopes da Silva F, eds. Event 151. Pfurtscheller G, Graimann B, Huggins JE, et al. Spatiotemporal
Related Desynchronization, Handbook of Electroencephalography and patterns of beta desynchronization and gamma synchronization
Clinical Neurophysiology, Revised Series vol. 6. Amsterdam: Elsevier in corticographic data during self-paced movement. Clin
Science; 1999:67–85. Neurophysiol. 2003;114(7):1226–1236.
90 Part I ■ Basic Principles

152. Sederberg PB, Kahana MJ, Howard MW, et al. Theta and gamma 167. Rapp PE, Albano AM, Schmah TI, et al. Filtered noise can mimic
oscillations during encoding predict subsequent recall. J low-dimensional chaos. Phys. Rev. E. 1993;47:2289–2297.
Neurosci. 2003;23:10809–10814. 168. Basar E. Chaotic dynamics and resonance phenomena in brain
153. Boeijinga PH, Lopes da Silva FH. Differential distribution of beta function: progress, perspectives, and thoughts. In: Basar E, Bullock
and theta EEG activity in the entorhinal cortex of the cat. Brain TH, eds. Chaos in Brain Function. Berlin: Springer; 1990:1–31.
Res. 1988;448:272–286. 169. Casdagli MC, Iasemidis LO, Savit RS, et al. Nonlinearity in inva-
154. Kay LM, Lancaster LR, Freeman WJ. Reafference and attractors in sive EEG recordings from patients with temporal lobe epilepsy.
the olfactory system during odor recognition. Int J Neural Syst. Electroencephalogr Clin Neurophysiol. 1997;102:98–105.
1996;7:489–495. 170. Elbert T, Ray WJ, Kowalik JE, et al. Chaos and physiology: determin-
155. Leung LS. Recurrent inhibition model of hippocampal CA in istic chaos in excitable cell assemblies. Physiol Rev. 1994;74:1–47.
vivo (letter to editor). Trends Neurosci. 1996;19:468–469. 171. Jansen BH. Is it and so what? A critical review of EEG chaos. In:
156. Freeman WJ. Feedback models of gamma rhythms (letter to edi- Duke DW, Pritchard WS, ed. Measuring Chaos in the Human
tor). Trends Neurosci. 1996;19:468. Brain. Singapore: World Scientific; 1991:49–82.
157. Whittington MA, Traub RD, Kopell N, et al. Inhibition-based 172. Pijn JP, Velis DN, Lopes da Silva FH. Chaos or noise in EEG sig-
rhythms: experimental and mathematical observations on net- nals: dependence on state and brain site. Electroencephalogr Clin
work dynamics. Int J Psychophysiol. 2000;38(3):315–336. Neurophysiol. 1991;79:371–381.
158. Perez Velasquez JLP, Carlen P. Gap junctions, synchrony and 173. Theiler J, Rapp PE. Reexamination of the evidence for lowdimen-
seizures. Trends Neurosci. 2000;23:68–74. sional nonlinear structure in the human electroencephalogram.
159. Traub RD, Whittington MA, Buhl EH, et al. A possible role for gap Electroencephalogr Clin Neurophysiol. 1996;99:213–222.
junctions in generation of very fast EEG oscillations preceding the 174. Varela F, Lachaux JP, Rodriguez E, et al. The brainweb: phase syn-
onset of, and perhaps initiating, seizures. Epilepsia. 2001;42:153–170. chronization and large-scale integration. Nat Rev Neurosci.
160. Pinto DJ, Jones SR, Kaper TJ, et al. Analysis of state-dependent 2001;2(4):229–239.
transitions in frequency and long-distance coordination in a 175. Pijn JP, Velis DN, van der Heyden M, et al. Nonlinear dynamics
model oscillatory cortical circuit. J Comput Neurosci. 2003;15(2): of epileptic seizures on basis of intracranial EEG recordings.
283–298. Brain Topogr. 1997;9(4):249.
161. Menon V, Freeman WJ, Cutillo BA, et al. Spatio-temporal corre- 176. Lopes da Silva FH, Pijn JP. Epilepsy: network models of genera-
lations in human gamma band electrocorticograms. Electroen- tion. In: Arbib MA, ed. The Handbook of Brain Theory and Neural
cephalogr Clin Neurophysiol. 1996;98:89–102. Networks. Cambridge, MA: A Bradford Book, MIT Press;
162. Babloyantz A. Strange attractors in the dynamics of brain activ- 1995:367–369.
ity. In: Haken H, ed. Complex Systems Operational Approaches. 177. Breakspear M. “Dynamic” connectivity in neural systems: theoret-
Berlin: Springer-Verlag; 1985:116–123. ical and empirical considerations. Neuroinformatics. 2004;2(2):
163. Schuster HG. Deterministic Chaos. Weinheim: PhysikVerlag; 1984. 205–226
164. Grassberger P, Procaccia I. Measuring the strangeness of strange 178. Lopes da Silva FH. Neural mechanisms underlying brain waves:
attractors. Physica. 1983;9D:189–208. from neural membranes to networks. Electroencephalogr Clin
165. Grassberger P, Schreiber T, Schaffrath C. Chaos. Int J Bifurc. Neurophysiol. 1991;79:81–93.
1991;1:521–547. 179. Freeman WJ. Dynamics of image formation by nerve cell assem-
166. Ott E. Chaos in Dynamical Systems. New York: Cambridge blies. In: Basar E, Flohr H, Haken H, et al., eds. Synergetics of the
University Press; 1993. Brain. Berlin: Springer-Verlag; 1983:102–121.
CHAPTER
Biophysical Aspects of EEG and
Magnetoencephalogram Generation
FERNANDO H. LOPES DA SILVA WITH AN APPENDIX BY AB VAN ROTTERDAM
5
T
he electrical activity of the brain consists of ionic cur- laminar current along the main axes of the neurons. The net
rents generated by biochemical sources at the cellular membrane current that results from the activation at the level
level. These ionic currents cause electric and magnetic of the synapse can be either a positive or a negative ionic cur-
fields that can be measured in the brain and surrounding tis- rent directed to the inside of the cell. Because there is no accu-
sues. The behavior of these fields can be predicted because they mulation of charge anywhere in the medium (see also
obey physical laws. This chapter is an introduction to the bio- Appendix, expression 5.1), the injected current at the synaptic
physical aspects of the generation of electroencephalogram level is compensated by other currents flowing in the medium,
(EEG) signals. It is convenient to consider the generation of as shown in Figure 5.1. At the level of the synapse and in the case
EEG signals in biophysical terms because this is the exact way to of an excitatory postsynaptic potential (EPSP), the synaptic cur-
determine the potential distribution at the scalp given a set of rent is carried by positive ions; in the case of an inhibitory
intracerebral current sources, that is, the so-called forward postsynaptic potential (IPSP), the corresponding current is
problem of electroencephalography. An understanding of this carried by negative ions. Because the direction of a current is
problem is necessary to discuss the inverse problem that consti- defined by the direction along which the positive charge is
tutes the main concern of clinical electroencephalography,
which is to determine the intracerebral sources given a meas-
ured potential distribution at the scalp. The inverse problem
has no unique solution, as shown long ago by Helmholtz (1);
therefore, it is essential to understand the forward problem so
that constraints of the inverse problem may be well examined.
This chapter treats the biophysical basis of the generation of
EEG potentials nonmathematically so that the general reader
may follow it more easily; in the theoretical appendix, a mathe-
matical treatment of the biophysical aspects is provided.

SPACE-DEPENDENT PROPERTIES
The electrical potential at a point in the brain in principle can
be computed if the microscopic cellular sources are known. In
expression 5.8 of the Appendix, the field potential is given as a
function of intracerebral current sources; in expression 5.10, it
is given in terms of the membrane potential.
In general terms, the field potential of a population of neu- Figure 5.1 Current flow patterns in and around an idealized neuron
rons equals the sum of the field potentials of the individual owing to synaptic activation. E, current flow caused by activation of a
neurons. To understand EEG phenomena, the activity of popu- synapse at the level of the apical dendrite resulting in depolarization of
lations of neurons must always be considered. EEG phenomena the membrane and flow of a net positive current. This current causes a
can be measured only at a considerable distance from the sink at the site of the synapse. The extracellularly measured excitatory
source if the responsible neurons are regularly arranged and postsynaptic potential (EPSP) is drawn at the left; it has a negative
activated in a more or less synchronous way. A typical regular polarity at the level of the synapse. At the soma there exists a distrib-
arrangement is the palisade, in which the neurons are distrib- uted passive source resulting in an extracellular potential of positive
uted with the main axes of the dendritic trees parallel to each polarity. I, current flow caused by activation of a synapse at the level of
other and perpendicular to the cortical surface. When in such a the soma resulting in hyperpolarization of the membrane and flow of a
population (e.g., the pyramidal neurons of layers IV and V of the net negative current. This results in an active source at the level of the
cortex) the neurons are more or less simultaneously activated soma and passive sinks at the basal and distal dendrites. The extracel-
by way of synapses lying at the proximal dendrites, extracellular lularly measured inhibitory postsynaptic potentials (IPSPs) at the soma
currents will flow; their longitudinal components (i.e., parallel and dendritic level are drawn. Note that an inhibition at the soma gen-
to the main axes of the neurons) will add, whereas their trans- erates about the same extracellular potential field as an excitation at the
verse components will tend to cancel out. The result is a apical or distal dendrites.

91
92 Part I ■ Basic Principles

transported, the ionic current is directed to the intracellular


medium with an EPSP; with an IPSP, it is directed to the extra-
cellular medium. Therefore, at the level of the synapse, there is
an active sink in the case of a positive ionic current (EPSP) or
an active source in the case of a negative ionic current (IPSP).
The extracellular potential at the sink is negative; at the source,
it is positive, as can be seen from expression 5.8. Along the cell
and at a distance from the synaptic level, there exists a distrib-
uted passive source in the case of an EPSP or a distributed pas-
sive sink in the case of an IPSP.
In addition to postsynaptic potentials, other relatively slow
variations of membrane potential, such as those associated with
depolarizing or hyperpolarizing afterpotentials and dendritic
events as calcium action potentials, may also be the sources of
extracellularly measurable potentials.
At a macroscopic level, it may therefore be stated that the
potential field generated by a synchronously activated palisade
of neurons behaves like that of a dipole layer. This is no more
than a rough model of reality. It may also be considered that an
active sink (for instance, at the level of the cell somata in the
cortex) is flanked by two passive sources, one lying more super-
ficially and the other deeper than the sink; in such a case, the
potential field should behave more like a quadrupole layer. This
is, however, an unnecessary complication because in general the
two sides of the quadrupole are rather asymmetrical; one is
usually dominant. This is to be expected because it can be seen
from histological sections of the cortex that there is a clear Figure 5.2 Examples of closed, open, and open-closed fields for differ-
asymmetry of the pyramidal cells along a direction perpendicu- ent types of neuron pools in the central nervous system. Left: The pop-
lar to the cortex. These cells present a morphological polariza- ulations are drawn. Right: Neurons representing the simultaneously
tion in the vertical direction because they are characterized by a activated pools are shown together with the arrows representing the
rather long vertically directed apical dendrite that ramifies in lines of flow of current at the instant when the impulses have evaded
the most superficial layers of the cortex and by basal dendrites the cell bodies. The zero isopotential lines (0) are also indicated.
distributed around the soma; the axon runs vertically down- A: Oculomotor nucleus represented schematically by only one neuron
ward to the white matter. For the sake of simplicity, it therefore with dendrites oriented radially outward. The isopotential lines are cir-
may be assumed that most potential fields generated in pal- cles; the currents flow entirely within the nucleus, resulting in closed
isades of neurons can be mimicked by simple dipole layers. field (all points outside the nucleus remain at zero potential).
Lorente de No (2) named this type of potential field the “open B: Superior olive represented by neurons each having a single dendrite
field” (Fig. 5.2C) (3), in contrast to those fields generated by oriented radially inward. The currents result in a closed field.
neurons with dendritic arborizations radially distributed about C: Accessory olive represented by only one neuron with a single long
the soma; according to his description, these would generate dendrite. The arrangement of sources and sinks in this structure permits
“closed fields” as shown in Figure 5.2A,B. In the latter case, the the spread of current in the volume of the brain and thus results in an
configuration of the radially oriented neurons is such that each open field. D: Two structures mixed together, generating an open-closed
neuron may be considered as generating a central source (or field. (Modified from Lorente de No R Action potential of the motoneu-
sink, depending on the type of synaptic activity) surrounded by rons of the hypoglossus nucleus. J Cell Comp Physiol. 1947;29: 207–287;
spherically distributed sinks (or sources). The field potential in Hubbard, J I, Llinas, R Quastel D M J Electrophysiological Analysis of
this case is equivalent to that of a distribution of radially ori- Synaptic Transmission. London: Edward Arnold Ltd; 1969.)
ented dipoles at the surface of a sphere. It can be seen intuitively
that, under these circumstances, not only the tangential but also
the radial components of current cancel each other; this can
also be proved by the volume conduction theory (Fig. 5.3) (4). variation caused by an action potential generates a field that is
Postsynaptic potentials in neuronal populations with an equivalent to that of a single dipole perpendicular to the mem-
appropriate spatial organization can be sources of field poten- brane because the piece of membrane that is depolarized at any
tials that can be measured at a distance and therefore are also instant of time is small. In contrast, that of an electrotonically
sources of EEG signals. Can other types of membrane poten- conducted postsynaptic potential extends, at any moment of
tials as action potentials also contribute to those EEG signals? time, over a larger portion of the membrane; thus, it generates
This is not generally the case for two reasons (see also a field that corresponds rather to that of a dipole layer with
Appendix). The first reason is that the membrane potential dipoles perpendicular to the membrane surface. The latter
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 93

Figure 5.3 Equipotential plots and depth profiles for open cortices. Intersection of base lines of the depth profiles is at
the center of the sphere. Tick marks represent the region of cells. Note that the 90-degree profile of the hemisphere and
45-degree profiles of the cap and punctured sphere run along the open edge of the cortex. (Adapted from Klee M, Rall W.
Computed potentials of cortically arranged populations of neurons. J Neurophysiol. 1977;40:647–666.)

attenuates with distance less rapidly than the former, as can be In those cases in which the action potentials occur simultane-
seen by comparing expressions 5.12 and 5.15 of the Appendix; ously in clusters (e.g., when a group of fibers is excited by a short
this is illustrated in Figure 5.10B and has also been discussed by stimulus), the field of these action potentials may also be
Humphrey (5). These properties have been analyzed more rig- recorded at relatively large distances in the form of what is gen-
orously by Pettersen and Einevoll (6) using realistic computer erally called a “compound action potential.”
models of pyramidal and stellate neurons with different mor- Quantitative measurements in vitro and computational neu-
phologies. These authors demonstrated that the neural mor- ronal models used to infer contributions of neuronal populations
phology and passive electrical parameters influence the width to MEG and EEG signals.
and amplitude of extracellular spikes. These modeling studies Studies using in vitro preparations allow the simultaneous
revealed that in all neurons there is a low-pass filtering effect, that measurement of electric and magnetic fields generated by neu-
is, the spike-width increases with increasing distance from soma. ronal activity. A calculation of the magnetic field around an
These authors showed quantitatively that the spike-amplitude axon was performed by Swinney and Wikswo (7) using a prepa-
decays with distance r depending on dendritic morphology, ration where an isolated axon was kept in a bath with saline, in
according to the rule 1/rn with 1 n 2 close to the soma and vitro, and was stimulated electrically. These authors found that
n 2 far away along the dendritic tree. These studies have also the magnetic field resulted mainly from the intracellular cur-
implications for understanding the relation between the extent of rent flowing inside the axon along the axial direction, whereas
fields produced by multiunit activity (MUA) and by synaptic the contribution of the return passive current that flows in the
potentials, which constitute the so-called local field potentials medium was two orders of magnitude smaller, and that of the
(LFPs or local EEG), since they substantiate why MUAs are more transmembrane current was negligible.
confined around the neural sources than the LFPs generated by More recently, Murakami and Okada (8) applied the same
synaptic currents. In the latter case the dipole layer lengths are kind of approach to the neocortex. This study is particularly
much longer than the typical action-potential current dipoles, relevant because it has yielded some results that may help to
and thus the attenuation with distance is weaker than for the interpret EEG and magnetoencephalogram (MEG) recordings
MUAs. The second reason that action potentials do not con- from the scalp. These authors made a computer model, based
tribute to EEG signals is that the latter, owing to their short dura- on that proposed by Mainen and Sejnowski (9), of the four
tion (1 to 2 msec), tend to overlap much less than do postsynaptic main types of cortical neurons taking into account their real-
potentials (EPSP and IPSP), which last longer (10 to 250 msec). istic shapes. Each neuron is described as a three-dimensional
94 Part I ■ Basic Principles

compartmental model, where each compartment has its typical populations (14–18); the interlocking takes place by way of
geometric dimensions, passive electrical properties (membrane recurrent collaterals forming different types of synapses that
capacitance and resistance, intracellular resistance), and five possibly include dendrodendritic synapses and even gap junc-
voltage-dependent ionic conductances. Neuronal activity was tions (19).
obtained by stimulating each neuron with an intracellular cur- These neural masses act as macroscopic sources of EEG sig-
rent injected at the soma. The intracellular current is repre- nals. For example, in the case of the generation of alpha
sented by a vector quantity, the magnitude of which is the rhythms, it has been shown that such a macroscopic source is
current dipole moment and is represented by the scalar quan- located within the cortex and behaves like a dipole layer
tity Q in [pA m]. directed perpendicularly to the cortical surface (20). The
The population Q for a group of pyramidal neurons was cal- dynamic properties of alpha rhythms, such as their frequency,
culated by computing the sum of the dot product of each cur- depend on the parameters of the neuronal populations, such as
rent dipole Qk and the unit vector orthogonal to the cortical feedback gains, strength of connections, time properties of
surface. The same was done for the stellate cells, with a modifi- synaptic potentials, and nonlinear properties (i.e., thresholds
cation due to the fact that these neurons are differently ori- and saturation).
ented. The magnetic field B and electrical potential V at the Does this rhythmic activity propagate along the cortex or
scalp can be expressed as functions of the intracellular currents, form a standing field? It has been shown experimentally (20)
called primary currents, and the return currents that flow in the that, in the visual cortex of the dog, small cortical areas appear
space around the sources. In the model the magnetic field B is to act as “epicenters” from which alpha rhythm activity
assumed to be generated by the tangential component of Q, “spreads” in different directions. The existence of this type of
with respect to the inner skull surface, and the electric field is spread has been concluded on the basis of phase differences in
due to both the tangential and radial components. This implies rhythmic components measured at different locations on the
that B is mainly caused by neuronal activity in sulci, since Q is cortical surface. It has been shown in a model study (21) that a
directed perpendicularly to the pial surface, whereas both radial neuronal chain, where the model neurons are interconnected by
and tangential components contribute to the electric field. This means of recurrent collaterals and interneurons such that the
investigation led to another result of practical interest, namely it connectivity functions are homogeneous and their strength
gave some direct indications about how to interpret the magni- decays as an exponential function of distance, shows propaga-
tude of the magnetic fields recorded in human. According to the tion of electrical activities characterized by frequencies in the
model the overall magnitude of Q for the activity of pyramidal alpha band with a velocity c in the order of magnitude of
neurons of layers V and II/III is in the order of 0.29 to 0.90 pA 0.3 m/sec, which is in the range of experimental results. The
m. Apparently this value deviates appreciably from that esti- dynamics of the activity of neuronal populations are treated in
mated previously for a cell by Hämäläinen et al. (10) that was more detail in Chapter 4.
just 0.02 pA m. That larger value, however, is of the same order The propagation of electric activity in the cortex may result in
of magnitude as those estimated for hippocampal pyramidal the interesting phenomenon of time dilation, that is, an EEG
neurons (0.2 pA m per cell in CA1 and 0.17 pA m per cell in transient recorded at the cortex will seem to be of shorter dura-
CA3, (11,12)). Murakami and Okada (8) point out that assum- tion than the transient recorded simultaneously at the scalp. This
ing a Q of 0.2 pA m per cortical pyramidal neuron, a population has been shown to occur in relation to epileptiform spikes (22).
of 50,000 synchronously active cells would generate a field with The phenomenon of time dilation is due to the fact that a mov-
a magnitude of 10 nA m, which corresponds precisely to the ing dipole seen from a distance can be observed over a longer
smallest value measurable from the human cortex according to time than when seen from nearby. The physical basis for this phe-
Hämäläinen et al. (10). Assuming that a cortical minicolumn nomenon is accounted for in the Appendix (expression 5.18).
with a diameter of 40 m contains 100 cells (13), the cortical
surface that would correspond to 50,000 cells should form a INFLUENCE OF INHOMOGENEITIES
patch with about 0.63 mm 2 area. If this cortical patch would
have a circular form its diameter would be about 0.9 mm. It has been assumed that the brain could be considered an infi-
In order to estimate the number of cortical cells correspon- nite homogeneous medium, but in reality this is not the case.
ding to a cortical column, the synchronous activity of which is The field potentials are influenced not only by the geometry of
responsible for the EEG/MEG signal measured, one has to the neuronal populations and the electrical properties of indi-
know, at least approximately, the density and the spatial distri- vidual neurons but also by the existence of regions with differ-
bution of pyramidal and other cell types within the cortex. ent conductivities in the head, that is, by the presence of
inhomogeneities. For an interpretation of field potentials meas-
TIME-DEPENDENT PROPERTIES ured at the scalp, therefore, it is important to take into consid-
eration the layers lying around the brain: the cerebrospinal fluid
We have emphasized above the importance of synchrony of (CSF), the skull, and the scalp. These layers account, at least in
neuronal activity for the generation of EEG signals. We consider part, for the attenuation of EEG signals measured at the scalp as
here the factors that cause this synchrony. The main factor is of compared to those recorded at the underlying cortical surface.
a structural nature. Neuronal masses are, in general, organized This can be formulated in mathematical terms, as in the
as combinations of interlocked excitatory and inhibitory Appendix (expression 5.19).
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 95

To compute the potential distribution at the surface of the consisting of five concentric shells, solved the forward problem
scalp caused by a dipole placed within the brain, it is necessary in an analytic form for the case in which the skull and the cor-
to compute the potential distribution at the surface of the differ- tex had anisotropic properties. The effects of the anisotropic
ent shells (i.e., to solve the boundary value problem). This can be conductivity of the skull were determined by Bertrand et al.
done by solving the Poisson equation (see equation 5.7) in polar (40) and by Van den Broek (41). The latter showed that the
coordinates as done in the Appendix. Already in the 1970s, the anisotropy of the skull causes the smearing out of the distribu-
influence on the EEG of the specific electrical properties of the tion of the EEG over the scalp, whereas the normal component
tissues surrounding the brain was recognized (23–27). The sys- of the MEG is not affected.
tematic study of Ary et al. (28) led to the proposal of a method In addition, it is relevant to emphasize that the skull does not
that would correct the errors introduced by variations in skull have a homogeneous surface, due to both the existence of
and scalp thickness. Initially, the models of the head volume regions with different thickness and the occipital and the eye
conductor consisted of concentric spheres, usually three to sockets openings (42). The existence of holes in the skull, for
account for the brain, skull, and scalp; in some models a fourth example, due to surgical interventions, influences also the dis-
layer was included representing the CSF. In the classic models, tribution of the EEG over the scalp as demonstrated by
the three main concentric spheres typically had radii of 90, 83, Bertrand et al. (40). In general, the localization of the dipoles is
and 78 mm. The values of the conductivities of the three layers then shifted toward the hole.
commonly used were proposed by Geddes and Baker (29): for
the brain 0.33, for the skull 0.0042, and for the scalp also REALISTIC MODELS OF THE HEAD
0.33 S.m 1; in case the CSF was included, it had a conductivity AND NUMERICAL METHODS
of 1.0 S.m 1. Recently more precise estimates of these conduc-
tivities were obtained by way of measurements in vivo, using an In the past, most solutions of the forward problem assumed
electric impedance tomography (EIT) method combined with a that the head could be reasonably approximated by a spherical
realistic model of the head (30–32). The results of the study of model with three or four shells. However, the geometry of this
Gonçalves et al. (32) showed that the ratio between the conduc- volume conductor is clearly not spherical. Therefore, since the
tivities of the skull and the brain lies between 20 to 50 (for six late 1980s, realistically shaped models were gradually intro-
subjects) rather than the traditionally assumed value of 80. The duced and applied (43–47). This was made possible by the
average values found in this investigation are the following: advances in magnetic resonance imaging (MRI) that allow
brain: 0.33 S.m 1; skull: 0.0081 S.m 1. It is also important to making realistic estimates of the three-dimensional geometry
note that the intersubject variance of the estimates decreased by of the brain and surrounding tissues (48). To solve the forward
half when a realistic model was used, in comparison with a problem in a realistic shaped volume of the head, it is necessary
spherical model. However, a factor of 2.4 was found between the to apply numerical methods. This involves decomposing the
subjects with the extreme values of the ratio of conductivities. volume conductor in a mesh consisting of a relatively large
This implies that these values should be estimated for each indi- number of discrete elements. The accuracy of the method
vidual to increase the reliability of the estimated conductivities. increases with the number of these elements, and, of course, is
In addition, it is known that the conductivity of the various inversely proportional to their size, but the computational
tissues is not homogeneous. The conductivity of the skull varies effort also increases accordingly. The most commonly used
with its thickness and bone structure. Law et al. (33) showed methods to compute the EEG potential distributions are the
that there is an inverse relation between skull resistivity and boundary-element method (49–53), which is appropriate to the
thickness. Cuffin (34) and Eshel et al. (35) investigated the case where the volume conductor is assumed to be isotropic
effects of varying the conductivity of the skull; the former and piecewise homogeneous, and the volume element methods,
determined that the effect of local variations in skull and scalp which are needed in case the assumptions of isotropy and
thickness was slightly larger on the EEG than on the MEG, piecewise homogeneity do not hold. Examples of the latter are
while the latter showed a correlation between interhemispheric the finite-volume (35), the finite-difference, and the finite-
asymmetry in skull thickness and the amplitude of the scalp element methods (for details see reviews by van Uitert,
EEG. The very thorough studies of Haueisen (36,37) on the Weinstein, and Johnson (54)). These methods are powerful, but
influence of various combinations of conductivities on both the they need reliable information about the structure of the brain
EEG and MEG showed that the scalp EEG is most influenced by and surrounding tissues, obtained from MRI scans, and of the
the conductivities of the skull and the scalp, while the MEG was corresponding conductivities, to be applied in a sensible way.
most influenced by the conductivities of the brain tissue and They are particularly suited to calculate the influence of specific
the CSF. We should note that the conductivity of the tissues of inhomogeneities such as the influence of the cerebral ventricles
the head can also present anisotropy. Indeed, it is known that in filled with CSF (41) and anisotropic conductivities (36).
brain tissue, the conductivity measured in a direction parallel to In a number of studies, the advantage of using realistic mod-
a fiber tract can be 10 times larger than in the perpendicular els in comparison with spherical models has been demon-
direction (38). It is, however, difficult to integrate this finding in strated; differences in dipole reconstructions using EEG data
global models of the whole head, since fiber tracts are organized were reported to reach 20 mm (55,56). Relatively smaller differ-
in a most complex way within the anatomical constraints of the ences were obtained with MEG data, in the order of 4 mm (57).
folds of the brain. Nevertheless, De Munck (39), using a model This implies that the necessity of using realistic models,
96 Part I ■ Basic Principles

particularly those accommodating inhomogeneous volumes, is distribution. The number of sensors, however, is much smaller,
stronger for EEG than for MEG recordings. In the latter case, in the order of a maximum of 128 for the EEG and 300 for the
most calculations are based on the boundary-element method. MEG. Thus, the problem of estimating brain sources of EEG or
MEG data is underdetermined. Regularization methods have
THE INVERSE PROBLEM: APPROACHES been proposed to help solve this major difficulty.
TO ESTIMATE SOLUTIONS For solving the inverse problem, the method of the least-
squares source estimation has been applied both to the activity
The introduction stated that the problem of calculating the measured at a single moment in time or within a time interval
intracerebral sources of the potentials measured at the scalp, that (65,66). In this context a convenient approach, proposed by De
is, the so-called inverse problem of electroencephalography, has Munck (39,67), is to split the set of parameters of the ED into
no unique solution. This means that different combinations of linear and nonlinear parameters. This procedure consists
intracerebral sources can result in the same potential distribution schematically of the following steps: first, the time functions
at the scalp. The only way to tackle this problem is to make spe- that describe the change of the source as function of time must
cific assumptions about the intracerebral sources that are be estimated for a given position and orientation of the ED; sec-
assumed to cause a given EEG potential distribution and to make ond, the orientations must be found given the dipole time func-
a model of the conductive media lying between the sources and tions and position. These two steps must be performed
the recording electrodes. This implies that first a particular type alternatively a number of times until the best dipole orientation
of forward problem is solved; thereafter, the theoretical scalp and time functions are found for given dipole positions.
potentials obtained in this way are compared with those recorded Thereafter, the nonlinear position parameters must be updated
experimentally. It should be noted that, on the one hand, a satis- and the process repeated until a best fit is obtained. This proce-
factory comparison does not necessarily imply that the assumed dure starts from the assumption that the position and orienta-
sources are those really responsible for the measured EEG poten- tion of the initial dipole are known, that is, it is based on the
tials. On the other hand, however, a bad comparison must lead to stationary dipole model proposed by Scherg and Von Cramon
the conclusion that the assumptions are not correct with respect (65), although it is not constrained to the initial choice.
to the sources or to the model, or both. If a reasonable assumption on the initial position and orien-
The numerical procedures by means of which the measured tation cannot be made, one may use the alternative approach of
EEG/MEG distribution over the scalp is compared with that estimating EDs for a number of successive time samples of a
computed using the forward approach are not trivial. Usually given potential, or magnetic, distribution (so-called moving
one expresses the difference between the real and the simulated dipole approach) as done by Stok (64) and Stok et al. (68) and
distributions as a summed squared difference. The question is many others. In this way a series of EDs is obtained, as shown in
to find a minimum of this function, which can be accomplished Figure 5.4. Those EDs that have about the same position and ori-
by several algorithms. The mostly used of this is that of entation can be clustered and the clustered dipoles can be aver-
Marquardt (58). In the last decades many solutions of the aged. If desired, these average ED clusters can be used as seed
inverse problem have been proposed, starting from the pioneer points for the procedure of de Munck as described earlier (69).
work of Schneider and Gerin (59), Schneider (25,26), Smith et We should note that, in practice, if the distribution of the
al. (60), Henderson et al. (61), and many others as reviewed by EEG potential or of the MEG field over the scalp is relatively
Scherg (62,63) and Stok (64). simple, a single ED may be an appropriate model. Of course,
According to this approach, the intracerebral source is one needs all available a priori information to speed up the
assumed to be an equivalent current dipole localized within the solution and to give it high reliability. For instance, if it is to be
brain. Here we consider, in general terms, those aspects of the expected that a given activity occurs simultaneously in both
inverse problem that are common to EEG and MEG. The aim hemispheres in areas well defined anatomically, it is advisable to
of the inverse procedure is to obtain an equivalent source, start by assuming two symmetrical dipoles at the appropriate
which in general is a dipole, determined by six parameters. This sites (63). An increase in the number of dipoles can easily lead
solution is called the equivalent dipole (ED), which must be to rather complex and ambiguous interpretations. Validation of
considered a mathematical abstraction. It represents the theo- ED models can be obtained in retrospect by relating the solu-
retical dipole, which generates a potential (or magnetic field) tions obtained to independent physiological or anatomical
distribution at the surface of the outer shell, that is the closest information. An example of an anatomical–physiological vali-
in the least square sense to the measured distribution at the dation is the case of the dipolar sources of alpha, mu rhythms,
scalp. It should be emphasized that the estimation of an ED and sleep spindles (70) that are distributed on brain areas as
located within the brain gives only a rough estimate of the expected on the basis of animal and human physiological
center of gravity of the cortical area active at a given moment. observations. An example of anatomical–pathological valida-
For example, in the case of the dipole layer shown in Figure 5.3 tion is given by the distribution of sources of epileptiform tran-
the corresponding ED would be located much deeper than the sients in patients with cortical lesions visible in MRIs (Fig. 5.5),
surface where the individual dipolar sources are placed. where the main epileptiform sources are located around the
Assuming that the activity of each cortical macrocolumn lesions (71,72).
would be described by an equivalent current dipole, one would A number of alternative methods have been developed to
need several thousands to account for an EEG or MEG scalp circumvent the obvious limitations of the ED approach. One of
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 97

Figure 5.4 Projection of equivalent dipoles (EDs) based on visual evoked potentials (VEPs) (left) and visual evoked fields
(VEFs) (right) during the on-response of subject JM, to the appearance of a checkerboard presented to the left half-visual
field. Each ED is shown in three projection planes, namely occipital (top), horizontal (middle), and sagittal (bottom). An
arrow indicates the direction and length of the projection of the dipole and the starting point indicates the dipole location.
Each arrow is identified by a small number that runs from 1 to 11. A group of arrows represents an interval of 50 msec, in
steps of 5 msec, spanning the time interval of 55 to 160 msec after start of the stimulus. Scale: one division indicates 10 mm
for the position and 6.7 10–9 A m, for the components. Each dipole is plotted with a line width corresponding to the
measure of fit 2: thick for 0.1, medium for 0.1 0.2, thin for 0.2 0.4, very thin for 0.4. (Modified
from Stok CJ, Spekreijse HJ, Peters MJ, et al. A comparative EEG/ MEG equivalent dipole study of the pattern onset visual
response. New trends and advanced techniques in clinical neurophysiology. EEG Suppl. 1990;41:34–50.)

these consists in applying spatial filtering to the data so that sig- issue by creating new algorithms based on the assumption that
nals from a given location may be privileged with respect to the the sources are at fixed locations within the brain, namely at a
rest. This is the so-called linearly constrained minimum vari- given node of the triangular tessellation of the cortical surface
ance beam forming (73) that has been applied in practice with as extracted from the subject’s MRI. In this way the inverse
some interesting results (74). Related methods are the synthetic solution is simplified to finding the linear parameters of
aperture magnetometry (SAM) method introduced by the sources. Nevertheless, this problem is still underdetermined.
Robinson and Vrba (75), and the parametric mapping method The methods used to solve this problem use different forms of
applied by Dale et al. (76). In addition, methods have been pro- regularization parameters (80). In this way particular forms of
posed to obtain estimates of multiple dipoles with little a priori inverse solutions can be obtained: minimum norm (MN)
information, such as the multiple signal classification (MUSIC) (81–84), weighted resolution optimization (WROP) method of
algorithm (77–79). A number of laboratories have pursued this Grave de Peralta Menendez et al. (85), and the low-resolution
98 Part I ■ Basic Principles

Figure 5.5 Top: Channel overlays and


topographic maps of the average magnetic
fields for the five spike clusters in patient
C. The field maps represent the magnetic
field distribution at the time of the marker,
indicated in the channel overlays by the
dashed vertical line. Above each map the
cluster number and, within brackets, the
number of spikes it contains. Bottom:
Position of equivalent dipoles. In the top
axial and sagittal magnetic resonance
imaging (MRI) slices the dipoles fitted to
the average magnetoencephalogram
(MEG) spike of clusters 1 and 2, com-
bined. In the bottom slices are the dipole
locations for cluster 5. For clusters 3 and 4
equivalent dipole sources with less than
10% residual error were not found. The
boundary of the structural lesion in the
right frontal lobe for this patient is marked
by dotted lines. (Adapted from Van’t Ent
D, Manshanden I, Ossenblok P, et al. Spike
cluster analysis in neocortical localization
related epilepsy yields clinically significant
equivalent source localization results in
magnetoencephalogram [MEG]. Clin
Neurophysiol. 2003;114(10):1948–1962.)

brain electromagnetic tomography (LORETA). The latter was compared, in a computer simulation study, the performance of
proposed by Pascual-Marqui et al. (86) and uses the discrete the surface Laplacian and a linear estimation approach with
spatial Laplacian operator for regularization. This method dipoles having free moment orientations, the so-called cortical
yields a very smooth inverse solution (87). potential reconstruction based on linear estimation (Fig. 5.6).
In any case, dipole models provide less ambiguous solutions Compared to the surface Laplacian method, the latter linear esti-
than the more sophisticated methods described above, since mation method provided a more accurate cortical image of the
they are based on simpler assumptions. Nonetheless, they yield sources for a comprehensive review of these methodologies, see
images that are less attractive than the latter methods. In any Ref. 94. Another alternative method consists in reconstructing the
case, dipolar methods are only meaningful if the scalp field has potential distribution on the inner boundary of the skull by the
approximately focal character. method of spatial deconvolution (89).
Alternatively, the scalp EEG may be described using the surface
Laplacian that estimates the local normal component of the cur-
rent through the skull (33) and can be computed by applying SPATIAL DISTRIBUTIONS OF SOURCES
either a local or a global approach. According to the local OF NEUROPHYSIOLOGICAL SIGNALS:
approach (88–90), the surface Laplacian is computed locally using THE CONTRIBUTION OF HEMODYNAMIC
a subset of neighboring sensors for differentiation. According to INFORMATION
the global approach (33,91,92), an interpolation function is first
applied to the measurements obtained over the whole head and Currently a number of groups are attempting to solve the
the differentiation is applied afterward. The relationship between problem of the inherent ambiguity of the EEG/MEG inverse
the surface Laplacian and the cortical potential distribution is only solutions by combining functional MRI (fMRI) data with
simple in case of homogeneous skull and scalp layers. Zanow (93) EEG/MEG data (76), but these approaches are still in
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 99

Figure 5.6 The EEG (top row), the brain


potential reconstructed by means of linear
estimation with minimum norm constraint
(center row), and the surface Laplacian
maps (bottom row) of the deviant tones
responses to a mismatch negativity exper-
iment are shown at two latencies: at 140
msec (left column) and 180 msec (right
column) after stimulus onset. Results were
computed with Advanced Source Analysis
(ASA) software package. [Adapted from
Zanow F. Laplacian maps (bottom row) of
the deviant tones responses to a mismatch
neg-1997. Realistically shaped models of
the head and their application to EEG and
MEG, p. 144. Ph.D. thesis, University of
Twente, Enschedé.]

development. A problem is that the time course of the fMRI mainly of synchronized synaptic potentials, either excitatory or
signal, that is, the blood-oxygen-level dependent (BOLD) sig- inhibitory, with a contribution from membrane oscillations
nal differs from the corresponding local neurophysiological and spike afterpotentials.
signals, since it is delayed with respect to the latter. Devor et al. (97) reported the results of a systematic study in
Nonetheless, Logothetis and Wandell (95) examined the rela- rat somatosensory cortex, to characterize the relationship
tion between LFPs (or local EEG), single- and multiunit activ- between changes in blood-oxygen content and the neural spik-
ity (MUA), and high spatiotemporal fMRI responses recorded ing and synaptic activity, showing that there is a nonlinear rela-
simultaneously in monkey visual cortex, and were able to tionship between electrophysiological measures of neuronal
demonstrate that while both LFP and MUA activity are corre- activity and the hemodynamic response.
lated with BOLD for short stimulation paradigms, this is not The methodologies developed to interpret the results of
the case for sustained oscillations, in the latter case only the combining EEG recordings and fMRI, particularly in epileptic
LFP signal is significantly correlated with the hemodynamic disorders, have been critically reviewed (98). More recently, a
response. The LFPs are commonly thought to be related to the study by Shmuel and Leopold (99) combining fMRI-BOLD and
input of a neural ensemble and dendritic processing within the intracortical neurophysiological signals confirmed the existence
network in contrast with the action potentials or “units” that of correlations between neuronal signals consisting of either the
consist of the output of neurons (96). Thus, LFPs consist firing rate of a small group of neurons, or of relative power
100 Part I ■ Basic Principles

changes in the MUA band, but particularly of LFPs in the In the section on the influence of inhomogeneities we have
gamma frequency band, with BOLD signals at a time lag (peaks seen that, to explain the EEG, we must assume the existence of
at about 6 sec lag) relative to neuronal activity. shells with different conductivities. The existence of these
shells affects the electric potential in two ways: it causes an
attenuation of the potential and it introduces ED layers at the
THE MAGNETOENCEPHALOGRAM (MEG) boundaries of the inhomogeneities as shown in expression
5.19. However, the brain and surrounding tissues behave as a
As discussed above, the ionic currents originating from bio- medium with constant magnetic permeability. Therefore, the
chemical sources at the cellular level in the central nervous sys- magnetic field, in contrast to the electric field, is not influ-
tem generate not only electric fields but also magnetic fields. enced by those layers; it is nevertheless also affected by the
Until Cohen’s work (100), magnetic fields could not be induced dipole layers existing at the boundaries of the inho-
detected owing to the extremely low strength of the MEG and mogeneities in conductivity, as shown in the Appendix
the absence of sufficiently sensitive transducers. The magnetic (expression 5.30).
field of the brain has a strength in the order of magnitude of Other properties of the MEG that should be mentioned are
10 8 gauss (G) or, in meter-kilogram-second (m kg s) units, of the following. Neither electrodes nor a reference point are nec-
10 12 Weber m 2 or tesla (W m 2 or T) (101), whereas the essary for recording the MEG; the transducers for the MEG
earth magnetic field has a strength in the order of magnitude of need not touch the scalp, because the magnetic field does not
0.5 G (102). However, with the introduction of the supercon- disappear where conductivity is zero (free space). From expres-
ducting quantum interference device (SQUID) magnetometer, sion 5.29 it can be seen that the vector potential has the same
based on a superconducting effect at liquid helium tempera- direction as the intracerebral current density. Because the mag-
ture, sensitivities in the order of magnitude of 10 10 G have netic field is perpendicular to the vector potential, we can con-
been obtained (101). clude that current dipoles directed perpendicularly to the
The earth magnetic field and the urban magnetic noise fields surface of the skull result in magnetic fields tangential to the
(10 4 to 10 3 G) can be practically removed while recording surface of the head and, vice versa, tangential current dipoles
the MEG by using the gradiometer technique. This is based on result in magnetic fields perpendicular to the skull.
the fact that the disturbing fields are almost constant over large Research on MEG has advanced considerably in recent years
distances, whereas the MEG falls off rapidly with distance. This (see Chapter 42). Both practical and theoretical aspects of magne-
is done by subtracting the induced potentials in two coils placed toencephalography have been elucidated, as summarized by
close to each other; in this way, the earth and urban noise mag- Cohen and Cuffin (104), Okada (105), Cuffin (106), Modena et al.
netic fields are suppressed. The magnetic field in stationary (107), Hari and Ilmoniemi (108), Hämäläinen (109), Lewine and
conditions obeys Ampere’s laws and can be derived from the Orrison (110), and de Jongh et al. (111). Some theoretical aspects
microscopic sources at the cellular level, as shown in the of general interest will be mentioned here. Although EEG and
Appendix (from expression 5.22 onward). Although the electri- MEG reflect in essence the same elementary phenomena, in prac-
cal potential reflects the strength and localization of the current tice the two types of measurement differ in a number of aspects.
sources, the magnetic field reflects not only these properties but First, the EEG is a relative measurement; it always needs a
also the direction of the current densities. This can be seen in reference electrode. The magnetic field measurement does not
the Appendix by comparing expression 5.29 for the magnetic need a reference point.
quantities with expression 5.8 for the electric potential. Second, the MEG is a measure of the magnetic fields perpen-
Whether the information about the direction of current densi- dicular to the skull, which are caused by tangential current
ties in the brain may be derived from the MEG depends on the dipoles. The radial component of the current dipole does not
practical possibility of recording the magnetic fields in three generate a magnetic field outside a sphere-shaped volume con-
directions. In general we may state that the EEG reflects mainly ductor, and thus does not contribute to the MEG. By contrast, the
the volume or extracellular currents, whereas the MEG is more EEG is a measure of both components. This means that the MEG
sensitive to the primary intracellular currents. There are very reveals tangential current dipoles in a clear way, which in the EEG
few experimental studies where magnetic and electric fields may be obscured by radial sources. The main intracortical dipo-
have been measured along with elementary electrophysiological lar current sources are perpendicular to the cortical surface
properties of simple brain preparations. The recent study of (Fig. 5.1). Therefore, Cohen and Cuffin (104) have pointed out
Murakami et al. (103) validates the assumption indicated that the MEG mainly measures the cortical current dipoles lying
above. This study examined whether evoked magnetic fields in the sulci and not on the convexity of the gyri. Thus, the former
and intra- and extracellular potentials from longitudinal hip- causes mainly tangential dipoles to the skull surface, whereas the
pocampal CA3 slices of guinea pig can be interpreted within a latter produces mainly radial dipoles (Fig. 5.4).
single theoretical framework that incorporates ligand- and volt- Third, the map representing the distribution of the mag-
age-sensitive conductances in the dendrites and soma of the netic field at the surface of the head caused by a tangential
pyramidal cells. The intracellular potentials in this validated current dipole is rotated by 90 degrees to the corresponding
model reveal that the spikes and slow waves of the magnetic EEG potential map as shown in Figure 5.7. This means that to
fields are generated in or near the soma and apical dendrites, localize a dipole-like source in the x and y directions, both
respectively. MEG and EEG should be used (104), but it cannot be stated
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 101

Figure 5.7 Examples of measured and computed maps corresponding to two time samples of a visual evoked potential
(VEP) and magnetic field (VEF). The stimulus was the appearance of a checkerboard pattern as described by Stok (64) and
Stok et al. (68). Top row: Measured VEF and VEP contour plots for two instants of time during the on-response: 100 and
130 msec. Middle row: Simulated VEF and VEP contour plots that correspond to the EDs that were estimated from the
measured VEP distributions. Bottom row: The simulated contour plots that correspond to the VEF-based EDs. The dashed
rectangles indicate the simulation area of 12 14 cm 2. Measures are in centimeters and with respect to the inion. In the
contour plots of measured data, negativity is indicated by dashed lines and in the contour plots of simulated data by thin
lines. The thick lines indicate the zero level. Note that these plots present theoretical and experimental examples of the
three main differences that can be expected between EEG and MEG contour plots. (Modified from Stok CJ, Spekreijse HJ,
Peters MJ, et al. A comparative EEG/ MEG equivalent dipole study of the pattern onset visual response. New trends and
advanced techniques in clinical neurophysiology. EEG Suppl. 1990;41:34–50.)

that one type of measurement is better than the other. significantly in respect to the case of the homogeneous volume
Differences in localization of sources with both methods may conductor under the influence of inhomogeneities in
be determined mainly by the direction along which the meas- conductivity. In certain cases of spherical symmetry, however,
urements were made. the magnetic field distribution due to a current dipole is
Fourth, the EEG represents a sum of the potentials caused by independent of spherically symmetrical variations in resistivity
primary and secondary sources or volume currents. In the of the volume conductor (114). Whether this will hold true for
Appendix, it is shown that the media of the head (brain, CSF, a real head is questionable.
skull, and scalp) have different conductivities; thus, secondary Nevertheless, Cohen and Cuffin (104) assume that the MEG
sources of electric (expression 5.19) and magnetic fields would not be influenced by the volume currents and that this
(expression 5.30) are introduced. It is often stated, however, would explain why the MEG pattern is more focal, or tighter,
that the MEG is not affected by these volume currents than the corresponding EEG potential distribution. It appears
(104,105). It is still debatable how much these secondary that still tighter MEG patterns may be obtained by using trans-
sources contribute to the EEG and MEG under the conditions ducers composed of a number of magnetic field–sensing coils
in which these signals are experimentally measured. Plonsey in an array.
(112) pointed out that both the electric and magnetic fields are Fifth, there are differences between MEG and EEG measure-
affected by secondary sources. Van Rotterdam (113), using a ments regarding the representation of some types of nondipo-
model of a volume conductor represented by a lattice including lar sources, as discussed in detail by Cuffin (106). Different
inhomogeneities, showed that the magnetic field changes types of sources give different results. For side-by-side line
102 Part I ■ Basic Principles

sources forming a dipole layer, the EEG gives a more accurate However, the question of whether the MEG offers real
representation of the actual sources than does the MEG; the practical advantages over the EEG has been raised (64). Stok
reverse is true for in-line sources, that is, a series of dipoles et al. (68) made a systematic theoretical and experimental
aligned along the same direction. However, it should be added comparison of EEG and MEG and concluded that the MEG is
that both EEG and MEG solutions of the inverse problems for not, in general, superior over the EEG, although the MEG
the side-by-side and in-line sources lie deeper in the head and localizes the equivalent sources slightly better than the EEG,
have a larger amplitude than the actual sources. In particular, but it estimates the dipole components slightly worse. Two
the sources that are in the form of lines and are more than 2 cm general conclusions of these studies should be put in evidence:
long give different results when measured using EEG or MEG; (i) when model assumptions are not violated, the MEG and
the MEG solutions are deeper for the side-by-side sources, and EEG lead essentially to the same inferences about the source
EEG solutions are deeper for the in-line sources. These differ- (excluding magnetically silent sources); and (ii) when model
ences may be helpful in identifying the type of such sources in assumptions are violated, the MEG and EEG respond, in gen-
the brain (106). eral, in different ways such that a comparison of the results
Sixth, MEG and EEG appear to differ in their capability of obtained with both methods may give clues regarding the
localizing a current dipole source, given a set of observations kind of model violation present. Therefore, it is important to
corrupted by noise. A single current dipole can be represented use both methods to obtain a higher reliability in functional
by six parameters; three determine its position inside the head localization studies (117).
and three define its components. It has been shown by Stok et This question has been addressed directly by comparing
al. (115) that the EEG gives better estimates of the three com- MEG and EEG maps obtained in patients where dipoles were
ponents of the current dipole than the MEG, based on obser- implanted, using indwelling intracranial electrodes that were
vations to which noise was added. The latter, however, gives placed for the diagnosis of an epileptogenic focus (55,118).
better estimates of the three position parameters. Moreover, These authors concluded, from a limited number of experi-
the inverse procedure, for cases with bad signal-to-noise ratios, mental measurements, that the average errors of localization
fails more often with EEG than with MEG data. The EDs esti- were 10 mm for the EEG and 8 mm for the MEG, and thus that
mated by the inverse procedure using EEG and MEG data sep- the MEG was not superior to the EEG. Nevertheless, the main
arately may be combined in order to obtain an average advantage of the MEG may be the fact that the MEG is not
estimate. Alternatively, the ED may be calculated directly by affected by radial sources and, in this way, the use of both MEG
determining the dipole that provides the least-squares fit to and EEG may complement each other and help to establish a
EEG and MEG simultaneously. It can be demonstrated that the better model of the source, when it cannot be described as
latter procedure provides slightly better results than the com- being a simple dipole.
bination of the two estimates into an average estimate. This In conclusion, more research is needed where the two meth-
reinforces the statement that the two types of measurement, ods are directly compared using the same data. In the tutorial
MEG and EEG, indeed provide complementary information book on “MEG Methods” edited by Hansen et al (119), some
and should be combined whenever precise source localization frequently asked questions about these issues are discussed in
is desired. more detail (120).
Seventh, spherical models of the head perform much bet-
ter with MEG than with EEG data, because the former is less APPENDIX: THE MATHEMATICAL BASIS
sensitive to the influence of volume currents, while these cur-
rents are affected appreciably by deviations from the ideal AB Van Rotterdam
head. This means that inverse solutions based on MEG data The ionic currents in the brain cause electrical and magnetic
may be simpler to obtain since spherical models may be more fields obeying Maxwell’s and Ohm’s laws. These fields have a
readily used in this case than with EEG measurements (116). direction and magnitude; they must, therefore, be represented
Nevertheless, volume currents may still affect the MEG as as vector functions. The electric field is given by E (r ,t) and the
shown by Van Uitert et al. (54). Certainly one of the benefits magnetic field by H (r ,t) , because these vectors depend on
that has been obtained by the introduction of the MEG tech- location r and on time t. (Vector quantities are indicated by an
nique is that a considerable amount of research on brain arrow above the symbol.)
activity by competent multidisciplinary groups has led to a In brain and surrounding tissues, the material constants
renewed interest in the biophysical study of the electric and are approximately the following: conductivity, ≈ 10–1 –1 m –1;
magnetic activity recorded from the scalp. This led to the dielectric constant, ≈ 10–9 F m –1; and magnetic permeability,
development of more sophisticated models of the neuronal ≈ 10–6 H m –1. Do potential changes taking place in the brain
sources and of the volume conductor. These developments at a frequency of, for example, 1 k Hz cause propagated elec-
have been of interest for obtaining a better understanding not tromagnetic waves in the medium that may be detected after
only of the MEG but also of the EEG. Initially the experimen- a certain time at a distant point such as the scalp? It can be
tal results obtained by the new technique have led to the proved (121) that, in a medium such as the brain with the
thought that the MEG is superior to the EEG in locating brain material constants indicated above, the propagation velocity
sources (108). of the electromagnetic waves is of the order of 105 m s–1,
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 103

change of current in the volume of the cube. When no net cur-


rent flows into the cube (i.e., the current that goes into the vol-
ume comes out again), this total change must be zero, as
indicated in expression 5.1. Thus, we may state that the current
is stationary. In the case of a stationary current, the electrical
field E(r ) , or simply E, is related to the electrical potential V(r ) ,
or simply V, by the following expression:

E gradV (5.3)

In this expression the operator gradient (grad) indicates dif-


ferentiation of the scalar function V, of space and time (time is
considered here to be fixed) in the following way:

V V V T
grad V a , , b (5.4)
x y z
where gradV is a vector quantity called the gradient of V.
Figure 5.8 Cubic volume with sides dx, dy, and dz. Current density In a conductive medium, Ohm’s law is valid; that is:
flowing in: Jz (x — dx, y — dy, z — dz); Jy (,y — dy) and Jz (,,z — dz)
or J(r dr ) . Current densities flowing out: Jx (x,y,z); Jy (,y,) and Jz J Ji sE (5.5)
(,,z) or J(r ) . When divJ 0, no charge is accumulated in volume
where J is the current density injected in the medium (e.g., a
dxdydz. Inset: Decomposition of current density vector J in components transmembrane current density caused by synaptic activity in a
in x, y, and z direction as Jz , Jy, and Jx. neuron).
Taking the divergence of both sides of expression 5.5 and
using expression 5.1, one may write:
which implies that one need not be concerned with the prop-
agation of electromagnetic waves caused by potential changes divJi
within the brain. This means that, in practical terms, the divE (5.6)
s
effects of those potential changes may be detected simultane-
ously at any point in the brain or surrounding tissues. This
Substituting E by – gradV according to expression 5.3, one
important statement leads to the conclusion that the currents
can write an expression that is called the Poisson equation for
caused by sources in the brain at any moment in time behave
the potential field owing to an injected current:
in a stationary way. This means that no charge is accumulated
at any time in the brain (122). 2 2 2
V V V divJi
Therefore, it can be stated that for the current density J(r ) divgrad V (5.7)
or, for simplicity, J, at any moment of time: x2 y2 z2 s

div J O (5.1) In case the medium where r o is situated is assumed to be


where the operation div J is called divergence of the vector J; infinite, isotropic, and homogeneous, it can be proved that the
J is the current density in A m –2. The operator div must be solution of the Poisson equation is the following:
understood as indicating differentiation of a vector; expression
5.1 can also be written in another way that may help in under- 1 divJi 3
V(r o ) dr (5.8)
standing its meaning. Let us consider the expression for J for 4 s R
vol
the case of an infinitesimally small cube with sides dx, dy, and
dz (Fig. 5.8). It holds that: which gives the values of the potential at a point r o in the
volume conductor from injected current densities Ji, at points
Jx Jy Jz
divJ dxdydz a b dx a b dy a dxdyb dz r , lying at a distance R from r o ( R 0r r o 0); the integral
x dydz y dydz z represents the summation over all current sources within the
(5.2) volume; note that divergence of the vector J gives a scalar
quantity.
In this expression the factors between brackets, such as Summarizing, it has been shown that the space-varying and
( Jx/ x dydz), represent the rate of change of current through the time-varying parts of the electrical variables can be sepa-
the surfaces dydz, dxdz, and dxdy. To account for the total rated; the time-varying part is the same everywhere in the
change of current, it is necessary that those factors are multi- medium owing to the large propagation velocity, and the space-
plied by the sides of the cube dx, dy, and dz, then these products varying part can be computed for every instant in time accord-
must be summed. Thus expression 5.2 represents the total ing to expression 5.8.
104 Part I ■ Basic Principles

This general expression is not the only way to define V(r o ) . To understand the effect of the solid angle in a more practi-
Two other expressions (5.10 and 5.12) are given below. These cal way, it is useful to derive an explicit expression for d :
are more convenient because they relate the extracellular
potential field directly to membrane processes of the individ- 1 # R cos
ual neurons in the brain. One of these expressions can be d (r r o) grada b dO (r ) #
3 dO (r ) dO
R R R2
derived assuming that the neuronal membrane can be consid-
(5.11)
ered as equivalent to a double layer with an inner (intracellu-
lar) membrane potential Vm and an outer (extracellular) where is the angle between R and dO; it should be noted that
potential V. In these terms, V( r o ) is given by the following R is the vector connecting the point r o where V is measured and
expressions: the point r where the dipole is located (see Fig. 5.8).
It should be noted in expression 5.9 that a change in mem-
1 1 # brane potential such as a depolarization in a small piece of
V(r o ) (s iVm (r ) s e(r ))grada b dO (r ) (5.9)
4 s R membrane potential results in an intracellular potential Vm and
s
an extracellular potential V. In this case, the membrane behaves
where i is the intracellular and e is the extracellular conduc- like a current dipole consisting of a sink at the outside and a
tivity, and dO is pointing to the extracellular medium; the inner source at the inside. Thus, there is a jump in potential across the
product grad (1>R) #dO is called the solid angle d (r r o )
membrane. A current dipole, or simply a dipole, is defined by its
strength and direction; it is represented by the vector p (r ) . In the
subtended by the surface element dO (r ) on the membrane sur- example described above, the dipole is oriented perpendicularly
face S and seen from the extracellular point r o. It must be to the membrane surface dO; thus, the dipole is represented as
emphasized that expression 5.8 takes the form of expression 5.9 p(r )dO. According to the above, another expression for the
only when the injected current densities Ji originate at the cell potential V(r o ) caused by one dipole can be derived (123):
membrane as derived by Plonsey (121). Expression 5.9 can be
simplified to the following approximate expression, because p(r ) 1 #
V(r o ) grad a b dO (r )
0s eV(r ) 0V 0s iVm (r ) 0: 4 s R

or
si
V(r o ) Vm (r )d (r ro) (5.10)
4 se p(r ) cos
s
V(r o ) dO (5.12)
4 s R2
The solid angle d subtended by a membrane surface dO
seen from a point r o can be interpreted in a geometrical way, as From expression 5.12 it can be concluded that in the case of
shown in Figure 5.9. a single dipole at the membrane, V decreases with the square of
the distant R to the source and it is also proportional to cos ;
thus, it is maximal when R is perpendicular to the surface dO.
In many cases, the membrane depolarization (or hyperpo-
larization) does not remain limited to a small piece of mem-
brane; rather, it spreads over a more or less extended membrane
surface. In this way the equivalent current source cannot be
accounted for in terms of single dipole. One must, instead,
assume that there exists a dipole layer at the membrane.
For a dipole layer located at the surface of the membrane,
one must sum the effects of all individual dipoles; therefore,
expression 5.12 becomes:

1 1 #
V(r o ) p(r )grad a b dO (r )
4 s s R

or

1
V(r o ) p(r )d (r ro) (5.13)
4 s s

Figure 5.9 Solid angle subtended by infinitesimal surface dO with


From expression 5.13, it can be noted that, if p is constant
direction dO on membrane cylinder S seen from the extracellular point over the closed membrane surface S, then V(r o ) 0, because
r o lying at a distance R 0ro r 0 from dO; the angle between the the integral of the solid angle d over an external closed surface
direction of the surface dO and r o r is . is zero. Thus, to obtain a value of V(r o ) different from zero,
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 105

Figure 5.10 A: Dipole layer of length 1 and height h in the y-z plane seen from extracellular point r o on the x axis.
B: Attenuation (relative potential, rel. pot.) of extracellular potential as a function of distance (r o) in a log–log plot for the
case of a single dipole and for the case of a dipole layer. Note that, in the latter case, two slopes can be distinguished, the
decay of the potential with distance being more accentuated in the far field (ro 1). The arbitrary values of h = 0.0625
and I = 1 used in this model are indicated on the horizontal axis.

p must vary over the membrane surface; in other words, the where the time behavior of the field is the same as that of the
membrane potential must vary over the surface owing, for sources.
example, to local synaptic activity. In general terms, to allow for time-varying sources, expres-
In this case, the attenuation of the potential in the volume sion 5.10 must be changed as follows:
conductor depends not only on distance R and angle , but also
on the geometry of the dipole layer. For instance, for a dipole 1
layer of constant strength with length 1 and height h located V(r o, t) Vm (r , t)d (r ro ) (5.16)
4 se s
at x = 0 in the y-z plane (Fig. 5.10A) the potential field at a point
r o along the x axis will be given by the following expression: This effect can be illustrated by the following example (Fig.
5.11A). Let us assume a cell that can be simplified to an equiva-
lent cylinder (124) oriented along the z axis where a membrane
1>2 ¢ h>2 ro depolarization front propagates along the cylinder with an arbi-
V(r o ) p 2 2 2 3>2
dyzy (5.14)
y 1>2 z ¢ h>2 (r 0 y z) trary velocity c. In this case, we may write that the resulting
potential V(r o ) is proportional to a time-varying solid angle.
For ro W h and h V 1, this can be estimated as follows: From expressions 5.11 and 5.16, the potential V(r o,t) caused
by a depolarization front along the cylinder is given by:
1
V(r o ) p¢ h (5.15)
ro5 1 2 1>2
(r o>1) 6 cos ( (t) )
V(r o, t) 2 dO (5.17)
0ctn z r o 0
It can be seen that in the far field, r o W 1, the potential tends
to decay as in the case of the single dipole with r 2o where for where n z is the unit vector in the z direction. Note that the
short distances h ro 1, the potential decays proportion- denominator in expression 5.17 represents the square of the
ally to distance ro. In Figure 5.10B, it can be seen that in the case distance R(t) between r o and the time-varying location of the
of the dipole layer the attenuation has two slopes depending on front.
the relation between r o and 1, whereas in the case of a simple At a point further away in the same direction, r 1 a r o, V
dipole there is only one slope. becomes:
Until now, the discussion has considered only the behavior
of standing sources of EEG signals, that is, sources that do not 1
change in space. However, there is experimental evidence that V(r 1, t) 2 V(r o, t>a ) (5.18)
a
leads one to assume that some macroscopic sources in the
brain do propagate in space (14,20,92). This propagation of Therefore, the potential at a far location r 1 is equal to the
sources has a particular effect on the behavior of the potential potential at location r o attenuated in amplitude and in a more
field in time. This effect does not appear for standing sources dilated time scale (Fig. 5.11B).
106 Part I ■ Basic Principles

a (s e s k )V(r )d (5.19)
k 1 sk

s e V(r )d
s

in which S1 represents the membrane surface that encloses the


(i)
first neuron with membrane potential Vm (r ) and intracellular
conductivity i; the extracellular conductivity is e except in the
regions bounded by the surfaces Sk, within these regions the
conductivity is k; s (r o ) is the conductivity for the region
where point r o lies: s (r o ) s k if r o is bounded by Sk and it is
e elsewhere, d are the solid angles of surfaces pointing to the
region with conductivity e.
The second term on the right describes the influence that
neurons have as inhomogeneities on the extracellular field; the
third term describes the distorting influences of the M inhomo-
geneities, whereas the fourth term expresses the influence of the
border of the medium.
Using expression 5.19, it is in principle possible to compute
the EEG measured on the scalp and generated by N neurons.
In the following model, the brain and surrounding tissues
are represented as a sphere with a number of shells within
which a single dipole is situated. The potential field in and on
the sphere is a solution of the Poisson equation obeying the
following boundary conditions: the current densities normal
to the surfaces of the shells are continuous and the tangential
components of the electric field at the boundaries are also
continuous.
Assuming radial symmetry, the solution for a radial dipole
can be written as a Legendre series, in polar coordinates as
Figure 5.11 A: Membrane cylinder oriented along the z axis in which follows (123):
a depolarization front propagates (with velocity c) along the z direc-
tion, r o and r 1 are two extracellular points at which extracellular q
potentials are measured r 1 a r o. Note that the potentials V at r o and V(r, f , ) (n 1)
bn r n 6P0n (cos ) (5.20)
a 5 an r
at r 1 are different, because and R differ when seen from r 1 instead n 0
of r o. The changes in and R as a function of time depend on
the propagation velocity c. B: Extracellular potentials V measured
where P0n ( cos ) is a Legendre polynomial of the nth order. For
simultaneously at r o and r 1 (plot with arbitrary scales) showing time
dilation. a tangential dipole, the solution is:

q
(n 1)
V(r, , ) a 5 cn r dnr n 6 P1n ( cos ) cos (5.21)
n 0

INFLUENCE OF INHOMOGENEITIES
Let us assume that the field potential V(r o ) generated by the where P1n (cos ) is an associated Legendre polynomial.
activity of N neurons situated in a finite homogeneous medium
The coefficients an, bn , cn , and dn for all n in both cases should
containing M structures with different conductivities and
bounded by surface S (outside the medium the conductivity is be evaluated on basis of the boundary conditions at the different
zero) can be expressed as follows (125): boundaries and the fact that the source is a dipole (23).

N
4 s (r 0 )V(r 0 ) sia (i)
Vm (r )d MAGNETIC FIELDS
i i si
N
In the stationary approximation of Maxwell’s equations for the
sea V(r )d electromagnetic fields generated by microscopic sources at
i i si the cellular level, the behavior of the magnetic field H or the
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 107

induction field B mH is governed by two experimental In this way, it can be seen that the magnetic vector potential
physical laws. There is Ampere’s law in the stationary A reflects the direction of the current densities Ji. If there are
approximation: inhomogeneities in conductivity, the expression becomes (126):

m Ji (r ) 3 V(r )
curlH J (5.22) A( r 0 ) c dr a (s e s k) dO (r )d (5.30)
4p vol R k Sk R
and
where Sk are surfaces enclosing regions with conductivity k;
divB 0 (5.23) elsewhere is the conductivity e. The surfaces dO point to the
region with conductivity e; is assumed to be constant.
which expresses the experimental fact that magnetic monopoles
do not exist. The term curl H is a differential operation called REFERENCES
curl or rotation on a vector function; it is defined as follows:
1. Helmholtz H. Ueber einige Gesetze der Verteilung elektrischer
0H y 0H z 0H z 0H x 0H x 0H y T Ströme in Körperlichen Leitern mit Anwendung auf die
curlH ca b #a b #a bd thierischelektrischen Versuche. Prog Ann Physik Chemie.
0z 0y 0x 0x 0y 0H y 1855;89:211–233, 353–377.
(5.24) 2. Lorente de No R. Action potential of the motoneurons of the
hypoglossus nucleus. J Cell Comp Physiol. 1947;29:207–287.
For conservative or rotation-free fields, that is, fields deriv- 3. Hubbard JI, Llinas R, Quastel DMJ. Electrophysiological Analysis of
able from vector potentials (e.g., E gradV), this differenti- Synaptic Transmission. London: Edward Arnold Ltd; 1969.
ation results in a zero vector. However, for vector fields directed 4. Klee M, Rall W. Computed potentials of cortically arranged popu-
lations of neurons. J Neurophysiol. 1977;40:647–666.
along a closed path (e.g., closed current density loops), the 5. Humphrey DR. Reanalysis of the antidromic cortical response. II.
operation results in a vector field perpendicular to the original On the contribution of cell discharge and PSPs to the evoked
vector field. It can be proved that expression 5.23 is equivalent potentials. Electroencephalogr Clin Neurophysiol. 1968;25:421–442.
6. Pettersen KH, Einevoll GT. Amplitude variability and extracellular
to the statement that B can be derived from a vector potential A
low-pass filtering of neuronal spikes. Biophys J. 2008;94(3):
as follows: 784–802.
7. Swinney KR, Wikswo JP Jr. A calculation of the magnetic field of a
B curlA (5.25) nerve action potential. Biophys J. 1980;32(2):719–731.
8. Murakami S, Okada YC. Contributions of principal neocortical
By substituting expression 5.25 in 5.22 and using the defini- neurons to magnetoencephalography and electroencephalography
tion of J as a function of impressed current densities (expres- signals. J Physiol. 2006;575(3):925–936.
sion 5.5), we obtain: 9. Mainen ZF, Sejnowski TJ. Influence of dendritic structure on fir-
ing pattern in model neocortical neurons. Nature.
1 1996;382:363–366.
curlcurlA Ji sE (5.26) 10. Hämäläinen MS, Hari R, Ilmoniemi R, et al. Magneto -
m encephalography: theory, instrumentation and applications to the
noninvasive study of human brain function. Rev Med Physics.
It can be shown that curlcurlA divgrad A grad divA, 1993;65:413–497.
so that 5.26 becomes 11. Kyuhou S, Okada YC. Detection of magnetic signals from isolated
transverse CA1 hippocampal slice of the guinea pig.
divgrad A mJi grad divA ms E (5.27) J Neurophysiol. 1993;70:2665–2668.
12. Okada YC, Wu J, Kyuhou S. Genesis of MEG signals in a mam-
One can simplify expression 5.27 by making use of the ambi- malian CNS structure. Electroencephalogr Clin Neurophysiol.
1997;103(4):474–485.
guity in the definition of A (123) and choose A such that 13. Mountcastle VB. The columnar organization of the neocortex.
divA ms V the two last terms of expression 5.27 then can- Brain. 1997;120(pt 4):701–722.
cel out; expression 5.27 becomes the Poisson equation that for 14. Freeman WJ. Mass Action in the Nervous System. New York, NY:
Academic Press; 1975.
each separate component of A can be expressed as follows:
15. Katchalsky AK, Rowland V, Blumenthal R. Dynamic patterns of
brain cell assemblies. Neurosci Res Prog Bull. 1974;12(1):90–94.
divgrad A mJi (5.28) 16. Lopes da Silva FH, Hoeks A, Smits A, et al. Model of brain rhyth-
mic activity. Kybernetik. 1974;15:27–37.
17. Lopes da Silva FH, Van Rotterdam A, Barts P, et al. Models of neu-
In an infinite medium with constant and , the solution of ronal populations. The basic mechanisms of rhythmicity. Prog
equation 5.28 can be proved to be (121): Brain Res. 1976;45:281–308.
18. Wilson HR, Cowan JD. A mathematical theory of the functional
m Ji (r ) 3 dynamics of cortical and thalamic nervous tissue. Kybernetik.
A(r 0 ) dr (5.29)
4 vol R 1973;13:55–80.
108 Part I ■ Basic Principles

19. Schmitt FO, Dev P, Smith BH. Electronic processing of informa- 40. Bertrand O, Thevenet M, Perrin F, et al. Effects of skull holes on.
tion by brain cells. Science. 1976;193:114–120. In: Proceedings of the 14th International Conference of the IEEE
20. Lopes da Silva FH, Storm van Leeuwen W. The cortical alpha Engineering in Medicine and Biology Society, Satellite Symposium
rhythm in dog: the depth and surface profile of phase. In: Brazier on Neuroscience and Technology. Lyon, France; November, 1992.
MA, Petsche H, eds. Architectonics of Cerebral Cortex. New York, pp. 42–45.
NY: Raven Press; 1978. 41. Van den Broek SP. Volume conduction effects in EEG and MEG.
21. Van Rotterdam A, Lopes da Silva FH, van den Ende J, et al. A Ph.D. thesis, University of Twente, Enschedé, 1997.
model of the spatial-temporal characteristics of the alpha rhythm. 42. Yan Y, Nunez PL, Hart RT. Finite element model of the human
Bull Math Biol. 1982;44:283–305. head: scalp potentials due to dipole sources. Med Biol Eng Comput.
22. Lopes da Silva FH, van Hulten K. Analyse quantitative de l’activite 1991;29:475–481.
intercritique en E.E.G. et S.E.E.G. dans l’epilepsie. Rev EEG 43. Hämäläinen MS, Sarvas J. Realistic conductivity geometry model
Neurophysiol. 1978;8:198–204. of the human head for interpretation of neuromagnetic data. IEEE
23. Hosek RS, Sances A, Jodat RW, et al. The contributions of intrac- Trans Biomed Eng. 1989;36(2):165–171.
erebral currents to the EEG and evoked potentials. IEEE Trans 44. He B, Musha T, Okamoto Y, et al. IEEE Trans Biomed Eng.
Biomed Eng. 1978;25:405–415. 1987;34:406–414.
24. Kavanagh RN, Darcey TM, Lehmann D, et al. Evaluation of meth- 45. Meijs JWH, Voorde BJ. ten, Peters MJ, et al. The influence of vari-
ods for three-dimensional localization of electrical sources in the ous head models on EEGs and MEGs. In: Pfurtscheller G, Lopes da
human brain. IEEE Trans Biomed Eng. 1978;25:421–429. Silva FH, eds. Functional Brain Imaging. Toronto: Hans Huber;
25. Schneider MR. A multistage process for computing virtual dipolar 1988:31–45.
sources of EEG discharges from surface information. IEEE Trans 46. Meijs JWH, Peters MJ, Boom HBK, et al. Relative influence of
Biomed Eng. 1972;19:1–12. model assumptions and measurement procedures in the analysis
26. Schneider MR. Effect of inhomogeneities on surface signals com- of the MEG. Med Biol Eng Comput. 1988;26:136–142.
ing from a cerebral current dipole source. IEEE Trans Biomed Eng. 47. Yvert B, Bertrand O, Echallier JF, et al. Improved forward EEG cal-
1974;21:52–54. culations using local mesh refinement of realistic head geometries.
27. Witwer JG, Tresek GJ, Jewett DL. The effect of media inhomo- Electroencephalogr Clin Neurophysiol. 1995;95:381–392.
geneities upon intercranial electric fields. IEEE Trans Biomed Eng. 48. Wieringa HJ, Peters MJ. Processing MRI data for electromagnetic
1972;19:352–362. source imaging. Med Biol Eng Comput. 1993;31:600–606.
28. Ary JP, Klein SA, Fender DH. Locations of sources of evoked scalp 49. Meijs JWH, Weier OW, Peters MJ, et al. On the numerical accuracy
potentials; corrections for skull and scalp thicknesses. IEEE Trans of the boundary element method. IEEE Trans Biomed Eng.
Biomed Eng. 1981;28:447–452. 1989;36:1038–1049.
29. Geddes LA, Baker LE. The specific resistance of biological; materi- 50. De Munck JC, Peters MJ. Mathematical aspects of biomagnetic
als—a compendium of data for the biomedical engineer and phys- modeling. In: Proceedings of the 8th International Conference on
iologist. Med Biol Eng. 1967;5:271–293. Biomagnetism. In: Hoke M, ed. Munster: Excerpta Medica; 1991:
30. Ferree TC, Eriksen KJ, Tucker DM. Regional head tissue conduc- 11–12.
tivity estimation for improved EEG analysis. IEEE Trans Biomed 51. Ferguson AS, Zhang X, Stroink G. A complete linear discretization
Eng. 2000;47:1584–1592. for calculating the magnetic field using the boundary element
31. Gonçalves SI, de Munck JC, Heethaar RM, et al. The application of method. IEEE Trans Biomed Eng. 1994;42:455–459.
electrical impedance tomography to reduce systematic errors in 52. Kybic J, Clerc M, Abboud T, et al. A common formalism for the
the inverse EEG problem—a simulation study. Physiol Meas. integral formulations of the forward EEG problem. IEEE Trans
2000;21:379–393. Med Im. 2005;24:12–28.
32. Gonçalves SI, de Munck JC, Verbunt JPA, et al. In vivo measure- 53. Kybic J, Clerc M, Faugeras O, et al. Generalized head models for
ment of the brain and skull resistivities using an EIT-based MEG/EEG: boundary element method beyond nested volumes.
method and realistic models of the head. IEEE Trans Biomed Eng. Phys Med Biol. 2006;51:1333–1346.
2003;50:754–767. 54. Van Uitert R, Weinstein D, Johnson C. Volume currents in forward
33. Law SK, Nunez PL, Wijesinghe RS. High-resolution EEG using and inverse magnetoencephalographic simulations using realistic
spline generated surface Laplacians on spherical and ellipsoidal head models. Ann Biomed Eng. 2003;31(1):21–31.
surfaces. IEEE Trans Biomed Eng. 1993;40:145–153. 55. Cuffin BN. EEG localization accuracy improvements using realis-
34. Cuffin BN. Effects of local variations in skull and scalp EEG’s and tically shaped head models. IEEE Trans Biomed Eng.
MEG’s. IEEE Trans Biomed Eng. 1993;40:42–48. 1996;43:299–303.
35. Eshel Y, Witman S, Rosenfeld M, et al. Correlation between skull 56. Roth BJ, Balish M, Gorbach A, et al. How well does a threesphere
reflects the direction of the current densities. IEEE Trans Biomed model predict positions of dipoles in a realistically shaped head?
Eng. 1995;42:232–249. Electroencephalogr Clin Neurophysiol. 1993;87:175–184.
36. Haueisen J. Methods of numerical field calculation for neuromag- 57. Menninghaus E, Lütkenhöner B, Gonzalez SL. Localization of a
netic source localization. Ph.D. thesis, Technische Universität bipolar source in a skull phantom: realistic versus spherical model.
Ilmenau, Shaker Verlag, Aachen, 1996. IEEE Trans Biomed Eng. 1994;41:986–989.
37. Haueisen J, Ramon C, Czapski P, et al. On the influence of volume 58. Marquardt DW. An algorithm for least squares estimation of non-
currents and extended sources on neuromagnetic fields: a simula- linear parameters. J Soc Indust Appl Math. 1963;11:431–441.
tion study. Ann Biomed Eng. 1995;23:728–739. 59. Schneider MR, Gerin P. Une methode de localisation des dipoles
38. Nicholson PW. Specific impedance of cerebral white matter. Exp cerebraux. Electroencephalogr Clin Neurophysiol. 1970;28:68–78.
Neurol. 1965;13:386–401. 60. Smith DB, Lell ME, Sideman RD, et al. Nasopharyngeal phase
39. De Munck JC. A mathematical and physical interpretation of the reversal of cerebral evoked potentials and theoretical dipole impli-
electromagnetic field of the brain. Ph.D. thesis, University of cations. Electroencephalogr Clin Neurophysiol. 1973;34:654–658.
Amsterdam, the Netherlands, 1989.
Chapter 5 ■ Biophysical Aspects of EEG and Magnetoencephalogram Generation 109

61. Henderson CJ, Buler SR, Glass A. The localization of equivalent 80. Baillet S, Garnero L. A Baysenian approach to introducing
dipoles of EEG sources by the application of electric field theory. anatamo-functional priors in the EEG/MEG inverse problem.
Electroencephalogr Clin Neurophysiol. 1975;39:117–130. IEEE Trans Biomed Eng. 1997;44:374–385.
62. Scherg M. Fundamentals of dipole source potential analysis. In: 81. Hämäläinen MS, Ilmoniemi RJ. Interpreting measured magnetic
Grandori F, Hoke M, Romani GL, eds. Auditory Evoked Magnetic fields of the brain: estimates of current distributions. Technical
Fields and Potentials. Advances in Audiology. Basal: S Karger; 1990;6: Report TKKFA559. Helsinki: University of Technology; 1984.
40–69. 82. Hämäläinen MS, Ilmoniemi RJ. Interpreting magnetic fields of the
63. Scherg M. Functional imaging and localization of electromagnetic brain: minimum norm estimates. Med Biol Eng Comp. 1994;
activity. Brain Topogr. 1992;5:103–111. 32:35–42.
64. Stok CJ. The inverse problem in EEG and MEG with application to 83. Wang JZ, Williamson SJ, Kaufman L. Magnetic source images deter-
visual evoked responses. Ph.D. thesis, University of Twente, The mined by a lead-field analysis: the unique minimum-norm least-
Netherlands, 1986. squares estimation. IEEE Trans Biomed Eng. 1992;39(7):665–675.
65. Scherg M, Von Cramon D. Two bilateral sources of the late AEP as 84. Wang JZ. Minimum-norm least-squares estimation: magnetic
identified by a spatiotemporal dipole model. Electroencephalogr source for a spherical model head. IEEE Trans Biomed Eng.
Clin Neurophysiol. 1985;62:32–44. 1993;40(4):387–396.
66. Wood CC. Application of dipole localization methods to source 85. Grave de Peralta Menendez R, Hauk O, Gonzalez Andino S, et al.
identification in human evoked potentials. Ann N Y Acad Sci. Linear inverse solutions with optimal resolution kernels applied
1982;388:139–155. to electromagnetic tomography. Hum Brain Map. 1997;5:
67. De Munck JC. The estimation of time varying dipoles on the basis 454–467.
of evoked potentials. Electroencephalogr Clin Neurophysiol. 86. Pascual-Marqui RD, Michel CM, Lehmann D. Low resolution elec-
1990;77:156–160. tromagnetic tomography: a new method for localizing electrical
68. Stok CJ, Spekreijse HJ, Peters MJ, et al. A comparative EEG/MEG activity in the brain. Int J Psychophysiol. 1994;18:49–65.
equivalent dipole study of the pattern onset visual response. New 87. Pascual-Marqui RD, Esslen M, Kochi K, et al. Functional imaging
trends and advanced techniques in clinical neurophysiology. EEG with low-resolution brain electromagnetic tomography
Suppl. 1990;41:34–50. (LORETA): a review. Methods Find Exp Clin Pharmacol. 2002;
69. Lopes da Silva FH, Spekreijse H. Localization of brain sources of 24(suppl C):91–95.
visually evoked responses: using single and multiple dipoles. An 88. Hjorth B. An online transformation of EEG scalp potentials into
overview of different approaches. In: Brumia CHM, Mulder G, orthogonal source derivations. Electroencephalogr Clin
Verbaten MN, eds. Event-Related Brain Research. Amsterdam: Neurophysiol. 1975;39:526–530.
Elsevier; 1991; EEG Suppl. 42:38–46. 89. Le J, Gevins AS. Method to reduce blur distortion from EEGs using
70. Manshanden I, De Munck JC, Simon NR, et al. Source localization a realistic head model. IEEE Trans Biomed Eng. 1993;40:517–528.
of MEG sleep spindles and the relation to sources of alpha band 90. Le J, Menon V, Gevins AS. Local estimate of surface Laplacian der-
rhythms. Clin Neurophysiol. 2002;113(12):1937–1947. ivation on a realistically shaped scalp surface and its performance
71. Schwartz DP, Badier JM, Vignal JP, et al. Non-supervised spatio- on noisy data. Electroencephalogr Clin Neurophysiol. 1994;26:
temporal analysis of interictal magnetic spikes: comparison with 193–199.
intracerebral recordings. Clin Neurophysiol. 114(3):438–449. 91. Babiloni F, Babiloni C, Carducci F, et al. A high resolution EEG
72. Van’t Ent D, Manshanden I, Ossenblok P, et al. Spike cluster method based on the correction of the surface Laplacian estimate
analysis in neocortical localization related epilepsy yields clinically for the subject’s variable scalp thickness. Electroencephalogr Clin
significant equivalent source localization results in magnetoen- Neurophysiol. 1997;103:486–492.
cephalogram (MEG). Clin Neurophysiol. 2003;114(10): 92. Nunez PL. Electrical Fields of the Brain. New York, NY: Oxford
1948–1962. University Press; 1981.
73. Baillet S, Mosher JC, Leahy RM. Electromagnetic brain mapping. 93. Zanow F. Realistically shaped models of the head and their appli-
IEEE Signal Process Mag. 2001;18(6):14–30. cation to EEG and MEG. Ph.D. thesis, University of Twente,
74. Gross J, Kujala J, Hämäläinen M, et al. Dynamic imaging of coher- Enschedé, 1997.
ent sources: studying neural interactions in the human brain. Proc 94. Knösche Th. Solutions of neuroelectromagnetic inverse problems
Natl Acad Sci U S A. 2001;98:694–699. an evaluation study. Ph.D. thesis, University of Twente, Enschedé,
75. Robinson SE, Vrba J. Functional brain imaging by synthetic aper- 1977.
ture magnetometry (SAM). In: Yoshimoto T, Kotani M, Kuriki S, 95. Logothetis NK, Wandell BA. Interpreting the BOLD signal. Annu
et al., eds. Recent Advances in Biomagnetism. Sendai, Japan: Tohoku Rev Physiol. 2004;66:735–769.
University Press; 302–305. 96. Berens P, Keliris GA, Ecker AS, et al. Feature selectivity of the
76. Dale AM, Liu AK, Fischl BR, et al. Dynamic statistical parameter gamma-band of the local field potential in primate primary visual
mapping: combining fMRI and MEG for high resolution imaging cortex. Front Neurosci. 2008;2(2):199–207.
of cortical activity. Neuron. 2000;26:55–67. 97. Devor A, Dunn AK, Andermann ML, et al. Coupling of total
77. Mosher JC, Leahy RM. Source localization using recursively and hemoglobin concentration, oxygenation, and neural activity in rat
projected (RAP) MUSIC. IEEE Trans Signal Process. 1999;47: somatosensory cortex. Neuron. 2003;39(2):353–359.
332–340. 98. Gotman J, Kobayashi E, Bagshaw AP, et al. Combining EEG and
78. Mosher JC, Lewis PS, Leahy RM. Multiple dipole modeling and fMRI: a multimodal tool for epilepsy research. J Magn Reson
localization from spatiotemporal MEG data. IEEE Trans Biomed Imaging. 2006;23(6):906–920.
Eng. 1992;39:541–557. 99. Shmuel A, Leopold DA. Neuronal correlates of spontaneous fluc-
79. Mosher JC, Baillet S, Leahy RM. EEG source localization using tuations in fMRI signals in monkey visual cortex: implications for
multiple signal classification approaches. J Clin Neurophysiol. functional connectivity at rest. Hum Brain Map. 2008;29(7):
1999;16:225–238. 751–761.
110 Part I ■ Basic Principles

100. Cohen, D. Magnetoencephalography. Evidence of magnetic fields 114. Nunez PL. The brain’s magnetic field: some effects of multiple
produced by alpha rhythm current. Science. 1968;161:784–786. sources on localization methods. Electroencephalogr Clin
101. Reite H, Zimmerman JE, Edrich J, et al. The human magnetoen- Neurophysiol. 1986;63:75–85.
cephalogram: some EEG and related correlations. Electro- 115. Stok CJ, Kouijzer WJJ, Peters MJ. Source localization based on
encephalogr Clin Neurophysiol. 1976;40:59–66. EEG’s and MEG’s. In: Proceedings of the 5th Conference on
102. Geselowitz DB. Magnetocardiography: an overview. IEEE Trans Biomagnetics. Vancouver, Canada; 1984.
Biomed Eng. 1979;26:497–504. 116. Leahy RM, Mosher JC, Spencer ME, et al. A study of dipole local-
103. Murakami S, Zhang T, Hirose A, et al. Physiological origins of ization accuracy for MEG and EEG using a human skull phan-
evoked magnetic fields and extracellular field potentials produced tom. Electroencephalogr Clin Neurophysiol. 1998;107:159–173.
by guinea-pig CA3 hippocampal slices. J Physiol. 2002;544(pt 1): 117. Dale AM, Sereno MI. Improving localization of cortical activity
237–251. by combining EEG and MEG with MRI cortical surface recon-
104. Cohen D, Cuffin BN. Demonstration of useful differences struction: a linear approach. J Cogn Neurosci. 1993;5:162–176.
between magnetoencephalogram and electroencephalogram. 118. Cohen D, Cuffin BN, Yunokuchi K, et al. MEG versus EEG local-
Electroencephalogr Clin Neurophysiol. 1983;56:38–51. ization test using implanted sources in the human brain. Ann
105. Okada YC. Inferences concerning anatomy and physiology of the Neurol. 1990;28:811–817.
human brain based on its magnetic field. Il Nuovo Cimento D. 119. Hansen P, Kringelbach M, Salmelin R, eds.MEG: An Introduction
1983;2:379–409. to Methods. New York, NY: Oxford University Press; 2009.
106. Cuffin BN. A comparison of moving dipole inverse solutions using 120. Lopes da Silva FH. Electrophysiological basis of MEG signals. In:
EEG’s and MEG’s. IEEE Trans Biomed Eng. 1985;32: 905–910. Hansen P, Kringelbach M, Salmelin R, eds. MEG: An Introduction
107.Modena I, Ricci GB, Barbanera S, et al. Biomagnetic measurements to Methods. New York, NY: Oxford University Press; 2009.
of spontaneous activity in epileptic patients. Electroencephalogr 121. Plonsey R. Bioelectric Phenomena. New York, NY: McGraw-Hill;
Clin Neurophysiol. 1982;54:622–628. 1969.
108. Hari R, Ilmoniemi R. Cerebral magnetic fields. CRC Crit Rev 122. Rosenfalck R. Intra and extracellular potential fields of active
Biomed Eng. 1986;14:93–126. nerve and muscle fibres. A physicomathematical analysis of dif-
109. Hämäläinen MS. Magnetoencephalography: a tool for functional ferent models. Acta Physiol Scand Suppl. 1969;321:1–168.
brain imaging. Brain Topogr. 1992;5:95–103. 123. Jackson JD. Classical Electrodynamics. New York, NY: Wiley;
110. Lewine JD, Orrison W Jr. Magnetoencephalography and mag- 1962.
netic source imaging. Functional Brain Imaging. St. Louis, MO: 124. Rall W. Electrophysiology of a dendritic neuron model. Biophys J.
Mosby Year Book; 1995:369–417. 1962;2:145–167.
111. de Jongh A, de Munck JC, Gonçalves SI, et al. Differences in 125. Van Rotterdam A. A one-dimensional formalism for the compu-
MEG/EEG epileptic spike yields explained by regional differences tation of extracellular potentials: linear systems analysis applied
in signal to noise ratios. J Clin Neurophysiol. 2005;22(2):153–158. to volume conduction. In: van Eijnsbergen B, Lopes da Silva FH,
112. Plonsey R. The nature of sources of bioelectric and biomagnetic eds. Progress Report No. PR6. Utrecht: Institute of Medical
fields. Biophys J. 39:309–315. Physics TNO; 1978:115–122.
113. Van Rotterdam A. Electric and magnetic fields of the brain: a sys- 126. Plonsey R. Capability and limitations of electrocardiography and
tems analysis approach. Ph.D. thesis, University of Amsterdam, magnetocardiography. IEEE Trans Biomed Eng. 1972;19:239–244.
The Netherlands, 1986.
CHAPTER

Analog Signal Recording Principles


CHARLES M. EPSTEIN 6
ANALOG AND DIGITAL RECORDING appearance in EEG and other neurophysiologic recordings. The
major entry points and barriers to noise consist of the electrode
Digital analysis and storage have transformed modern neuro- interface, the amplifier inputs, and analog filters. The following
physiology in ways that could only be imagined and envied by sections describe from front to back the different components
the founders of EEG. But the “front end” of all recording sys- of the system and the features that contribute to accurate
tems, the portion that interfaces with the patient, remains ana- recording of neurophysiologic signals.
log; and understanding the operation of analog components
can also give insight into aspects of signal processing that are
now performed mostly by digital techniques. Analog refers to
ELECTRODES
methods of handling data that are physically similar to the orig- EEG and other recording electrodes provide an interface
inal signal. For example, the moving pens and ink traces of the between lead wires, which conduct current as the flow of elec-
original electroencephalographs produced waveforms that mir- trons and human tissues, which transmit it through the move-
rored the electrical potential differences on the scalp, and the ment of ions. Because cutaneous oils and keratin are good
wavy grooves of old-fashioned vinyl records follow the frequen- insulators, placing dry metal electrodes directly on dry skin is
cies and amplitudes of the sound waves produced by stereo sys- usually ineffective. Keratin is often removed by rubbing the
tems. Simple amplification of the microvolt potentials in EEG is skin gently with mild abrasives, and sometimes oils are
an analog process. Analog data “looks like” the original signal, removed separately with alcohol wipes. (Despite such prepara-
which the zeroes and ones of digital representations do not. tions, the epidermis remains the single greatest barrier to cur-
Ideally, each type of data processing would produce a perfect rent flow between the body and the recording system.) In
reflection of the original waveforms. In practice, each is subject addition an ionic solution, or electrolyte, is placed between the
to its own limitations and potential for inaccuracy, which will skin surface and the electrode. As with human extracellular
be discussed in the following chapters. More detailed discussion fluids, the most abundant ions in the electrolyte are sodium
of the material here can be found online in the Guidelines of and chloride. The consistency of the electrolyte ranges from
the American Clinical Neurophysiology Society (1) and in those free-flowing solutions to thick sticky pastes. The latter are used
of the International Federation of Clinical Neurophysiology (2). to help hold cup electrodes mechanically against the scalp. The
former are chosen when the cup is attached by other means
THE HOSTILE WORLD OF CLINICAL and must be filled with electrolyte through a small hole in the
NEUROPHYSIOLOGY middle.
Contact between the electrolyte and the electrode causes
Figure 6.1 shows the approximate amplitudes of electromyo- opposite charges to line up at the metal–ionic boundary. The
graphic activity (EMG), EEG, and evoked potential signals in simplest model for this arrangement is the “electrical double
relation to the many sources of noise that constantly threaten to layer,” first proposed in the 1850s by Helmholtz. The double
overwhelm them. This vast range of voltages requires the use of layer contributes to the stability of the interface and also rep-
a logarithmic scale. So, for example, ECG recorded from the resents a capacitance that augments the apparent current
head can be a thousand times larger than a brainstem evoked flow. At times, especially with new electrodes, the electrical
potential. Within electrified buildings, the 50- or 60-Hz electric double layer undergoes sudden, poorly understood transi-
field on human bodies can be 10,000 times larger than the EEG. tions that produce electrode “pops” and a variety of bizarre
Some physiologic signals, such as EMG and the standing poten- oscillations.
tial on the retina, can also represent noise in relation to smaller The junction between the metal electrode and the ionic elec-
signals that we need to see behind them. trolyte also develops a half-cell potential, which reflects the
At first glance, it may seem impossible that any interpretable electrical force of incipient chemical reactions between them. It
recording could be performed through noise that is orders of also represents half of an electrical battery. The half-cell poten-
magnitude larger than the signal of interest. This chapter dis- tial can be much smaller than that in a flashlight battery and
cusses some of the ubiquitous artifacts in Figure 6.1, the fea- still be enormously larger than the EEG. Ideally this would not
tures of the analog front end that are designed to defeat them be problem, because the half-cell potentials on any pair of
(while allowing faithful recording of the desired signal) and the matching electrodes would be identical, and thus would simply
ways in which they may nonetheless make an unwanted cancel out when those electrodes are connected to an amplifier.
111
112 Part I ■ Basic Principles

Intracranial strip and depth electrodes are generally made


from stainless steel or platinum. The more favorable electrical
properties of platinum tend to be offset by its greater cost.
With modern amplifiers the high impedance and polarization
of stainless steel represent less of a problem than they did in
the past.

LEAD WIRES, JACKBOX, AND CABLES


The lead wires, jackbox, and cables are basically passive compo-
nents whose function is transparent. Excessive separation of the
lead wires should be avoided. The capacitance of extremely long
cables can attenuate very high frequencies. These effects can be
ameliorated by placing the amplifiers inside the jackbox or even
directly on the electrodes and by using fiberoptic connections
to replace electrical cables. In exceptional circumstances, such
Figure 6.1 Potential sources of noise in neurophysiologic recording. as inside the bore of a high-field MRI system, resistors should
Sources may represent noise in some contexts and signals in others. be placed in series with every electrode lead to prevent induc-
Note the logarithmic scale. tion of excessive current. When troubleshooting, it is worth
remembering that if things mysteriously go wrong, the com-
monest culprit is a bad lead wire, cable, or connector.
However, half-cell potentials can be altered in a number of
ways, including drying out of the electrolyte, its dilution by ELEMENTARY ELECTRONICS
sweat or other fluids, relative differences in temperature, or
exposure of different metallic surfaces by wear and tear. These Classically the building blocks of the EEG front end are ampli-
potentials are also affected by movement of the electrode fiers, filters, switches, and potentiometers. The top portions of
against the electrolyte and the skin, which disrupts the electri- Figures 6.2 through 6.6 sketch the simplified “transfer functions”
cal double layer. The effect of movement is reduced by using of an amplifier, a voltage divider, a high filter (which, confusingly,
cup-shaped electrodes, which hold a larger reservoir of moist
electrolyte.
Chemical interactions at the electrode surface consist of
redox reactions, in which an electron is donated or removed
from an ion. Since the product of such interactions is com-
monly a toxic species, such as hydroxyl ions or chlorine gas, care
must be taken to avoid injury of the underlying tissues. The
safest operating zone for recording and stimulating electrodes is
one where they carry only a limited alternating or balanced
pulse current, small enough to be transmitted entirely through
the double-layer capacitance rather than through redox reac-
tions. Passage of direct current through the double layer will
more readily produce chemical changes at the metal surface,
including electrical polarization, which impedes the free flow of
current in one direction more than another, increases imped-
ance at low frequencies, and may be associated with a substan-
tial change in the half-cell potential.
Scalp and other cutaneous electrodes are usually made
from gold—which is inert and therefore produces smaller
half-cell potentials—or silver coated with a layer of silver Figure 6.2 Amplifier. The top sketch shows the amplifier as just a
chloride. Disposable stick-on electrodes almost always use “black box” that magnifies the size of the signal. At bottom left is the
a silver–silver chloride interface. Combined with a chloride standard circuit symbol for an amplifier, with two inputs on the left and
electrolyte, the latter represents the most stable electrode an output on the right. At bottom right is a graph of the output. It shows
available for neurophysiologic applications, resistant to polar- a straight line representing a gain of 2.0 at the top of the y-axis, because
ization and with the best characteristics for low-frequency and the gain is the same at all frequencies. This simple “single-ended”
direct-current recording. Occasionally the exposure of metallic amplifier has asymmetric inputs: one terminal is “hot” and the other is
silver to the light pulse from photic stimulators produces usually connected to system ground (the bent trident symbol). The
synchronous photoelectric discharges, which may be puzzling inherent asymmetry prevents accurate rejection of noise signals, even
to novice interpreters. when they are equal at both inputs.
Chapter 6 ■ Analog Signal Recording Principles 113

Figure 6.3 Resistance bridge. The circuit symbol for a resistor [R] is a
cylinder. The two resistors here are labeled R1 and R2, and both have
the same value. Current from the AC input signal at the top flows
through the resistors to system ground (the bent trident symbol) at the
Figure 6.5 Sketch of how changing frequencies produce the LF behav-
bottom. The output voltage graph is a straight line with a gain of 0.5,
ior graphed in Figure 6.4. At low frequencies (A) the capacitor imped-
because a pure resistive circuit has the same gain at all frequencies.
ance becomes very high, so the capacitor and signal source practically
drop out of the circuit. At high frequencies (B) the capacitor impedance
becomes very low, and the resistor practically drops out of the circuit.
an engineer would call a low pass filter), and a low filter (LF)
(which an engineer would call a high pass filter). The amplifier
(Fig. 6.2) makes the desired signal larger. The purpose of the LF Amplifiers (Fig. 6.2) have the primary purpose of magnify-
is to block frequencies so low that they carry more noise than ing the signal from a range measured in microvolts up to sev-
signal, and thus are better removed. The high filter blocks fre- eral volts, which can do something useful such as moving
quencies so high that they carry more noise than signal. These mechanical pens or undergoing analog-to-digital conversion to
simple functions remain the basic components of analog a stream of numbers. The ratio of the signal coming out of the
recording. For this discussion the amplifier will remain mostly amplifier to its size going in is called gain. For both analog and
a “black box,” which is the way electrical engineers prefer to use digital systems gain should be in the range of 100,000 or more.
modern integrated circuits anyway. The voltage divider and fil- In neither case, however, is gain easily apparent from the system
ters are represented in the form of their simplest analog com- output. This is because the EEG signal is universally shown in
ponents: resistors and capacitors. terms of sensitivity, the ratio of microvolts to millimeters on a
paper or electronic display.
Figure 6.3 shows a simple voltage divider or potentiometer
in the form of a two-resistor network. A resistor is a circuit ele-
ment with two characteristics: it partially blocks the flow of cur-
rent according to Ohm’s law (voltage = current resistance),
and this relationship is independent of signal frequency.
Potentiometers are useful when for any reason voltage must be
reduced rather than increased (i.e., the gain should be less than
one). The signal enters the two-resistor network as the AC volt-
age source at the top and passes through the resistors to system
ground. According to Ohm’s law, if the resistors share the same
current and have equal values, the voltage output at the middle
of the network must be half the voltage at the top. Since the
function of the resistors is the same at all frequencies, the graph
shows a steady gain of 0.5 all the way across.
The middle and bottom of Figure 6.4 show an LF composed
of one resistor and one capacitor. A capacitor consists basically of
two metal plates that are close together but separated by an insu-
Figure 6.4 Low filter (LF). The low-frequency waveform coming out is lator. Current cannot cross the insulator directly, but positive
smaller than the one going in. The circuit symbol for the capacitor (C) and negative charges attract and repel electrostatically on the
is a pair of parallel lines. Current from the AC input signal at the top plates, producing AC current flow in and out of the capacitor.
flows through the capacitor and the resistor [R] to system ground at the The behavior of the capacitor changes predictably with fre-
bottom. The log–log output voltage graph shows a horizontal line indi- quency, according to the equation Z = 1/(2 FC). Here Z repre-
cating a gain of one on the right side, where frequencies are above the sents the impedance of the capacitor to current flow. Impedance,
cutoff. Below the cutoff frequency, gain drops steadily as frequency which is a generic term for all obstacles to electric current, goes
increases. down as either frequency F or capacitance C goes up.
114 Part I ■ Basic Principles

AMPLIFIERS FOR NEUROPHYSIOLOGY


In addition to gain, neurophysiologic amplifiers require three
other essential properties: linearity and flatness across the entire
frequency range of possible signals, high common-mode signal
rejection, and high input impedance. Any reasonable commer-
cial device should have excellent linearity. The other require-
ments are explained below.
Removing the enormous fields caused by AC wiring—and
by other electrical noise (Fig. 6.1)—is the purpose of the
Faraday cages used with the earliest electroencephalograms and
of shielded rooms still required for modern magnetoen-
cephalograms. As an alternative, patients have occasionally
been connected directly to earth ground. Faraday cages are
unacceptably clumsy and grounding patients is prohibitively
dangerous in medical environments, so more practical alterna-
Figure 6.6 High filter (HF). The circuit is simply an inverted LF. The tives have been found. The key to effective recording of small
high-frequency waveform coming out is smaller than the one going in. neurophysiologic signals is the fact that, large as the noise may
The log–log output voltage graph is a horizontal line for a gain of one be, most sources of electrical noise are of similar amplitude in
on the left side, where frequencies are below the cutoff. Above the cut- nearby regions of the body. Thus the ECG potentials at adjacent
off frequency, gain drops steadily as frequency decreases. EEG electrodes are almost identical—at least, in most normal
individuals. The large AC field produced by capacitive coupling
between protoplasm and AC power systems is similar all over
LF gain changes strikingly with frequency. The right side of the body. Potentials that are similar at different electrode sites
the graph in Figure 6.4 shows a value of 1, meaning that the sig- are called common-mode signals. Common-mode signals are
nal passing through is unchanged. On the left side of the graph removed by very precisely subtracting the whole combination
the signal comes through at lower and lower amplitude as the of potentials at one electrode site from the combination of
frequency drops—essentially, low frequencies are filtered out. potentials at another, leaving behind only those potentials
In the middle of the graph is a break point where the imped- that are different at the two “hot” electrodes—the so-called
ance of the capacitor matches the resistance of the resistor. This differential-mode signal. This process is illustrated in Figure 6.7.
break point is the nominal filter frequency, cutoff frequency, or The simpler amplifiers used in radios, stereos, and other famil-
3-dB point.1 iar electronics (sometimes referred to as single-ended ampli-
The behavior of analog filters far from the cutoff frequency fiers) are incapable of the very precise subtraction needed, so
is sketched for an LF in Figure 6.5. It may be understood most that all neurophysiologic amplifiers have some version of the
easily by remembering the equivalence of the capacitor imped- symmetrical inputs in Figure 6.7. The latter are called differen-
ance and the resistor resistance at the cutoff point in Figure 6.4. tial, balanced, or instrumentation amplifiers to distinguish
At lower frequencies the impedance of the capacitor is so high them from the single-ended variety. The two “hot” inputs in
that the AC signal has less and less influence on the output ter-
minal, and the output approaches a short circuit to system
ground. At higher frequencies, however, the impedance of the
capacitor becomes much lower than the resistance, so that the
system ground has less and less influence on the output
terminal—which is now practically short-circuited to the signal
source. The flat passband with a gain of 1.0 reflects this short
circuit.
The high filter (HF) circuit (Fig. 6.6) and its transfer func-
tion are simply the reverse of the LF. The elegant symmetry of
the filter curves is seen only in log–log displays, which is one
reason for preferring this type of graph over linear representa-
tions. Merging the HF and LF curves would form a complete
passband of 1.0 to 100 Hz.
Figure 6.7 Differential amplifier with balanced “hot” inputs and sys-
tem ground. The vertical symmetry of this more complicated circuit
1Readers familiar with decibel notation may notice that the output of the RC fil- allows precise rejection of large common-mode signals that are equal at
ter is reduced by only the square root of 2 when the resistive impedance equals
both “hot” inputs. The relatively small differential-mode signal—the
the capacitive impedance, whereas it is attenuated by a factor of 2 in the two-
resistor network. Explaining the difference requires the use of imaginary num- small square at the far left—is the only signal that passes through the
bers, which will not be attempted here. amplifier.
Chapter 6 ■ Analog Signal Recording Principles 115

Figure 6.7 are symmetrical and equally active, despite the uni- would have infinite input impedance. Although perfect ampli-
versal practice of labeling one of them as “active” and the other fiers do not exist, the highest practical impedance is preferred
as “reference” in many EEG recording montages and in for neurophysiologic recording. Figure 6.8 shows why. Imagine
EMG/NCV studies. Their electrical behavior is indistinguish- an amplifier in which the two inputs have an impedance of
able; the only difference is in terminology. The “ground” or 10,000,000 , connected to two “hot” electrodes with imped-
neutral connection in Figure 6.7 is normally present during all ances Z E1 of 1,000 and ZE2 of 10,000 , respectively. Both elec-
neurophysiologic recording (although modern amplifiers may trodes are picking up an equal 60 Hz common-mode noise
appear to function passably without it). signal of 10 mV. Assuming that the input impedances of the
In EEG, a single “ground” or system reference is shared by all amplifier are still much larger than the electrode impedances,
recording channels and is almost universally placed on the fore- the simplified equation in Figure 6.8 allows calculation of the
head. The ground must be distinguished from the recording ref- result. The unequal electrode impedances will produce a differ-
erence, used in constructing referential EEG montages. The ential-mode noise signal at the amplifier inputs of 0.01
“reference” in referential montages is the second hot terminal of (10,000 – 1,000)/10,000,000 = 0.000009 V or 9 V. This noise
the three-terminal differential amplifier, not ordinarily the will be faithfully magnified by the differential amplifier to pro-
“ground.” For digital EEG, the initial recording is most often duce visible widening of the EEG tracings—just as seen in
performed using a recording reference in the vicinity of the Figure 6.9B.
central-parietal region. With modern electronics, it is possible Since it would be impossibly cumbersome to make all the
to use a single digital recording reference (or one of the regular electrode impedances exactly equal, we make sure all electrode
scalp electrodes) rather than a separate recording reference and impedances are low—in which case the difference between
ground. But noise rejection will be better using both, provided them cannot get too large—and make the amplifier input
there is provision for them in the jackbox. impedances as high as practical. Recent guidelines specify that
The classic differential amplifier takes the common-mode the differential input impedance of the amplifier must be at
noise signal as it comes—whatever its size—and then attempts least 100 M (1). The CMRR should be at least 80 dB
to subtract it out. But smaller common-mode voltages would (10,000:1) at the highest sensitivity of the amplifier when the
make the task easier. A more recent technique takes the common- common-mode signal is applied between both inputs and neu-
mode signals seen by all of the amplifiers on the head, averages tral (1). Amplifier input impedances higher than this can be
and inverts them, and injects the result back into the body obtained without much difficulty, but their performance is
through the former “ground” electrode. Since the necessary cur- liable to be degraded by the capacitance between conductors in
rents are measured in microamperes, no risk is involved. This the lead wires and cables.
method, still referred to anachronistically from its original ECG If the only problem caused by unbalanced (i.e., excessively
usage as “driven right leg,” can cancel and reduce excess noise in large) electrode impedance were 50 or 60 Hz noise, it would be
some applications. It also removes the “ground” label and with it easy to deal with using notch filters. But Figure 6.9 illustrates a
the possibility for a misunderstanding to harm the patient (see
section “Electrical Safety and Ground”).
The ability of an amplifier to pass differential-mode signals
and block common-mode signals is described by its common-
mode rejection ratio (CMRR) and is usually expressed in deci-
bels (dB). Thus an amplifier that reduces the common-mode
signal by a factor of 100,000 (105) relative to the differential-
mode signal has a CMRR of 100 dB. Modern integrated circuit
amplifiers can come off the assembly line with CMRR specifi-
cations of 120 dB (1,000,000:1) or higher. However, many
extraneous factors can defeat them and allow noise to get
through despite the nominal superb performance of the differ-
ential amplifier. One mundane consideration is simply that
common-mode noise is not absolutely identical at different
electrode sites, and the difference grows larger as the electrodes
are moved farther apart. Another example is the induction of
AC potentials in the electrode lead wires if they become widely
separated and form an open loop. A third example is the stim-
ulus artifact from the electrical stimulators used in nerve con-
duction studies and evoked potentials. At hundreds of volts, the Figure 6.8 Model of the differential amplifier showing the impedances
latter can produce an enormous difference at the two hot of the electrodes and the two amplifier inputs. Making some reasonable
amplifier inputs. And very commonly, as discussed below, poor approximations, the equation at the upper right represents the way in
electrode application can defeat the differential amplifier. which unequal electrode impedances ZE1 and ZE2 convert a portion of
An ideal amplifier senses electrical potentials without any the common-mode noise voltage VCM into a differential-mode voltage
current actually passing through the inputs. This perfect device VDM, which passes through the amplifier.
116 Part I ■ Basic Principles

Figure 6.9 ICU EEG study with ground recording artifacts in the F4 channels, including apparent slowing and a spike at
the tip of the arrow (A). 60-Hz noise with the notch filter off (B) is an important clue to the existence of a serious prob-
lem. See text for further discussion.

far more insidious consequence. Panel A of this ICU recording lead on the forehead and magnified across the now-unbalanced
appears to show focal delta slowing involving electrode F4, cul- amplifier inputs. Recognizing the presence of ground recording
minating in an apparent spike-wave discharge. Panel B, with the artifacts, and the importance of AC power line artifacts in
60 Hz notch filter turned off, indicates that in fact there is a assisting that recognition, is vital exactly because these artifacts
poor, high-impedance connection at F4 and that 60-Hz com- may not be obvious. In several decades of teaching EEG, none
mon-mode AC noise has entered the involved channels. The of our residents or fellows has ever recognized or deciphered
crucial message of Figure 6.9 is that the apparent focal abnor- ground recording independently. Recording routinely with the
malities also represent common-mode noise. The F4 slowing is AC notch filter on badly skews the odds that many electroen-
actually time-locked to the ECG and reflects an obscure compo- cephalographers ever would.
nent of the t-wave. The apparent spike is due to the small pha- One corollary of the preceding discussion is noteworthy
sic EMG artifact seen in other leads, picked up by the ground for being even more counterintuitive. Poor, high-impedance
Chapter 6 ■ Analog Signal Recording Principles 117

electrode connections do not generally reduce the size of the earth ground at the power station.2 In modern neurophysiology,
EEG signal. Indeed, as in Figure 6.9, channels 9 to 10, they this feature of the power system is the most important concept
commonly make it look bigger. for electrical safety. To operate electrical devices plugged into
the AC wall jack, current flows from the “hot” wire of the power
RANDOM BACKGROUND NOISE line to the “neutral” wire at earth ground. If patients are con-
nected to earth ground, any stray currents (especially those from
In addition to the physiologic and man-made artifacts the hot power line) will happily flow through the patient on the
described above, several sources of random noise can interfere way there. A third wire, which represents a second and more
with recording of very small electrical signals. Of these the direct earth ground connection, is required in hospital equip-
most irreducible is thermal or “Johnson” noise, which repre- ment as well. This additional ground, commonly referred to as
sents Brownian movement of electrons in resistive conduc- “the” ground, increases safety in some situations—but also con-
tors. The magnitude of thermal noise increases with stitutes a risk if the patient is connected to it. Modern safety
temperature, circuit resistance, and recording bandwidth. At standards require that the recording ground on the patient cannot
body temperature, with a high filter of 70 Hz and total elec- form a direct connection to the power line ground(s) or to other
trode resistances of 1000 , thermal noise is about 0.035 V sources of earth ground, including “the” ground wire.
and unlikely to make any difference in recording. But in a Traditionally discussions of electrical safety have focused on
brainstem auditory evoked potential (BAEP) or concentric “ground loops,” which may be formed when the patient is con-
needle EMG study, high electrode resistance can raise the nected to more than one ground electrode through different
minimum noise to the approximate magnitude of the desired pieces of medical equipment. Decades ago, currents flowing
signal. Some examples of possible recording conditions are through the power line ground system could place the different
given in Table 6-1. Note that other noise sources in the ampli- components of that system at somewhat different voltages—in
fier may increase actual noise figures several times above those which case the voltages would attempt to equalize through the
listed. Recent guidelines specify that the noise level of an EEG body of the patient. But the modern requirement that record-
amplifier must not exceed 2 V rms with the inputs con- ing ground on the patient must never be connected to earth
nected to neutral and with a bandpass of 0.1 to 5000 Hz (1). ground means that ground loops can seldom present a safety
risk using commercial systems. However, ground loops may
ELECTRICAL SAFETY AND GROUND contribute substantially to electrical noise.

In electronic circuits “ground” is a reference point that remains


FULL ANALOG RECORDING
at a virtual zero voltage. Without such a reference point, circuit
operation easily becomes unstable. Many years ago the circuit Prior to the digital era, the vast amount of information con-
ground, and with it the ground electrode on the head, was actu- tained in even a routine EEG could be stored only on paper. The
ally connected to real earth ground through metal plumbing or original signal was amplified up to 1,000,000 times, enough to
other low-resistance conduits. Earth ground is a wonderful power an electromagnetic coil that moved a long mechanical
zero-reference point—a near-infinite reservoir of electrons— pen which carried ink onto fanfold paper. Mechanical pens
which will never be changed by local circuit operation. It is also could move only so fast, setting the upper frequency limit
an unacceptable connection for human bodies, especially in around 70 Hz. They could move only so far, causing the wave-
medical environments. This is because one of the two wires in forms to “square-off” at the limits of the pen range (Fig. 6.10).
alternating current power lines is almost always connected to Special circuitry was used to prevent pens from colliding and
tangling together. The fanfold paper advanced from one tray to
Tabl e 6 . 1 another via a mechanical chart drive with rollers, which could
move at speeds of 15, 30, and 60 mm/sec. The paper was prone
Minimum System Noise Under Representative to ripping or jamming. Ordinarily, EEGs were recorded at
Recording Conditions 30 mm/sec on pages 300 mm wide. The resulting scale of
10 sec/page has been carried over to digital systems. The obso-
Bandwidth Electrode Thermal lete designation of paper speed in mm/sec is so engrained in
Modality (Hz) Resistance Noise older technologists and electroencephalographers that even
( ) ( V) now computer displays are labeled anachronistically in the
same manner.
EEG 35 1,000 0.024
In fully analog EEG systems, constructing different mon-
EEG 70 10,000 0.13 tages requires the use of elaborate mechanical switches. Rows of
BAEP 1,000 1,000 0.13 individual switches are also needed for every channel to set
gain, HF, and LF, and AC notch filters. The available range of
BAEP 3,000 10,000 0.72
EMGa 10,000 100,000 4.1 2Grounding of the power line is necessary because of the astonishingly large
voltage gradients that can develop at different elevations in the atmosphere and
aConcentric needle electrode. over long distances across the earth’s surface.
118 Part I ■ Basic Principles

as compared to about 30 pixels/cm for conventional computer


monitors. To give equivalent resolution, while leaving room for
data labeling, digital monitors would have to be about a meter
wide and incorporate pixel counts that only now are being
approached by high-definition television. Furthermore, paper
can easily be unfolded to provide an overview of several min-
utes’ data while maintaining the same exquisite detail. The
inability of computer displays to show more than 10 seconds
worth of data without severe compromise in frequency resolu-
tion is an underappreciated handicap. It can be overcome only
by knowledgeable adjustment of the display on the part of
Figure 6.10 Detail of two channels from an analog paper writeout. interpreters who understand its limitations for slowly evolving
During a burst of high-voltage frontal theta, the pen in the second chan- activity. Finally, there is no recorded instance of an entire paper
nel reaches the limits of its mechanical range, causing the waveform to EEG tracing vanishing forever on its way to the server, when
“square-off” in the vicinity of the shaded area. Within the shaded area somebody accidentally pushed the “delete” button or when the
is a very narrow spike-like artifact, apparently caused by the collision- operator forgot to hit “record.” And the recent history of digital
avoidance system meant to protect the EEG pens. Note the curvature of recording media suggests that ancient paper records will still be
the high-voltage waveforms and of the artifact, due to the radial nature quite readable when many digital studies can only be viewed in
of the pen movement. a museum.

ARTIFACTS UNIQUE TO
settings is consequently limited, most notably to the legendary ANALOG RECORDING
HF choices of 15, 35, and 70 Hz. A few generations of “hybrid” Artifacts related to the electrode interface, poor common-
systems are likely to still be in use. In hybrid systems switching mode rejection, and ground recording have been discussed pre-
and the associated notations are done electronically, but the viously. Mechanical pen writers are subject to radial
final writeout remains pen and ink. displacement due to the pens moving in an arc rather than in a
In order to produce a comprehensive overview of cerebral straight line (Fig. 6.10)—at times causing inaccurate alignment
activity, several montages must be used during the course of of high-voltage, sharp-contoured waveforms. The ink may skip
each analog recording. Because the physical settings are not or puddle on the page. The moving paper can transiently slip or
noted automatically on the paper record, analog interpreters jam on the roller, stretching or compressing the EEG wave-
rely on the technologist to write by hand every setting and every forms. Figure 6.10 also shows an unusual artifact apparently
change in parameters at the time of recording. Traditionally, caused by the collision-avoidance system in a hybrid electroen-
every combination of parameters is then replicated during a cephalograph. What appeared at first glance to be spike wave
terminal calibration. Producing a true calibration, which faith- consists only of high-voltage theta slowing plus a spike-like
fully displays the performance of the entire pathway from artifact. When the pen reaches the limit of movement, it para-
inputs to writeout, is a virtue of analog recording that not every doxically moves across its full range in the opposite direction
digital system properly duplicates. (On the other hand, the and quickly back. We have observed this behavior in several dif-
mechanical writeout, the weakest link in the analog recording ferent brands of paper electroencephalograph, where it was
chain, has been abolished from digital systems.) misinterpreted as evidence of epilepsy.
Perhaps the greatest limitation of analog recording is the
dependence on the technologist to make notations accurately,
to recognize abnormalities “online,” and to optimize their dis- REFERENCES
play for the reader by making appropriate changes in montages 1. American Clinical Neurophysiology Society Guidelines.
and recording parameters. This feature does, however, have a https://www.acns.org.
silver lining. The technologist by necessity is far more engaged 2. Ebner A, Sciarretta G, Epstein CM, et al. EEG instrumentation.
in the recording and its eventual interpretation. The International Federation of Clinical Neurophysiology.
Other virtues of paper should not be entirely dismissed. The [Practice Guideline]. Electroencephalogr Clin Neurophysiol Suppl.
spatial resolution of paper EEG is in the range of 100 dots/cm, 1999;52:7–10.
CHAPTER

Digital EEG
DOUGLAS MAUS, CHARLES M. EPSTEIN, AND SUSAN T. HERMAN 7
INTRODUCTION AND HISTORY and epileptologists in particular. Practical digital computation
OF DIGITAL EEG began with the development of ENIAC around 1943 to 1944
(2). As early as the 1950s, the potential advantages of computer
Digital electroencephalography (EEG) refers to the recording, analysis of EEG were foreseen (3). However, the routine use of
storage, and review of the EEG on digital equipment (i.e., computers for EEG acquisition and analysis required several
computers). The process requires conversion of the continuous decades for commercial digital electronics to achieve bench-
analog EEG signal, as described in Chapter 6, into discrete marks that equal or surpass the capabilities of analog EEG
numerical values. Over the past 15 years, digital EEG has grad- recording apparatus. Reliable, power-efficient computation
ually replaced analog systems and is now the standard for clin- began with the semiconductor transistor, invented at Bell Labs
ical EEG. Digital acquisition and processing of EEG allows by William Shockley in 1947. The development of integrated
many advantages, but at the same time imposes limitations that circuits in the 1950s permitted mass production and commer-
should be understood by the clinician to avoid inaccurate inter- cialization. A notable computer algorithm for spectral analysis,
pretation. The practical advantages of digital EEG over analog the Tukey–Cooley version of the discrete fast Fourier transform
EEG are summarized in Table 7.1. In this chapter, we will out- (FFT), was published in 1965 (4). First-generation digital EEG
line the principles of digital signal acquisition and processing systems, with limited digitization technology and screen resolu-
that are important in the design and proper use of clinical EEG. tion, could not match the display properties of paper-based
Clinical digital EEG represents the convergence of two scien- EEG (5), but digital electronics gradually achieved fast sam-
tific fields that evolved in parallel for the last 70 years. In 1929, pling rates and larger data throughput. Finally, in the late 1990s,
Hans Berger first reported his studies on the electroencephalo- digitization rates and computer storage available for reasonably
gram (1). The utility of EEG for clinical diagnosis was quickly priced digital commercial products were sufficient to match
established, and EEG has flourished as a tool of neurologists, routine analog EEG machines.

Tabl e 7 . 1

Advantages of Digital EEG

Efficiency Computer-based display generally permits quick review of many studies


Nondestructive processing Raw data are stored digitally, allowing reconstructed display with different montages, different
filters, and different sensitivity
Precision Can precisely measure frequencies and amplitudes for longitudinal or intergroup comparisons
Archiving Easily make exact copies, retain for easy access on disk drive, or save on optical media
(CD, DVD)
Transmission/comparison Digital copies can be easily and quickly transmitted for second opinion or conferences
Increased frequency range Current and near-future digital sampling rates easily exceed the frequency range accessible
by traditional analog EEG
Reliability Stable and reliable electronics, controlled by microprocessors, decreases likelihood of technical
artifacts compared to analog equipment requiring manual calibration for each channel
Portability Amplifiers can be small enough for patients to carry during ambulatory and long-term studies
Digital signal processing Allows extensive on-line or postacquisition analysis, such as automated detection of spikes and
seizures, topographic maps, and graphical displays of quantitative EEG
Associated software May include patient information databases, report writing programs, and integration with
hospital electronic medical records

119
120 Part I ■ Basic Principles

DIGITAL EEG COMPONENTS Routine digital EEG has no requirements for specialized elec-
trodes. Current protocols for experimental digital EEG acquisi-
Figure 7.1 shows the basic components of the digital EEG sig- tion at high sampling rate with intracranial implantation may
nal path from the electrodes to the display. Several components derive benefit from specialized microelectrodes that have a
(jackbox, differential amplifier, display) parallel those encoun- smaller contact area and may be able to detect spatially local-
tered in analog EEG machines (Chapter 6). The features of ized multiunit or even single-unit firing trains.
these components that differ from analog EEG will be high-
lighted in the sections below. Components unique to digital Electrode Input Board or Jackbox
EEG systems include (i) the analog-to-digital converter (ADC), Jackboxes for digital EEG typically have a greater number of
which samples the analog EEG signal and converts it into dis- electrode inputs than analog machines. The standard number
crete digital data; (ii) the computer or microprocessor, which of inputs for routine EEG is 32, but jackboxes accepting 64, 128,
stores and manipulates the digital data for optimal review; or even 256 inputs are common for epilepsy monitoring and
(iii) monitors for display of EEG data; and (iv) digital storage intracranial EEG recordings. Additional inputs can accommo-
devices and networking equipment, which allow archiving of date additional EEG electrodes, such as 10-10 modified
EEG data and transmission to remote sites. Some systems International System placements, as well electro-oculogram,
contain video processors and sound cards for simultaneous electromyogram, or other polygraphic inputs (6). Jackbox
acquisition and synchronization of video and audio data. The inputs are labeled with the location of the electrode on the head
hardware and software capabilities of current digital EEG sys- according to the International 10-20 System (6) or numbered.
tems vary significantly between vendors. This review cannot In addition, there are inputs for ground and for a machine ref-
describe all possible digital system specifications but aims to erence (sometimes called recording reference) electrode, which is
provide a summary of the most important features. usually placed in a location not susceptible to large artifacts,
such as the midline central-parietal region.
Electrodes In contrast to analog systems using electrode selectors, most
Along the signal acquisition path, we turn to the first compo- digital amplifiers are “hard-wired,” with the signal from each
nent—the electrodes. These are discussed in detail in Chapter jackbox position going to Input 1 of a specific amplifier. Input
6. One significant difference from analog EEG is the require- 2 for all amplifiers is the machine reference electrode. Some
ment for one additional electrode, the machine reference, digital systems have electronically switched jackboxes to allow
which we shall discuss below in the section on (differential) amplifier input channels to be switched among different elec-
amplification. As with analog EEG, a good recording depends trodes. Because all EEG activity is acquired in relation to the
on low-impedance electrical contact with the scalp. Most machine reference electrode, the data in all channels may be
current commercial EEG acquisition machines have function- distorted if this reference electrode is of high impedance. With
ality for checking electrode impedances. In general, these are a high impedance reference, external noise may overwhelm the
software automated. For each electrode, the impedance is esti- EEG signal, producing a very low amplitude recording with lit-
mated by forming a complete circuit, starting with that elec- tle signal. (This is different from effects of high impedance in
trode, then creating a return path with all the other electrodes any other electrode, which can increase the apparent EEG
joined (so that section of the circuit has an impedance equiva- amplitude through ground reference recording, in addition to
lent to all other electrodes in parallel, and thus small). A small showing the noise.) To prevent this problem, the EEG should be
(microvolt) voltage (typically oscillatory, on the order of 30 reviewed during the recording in a machine reference montage
Hz) is applied, and the current is measured. The impedance of to check for excessive noise in the reference electrode.
that single electrode contact is thereby estimated by the voltage Digital EEG jackboxes may be housed in the same enclosure
(known) divided by the current (measured). For standard as other digital components, such as preamplifiers (see the sec-
scalp electrodes applied with electrolytic paste, impedances tion “Amplifiers”). The short distance between the scalp elec-
should be less than 5 k . This technique relies, of course, on trode and this first amplification step reduces electrical
the assumption that at least some of the electrodes are well interference and artifact in the EEG signal. ADCs may also be
connected. If all electrodes have poor contact, spurious results present in the jackbox itself, as in systems for ambulatory
can be obtained. recording (7). The jackbox also contains or is connected to an

Electrode
Figure 7.1 The schematic of a digital EEG
machine shows components of the signal Software:
"Jackbox" Differential Anti-alias Analog montage
path from the electrode to the display. digital filter Display
preamp amplifier filter
converter sensitivity
time-scale
Reference
Digital
storage
Chapter 7 ■ Digital EEG 121

Paper Pens Low-cut High-cut Montage


Figure 7.2 Schematic of amplifier inputs for analog EEG for
(fixed) (fixed) (fixed)
a longitudinal bipolar montage. One additional electrode
Fp1−F7 + Fp1
Fp1−F7 0.5 Hz 70 Hz input—the ground—is omitted for simplicity.

F7
F7−T3 +
F7−T3 0.5 Hz 70 Hz

T3
T3−T5 +
T3−T5 0.5 Hz 70 Hz

T5
T5−O1 +
T5−O1 0.5 Hz 70 Hz
− O1

impedance meter to allow measurement of electrode imped- (In our figures, we show the electrical symbol for an op-amp,
ance (see the section “Electrodes”) without unplugging the short for operational amplifier. Op-amps do amplify the differ-
electrode wires from the electrode input panel. ence between their inputs, but this is a simplification. Actual
hardware differential amplifiers are more complex circuits, using
Amplifiers a combination of op-amps with other circuit elements, specifi-
The first active components in the signal path are the cally designed and tuned to amplify the difference in voltage
(pre)amplifiers. Prior to digitization, the signals—which are in even when both voltages may be quite large in absolute value.
the microvolt range—must be amplified. This may be done via See more about common-mode rejection below.)
one or more cascaded amplifiers. Modern amplifiers are small, In analog EEG, the montage is fixed at acquisition time by
low-power, single-chip multichannel devices with solid-state the fact that the electrodes are mechanically connected to a set
integrated circuits. Noise (generated by random movement of of differential amplifiers for that montage. Figure 7.2 shows a
electrons within amplifier circuits such as resistors and semi- simple diagram of the arrangement of differential amplifiers for
conductors) should be less than 2 V peak to peak. Amplifiers analog EEG for a longitudinal bipolar montage. Digital EEG
are electrically or optically isolated to prevent conduction of also employs differential amplifiers, but with a significant dis-
current from the EEG machine to the patient through the EEG tinction. Instead of determining the difference between elec-
electrodes. trode inputs at acquisition time, each electrode is acquired
Properly designed, the first amplifier (often called a preampli- relative to one fixed common electrode, the machine reference.
fier) has high input impedance (on the order of mega- to tera- These individual electrode channels are then digitized and
ohms), which allows the source signals to be precisely captured, stored. Later review with tracings displayed in different mon-
without drawing significant current which would create distor- tages can be performed by digitally reconstructing the mon-
tions in the voltage to be measured (8). They also raise the signal tages. Figure 7.3 shows a simple diagram highlighting this
into a range where stray device noise (powerline, television, difference from analog EEG. Note that the vertical dashed line
radio) is negligible and so cannot disturb the signal. Finally, com- separates the fixed, unalterable acquired data on the left, from
mercial ADCs have an expected input range, such as 5 V. To computations on the right showing the data in one of many
take full advantage of the amplitude resolution of the ADC, input possible different montages, which can be easily reconstructed.
signals must be brought up to this voltage range by amplification Also note that the input to the inverting contact for each differ-
(though not over it, since this would “clip” the signal). ential amplifier is the machine reference for all the electrode
For EEG, one of the most important aspects of the amplifi- channels recorded.
cation stage is the use of differential amplifiers. Both analog and There are several other aspects of amplifiers that could
digital EEG employ differential amplifiers. Differential ampli- impact the data acquisition. The common-mode-rejection ratio
fiers have three input connections: (i) inverting input; (ii) non- (CMRR) is a measure of how well the differential amplifier
inverting input; and (iii) ground, typically placed on the reflects the difference between the inputs, especially when both
forehead, and a single output. The output is the difference inputs are large in magnitude. Real differential amplifiers can
between the two inputs multiplied by some constant—the be best modeled as generating an output voltage V0 as
amplification factor, or gain. Gain for clinical EEG machines is 1
usually in the range of 2000 to 20,000, but can be up to V0 Ad (V V ) 2As(V V )
1,000,000. The gain equals output voltage divided by input volt- The coefficient Ad is the differential gain and As is called the
age. Gain is typically expressed as a logarithmic ratio in decibels common-mode gain. The common-mode rejection ratio is
(dB), with gain (dB) = 20(log(Vout/Vin)). For instance, with
inputs of 106 V (noninverting) and 104 V (inverting), and Ad
CMRR 20log10 a b
amplification factor (gain) of 20, the output will be 40 V. As
122 Part I ■ Basic Principles

Figure 7.3 Differential amplifiers Digital


for digital EEG, with each electrode Transmission
Montage
input amplified compared to the +
/Storage
Fp1
machine reference. Computer soft- ADC …00101010… ‘Fp1’

ware then converts this referential ‘Fp1−F7’
montage to a longitudinal bipolar 10111000
F7 +
montage. See text for details. The ADC …01110010… ‘F7’
− Computer Display
ground electrode is omitted for
‘F7−T3’
simplicity. 00001001 Fp1−F7
T3 +
ADC …01101001… ‘T3’ F7−T3

‘T3−T5’ T3−T5
01010110
T5 + T5−O1
ADC …00010011… ‘T5’

‘T5−O1’
10111111
O1 +
ADC …01010100… ‘O1’

Ref

So, a CMRR of 80 dB, as recommended by American Clinical more components to achieve a faster roll-off in the transition
Neurophysiology Society (ACNS) (9), indicates that the ampli- band, thus a “sharp” cutoff. Such analog filters may be designed
fier will begin to lose accuracy when the absolute magnitude is according to Butterworth or Chebyshev topologies (11).
10,000 (104) times the difference in magnitudes (e.g., 10,001 Other filters can be inserted prior to digitization. DC is not
V – 10,000 V). Most of the common mode signal is 60 Hz commonly used in clinical EEG but may be used for research
noise, and the magnitude of that noise is determined more by purposes. In most commercial systems, filters are inserted to
the electrode impedances than circuit specifications. remove the DC (constant) and very low-frequency components
( 0.5 Hz), which make the record harder to read and require a
Analog Filters larger magnitude resolution for the ADC. These have the addi-
Prior to digitization, analog electrical filtering is applied. The tional benefit of maximally exploiting the ADC resolution, since
fundamentals of analog filter design are discussed in Chapter 6. the average signal is then zero, and the greatest positive and neg-
In contrast to analog machines, digital EEGs are acquired using ative excursions can be designed to fit within the digitizer input
broad bandpass filter settings, typically 0.1 to 70 or 100 Hz. range to take full advantage of the magnitude resolution of the
However, for specialized applications, the bandpass may ADC. Another filter that may appear before digitization is a
include very low frequencies (0.001 Hz) or even direct current power line “notch” filter, which is specifically tuned to eliminate
(DC), as well as high frequencies up to 3000 to 10,000 Hz (10). the frequencies matching that of the power line: 60 Hz in the
The most important filter in this stage is the anti-aliasing fil- United States, 50 Hz in Europe and other countries.
ter as described below. This requires a low-pass filter (passes
low-frequency signals but attenuates high-frequency signals). Analog-to-Digital Conversion
The simplest low-pass analog filter is a resistor (R) and capaci- The stage of conversion of the analog signal to digital represen-
tor (C) in series, with the voltage across the capacitor transmit- tation is the most crucial step in the acquisition process. Design
ted to the next stage, as diagrammed in Figure 7.4. This of an acquisition system requires that the components be
first-order low-pass filter will attenuate high frequencies and matched and appropriate as a whole, so that the final data are
has a cutoff frequency (in Hz) of 1/(2 RC). Commercial anti- not hindered by poor design of any one element in the path.
alias filters may be this simple but are often engineered with Because parameters of the digitization impact acquisition com-
ponents both earlier and later in the signal path, thorough
R understanding of digitizer capabilities and limitations is crucial.
The act of digitization collapses an EEG signal in two independ-
ent axes: time and magnitude.

Vin C Vout Time


Typically, a time-varying signal is digitized at fixed (constant)
discrete intervals. The time intervals are described by sampling
rates. Typical sampling rates are on the order of several hundred
Figure 7.4 A low-pass analog filter is a resistor (R) and capacitor (C) in samples per second (Hz). For 200 Hz, this implies that samples
series, with the voltage across the capacitor transmitted to the next stage. are taken every 5 msec. The time between subsequent samples
Chapter 7 ■ Digital EEG 123

A Figure 7.5 Aliasing. A: A pure sinusoid at 11 Hz


+1
(the solid curve) sampled at 16 samples per second
(Nyquist frequency is then 8 Hz). The sampled data
points can be seen to equivalently match a sinusoid
at 5 Hz (dashed curve). When displayed on a screen
(connected by straight lines), these points would
more closely match the 5-Hz sinusoid than the origi-
nal 11-Hz sinusoid. B: Frequencies above the Nyquist
0 are “folded back” across the Nyquist, as if the paper
0.5 1 were folded with the crease at the Nyquist fre-
quency—best exemplified by the solid curves of the
frequencies between the Nyquist and the sampling
rate. The heavily darkened curve represents the alias-
ing of 11 to 5 Hz from A.

−1
Seconds

0 Nyquist Sample
(8 Hz) rate

is called the dwell time. The choice of sampling rate impacts (solid curve). Indeed, in practice, when displayed on a screen
how well the obtained data reflect the analog source, with faster (connected by straight lines), these points would more closely
sampling roughly giving more fine detail. A reasonable question match the 5-Hz sinusoid than the original 11-Hz sinusoid. This
is: How fast is fast enough? A well-known theorem from signal phenomenon is known as “aliasing.” Figure 7.5B shows the pat-
processing literature provides a quantitative answer to this tern by which frequencies above the Nyquist are “folded back”
question. The Nyquist theorem states that the highest frequency into the range between 0 and the Nyquist frequency, with the
distinguishable is one half the sampling frequency. The down- example of 11 Hz being folded back to 5 Hz at sampling rate of
side of more rapid sampling is the creation of larger EEG files. 16 samples per second emphasized with the bold arc.
Armed with the Nyquist theorem, one knows the limitations Figure 7.6 shows the deleterious effect of noise above the
of what signal components in the source are capturable, but the Nyquist frequency. Fortunately, methods to eliminate this artifact
question is naturally raised as to the consequence of signals that are quite simple. Electrical filters—specially designed circuits
are above this Nyquist limit. If these were simply eliminated, the composed of elements such as resistors and capacitors—can be
issue would be moot, but sadly high-frequency components used to eliminate undesired frequencies in the analog signal prior
above the Nyquist can actually create nefarious artifacts in the to digitization. In particular, ADC systems incorporate an analog
digitized output. Figure 7.5A shows an example of a pure sinu- low-pass filter prior to digitization, with this filter designed to
soid at 11 Hz (the dashed curve) being sampled at 16 samples pass frequencies below the Nyquist frequency and eliminate
per second (Nyquist frequency of 8 Hz). The sampled data those above it. Such a low-pass filter in this context is termed an
points can be seen to equivalently match a sinusoid at 5 Hz “anti-aliasing” filter.
124 Part I ■ Basic Principles

Spectrum

Original
0 Hz 256
Low−pass filtered
Filter

0 0

0 2.0 0 2.0
Noise folded
Digitize Digitize
128 Hz 128 Hz

0 0

0 Seconds 2.0 0 2.0

Figure 7.6 Deleterious effect of noise above the Nyquist frequency. Top left panel shows 2 seconds of “original” (simu-
lated) data composed of a signal at 5 Hz, plus noise with significant components up to 256 Hz. The inset (just above and
to right) shows the power spectrum of this data, showing the white noise and the large signal at 5 Hz. Bottom left panel
shows what would be the result of naïve direct digitization at 128 samples per second, rendering the signal at 5 Hz nearly
unrecognizable within the abundant noise. The signal-to-noise ratio (SNR) has actually been worsened because the noise
above the Nyquist limit (64 Hz = half of 128 samples per second) has been folded back, adding to the noise in the fre-
quencies below the Nyquist, as illustrated in its power spectrum. The right side panels show how anti-aliasing filtering
prior to digitization can prevent this problem. Top-right panel shows the tracing after a low-pass filter (modeled as
Butterworth third order) with cutoff at 50 Hz has been applied. Bottom-right panel shows the result of digitization (again
at 128 samples per second) of this filtered tracing. Frequencies above the Nyquist (64 Hz) have been attenuated by the
low-pass filter and are therefore not folded back, so digitization in this case yields a faithful reproduction of the signal.

The anti-alias filter and ADC in Figure 7.1 are deliberately reflects the fact that two sinusoids with some difference in fre-
joined. The analog hardware anti-alias filter must obviously quency will become out of phase with each other over that time
precede the ADC. The anti-alias filter cutoff must be appropri- frame. For instance, to reliably distinguish a 0.1 Hz difference in
ate for the sampling rate of the joined ADC. The filter must frequency requires a total acquired signal on the order of 1/(0.1
adequately attenuate frequencies above the Nyquist, or else Hz) = 10 seconds. This applies irrespective of the absolute fre-
high-frequency noise will be aliased into the digitized signal. If quency—that is, distinguishing 8.3 from 8.4 Hz or 124.7 from
the cutoff is too low, then signals with frequencies above this 124.8 Hz both require approximately 10 seconds of acquisition.
cutoff and below the Nyquist will be unnecessarily attenuated
and lost, and expense will have been wasted in using an ADC Magnitude or Amplitude
with higher sampling rate than necessary. Digitization occurs along the magnitude axis as well. In this
Another point regarding the time axis regards the “frequency respect, the effect of digitization is typically discussed in terms of
resolution,” a factor important for quantitative analysis of EEG. digitization “depth.” Digitizers presume that each successive
The sampling rate does not impact whether one can distinguish a point is at a known time interval, so this time interval does not
signal as being one of two closely spaced frequencies, for example, need to be stored. Instead, only the value or magnitude at each
8.3 Hz versus 8.4 Hz. Frequency resolution is determined by the successive point is stored. Digital equipment is optimized when
duration of the acquired signal. In general, the duration required storing each successive magnitude in a predefined length. To be
is approximately the inverse of the difference in frequency. This concrete, the output is a stream of 0’s and 1’s. The number of
Chapter 7 ■ Digital EEG 125

Figure 7.7 The practical effect of 4-, 6-, and 8-bit


digitization depth. Note carefully that for the 6-bit
depth, the digital step is most apparent at the peak
and trough, where for each time step the voltage
change is small, but these changes are smaller than
the smallest discrete step and so cannot be repre-
sented.

4-bit 6-bit 8-bit


−7 to +7 −31 to +31 −127 to +127

binary digits (bits) set aside for each measurement is the same. the minimum, with 12 bits or more being preferred. The mini-
This storage length can differ between equipment manufacturers. mum amplitude resolution is recommended to be 0.5 V.
Early digitizers used 8, 10, or 12 bits to store each measurement. Regarding the sampling rate, the guideline has no absolute min-
Digitizers now routinely use 16 to 24 bits. The number of bits imum sampling rate but states that “Higher rates are prefer-
determines explicitly the number of distinguishable magnitudes able.” The guideline does suggest that “minimum sampling rate
of the measurements. For example, 8 bits per measurement can (be) three times the high-frequency (anti-aliasing) filter,” which
store up to 28 = 256 different values (0 to 255 unsigned, or –127 is equivalent to saying that for a given sampling rate, the anti-
to 127 signed two’s complement system). Figure 7.7 illustrates aliasing filter cutoff be at 2/3 the Nyquist frequency. Since the
the practical effect of 4-, 6-, and 8-bit digitization depth. ADC rate and anti-aliasing filter are hardware determined,
The three parameters—maximum value (or dynamic range), these are under the control of the manufacturer but should be
minimum distinguishable difference, and digitization depth— investigated and documented clearly when purchasing deci-
are interconnected, and not independent. Choosing any two sions are considered.
specifies the third. With a 12-bit linear digitizer (212 = 4096 dif-
ferent levels), in order to be able to represent magnitudes up to ADC Hardware
1 mV, then the “resolution”—the smallest detectable differ- Some of the basic principles of the hardware involved in AD
ence in magnitude—will be (2000 V/4096) ~0.5 V. The more conversion are worth mentioning. First, no AD converter oper-
bits available, the more accurate each sample. For very low- ates instantaneously. The input voltage is typically measured
amplitude signals (e.g., electrocerebral inactivity recordings), over some finite time (obviously less than the time between
ADC resolution less than 0.5 V is necessary (12). samples). The time over which the signal is actually measured is
called the conversion time. Sampling skew, or loss of time axis
ADC Effects on File Size integrity, can occur if the ADC samples each channel sequen-
Both sampling rate and digitization depth have an impact on tially, since it takes some time to sample one channel and con-
acquired file lengths. For example, for 32 channels with digiti- vert the amplitude to a numerical value before it moves on to
zation of 16 bits (2 bytes) per sample and a sampling rate of 400 the next channel’s sample. Sampling skew is most problematic
samples per second, 1 hour of EEG (3600 seconds) will gener- with high-frequency activity and may cause misalignment of
ate (32 2 400 3600) = 92,160,000 bytes = 90,000 kB = high-frequency events such as spikes between the first and the
87.89 MB. A full day of EEG with these settings would generate last channels sampled. Most ADCs use an input circuit called a
~2 GB. Increasing the sampling rate to 1000 Hz would increase sample-and-hold, employing a capacitor to “hold” the voltage
the file sizes by 2.5, generating ~5 GB per day. during this conversion time. Then, comparator circuits are
applied to bracket the “held” voltage to the desired precision. A
Guidelines second method is to have a series of ADCs, one for each chan-
There are specific guidelines for minimum requirements of dig- nel, all triggered by a single computer command. Some digital
ital EEG acquisition, specifically touching on ADC. The ACNS instruments have combinations of these two methods, with
guideline (9) suggests that 11-bit depth (including sign bit) is ADCs for blocks of four to eight channels using the “sample
126 Part I ■ Basic Principles

and hold” method. Since large ADCs with high throughput are amplifier to be plugged into a network jack near the patient and
expensive, multiple ADCs may be more cost-efficient. to stream data to an acquisition machine anywhere on the net-
There are various ADC hardware structures available. These work. Advanced LTM recorders may have bluetooth connectivity
structures include direct conversion or flash, successive approx- and can wirelessly stream data to nearby storage (15) and often
imation, ramp-compare, delta-encoded, pipeline, and sigma- have battery backup and local storage to allow patients increased
delta. Some of these structures actually involve complex mobility while ensuring uninterrupted data acquisition.
combinations of simpler ADC designs. For example, the
sigma–delta converter first oversamples at a very high rate, dig- Video and Audio Acquisition
itally filters and may use a Flash ADC, then finally downsamples Simultaneous video and audio recording is most commonly
to the desired sampling rate. done in LTM for epilepsy and in the intensive care unit, to allow
The most obvious trade-off of AD converters is between correlation of EEG features with behavioral events (16). Most
sampling rate and number of channels. The product (rate digital EEG systems today utilize digital cameras and store EEG
channels) is basically a fixed constant, with this constant being and video data to computer hard drives, although some older
proportional to the expense of the digitizer. With current hard- systems with analog video and audio recorded to videotape are
ware digitizers, it is often possible to sample very fast (2 kHz) still in operation (17). Early proprietary video capture systems
on a handful (~16) of channels, or at slow rates (256 Hz) on have largely been replaced by standard digital video recording
several dozen channels (64 to 256). Most commercial EEG equipment, markedly decreasing the cost and increasing the
machines aim for a compromise with the ability to monitor 32 efficiency of video acquisition and review. Digital video
to 64 channels at sampling rates of 256 to 1024 Hz, and these recorded directly to hard drives can be accessed randomly,
rates can be user programmed according to the task at hand. allowing the reviewer to quickly jump to any portion of the
record, rather than having to fast-forward or rewind a series of
Calibration videotapes. Video equipment can be customized to optimize
Digital calibration differs significantly from analog calibration. recording for specific types of monitoring. Camera options
The ADC must be calibrated so that it “knows” how large to include analog versus digital, color versus black-and-white, low
represent each voltage point. A 50- or 100- V signal (usually a light versus infrared, fixed versus pan-tilt, and wide-angle ver-
30-Hz sine wave) is passed through each amplifier. The ADC sus remote zoom. Many systems have controls for remote pan-
takes the amplifier outputs and assigns a 50 V value to the tilt and zoom functions. While early systems required custom
peak of each channel. All other amplitude values are deter- cabling for video, modern systems can transmit and control
mined based on this initial calibration. Calibration information digital video over standard TCP/IP cables.
is saved with each EEG record. The digital EEG software should Video and audio are usually encoded into MPEG, MPEG2, or
contain a voltage cursor that allows the user to determine the MPEG4 formats. These are standard formats for digital video,
voltage of individual waveforms. This cursor is then applied to differing in resolution and compression algorithms. All data
the known signal from the signal generator to determine should include time markers so that EEG, video, and audio can
whether the system is properly calibrated. be synchronized precisely (18). Video should be synchronized to
Square wave calibration pulses can sometimes be generated EEG in the millisecond range, or frame-by-frame. Video files
using a digital EEG machine to verify the amplifier and analog contain large amounts of data; 24 hours of video recording can
filter functioning. The calibration signal should be time-locked be 8 to 30 GB, depending on resolution (usually 320 240 pix-
to the ADC clock (13). The square wave calibration should be els or 640 480 pixels), color depth, frame rate, and data com-
examined in the machine reference montage with all digital fil- pression algorithm. EEG machines for video-EEG recording
ters off. Biologic calibration may not be possible with digital should be able to store at least 24 hours of continuous video and
EEG machines. EEG data. Currently available systems, with hard drive capacities
of 200 GB or more, may allow continuous recording for more
Ambulatory and Long-Term EEG Portable Recorders than a week. Because of the size of video files, the entire video is
Systems for acquisition of long-term EEG (hours to days) can not usually archived; rather, video segments of interest (seizures
have a variety of configurations. The number of hours of EEG and behavioral events) are clipped and archived.
that can be stored depends on the number of channels recorded,
recorder battery life, and memory storage capacity. Fully portable Computer and Software
ambulatory recorders, designed for home EEG recording, are The EEG acquisition and digitization devices described above are
small enclosures containing an amplifier, ADC, batteries, and connected to a central computer, which can be either a standard
data storage (usually flash memory card) (7,14). Modern systems desktop personal computer or a laptop computer. The computer
can store up to 96 hours of continuous 16–32-channel EEG data. contains a central processing unit (CPU) that performs calcula-
When recording is complete, data are downloaded to a computer tions, random access memory (RAM) which holds data tem-
via a universal serial bus (USB) or proprietary connection for porarily while they are being manipulated, and a hard disk drive
review. for permanent data storage, as well as a monitor, keyboard,
In systems for long-term monitoring (LTM) of epilepsy, mouse, network connectivity devices, and various connections for
amplifiers/ADCs can be connected directly to a computer via removable media such as CDs, DVDs, and external hard drives.
USB or other cable, or have TCP/IP connectivity that allows the Most EEG vendors currently use standard commercially available
Chapter 7 ■ Digital EEG 127

computers, running on commonly used software platforms such


as Microsoft Windows or Macintosh operating systems.
Most of the variability in modern digital EEG systems comes
from proprietary software applications that allow customization
of EEG acquisition as well as manipulation and display of the EEG
and video data through a graphical user interface (GUI). Features
of review software are discussed in more detail below. In addition,
vendor-provided software applications may include databases for
patient and study information, interfaces to hospital patient infor-
mation systems, report-generation software, security packages,
and a variety of postacquisition signal processing software.
Because the central machine is a standard computer, review sta-
tions can also run other software that may enhance laboratory
productivity such as word processors, voice recognition software,
and hospital electronic medical record systems.

Display
Digital EEG can be displayed in a variety of methods. The
recorded signal can be output to an oscilloscope, printed out on
paper, reconverted to analog signal via digital-to-analog con-
version (DAC) and written by traditional pen-writing systems,
or viewed on a computer monitor. The most practical and ver-
satile method is display on a computer monitor.
Visual display on screen is also a digital process. Whether dis-
played on older CRT (cathode ray tube) or modern digital LCD,
images are drawn as discrete pixels. In fact, current display pixel
resolution may be the limiting factor impeding precise represen-
tation of the signal characteristics to the electroencephalogra-
pher (19). The EEG signal consists of points discretized in time
and voltage but obviously not necessarily in adjacent pixels, so
intermediate pixels are filled to construct lines connecting the
points. The top diagram in Figure 7.8 is an idealized example of
a set of such points and where they might lie on a pixel display. Figure 7.8 Effects of horizontal pixel resolution on EEG display.
Modern software draws lines between these points using line- Supposing two time steps per pixel-width (e.g., 200 Hz digital sampling
drawing routines based on Bresenham’s algorithm (20). rate and 1000 pixel-widths for 10 seconds = 100 pixel-widths per sec-
Screen resolution is described in terms of the number of hor- ond). Top panel shows idealized signals at 12.5, 25, 50, and 100 Hz (the
izontal and vertical points or pixels. In 2010, typical mid-range digitization Nyquist frequency). Bottom panel shows how these fre-
display monitors may have 1280 pixels across with 1024 pixels quencies would be actually drawn with pixels. As can be seen, 50 Hz is
vertically. We examine horizontal resolution first. Display of 10 the highest frequency that can be visualized with these pixel settings
seconds of EEG on a screen implies each second being allotted (and not terribly well) with frequencies above 50 Hz appearing
128 pixels, in the best case. If the sampling rate was 256 samples “smudged.” In analogy with the digitization Nyquist frequency, the
per second, then horizontally there would be two EEG points highest frequency represented by pixels can be said to be half the num-
per pixel. Suppose there were (for simplicity) 100 pixels per sec- ber of pixels per second (100 pixel-widths per second can represent
ond, and an EEG channel sampled at 200 Hz, with signals up to signals up to 50 Hz).
100 Hz (which is at the Nyquist frequency, and representable in
the digitized data stream). Figure 7.8 shows a set of points with
sine waves up to 100 Hz. Unfortunately, by the Bresenham line occur when viewing fast frequency activity, such as 60-Hz arti-
drawing algorithm, these signals will not be able to be traced fact or EMG activity, and can occur even when following rec-
out, instead resulting in blurred uninterpretable lines, as shown ommended guidelines for video resolution. When such video
in Figure 7.8 (bottom). In general, the maximum frequency aliasing artifacts are suspected, decreasing the number of sec-
faithfully representable on a pixel display is one half the number onds displayed per page (e.g., from 10 to 5 seconds) or increas-
of pixels per second. In this case, that would be (100/2) = 50 Hz. ing screen resolution will allow display of all samples in each
Display of EEG on monitors with inadequate resolution (i.e., second and resolve the artifact (Fig. 7.9).
lower than the sampling rate) can result in aliasing, the intro- We now turn to the vertical display resolution. The dimen-
duction of spurious waveforms caused by “undersampling” the sion of pixels on current LCD screens is on the order of 0.25
EEG signal (21). This is analogous to violating the Nyquist the- mm. If we try to replicate the vertical scale of analog EEG,
orem when choosing a sampling rate. Aliasing artifacts typically then 7 V/mm translates into 7 V over 4 pixels. Thus, each
128 Part I ■ Basic Principles

Figure 7.9 Aliasing by inadequate moni- A


tor resolution. A: EEG was viewed on a
hospital computer with monitor resolution
set at 800 600 pixels. An unusual 9.5-Hz
artifact is seen in channel T6 – reference.
B: When the EEG was viewed on a moni-
tor with screen resolution 1600 1200
pixels, this signal is seen to be a sinusoidal
60-Hz artifact.

50 µV
1 sec
B

50 µV
1 sec

pixel spans about 2 V in vertical height. With current digi- the sensitivity to 2 V/mm. With our 1024 pixels in the ver-
tizers (12- or 16-bit), the amplitude resolution is typically tical dimension, and viewing 20 channels, we see that we have
better than 0.5 V, perhaps as low as 0.03 V. Figure 7.10 approximately 50 pixels per channel. Using our standard 7
shows an idealized example with a low-amplitude signal that V/mm, EEG signals of 50 V can be displayed. Signals
ranges from 1 V to –1 V. This variation is lost with pix- larger than 50 V will begin to spill over into adjacent
els of 0.25 mm at a sensitivity of 7 V/mm, resulting in a flat channel display, causing overlap of tracings in adjacent chan-
line. Conversely, to achieve 0.5 V per pixel requires setting nels and making the EEG more difficult to interpret.
Chapter 7 ■ Digital EEG 129

should be able to print at resolutions of 600 dots per inch


(DPI), or approximately 350 samples per second. Optimally, the
system should be able to print a subset of channels on each page
to maximize vertical resolution.

COMPUTER ACQUISITION AND REVIEW


Digital EEG has several advantages over analog recordings (22). In
routine clinical practice, the ability to reformat montages, change
sensitivities and “paper speed,” and manipulate filter settings dur-
ing EEG review are some of the major advantages. For analog
EEG, what the technologist records is what the electroencephalo-
grapher reviews, with no ability to change or reformat data to bet-
ter localize abnormalities or exclude artifacts. Digital EEG allows
review of the data “as acquired” (as the technologist viewed the
data as they were being recorded) or with a nearly limitless array
of post hoc montage, sensitivity, and filter settings.
Digital reformatting improves EEG interpretation. Levy et al.
(23) demonstrated that interrater agreement for “as-acquired”
digital EEG was the same as for paper EEG, with weighted kappa
scores of approximately 0.65 (good agreement). When elec-
troencephalographers were given the ability to reformat the
digital EEG, however, interpreter agreement improved to kappa
scores of 0.8 (very good agreement). Montage reformatting was
most useful for distinguishing normal variants from abnormal
patterns, identifying artifacts, and classifying abnormalities as
focal or generalized. Correlation of digital EEG with simultane-
ously recorded video also enhances the ability to detect and cor-
Figure 7.10 Effects of vertical display resolution on EEG. Idealized
rectly identify environmental and biologic artifacts.
example with a low-amplitude signal that ranges from 1 to –1 V.
Digital EEG can be reviewed page by page (usually 10 seconds
This variation is lost with pixels of 0.25 mm at a sensitivity of 7
per page) or in continuous “scroll” mode. Scroll mode is typically
V/ mm, resulting in a flat line.
used during acquisition or when reviewing simultaneous video
and EEG, and page mode is used during review. Page mode is
much faster; up to 5 to 10 pages can be presented each second.
Several methods can be used to improve signal display. Only superficial features of the EEG can be determined with this
Screen size for review stations should measure 20 diagonally or rapid paging speed, but it may be useful for screening for seizures
more. Dot pitch (the diagonal distance between display pixels) or determination of sleep states. In page mode, EEG abnormali-
should be as small as possible. Display is best when the moni- ties or comments at the edge of the page may be missed. This can
tor’s “native” screen resolution is used. Large (20 or 24 ) mon- be avoided by overlapping the pages by several seconds.
itors with high resolution (1600 1200 pixels, or widescreen
1920 1200 pixels) are currently reasonably priced, and higher Patient Information
resolution larger monitors are available for those with larger At a minimum, the patient’s name, date of birth, medical record
budgets (Table 7.2). A minimum horizontal resolution of 100 number, laboratory EEG number, and the date the study was
pixels per second is recommended (9). For lower resolution performed should be recorded and stored with the EEG (24).
monitors, decreasing the number of seconds per screen can This information can be stored in the same file as the EEG data
allow display of every data sample point. Similarly, for vertical itself, as a separate text file, or in a database linked to the EEG
resolution, one can reduce the number of channels displayed or data file. Most systems allow recording of more detailed infor-
use a “zoom” feature to hone in on waveforms of interest. Dual mation including patient history, medications, and recording
monitor displays may be a particularly attractive option. Two conditions. More advanced systems may include report-gener-
side-by-side 24 widescreen WUXGA monitors, rotated 90 ation software, allowing reports to be stored with the digital
degrees, give an effective resolution of 2400 1920 pixels. This EEG record.
allows viewing of nearly every data point in 10 seconds of EEG
sampled at 250 samples per second. Data Labeling
Digital EEG signals can also be printed on paper. Printers During recording, the EEG data should be automatically
using fan-fold paper allow printing of a continuous EEG record labeled with time of day, all montage, sensitivity, and filter set-
similar to that acquired with analog systems. Laser jet printers tings used by the technologist during recording, and start and
130 Part I ■ Basic Principles

Tabl e 7 . 2

Commonly Used Computer Monitor Resolutions

Code Name Aspect Ratio Resolution


VGA Video Graphics Array 4:3 640 480
SVGA Super Video Graphics Array 4:3 800 600
XGA eXtended Graphics Array 4:3 1024 768
SXGA Super eXtended Graphics Array 5:4 1280 1024
WXGA Widescreen eXtended Graphics Array 16:9 1366 768
UXGA Ultra eXtended Graphics Array 4:3 1600 1200
WSXGA Widescreen Super eXtended Graphics Array Plus 16:10 1680 1050
WUXGA Wide Ultra eXtended Graphics Array 16:10 1920 1200
WQXGA Wide Quad eXtended Graphics Array 16:10 2560 1600
WQUXGA Wide Quad Ultra eXtended Graphics Array 16:10 3840 2400

end of procedures such as photic stimulation and hyperventila- Montage reformatting allows the electroencephalographer
tion. The system should support entry of comments by the to mentally reconstruct the three-dimensional EEG by combin-
technologist, both as stored “codes” for common events (e.g., ing views of three or more montages, such as longitudinal bipo-
“eyes open,” “awake,” or “seizure”) and as free text. Comments lar, transverse bipolar, and referential montages.
can also be generated by digital analysis programs, such as spike Additional reference montages can be easily constructed by
and seizure detection algorithms. taking the average of data from two or more electrodes (25).
During review, the electroencephalographer should be able to Laplacian montages (source current density montages) are also
view all notations made during acquisition, as well as add com- easy to implement using “nearest neighbor” methods to
ments. A scrollable list of comments can allow the reviewer to approximate the laplacian (26). Montages can include elec-
immediately find an EEG segment of interest. A particularly use- trodes of the modified 10-10 International System, as well as
ful feature is the ability to create a “context-sensitive” comment, other physiologic monitors such as electro-oculogram, electro-
which stores both a text annotation and the montage, sensitivity, cardiogram, oxygen saturation, and electromyogram.
and filter settings at which the EEG was originally viewed. In addition to changing montages, digital EEG software usu-
ally allows the reviewer to optimize vertical resolution by select-
Montage Reformatting ing only certain channels or groups of channels for display or
Since digital EEG data are acquired using a hard-wired referen- “hiding” channels contaminated by artifact. Individual or
tial montage (input 2 for all channels is the machine reference, groups of channels can be moved or reordered to improve com-
or REF), the computer can perform calculations on the stored parisons between homologous brain regions. Extra space or
data to create any desired montage, nearly instantaneously. For “gaps” between channel groups can facilitate review, albeit at the
example, transforming the recorded montage to a bipolar mon- expense of vertical resolution. Using different colors (red vs.
tage requires the following calculations: blue) for left versus right hemisphere channels can help the elec-
troencephalographer quickly localize waveforms but may intro-
Channel Recording Calculations Display duce subtle visual illusions that can mislead the interpreter.
Montage Montage
Sensitivity Changes
1 Fp1-REF Fp1-REF – Fp1-F7
(F7-REF) In analog EEG, sensitivity is the input voltage ( V) divided by
the output pen deflection (mm). For example, a 50- V signal at
2 F7-REF F7-REF – F7-T3 a sensitivity of 7 V/mm would be 7.1 mm high. In digital EEG,
(T3-REF) the same waveform on the display may not actually measure 7.1
3 T3-REF T3-REF – T3-T5 mm. Rather, the amplitude scale is displayed as a vertical icon
(T5-REF) (calibration line) on the monitor, and the amplitude of EEG
4 T5-REF T5-REF – T5-O1 signals is determined relative to this reference amplitude. The
(O1-REF) scale can be changed to provide a variety of sensitivity settings.
Sensitivity changes can be performed in two ways with a
5 O1-REF digital EEG machine. Rarely, the technician can use a software
Chapter 7 ■ Digital EEG 131

sensitivity switch during EEG acquisition to change the actual channel. Others contain a calibration icon that shows the size of a
amplifier sensitivity (e.g., for intracranial vs. surface EEG 50- or 100- V signal. The calibration icon can be moved around
recordings). More commonly, the display sensitivity is changed the screen and compared to various waveforms. Most programs
during acquisition or review. To decrease the amplitude of the also contain a feature that allows a user to draw a line from peak
waveforms on the page, the software program plots each data to trough of any given waveform to get a numerical value for the
point on the monitor as if it were half as large. Sensitivity amplitude in microvolts, or the duration in milliseconds. Finally,
changes can be made for a single channel, groups of channels, or more advanced waveform measurements may include average
the entire page. amplitude or frequency for a selected segment of a channel of
EEG, or even a spectrogram of the component frequencies.
Time Scale Changes
Most EEG software programs display EEG at 10 seconds per Digital Filtering
page, or for larger or widescreen monitors, 25 to 35 mm per sec- One of the major advantages of digital EEG is the ability to
ond. The number of seconds displayed per page can be apply, remove, and reapply a variety of digital filters to enhance
adjusted, however, to optimize interpretation. For example, dis- or minimize activity in certain frequency bands, without alter-
playing 20 seconds or more per page will enhance slow activity ing the original EEG data. This is analogous to inserting electri-
and allow analysis of slow periodic complexes or prolonged cal filters in the acquisition path (see Chapter 6). The field of
events such as seizures. Spreading out the EEG to show only 2 digital time domain filters is vast and complex, but some basic
to 5 seconds per page will spread out faster frequencies, similar principles are important to illustrate (Table 7.3).
to increasing the paper speed on an analog machine. This The three common methods of digital filtering include two
allows more precise analysis of time relationships of signals in time domain methods (finite impulse response [FIR] and
adjacent channels, for example, to analyze the propagation of infinite impulse response [IIR]) and one frequency domain
epileptiform spikes. In addition, time cursors can usually be method using the fast Fourier transform. While most commer-
placed on the screen to measure time relationships exactly. cial systems do not incorporate all the filter choices discussed
Many software programs allow the number of seconds per page below, it is important to understand how different digital filter
to be varied from 1 to 100 seconds. types can be designed to mimic (or improve upon) the analog
Unfortunately, even the largest monitors cannot adequately filters discussed in Chapter 6. Signal processing programs and
display more than 20 to 30 seconds of EEG, limiting simultane- toolboxes used for quantitative EEG analysis (e.g., MatLab) will
ous analysis of long segments of EEG (e.g., evolution of seizures incorporate a larger number of filters and the ability to design
over several minutes). Printing the EEG to fan-fold paper may custom digital filters.
enhance the interpretation of long EEG epochs. Alternatively,
most EEG software programs allow split-screen views to allow Time Domain Filters
side-by-side comparison of noncontiguous segments of EEG. Processing discrete time sequences to accentuate or diminish
frequency bands is made possible by a technique called digital
Waveform Measurements filtering. We begin with an extremely simple example, the
Waveforms can be measured in several ways. Some software dis- two-point moving average. If one takes a sequence and
plays the maximal “peak-to-peak” amplitudes available for each replaces each value by the average of it and the previous value,

Tabl e 7 . 3

Time Domain Digital Filters

Filter Type Pros Cons


FIR Absolutely stable Computationally more expensive
Linear phase, constant group delay Require higher order for equivalent
(peaks and other features of wave performance
shapes will have the same delay Difficult to reproduce effect of analog
in two channels processed by the filters
same filter)
IIR Computationally cheaper Potentially unstable (stable in typical
practice)
Can obtain good performance with Nonlinear phase, group delay is
low order frequency dependent
Can simply and exactly reproduce
analog filters
132 Part I ■ Basic Principles

one generates a new sequence. If xk is the input at time t k and the phase delay (in radians and degrees) as a function of (reduced)
yk is the output at t k, then frequency. Note also that the phase shift is a straight line (linear).
1 This simple algorithm (average of 2 points) acts as a filter that
yk 2 (xk xk 1)
attenuates higher frequency inputs and preserves low frequencies
Figure 7.11 shows this concept applied to pure sinusoids of and is thus termed a low-pass filter. A four-point moving average
several different frequencies, up to the Nyquist limit. The squares would act similarly but would more dramatically attenuate high
(connected by solid curve) are the original data points. The cir- frequencies. A simple filter that would attenuate low frequencies
cles (connected by dashed curve) are the points that result from and preserve high frequencies would be a two-point difference fil-
the two-point moving average. We note several features: (i) For ter (computing the difference between the current input point
an input of a pure sinusoid, the amplitude of the output may be and the previous input point).
altered, but the frequency of the output is the same as the input. The graph of the magnitude of the output as a function of the
This is a general feature. (ii) The amplitude of the output dimin- input frequency is known as the transfer function. Several differ-
ishes with higher frequency. This is specific to our example. ent regions or bands in this transfer function have specific names.
Other filters might attenuate low frequencies and preserve high The frequency band in which the output is preserved (ratio of
frequencies. (iii) The output is shifted to the right. In particular, output to input near 1) is called the passband. The frequency
the peaks in the output are all shifted ½ point to the right relative band in which the output is severely attenuated (ratio near 0) is
to the input sinusoid. However, the apparent shift in phase termed the stopband. The frequency band inbetween is known as
increases with higher frequency. (iv) At the Nyquist frequency, the transition band. The slope of the transfer function in the tran-
the output is identically zero, for this example. sition band is called the roll-off. The graph of the phase shift ver-
Figure 7.11B shows graphs of these effects. The top shows the sus frequency is known as the phase plot. There is a delay in the
ratio of the output amplitude over the input amplitude, as a func- output, which is named the group delay and is generally expressed
tion of frequency (in this case, the absolute frequency divided by in number of sample points. The group delay may vary as a func-
the sampling frequency, as this effect does not depend on absolute tion of frequency. In general, the group delay at a given frequency
sampling rate, so 0.5 is the Nyquist frequency). The bottom shows is the (negative) slope of the phase plot at that frequency. The

A
100

B
0 1
Magnitude

−100
0 Seconds 1
100
0.5

−100 0
0 Seconds 1 0 Frequency 0.5
100

0 0 0°
Phase

−100
0 Seconds 1
100
−π/4 −45°

−100 −π/2 −90°


0 Seconds 1 0 Frequency 0.5

Figure 7.11 Filtering by FIR filter, two-point moving average. See text for explanation.
Chapter 7 ■ Digital EEG 133

order of a digital filter is the number of prior input values that are choices yield a low-pass filter. Note that these coefficients yield
used in the computation of the output. Thus, a two-point aver- a small passband and a cutoff near 0.05 (i.e., low frequencies
age is first order and a four-point moving average is third order. below 0.05 sampling rate are passed, but frequencies above
These are the principal features that are compared and contrasted 0.1 sampling rate are mostly attenuated).
between different digital filters. In this manner, given a set of filter coefficients, one can com-
pute the transfer function and phase plot. There are algorithms for
Finite Impulse Response. The examples described so far are part
generating the coefficients corresponding to traditional analog
of a class known as FIR filters. When the input consists of an
circuits (Butterworth, Chebyshev type 1 and 2, Bessel, etc.) with a
“impulse”—a single nonzero input value in a sequence of zero
given cutoff frequency. The inverse problem—having a desired
inputs—the output sequence (response) will be nonzero for
transfer function, and computing the required order and coeffi-
only a finite number of elements before it necessarily returns to
cients—is beyond the scope of this chapter. For details, see Ref. 27.
zero. This is a direct consequence of the fact that the output yk
In general, for IIR filters, phase shift is nonlinear, so group
depends only on current and prior inputs xk, xk – 1, … This fea-
delay varies with frequency. In fact, particular coefficients for
ture defines the FIR class of filters. FIR filters can be designed to
IIR filters can create a situation in which two finite sinusoidal
be low-pass, high-pass, band-pass, and many other forms. A
signals of different frequencies can appear reversed in order
particular feature of the FIR class of filters is that the phase shift
after being filtered, compared to the input sequence.
is a linear function of frequency, and thus the slope is constant,
Techniques to mitigate group delay for both FIR and IIR filters
so the group delay is the same at all frequencies.
are discussed below.
Infinite Impulse Response. The other class of digital filters is
known as IIR filters. They are also known by other names Undesired Effects
including autoregressive (AR) or recursive. A simple example is Group delay can produce unexpected distortions in digital sig-
1 4 nals, particularly for multichannel comparisons, as with EEG.
yk xk yk 1 Figure 7.13 shows the distorting effect of different filter settings
5 5 when visually comparing channels. Group delay can be prob-
That is, the output (yk) is computed as 1/5 the current input
lematic, and it affects both FIR and IIR filters. As mentioned
(xk) plus 4/5 the prior output.
previously, the group delay generally increases with increasing
Using prior output values is what gives this filter class its
order of the filter. Since FIR filters typically require significantly
important characteristics. For an input sequence that contains an
higher orders to obtain the same transfer function attenuation
impulse (a single nonzero value), this filter will yield an output
as IIR filters, group delay can quickly become very problematic
that continues to have nonzero outputs. Figure 7.12A shows such
for FIR filters. Techniques to eliminate it have been developed.
an IIR (though the values trend toward and approach zero).
Filters that are acausal can eliminate group delay. Acausal filters
This filter applied to another input sequence known as a step
are ones in which future time points as well as past time points
function is shown in Figure 7.12B. This plot will probably
are included in the computation, as opposed to the filters dis-
remind many readers of the graph of voltage charging a capac-
cussed previously that include only past time points, which are
itor. In fact, this filter is the digital analogy of a simple single
called causal. While causal filers can be used to filter live on a
resistor and capacitor circuit arranged as a low-pass filter.
point-by-point basis (but introducing the group-delay artifact),
Digital filters are specified by the coefficients that multiply
acausal filters cannot be implemented live. They can, however,
prior inputs and output to compute the current output. Given
be implemented with a short delay.
a set of coefficients, it is straightforward (though possibly com-
If the entire time series of points is known and immutable,
plicated) to determine the transfer function and phase plot. In
having been acquired and stored, then acausal filters are simple
general, the equation for a filter of order n follows:
to implement and can completely eliminate the group delay. In
yk 5 b0xk 0 b1xk 1 b2xk 2 p bnxk n6 fact, a simple scheme is to filter the entire time series in the for-
5a1yk 1 a2yk 2
p anyk n 6 ward direction, then reverse and use the same filter coefficients
in the backward direction. This eliminates the group delay and
The transfer function (H) and phase are generated by calcu- can even eliminate the phase distortion, achieving zero-phase
lating (or having a graphing program display) distortion. This forward–backward zero-phase filtering works
5b0 b1e i2 x
b2e 2i2 x p 6 for both FIR and IIR filters. A further trick can be employed to
H ` i2 x 2i2 x p 6` minimize transients at the end of the time series, producing the
51 a1e a2e
same result whether filtering forward first or backward first (28).
and
Frequency Domain Filters
1
Imag(H)
phase tan e f Frequency domain filters use FFT to remove unwanted fre-
Real(H) quencies from the signal (4,29). According to Fourier’s theo-
For our simple example, b0 = 0.2 and a1 = 0.8. With these rem, any time series waveform can be modeled as the sum of a
coefficients, the transfer function (solid) and phase (dashed, in set of sinusoidal waveforms, each with different frequency,
radians) are graphed in Figure 7.12C. It demonstrates that these amplitude, and phase. In frequency filtering, the input signal is
134 Part I ■ Basic Principles

A C
+1 1

Magnitude

0.5

0 0
0 10 0 Frequency 0.5

B
+1 0 0°

Phase

−π/4 −45°

0 −π/2 −90°
0 10 0 Frequency 0.5

Figure 7.12 Infinite impulse response (IIR) filter. A: Filter response to an impulse input. The open squares connected by
wide-dash line shows the input, which is zero everywhere but time point 0 where it is 1, and the filled squares connected
by narrow-dash line plots the output, which approaches but never reaches zero, having a persisting, infinite response to
an impulse input, thus the term “infinite impulse response.” B: IIR filter applied to another input sequence known as a
step function. Again, the open squares connected by wide-dash line shows the input, and the filled squares connected by
narrow-dash line plots the output. C: Transfer function and phase plot of IIR low-pass filter. See text for details.

broken into short segments and then transformed into its com- the signal will remain stationary during the entire epoch.
ponent sine wave signals using FFT. To filter the signal, the coef- Breaking the EEG into short epochs, however, may introduce dis-
ficients of the unwanted frequencies above or below a specified continuities between subsequent epochs. “Windowing” proce-
cutoff frequency are set to zero, and the inverse Fourier trans- dures can minimize such discontinuities. Windowing is the
form is then computed to obtain the original EEG signal minus application of a weighting function (e.g., Hamming window) to
the unwanted frequencies (Fig. 7.14). taper the ends of each epoch toward zero. Because windowing
Frequency domain filters have several limitations. Frequency necessarily loses some information at the ends of the epochs,
filtering cannot be performed until the full epoch to be filtered analysis is commonly performed on multiple epochs overlapping
has been acquired, precluding true real-time filtering. To improve each other by 25% to 75%. The length of the analyzed epoch, the
computational speed, certain limitations may be placed on the windowing function utilized, and the degree of overlap all influ-
time series, such as a requirement to contain exactly 2n members ence the computational speed of frequency domain filtering.
(where n is a positive integer). Finally, if the EEG signal is not sta-
tionary (i.e., has variable frequency components) over the epoch, Digital Analysis and Data Reduction
the FFT may introduce artifactual high-frequency noise (30). For Digital EEGs can be very rapidly reviewed by displaying pages
this reason, frequency domain filtering is typically performed at rates of 1 to 10 pages (10 seconds of EEG) per second. The
over short epochs (e.g., 1 second) to improve the likelihood that maximal speed of review is determined by the speed of the
Chapter 7 ■ Digital EEG 135

which can be performed in real time by the computer proces-


sor, while off-line analyses can be more complex. Digital pro-
cessing can highlight specific features of the EEG using
graphical displays, remove or correct artifacts, or detect com-
mon patterns such as seizures or sleep states.

Spike and Seizure Detection


Original signals Algorithms to detect spikes and seizures are commonly used in
long-term video-EEG monitoring, intensive care unit monitor-
ing, and ambulatory EEG. The algorithms are designed to detect
specific features in the digital data, such as the sharpness and
field of an epileptiform discharge or the change in frequencies or
amplitude at the beginning of a seizure (34,35). Other methods
use neural networks that can be trained and updated with a
database of seizures to improve detection accuracy (36,37). The
detection algorithms mark the sections of data which contain
events of interest. The electroencephalographer can then review
this prescreened data rather than the entire EEG recording. The
Delay
detection algorithms are not perfect, and may either miss
seizures or spikes, or have high numbers of false detections.
Filtered Topographic Display (Mapping)
Topographic maps display the EEG voltage at various locations on
the head (38). In order to construct smoothed maps, interpolation
Not filtered
techniques such as spline interpretation or nearest neighbor rules
are used (39,40). The maps are most accurate when large numbers
of electrodes are used. Topographic maps can be used to display
the voltage topography of epileptiform activity (41).

Dipole Source Modeling


0 Seconds 1.0
This technique aids in localizing the generators of EEG events,
such as epileptiform discharges (42,43). Multichannel digital
Figure 7.13 Distorting effect of different filter settings when visually EEG is modeled as being generated by a single (or a small num-
comparing channels. The top panel shows (synthesized) traces for two ber) dipole source. Each dipole source produces particular scalp
sequential channels from a bipolar derivation with an idealized sharp- voltage waveforms when placed in a mathematical model of the
and-slow wave complex, but with significant noise on the top channel. head. The locations and orientations of the dipoles are adjusted
It is tempting to try to eliminate the noise in that one channel by selec- to produce the best fit between the mathematical model and the
tively applying a low-pass filter. The bottom panel shows the result of actual EEG. Source localizations can be coregistered with data
an FIR low-pass (cutoff 25 Hz) filter, with order of 50 (a not unreason- from MRI.
able order for an FIR filter). The high-frequency noise is indeed
reduced, but a delay is induced by the filter. The (downgoing) peak of Spectral Analysis/Graphical Displays of Quantitative EEG
the top tracing is delayed by 1/ 8th of a second. All traces are 1-second Spectral analysis involves analyzing the various frequencies that
duration at 200 samples per second. make up the digital EEG signal (44). Typically, the fast Fourier
transform is used to calculate the frequencies present in the EEG.
The spectral analysis of a segment of EEG can then be plotted on
a topographic map or displayed as a graph of power in a partic-
network, computer processor, and graphics card. Even at very ular frequency versus time. This application is increasing in use
rapid review speeds, however, review of an entire 24-hour EEG for long-term EEG monitoring in the intensive care unit (45).
(8640 10-second pages) will take 15 to 60 minutes or more. A
variety of data reduction methods can be used to identify EEG
segments of interest for more rapid interpretation. DATA SHARING, STORAGE, AND
Digital EEG can be analyzed quantitatively to assist interpre- NETWORKING
tation, automatically detect events, and produce graphical dis-
plays (31–33). A full discussion of these techniques is beyond EEG File Formats and Data Sharing
the scope of this chapter, but they are included in later chapters. EEG files acquired with current commercial EEG systems are
Signal processing can be performed either on-line, as EEG is typically stored in proprietary formats that require vendor-
acquired, or off-line. On-line analyses are limited to those specific software to open and view the files. This limitation can
136 Part I ■ Basic Principles

Spectrum − power
Figure 7.14 Filtering in the fre- Original FFT
quency domain. The top left shows
the original signal (this example has
512 samples per second). The fast
(discrete) Fourier transformation 0
(FFT) converts this signal into a fre-
0 Hz 256
quency domain representation (top
right). The spectrum is multiplied by × (multiply by)
a desired filter function (middle
right), in this case preserving all fre- 0 Seconds 1
quencies between 2 and 32 Hz—a
bandpass—and zeroing all other fre-
quency components. The resulting Filtered
spectrum (bottom right) is then 0 Hz 256
inverse Fourier transformed back into
the time domain, yielding the filtered
signal (bottom left). The Fourier 0
Inverse
domain representation also has FFT
phase information—this has been
omitted for clarity of illustration.

0 Seconds 1
0 Hz 256

cause significant problems in transferring data between differ- EEGs can be archived on a variety of media, including CD,
ent EEG laboratories for review, and even for maintaining read- DVD, tape, hard drives (internal or external), servers, or network-
able archives as file formats are changed over time. There are attached storage devices. These media vary greatly in size
several approaches to overcome this Tower of Babel problem (amount of data which can be stored), cost, convenience (time
(46). Most commercial systems can export EEGs to a compact required to archive and ease of use), and durability. Use of digital
disk (CD) or digital video disk (DVD) with a limited-function media has significantly decreased space requirements and cost of
version of the EEG reader software, so the disk becomes a self- EEG storage. CDs (storing up to 700 MB) and DVDs (4.7 GB,
contained reader station usable on any computer. Others pro- dual layer 9 GB) are most commonly used when EEGs need to be
vide a “light” version of review software that can be installed on transferred to another electroencephalographer for interpreta-
multiple computers at low cost. Some allow EEGs to be con- tion. Archiving to removable media is low cost but inefficient, as
verted to open-source formats (e.g., European Data Format each disk holds only a small amount of data, must be “burned”
[EDF], European Data Format plus [EDF ] (47), Extensible individually, must be placed back in the computer for the data to
Biosignal [EBS] (48), ASCII (49), or the American Clinical be reviewed, and may become unreadable over years. External
Neurophysiology Society’s Technical Standard 1 (50)) that can tape drive storage devices have large capacity and rapid backup,
be read by a variety of freely available reader programs and but data retrieval can be slow because the tape must be trans-
some commercial systems. These open formats may not repli- ferred back to the review station hard drive before review. Tapes
cate all of the features available in the original EEG software, may also deteriorate over time. External hard drives are inexpen-
and most have been adopted by only a few vendors. Finally, sive and provide more rapid access to data than tape drives. Hard
there are several commercial programs that can read EEG files drive costs have decreased significantly over recent years, making
collected by different commercial EEG systems, but these may storage of large amounts of data on servers or network-attached
not read all EEG file types and can be quite expensive. storage devices economically feasible. EEG data stored on a server
can be rapidly retrieved from any computer on the network.
Data Storage
Maintaining digital EEGs archives (e.g., updating to new media
Once EEG is reviewed, video and EEG data are typically clipped or to new file formats) can be costly and time-consuming.
for events of interest and archived to long-term storage. The size of an EEG file depends on the number of channels
Archiving schemes vary; most labs save all routine EEGs, but recorded, sampling rate of the ADC, and the duration of the
only clips of significant events for long-term video-EEG moni- recording. One hour of 32-channel EEG sampled at 200 sam-
toring. Some save all continuous 24-hour EEG data, but contin- ples per second produces an approximately 50-MB file, while 1
uous video files are too large to store in a cost-effective manner. hour of video data can be nearly 1 GB, depending on resolution
A laboratory performing 2000 routine EEGs, 1000 video-EEG and compression. Compression techniques can be used to
studies, and 500 ambulatory EEGs will produce more than a reduce the number of bits necessary to represent information,
terabyte of digital data each year. by removing repeated bits. For example, instead of encoding 10
Chapter 7 ■ Digital EEG 137

Tabl e 7 . 4

Transmission Speed of Digital EEG and Video

EEG Video
Type Data Rate Transfer Time Real Time Transfer Time Real Time
Wired
Gigabit Ethernet 1 Gbps 0.5 sec Yes 5.3 sec Yes
T4 (fiberoptic 274.76 Mbps 1.7 sec Yes 19.2 sec Yes
trunk line)
Fast Ethernet 100 Mbps 4.8 sec Yes 52.8 sec Yes
Ethernet 10 Mbps 48 sec Yes 8 min 48 sec Yes
Cable modem 2.5–6 Mbps 1 min 20 sec– Yes 14 min 40 sec– Yes
3 min 12 sec 35 min 20 sec
T1 (trunk line) 1.54 Mbps 5 min 20 sec Yes 57 min 8 sec Yes
DSL 1–3 Mbps 2 min 40 sec–8 min Yes 29 min 20 sec– Maybe
88 min
Modem 56 kbps 2 hr 38 min No 26 hr 20 min No
Wireless
802.11n 100 Mbps 4.8 sec Yes 52.8 sec Yes
802.11a/g 54 Mbps 8.9 sec Yes 1 min 38 sec Yes
801.11b 11 Mbps 43.6 sec Yes 8 min Yes
4G cellular 6–100 Mbps 4.8 sec–1 min 20 sec Yes 52.8 sec– Yes
14 min 40 sec
3G cellular 144 kbps–2.4 Mbps 3 min 20 sec– Yes 36 min 20 sec– Maybe
55 min 33 sec 10 hrs 10 min

Digital EEG, 32 channels, 16-bit resolution, sampled at 256 Hz, 1 hour = 60 MB = 480 Mbit. MPEG4 color video, 640 pixels 480 pixels resolution, 30 frames per
second, compressed, 1 hour = 660 MB = 5280 Mbit.

binary 1 and 5 binary 0 values, the compression method width 11 to 100 Mbits/sec), although the latter does not incur
encodes that a binary 1 value is repeated 10 times and binary 0 high costs of running cable to all EEG locations and may be
value 5 times (111111111100000 vs. 10-1 5-0). The destination adequate for low-volume laboratories.
computer then decodes the information and expands it back to An efficient and organized network requires careful plan-
the original number of bits. While it is possible to compress dig- ning, usually in conjunction with trained and certified hospital
ital EEG signals slightly, in general the savings are small and network managers. The design of a network for EEG is deter-
may not justify computing overhead (decompressing data) in mined by a few basic principles: (i) function, (ii) geography
these systems. Digital video compression is essential, and this (number of users, distance of the farthest users from the
technology continues to evolve. server), (iii) speed or bandwidth, (iv) budget, (v) expandabil-
ity/flexibility, and (vi) requirements for network administra-
Networking tion. The first step is to identify the purpose(s) of the network,
One of the most useful features of digital EEG is the ability to such as (i) data acquisition, (ii) transmission of data to central
connect acquisition machines to computer networks for data review area, (iii) provision of access to data from multiple loca-
transmission and communication. A network is a group of tions, sometimes simultaneously, (iv) interpretation of studies,
computers and other devices that are connected together to (v) generation of reports, and (vi) data archiving.
facilitate the transfer of information and sharing of resources. A server, a computer specialized for storage, transmission,
The type of network and its bandwidth, or speed, will deter- and security of data, can provide a central repository of files,
mine how quickly EEG studies can be accessed and reviewed most importantly EEG and video files, EEG databases, and EEG
(Table 7.4). Wired networks are typically faster (100 to 1000 reports, which can then be accessed from multiple network
Mbits/sec) and more reliable than wireless networks (band- locations. The EEG can be streamed continuously to the server
138 Part I ■ Basic Principles

as it is acquired, or transferred after the study is completed. any device with Internet access, such as a home computer,
Central location of the data facilitates security measures and laptop, wireless personal digital assistant, or digital cellular
improves the efficiency of data backups and archiving. The telephone. Cellular wireless systems are slower than cable or
server is typically located locally in the EEG laboratory or in the DSL connections but may be adequate to quickly review
hospital’s server farm. emergency studies.
Networks used in hospitals usually consist of a long length of Remote access allows users to connect to the network and
very high bandwidth cable media that connects the servers, perform tasks as if they were directly connected to the net-
called the backbone, with branches off the backbone called seg- work, even when they are far away. Remote access can be
ments (Fig. 7.15). Segments (such as to EEG labs) usually con- accomplished via a dial-up connection to a remote access
sist of lower bandwidth cable media, which again branch to server or via a virtual private network (VPN) tunneled
individual users. EEG machines connected to the network can through the Internet. The VPN tunnel maintains the confiden-
be accessed remotely to review ongoing studies, update patient tiality of the data through encryption. Transmission of EEG
databases, and adjust recording parameters (e.g., montage, data to remote locations may be very slow (see Table 7.4). A
camera position). The bandwidth (or speed) of the network terminal server system such as Windows Terminal Server or
will determine the speed of remote review. Citrix Metaframe allows multiple remote users to run applica-
Finally, the need to review data from several different sites tions from anywhere with an Internet connection, nearly iden-
(remote EEG labs, physicians’ offices, home, etc.) may tical to when sitting in front of a computer on the network.
require additional resources, such as remote access servers, The remote user logs in and is authenticated, then can open
as well as Internet service providers outside the hospital or the EEG software application. The terminal server sends
health care facility. Users can access the network using nearly remote keystroke and mouse movements to the application

Research Home Remote Site

Remote
Access
Server
Internet Service Provider
Backup /
Storage

To Remote Sites on Hospital


Backbone: Pediatric Server(s)
EMU, etc.
Backbone

Epilepsy EEG
OR ICU Wada, PET Collectors Collectors

Reading
Room

Fellows

Attending

Figure 7.15 A complex EEG network connected to the hospital backbone. EEG data are collected from patients in vari-
ous locations and sent to a central server. The backbone provides for high-speed transmission of files, so the review loca-
tion can be quite distant from the collection locations. The network also provides remote access for users outside the
hospital. The hospital server can provide a backup for the EEG server.
Chapter 7 ■ Digital EEG 139

and sends screenshots of the application to the remote user. information by directly viewing unsecured workstations, or
Because only screenshots, not the actual EEG data, are trans- obtaining passwords written on the stations themselves, key-
mitted, review is typically much faster. stroke capture malware, or social engineering (requesting
An information technology specialist or network adminis- passwords from authorized personnel by using the pretense of
trator is often helpful for troubleshooting of computer and being an Information Technology employee). Some strategies
software problems and to provide network security and disaster applicable to digital EEG to mitigate these risks include:
protection/recovery. Network security is especially important if Computers on which EEG data are stored must be password
patient information is stored in the network, as law protects protected. Automatic logins, in which a login password is
patient confidentiality. The security architecture must be com- bypassed on computer startup, should be disabled. Each user
pliant with the Health Insurance Portability and Accounting should have an individual password. Vendors of EEG acquisi-
Act (HIPAA) of 1996 guidelines. Adequate security measures tion and reading stations should work toward the goal of
must protect against unauthorized users, computer viruses, avoiding the practice of a common username and password
accidental destruction of files, and other forms of attack. for all technicians and physicians using the stations. Ideally,
Sensitive files may need to be encrypted both on the computer each user should have individual account, with shared config-
disk and during network transmission. Networks connected to uration settings placed in shared folders and personalized
the Internet or other outside connections are especially vulner- preferences stored in individual accounts. Antivirus and anti-
able to outside attack and need a firewall or proxy to create a malware software should be installed and kept current on
barrier around the internal network. Protecting data from cat- each workstation to mitigate the possibility of keystroke
astrophic data loss includes several considerations: (i) power capture of passwords.
backup with surge protectors or universal power supplies, (ii)
data backup on a regular and reliable schedule, (iii) mainte- Storing
nance of backup files in a secure location separate from the Digital EEG involves storing/archiving vast amounts of data, and
original files, (iv) disk fault tolerance by providing multiple while PHI is again a small minority, risks remain. Laptops con-
copies of the same information on a array of hard drives, and taining digital EEG could be stolen. Portable hard drives could
(v) server fault tolerance by providing multiple servers in case likewise be stolen. Digital EEG records could be recovered from
of server malfunction. local cached files when accessed from computers outside the hos-
pital. Strategies to minimize these risks include: Laptops and
Security desktops could be stored in locked rooms when not in use.
HIPAA (http://www.hhs.gov/ocr/hipaa/) instituted a set of Laptops could be secured by “locking mechanisms” to fixed walls.
rules (the “Security Standard”) that impact digital EEG storage, Laptops (and even desktops) could have full hard disk drive
review, transmission, and archiving. The Department of Health encryption. Without the disk encryption password, the plaintext
and Human Services has published a guideline document information would not be recoverable. Of course, this leads to
(http://www.cms.hhs.gov/SecurityStandard/Downloads/Securi additional passwords, so a clear password policy and password
tyGuidanceforRemoteUseFinal122806.pdf) that provides a management system becomes even more necessary. Prevention of
concise description of many of the issues that relate to HIPAA cached records on outside computers is more difficult and would
rules on network transmission and storage of protected health likely involve software implementation by vendors.
information (PHI) and which can be applied directly to digital
EEG. We review some general issues and recommendations and Transmitting
describe some of the specifics of the HHS guideline. This is probably the aspect of security with digital EEG that
One goal intended by HIPAA was protection of patient pri- requires the most attention. There is no doubt that remote
vacy and prevention of abuse of patient information. While access (physicians at off-sites, home, conferences, etc.) and dis-
the vast majority of data in digital EEG files are the digitized tant second opinions could add much value to patient care.
voltage time series and are fairly useless to identity thieves, However, remote access then requires careful consideration of
some portions of the files (depending on the vendor) can con- protection from data security breaches. Electronic PHI could be
tain electronic protected health information (ePHI), such as intercepted when in network transit, either outside hospital
name, date of birth, address, doctor, and diagnoses. These network (external), or even within (internal). Strategies to min-
pieces of information could be used to harm patients if imize the risk of network interception include: Digital EEG data
obtained by individuals with ill intent. Strategies for protec- transmitted over networks should be encrypted. For external
tion of this information must be addressed by the modern network access, most sites use VPN protocols. Ideally, auto-
digital EEG laboratory. mated network analysis by the hospital IT staff should prompt
The HHS guideline groups electronic processing into three alerts for unusual traffic, such as frequent logins, simultaneous
areas: accessing, storing, and transmitting. logins from disparate locations, logins from other continents,
and so on Web-based (HTTP) protocols are also in some use.
Accessing For these, at a minimum HTTPS (HTTP-secure) should be
With regard to digital EEG, an example relating to accessing used. For HTTPS, modern browsers use TLS (transport layer
might be the login accounts of acquisition and reviewing security, which superseded the older SSL/secure sockets layer
workstations. Unauthorized persons might access patient protocol) for bidirectional encryption.
140 Part I ■ Basic Principles

CONCLUSIONS 11. Horowitz P, Hill W. The Art of Electronics. 2nd ed. Cambridge,
England: Cambridge University Press; 1989.
Rapid advances in digital technology have greatly improved the 12. Nuwer MR, Comi G, Emerson R, et al. IFCN standards for digital
efficiency and reliability of clinical EEG recordings and offer recording of clinical EEG. The International Federation of Clinical
new possibilities for research applications. New electrode sys- Neurophysiology. Electroencephalogr Clin Neurophysiol Suppl.
tems that use dry electrodes or require limited skin preparation 1999;52:11–14.
may allow recordings to be initiated easily in nontraditional 13. Blum DE. Computer-based electroencephalography: technical
basics, basis for new applications, and potential pitfalls.
locations, such as the emergency room, ambulances, or even
Electroencephalogr Clin Neurophysiol. 1998;106(2):118–126.
patients’ homes. EEG data could then be transmitted wirelessly 14. Gilliam F, Kuzniecky R, Faught E. Ambulatory EEG monitoring. J
to central locations for access and review by electroencephalog- Clin Neurophysiol. 1999;16(2):111–115.
raphers. Small, light, wireless amplifiers may allow recording of 15. Budinger TF. Biomonitoring with wireless communications. Annu
ambulatory patients over weeks to months, analogous to car- Rev Biomed Eng. 2003;5:383–412.
diac monitoring with implantable loop recorders. Such data 16. Guideline twelve: guidelines for long-term monitoring for
could help to elucidate patterns of seizure occurrence or the epilepsy. Am J Electroneurodiagnostic Technol. 2008;48(4):265–286.
development of epileptiform activity or seizures after poten- 17. Ives JR. Video recording during long-term EEG monitoring of
tially epileptogenic brain injuries. Current network and remote epileptic patients. Adv Neurol. 1987;46:1–11.
access solutions allow rapid EEG review from any location, 18. Burgess RC. Design and evolution of a system for long-term elec-
making “real-time” monitoring of brain activity (e.g., for criti- troencephalographic and video monitoring of epilepsy patients.
Methods. 2001;25(2):231–248.
cally ill neurology patients) an achievable goal. Decreased cost
19. Risk WS. Viewing speed and frequency resolution in digital EEG.
and increased capacity of storage media allows collection and Electroencephalogr Clin Neurophysiol. 1993;87(6):347–353.
archiving of vast amounts of EEG data. Modern ADCs can sam- 20. Bresenham J. Algorithm for computer control of a digital plotter.
ple at thousands of times per second from hundreds of chan- IBM Systems J. 1965;4(1):25–30.
nels, facilitating collection of multichannel EEG from 21. Epstein CM. Aliasing in the visual EEG: a potential pitfall of
microelectrodes to reveal pathophysiological mechanisms of video display technology. Clin Neurophysiol. 2003;114(10):
epilepsy and other neuronal activity at the single neuron or 1974–1976.
localized network level. Finally, EEG data can be analyzed by an 22. Swartz BE. The advantages of digital over analog recording
ever-increasing array of signal analysis techniques, coregistered techniques. Electroencephalogr Clin Neurophysiol. 1998;106(2):
with imaging procedures for source localization or used to cre- 113–117.
ate statistical databases of normal and abnormal findings. 23. Levy SR, Berg AT, Testa FM, et al. Comparison of digital and conven-
tional EEG interpretation. J Clin Neurophysiol. 1998;15(6):476–480.
These techniques are described in detail in later chapters.
24. Guideline fourteen: guidelines for recording clinical EEG on
digital media. American Electroencephalographic Society. J Clin
REFERENCES Neurophysiol. 1994;11(1):114–115.
25. Jayakar P, Duchowny MS, Resnick TJ, et al. Localization of epilep-
1. Berger H. Uber das Elektrenkephalogramm des Menschen IV. togenic foci using a simple reference-subtraction montage to doc-
(English translation by P. Gloor: On the Electroencephalogram of ument small interchannel time differences. J Clin Neurophysiol.
Man, Electroenceph Clin Neurophysiol 1969, Suppl 28). Arch 1991;8(2):212–215.
Psychiat Nervenheil. 1929;97:6–26. 26. Klein SA. Inverting a Laplacian topography map. Brain Topogr.
2. Burks AW, Burks AR. The ENIAC: first general-purpose electronic 1993;6(1):79–82.
computer. 1981. MD Comput. 1995;12(3):206–212. 27. Oppenheim AV, Shafer P, Buck JR. Discrete-Time Signal Processing.
3. Farley B, Clark W, Gilmore J, et al. Computer techniques for the study 2nd ed. Englewood, NJ: Prentice-Hall, Inc.; 1998.
of patterns in the electroencephalogram: Lincoln Laboratories, MIT, 28. Gustafsson F. Determining the initial states in forward-backward
1957. Contract No. 337. filtering. IEEE Trans Signal Process. 1996;44(4):988–992.
4. Cooley JW, Tukey JW. An algorithm for the machine calculation of 29. Brigham EO. The fast fourier transform and its applications. In:
complex Fourier series. Math Comput. 1965;19:297–301. Oppenheim AV, ed. Englewood Cliffs, NJ: Prentice Hall; 1988.
5. Technology and equipment review. Digital electroencephalo- 30. Cook EW III, Miller GA. Digital filtering: background and tutorial
graphs. J Clin Neurophysiol. 1993;10(3):378–392. for psychophysiologists. Psychophysiology. 1992;29(3):350–367.
6. Guideline Five: Guidelines for Standard Electrode Position 31. Colditz PB, Burke CJ, Celka P. Digital processing of EEG signals.
Nomenclature. American Clinical Neurophysiology Society; 2006 IEEE Eng Med Biol Mag. 2001;20(5):21–22.
[updated February 10, 2006]. Available from: https://www.acns.org/. 32. Duffy FH, Hughes JR, Miranda F, et al. Status of quantitative EEG
7. Schomer DL. Ambulatory EEG telemetry: how good is it? J Clin (QEEG) in clinical practice, 1994. Clin Electroencephalogr. 1994;
Neurophysiol. 2006;23(4):294–305. 25(4):VI–XXII.
8. Van Drongelen W. Signal Processing for Neuroscientists. Burlington, 33. Nuwer MR, Lehmann D, da Silva FL, et al. IFCN guidelines for topo-
MA: Academic Press; 2007. graphic and frequency analysis of EEGs and EPs. The International
9. Guideline Eight: Guidelines for Recording Clinical EEG on Digital Federation of Clinical Neurophysiology. Electroencephalogr Clin
Media. American Clinical Neurophysiology Society; 2006 [updated Neurophysiol Suppl. 1999;52:15–20.
Februray 10, 2006]. Available from: https://www.acns.org/. 34. Gotman J. Automatic detection of seizures and spikes. J Clin
10. Vanhatalo S, Voipio J, Kaila K. Full-band EEG (FbEEG): an emerg- Neurophysiol. 1999;16(2):130–140.
ing standard in electroencephalography. Clin Neurophysiol. 2005; 35. Qu H, Gotman J. A seizure warning system for long-term epilepsy
116(1):1–8. monitoring. Neurology. 1995;45(12):2250–2254.
Chapter 7 ■ Digital EEG 141

36. Wilson SB, Turner CA, Emerson RG, et al. Spike detection II: auto- 44. Alarcon G, Binnie CD, Elwes RD, et al. Power spectrum and
matic, perception-based detection and clustering. Clin Neurophysiol. intracranial EEG patterns at seizure onset in partial epilepsy.
1999;110(3):404–411. Electroencephalogr Clin Neurophysiol. 1995;94(5):326–337.
37. Wilson SB. A neural network method for automatic and incre- 45. Scheuer ML, Wilson SB. Data analysis for continuous EEG monitor-
mental learning applied to patient-dependent seizure detection. ing in the ICU: seeing the forest and the trees. J Clin Neurophysiol.
Clin Neurophysiol. 2005;116(8):1785–1795. 2004;21(5):353–378.
38. Nuwer MR. The development of EEG brain mapping. J Clin 46. Rapoport DM, Ayappa I, Norman RG, et al. NPSG data inter-
Neurophysiol. 1990;7(4):459–471. change-dealing with the Tower of Babel. Sleep. 2006;29(5):599–600.
39. Perrin F, Pernier J, Bertrand O, et al. Spherical splines for scalp 47. Kemp B, Olivan J. European data format ‘plus’ (EDF ), an EDF
potential and current density mapping. Electroencephalogr Clin alike standard format for the exchange of physiological data. Clin
Neurophysiol. 1989;72(2):184–187. Neurophysiol. 2003;114(9):1755–1761.
40. Soong AC, Lind JC, Shaw GR, et al. Systematic comparisons of 48. Hellmann G, Kuhn M, Prosch M, et al. Extensible biosignal (EBS) file
interpolation techniques in topographic brain mapping. format: simple method for EEG data exchange. Electroencephalogr
Electroencephalogr Clin Neurophysiol. 1993;87(4):185–195. Clin Neurophysiol. 1996;99(5):426–431.
41. Ebersole JS, Wade PB. Spike voltage topography identifies two types 49. Designation AS. E1467-92 specification for transferring digital
of frontotemporal epileptic foci. Neurology. 1991;41(9):1425–1433. neurophysiological data between independent computer systems.
42. Gotman J. Noninvasive methods for evaluating the localization and Available from: ASTM, 1916 Race St, Philadelphia, PA 19103. 1992.
propagation of epileptic activity. Epilepsia. 2003;44(suppl 12):21–29. 50. Guideline TS1: Standard for transferring digital neurophysiological
43. Scherg M, Ebersole JS. Brain source imaging of focal and multifo- data between independent computer systems. J Clin Neurophysiol.
cal epileptiform EEG activity. Neurophysiol Clin. 1994;24(1):51–60. 2008.
CHAPTER

Polarity and Field Determinations


BRUCE J. FISCH 8
INTRODUCTION cortex. To perform the latter requires an understanding of how
the EEG signal arises and is conducted to the scalp. The relation-
Accurate EEG interpretation is not possible without a thorough ship between the scalp signal and its origin within the brain is a
understanding of EEG scalp localization. In contrast to most complex topic that has been approached by a variety of mathe-
other aspects of EEG interpretation, the process of scalp locali- matical models (and is covered in greater detail in other chapters
zation is deductive and does not present the problem of reason- of this book). In routine practice localization is approximated
able disagreement between readers. according to a basic understanding of cortical anatomy and
The development of digital EEG recording has made it pos- volume conduction theory.
sible to view the same segment of recording retrospectively The spontaneous EEG signal in the routine scalp recorded
using different frequency filters and spatial filters (montages). EEG arises from cortical pyramidal cell postsynaptic potentials.
This retrospective analysis greatly increases the ability of the Although other sources (e.g., other neurons and glial cells) and
electroencephalographer to localize the electrical fields of the forms of electrical potentials (e.g., action potentials) within the
scalp EEG and thereby provide more accurate interpretation. It cortex exist, their contributions to extracranial electrical fields
is not an overstatement to say that it has changed the practice of appear to be very limited. Pyramidal cells that generate postsy-
electroencephalography. naptic potentials have the biophysical advantage of firing in
There are several reasons why localizing EEG activity is an synchrony (i.e., they summate in time) with similar radial ori-
essential skill. First, the identification and classification of EEG entation (i.e., they are aimed in the same direction toward the
activity is always based to some degree on localization. For cortical surface) and long duration, making them ideal for pro-
example, a high amplitude, sharply contoured, anterior tempo- ducing a large enough signal to be conducted through the skull
ral waveform that clearly stands out from the background and and recordable at the scalp. As illustrated in Figure 8.1, this usu-
is activated by sleep has a high likelihood of being an abnormal ally means that the recording electrode that detects the highest
epileptiform spike. However, the same waveform appearing amplitude signal is overlying the cortical source (1). However,
over the central head region during sleep is far more likely to be there are contributions from the walls of cortical sulci that are
a benign vertex wave of normal sleep. Second, localization is not oriented perpendicular to the scalp and may affect adjacent
often the only feature that allows the electroencephalographer electrode positions. More notable exceptions to this simple spa-
to determine if an activity is cerebral in origin or an artifact. A tial relationship can occur, particularly if:
sharp rhythmic pattern that evolves in frequency and morphol-
ogy over a left frontal head region may be likely an electro- (1) the source is in a fissure
graphic seizure, whereas the same pattern that appears (2) the source is primarily in sulci
simultaneously over both the left frontal head region and the (3) cortical anatomy and orientation have been altered (most
right occipital head region without involvement of electrodes commonly in cerebral palsy)
between these regions is far more likely to be an artifact. Third, (4) skull thickness has been altered (there is relative thinning as
the clinical correlation portion of EEG interpretation always in early infancy, thickening as in certain bone diseases, or
depends on the cortical localization of the EEG findings. Lastly, gap in the skull from prior trauma or surgery).
localization is the one skill that greatly increases the likelihood The way in which electrical current is transmitted throughout
of correctly interpreting an EEG pattern the reader has never the body is referred to as volume conductor theory. Figure 8.1
encountered before. illustrates an example of volume conductor theory applied to
EEG in which the outer layers of the cortex are producing a
THE EEG SIGNAL: CORTICALLY GENERATED momentary signal that is negatively charged. In the example, the
SCALP CURRENTS EEG source involves an entire gyrus. Because the pyramidal cell
postsynaptic potentials generate a circuit of current flow
The skill of localization in routine clinical EEG interpretation throughout the length of each cell, one end of the pyramidal cell
consists of two steps: (1) localizing EEG activity to specific always has the opposite electrical polarity from the other (in Fig.
electrode positions on the scalp and then (2) relating the scalp 8.1 the outer cortex is negative and the inner cortex is positive).
localization to the likely source in the underlying cerebral In Figure 8.1, the addition of all polarities oriented toward the

143
144 Part I ■ Basic Principles

Figure 8.2 This figure illustrates the steps outlined in the text
(A through F) for measuring the head for the placement of the 10-20
system. (From Fisch BJ. Fisch and Spehlmann’s Digital and Analog EEG
Primer. Amsterdam: Elsevier; 1999.)

Figure 8.1 This figure illustrates volume conductor theory for a cortical determined by changing currents arising from underlying
source that occupies a gyrus. The distribution of the amplitude of the pyramidal cell postsynaptic potentials. Electrode placement is
voltage that reaches the scalp surface is shown in line S. The current that used to accurately map the contours of the changing voltages
reaches the electrode from the cortical source is equivalent to the area over the scalp surface. Each electrode, therefore, represents a
contained by the lines that begin at the cortex and converge on the elec- spatial voltage sample point. The more electrodes used, the
trodes P1 or P2. For P1, the gyral convexity source projects only negativ- more faithfully they will map the actual contours of the scalp
ity. For P2, the source projects both negativity from the outer cortex and potentials. If too few electrodes are used, then the presence or
positivity from the inner cortex of the sulcus (shown by the area between absence of EEG changes in some areas of the scalp may be
the dashed lines that converge on P2). The cancellation effect of the inner missed. This is somewhat analogous to digital sampling of EEG
cortex and the outer cortex (negativity added to positivity; the area within or audio signals. If too few samples are taken, then high fre-
the dashed lines subtracted from that within the solid lines converging on quency waveforms cannot be recorded. If too few electrodes are
P2) reduces the voltage of the signal detected by P2 as shown in line S. used, then voltage changes over areas in between will be com-
Although the figure is drawn in two dimensions, it is intended to repre- pletely missed. A certain number of electrodes is necessary to
sent three-dimensional space with the lines leading from the cortex to avoid spatial undersampling.
electrodes representing the sides of an approximately conical space. The Routine EEG recording uses 21 scalp electrode positions.
amount of current reaching P1 and P2 then depends on the solid angle These are placed according to the international 10-20 system of
(signified by the omega characters in the figure) subtending the volume. electrode placement described in Figure 8.2 (2). Although this
(From Gloor P. 1985 Neuronal generators and the problem of localization is sufficient for routine clinical recording, to accurately sample
in electroencephalography: application of volume conductor theory to the scalp surface for the detection of small signals generated by
electroencephalography. J Clin Neurophysiol. 2:327–354.) evoked potential recordings, or for research in quantitative EEG
analysis and intracranial localization, it is estimated that at least
100 electrodes would be needed (3).
recording electrode determines the final voltage at the scalp Spatial sampling in neonates also follows the 10-20 system,
(negative in the outer cortex pointing toward the recording elec- but because of the reduced head size fewer electrodes are used.
trodes, and positive in the deeper cortex pointing toward the F7, F3, F8, F4, P7, P3, P8, and P4 are excluded. Fp1 and Fp2 are
recording electrodes). replaced by more posterior electrodes, Fp3 located midway
The scalp EEG can be imagined as a constantly changing between Fp1 and F3 and Fp4 located midway between Fp2 and
relief map in which the map is composed of hills and valleys F4, consistent with the relatively more posterior location of the
whose height or depth represents a particular voltage that is frontal lobes in relation to skull measurements at that age (4). If
Chapter 8 ■ Polarity and Field Determinations 145

Once that measurement is obtained, Fp1 is placed at 5% of


the total distance on the circumferential line to the left of
the midline. Then marks are placed at intervals of 10% of
the total distance around the head with the last mark at the
Fp2 position. These marks establish the positions for F7,
T7, P7, O1, O2, P8, T8, F8, and Fp2.
(4) The front to back distance from Fp1 and Fp2 running
through C3 and C4 to O1 and O2 respectively, is measured
on each side. At half the distance between Fp1 and C3 on
the left, and Fp2 and C4 on the right, F3 and F4 positions
are marked, respectively. Similarly, the midpoints between
C3 and O1 on the left, and C4 and O2 on the right give the
positions for P3 and P4.
(5) Next, the measurements are taken from F7 to F8 through Fz
to define the transverse coordinates for F3 at the midpoint
between F3 and Fz, and for F4 half way between Fz and F8.
Similarly, measurements from T5 to T6 through Pz locate
the transverse coordinates for P3 midway between T5 and
Pz and for P4 midway between Pz and T6.
Figure 8.3 The 10% system of electrode placement is shown, with T7, If an electrode cannot be placed (due to scalp defects, bandages,
T8, P7, and P8 replacing the formerly named T3, T4, T5, and T6 elec- or monitors), then the nearest available electrode site of the
trodes to maintain naming consistency. The added electrodes at 10% “10-10” combinatorial system should be used, and the corre-
distances are named according to the surrounding 10-20 electrodes. sponding electrode on the other side of the head should be
Note that in recording an anterior temporal source that FT9 and FT10 moved to the same mirror image position.
placement is usually superior to F7 and F8, which are located over the It should be kept in mind by the electroencephalographer
posterior lateral frontal lobes. that there is no substitute for the careful approach to measure
the head outlined above. Unfortunately, it is also the procedure
in which short cuts are taken by some EEG technologists. With
A1 and A2 cannot be applied to the earlobe then the mastoid
the advent of digital EEG and video, it is reasonable to request
position is used (5).
that the technologist include the electrode measurement and
As illustrated in Figures 8.2 and 8.3, each recording electrode
application process in the video recording of each EEG as a
is named with a letter indicating the head region and a sub-
quality control measure.
script number indicating the side of the head (odd numbers are
In some clinical situations (e.g., presurgical epilepsy moni-
left and even right) or z for the midline. The further the elec-
toring), better localization may require using more than 21
trodes are placed laterally from the midline, the higher the sub-
electrodes. In such cases, selected additional electrodes are
script number (e.g., P7 is lateral to P3). Measurements of the
placed according to the 10-10 system described in Figure 8.3.
head are made with a measuring tape (calipers or other meth-
Physiologic electrical currents are detected using differential
ods should never be used). Measurements are all based on four
amplifiers. Each amplifier has two electrode inputs, named input
landmarks: the nasion (lower forehead between the eyes), the
terminal 1 and input terminal 2. The amplifiers subtract the volt-
inion (bone protruberance in the midline back of the head),
age of the input 2 electrode from the input 1 electrode (input 1 –
and the left and right preauricular point (immediately anterior
input 2 = recorded EEG signal), hence differential amplification.
to the auditory canal). Measurements are made sequentially as
Differential amplification is used for the recording of all bioelec-
shown in Figure 8.2 and summarized as follows:
trical signals (ECG, EMG, EEG). The advantage of differential
(1) The midline distance between the inion and nasion is amplifiers is that they cancel external noise. The most common
measured. Beginning at 10% of the total distance above the sources of noise arise from other electrical devices. In environ-
inion (or nasion), a scalp mark is placed with a wax pencil. ments where current is provided at 60 or 50 cycles/sec, the electri-
Then three more marks are placed at intervals of 20% of cal interference nearly equally contaminates both input 1 and 2
the total nasion to inion distance to establish the positions electrode wires with 60 or 50 cycle signals. Because the differential
for Fz, Cz, and Pz. amplifier subtracts the input 2 signal from the input 1 signal, the
(2) The coronal distance between the left and right preauricular electrical interference that is equal in both inputs cancels out and
points intersecting with Cz is measured. Points at 20% and is removed from the signal. However, because differential amplifi-
40% of the total distance are then placed from Cz on the left cation is used, the absolute voltage value at electrodes attached to
and right to mark the positions for C3, C4, T7, and T8. either input 1 or 2 is never known, only the difference between
(3) The distance around the head that passes through T7, T8, them is known. Fortunately, by comparing a number of electrode
and the points 10% above the inion and nasion is measured positions, the EEG can give a good approximation of the activity
by placing the measuring tape around the patient’s head. at a particular scalp location relative to other locations.
146 Part I ■ Basic Principles

DIFFERENTIAL AMPLIFIER POLARITY signal deflection? Which of the following is correct? (A) +10
and downward, (B) +10 and upward, (C) 150 and downward,
As noted above and illustrated in Figure 8.1, the currents flow- (D) 150 and upward, and (E) +/ 0. The answer is located at
ing through the scalp have a polarity that is either positive or the very end of the chapter. Do not look until you have tried to
negative. The comparison of electrodes by differential amplifi- answer the question on your own.
cation yields a difference in voltage and assigns the output a
polarity (negative or positive). As described above, the signal SCALP CURRENT FIELD LOCALIZATION:
output and its polarity are achieved by subtracting the voltages
between input 1 and 2. The voltage is displayed on the EEG
SPATIAL FILTERING
monitor or recording paper by the distance the signal deviates Scalp potential fields overlap and vary in size and location.
from the zero baseline. The polarity is displayed according to Widely distributed fields are often higher in amplitude than
the direction the waveform deflects (above or below the base- fields that occupy a small area of the scalp. When viewed simul-
line). If the voltage in the electrode in input 1 is 50 V and taneously, smaller fields are frequently obscured by or lost in the
the voltage in the electrode in input 2 is +50 V, then the EEG higher amplitude widespread fields. However, there is a way to
signal will be input 1 minus input 2, or in this case ( 50) spatially filter out the more widespread fields so that the more
(+50) = 100 V. In addition to knowing that input 1 is sub- localized ones can be easily seen. The use of spatial filters is
tracted from input 2, the electroencephalographer has to extremely important because many EEG signals, such as epilep-
understand how the amplifier orients the direction of the wave- tiform spikes, often occupy small scalp areas.
forms upward or downward to indicate the relative polarity of The electrodes in inputs 1 and 2 of each line of amplified
the signal. EEG signal channel displayed are referred to as the electrode der-
As shown in Figure 8.4, the convention for biologic recording ivation. Derivations are written with the name of the electrode
is that, if input 1 is more negative than input 2, the output signal in input 1 followed by the name of the electrode in input 2, sep-
will deflect upward, whereas if input 1 is more positive than arated by a minus sign. Fp1-F3 is an example of a derivation. All
input 2, then the output signal will deflect downward. This is the derivations are also mathematical expressions. In the case of
same as stating that if input 2 is more positive than input 1, then Fp1-F3, the output of the differential amplifier displayed on the
the signal will deflect upward, and if input 2 is more negative monitor screen will be the voltage detected at Fp1 minus the
than input 1, then the signal will deflect downward. Also, if the voltage detected at F3 or Fp1-F3.
polarity and voltage in the input 1 electrode is the same as in the A derivation is referred to as bipolar if it consists of adjacent
input 2 electrode, then the output signal will be a flat line (zero). electrodes on the scalp. Fp1-F3 is therefore a bipolar derivation
At this point it is recommended that the reader test their because Fp1 and F3 are located next to each other. In contrast
understanding of the information presented in Figure 8.4 by to bipolar derivations, those with longer interelectrode dis-
answering the following question: tances are referred to as referential. An example of a typical ref-
A potential of 70 V in input 1 and 80 V in input 2 of erential derivation is Fp1-A1.
an EEG amplifier yields what voltage and what direction of A display that consists of a combination of derivations is
referred to as a montage. Montages that use long interelec-
trode distances are reference montages, whereas those that
use bipolar derivations are referred to as bipolar montages.
The use of bipolar versus reference montages allows the elec-
troencephalographer to visualize localized versus widespread
scalp fields.
The concept of spatial filtering is illustrated in Figure 8.5.
The bipolar derivation (closely spaced electrodes) on the left
side of Figure 8.5 shows the electrode in input 1 immediately
overlying a cortical generator represented by the black oval area,
and the input 2 electrode is outside the area of the black cortical
generator. Both electrodes are within the field of the larger
underlying cortical generator represented by the gray oval area.
As input 2 is subtracted from input 1 the activity in the black
area is accurately represented as the difference between the two
Figure 8.4 Biological amplifiers use differential amplification in which electrodes (black dashed line waveform at the amplifier out-
the voltage in the electrode plugged into input 2 is subtracted from the put). In contrast, the larger gray field produces a similar voltage
voltage in the electrode(s) plugged into input 1. In EEG, EMG, and at both input 1 and 2 electrodes in the bipolar derivation and is
evoked potential recording, by convention if the relative voltage differ- therefore filtered out by cancellation.
ence is negative the signal will deflect upward and if positive, down- The referential derivation on the right side of Figure 8.5 has a
ward. However, as shown in the figure, the voltage source in the brain much larger interelectrode distance. The input 1 electrode is
can produce opposite deflections depending on whether the input 1 or within the field of both the small black cortical area and the larger
input 2 electrode is closest to the scalp source. gray cortical area, whereas the input 2 electrode is outside of both
Chapter 8 ■ Polarity and Field Determinations 147

Figure 8.6 A longitudinal bipolar montage records a left hemisphere


Figure 8.5 This figure illustrates the filtering effects of interelectrode
seizure that is not visible. (From Fisch BJ, Padin-Rosado J. EEG local-
distance on the recording of underlying EEG scalp potential fields. The
ization and interpretation. In: Fisch BJ, ed. Epilepsy and Intensive Care
closer spaced bipolar electrodes (left) filter out the more widespread
Monitoring. New York, NY: Demos Medical Publishing; 2010:57–76.)
field (gray cortical area) because both inputs see the same voltage. The
more localized field (black cortical area) is clearly displayed. The more
widely spaced referential electrodes display a combination of both fields recording. A widespread left frontal seizure is not visible in the
in which the more localized (black cortical area) is obscured by the longitudinal bipolar montage (Fig. 8.6), but becomes easily visi-
larger (gray cortical area) field but the larger field is now seen (see also ble in the transverse bipolar montage (Fig. 8.7) and in the refer-
the discussion in the text). ential montage (Fig. 8.8). The input 2 reference of the referential
montage consists of an electrode placed at the top of the ster-
num and an electrode placed over the C6 vertebral protruber-
fields. This means that it will amplify a summation of both fields. ance and is referred to as a noncephalic neck-chest reference. It
The advantage is that it “sees” the signal from the larger gray cor- has the advantage of eliminating reference contamination by the
tical generator that is eliminated from the bipolar recording. The scalp, but is sometimes overwhelmed by the ECG signal. This
disadvantage is that by combining the two generators, the local- illustration also makes the important point that for some
ized black field is obscured in the larger signal produced by the patients it is not sufficient to review the record using only one
gray field. The spatial filtering effect shown in Figure 8.5 explains montage or important electrographic events can be missed.
how widespread fields can be removed from the recording. It also
illustrates that overall EEG amplitude increases with increasing
interelectrode distances. This tends to occur with distances up to
about 10 cm. This is also the reason why EEGs performed for the
determination of electrocerebral inactivity (brain death) are
recorded with long interelectrode distances.
Early in the history of EEG, clinical schools developed that
strongly stressed the exclusive use of either bipolar or referential
recording. Neither is superior to the other, they are simply viewed
as different spatial filters that emphasize different aspects of the
scalp topography of EEG voltage fields.
Montages are constructed in either a linear anterior to pos-
terior or transverse pattern across the scalp to simplify interpre-
tation. Bipolar montages are constructed in overlapping chains
(e.g., Fp1-F3 followed by F3-C3) in either an anterior to poste-
rior direction (longitudinal bipolar montages) or in a transverse
left to right direction (transverse bipolar montages). Referential
montages are usually displayed with electrodes in successive
channels in an anterior to posterior direction, with the same
reference electrode (or electrodes) in input 2 of every channel.
Longitudinal and transverse bipolar montages are both used
because scalp voltage fields can be asymmetric along either ante- Figure 8.7 The same recorded epoch as in Figure 8.6 using a trans-
rior to posterior or transverse axis. Such asymmetries can only verse bipolar montage now clearly reveals the presence of an electro-
be reliably assessed by either using both a longitudinal and a graphic seizure most easily seen in the fifth channel (F3-Fz). (From
transverse bipolar montage, or by using one bipolar montage Fisch BJ. EEG localization and interpretation. In: Fisch BJ, ed. Epilepsy
and reference montage. This is dramatically illustrated in Figures and Intensive Care Monitoring. New York, NY: Demos Medical
8.6 through 8.8 (6). Each figure shows the same epoch of EEG Publishing; 2010:57–76.)
148 Part I ■ Basic Principles

Figure 8.8 The same recorded epoch as in Figures 8.6


and 8.7 using a noncephalic neck–chest electrode pair
reference reveals the widespread seizure with maximal
amplitude epileptiform spikes at F3 and very little activ-
ity at Fz. (From Fisch BJ. EEG localization and interpre-
tation. In: Fisch BJ, ed. Epilepsy and Intensive Care
Monitoring. New York, NY: Demos Medical Publishing;
2010:57–76.)

SELECTING SPATIAL FILTERS: trodes O1 and O2, not because there are eye movement poten-
MONTAGE DESIGN tials in O1 and O2, but because the activity is actually coming
from the reference electrodes that include the activity detected
There are five commonly used montage designs and each has a by Fp1 and Fp2. Therefore, it is important to be aware that
unique spatial filtering characteristic. The montages that can pro- waveforms that appear upside down in distant head regions in
duce the severest filtering of widespread fields and the greatest an average reference montage are likely to be due to reference
enhancement of highly localized fields are the longitudinal bipo- contamination. However, the electrodes with the highest ampli-
lar and transverse bipolar montages (described earlier). These tude are almost always closest to the true source of the activity.
montages are followed in order of greatest to least in filtering of The common reference montage consists of the same one or
widespread fields as follows: the Laplacian montage, the average two electrodes in input 2 in all channels of the montage. The
reference montage, and the common reference montage. common reference montage produces the least attenuation of
The Laplacian montage is implemented as a local average ref- widespread fields, particularly when the reference electrodes are
erence montage in which the electrode in input 1 is referred to not placed on the head (noncephalic neck-chest reference). A
a combination of the immediately surrounding electrodes in widely used reference montage uses the A1 and A2 ear reference
input 2, for example, Cz ¼ (Fz + C3 + Pz + C4). Each chan- electrodes. All left-sided electrodes are often referenced to A1
nel of the montage has a different reference combination in with all right-sided electrodes referenced to A2. A1 and A2 refer-
input 2 that depends on the location of the electrode in input 1. ence electrodes are particularly useful during sleep when high
The Laplacian montage compliments the bipolar when identi- amplitude activity is occurring over the central head regions that
fying highly localized fields with low amplitude, such as a focal would otherwise contaminate the reference electrodes with
electrographic seizure onset. A reduced filtering effect that pro- excessive activity. In contrast, during wakefulness, a common
duces less suppression of regional fields can be achieved by a reference montage using Cz is often used because central activity
Laplacian montage that incorporates all electrodes on the scalp is generally low in amplitude and the relative absence of muscle
except the input 1 electrode into the reference. This is done by at the vertex makes it less likely that Cz will be contaminated
weighting the contributions of the reference electrodes accord- with muscle artifact. The advantage of the common reference
ing to their distances from the input 1 electrode (the farther montage, particularly the noncephalic neck-chest reference is
away the input 2 electrode is from the input 1 electrode, the less that it produces a simple view of the scalp field, where the input
it contributes to the combined voltage of the input 2 elec- 1 electrode whose channel has the highest amplitude identifies
trodes). This montage is not commercially available but can be the scalp location with the field maxima. As will be seen in the
implemented on many systems using a spreadsheet (7). following section, this works well unless the reference electrode
The average reference montage consists of a single electrode is within the field of interest (reference contamination).
in input 1 referred to a combination of all or most of the
remaining scalp electrodes. The average reference montage is LOCALIZING CURRENT FIELDS: PHASE
good for viewing highly localized medium to high amplitude CHANGES AND DIFFERENTIAL
fields and somewhat more widespread regional fields. When a AMPLIFICATION
high amplitude or very widespread activity occurs, the average
reference itself will contain that activity, sometimes producing Localization using bipolar montages is accomplished by analyzing
a confusing picture. For example, eye movement waveforms phase orientation (the upward or downward deflection of the
that typically have a voltage field maximum at Fp1 and Fp2 will waveform) and amplitude. Localization using reference mon-
often appear upside down simultaneously in the occipital elec- tages is accomplished mainly by analyzing amplitude. Bipolar
Chapter 8 ■ Polarity and Field Determinations 149

Figure 8.9 Instrumental phase reversal. Figure 8.10 Instrumental phase reversal with cancellation in an inter-
vening channel.

localization depends on changes in phase between channels. Phase Figure 8.9 illustrates the concept of instrumental phase reversal
refers to the upward or downward direction of a waveform. A because it is the instrument (i.e., the way the electrodes are
phase reversal has occurred if the phase has changed direction arranged with the input 2 electrode becoming the input 1 elec-
from up to down or from down to up. Figure 8.9 shows an exam- trode in the next channel) that causes the phase reversal. In con-
ple of a phase reversal between channels 1 (derivation Fp1-F3) trast, a true phase reversal would mean that two different polarities
and 2 (derivation F3-C3). are simultaneously present in adjacent cortical areas. The occur-
The rules for localization in bipolar montages are summa- rence of a true phase reversal of cortical polarity would produce
rized in Table 8.1. Examples of these rules are illustrated in two instrumental phase reversals in a linear chain of bipolar elec-
Figures 8.9 through 8.12. The electrodes in these figures are trodes. In the event that there is no phase reversal in a bipolar
shown as black circles and the lines leading from them plug into chain (i.e., all the waveforms point in the same direction), then the
input 1 (the higher line going into the derivation) or input 2 field maxima is closest to the electrode at the end of the chain that
(the lower line going into the derivation). They are arranged as shows the most rapid change in voltage (highest amplitude) in
a longitudinal bipolar montage. successive channels. This is referred to as the end of chain phe-
Figure 8.9 shows an instrumental phase reversal between nomenon. It is localizing to the electrode at the end of the elec-
channels with the derivations Fp1-F3 and F3-C3. The field over trode chain and does not suggest the source is deep in the brain.
the scalp is displayed by the dashed lines with the highest voltage Figure 8.10 illustrates an instrumental phase reversal with
not covered by an electrode. The field potential is negative. cancellation in an intervening channel. The field on the scalp
Therefore, the channel 1 deflection is downward (input 1 is more has a negative polarity with maximal voltage in the center of the
positive than input 2) whereas the deflection in channel 2 is dashed lines. The first channel deflection is downward because
upward (input 1 is more negative than input 2) (see Fig. 8.4). The input 1 is more positive than input 2. The second channel is flat
third channel deflection is also upward because input 1 is more because the input 1 and 2 electrodes, F3 and C3, see the same
negative than input 2. The last channel shows no deflection voltage. The third channel deflects upward and is reversed from
because both electrodes, P3 and O1, are the same. The electrode the first channel because input 1 is more negative than input 2.
common to both channels that reverse phase localizes the maximum The intervening channel with cancellation (F3-C3) localizes the
field potential to that common electrode. In this case that is F3. field maximum between the electrodes in that channel.

Tabl e 8 . 1

Rules of Localization: Bipolar Montages

The electrode with the highest voltage potential is found by


1. Instrumental phase reversal
2. Instrumental phase reversal with cancellation in an
intervening derivation
3. End of chain phenomenon or the absence of phase
reversal
4. End of chain phenomenon with cancellation
5. True phase reversal (double phase reversal)
Figure 8.11 End of chain phenomena with cancellation.
150 Part I ■ Basic Principles

Figure 8.12 Ipsilateral ear reference with the negative field maximum
closest to F3.

Figure 8.11 illustrates the end of chain phenomena with can-


cellation. Channel 1 electrodes detect the highest voltage, but
because it is the same in both electrodes the output is zero, a flat
line. As in Figure 8.10 this localizes the field maximum between
those two electrodes (Fp1 and F3) at the end of the bipolar Figure 8.13 Tangential dipole with cancellation in bipolar derivations
chain. The next channel shows an upward deflection because and true phase reversal in common reference derivations; anterior max-
input 1 is more negative than input 2. The same applies to the imal negativity with posterior positivity. (From Fisch BJ, Padin-Rosado
third channel. The fourth channel is flat because there is no J. EEG localization and interpretation. In: Fisch BJ, ed. Epilepsy and
voltage difference between those electrodes. Intensive Care Monitoring. New York, NY: Demos Medical Publishing;
Without the accompanying head figure it is reasonable to ask 2010:57–76.)
why the changes in this montage are not interpreted as a posi-
tive field in the back of the head instead of negative field in the
front of the head. While that is possible it is unlikely. The rate of input 1 electrode closest to the field maxima. If all channels
change in voltage (i.e., the amplitude of the waveform) between containing a reference show the same signal output then the
two electrodes is usually greatest near the field maxima and activity is coming from the reference, not the input 1 electrode.
least at a distance from it. In this case, the presence of a higher Figure 8.12 illustrates a reference recording of a negative
amplitude in channel 2 than in channel 3 localizes the signal field in which the reference electrode, A1, is outside of the scalp
source (field maxima) to the front of the head. field. In this case, the derivation with the highest amplitude sig-
A true phase reversal is shown in Figure 8.13. True phase nal shows the input 1 electrode that is closest to the field max-
reversals are rarely encountered except in situations where the ima (F3). The deflections are all upward because input 1 is more
cortical anatomy has been disturbed (e.g., cerebral palsy) or more negative than input 2.
often as an identifying feature of benign childhood epilepsy with Figure 8.14 illustrates reference contamination. In this
centromidtemporal spikes (BCECTS). In these patients the example, the reference electrode A1 is within the positive scalp
epileptiform spike arises at an angle that is parallel to the cortical field. The other electrodes are outside of the field. Therefore, the
surface, allowing both the negativity from the outer cortex and deflection seen in all channels is the same and is upward
positivity from the deeper cortex to be seen simultaneously. In because for each channel, input 1 is more negative than input 2.
BCECTS the positivity projects anteriorly, for example, appearing
as an upward deflection in Fp1-F3 and downward in F3-C3. The
negativity appears more posteriorly and in this example would
appear as a downward deflection in C3-P3 and upward in P3-O1.
The anterior positivity is typically lower in voltage than the pos-
terior negativity and may require increasing the gain or the use of
a neck-chest reference to be visualized.
As noted above, common reference montages are most use-
ful for identifying widespread scalp fields and least useful for
identifying highly localized low amplitude fields. Although
localization is usually simple with a reference montage, a con-
fusing picture arises when the reference electrode(s) are within
the field of the activity of interest. In that case, the activity in a
channel may be coming from the reference instead of the input Figure 8.14 Ipsilateral ear reference with reference contamination.
1 electrode. If the reference is not in the scalp field of interest, The signal is from the reference electrode as the other electrodes are
then the channel with the highest amplitude identifies the outside of the scalp field.
Chapter 8 ■ Polarity and Field Determinations 151

EEG INSTRUCTION IN LOCALIZATION


Those who teach electroencephalography should be aware that
many, if not most, physicians who read EEGs do not under-
stand how to localize EEG findings. It is a disservice to patients
with neurologic disorders when teachers fail to emphasize the
importance of localization and practicing physicians continue
to believe that EEG interpretation is only a matter of pattern
recognition. Although it is not possible to reach physicians who
have completed their training and are already out in practice, it
is possible to improve the skills of those currently being trained.
When training residents and fellows in EEG localization it is
Figure 8.15 Reference contamination.
helpful to ask certain questions that assess their progress. First,
what is the polarity of an upward deflection in a single channel of
Figure 8.15 illustrates how confusing the picture can become recording? Most students initially believe that an upward deflec-
when the reference is contaminated with activity also detected tion in a channel is negative. Of course, the correct answer is that
by the input 1 electrodes. Channel 1 deflects downward because there is no way of knowing. In a single channel one never knows
the reference is relatively negative (input 1 is more positive than if an upward deflection is caused by a negative potential in input
input 2). Channel 2 is flat because input 1 and input 2 see the 1 or a positive potential in input 2. Any misconceptions can usu-
same negative voltage. Channel 3 deflects upward because input ally be further resolved by a discussion of positive occipital sharp
1 is more negative than input 2, and channel 4 deflects down- transients of sleep (POSTS), because they always deflect upwards
ward because input 1 is more positive than input 2. in the longitudinal bipolar montages. Second, ask, is a phase
A field with a single polarity will not produce waveforms of reversal an abnormal finding? It is a surprising how often stu-
opposite phase in a referential recording between adjacent dents believe phase reversals are abnormal. This occurs because
channels unless the reference is contaminated. Two phase rever- the student never understood the importance of studying local-
sals as shown in Figure 8.15 (channel 1 vs. channel 3 and chan- ization and the teacher invoked the concept of phase reversal only
nel 3 vs. channel 4) virtually always assure the reader that in the setting of EEG abnormalities. This situation can be quickly
reference electrode contamination is occurring. A single-phase resolved by discussing phase reversals of vertex waves (or other
reversal in a common reference recording without reference normal waveforms) in bipolar montages. Third, ask, what is the
contamination, as shown in the lower three channels in Figure difference between an instrumental phase reversal and a true
8.13, represents a true (not instrumental) phase reversal, in that phase reversal? If the student can correctly answer these basic
it indicates the presence of a horizontal (tangential) cortical questions then they are well on their way to understanding EEG
dipole. localization. Lastly, never allow students to speak in terms of head
An example of how to approach localization is shown in regions when localizing EEG findings. Instead insist that they
Figure 8.13. In the initial longitudinal bipolar recording speak in terms of actual scalp spatial sampling points by using
epileptiform spikes are present in only one channel, creating a electrode names. (The answer to the question in the section on
complex problem in localization. The problem is quickly differential amplification and polarity is A).
solved by using a reference montage. At the time of EEG inter-
pretation new derivations were selected using a distant refer- REFERENCES
ence electrode unlikely to be contaminated by the field of the
1. Gloor P. Neuronal generators and the problem of localization in
epileptiform activity. In this case the reader selected the most electroencephalography: application of volume conductor theory
distant electrode, P8 (opposite side of the head, lateral and to electroencephalography. J Clin Neurophysiol. 1985;2:327–354.
posterior) for the reference to explore the activity in Fp1, F7, 2. Fisch BJ. Fisch and Spehlmann’s Digital and Analog EEG Primer.
and T7. The added derivations are in the last three channels in Amsterdam: Elsevier; 1999.
Figure 8.13. They show that there are similar amplitude spikes 3. Srinivasan R, Tucker DM, Murias M. Estimating the spatial
in Fp1 and F7. This explains why the first channel is relatively Nyquist of the human EEG. Behav Res Methods Instrum Comput.
flat (very low amplitude upward deflections are present), there 1998;30:8–19.
is cancellation between Fp1 and F7 (if input 1 and 2 are nearly 4. American Electroencephalographic Society Guidelines in EEG,
equal, the output is nearly zero). T7-P8 demonstrates a phase 1–7 (Revised 1985). J Clin Neurophysiol. 1986:3131–3168.
reversal with positivity arising from T7. If opposite polarities 5. Park KL, Nordli DR. Special considerations in neonatal monitor-
ing. In: Fisch BJ, ed. Epilepsy and Intensive Care Monitoring. New
are subtracted as shown in the left lower part of Figure 8.4,
York, NY: Demos Medical Publishing; 2010:91–103.
then the resulting signal will summate to create a higher 6. Fisch BJ. EEG localization and interpretation. In: Fisch BJ, ed.
amplitude signal than that in either input alone. This effect Epilepsy and Intensive Care Monitoring. New York, NY: Demos
creates the high amplitude spikes in channel 2 (F7-T7). T7 Medical Publishing; 2010:57–76.
and P7 share a similar amplitude positivity and therefore can- 7. Lemos ML, Fisch BJ. The weighted average montage. Electroenceph
cel out in the third channel. The positivity of P7 in relation to Clin Neurophysiol. 1991;79:361–370.
O1 can barely be seen in the fourth channel P7-O1.
Part II Normal EEG
CHAPTER
Normal EEG and Sleep:
Preterm and Term Neonates
ELI M. MIZRAHI, SOLOMON L. MOSHÉ, AND RICHARD A. HRACHOVY
9
INTRODUCTION TECHNICAL CONSIDERATIONS
The interpretation of the neonatal electroencephalogram con- The general principles of recording the electroencephalogram
sists of related phases of analysis most of which are conducted in neonates are similar to those of older children and adults
in parallel. Initially, the EEG is assessed to determine the con- with some important additions and exceptions. Guidelines for
ceptional age (CA) of the infant. In concert with this, the back- the recording of the neonatal EEG have been established by the
ground activity is assessed to determine whether it is normal, American Clinical Neurophysiology Society (13) and the
and if not, to characterize the abnormal features. In addition, International Federation of Clinical Neurophysiology (14).
the determination of CA provides the basis for the understand- Critical to the recording of neonatal EEG is a well-trained
ing of the range of both diffuse and focal abnormalities that staff of electroneurodiagnostic technologists (ENDTs) with
may be anticipated within each age-dependent epoch. Thus, expertise in the recording of newborn and young infants. To
visual analysis of the neonatal EEG begins with the recognition insure maximum clinical relevance, the ENDT should obtain
of the CA-dependent features characteristic of specific epochs basic information about each neonate to be recorded including
of development. This is followed by inspection for EEG abnor- standard demographic data, description of the recording envi-
malities based on an understanding of possible age-dependent ronment, documentation of reason for referral, details of the
findings. Taken together these aspects of visual analysis of the medical history, a list and timing of medications, and the
neonatal EEG provide the basis for the determination of CA of specifics of the infant’s general medical condition. In addition,
the infant and the assessment of the degree to which the EEG is the technologist must consider the state of each infant and
normal. determine the best method to make the infant as comfortable as
Two additional principles of interpretation of the neonatal possible in order to obtain a complete recording. This may
EEG relate to age-dependent changes over time and to find- require having the infant fed, having diapers changed, adjusting
ings of indeterminate significance. Typically, the EEG-deter- room temperature and, often, just prolonging the recording
mined age of the infant is given in CA—considered the age until the infant becomes comforted.
from the time of conception. The gestational age (GA) is the The International 10-20 System of Electrode Placement has
time from conception to the time of birth. The chronologic or been modified for recording neonates. Typically, a minimum of
legal age is the time from birth to the time of the EEG. The CA nine scalp positions are utilized (F1, F2, C3, C4, CZ, T3, T4, O1,
is the GA plus the legal age. A basic principle of interpretation and O2). In addition, electrodes are placed at A1 and A2 and a
of the neonatal EEG is that the immature brain develops at the ground electrode is placed either at mid-forehead or on a mas-
same rate whether in utero or following delivery. Thus, the toid region. Since digital recordings are fundamentally referen-
EEG recorded several weeks after an infant was born prema- tial, an additional reference electrode position may be needed
turely would have similar features to an EEG recorded on the (typically noncephalic), although some instruments provide a
first day of life of infant whose CA equaled the GA plus the so-called “internal” reference. The F1 and F2 electrode posi-
legal age of the premature infant. Another important consid- tions are 20% of the inion–nasion distance above the nasion
eration is that the clinical significance of all features of the and 10% of the circumferential measurement from the midline.
neonatal EEG has not been determined. Thus, the challenge is Their placement allows for the foreshortened position of the
to differentiate the known from the unknown and focus inter- neonatal immature frontal lobes. Placement of all of the stan-
pretation on those aspects of the EEG for which accurate data dard electrodes of the International System would result in such
are available. close spacing on the neonate’s scalp that electrode recording
A number of investigators have, over the years, contributed may overlap with redundant electrical fields. Paste, not collo-
to our understanding of the normal neonatal EEG beginning dion, is utilized for electrode placement.
with the pioneering work of the French school (1–5). More Polygraphic measures are integral to the recording of the
recent discussions of normal neonatal electroencephalography EEG in order to assist in characterizing sleep states, eye move-
include those by Hrachovy et al. (6), Watanabe et al. (7), Clancy ments, muscle contractions, cardiac rhythms, and respiratory
et al. (8), Mizrahi et al. (9), Plouin et al. (10), Vecchierini et al. patterns and to assist in identification of noncephalic artifact.
(11), and André et al. (12). The polygraphic measures include the electrooculogram

153
154 Part II ■ Normal EEG

(EOG), submental electromyogram (EMG), electrocardiogram a neonatal EEG. The record should be annotated when behav-
(ECG), and respiratory monitors such as a strain gauge, bipolar ioral or autonomic changes occur or when other events happen
electrodes, or pneumograph. that may affect the record.
The recording of the neonatal EEG includes the documenta- A final technical consideration relates to the duration of the
tion of wake/sleep cycles, the characterization of reactivity of neonatal EEG recording. There are two considerations: the time
the record to stimulation and identification of age-dependent it may take for the infant to experience the full cycles of sleep
waveforms and other features. These are best achieved utilizing and wakefulness, and a sufficient period of time for the infant
a single, bipolar montage with broad coverage over the scalp. to experience clinical or electrical events. The typical minimum
Digital recording of the neonatal EEG provides the opportunity duration of recording for a neonatal EEG is 1 hour. However,
to examine various waveforms with different montages after the recording duration is an interactive process based on clini-
recordings are complete. cal circumstances and the unfolding EEG.
Instrument settings used at the onset of recording are listed
in Table 9.1. The filter settings and sensitivity of the EEG chan- AGE-DEPENDENT DEVELOPMENTAL
nels should be the same as that for EOG in order to allow accu- FEATURES
rate comparison of waveforms to differentiate cerebral activity
from that of ocular origin. Paper speed is set at 30 mm/sec for Initial analysis includes the assessment of the degree of conti-
analog recordings or 10 sec/screen or “page” for digital record- nuity of background activity and the degree of interhemi-
ings. These paper speed settings are utilized in many laborato- spheric synchrony of the background activity. Further analysis
ries in the United States. However, several laboratories, consists of inspection for specific age-dependent waveforms
particularly those with ties to the French School of Recording, and patterns (so-called “grapho-elements”) (Table 9.2) and the
utilize a slow speed: 15 mm/sec or 20 sec/“page.” presence and character of sleep/wake cycles. In addition, there
The most successful and clinically relevant neonatal EEG is recognition of some special waveforms and patterns that
recordings are those in which objectives and strategies are iden- occur in the near-term or term infant that are considered devel-
tified prior to the beginning of each study. Advance planning is opmental milestones and variations of the normal EEG.
essential. The basic tasks are to obtain historical data, determine
reason for referral, initiate technical recordings, examine the Continuity
EEG in real time, observe the infant for clinical behaviors, The earliest appearance of electrical activity on the EEG is char-
record the infant in sleep and wakefulness, and attempt to pro- acterized by a pattern of discontinuity, with long periods of qui-
voke abnormal paroxysmal clinical events. Close observation of escence (Fig. 9.1). This pattern may be present in all states of
the infant at all times is particularly important when recording waking and sleep depending on CA and has been referred to as

Tabl e 9 . 1

Initial Instrument Settings for Recording Neonatal EEG

EEG channels
Sensitivity 7 V/mm
Time constant 0.3 sec
High-frequency filter 70 Hz
Notch filter (60 Hz, 50 Hz) Off
Display time
Paper speed 30 mm/sec*
Screen display 10 sec/“page”*
EOG** EMG*** Respir.*** ECG***
Polygraphic channels
Sensitivity 7 V/mm 7 V/mm 7 V/mm 300 V/mm
Time constant 0.3 sec 0.01 sec 0.3 sec 0.3 sec
High-frequency filter 70 Hz 70 Hz 70 Hz 70 Hz
Notch filter (60 Hz, 50 Hz) Off Off Off Off

*Some laboratories may choose to record at “half paper speed” with paper speed setting of 15 mm/sec or screen display of 20 sec/“page.”
**EOG settings remain the same as EEG settings for comparison of simultaneously recorded waveforms.
***EMG, Respir. (pneumograph), and ECG settings may be adjusted to optimize display.
Tabl e 9 . 2

Age-Dependent Features of the Neonatal EEG

CA (weeks) Continuity Synchrony Landmarks Wake/Sleep Cycles Reactivity


24–26 Discontinuous Asynchronous Beta–delta complexes may Limited behavioral or No reaction to stimulation
(trace discontinu) be present, although polygraphic changes
absence is not abnormal No cyclical EEG changes
27–28 Discontinuous Asynchronous Beta–delta complexes first Limited behavioral or No reaction to stimulation
Interburst intervals Traditionally appearing in the central polygraphic changes
are CA dependent characterized regions No cyclical EEG changes
and less than as an epoch of Temporal theta bursts
preceding epoch hypersynchrony (rudimentary)
29–30 Discontinuous Asynchronous Beta–delta complexes in Tendency for greater continuity No reaction to stimulation
Interburst intervals Greater than 50% the central regions of background activity
are CA dependent activity Temporal theta bursts during behavioral REM sleep
and less than asynchronous on (most consistent during
preceding epoch the two sides this epoch)
Some brief periods of
relative continuity
31–33 Discontinuity during Asynchronous Beta–delta complexes more Continuity during REM sleep No reaction to stimulation
wakefulness Degree of synchrony prominent in temporal Discontinuous during
Continuity during greater than and occipital regions wakefulness and NREM sleep
behavioral sleep previous epoch Temporal theta bursts
until 32 weeks CA
Temporal alpha bursts
replace theta bursts
at 33 weeks CA
34–35 Discontinuity during Asynchronous Frontal sharp transients EEG and physiologic features Background activity is
NREM, less than the Degree of synchrony (enchoches frontales) of wakefulness, REM, and reactive to stimulation
previous epoch greater than Beta–delta complexes in NREM sleep clearly defined —state dependent
Continuity during previous epoch occipital-temporal region,
wakefulness most often in NREM sleep
and REM sleep
36–37 Continuity during Residual asynchrony Frontal sharp transients persist EEG and physiologic features Background activity is
wakefulness and Degree of synchrony Beta–delta complexes of wakefulness, REM, and reactive to stimulation
REM sleep about 80% on the become less frequent NREM sleep clearly defined —state dependent
Episodes of discontinuity two sides Bifrontal delta may be present
during NREM sleep
38–40 Continuity in all states Synchronous on the Frontal sharp transients persist EEG and physiologic features Background activity is
two sides by 40 Beta–delta complexes of wakefulness, REM, and reactive to stimulation
weeks CA disappear from NREM sleep NREM sleep clearly defined —state dependent
Appearance of trace alternant
41–44 Continuity in all states Synchronous on Frontal sharp transients persist EEG and physiologic features Background activity is
the two sides No other immature of wakefulness, REM, and reactive to stimulation
waveforms are present NREM sleep clearly defined —state dependent
Trace alternant begins to
resolve by 44 weeks CA

155
156 Part II ■ Normal EEG

Figure 9.1 26 to 27 weeks CA. This EEG


demonstrates a trace discontinu pattern with a
burst of bilateral polyfrequency activity. The
burst is somewhat asynchronous and has fea-
tures of beta–delta complexes. (Reprinted with
permission from Mizrahi EM, Hrachovy RA,
Kellaway P. Atlas of Neonatal Electroencephalo-
graphy. 3rd ed. Philadelphia PA: Lippincott
Williams & Wilkins; 2004.)

trace discontinu. As the CA increases, periods of inactivity At approximately 30 weeks CA, continuous activity first
shorten. The identification of interburst intervals (IBIs) is pri- appears, but is state dependent: present only during behavioral
marily based on the degree of attenuation between bursts, rapid eye movement (REM) sleep. At about 34 weeks CA, the
although there is no real consensus among investigators regard- EEG also becomes continuous in the apparent awake state. By
ing a definition of the maximum amplitude of the IBI, ranging 37 to 38 weeks CA, continuity appears in behavioral sleep state
from 30 V (15) to 15 V (16), with 30 V recommended as characterized as non-REM (NREM). However, from that time
the upper limit (17). until about 5 to 6 weeks post-term, during periods of NREM
Several groups of investigators have measured IBIs in order to sleep the EEG demonstrated semi-periodic episodes of gener-
establish normative data at the various CAs. Although all have alized voltage attenuation, not quiescence, lasting from 3 to 15
noted the trend of shortened IBIs with increasing CA, there is seconds; a pattern that has been called trace alternant. These
much variation in mean and maximum IBI values at the earliest periods of attenuation differ from those of trace discontinu
CAs (1,2,15–20). The variation in findings may relate to method- since the trace alternant is characterized by persistence of
ology differences that include amplitude of activity during the waveforms at lower voltage, whereas trace discontinu features
IBI (the lower the allowable amplitude, the shorter the attenuated
interval), localization of attenuated activity of IBI (all channels
simultaneously vs. allowable activity in some channels), method
of measurement of burst onset, and the presence and degree of
underlying central nervous system dysfunction such as hypoxic-
ischemia (which may prolong IBIs, even at subclinical levels).
In most studies of IBI duration, the earliest CA characterized
has been approximately 27 to 28 weeks. There are limited data
concerning the IBIs of younger neonates. Hayakawa et al. (21)
studied infants beginning as young as 21 to 22 weeks CA and
observed that the mean and maximum IBI duration also
decreased with increasing CA. Vecchierini et al. (22) concluded
that in neurologically normal 24- to 26-week-CA infants the
IBIs do not exceed 60 seconds. In addition, Clancy et al. (8) sug-
gested that a starting point for mean IBI in the very premature
infant (24 weeks CA) is 10 seconds.
Figure 9.2 represents a composite of data of maximum IBIs Figure 9.2 Average duration of interburst intervals in NREM sleep in
based on various studies (23) beginning with a CA of approxi- prematurity. Composite of findings from studies of interburst intervals.
mately 26 weeks. Within an individual recording the duration (From Hrachovy RA. Development of the normal electroencephalo-
of IBIs may vary. However, in clinical practice, the IBIs with the gram. In: Levin KH, Luders HO, eds. Comprehensive Clinica l
longest duration are most clinically relevant. Neurophysiology. Philadelphia, PA: WB Saunders; 2000:387–413.)
Chapter 9 ■ Normal EEG and Sleep: Preterm and Term Neonates 157

Figure 9.3 29 to 30 weeks CA. Beta–delta com-


plexes are present predominantly in the left central
region (shaded oval). Temporal theta bursts are
present independently on the left and right (shaded
square). The EEG is discontinuous and asynchro-
nous. (Reprinted with permission from Mizrahi EM,
Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia, PA:
Lippincott Williams & Wilkins; 2004.)

periods of electrical quiescence with virtual absence of electri- delta bursts,” “brushes,” “spindle-like fast waves,” and “ripples of
cal activity. prematurity.” Dreyfus-Brisac et al. (24), who first described the
complexes, referred them as “rapid bursts,” emphasizing the fast
Bilateral Hemispheric Synchrony component of the complexes.
It has often been reported that at or prior to 27 to 28 weeks CA, They are present from about 26 to 38 weeks CA and consist of
bursts of activity in the two hemispheres between periods of randomly occurring 0.3- to 1.5-Hz waves of 50 to 250 V, with
quiescence occur as generalized bisynchronous bursts with a superimposed bursts of low- to moderate-voltage fast activity
discontinuous background (2,19). However, others have (Fig. 9.3). The frequency of the fast activity may vary, even in the
reported that at 27 to 28 weeks CA the activity is generally asyn- same infant. Two frequencies predominate: 8 to 12 Hz and, more
chronous in homologous regions of the hemispheres (9). The commonly, 18 to 22 Hz. The voltage of the fast activity varies
greater the recording distance from the midline, the greater throughout each burst but rarely exceeds 75 V. Until 32 weeks
the degree of asynchrony (Fig. 9.3). With increasing maturity, CA, the fast component has a predominant frequency of 18 to 22
the degree of asynchrony diminishes. The degree of asynchrony Hz; thereafter, the slower frequency is most often present.
reflects not only maturation but also wake/sleep state of the The complexes first appear as a dominant feature in the EEG
infant. Asynchrony is most prominent in NREM sleep and is at about 26 weeks CA. When first present they occur infre-
least prominent in REM sleep. The only exception to these gen- quently, predominantly in the central regions. During the next
eral rules is that from the time frontal sharp waves first appear, 5 to 6 weeks, they become progressively more persistent, and
at about 35 weeks CA, they are bilaterally synchronous. the voltage of the fast component usually increases. From 29 to
33 weeks CA, the pattern is a prominent feature during behav-
Age-Dependent Waveforms ioral REM sleep. The spatial distribution of the beta–delta com-
There is an orderly appearance and disappearance of specific plexes is also age dependent, becoming more prominent in the
waveforms and patterns with increasing CA. These age-depend- occipital and temporal areas with increasing age. Beta–delta
ent waveforms have been referred to as “grapho-elements” and complexes typically occur asynchronously in derivations from
are essential landmarks in assessing CA and determination of homologous areas and show a variable voltage asymmetry on
the degree to which the background EEG is normal. the two sides.
At 33 weeks CA, beta–delta complexes are maximally
Beta–Delta Complexes expressed in NREM sleep, rather than REM sleep, and appear
These constitute the principal landmarks of prematurity. more prominently in the temporal-occipital areas. From 33 to
Various names have been given to these complexes: “spindle- 38 weeks CA, beta–delta complexes continue to occur primarily
158 Part II ■ Normal EEG

Figure 9.4 33 weeks CA. Temporal alpha bursts


are present on the left (shaded square) and
beta–delta complexes are present in the occipital
(shaded oval) and temporal regions (shaded
oval). The EEG is discontinuous and asynchro-
nous. (Reprinted with permission from Mizrahi
EM, Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia, PA:
Lippincott Williams & Wilkins; 2004.)

in NREM quiet sleep. The disappearance of the beta–delta com- relation to other developmental features. The occurrence of
plexes when the infant appears behaviorally awake constitutes temporal alpha bursts defines a more narrow CA range at 33
one marker of 36 to 37 weeks CA. weeks.

Temporal Theta and Alpha Bursts Frontal Sharp Waves


Temporal theta bursts appear at about 26 weeks CA and are These are characterized as isolated sharp, but bluntly config-
maximally expressed between 30 and 32 weeks (Fig. 9.3). They ured waves. They usually have an initial surface-negative phase
then rapidly disappear—replaced by temporal alpha bursts, a followed by a surface-positive phase. They have been referred to
very specific marker for 33 weeks CA. Temporal theta bursts are as “enchoches frontales.” Frontal sharp waves may first appear
characterized by rhythmic 4.5- to 6.0-Hz waves occurring inde- at 34 weeks CA but attain maximum expression at about 35
pendently in short bursts of rarely more than 2 seconds from weeks CA (Fig. 9.5). When they initially appear they are low
the left and right midtemporal areas. Voltage varies from voltage and infrequent in occurrence, with a less well-defined
roughly 20 to 200 V. Some individual waves often have a sharp waveform than when they are maximally expressed later. After
configuration. Temporal alpha bursts have similar features 44 weeks CA they diminish in number and voltage and are only
except the frequency of their waveforms is greater than 6.0 Hz rarely seen in infants older than 6 weeks post-term.
(Fig. 9.4). Frontal sharp transients are bilaterally synchronous and
In practical terms, strongly expressed temporal theta bursts symmetrical from the time of their first appearance. The initial
can serve as a useful developmental landmark in determining surface-negative component lasts about 200 msec. The succeed-
CA between 30 and 32 weeks—particularly when considered in ing surface-positive component lasts somewhat longer and is

Figure 9.5 36 to 37 weeks CA. The awake EEG


is characterized by frontal sharp transients that
occur bilaterally, synchronously, and simultane-
ously (shaded square). The background activity
is continuous and synchronous, with some per-
sistent beta–delta complexes in occipital regions.
(Reprinted with permission from Mizrahi EM,
Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia, PA:
Lippincott Williams & Wilkins; 2004.)
Chapter 9 ■ Normal EEG and Sleep: Preterm and Term Neonates 159

variable. Although typical blunt in configuration, when they are awake state and eye closure is associated with sleep: relatively
maximally expressed, the waveforms may be quite sharp in unreliable criteria. Regular respiration, random eye move-
morphology. Frontal sharp transients typically occur randomly ments, and variable muscle tone are associated with NREM
as single events, predominantly in transitional rather than in sleep. Irregular respiration, rapid eye movements, and
REM or NREM sleep. However, they may also recur in brief decreased muscle tone are associated with REM sleep.
runs and may also be mixed with bifrontal delta activity, which
is another normal feature of near-term infants (see below). Wakefulness
At about 30 weeks CA, the background activity is continuous in
STATE CHANGES behavioral and polygraphic determined REM sleep and discon-
tinuous during wakefulness and behavioral and polygraphic
The EEG in Wakefulness and Sleep NREM sleep. In all states, beta–delta complexes are present with
Until 36 to 37 weeks CA, distinguishing the various states of the their age-dependent characteristics of abundance, spatial distri-
wake/sleep cycle is based on infant behavior and findings of bution, and degree of synchrony. By 36 to 37 weeks CA, there is
polygraphic recordings rather than EEG features, although a clear distinction between the waking EEG and the sleep EEG,
there may be EEG changes associated with behavioral state without reliance on clinical or polygraphic recordings
changes before that epoch. Eye opening is associated with the (Fig. 9.6). In the awake EEG age-dependent features persist and

Figure 9.6 34 to 35 weeks CA. A:


Awake: The background activity is
continuous with intermittent
beta–delta complexes in the occipital
regions (same recording as B). B:
NREM sleep: Discontinuity during
NREM sleep with otherwise similar
developmental features as during
wakefulness (same recording as A).
(Reprinted with permission from
Mizrahi EM, Hrachovy RA, Kellaway
P. Atlas of Neonatal Electroencepha -
lography. 3rd ed. Philadelphia, PA:
Lippincott Williams & Wilkins;
2004.)
160 Part II ■ Normal EEG

Figure 9.7 40 to 44 weeks CA.


The EEG is characterized by
NREM sleep. The fluctuating
amplitude of the background
activity is typical of the trace
alternant pattern of quiet sleep.

the background activity consists chiefly of continuous polyfre- occur in the transition from wakefulness to sleep and be a rel-
quency activity. This polyfrequency activity is characterized by atively brief period or may be more prolonged between
random, very slow, low-voltage activity best described as states.
baseline shifting, with superimposed semirhythmic 4 to 8 Hz
activity in all regions. In addition there may be very low voltage Reactivity to Stimulation
(18 to 22 Hz) activity and very low voltage (2 to 3 Hz) activity Between 33 and 34 weeks CA, changes in EEG activity in
in frontal regions. At 36 to 37 weeks, during wakefulness, the response to stimuli appear. By 37 weeks CA, these responses
beta–delta complexes are no longer present. may be easily and more reliably elicited. Typically, the response
to a stimulus is related to the character of the ongoing activity
NREM Sleep at the time of the stimulus. If high-voltage, very slow activity is
Prior to about 36 weeks CA, the background activity in behav- present an effective stimulus produces abrupt and pronounced
ioral and polygraphic determined NREM sleep is discontinu- generalized attenuation of voltage lasting upto several seconds
ous. Between 36 and 38 weeks CA, two patterns of NREM sleep (Fig. 9.8). In addition, there may be spontaneous episodes of
emerge: continuous high-voltage slow wave activity in all attenuation associated with self-arousal. These may occur in
regions and trace alternant (Fig. 9.7). The latter is characterized infants until about 2 weeks post-term and should not be inter-
by a modulation of activity with alternating periods of high- preted as evidence of immaturity or be confused with the repet-
and low-voltage activity. This pattern may be present upto 44 itive episodes of generalized or regional attenuation that occur
weeks CA. Thereafter, NREM sleep is characterized by continu- in neonatal encephalopathies.
ous slow wave activity with the eventual emergence of sleep
spindles after about 46 weeks CA (although rudimentary spin-
SPECIAL WAVEFORMS AND PATTERNS
dles may occur earlier).
Some special waveforms and patterns are present, particularly
REM Sleep in the near-term and term infant, which are considered to be
The character of the EEG during behavioral REM sleep is sim- within the range of normal variation.
ilar to that of the awake recording beginning from about 30
weeks CA. Infant behaviors and polygraphic recordings of res- Bifrontal Delta Activity
piration, eye movements, and submental EMG are used to dis- Bifrontal delta activity appears in the near-term or term infant
tinguish the awake state from REM sleep. as intermittent semirhythmic 1.5 to 2 Hz moderately high to
high voltage activity in the frontal regions bilaterally (Fig. 9.9).
Transitional or Indeterminant Sleep These waveforms may occur in close association with frontal
These terms are utilized to characterize the EEG when the sharp transients and are most prominent during transitional
state of the infant cannot be precisely determined. This may sleep. Although the pattern has been referred to as “anterior
Chapter 9 ■ Normal EEG and Sleep: Preterm and Term Neonates 161

Figure 9.8 38 to 40 weeks. A


transient arousal is characterized
by a brief episode of generalized
voltage attenuation. The back-
ground activity is consistent with
NREM sleep before and after the
arousal. (Reprinted with permis-
sion from Mizrahi EM, Hrachovy
RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed.
Philadelphia, PA: Lippincott
Williams & Wilkins; 2004.)

dysrhythmia,” it does not suggest nor has it been associated Criteria for abnormality include complex morphology, posi-
with any abnormality. tive polarity, persistent localization, and occurrence during
wakefulness.
Temporal Sharp Waves
Temporal sharp waves are typically discussed as abnormalities CONCLUSION
or findings of uncertain diagnostic significance (see Chapter
25). However, some temporal sharp waves may appear in oth- The interpretation of the neonatal electroencephalogram is
erwise normal neonatal EEG recordings and are considered based on the recognition of age-dependent features. For the
findings within the range of normal variation. Temporal sharp neonate with, ultimately a normal EEG, this provides the
waves may be considered normal, which meet the following basis for determination of CA and the assessment of
criteria: simple diphasic morphology, with primarily surface- expected background and focal features. In addition, the
negative polarity; occur randomly and, usually, asynchro- understanding of CA-dependent features will suggest the
nously on the two sides; and occur during sleep (Fig. 9.10). range of both diffuse and focal abnormal features that may

Figure 9.9 38 to 40 weeks.


Bifrontal delta activity is present
(shaded oval), mixed with frontal
sharp transients (shaded square)
in this recording of transitional or
indeterminant sleep. The EEG is
continuous and synchronous on
the two sides. There are no imma-
ture features. (Reprinted with per-
mission from Mizrahi EM,
Hrachovy RA, Kellaway P. Atlas of
Neonatal Electroencephalography.
3rd ed. Philadelphia, PA:
Lippincott Williams & Wilkins;
2004.)
162 Part II ■ Normal EEG

6. Hrachovy RA, Mizrahi EM, Kellaway P. Electroencephalography of


the newborn. In: Day DD, Pedley TA, eds. Current Practice of
Clinical Electroencephalography. 2nd ed. New York, NY: Raven
Press; 1990:201–242.
7. Watanabe K, Hayakawa F, Okumura A. Neonatal EEG: a powerful
tool in the assessment of brain damage in preterm infants. Brain
Dev. 1999;21:361–381.
8. Clancy RR, Bergqvist C, Dlugos DJ. Neonatal electroencephalogra-
phy. In: Ebersole JS, Pedley TA, eds. Current Practice of Clinical
Electroencephalography. 3rd ed. Philadelphia, PA: Lippincott
Williams & Wilkins; 2003:160–234.
9. Mizrahi EM, Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia, PA: Lippincott
Williams & Wilkins; 2004:250.
10. Plouin P, Kaminska A, Moutard ML, et al. L’EEG en pediatrie.
United Kingdom: John Libbey; 2005:185.
11. Vecchierini MF, André M, d’Allest AM. Normal EEG of premature
infants born between 24 and 30 weeks gestational age: terminol-
ogy, definitions and maturation aspects. Neurophysiol Clin.
2007;37:311–323.
12. André M, Lamblin MD, d’Allest AM, et al. Electroencephalography
in premature and full-term infants. Developmental features and
glossary. Neurophysiol Clin. 2010;40:59–124.
13. American Clinical Neurophysiology Society. Guideline 2.
Minimum technical standards for pediatric electroencephalogra-
phy. J Clin Neurophysiol. 2006;23(3):92–96.
Figure 9.10 38 to 40 weeks. A temporal sharp wave is present on the 14. De Weerd AW, Despand PA, Plouin P. Neonatal EEG: recommen-
left (shaded square). It is randomly occurring, moderate in voltage, sim- dations for the practice of clinical neurophysiology: guidelines for
ple in morphology occurring during transitional or indeterminant sleep. the International Federation of Clinical Neurophysiology.
The background activity is within the range of normal for CA. Electroencephalogr Clin Neurophysiol. 1999;52(suppl):149–157.
(Reprinted with permission from Mizrahi EM, Hrachovy RA, Kellaway 15. Biagioni E, Bartalena L, Boldrini U, et al. Background EEG activ-
P. Atlas of Neonatal Electroencephalography. 3rd ed. Philadelphia, PA: ity in preterm infants: correlation of outcome with selected mat-
urational features. Electroencephalogr Clin Neurophysiol. 1994;91:
Lippincott Williams & Wilkins; 2004.)
154–162.
16. Hahn JS, Monyer H, Tharp BR. Interburst interval measurements
in the EEGs of premature infants with normal neurological out-
comes. Electroencephalogr Clin Neurophysiol. 1989;73:410–418.
be present at specific epochs. An orderly approach to the
17. Selton D, Andre M, Hascoët JM. Normal EEG in very premature
visual analysis of the neonatal EEG will provide for its great- infants: reference criteria. Clin Neurophysiol. 2000;111:2116–2124.
est clinical utility. 18. Monod N, Tharp B. Activité electroencéphalographique normale du
nouveau-né du pré`maturé au cours des états de veille et de som-
REFERENCES meil. Rev Electroencephalogr Clin Neurophysiol. 1977;7:302–315.
19. Anderson CM, Torres F, Faoro A. The EEG of the early premature.
1. Dreyfus-Brisac C. The electroencephalogram of the premature Electroencephalogr Clin Neurophysiol. 1985;60:95–105.
infant. World Neurol. 1962;3:5–15. 20. Connell JA, Oozeer R, Dubowitz V. Continuous 4-channnel EEG
2. Dreyfus-Brisac C. The electroencephalogram of the premature monitoring: a guide to interpretation, with normal values, in
infant and full-term newborn: normal and abnormal development preterm infants. Neuropediatrics. 1987;18:138–145.
of waking and sleeping patterns. In: Kellaway P, Petersén I, eds. 21. Hayakawa M, Okumura A, Hayakawa F, et al. Background elec-
Neurological and Electroencephalographic Correlative Studies in troencephalographic (EEG) activities of very premature infants
Infancy. New York, NY: Grune & Stratton; 1964:186–206. born at less than 27 weeks gestation: a study on the degree of con-
3. Dreyfus-Brisac C, Fischgold H, Samson-Dollfus D, et al. Veille, tinuity. Arch Dis Child Fetal Neonatal Ed. 2001;84:F163–F167.
sommeil, réactivité sensorielle chez le prématuré, le nouveau-né et 22. Vecchierini MF, d’Allest AM, Verpillat P. EEG patterns in 10
le nourrisson. Electroencephalogr Clin Neurophysiol. 1957;6(suppl): extreme premature neonates with normal neurological outcome:
417–440. qualitative and quantitative data. Brain Dev. 2003;25:330–337.
4. Dreyfus-Brisac C, Monod N. Sleeping behavior in abnormal new- 23. Hrachovy RA. Development of the normal electroencephalogram.
born infants. Neuropädiatrie. 1970;1:354–366. In: Levin KH, Luders HO, eds. Comprehensive Clinical Neurophysi-
5. Dreyfus-Brisac C, Larroche JC. Discontinuous EEGs in premature ology. Philadelphia, PA: WB Saunders; 2000:387–413.
and full-term neonates. Electro-anatomo-clinical correlations. 24. Dreyfus-Brisac C, Blanc C. Électroencéphalogramme et matura-
Electroencephalogr Clin Neurophysiol. 1972;32:575. tion cerebrale. Encephale. 1956;45:205–241.
CHAPTER
Normal EEG and Sleep:
Infants to Adolescents
JAMES J. RIVIELLO, JR, DOUGLAS R. NORDLI, JR, AND ERNST NIEDERMEYER
10
GENERAL PRINCIPLES of 30 years), at a time when biologic aging of body tissues has
already started and physical capabilities or athletic performances
Electroencephalogram (EEG) interpretation depends on accu- have clearly passed their peak. However, cerebral maturation is
rate pattern recognition. One of the first lessons the novice elec- not a smooth process of steady growth, since quantum changes
troencephalographer learns is that EEG pattern interpretation occur in prematurity, as well as in infancy and childhood (1).
must take into account the patient age and the level of vigilance,
or state. EEG patterns vary according to central nervous system HISTORICAL ASPECTS
(CNS) development and maturation. This process evolves over
time, starting with the early development and maturation of the Berger (2) performed the first EEG studies in children and dis-
nervous system (an evolution) to a peak of maturity, followed covered age-dependent changes. The EEG in normal and
by an involution. Basic differences exist between the ascending abnormal children has been well studied and has been
(developmental) and the descending (involutional) portions of expanded with quantified EEG (QEEG). The developmental
this curve. This chapter discusses the influence of age and thus aspect of EEG interpretation is especially important in the
the ontogenetic evolution or maturation of the EEG. interpretation and study of the neonatal EEG. The technical
The declining curve of life is not determined by a reverse performance of the EEG and its interpretation in the premature
process such as “dematuration” (certainly not by demyelination), and full-term newborn requires such specialized knowledge
although there is degeneration of CNS structures. CNS aging is that neonatal electroencephalography is now a special branch of
caused predominantly by a variety of pathologic processes. The electroencephalography, neonatal electroencephalography.
awareness of this basic difference between the ascending and the Neonatal EEG is described in Chapter 9.
descending portions of life is of great significance in practical The EEG evolution from infancy to adolescence in a large
electroencephalography. During the maturational phase, the EEG number of healthy adolescents has been extensively studied by
must be interpreted knowing the conceptual age (CA) and Swedish authors (3–7), whose work set the standards for develop-
according to statistical norms, whereas in the aged individual, the mental electroencephalography. In a remarkable study of 1416
EEG is interpreted using the ideal or optimal norm, because the healthy subjects with an age range from 6 to 39 years using QEEG,
optimal situation, even in advanced age, is absence of pathology. Matsuura et al. (8) investigated the development of various fre-
Using a statistical norm, a mentally alert octogenarian with the quency ranges over the occipital, central, and frontopolar regions.
frequency pattern of a normal middle-aged adult would have to Gasser et al. (9,10) investigated the development of the EEG in
be read as abnormal. This would be an absurdity. children and adolescents by frequency analysis and topography.
Figure 10.1 demonstrates the maturational period, the peak EEG maturation has also been studied in animals. A study in
of maturity, and the decline caused by pathology. The peak the dog by Pampiglione (11) and the work of Caveness (12) in
period of EEG maturation lies well into adulthood (after the age the rhesus monkey deserve special attention. Table 10.1 provides

Figure 10.1 Cerebral maturation (infancy, childhood, and


adolescence) versus pathology with regard to repercussions
on the EEG.

163
164 Part II ■ Normal EEG

Tabl e 10 . 1

A Condensed View of EEG Maturation

Premature Premature Premature Full-Term Newborn


(24–27 Wk) (28–31 Wk) (32–35 Wk) (36–41 Wk)
Continuity Discontinuous, long flat Discontinuous Continuous in waking state and Continuous except for tracé
stretches REM sleep, discontinuous in alternant in non-REM
non-REM sleep (quiet) sleep
Interhemispheric Short bursts in synchrony Mostly asynchronous Partly synchronous, especially in Minor asynchronies still present
synchrony activity leads occipital
Differentiation of waking Undifferentiated Undifferentiated Waking distinguished from sleep Good
and sleeping early in the period, then
differentiation of non-REM
and REM sleep
Posterior basic (alpha) None None None None
rhythm

Slow activity (awake) Very slow bursts, high Very slow activity Slow (delta) with occipital Slow (delta), mostly of moderate
voltage (state of vigilance predominant maximum voltage
undifferentiated)
Temporal theta Present and increasing Prominent Decreasing and disappearing Disappearing or absent
Occipital theta Prominent Decreasing Decreasing Absent
Fast activity (awake) Very little beta activity Frequent ripples or Frequent ripples or brushes Decreasing ripples, sparse fast
brushes around (16–20 per second) activity
16 per second
Low voltage Long flat stretches Flat stretches, mainly Low-voltage record suspect of Very low voltage records are due
asynchronous serious cerebral pathology to severe cerebral pathology;
prognosis ominous
Hyperventilation Not feasible Not feasible Not feasible Not feasible

Intermittent photic Unknown Unknown Unknown Driving response below 4 flashes/


stimulation second may occur, not easily
elicited
Drowsiness Undifferentiated Undifferentiated Undifferentiated Undifferentiated

Tracé alternant None None Present in non-REM (quiet) sleep Present in non-REM (quiet) sleep

Spindles None None (but ripples present) None (but ripples present) None (but scanty ripples)

Vertex waves and K None None None None


complexes

Positive occipital sharp None None None None


transients of sleep
Slow and fast activity in Slow activity of high Much slow activity, more Irregular slow activity of occipital Much delta and theta activity,
sleep voltage, little slow activity irregular; little fast predominance continuous in REM sleep
(stage of vigilance activity
undifferentiated)
REM sleep Undifferentiated Undifferentiated Continuous slow activity; Continuous slow activity, REM in
oculographically, REM present EOG (more REM or “active”
than non-REM sleep)
Rhythmical frontal theta None None None None
activity (6–7 per second)
14 and 6 per second None None None None
positive spikes
Psychomotor variant None None None None
(marginal abnormality)
Sharp waves, spikes Some intermixed sharp Some intermixed sharp Often prominent sharp Some minor sharp transients
activity in bursts (normal) activity (normal) waves or spikes (normal) (normal) (abnormal spikes
more consistent and
prominent)
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 165

Early childhood Preschool age Older children Adolescents


Infancy (2–12 mo) (12–36 mo) (3–5 yr) (6–12 yr) (13–20 yr)
Continuous Continuous Continuous Continuous Continuous

No significant asynchrony No significant asynchrony No significant asynchrony No significant asynchrony No significant asynchrony

Good Good Good Good Good

Starting at age 3–4 mo at 4 per Rising from 5–6 per second Rising from 6–8 per second Reaching 10 per second at Averaging 10 per second
second, reaching about to 8 per second (seldom to 7–9 per second age 10 yr
6 per second at 12 mo 9 per second)
Considerable Considerable Marked admixture of Varying degree of posterior Posterior slow activity
posterior slow activity slow activity mixed with diminishing
(to alpha rhythm) alpha
None None None None None
None None None None None
Very moderate Mostly moderate Mostly moderate Mostly moderate Moderate, except for low
voltage fast records

Uncommon, usually Uncommon, usually Uncommon, usually Seldom as variant Occasionally and (at end of
abnormal abnormal abnormal of normalcy teenage period more often)
as variant of normalcy
Not feasible Mostly not feasible Often marked delta response Often marked delta response Delta responses become less
impressive
Improving driving to low Often good driving response Often good driving response Often good driving response, Often good driving response,
flash rates after age 6 mo to low flash rates to low flash rates chiefly at medium flash chiefly at medium flash
rates (8–16 per second) rates
Around age 6 mo, appearance Marked “hypnagogic” Rhythmical theta gradually Gradual alpha dropout with Gradual alpha dropout with
of rhythmical theta rhythmical theta vanishing, other types of increasing slow activity low-voltage stretches
(4–6 per second) slow activity predominant (mainly slow)
Disappears in first (seldom None None None None
second) month
Appear after second In second year, sharp and Typical vertex maximum Typical vertex maximum Typical vertex maximum
month; 12–15 per shifting, then symmetrical
second, sharp, shifting with vertex maximum
Appear mainly at 5 mo, Large, becoming more Large with an increasingly Large with a prominent Not quite as large, sharp
fairly large, blunt pointed impressive sharp sharp component component not quite
component as prominent
None Poorly defined Poorly defined Still poorly defined but Often very well developed
gradually evolving
Much diffuse 0.75–3 per Marked posterior maximum Predominant slowing but Much diffuse slowing, Much diffuse slowing with
second activity with of slow activity; often a less prominent posterior slightly decreasing further attenuation of
posterior maximum; good deal of fast activity maximum voltage voltage
moderate fast activity
REM portion decreasing; Mostly slow, starting to Slow activity with some Less slowing and increasing Mature desynchronization
mostly slow activity become more desynchronization desynchronization
desynchronized
None Seldom in third year of life May occur, not very common A bit more common A bit more common,
declining at end of period
None Rare May occur, not very common Fairly common Fairly common

None None Probably none Uncommon More common (although


relatively rare)
Essential as abnormal Spikes in seizure- free Spikes in seizure-free Spikes in seizure-free Benign Rolandic spikes
phenomena children, mainly occipital children, mainly children, mainly Rolandic usually disappear before
(mild abnormalities) occipital, also (central-mid-temporal), beginning of this period
Rolandic (slight slight to moderate
abnormalities) abnormalities; physiological
occipital spikes in
congenitally blind children
166 Part II ■ Normal EEG

a condensed presentation of a number of EEG variables and night before and allow the infant to lie on a bed or in the
their developmental aspects. mother’s arms. Sweeney et al. (16) compared sleep deprivation
versus sedation. The recordings had fewer artifacts and were off
DISAPPEARANCE OF NEONATAL PATTERNS a higher quality with sleep deprivation. Guidelines have been
established for sleep deprivation (17). The use of sleep depriva-
Neonatal EEG patterns generally end between 46 and 48 weeks tion has been confirmed by Ong et al. (18). In a sedated infant,
CA and transition to predominantly infantile EEG patterns end it is often impossible to secure a waking tracing. Using the sec-
by 3 months of age. The trace alternant pattern is the EEG ond approach, the record routinely starts while the baby is
correlate of quiet sleep (non-REM sleep) and is common dur- awake. It is extremely difficult to place electrodes under these
ing the first 1 to 3 weeks of life in a full-term newborn. Active circumstances as most infants resent this type of manipulation.
sleep (REM sleep) occurs more commonly in the sleeping new- Some EEG technologists have become extremely experi-
born (64% of sleeping time, according to Passouant et al. (13)). enced in the art of handling a waking infant, and often succeed
The healthy full-term newborn demonstrates active sleep (REM in obtaining both waking and spontaneous sleep records.
sleep) at sleep onset. Further information about the management of infants and chil-
Quiet sleep onset is well established 1 month after full-term dren in North American EEG laboratories is found in the study
delivery (37 days, after Ellingson (14)) and gradually emerges of Leonberg (19).
as the predominant type of sleep with the development of The International 10-20 Electrode System should be used for
stages 2, 3, and 4. Trace alternant patterns disappear about 3 electrode placement, using a full set of 21 EEG electrodes, 19
to 4 weeks after full-term birth (15), although they have been scalp electrodes, and two ear reference electrodes, according to
observed up to day 47 (14). Intervening CNS insults, either the American Clinical Neurophysiology Society guidelines (20).
primary or secondary (systemic), may interfere with CNS The use of ocular leads, respiratory monitoring, and electrocar-
function and temporarily cause developmental regression, diogram (ECG) are helpful in the detection of artifacts, as well
which may be expressed with a return to a less mature EEG as in the assessment of sleep stages.
pattern. Although some laboratories use rubber caps in small infant
With the disappearance of neonatal EEG patterns and the sizes, this technique is not conducive to sleep, and collodion is
emergence of patterns such as the posterior basic (the posterior preferable, especially in the larger pediatric hospitals. Bentonite
dominant rhythm), the forerunner of alpha rhythm, and sleep paste (recommended by Fois (21)) is no longer available. Excellent
spindles, the EEG begins to demonstrate the more mature new types of commercial paste are now available, but when used,
patterns seen in adults. During the waking state, a generally it is possible that rapid drying and thus deteriorating electrode
rhythmical 3 to 4 Hz posterior activity occurs, a precursor of function may occur, resulting in high electrode impedance values,
the posterior alpha rhythm, or the posterior occipital or domi- in addition to the natural higher impedance of the scalp in new-
nant rhythm. This posterior rhythm demonstrates reactivity to borns and early infancy. Needle electrodes are now rarely used.
eye opening and closing: blocking with eye opening and activa-
tion with eye closing. EEG Characteristics in the Waking State
In early infancy (around the age of 2 months), irregular delta
EEG IN INFANCY (2 TO 12 MONTHS) activity of 2 to 3.5 Hz and medium to high voltage (50 to 100
V) is widely preponderant. As noted above, rhythmical occip-
General Considerations and Technical Aspects ital 3- to 4-Hz activity is often noted at the age of 3 to 4 months;
It is not easy to obtain a waking EEG recording in an infant. As this activity can be blocked by eye opening.
infants tend to keep their eyes open, in order to obtain a poste- This posterior basic (dominant) rhythm becomes more sta-
rior dominant rhythm, Dreyfus and Curzi-Dascalova (15) sug- ble at the age of 5 months and increases over time, first to 5 Hz
gest gentle passive occlusion of the eyes (passive eye closure) for (15) and subsequently to an average frequency of 6 to 7 Hz
short periods to activate the posterior basic rhythm. Temporary (occasionally 8 Hz) by age 12 months. The amplitudes range
use of a short-time constant (0.1 second) may be helpful to sep- from 50 to 100 V.
arate the posterior rhythm from movement artifacts. This Some rhythmical rolandic (central) activity of 5 to 8 Hz may
rhythm may also be present in a crying infant (associated with be present as early as the age of 3 months, with an amplitude
forceful closure of eyes and concomitant frontal muscle arti- around 25 to 50 V. This activity is the precursor of the mu
facts), as well as in a quiet infant with open eyes. The infant usu- rhythm and is stable during the first year of life (15), but may
ally closes the eyes as a sign of impending drowsiness. be dependent on somatosensory stimulation (22). These visual
There are two basic philosophies to EEG recording in analysis findings have been confirmed with QEEG (23–25).
infancy. In the first, the recording is started in a sleeping baby.
Spontaneous sleep is desirable, and this may be achieved when EEG Characteristics in Drowsiness
the recording is scheduled shortly after feeding. In the past, Prior to the age of 5 to 8 months, the transition from the waking
sedation was commonly used, usually chloral hydrate. Because state to sleep is a gradual process characterized by progressive
of concerns for adverse events with conscious sedation, such as slowing into the delta frequency range. No specific drowsy state
apnea, the use of conscious sedation for EEG has been disap- is identified in this smooth progression of slow activity to the
pearing. Many laboratories now favor sleep deprivation the sleep stage. Drowsiness is recognized by a distinct pattern
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 167

between the age of 6 and 8 months: a hypersynchronous rhythm month. With the third month, the amplitude increases and the
in the lower theta range, around 4 Hz, with gradual acceleration duration of the runs becomes much longer (30). The intervals
to 5 and 6 Hz over the ensuing months. This impressive theta between spindle trains become shorter, but we do not agree
rhythmicity is known as “hypnagogic hypersynchrony” (26). that spindling is almost continuous between the age of 2 and
This drowsy pattern seems to develop from the posterior 6 months (24). Spindle bursts may reach a duration of 10 sec-
basic (dominant) rhythm. The transition from wakefulness to onds in the second half of the first year, but there is a decreased
drowsiness is associated with a change in the amplitude distri- duration of each spindle run, while the overall number of runs
bution. The maximum rhythmical theta activity moves into the increases.
centroparietal region where amplitudes commonly reach 100 to Dreyfus and Curzi-Dascalova (15) report that the complete
250 V. EEG amplitude values depend on the interelectrode absence of spindles at the age of 3 to 8 months represents a
distance and may vary from montage to montage and are best severe abnormality. However, this finding should be interpreted
measured with a reference montage. with great caution, as enough sleep must have been obtained
According to Dreyfus and Curzi-Dascalova (15), the occipi- during the recording in order “to give the baby a chance to pro-
tal rhythm may be somewhat slower than the diffusely predom- duce spindles.” At this age, there is still a fair chance that the
inant theta rhythm (possibly indicating a basic difference sleep recording is limited to REM sleep; this demonstrates the
between two coexisting rhythmical theta patterns). In rare importance of oculographic and pneumographic recording.
cases, the hypnogogic hypersynchrony may not occur. An excellent demonstration of the development of sleep
spindles from the age of 10 weeks to 1 year was presented by
Sleep EEG and Non-REM Sleep Hughes (31) (Fig. 10.3).
During the first 3 months of life, sleep may begin in a peculiar Vertex waves and K complexes are usually seen around the age
manner. Curzi-Dascalova et al. (27) have shown that some of 5 months, although rudiments may occur much earlier.
infants fall asleep without eye closure, others with half-closed Vertex waves may be quite large at the age of 5 to 6 months. At
eyes, and others with brief alternating opening and closing of this age, K complexes are of considerable voltage, but the initi-
the eyes. The EEG and polygraphic data indicate sleep onset ating sharp component is poorly developed and somewhat
with active (REM) sleep in neonates and gradual evolution of “blunted” (32). In contrast, Metcalf et al. (33) have stressed the
sleep onset with quiet (non-REM) sleep during the ensuing comparatively low voltage of K complexes in infancy. These
weeks. The “slow” sleep of the infant is dominated by diffuse authors also noticed the appearance of K complexes in infants
0.75- to 3-Hz activity with a maximum amplitude (100 to 150 aged 5 to 6 months, but the complexes may be obscured by
or 200 V) over the occipital area, “occipital delta.” This occip- background activity (Figs. 10.4 and 10.5). A vertex wave may be
ital delta activity may be quite prominent during the first year frontally dominant or extend into the lateral frontal regions;
of life. The amplitudes increase with deepening slow sleep. this is called an F-wave (34) (Fig. 10.6).
There are some intermixed theta, alpha, and beta frequencies of In normal infants in the first year of life, Ellingson et al. (35)
smaller amplitudes. recorded brief apneic episodes (“respiratory pauses” lasting 3 to
Sleep spindles usually appear during the second month of 10 seconds) during sleep. These pauses are more common in
life; occasionally, spindle fragments may be seen somewhat ear- REM sleep.
lier (28). The spindle frequency ranges from 12 to 15 Hz, with
14 Hz the most commonly encountered frequency. Throughout Sleep EEG and REM Sleep
infancy, spindles are maximal over central and parietal areas REM sleep abundance decreases during the first year of life
with shifting asymmetries. A clear-cut midline (vertex) maxi- (36,37), evolving from approximately 50% at birth, falling to
mum does not exist at this age. Spindles of infancy usually show 40% at 3 to 5 months and 30% between 12 and 24 months.
a negative sharp component, whereas the positive component is Dittrichova et al. (38) and Dreyfus and Curzi-Dascalova (15)
rounded. The sharp spindle configuration (shown by Fois (21)) reported the occurrence of sharply contoured occipital activity
is a typical hallmark of sleep in infancy. The comb-like shape of in the REM sleep of infants. This activity shows a frequency
these runs may be erroneously interpreted as 14-Hz positive around 2 Hz at 6 weeks and 2 to 4 Hz at 12 to 16 weeks of age.
spikes, but a careful analysis of polarity clearly shows the nega- There is some intermixed delta and theta activity associated
tivity of the spiky components. The spatial distribution also is with the occipital sharp transients.
different: spindles are rolandic, whereas 14-Hz positive spikes The REM sleep latency (time span from sleep onset to the
are predominant in a posterotemporal location. Finally, the first REM period) gradually lengthens during the first year of
14-Hz or 14- and 6-Hz positive spike pattern is extremely rare life. Schulz et al. (39) has shown that the REM sleep latency
before the age of 2 years and virtually nonexistent during the underlies diurnal rhythmical changes, with the longest latencies
first year of life (Fig. 10.2). between noon and 4 PM and the shortest between 4 AM and 8 AM.
Spindles may show sharp negative and positive phases after
the age of 6 months Katsurada (29). This sharp positive spindle Reactivity and Evoked Responses
polarity is less common than spindles with a strictly negative The evolution of evoked responses in newborns and infants are
sharp component. discussed in Chapter 49.
The spindle train duration varies with age. Spindle trains The blocking response of the posterior basic (dominant)
are of short duration and low voltage during the second rhythm (obtained by passive eye closure, see General
168 Part II ■ Normal EEG

Figure 10.2 A: Patient age 10 months. Light non-REM sleep. Typical spindles of infancy,
sharp and shifting. Normal posterior voltage maximum of slow activity (channels 3 to 7
from the top). B: Patient age 9 months. More prolonged trains of infantile spindles.

Considerations and Technical Aspects) is usually present at the prominent in the theta band (41,42). Occipital lambda wave
age of 3 to 4 months. The central mu rhythm does not react to activity occurs rarely during the first year of life.
eye opening, which permits differentiation when there is ante-
rior spreading of the posterior dominant rhythm. A true EEG IN EARLY CHILDHOOD
rolandic mu rhythm cannot be identified during the first year (12 TO 36 MONTHS)
of life, although forerunners of this activity are likely to be pres-
ent. The authors’ earliest observation of unmistakable rolandic Waking Record
mu rhythm was made in a 20-month-old child (40). Readable waking records are not easily obtained in the second
A photic driving response to flickering light may be obtained and third years of life. At this age, many children resent being laid
as early as 3 to 4 months after delivery; the responses are most down in a supine position for electrode placement and recording
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 169

Figure 10.3 Sleep spindles at different ages. At 10 weeks note the very
low amplitude at the beginning and end of the burst (underlined) with
a duration of 5 seconds; at 13 weeks a long lasting spindle of 6.5 sec-
onds; at 17 weeks, 3.5 seconds; at 39 weeks, 2 seconds; and 52 weeks,
1.5 seconds. Time (1 second) and voltage calibration (50 V) are
shown. Montage is bipolar linkage of the C3-P3 electrodes. Arrows des-
ignate the beginning and ending of typical spindles at the given age.
(From Hughes JR. Development of sleep spindles in the first year of life.
Clin Electroencephalogr. 1996;27:107–115.)

and may be too heavy to be comfortably held. These children may


successfully fight sleepiness and sleep, and if sedated, the waking
portion is a postarousal tracing, in which it must be determined
that the child is truly awake. Some children may pull off their
electrodes almost immediately after awakening.
The posterior basic (dominant) rhythm increases from the Figure 10.5 A K complex in a patient age 13 months; quite large with
upper theta to the lowest alpha range and is most commonly a blunted sharp component.
found between 6 and 7 Hz during the second and 7 and 8 Hz
during the third year of life. The blocking response to eye open-
ing intensifies, although the degree of this reactivity, a more
impressive response, is considerably determined by darkness or
brightness of the recording room.
The posterior basic (dominant) rhythm frequency is subject
to considerable variation in the second and third year of life.
According to Lindsley (43), a frequency range from 5 to almost
10 Hz can be observed in the second year, whereas Eeg-
Olofsson (3) reported a surprisingly high mean frequency of
8 Hz at this age.
Eye blinks are associated with a biphasic (negative–positive)
potential of medium to high voltage in bilateral synchrony over
occipital areas (44).
The amount of fast activity varies considerably. Unusually
pronounced activity in the 18 to 25 Hz range in unmedicated
children may reach abnormal degrees in the waking state as well
as in sleep; such records suggest a nonspecific abnormality and
may be seen in the study of Gibbs and Gibbs (45).
Frequencies slower than the posterior basic (dominant) rhythm
are practically always noted in the waking state at this age,
mostly in the 2 to 5 Hz range and widely scattered. The amount
of this activity is also quite variable and may be accentuated by
Figure 10.4 Patient age 9 months. Light non-REM sleep. A large vertex crying due to a hyperventilation effect.
wave after auditory stimulus. The response is somewhat complex and The reactivity of the waking EEG becomes more prominent;
resembles a K complex. the blocking response of the posterior basic (dominant) rhythm
170 Part II ■ Normal EEG

Figure 10.6 The F wave: a more


frontal (anterior) vertex wave.

is more clearly demonstrable than in the first year. A mu Sleep Record


rhythm over the rolandic region may be identifiable in the Sleep begins with non-REM sleep. Because of the usual rela-
second year of life (40). The photic driving response to the tively short duration of a sleep recording in the EEG laboratory
strobe light remains accentuated in the low-frequency range. in daytime, REM stages are usually not observed. The hypna-
gogic hypersynchrony of drowsiness is replaced by diffusely
Drowsiness
preponderant, irregular, high-voltage (1 to 3 Hz and medium-
Generalized high-voltage (mostly 4 to 6 Hz) theta activity is the to high-voltage 4 to 6 Hz) activity. The maximum amplitude is
hallmark of the drowsy state in early childhood. This drowsy almost invariably found over the occipital area, where 0.75- to
pattern is called hypnogogic hypersynchrony. It is most pro- 2-Hz waves may reach very high voltage (Fig. 10.2). A different
nounced in the central and parietal leads, is less impressive over type of voltage distribution or even an anterior voltage maxi-
the temporal areas, and sometimes least developed over the mum should warn the electroencephalographer; residual dam-
occipital region. The rhythmicity may assume quite monoto- age to the posterior regions is a good possibility, although none
nous character; the in-phase character of the ubiquitous pattern of the typical EEG abnormalities may be present (Fig. 10.8).
makes it tempting to theorize about an enormously extended This anterior to posterior amplitude difference is referred to as
generator area (Fig. 10.7). the frequency amplitude gradient (48).
Gibbs and Gibbs (46) paid special attention to a variant of The varying degree of intermixed fast activity without pre-
the rhythmical theta activity of drowsiness. In a large number medication was mentioned earlier. Spindle activity in the 12- to
of small children, bursts of 4- to 5-Hz activity may be present
instead of the steady rhythmical theta activity. This pattern
extends into the fourth year of life and shows mild paroxysmal
activity without representing a true abnormality. Occasionally,
small sharp or spiky discharges may be interspersed between
the theta waves; the electroencephalographer must refrain from
calling these discharges epileptic or spike–wave complexes.
Brandt et al. (47) determined that these admixed sharp or spiky
components occur in a small percentage (approximately 5 to
10%) of normal children at all ages. This was confirmed by Eeg-
Olofsson et al. (5). Unless unquestionable spikes are detectable,
the presence of such small spiky potentials is harmless, does not
represent a definite abnormality, and should be interpreted as a
normal finding. The mature form of drowsy EEG activity with
dropout of the posterior basic (dominant) rhythm and general Figure 10.7 Patient age 3 years. Drowsiness. Rhythmical hypnagogic
decline of the voltage output is quite common at this age. theta activity, 4 to 5 per second.
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 171

Figure 10.8 Patient age 9 months. Cerebral palsy, diplegic. Lack of


posterior voltage maximum. Otherwise a normal record with sharp
spindles of infancy.

14-Hz range is now found in a transitional stage; the shifting


spindle runs occurring over centroparietal areas with a nega-
tive spiky component are gradually replaced by symmetrical
spindle activity with a maximum over vertex. A 14-Hz spindle Figure 10.9 Patient age 22 months. REM sleep.
type appears first over the vertex, and with deepening sleep
(transition stage 2 to 3), spindles in the 12-Hz range appear The fast spiky spindle variant (51) is a very rare pattern
over frontal midline and upper frontal regions. Both types of (about 1 case in 2000 to 3000 EEGs of children). It consists of
spindles may occur concomitantly, but have a different spatial spindle-like activity in the range of 16 to 20 Hz and may occur
distribution. over a variety of areas (central, parietal, vertex, or mid-
Vertex waves, also known as “biparietal humps” (46), are temporal) in stage 2 or 3 of sleep. It is seen mainly in early child-
quite prominent at this age; their amplitude is impressive, but hood (around the age of 3 years) but also occurs in older chil-
their rise is not as abrupt and the configuration is not as sharp dren and adolescents. This fast spiky spindle variant is usually
as in later years (the age of 4 to 12 years). This same is true for seen in children with static or progressive brain dysfunction
K complexes, which are abundantly noted in the stages of non- and probably indicates a mild abnormality.
REM sleep. The 14- and 6-Hz positive spike discharge is very uncommon
Positive occipital sharp transients of sleep (occipital positive before the age of 3 years and will be discussed in Chapter 26. It
sharp activity, lambdoid activity) are absent or very poorly is now called the 14- and 6-Hz positive burst.
developed at this age.
REM sleep at this age rarely occurs during a standard EEG,
but is seen during a standard sleep study; it starts to show signs
of EEG desynchronization, but slow activity (mostly 2 to 5 Hz,
according to Cadilhac (49)) is still preponderant (Fig. 10.9).

Unusual and Variant Patterns in Sleep


Extreme spindles (45,50) represent an unusual variant of sleep
spindle activity with high voltage, a wide frequency range (6 to
18 Hz), and occasional paroxysmal traits. In a diminutive form,
these spindles may even occur while the child is awake (coexist-
ing with a posterior alpha rhythm) (Fig. 10.10).
According to Gibbs and Gibbs (45), this pattern occurs in
0.05% of normal children, but the prevalence reaches 5% to 18%
in children with mental retardation and/or cerebral palsy. This pat-
tern is seen from the age of 1 to 12 years, with a peak at the age of
3 years; after the age of 12 years, the incidence falls to almost zero.
Following the original studies of Gibbs and Gibbs, there have
been very few reports dealing with this pattern. When clearly
separable from typical spindle activity, extreme spindles might Figure 10.10 Patient age 22 months. Extreme spindles over central
represent a mild abnormality. region; a somewhat slower type of spindles over the frontal area.
172 Part II ■ Normal EEG

Arousal from Sleep


Arousal from sleep is characterized by a similar marked and We hypothesize that there is a limited number of EEG
prolonged high-voltage (4 to 6 Hz) activity in all leads with responses to cerebral impairment prior to the age of 21 months,
some intermixed slower frequencies (52,53). This is called with a lack of fine intermediate nuances of abnormality. It is
hypnopompic hypersynchrony. White and Tharp (54) reported possible that minor abnormalities are not so rare after all and
the observation of prolonged rhythmical sharp or spiky activity that new valid criteria will evolve for the interpretation of the
over the frontal areas in children between the age of 2 and 12 EEG infancy and early childhood. Recent progress in neonatal
years. This pattern was seen only in children with minimal cere- EEG has shown that classical signs of abnormality such as
bral dysfunction. In the authors’ experience, this pattern has a spikes, focal or diffuse slowing, or local voltage depression can-
very definite maximum over the frontal midline and is seen not serve as the only guideline; the sequences of sleep stages and
mainly from the second to the fourth year of life. Its mildly their adequacy for the gestational age are important criteria for
abnormal character is likely, but it is now considered a normal the newborn. In the older child and adult, an example of a new
variant (Fig. 10.11). criteria for normality is the frequency amplitude gradient:
However, in a study of 50 children with “frontal arousal lower amplitude, faster frequency waveforms in the anterior
rhythm,” it is rare, occurring in 0.22% of children, and epileptic regions with higher amplitude, slower frequency waveforms in
seizures were present in 35 (70%) of the cases (55). Hughes (56) the posterior regions, reported in sleep by Slater and Torres (48)
also reported a case in which this frontal arousal rhythm was (also see Fig. 10.8). However, this gradient is also present in the
associated with a clinical seizure consisting of eyelid flutter and waking state.
chewing.
PRESCHOOL-AGE EEG (3 TO 5 YEARS)
Scarcity of Minor Abnormalities during
the First 2 Years Waking Record
When abnormalities are carefully and consistently graded as A waking record is much easier to obtain, and with good han-
minimal, slight, moderate, marked, and severe, there is a sta- dling by the technologist, many children are quite cooperative.
tistically significant (p .001) small number of minor (i.e., At the age of 3 years, the posterior basic (dominant) rhythm has
minimal, slight, and moderate) abnormalities prior to the age reached 8 Hz and thus the alpha range. The alpha amplitude is
of 21 months. In other words, there is no real continuum of almost always higher than in adolescence and adulthood.
various forms of minor abnormalities between normal Leissner et al. (58) correlated the alpha amplitude with skull
records on one side and markedly abnormal records on thickness by ultrasound. The alpha amplitude may reach 100
the other side (57). After the age of 21 months (~2 years), V and tends to increase from the age of 3 to a peak at 8 or 9
minor abnormalities become much less frequent and the years. Amplitudes are usually higher over the nondominant
aforementioned continuum of graded abnormalities is pres- hemisphere (6).
ent. This data suggests that perinatal brain damage shows The train of posterior alpha waves is frequently interrupted
accompanying EEG abnormalities in the neonatal period, fol- by admixed slow waves, mostly in the range of 1.5 to 4 Hz,
lowed by a silent period with normal EEG findings yet ongo- extending from occipital into the posterior temporal and, less
ing clinical problems until the end of the second year of life, impressively, into the parietal regions. These admixed slow
when EEG abnormalities tend to reappear. waves, fused with overriding alpha, are called posterior slow
waves of youth. Alpha voltage, distribution, and admixture of
slow activity create a picture that makes the tracing almost
immediately identifiable as a record of childhood.
Posterior slowing may show a variety of forms. Most com-
mon is the irregularly interspersed type of slow activity.
These slow waves are often preceded by a sharply contoured
potential that blends together with the ensuing slow wave
“slow fused transients” (1). The slowing is more often later-
alized to the left hemisphere, according to Petersén and
Eeg-Olofsson (6); in the author’s experience, however, later-
alization to the right seems to be more common. Posterior
slow activity can reach abnormal degrees, especially with
towering amplitudes or abnormal rhythmicity such as pro-
longed trains of rhythmical slow waves. The differentiation of
such abnormal forms from physiologic posterior slow activ-
ity is usually not difficult. Posterior slow waves of youth react
to eye opening and closing, as does the posterior dominant,
or alpha, rhythm.
Figure 10.11 Patient age 16 months. Arousal from sleep. Pronounced In the absence of posterior slow activity, the posterior alpha
rhythmical 6 per sec activity with a sharp component, maximal over rhythm assumes a more “mature” character with unbroken
frontal midline. prolonged stretches. Eeg-Olofsson (4) and Petersén and Eeg-
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 173

Olofsson (6) list such tracings as “supernormal,” which is a childhood. Sleep spindles have lost their sharp and bilaterally
term comprehensible to every electroencephalographer. Such shifting feature of infancy and now show a well-defined
records suggest a hastened process of maturation, and a maximum over the vertex. Frequencies around 14 Hz are seen
5-year-old child may show the adult features of posterior alpha. before more anterior spindles in the 10- to 12-Hz range occur
Anterior rhythmical 6- to7-Hz theta activity is not uncom- with deepening sleep.
monly found at preschool age but reaches its peak somewhat Vertex waves and K complexes show a more prominent sharp
later. The clinical significance of this pattern is discussed in the component than in earlier years. Positive occipital sharp transients
next section. of sleep (lambdoid activity) are absent or very poorly delineated
Rolandic mu rhythm is clearly on the rise at this age before at this age. REM sleep is rarely recorded under regular laboratory
peaking early in the second decade of life. conditions. The EEG shows little desynchronization at this age.
At this age, low-voltage records with activity consistently
below 25 to 30 V are not seen as a variant of normalcy. Such
tracings may be associated with states of cerebral palsy (45). EEG IN OLDER CHILDREN (6 TO 12 YEARS)
Excessively fast records usually represent drug effect; otherwise,
General Considerations
such records fall into the mildly to moderately abnormal range.
Normal children are fairly cooperative. Complete records with
Activations a resting awake portion, hyperventilation, intermittent photic
At the age of 4 years, children usually become very cooperative stimulation, and sleep are easily obtained.
for hyperventilation, although one seldom finds a cooperative 3- This is a well-explored age range. The most authoritative
year-old child. Four-year-olds seem to enjoy the test and are study in this field was carried out in Gšteborg, Sweden, by
sometimes hard to stop. The use of a pinwheel is very helpful. Eeg-Olofsson (3) and Petersén and Eeg-Olofsson (6) (also
High-amplitude slowing to hyperventilation can be very see (7). To secure a population of truly healthy children, a
pronounced at this age and must be considered normal unless remarkable set of strict criteria was used, excluding those
unequivocal epileptic discharges or marked asymmetries are with even minute signs of neurologic, autonomic, and psy-
found. Protracted slowing (either as rhythmical delta bursts or chological dysfunction.
as continuous irregular delta–theta activity) after termination
of hyperventilation usually indicates that the child continues Waking Record
overbreathing. In such cases, the effect of hypoglycemia must The posterior alpha rhythm gradually reaches a mean frequency
also be taken into consideration. It may be important to of around 10 Hz, which equals the mean frequency of the
document the time of the last meal. Special studies in this field mature adult EEG. This frequency is reached around the age of
were done by Gibbs et al. (59) and by Petersén and Eeg- 10 years. In the work of Petersén and Eeg-Olofsson (6), the
Olofsson (6). A prolonged response to hyperventilation may mean value of 9 Hz was reached at the age of 7 and 10 Hz at
indicate vascular disease, as has been specifically reported in 15 years. Interestingly, girls showed a statistically significant
Moyamoya syndrome (60). faster acceleration of the posterior alpha frequency. These
The occipital driving response to intermittent photic stimula- authors reported an increase of the alpha amplitude during
tion is quite often best obtained with a flash frequency in the early childhood until a peak was reached at the age of 6 to
lower flicker frequency range; responses are most prominent 9 years, with subsequent gradual decline. There was no gender
below 8 Hz (6). Paroxysmal responses to flicker (even in the difference. The alpha amplitude was determined by a bipolar
normal child) are more likely to occur in school-age children. occipitotemporal array, and a mean of 56 V was found; such
precise values are completely dependent on electrode selection
EEG Findings in Drowsiness and interelectrode distance.
Past 3 years of age, the previously mentioned hypnagogic The posterior alpha rhythm is usually of higher amplitude
hypersynchrony disappears; it is seldom seen in healthy chil- over the nondominant hemisphere and thus larger on the right
dren at the age of 6 years. Paroxysmal theta bursts are also on side. According to Petersén and Eeg-Olofsson (6), these asym-
the decline. The admixture of posterior slow activity tends to metries seldom exceed 20 V. These authors were also unable to
increase in early drowsiness, and increased delta and theta correlate alpha asymmetries with handedness.
activity soon occurs in all leads. Although the mature alpha frequency of 10 Hz may be
Anterior rhythmical 6- to 7-Hz theta activity increases in early reached at the age of 10 or at least at age 15 years, there is still a
drowsiness but disappears in deep drowsiness. This pattern occurs considerable admixture of posterior slow activity interspersed
more often at school age and is discussed in Waking Record. between trains of alpha waves. The maturational process of the
The 14- and 6-Hz positive burst manifests itself in deep second decade of life is thus characterized by the gradual disap-
drowsiness and vanishes in light sleep. This pattern is not very pearance of the admixed posterior slow activity, excluding a few
common at this age and is discussed in Chapter 26. other minor changes in EEG evolution (Fig. 10.12A).
Posterior slow activity is still quite prominent between the
Sleep EEG Records age of 6 and 12 years; “slow fused transients” (61) with sharp
The posterior maximum of diffusely predominant delta (1 to transients preceding single large slow waves are quite common.
3 Hz) activity is not as pronounced as in infancy and early In addition to the common type of posterior slowing as randomly
174 Part II ■ Normal EEG

Figure 10.12 A: “Supernormal” posterior alpha rhythm in a patient aged 9 years. The only intermingled slow waves are
indicated by an arrow. B: Patient age 8 years. Considerable intermixed posterior slowing, mostly 2.5 to 3.5 per second.

intermixed slow potentials, there are also other and more rhyth- 5-Hz rhythm, which is a special entity of posterior slowing,
mical forms of posterior slow activity. is not demonstrable during the first decade. Very rhythmical
Waveforms of posterior slow activity are broken down into high-voltage 3- to 4-Hz waves may occur in prolonged runs in
arrhythmical and rhythmical slowing. This division also applies children with absence epilepsy. This is referred to as occipital
in adolescence and adulthood, although certain forms of poste- intermittent rhythmic delta activity (OIRDA) (66). This pattern
rior slowing are more common in children and others in adults. is clearly abnormal and is discussed in Chapter 26.
This subject has been extensively studied and discussed by Aird Figure 10.12B shows slow waves often preceded by a sharp-
and Gastaut (62), Petersén et al. (7), and Kuhlo (63). contoured large alpha wave (fused forms). Record is within nor-
In the age range from 6 to 12 years, posterior slow waves of mal limits of variability for age.
youth, or the arrhythmical type of “slow, posterior waves found Fast activity usually does not play a major role at this age.
predominantly in youth” (62), are most commonly encountered. Rolandic mu rhythm steadily increases until a peak is
This pattern consists of waves in the delta and theta range, of vari- reached between the age of 11 and 15 years (40). Occipital
able form, lasting 0.35 to 0.5 second or longer without any consis- lambda waves are demonstrated more easily in older than in
tent periodicity. Alpha waves are almost always intermingled or younger children but still lack the crisp and unmistakable
superimposed. Posterior slow waves of youth react to eye opening contour of adulthood.
with blocking, similar to the posterior dominant rhythm. The prevalence of anterior rhythmical 6- to 7-Hz theta activ-
Among the rhythmical forms, the slow alpha variant repre- ity increases between the age of 6 and 12 years before reaching
sents a subharmonic of the alpha frequency (mostly 4 to 5 Hz), a peak at the age of 13 to 15 years (67). These authors consider
often with intermixed alpha waves. Unilateral alpha subharmon- this pattern not an abnormality as such, but “of clinical interest
ics (left side, 4.5 Hz) were noted (64) in a neurologically normal because of its epileptologic implications” (Fig. 10.13).
10-year-old girl. Forms of pronounced rhythmical high-voltage This view differs from the opinion of Petersén et al. (7), who
(3- to 4-Hz) activity over occipital areas and adjacent regions are found a relatively high prevalence of anterior theta rhythm in
not uncommon at this age; these waves are often sharply con- healthy young individuals (21%). This figure is much larger than
toured, and the moderately paroxysmal character of these slow that of Palmer et al. (67), who found up to 4%. Interestingly,
runs places this activity into the mildly abnormal range. Petersén et al. (7) found a gender difference in the occurrence of
According to the extensive work of Kuhlo et al. (65), the 4- to this rhythm, with 24.9% in boys and 16.5% in girls.
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 175

Drowsiness
Drowsiness is characterized, at this age, by increasing theta and
delta frequencies along with gradually fading posterior rhyth-
mical alpha activity. The mature type of onset of drowsiness
with gradual alpha dropout and mixed low-voltage slow and
fast activity usually does not occur in the first decade and slowly
makes its appearance with early adolescence. Alternatively, hyp-
nagogic hypersynchrony seen in early childhood usually disap-
pears around the age of 6 years.

Sleep
Under regular EEG laboratory conditions (leaving aside noc-
turnal or all-day sleep studies), non-REM sleep in its various
stages is almost exclusively found unless the recording is pro-
longed to 1 hour or more.
Prior to the appearance of the first spindle trains, vertex waves
are noted (transition from stage 1 to 2). These vertex waves are of
remarkably high voltage and may have a very sharply contoured
morphology; at times, their sharp character may be poignant,
bordering on spikes. These potentials are also known as “bipari-
etal humps” because, in the montages of Gibbs and Gibbs (46),
the maximum was over the parietal region, because neither vertex
Figure 10.13 Rhythmical 6 to 7 per second activity over frontal
nor strictly central-rolandic electrodes were used (Fig. 10.14).
midline and vicinity in a 12-year-old child.
In school-age children, vertex waves may show moderate (sel-
dom considerable) amplitude asymmetries with asymmetrical
A 4- to 6-Hz theta rhythm in children with predisposition to
primary generalized epilepsy (spike–wave absences, etc.) has
been described by Doose et al. (68–70). This activity was seen
mainly below the age of 8 years and was common in seizure-free
siblings of children with petit mal absences. The predominantly
centroparietal theta activity is considered abnormal (after care-
ful differentiation from physiologic activity and drowsiness).
Low-voltage records as a variant of normalcy are very rare at
this age. Petersén and Eeg-Olofsson (6) did not find a single
case in their series. Schauseil-Zipf et al. (71) reassessed the EEG
at the age of 7 to 12 years in children who were regarded as
“small-for-date” babies. Out of a total of 56 children, the EEG
was normal in 36 and slightly to moderately abnormal in the
remaining cases.

Activations
Hyperventilation shows particularly pronounced high-ampli-
tude slowing at this age. The slowing may start over posterior
areas and gradually becomes diffusely distributed with a frontal
maximum; rhythmical slow activity usually ranges from 1.5 to
4 Hz. A variety of responses has been reported by Daute et al.
(72) (with concomitant evaluation of pH and pCO2 changes)
and by Petersén and Eeg-Olofsson (6). The normal character of
the rhythmical and arrhythmical slow responses cannot be
overemphasized (see Activations [Adolescents], below, for more
details).
Intermittent photic stimulation shows a more mature type of
occipital driving response, less prominent at low flash rates, and
more impressive in the medium range (6 to 16 Hz). The work
of Herrlin (73), Doose et al. (74), and Petersén and Eeg-
Olofsson (6) has been essential in the investigation of these Figure 10.14 Patient age 10 years, non-REM sleep stage 2. Well-
responses. developed spindles, vertex waves, and K complexes.
176 Part II ■ Normal EEG

spread into the vicinity. Under these circumstances, the physio-


logic vertex activity may be confused with cerebral (rolandic)
spikes; with greater experience, however, such errors can be
avoided. The distinction between vertex waves and rolandic
spikes, usually picked up by electrodes C3 or C4 over central
areas, may occasionally arise from the vertex itself. When such a
coexistence of vertex potentials exists, the physiologic vertex
waves are usually of longer duration and of somewhat higher
voltage than the abnormal spikes.
Prior to onset of stage 2 (light sleep), positive occipital sharp
transients of sleep may already be noticeable. This pattern, how-
ever, is less common and less prominent in children than in
adolescents and adults. A more detailed discussion is presented
in the section dealing with the sleep EEG in general.
Sleep spindles show the features of maturity, with a well-
defined vertex maximum. Its initial frequency of 14 Hz or 12
to 14 Hz may decrease to 10 to 12 Hz with deepening sleep;
the superior frontal electrodes (F3, F4) may show the spindles
somewhat better than the central electrodes (C3, C4). The spin-
dle activity maximum remains over the midline; a vertex
maximum may or may not be replaced by a maximum over
frontal midline (FZ) with deepening sleep (transition from
stage 2 to 3). The spindle trains are usually shorter than 1 second. Figure 10.15 Evolutional and involutional phases of the K complex.
The waves are rounded; the physiologic negative spiky spindle Left tracing, patient age 5 years. Prominent initiating sharp component.
character has disappeared much earlier (at the age of 2 years) Upper right tracing, patient age 8 years. Note very pronounced sharp
(Fig. 10.14). component of K complex. Far right, a single vertex wave is shown.
K complexes are often seen in association with spindles. The Lower right, patient age 47 years. K complex of moderate voltage; the
complex wave morphology of this pattern is extensively discussed initiating sharp component is markedly blunted. The slow component
in conjunction with sleep in general and again in Chapter 26. is inconspicuous; superimposed spindles are barely visible. This decline
K complexes and vertex waves represent fascinating responses to in the K complex suggests some cerebral pathology.
intrinsic or extrinsic stimuli discussion (Fig. 10.15).
REM sleep (rarely seen in a regular EEG laboratory) shows
less slow activity and increasing desynchronization with mixed dominantly seen in adolescents and young adults, but it is not
activities in the theta, alpha, and beta frequency range. uncommon in older children. The wave morphology of this dis-
Arousal from sleep is characterized by an increasingly shorter charge is presented in other sections. The psychomotor variant
transition from sleep to the waking state and decreasing length occurs almost exclusively in very light drowsiness and disap-
of high-voltage theta activity. pears with deepening level of awareness. This pattern has also
been called rhythmic mid-temporal discharges (80). This pat-
Variant Patterns of Uncertain Clinical tern does not have the evolution of frequencies and waveforms
Significance in Drowsiness and Sleep seen with an actual electrographic seizure discharge (76).
There are EEG waveforms that are now considered normal vari-
ants. These waveforms typically occur in a rhythmic manner,
with an admixed “epileptiform” morphology. These may repre- EEG IN ADOLESCENTS (13 TO 20 YEARS)
sent a superimposition of background frequencies (75). These
patterns are now referred to as normal variants, benign vari- General Considerations
ants, or patterns of uncertain clinical significance (76,77). Although the transition from childhood to puberty is a major
The 14- and 6-Hz positive burst is a common finding at this step in biologic development, EEG maturation shows no dra-
age. The sections on the sleep EEG and on abnormal (epileptic) matic changes in these years. In other words, there are no
patterns in the EEG discuss more extensively this type of striking differences between the record of a healthy teenager
activity, which was widely considered a genuine abnormality and a healthy older child. Nevertheless, there are a number of
after the first report of Gibbs and Gibbs (78). Subsequently, subtle changes.
accumulating evidence demonstrates that this pattern is not Eeg-Olofsson (4) carried out a special study of the EEG
categorically abnormal and is considered a normal variant development of normal adolescents and selected 185 persons
unless it appears in enormous abundance. from the age of 16 to 21 years, using the same strict criteria that
The rhythmic temporal theta bursts of drowsiness, previ- were applied to the younger children. Earlier studies mostly
ously known as the psychomotor variant pattern (45,79), is pre- included mixed ages, either adolescents or children.
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 177

Waking Record cents’ slow frequencies may show a wider distribution and their
The posterior alpha rhythm shows a mean frequency of around 10 peak over the posterior regions is not quite as impressive as in
Hz; according to Petersén and Eeg-Olofsson (6), the mean older children. “Slow fused transients” (61) are not necessarily
frequency lies at 10.2 Hz in the male and 10.3 Hz in the female. normal at this age.
The entire alpha range, as reported by Petersén and Eeg- A special form of posterior slow activity is the posterior 4- to
Olofsson, stretches from 8 to 12 Hz; these authors observed a case 5-Hz rhythm (Fig. 10.16), which probably constitutes a mild
of 8 Hz alpha rhythm in a healthy 17-year-old boy (this would abnormality. This pattern is uncommon, with probably only
raise the suspicion of a minimal abnormality, in the opinion of one case occurring in 1000 or 2000 tracings from a large EEG
some electroencephalographers). With the use of frequency laboratory. The mean age at first examination was 24.15 years
analysis, Samson-Dollfus and Goldberg (81) found mean alpha (65), which indicates that this is a pattern of adolescence and
frequencies from 9.64 to 9.94 Hz in subjects at the age of 15 years. young adulthood. Acquired cerebral pathology (especially cran-
The posterior alpha rhythm is of moderately higher ampli- iocerebral trauma), as well as genetic predisposition (65), may
tude than in adulthood, but a slight decline from the height of play a determining role.
alpha waves in childhood is noticeable. Alpha waves may show Another form of rhythmical posterior slow activity consists
a negative sharp component (classically present in mu rhythm) of rhythmical 3- to 4-Hz waves of high voltage over the occipital
that is due to a hidden or overt admixture of fast activity. Such region and its immediate vicinity. This is referred to as OIRDA,
sharp alpha waves are common in older children, adolescents, and it is activated by hyperventilation, eye closure after a latency
and young adults; this pattern is perfectly normal, a fact already period of 300 to 500 msec and lasts for about 1.5 to 3 seconds,
stressed by Gibbs and Gibbs (46). Amplitude asymmetries are drowsiness, and photic stimulation. The rhythmical 3- to 4-Hz
the rule, with amplitudes usually somewhat higher on the right waves exhibit a more or less obvious sharp component,
side but, according to Petersén and Eeg-Olofsson (6), the differ- sometimes a “notch” in the slow wave. This type of posterior
ence seldom exceeds 20% of the greater amplitude. slowing has been investigated by Belsh et al. (82). These authors
The admixture of posterior slow activity constantly dimin- found this pattern only in children and adolescents from the age
ishes during the period of adolescence. In general, the amount of 6 to 16 years. This pattern occurs in children and juveniles
of intermixed delta–theta activity (between alpha waves) lies with or without epileptic seizure disorder. One must agree with
around 10% to 15%; slightly or moderately higher figures are Belsh et al., who think that “such a discharge should not be
still acceptable as compatible with normalcy. Healthy adoles- interpreted as epileptiform activity,” although Riviello and

Figure 10.16 Patient age 21 years. An example of the


4- to 5-Hz rhythm over the posterior region. Note that
eye closure is followed by a few 12- to 13-Hz alpha
waves before a prominent posterior 4.5- to 5-Hz rhythm
emerges. This rhythm apparently vanishes shortly
before eye opening. The record was otherwise normal.
The patient was referred because of suspected psy-
chogenic seizures.
178 Part II ■ Normal EEG

Foley reported OIRDA as an ictal waveform (66). This pattern delta activity was found in 22% of the healthy adolescents stud-
is also known as the “phi rhythm” (83). ied by Petersén and Eeg-Olofsson (6). It should be noted that
Fast activity is more widely seen in adolescents than in older hyperventilation is a poorly standardized procedure in which
children. The maximum fast activity is most commonly found motivation and effort play a paramount role.
over the frontal areas. Well-defined and circumscript beta waves Intermittent photic stimulation shows essentially the mature
over the rolandic regions are seen, usually in conjunction with type with a peak driving response in the medium and fast rate
the central mu rhythm. of flicker (mostly 6 to 20 per second). In adolescents, the predis-
The rolandic mu rhythm is fairly common in adolescents and position to paroxysmal flicker responses must not be underrated;
gradually declines from a peak reached between the age of 11 such responses of the photomyoclonic as well as of the photo-
and 15 years (40). Occipital lambda waves show their typical convulsive type (87) may occasionally occur in apparently
mature configuration. Anterior rhythmical 6- to 7-Hz theta healthy individuals. Further investigation of such persons
activity gradually decreases from its peak prevalence (the age of sometimes unearths signs of mental, emotional, and autonomic
13 to 15 years; see Fig. 10.16). A rare phenomenon is rhythmical nervous system instabilities.
4-Hz activity over the vertex (“4-Hz vertex spindles”) described
by Van Huffelen and Magnus (84); this occurs in the waking Drowsiness
state only, mostly in adolescents, and especially in patients with The transition from wakefulness to sleep (stage 1) shows, in
vasomotor instability, such as syncope. general, the mature adult type with gradual alpha dropout and
Low-voltage records, very rare in childhood, are more com- long stretches of a diffuse low-voltage pattern with mixed slow
mon in adolescents and may occur in about 5% of patients; and fast activity. With deepening drowsiness, vertex waves
these records are usually dominated by beta frequencies. This appear, and positive occipital sharp transients of sleep may be
type of low-voltage fast record has been found more often in present.
females (6,85,86). In healthy adults, the prevalence of low-
voltage records increases to 11.6% (46). Sleep
In the commonly observed non-REM sleep onset recording in
Activations the EEG laboratories, there is little difference from the activity
Hyperventilation does not yield the dramatic high-amplitude found in adults. The sleep spindle amplitude is somewhat
slowing that is seen in children. Marked bilateral synchronous greater than in adulthood, and this is also generally true for this

Figure 10.17 Phantom spike–wave complex. The high-amplitude wave is the prominent component of this spike–wave
complex.
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 179

amplitude of vertex waves and K complexes. Their sharp REFERENCES


component is not quite as sharp as in older children. Positive
occipital transients of sleep may be abundant in stage 2. REM 1. Kellaway P. Ontogenetic evolution of the electrical activity of the
brain in man and animals. Fourth International Congress of
sleep and arousal from sleep show the mature type with very
Electroencephalography and Clinical Neurophysiology. Acta Med.
rapid transition from sleep patterns to waking activity.
Belg. 1957:141–154.
2. Berger H. Über das Elektroenzephalogramm des Menschen. 5.
Variant Patterns of Uncertain Clinical Mittlg. Arch. Psychiatr. Nervenkr. 1932;98:231–254. [English trans-
Significance lation: Gloor, P. 1969. Hans Berger. On the Electroencephalogram of
Man. Amsterdam: Elsevier (see pp. 157–160).]
The 14- and 6-Hz positive burst is not uncommon at this age.
3. Eeg-Olofsson O. The development of the EEG in normal children
The incidence starts to decline, as demonstrated by Gibbs and from age 1 to 15 years. The 14 and 6 Hz positive spike phenome-
Gibbs (45) (15.8% at the age of 5 to 9, 20.8% at 10 to 14, and non. Neuropaediatrie. 1971;4:405–427.
1.65% at 15 to 19 years; there is marked further decline to 4. Eeg-Olofsson O. The development of the electroencephalogram in
8.75% between the age of 20 and 24 years and 1.4% between 25 normal adolescents from the age of 16 through 21 years.
and 29 years). Eeg-Olofsson confirmed these findings (3). Deep Neuropaediatrie. 1971;3:11–45.
drowsiness and light sleep facilitate this pattern, which is most 5. Eeg-Olofsson O, Petersén I, Selldén U. The development of the
prominent over the posterior temporal and occipital regions. electroencephalogram in normal children from the age of 1
Lombroso et al (88) found 14- and 6-Hz positive bursts in 58% through 15 years. Paroxysmal activity. Neuropaediatrie. 1971;
of 13- to 15-year-old normal high school boys. They referred to 2:375–404.
them as ctenoids, after the Greek, for comb. 6. Petersén I, Eeg-Olofsson O. The development of the electroen-
cephalogram in normal children from the age of 1 through 15
The 6-Hz spike–wave complex, now typically referred to as the
years. Neuropaediatrie. 1971;2:247–304.
phantom spike and wave, is occasionally seen in adolescents; 7. Petersén I, Selldén U, Eeg-Olofsson O. The evolution of the EEG in
Gibbs and Gibbs (45) found this pattern in 2.8% of their 3476 normal children and adolescents from 1 to 21 years. In: Lairy GC, ed.
control subjects, a higher figure than at any other age. Handbook of Electroencephalography and Clinical Neurophysiology.
Considered a mild or moderate abnormality in the past, it is Vol 6B, ed.-in-chief, A. Remond. Amsterdam: Elsevier; 1975:31–68.
now considered a normal variant or a pattern of uncertain clin- 8. Matsuura M, Yamamoto K, Fukuzawa H, et al. Age development
ical significance. It is called the phantom spike and wave and sex differences of various EEG elements in healthy children
because the wave component has a much higher amplitude and adults. Quantification by a computerized wave form recog-
than the spike (Fig. 10.17). It is discussed in Chapter 26 with nition method. Electroencephalogr Clin Neurophysiol. 1985;60:
epileptic seizure discharges. 394–406.
The rhythmic temporal theta bursts of drowsiness, or the 9. Gasser T, Verleger R, Bacher P, et al. Development of the EEG of
school-age children and adolescents. I. Analysis of band power.
psychomotor variant pattern, is much less common and reaches
Electroencephalogr Clin Neurophysiol. 1988;69:91–99.
a slight peak at the age of 15 to 19 years (0.4%), according to 10. Gasser T, Jennen-Steinmetz C, Sroka L, et al. Development of the
Gibbs and Gibbs (45). EEG of school-age children and adolescents. II. Topography.
Electroencephalogr Clin Neurophysiol. 1988;69:100–109.
11. Pampiglione G. Development of Cerebral Function in the Dog.
CONCLUDING STATEMENTS London: Butterworths; 1963.
This chapter provides a condensed description of EEG matura- 12. Caveness WF. Atlas of EEG in the Developing Monkey (Macaca
tion from birth through young adulthood. The basic rules of this mulatta). Reading, MA: Addison-Wesley; 1962.
13. Passouant P, Cadilhac J, Delange M. Le sommeil du nouveauné.
EEG maturation are not carved in stone and must be viewed with
Considérations sur la période de mouvements oculaires. Arch Fr
a certain degree of flexibility. It is strongly recommended that Pediatr. 1965;22:1087–1092.
terms such as within broad (very broad) normal limits of variabil- 14. Ellingson RJ. EEGs of premature and full-term newborns. In: Klass
ity for age be used in mildly deviant but not definitely abnormal DW, Daly DD, eds. Current Practice of Electroencephalography. New
tracings, rather than making rigid distinctions between the “nor- York: Raven Press; 1979:149–177.
mal” and “abnormal” character of a given record. 15. Dreyfus C, Curzi-Dascalova L. The EEG during the first year of
The “clinician within ourselves” will demand statements life. In: Lairy GC, ed. Handbook of Electroencephalography and
concerning the clinical significance of mild deviations or defi- Clinical Neurophysiology. Vol 6B, ed.-in-chief, A. Remond.
nite abnormalities in the maturational process. The aforemen- Amsterdam: Elsevier; 1975:24–30.
tioned normal variant patterns, such as the 14- and 6-Hz 16. Sweeney D, Beckman D, Calmese F, et al. The use of modified sleep
positive spike discharge, rhythmic temporal theta bursts of deprivation to facilitate pediatric electroencephalographic record-
ings. Am J END Technol. 1997;37:218–230.
drowsiness, and 6-Hz spike–wave discharges, must be inter-
17. Brown V, Haak N, Egel RT. Technical tips: Age-specific sleep
preted in a prudent and circumspect manner. deprivation guidelines. Am J END Technol. 1997;37:231–235.
It is also very difficult to make a firm and solidly founded 18. Ong H-T, Lim KJL, Low P-C, et al. Simple instructions for partial
statement as to whether deviations from the rules of EEG mat- sleep deprivation prior to pediatric EEG reduces the need for seda-
uration (for instance, excessive posterior slowing at a given age) tion. Clin Neurophysiol. 2004;115:951–955.
are simply a sign of immaturity or a sign of possible sustained 19. Leonberg SC. Management of infants and children for electroen-
structural insult to the brain. cephalography. Clin Electroencephalogr. 1984;15:202–207.
180 Part II ■ Normal EEG

20. Epstein CM. Guideline Two: Minimum technical standards for 44. Westmoreland BF, Stockard JE. The EEG in infants and children:
Pediatric Electroencephalography. J Clin Neurophysiol. 2006;23: 92–96. normal patterns. Am J EEG Technol. 1977;17:187–206.
21. Fois A. The Electroencephalogram of the Normal Child. Springfield, 45. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 3.
IL: Charles C Thomas; 1961. Reading, MA: Addison-Wesley; 1964.
22. Stroganova TA, Orekhova EV, Posikera IN. EEG alpha rhythm in 46. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 1.
infants. Clin Neurophysiol. 1999;110:997–1012. Cambridge, MA: Addison-Wesley; 1950.
23. Hagne I. Development of waking EEG in normal infants during 47. Brandt H, Brandt S, Vollmund K. The electroencephalographic
the first year of life. In: Kellaway P, Petersén I, eds. Clinical patterns in young healthy children from 0 to five years of age. Acta
Electroencephalography of Children. New York: Grune & Stratton Psychiatr Scand. 1955;30:77–89.
(Stockholm: Almqvist & Wiksell); 1968:97–118. 48. Slater GE, Torres F. Frequency-amplitude gradient. A new param-
24. Hagne I, Persson J, Magnusson R, et al. Spectral analysis via fast eter for interpreting pediatric sleep EEGs. Arch Neurol.
Fourier transform of waking EEG in normal infants. In: Kellaway 1979;36:465–470.
P, Petersén I, eds. Automation of Clinical EEG. New York: Raven 49. Cadilhac J. Ontogenesis and phylogenesis of sleep. In: Passouant P,
Press; 1972:3–48. ed. Handbook of Electroencephalography and Clinical Neurophysio-
25. Ohtahara S. Development of electroencephalogram during logy. Vol 7A, ed.-in chief, A. Remond. Amsterdam: Elsevier; 1975:
infancy and childhood. Proceedings of the Japanese EEG Society. 18–25.
1964:18–23. 50. Gibbs EL, Gibbs FA. Extreme spindles: correlation of electroen-
26. Samson-Dollfus D, Nogues B, Delagree E. EEG recording during cephalographic sleep pattern with mental retardation. Science.
drowsiness in normal babies from 2 to 12 months of age. 1962;138:1106–1107.
Electroencephalogr Clin Neurophysiol. 1981;52:58P[abstract]. 51. Niedermeyer E, Capute AJ. A fast and spiky spindle variant in chil-
27. Curzi-Dascalova L, Monod N, Guidasci S, et al. Waking-sleeping tran- dren with organic brain disease. Electroencephalogr Clin
sition in the newborn baby and in infants before the age of 3 months. Neurophysiol. 1967;23:67–73.
Electroencephalogr Clin Neurophysiol. 1981;52:57P [abstract]. 52. Hess R. The electroencephalogram in sleep. Electroencephalogr
28. Metcalf DR. The effect of extrauterine experience on the ontogen- Clin Neurophysiol. 1964;16:44–55.
esis of EEG sleep spindles. Psychosom Med. 1969;31:393–399. 53. Kellaway P, Fox BJ. Electroencephalographic diagnosis of cerebral
29. Katsurada ML. Electroencephalographic study of sleep in infants pathology in infants during sleep. I. Rationale, technique and the
and young children. I. Development of spindle waves (in characteristics of normal sleep in infants. J Pediatr. 1952;41:
Japanese). Ann Paediatr Jpn. 1965;2:391–394. 262–287.
30. Lenard HC. The development of sleep spindles during the first two 54. White JC, Tharp BR. An arousal pattern in children with organic
years of life. Neuropaediatrie. 1970;1:264–276. cerebral dysfunction. Electroencephalogr Clin Neurophysiol. 1974;
31. Hughes JR. Development of sleep spindles in the first year of life. 37:265–268.
Clin Electroencephalogr. 1996;27:107–115. 55. Hughes JR, Daaboul Y. The frontal arousal rhythm. Clin
32. Niedermeyer E. The Generalized Epilepsies. Springfield, IL: Charles Electroencephalogr. 1999;34:13–14.
C Thomas; 1972. 56. Hughes JR. The frontal arousal rhythm (FAR) is an ictal pattern: a
33. Metcalf DR, Mondale J, Butler FK. Ontogenesis of spontaneous K case report. Clin Electroencephalogr. 2003;34:13–14.
complexes. Psychophysiology. 1971;8:340–347. 57. Niedermeyer E, Yarworth S. Scarcity of minor EEG abnormalities
34. Westmoreland BF. Electroencephalography: Adult, Normal, and during the first two years of life. Electroencephalography. 1978;
Benign Variants. Clinical Neurophysiology. 3rd ed. Oxford: Oxford 9:20–28.
University Press; 2009:119–136. 58. Leissner P, Lindholm LE, Petersén I. Alpha amplitude dependency
35. Ellingson RJ, Peters JF, Nelson B. Respiratory pauses and apnea on skull thickness as measured by ultrasound technique.
during daytime sleep in normal infants during the first year of life: Electroencephalogr Clin Neurophysiol. 1970;29:392–399.
longitudinal observations. Electroencephalogr Clin Neurophysiol. 59. Gibbs FA, Gibbs EL, Lennox WG. Electroencephalographic
1982;53:48–59. response to overventilation and its relation to age. J Pediatr. 1943;
36. Roffwarg HP, Muzio JN, Dement WC. Ontogenetic development 23:497–505.
of the human sleep dream cycle. Science. 1966;152:604–619. 60. Sunder TR, Erwin CW, Dubois PJ. Hyperventilation induced
37. Stern E, Parmelee AH, Akiyama Y, et al. Sleep cycle characteristics abnormalities in the electroencephalogram of children with
in infants. Pediatrics. 1969;43:65–70. Moya-Moya disease. Electroencephalogr Clin Neurophysiol. 1980;
38. Dittrichova J, Paul K, Pavlikova E. Rapid eye movements in para- 49:414–420.
doxical sleep in infants. Neuropaediatrie. 1972;3:248–257. 61. Kellaway P, Crawley JW, Kagawa N. A specific electroencephalo-
39. Schulz H, Salzarulo P, Fagioli I, et al. REM latency: development in graphic correlate of convulsive equivalent disorders in children.
the first year of life. Electroencephalogr Clin Neurophysiol. J Pediatr. 1959;55:582–592.
1983;56:316–322. 62. Aird RB, Gastaut Y. Occipital and posterior electroencephalographic
40. Niedermeyer E, Koshino Y. My-Rhythmus: Vorkommen und klin- rhythm. Electroencephalogr Clin Neurophysiol. 1959;11:637–656.
ische Bedeutung. Z EEG-EMG. 1975;6:69–78. 63. Kuhlo W. Slow posterior rhythms. In: Handbook of Electroenc-
41. Hrbek A, Vitova Z, Mares P. Optic evoked potentials in children ephalography and Clinical Neurophysiology. Vol 6A, Ed.-in-chief, A.
at different stimulation frequencies. Physiol Bohemoslov. 1966;15: Remond. Amsterdam: Elsevier; 1976:89–104.
201–204. 64. Attarian HP, Pacquiao PA, Erickson SM. Unilateral alpha subhar-
42. Vitova Z, Hrbek A. Developmental study on the responsiveness of monics: a case report. Clin Electroencephalogr. 2001;32:32–35.
the human brain to flicker stimulation. Dev Med Chid Neurol. 1972; 65. Kuhlo W, Heintel H, Vogel F. The 4–5/sec rhythm. Electroen-
14:476–486. cephalogr Clin Neurophysiol. 1969;26:613–618.
43. Lindsley DB. A longitudinal study of the alpha rhythm in normal 66. Riviello JJ, Foley CM. The epileptiform significance of intermittent
children: frequency and amplitude standards. J Genet Psychol. 1939; rhythmic delta activity (IRDA) in childhood. J Child Neurol. 1992;
55:197–213. 7:156–160.
Chapter 10 ■ Normal EEG and Sleep: Infants to Adolescents 181

67. Palmer FB, Yarworth S, Niedermeyer E. Frontal midline theta 78. Gibbs EL, Gibbs FA. Electroencephalographic evidence of thala-
rhythm. Electroencephalogr Clin Neurophysiol. 1976;7:131–138. mic and hypothalamic epilepsy. Neurology (Minneapolis). 1951;1:
68. Doose H, Gerken H, Horstmann T, et al. Genetics of centren- 136–144.
cephalic epilepsy in childhood. Epilepsia (Amsterdam). 1968;9: 79. Gibbs FA, Rich CL, Gibbs EL. Psychomotor variant type of seizure
107–115. discharge. Neurology. 1963;13:991–998.
69. Doose H, Gerken H, Horstmann T, et al. On the genetics of EEG 80. Lipman IJ, Hughes JR. Rhythmic mid-temporal discharges. An
anomalies in childhood. I. Abnormal theta rhythms. Neuropaediatrie. electro-clinical study. EEG Clin Neurophysiol. 1969;27:43–47.
1972;3:386–401. 81. Samson-Dollfus D, Goldberg P. Electroencephalographic quantifi-
70. Doose H, Gerken H, Horstmann T, et al. Genetic factors in spike- cation by time domain analysis in normal 7–15 year old children.
wave absences. Epilepsia (Amsterdam). 1973;14:57–75. Electroencephalogr Clin Neurophysiol. 1979;46:147–154.
71. Schauseil-Zipf U, Hamm W, Mandl-Kramer S. EEG findings in 82. Belsh JM, Chokroverty S, Barabas G. Posterior rhythmic slow activ-
“small-for-date babies” at 7–12 years of age. Electroencephalogr ity in EEG after eye closure. Electroencephalogr Clin Neurophysiol.
Clin Neurophysiol. 1984;57:33P–34P[abstract]. 1983;56:562–568.
72. Daute KH, Frenzel J, Klust E. Über den unspezifischen 83. Silbert PL, Radhakrishnan K, Johnson J, et al. The significance of the
Hyperventilationseffekt im EEG des gesunden Kindes. I. Stärkegrad. phi rhythm. Electroencephalogr Clin Neurophysiol. 1995;95: 71–76.
Z Kinderheilkd. 1968;104:197–207. 84. Van Huffelen AC, Magnus O. 4c/sec vertex spindles. Electroen-
73. Herrlin KM. EEG with photic stimulation: a study of children with cephalogr Clin Neurophysiol. 1973;34:543–546.
manifest or suspended epilepsy. Electroencephalogr Clin Neurophysiol. 85. Mundy-Castle AC. Theta and beta rhythm in the electroencephalo-
1954;6:573–589. gram of normal adults. Electroencephalogr Clin Neurophysiol.
74. Doose H, Gerken H, Hien-Volpel KIF, et al. Genetics of photosen- 1951;3:477–486.
sitive epilepsy. Neuropaediatrie. 1969;1:56–73. 86. Vogel F, Fujiya Y. The incidence of some inherited EEG variants
75. Klass DW, Westmoreland BF. Nonepileptogenic epileptiform elec- in normal Japanese and German mates. Hum Genet. 1969;7:
troencephalographic activity. Ann Neurol. 1985;18:627–635. 38–42.
76. Westmoreland BF. Benign EEG variants and patterns of uncertain 87. Bickford RG, Sem-Jacobsen CW, White PC, et al. Some observa-
clinical significance. In: Ebersole JS, Pedley TA, eds. Current tions of the mechanisms of photic and photo-metrazol activation.
Practice of Clinical Electroencephalography. 3rd ed. Philadelphia: Electroencephalogr Clin Neurophysiol. 1952;4:275–282.
Lippincott Williams & Wilkins; 2003:235–245. 88. Lombroso CT, Schwartz IH, Clark DM, et al. Ctenoids in healthy
77. Tatum WO, IV, Husain AM, Benbadis SR, et al. Normal adult EEG youths: controlled study of 14 and 6 per second positive spiking.
and patterns of uncertain significance. J Clin Neurophysiol. 2006; Neurology. 1966;16:1152–1158.
23:194–207.
CHAPTER
Normal EEG and Sleep:
Adults and Elderly
BERNARD S. CHANG, DONALD L. SCHOMER, AND ERNST NIEDERMEYER
11
INTRODUCTION The frequency range of the EEG has a fuzzy lower and upper
limit. There are ultraslow and ultrafast frequency components
There is perhaps nothing as compellingly beautiful to a clinical that play no significant role in the clinical EEG, with the
neurophysiologist as the appearance of a normal adult elec- exception of ultraslow activity in profound coma and near-ter-
troencephalogram (EEG) recording, in wakefulness and sleep. minal states. For these reasons, the frequency-response curve of
But the relative order, symmetry, and transitions that populate an EEG apparatus concentrates on the clinically relevant fre-
such a recording and are so welcome to the clinically trained eye quency range, which is also the most important from the psy-
can lead us to forget the fundamentally remarkable phenomena chophysiologic viewpoint. This range lies between 0.1/sec (or
that electrical activity from the human brain can indeed be cycles per second [cps] or Hz) and 100/sec and, in a more
measured routinely by electrodes placed only on the scalp; that restricted sense, between 0.3 and 70/sec. In the normal adult,
defined, well-catalogued abnormalities in the resulting meas- the slow ranges (0.3 to 7/sec) and the very fast range (above
urements can be identified and interpreted with clinical mean- 30/sec) are sparsely represented; medium (8 to 13/sec) and fast
ing; and that our entire understanding of this field has (14 to 30/sec) ranges predominate.
essentially arisen within only the past century. These frequencies are broken down into the following bands
A chapter on the normal EEG is actually more difficult to or ranges:
organize than it might seem. Many clinical electroencephalog-
raphers would define normality on an EEG as being simply the Delta below 3.5/sec
absence of identifiable abnormalities. In other words, there is Theta 4 to 7.5/sec
no single rhythm, feature, or characteristic of an EEG that Alpha 8 to 13/sec
must be present to make it normal (except, perhaps, reactiv- Beta 14 to 30/sec
ity), and, indeed, no combination of any such findings either. Gamma above 30/sec
Such is the nature of human EEG, and this must be kept in The sequence of these Greek letters is not logical and can be
mind in the clinical interpretation of tracings. Having said understood only in the historical view. The terms alpha and
that, decades of EEG study, beginning with Berger’s early fun- beta rhythm or waves were introduced by Berger (1); the term
damental descriptions and continuing now with advanced gamma rhythm was subsequently used by Jasper and Andrews
power spectral analyses, have shown us that there are certain (2) to designate frequencies above 30 or 35/sec; these were
basic recognizable patterns, seen at least some of the time in essentially 35 to 45/sec and superimposed on the occipital alpha
almost all healthy individuals, that together constitute what rhythm (see Ref. 3). This term was temporarily abandoned, and
we would consider to be representative of a normal EEG. It is gamma frequencies became a part of the beta range.
the goal of this chapter to define and describe these recogni- The use of the term gamma rhythm or gamma frequency
zable elements, with attention to their clinical implications and range made an impressive comeback during the 1990s. The use
their significance for our understanding of underlying cerebral of a fast beta range and a very fast gamma range might have
physiology. been convenient for those who utilize frequency analysis with
power spectra. Furthermore, EEG rhythm research in the 1990s
EEG FREQUENCIES unearthed the all-but-forgotten term gamma rhythm (4–7). In
recent EEG rhythm research, rhythmic activities of the brain are
The EEG contains a wide frequency spectrum, but it is not often conceived mainly as induced rather than as spontaneous
simply a hodgepodge of frequencies. Rhythmicity seems to rhythms.
create some law and order among waves of various lengths The term delta rhythm was introduced by Walter (8) to des-
and amplitudes. The impression of prevailing rhythmicity and ignate all frequencies below the alpha range. Walter himself,
organization, however, is not a yardstick for the normality of however, found a need to introduce a special designation for the
an EEG. Pronounced rhythmicity may be a sign of abnorma- 4 to 7.5/sec range and used the letter theta. He thus bypassed
lity, and a chaotic appearance does not necessarily imply the Greek letters epsilon, zeta, and eta; he chose theta to stand
abnormality. Reactivity may be the magic word in such cases; for thalamus because he presumed a thalamic origin of these
an EEG of mixed frequencies may be quite responsive to cer- waves (also see Ref. 9).
tain stimuli.

183
184 Part II ■ Normal EEG

The term pi rhythm has been used for the designation of whereas the vast majority of low-voltage records are “desyn-
posterior slow rhythms (3 to 4/sec) without harmonious rela- chronized” (discussed later) and are a variant of normalcy.
tionship to the posterior alpha rhythm according to Dutertre
(3) who recommended the preferable (although certainly less
BASIC EEG PATTERNS SEEN
precise) term posterior slow rhythms. Hardly anyone has used
the term pi rhythm since the 1990s. IN WAKEFULNESS
The term phi rhythm was suggested (according to Ref. 10) for Alpha Rhythm
the designation of monorhythmic posterior delta waves (less
than 4/sec), distinct from the background and occurring within Definition
2 seconds of eye closure. This rhythm was also described by The International Federation of Societies for Electro -
Belsh et al. (11) as “posterior rhythmic slow activity after eye encephalography and Clinical Neurophysiology (IFSECN)
closure.” (committee chaired by G. E. Chatrian; see Ref. 18) proposed the
The alpha-like anterior temporal kappa rhythm (12) is a con- following definition of alpha rhythm (Fig. 11.1):
troversial pattern that is discussed later in this chapter. Kugler Rhythm at 8 to 13 Hz occurring during wakefulness over the
(13) has been using the term sigma activity instead of sleep spin- posterior regions of the head, generally with higher voltage over
dles and, furthermore, the term sigma rhythm for activity in the the occipital areas. Amplitude is variable but is mostly below 50
11 to 15/sec range. The term rho waves was used for the activity µV in adults. Best seen with eyes closed and under conditions of
known as positive occipital sharp transients of sleep (POSTS) physical relaxation and relative mental inactivity. Blocked or
(14), but it has disappeared. attenuated by attention, especially visual, and mental effort (18).
Other Greek letters have been proposed for the designation This committee also has pointed out that the term alpha
of distinct EEG activities. Mu rhythm and lambda waves are rhythm must be restricted to rhythms fulfilling all of the above
discussed in this chapter. The term tau rhythm denotes a phys- criteria. Rolandic mu rhythm may have the same frequency
iologic alpha rhythm of the temporal region (perhaps identical range, but its topography and reactivity are different (also
with the “third rhythm” discussed in this chapter). “Zeta wave” see Ref. 19).
simply denotes a certain type of delta wave (rather than a
rhythm) with some sharp configuration (15,16). Frequency
Thus, 14 of the 23 letters of the Greek alphabet are being EEG maturation shows the gradual frequency increase of a pos-
used in the EEG terminology, and this number could be even terior basic rhythm that is detectable around the age of 4 months
higher. In our opinion, it might be better to limit the Greek with a frequency of approximately 4/sec. This posterior
terms to the classical EEG frequency ranges retaining solely the dominant rhythm shows a progressive frequency increase with
letters alpha, beta, gamma, delta, and theta.

EEG AMPLITUDES
The EEG denotes voltage plotted against time. The voltage of
the EEG signal determines its amplitude. The passage of the
cortical EEG signal through leptomeninges, cerebrospinal fluid,
dura mater, bone, galea, and scalp has a strongly attenuating
effect on the original signal (17). Corticographic discharges
show amplitudes of 500 to 1500 µV (0.5 to 1.5 mV) and several
millivolts in prominent spiking. The amplitudes of the scalp
EEG are markedly reduced and lie between 10 and 100 µV (in
adults, more commonly between 10 and 50 µV).
The EEG amplitudes are measured from peak to peak.
Precise determination of the voltage of each wave is unneces-
sary and should be discouraged as pseudoaccuracy; too many
variables are involved (above all, the interelectrode distance and
the type of montage, whether bipolar or referential recording).
Electroencephalographers may indicate in their reports a cer-
tain amplitude range, such as “alpha rhythm from 20 to 30 µV,”
or, even better, limit themselves to statements such as “of
medium voltage” or “of low to medium voltage.” Figure 11.1 Normal record in a patient aged 27 years. Posterior 10 to
A given frequency can be rendered abnormal by excessive 12/ sec alpha rhythm. Good blocking response to eye opening and reac-
voltage. This is true for all frequencies, and it is particularly tivation of alpha rhythm with eye closure. Note low-voltage fast activ-
important for the fast (beta) band. The problem of low voltage ity and some return of alpha rhythm while eyes are kept open. Also
will be thoroughly discussed, because low amplitudes can indi- note typical artifacts with eye opening, eye closure, and eye blink
cate a life-threatening decline of cerebral voltage output, (caused by eye potential shifts).
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 185

average values of around 6/sec at the age of 12 months and 8/sec (28) found amplitudes between 20 and 60 µV in 66% of their
at the age of 3 years. At that time, the alpha frequency band is subjects; values below 20 µV were found in 28% and above 60
reached, and there is justification for the use of the term alpha µV in 6% (also see Ref. 29).
rhythm. The frequency reaches a mean of about 10/sec at the age Higher alpha amplitudes are more likely to be found in asso-
of 10 years. This is essentially the mean alpha frequency of adult- ciation with slower alpha frequencies (30,31). There is good
hood; in other words, the progressive alpha rhythm acceleration evidence of a mild-to-moderate alpha amplitude asymmetry
usually ends around the age of 10 years, but the second decade with a higher voltage on the right (20,27,29,31–33). This seems
of life (and to some degree also the third decade) features a con- to indicate that the alpha rhythm is of greater amplitude over
stant decline of intermixed posterior slow activity that is usually the nondominant hemisphere, but no convincing conclusion
present in considerable quantity at the age of 10 years. concerning handedness can be derived from this asymmetry
The frequency of the alpha rhythm tends to decline in eld- (20). This physiologic asymmetry has been confirmed by
erly individuals. This decline apparently reflects some degree of Matousek et al. (34), who also found a reversal of this rule (i.e.,
cerebral pathology, which may be vascular or degenerative, in higher voltage on the left occiput in patients with endogenous
many instances. Healthy and vigorous elderly people may show depression).
little or no alpha frequency decline, even in the ninth decade. Amplitude asymmetries must be demonstrated in both ref-
The figure of 10.2 ± 0.9/sec has been indicated as the mean erential and bipolar montages from two or more posteriorly
adult alpha frequency (20). An element of instability of the placed electrodes (such as parietal and posterior temporal)
alpha frequency must be taken into consideration; according to before they are considered significant (19). It may be difficult to
Townsend et al. (21), the alpha rhythm frequency can be stabi- distinguish between physiologic and truly abnormal alpha
lized by sinusoidally modulated light. Extreme upward gaze amplitude asymmetries.
tends to facilitate the posterior alpha rhythm (22,23). Lateral
eye deviations may have similar effects (24). Morphology
An alpha rhythm frequency shift to the faster portion of the The alpha rhythm is usually characterized by rounded or sinu-
band is not uncommon and is essentially within normal limits, soidal wave forms. However, a sizable minority of individuals
as will be discussed later. The similarities between the frequen- have sharp alpha configuration. In such cases, the negative
cies of alpha rhythm and the physiologic finger tremor have component appears to be sharp and the positive component
been discussed by Isokawa and Komisaruk (25). Immediately appears to be rounded, similar to the wave morphology of
after eye closure, the alpha frequency may be accelerated for a rolandic mu rhythm.
moment (“squeak effect,” after Ref. 26). The sharp configuration of posterior alpha waves is by no
means an abnormality. It is a common finding, especially in
Amplitude young adults, adolescents, and older children. An admixture of
Alpha rhythm amplitudes vary considerably from individual to beta waves is usually the cause of the sharp configuration; drug
individual and, in a given person, from moment to moment. effects from sedatives or minor tranquilizers must sometimes
The electroencephalographer, therefore, should look for be suspected in such cases.
stretches of optimal alpha output. A referential montage to the
ipsilateral ear is usually most suitable for the determination of Spatial Distribution
the alpha rhythm amplitude, but the interelectrode distances The alpha rhythm is clearly a manifestation of the posterior half
must always be considered. The maximum alpha voltage is usu- of the head and is usually found over occipital, parietal, and
ally over the occipital region as such, but a bipolar montage posterior temporal regions. This observation of Adrian and
with a parasagittal array may obscure rather than reveal the true Matthews (35) was doubted by Berger (36), whose concept of
alpha maximum. The alpha amplitude may be quite small in alpha rhythm as a global cerebral rhythm was an erroneous
the channels displaying P3–O1 and P4–O2 because of massive conclusion from his fronto-occipital bipolar recording tech-
homophasic activity, which results in canceling out. nique. The alpha rhythm may extend into central areas, the ver-
The alpha amplitudes tend to show constant waxing and tex, and also the midtemporal region. When the central region
waning. For this reason, trains of alpha waves show a typical is strongly involved, the alpha rhythm must be distinguished
spindle shape with a belly and a thin portion. However, the from possibly coexisting rolandic mu rhythm. This is usually
term spindle has been reserved for a classical pattern of sleep easily demonstrable with eye opening, which should lead to
and should not be used in this context. attenuation of the alpha rhythm but not the mu rhythm.
Berger (1) found alpha rhythm voltages of 15 to 20 µV; these The alpha rhythm may occasionally extend slightly into the
are small values when one considers his fronto-occipital record- superior frontal leads (F3, F4). Extension into the frontopolar
ing technique; the smallness was probably due to the limitations region (Fp1, Fp 2) is practically unheard of. Apparent alpha
of his Edelmann string galvanometer. According to Cobb (27), rhythm in the frontopolar leads may be very prominent in ref-
the alpha rhythm voltage fluctuates between 0 and 40 to 50 µV erential (unipolar) montages if the referential ear electrode
in the individual record; values above 100 µV are uncommon in picks up the posterior alpha rhythm. This is particularly com-
the adult, whereas maximums of 5 to 10 µV are frequently seen mon when the mastoid region is used instead of the ear lobe
(27). The work of Simon (also known as Simonova) and her (the mastoid being a preferred place with paste technique).
coworkers has shed more light on this subject; Simonova et al.
186 Part II ■ Normal EEG

Another source of confusion is eyelid flutter, with closed eyes No EEG is complete without certain reactivity tests.
giving rise to frontal artifacts in alpha frequency. Although alpha rhythm disappears with the earliest approach
In depth electroencephalography and with occipital of drowsiness, there are exceptional cases of persisting posterior
implants, posterior alpha rhythm can be demonstrated alpha rhythm in profoundly comatose patients with pontine
throughout the depth of the occipital lobe and even in the vascular lesions; their alpha rhythm shows no reactivity, even
vicinity of the lateral geniculate body. According to Albe- with strong nociceptive stimuli. According to Kiloh et al. (33),
Fessard (37), alpha rhythm may be recorded from the medial “complete unresponsiveness of the alpha rhythm to visual stim-
pulvinar but not from the remaining thalamic nuclei in the uli is a rare and unequivocally abnormal finding.”
human (also see Ref. 38). In the dog, Lopes da Silva et al. (39)
demonstrated that alpha rhythm of the same peak frequency, Interindividual Differences
bandwidth, and reactivity can be recorded from the visual cor- When Adrian and Matthews demonstrated their own EEGs on
tex as well as from the visual thalamus (lateral geniculate body, May 12, 1934, at a meeting of the astonished members of the
pulvinar nuclei). Physiological Society in Cambridge, England, it was found
that Adrian’s 10/sec alpha rhythm was quite impressive,
Reactivity whereas Matthews produced “no regular waves” (49). Studies
The posterior alpha rhythm is temporarily blocked by an of further subjects showed that Adrian’s alpha development
influx of light (eye opening), other afferent stimuli, and men- was that of the majority, whereas Matthews, the ingenious
tal activities. The degree of reactivity varies; the alpha rhythm engineer and designer of Adrian’s instrumentation, belonged
may be completely blocked, suppressed, or attenuated with to a minority of persons with little or no organized alpha
voltage reduction. The alpha blocking response to eye opening rhythm.
was discovered by Berger and described in his first report Davis and Davis (50) distinguished four types of records: (a)
(1) on the human EEG; it came as a great surprise for investi- dominant alpha (found in 20% of healthy adults); (b) subdom-
gators who were searching for action potentials and hence inant alpha (35%); (c) mixed alpha (20%); and (d) rare alpha
would have expected enhancement of EEG voltage with influx (25%). Golla et al. (51) distinguished three alpha types: M for
of light. Berger’s own explanation was the concept of a zone of minus or minimal, P for persistent, and R for responsive. The P
inhibition surrounding the area of excitation by the afferent type shows no real persistence of alpha, but has a very short
stimulus (40,41). blocking response to eye opening (also see Ref. 33). Another
It is noteworthy that in some subjects the alpha suppression type of alpha rhythm is the “monotonous high voltage alpha,”
is more pronounced than in others. These persons may have an which shows little or no waxing and waning of the amplitude
alpha-free stretch of desynchronized and chiefly fast EEG activ- (52). These alpha traits may be, to some degree, genetically
ity for the duration of the eye opening, even in rather dim light. transmitted (50,53,54); marked similarities have been reported
In others, the alpha blockage lasts for less than 1 second. in identical twins. Small differences in the EEG of males and
According to Gibbs and Gibbs (42) “after the eyes have been females have been reported by Veldhuizen et al. (55).
held open for a few minutes, low voltage alpha waves … usually A detailed review of historical EEG literature on the alpha
reappear, unless the subject continues to look at something rhythm inevitably uncovers multiple publications suggesting a
which holds his interest.” The amplitude ratio between eyes relationship, or lack thereof, between alpha rhythm traits and
closed (well-developed alpha) and eyes open (beta of much particular personality features (56–61), with little evidence of
smaller voltage) declines with advancing age (43). reproducible findings. Similarly, attempts at simple correlations
Alpha attenuation due to auditory, tactile, and other between alpha rhythm and intelligence have largely been futile
somatosensory stimuli or heightened mental activity (such as or unconvincing (36,45,62–67), but with the use of power spec-
solving difficult arithmetical problems) is usually less pro- tral analysis, Gasser et al. (68) found good correlations between
nounced than the blocking effect with eye opening. Indeed, alpha rhythm (plus other EEG criteria) and intelligence, and
the blocking or attenuating effect of mental arithmetic on the individuals who are faster in retrieving information from mem-
posterior alpha rhythm may not be present in most cases (44). ory tend to have higher alpha rhythm frequencies (69).
In a study done in 1280 patients (598 with normal EEG
tracings, 682 with various degrees of EEG abnormality), alpha Intraindividual Differences
suppression or attenuation with calculation (serial sevens) A person’s EEG traits and alpha rhythm development must not
was noted in only 21 (1.6%) of the patients. On the other be considered as permanent and unchangeable features like fin-
hand, Berger (40,45–48) showed fine examples of arithmetic- gerprints. Every EEG tracing of some length gives testimony to
induced alpha blocking. A possible explanation for the a certain degree of variability, even within the state of relaxed
discrepancy lies in the difficulty of the task: the arithmetical vigilance. This is probably the effect of a large number of phys-
problems given to Berger’s two teenage children, Klaus iologic and psychophysiologic variables. To mention just one
and Ilse, were more challenging. Other factors may also be example, the waxing phase of alpha amplitudes has been related
at work: even simple arithmetic may cause alpha blocking if to the waning phase of afterimages (70). With advancing age,
the subject shows great motivation trying to please the exam- the alpha frequency tends to decrease (71) but this is probably
iner. With a nonchalant approach, there is usually no alpha the effect of cerebral pathology (vascular or degenerative)
blocking. occurring at old age.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 187

Circadian studies of the EEG have been done sparingly Body temperature also modifies the alpha frequency; the
(72,73) and with inconclusive results (for further data, see alpha rhythm accelerates with increasing body temperature
Ref. 74). The menstrual cycle has been thought to influence the (75). Therapeutic hyperthermia (up to 41°C) in cancer patients
EEG and especially the alpha rhythm frequency; Harding and slows down the EEG to the delta range and depresses the over-
Thompson (74) have summarized the work in this field as fol- all EEG output (76).
lows:
Preovulatory phase (days 5 to 14): Alpha frequency increased, Relationship to Vigilance and Anxiety
amount of beta increased, photic driving reduced. Alpha rhythm is the classical EEG correlate for a state of relaxed
Postovulation or luteal phase (days 15 to 23): Alpha slower, wakefulness best obtained with the eyes closed. A degree of higher
amount of alpha increased, less beta and more theta activity, alertness attenuates or suppresses the alpha rhythm, which is then
photic driving increased. supplanted by “desynchronized” low-voltage fast activity.
Premenstrual phase (days 23 to 28): Alpha frequency increased, The earliest stage of drowsiness is characterized by “alpha
amount of alpha reduced, more beta, less theta activity. dropout” (Fig. 11.2). The trains of alpha waves become less and
Menstrual phase (days 1 to 5): Alpha frequency slowed and less continuous, and the last alpha fragments finally give way to
amount increased, less beta, more theta activity. a low-voltage pattern of mixed slow (mostly theta range) and
fast frequencies. This type of alpha dropout is a hallmark of a

Figure 11.2 A–C: Three examples of alpha dropout at the earliest stage of
drowsiness in normal adults. The close-up shows clearly the mixture of low-
voltage fast and slow activity following alpha dropout. This is different from
the low-voltage fast activity in the alpha blocking response to eye opening and
heightened alertness.
188 Part II ■ Normal EEG

normal adult EEG; in children and infants, various types of further insight into the possibility of multiple sources of alpha
slow patterns appear. In adults with neurodegenerative disease, generation (99). Further work on interhemispheric phase dif-
a posterior alpha rhythm of normal appearance may be ferences between alpha waves has been carried out with topo-
replaced by activity in the theta and delta range; a special pat- scopic analysis (100) and cross-correlation technique (101).
tern with rhythmical widespread (but mainly parietotemporal) Posterior alpha generation and spread in posteroanterior
5 to 7/sec activity has been individualized recently by direction (97) is generally acknowledged, but has been chal-
Westmoreland and Klass (77), who consider this pattern lenged by Inouye et al. (102), who feel that alpha spread occurs
“benign.” in an anteroposterior direction in the dominant as well as in the
Voluntary control of the alpha rhythm and the use of alpha nondominant hemisphere, based on the method of entropy
feedback methods have been widely discussed topics since the analysis.
late 1960s. This work was presumably prompted by the obser- Our comprehension of alpha-rhythm genesis has not strik-
vation of well-modulated alpha during meditation practiced by ingly increased in past several decades. It may be assumed that
yogis (78,79) or zen buddhists (Hirai, 1968, and Kasamatsu and there are corticocortical and thalamocortical systems that inter-
Hirai, 1966, both articles cited in Ref. 80). Nowlis and Kamiya act in the generation of cortical alpha rhythms (103). From the
(81) and Brown (82) associated alpha rhythm enhancement experimenter’s as well as from the clinical electroencephalogra-
(on a “voluntary” basis) with a pleasant mood. Further work in pher’s viewpoint, there is good reason to presume that alpha
this field was done by Wallace (83), Wallace et al. (84), and rhythm is most definitely a cortical phenomenon but there has
Banquet (85), who studied states of transcendental meditation. been, thus far, no evidence of a synchronizing mechanism at the
Alpha amplitudes increased or decreased; in some subjects cortical level. No single neurophysiologic alpha rhythm theory
there were periods of low-voltage theta activity. Knott (67) feels has yet found general acceptance, and there are still uncertain-
that such states are in essence periods of drowsiness and doubts ties about the origin and psychophysiologic significance of this
the validity of the view that the high alpha state is a desirable remarkable phenomenon. And yet, our insights into the nature
condition. According to Stigsby et al. (86), there is no consistent of the alpha rhythm (and other EEG rhythms) have been deep-
EEG pattern associated with successful or unsuccessful tran- ening.
scendental meditation. In the end, the alpha rhythm as studied
here may simply reflect the level of vigilance (see Ref. 87). Mu Rhythm
Because a relaxed waking state is the optimal condition for the Rolandic (central) mu rhythm is in frequency and amplitude
posterior alpha rhythm, however, it is hence reasonable to related to the posterior alpha rhythm, but its topography and
assume that emotional tension attenuates or blocks the alpha physiologic significance are quite different (Fig. 11.3).
rhythm. This seems to hold true for the state of emotional ten- Historically, the existence of a special central rhythm was pre-
sion in patients or subjects with pending litigation, after a head sumed by Jasper and Andrews (2) (“precentral alpha rhythm”),
injury, or after other forms of physical damage, for example Maddocks et al. (104) (“alphoid activity”), and Schütz and
(88). Müller (105) (“high voltage rolandic alpha”). The features of
mu rhythm were first described in detail by Gastaut et al. (106)
Cerebral Generators of the Alpha Rhythm and Gastaut (107), who also included electrocorticographic
The alpha rhythm is of cortical origin, but the theory of a thal- tracings; these authors introduced the term rhythme rolandique
amic pacemaker function has frequently surfaced since the en arceau. The epithet en arceau alludes to the arch-shaped wave
work of Berger (40), who presumed cortical genesis but thala- morphology, which has also prompted the term wicket rhythm
mic governance of the alpha phenomenon. Bishop (89) pro- (108). Magnus (109) used the term central alpha (see also Ref.
posed the concept of corticothalamic reverberating circuits, 2). Other terms are arcade rhythm (110), comb rhythm (27), and
and Andersen and Andersson (90) are the proponents of a thal- somatosensory alpha rhythm (111).
amic theory that is based on presumed similarities between Mu rhythm is not detectable in every mature subject; its
human alpha rhythm and experimental barbiturate spindle prevalence is limited unless the “hidden” mu rhythm is visual-
activity in animals. According to this theory, the alpha rhythm ized with special methods. Mu stands for motor; this rhythm is
is driven by presynaptic input to cortical neurons from the thal- strongly related to functions of the motor cortex, but the con-
amic level (also see Refs. 91 and 92). This concept has been tribution of the adjacent somatosensory cortex must not be
challenged by Lopes da Silva et al. (39,93). Watanabe (94) has ignored.
postulated a “somewhat loose but stable oscillator system” sub- Mu rhythm and associated beta activity over the sensorimo-
serving the generation of alpha rhythm. tor cortex have become a topic of special interest in the past two
Adrian and Yamagiwa (95) regarded the alpha rhythm as decades. Indeed, mu rhythm as an interesting clinical EEG phe-
cortical with maximal involvement of the visual area. nomenon has transcended into a powerful contributor to the
Important new vistas were opened with the demonstration of understanding of motor activity in general. This development
some degree of interhemispheric asynchrony between alpha started with the work of Pfurtscheller on event-related desyn-
waves (96). It has been presumed that there is more than one chronization (ERD) (112,113). The ERD pertains even more
alpha generator within the posterior regions of the cerebrum strongly to the rolandic mu rhythm (and furthermore to vari-
(97); this was further substantiated by depth EEG studies in the ous cognitive tasks and their cortical EEG accompaniment)
human (98). The technique of chronotopography has added (114–119). In other words, the ERD has become a new criterion
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 189

healthy young male adults, Gastaut et al. (125,126) found mu


rhythm in 14.4%. Dongier and Dongier (127) noted mu
rhythm in 18% of a population of neurotic patients. Figures of
12% were reported by Picard et al. (128) and Simonova et al.
(129). In the patients of Niedermeyer and Koshino (130), the
prevalence of mu rhythm was 8.1% (182 of 2248); broken down
into age ranges, there were 9.0% between ages 0 and 10 years,
13.8% between 11 and 20 years, 8.4% between 21 and 40 years,
and 4.5% above 41 years. These authors demonstrated rolandic
mu rhythm in a 20-month-old child; this was thought to be an
exceptionally early manifestation of mu rhythm, although the
data of Stroganova et al. (131) demonstrated central mu
rhythm (during a state of attention) in the tracing of an 8-
month-old baby with a frequency of about 6 to 8.8/sec. The
authors speculated that mu rhythm tends to appear before the
occipital alpha-equivalent because, unlike visual stimulation,
somatosensory stimulation is present within the uterus. With
the use of frequency analysis, the prevalence of mu rhythm
reaches values close to 100% (132). Familial occurrence of mu
rhythm has been reported by Koshino and Isaki (133).

Morphology, Frequency, and Spatial Distribution


Older synonyms such as rhythme en arceau or wicket rhythm
pertain to the wave morphology; mu rhythm shows in most
instances a sharp (or spiky) negative and a rounded positive
phase. There are a few patterns with a monophasic spiky
appearance. Posterior alpha rhythm may have a similar config-
uration: “wicket spikes” have a similar appearance. The same is
true for 14 and 6/sec positive spikes, except for the spike posi-
tive and rounded negative component. All these patterns show
Figure 11.3 A: Rolandic mu rhythm, in a patient aged 13 years. Stretches a spatial distribution that is different from the mu rhythm or
of central mu rhythm while eyes are kept open. These runs occur asyn- occurs at levels of vigilance that are usually incompatible with
chronously on both sides and are blocked with clenching of right fist. mu rhythm. The amplitudes of mu rhythm are comparable to
Some posterior alpha rhythm persists after eye opening. Possible spread those of the posterior alpha rhythm.
of mu rhythm into the parietal area makes differentiation of alpha and Mu rhythm usually occurs in short stretches. Persons with
mu difficult in right parieto-occipital lead (eighth channel from the top). pronounced mu rhythm show long trains of mu rhythm. The
B: The close-up view of the same patient shows independent bilateral most common frequency of the mu rhythm is 10/sec.
central mu rhythm. Frequencies may lie below 9/sec and above 11/sec; a mu rhythm
of less than 8/sec is probably a mild abnormality. Mu rhythm is
often mixed with local activity around 20/sec. According to
for the assessment of cortical functioning, not only over the Storm van Leeuwen et al. (134), mu frequencies are slightly
motor cortex but also over various cortical regions of strong higher than alpha frequencies. The spatial distribution is essen-
afferent input related to neurocognitive activities. Further detail tially confined to the precentral–postcentral region; some spread
on the ERD is found in the work of Pfurtscheller and Lopes da into parietal leads is not uncommon. The C3 and C4 electrodes
Silva (120,121). are mostly involved; occasionally, a vertex (Cz) maximum is
noted. A special vertex mu rhythm with special reactivity (lower
Age and Prevalence limb) was described in aged patients by Farnarier et al. (135).
Central mu rhythm used to be considered scarce. The introduc- When recorded through bone defects, the mu rhythm is more
tion of the International Electrode System (10-20 system) has widely distributed. According to Kuhlman (111), the maximum
contributed to a much greater awareness of this pattern. The C3 lies over the postcentral rather than the precentral cortex.
and C4 electrodes are located over the precentral gyrus in an The alphoid (around 10/sec) and the fast (around 20/sec)
optimal location for picking up central mu rhythm. The use of component of the mu rhythm seem to be inseparably intermin-
a different electrode system revealed a prevalence of only 3.2% gled but, with more advanced technology, spatial separation is
(122) and 2.9% (123), figures that lie well below those found in possible. According to Salmelin and Hari (136,137) the beta
other studies. Gastaut et al. (108) found mu rhythm in 10% of component arises from the motor cortex and the alphoid com-
their adult patients; Beek (124) observed this rhythm in 13% of a ponent from the sensory cortex. These magnetoencephalo-
predominantly psychiatric patient population. In 500 essentially graphic findings have been confirmed by Nashmi et al. (138).
190 Part II ■ Normal EEG

In most persons with mu rhythm, the activity is bilateral but rolandic region contralateral to the site of movement; the effect
tends to shift from side to side. Coherence function studies have appears prior to the onset of muscular contraction
shown lack of bilateral coherence of mu rhythm in normal sub- (106,107,122,144,145). According to Chatrian et al. (145), there
jects (134). Strictly unilateral mu rhythm must be scrutinized are delays of 50 msec to 7.5 seconds (average, 1.5 seconds) at the
for the possibility of an ipsilateral rolandic disturbance as, for onset of the mu blocking effect after the initiation of the spon-
instance, an early stage of parasagittal meningioma, an arteri- taneous flexion of the contralateral thumb, with the ipsilateral
ovenous malformation, or other types of neoplasms (139). In lagging behind the contralateral response. Gastaut et al. (106)
such cases, even the possibility of a contralateral rolandic lesion and Ciganek (146) demonstrated a somatotopic distribution of
must be taken into consideration (107,140,141). Above all, the the blocking effect (according to the functional anatomy of the
possibility of a local cranial bone defect, surgical or traumatic, rolandic cortex), but this observation could not be confirmed
must be ruled out; a single burr hole in the rolandic region by Chatrian et al. (145).
enables otherwise hidden mu rhythm to become manifest on With the use of a gamma band (38 to 40/sec) frequency
the scalp (Fig. 11.4). Thus, mu rhythm represents at least a por- analysis focusing on the event-related synchronization (ERS) of
tion of rhythmical activity beneath bone defects (“breach the gamma component, Pfurtscheller et al. (119) beautifully
rhythm,” Ref. 142). demonstrated the locus for right and left index finger move-
ment, right toes, and the rather broad and bilateral area for
Reactivity tongue movements.
Mu rhythm is blocked by movements. These movements may The mu blocking response is likely to be related to the con-
be active (voluntary), passive, or reflexive (106,107,143–145). ceptual design of the movement to be executed. Mere thoughts
The blocking effect is bilateral but more pronounced on the about performing movements and readiness to move block the
mu rhythm (106,107,122,143–145). Mu blocking responses
have also been demonstrated in persons with amputations of
extremities; these mental activities concern movement of a
phantom limb (122,147).
Light tactile stimuli also produced a mu rhythm blocking
effect that is most evident over the contralateral rolandic region
(106,107,109,143–145). Kuhlman (111) has placed even greater
emphasis on the sensory component and regards mu rhythm as
the “idling” of the cortical sensory region, in analogy to the
alpha rhythm as “idling” of the visual cortex and its vicinity. His
work was carried out with power spectral analysis. This tech-
nique shows that central mu rhythm is a more common phe-
nomenon than one would expect from the visual analysis of the
scalp EEG (also see the power spectra study of Ref. 112). These
authors also showed simultaneous posterior alpha desynchro-
nization (112,113). Coherence functions have also been used
for the demonstration of rolandic mu rhythm (132,134).
It has been noted that rolandic mu rhythm is enhanced dur-
ing intermittent photic stimulation (148) and pattern vision
(149). The latter observation would contradict the basic law of
mu rhythm as a rhythm of immobility, but would be consistent
with extraocular movement control being neuroanatomically
distinct from the rolandic motor cortex.

Cerebral Generators of the Mu Rhythm


Jasper and Penfield (150) found evidence of a strictly localized
beta (around 20/sec) over the human motor cortex with
electrocorticographic recording technique in the locally
anesthetized patient. This rhythm could be blocked like mu
rhythm with movement, especially contralateral movement,
and also with thinking about the execution of movement. This
Figure 11.4 Patient aged 51, 2 years after evacuation of a right-sided rhythm is obviously the cortical equivalent of rolandic mu
subdural hematoma. A right-sided central mu rhythm shows admixture rhythm on the scalp. The mu waves on the scalp often show
of fast frequencies and spiky potentials. All components of this rhythm some notching or are imbedded in local fast activity, which is
show a prominent and persistent blocking response to contralateral also blocked by movement. Gastaut (107) confirmed the corti-
hand movements. (From Niedermeyer E, Koshino Y. My-Rhythmus: cal precentral fast activity in electrocorticographic tracings.
vorkommen und klinische bedeutung. Z EEG-EMG. 1975;6:69–78.) These data conflict with Kuhlman’s (111) observation of a
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 191

postcentral maximum of rolandic mu rhythm (scalp recordings and even single spikes may stand out. Children with benign
with power spectral analysis). On the other hand, Graf et al. rolandic epilepsy may gradually develop mu rhythm when
(151) demonstrated precentral 8 to 10/sec mu rhythm in the rolandic spike activity subsides and the seizures appear to be
electrocorticogram of an adult epileptic. under control. Interestingly, in some children with benign
As mentioned earlier, mu rhythm around 10/sec on the scalp rolandic epilepsy, cerebral spikes can be blocked by contralat-
and cortical rolandic 20/sec activity appear to be harmonically eral movements in the same manner as mu rhythm is blocked
related; such a relationship, however, does not exist between mu (165,166). This response is not present in all children with
rhythm and rolandic beta activity on the scalp according to rolandic spikes; this explains negative responses to motor activ-
Pfurtscheller (114) who used power spectral analysis. ity reported by Isch-Treussard (167).
Quantitative EEG studies have also established a clean separat- As with many other EEG patterns considered to be within
ing line between mu rhythm and posterior alpha rhythm (152). the range of normal, mu rhythm has been associated in the
A study of the functional significance of the mu rhythm historical literature with a predisposition to any number
from the human cortex was carried out with subdural elec- of neurologic and non-neurologic pathologic conditions,
trodes by Arroyo et al. (153). This work also showed the rela- including headache, hypertension, hyperthyroid states, psy-
tionship of the cortical mu rhythm to the well-known chiatric disorders, and even eczema (108,123,127,154,168), all
somatotopic arrangement of the sensorimotor cortex. with varying degrees of correlation and plausibility. It is
Three historical theories have governed the theoretical known that essentially all healthy adolescents and adults
approach to the mu rhythm: (a) the hypothesis of “neuronal harbor central mu rhythm when power spectra and coherence
hyperexcitability” (“hyperexcitabilité neuronique”), either dif- tests are utilized (132). Why then is mu rhythm evident in the
fuse or locally restricted to the rolandic cortex conventional EEG in just a minority of persons? It may be that
(106,107,110,124); (b) the hypothesis of superficial cortical their mu rhythm is more powerful than that of average
inhibition (“inhibition corticale superficielle”), comparable to persons in order to be picked up from the scalp. There is no
aforementioned theories of “cortical idling”; this theory would good evidence that this represents a clinically pathologic sign,
explain the suppression of mu rhythm with motor activity however.
(154–156,147); and (c) the hypothesis of somatosensory “corti- In cases of strictly unilateral mu rhythm, a careful search for
cal idling,” turning mu rhythm into an afference-dependent bone defects (burr holes) and/or local pathology is indicated. In
phenomenon (111). the wake of ischemic hemiplegic insults, central mu rhythm
may disappear or its reactivity may vanish (169).
Conditioning, Feedback Training,
and Relationship to Vigilance The “Third Rhythm” (Independent
An impressive international study on mu rhythm and the pha- Temporal Alphoid Rhythm)
sic evolution of conditioning was presented by Gastaut et al. Rhythmical activity in the alpha and upper theta range can be
(157). Mu rhythm feedback training has been used for various picked up from epidural electrodes over the midtemporal
purposes, including the treatment of epileptic seizure disor- region (170–174). This rhythm is not detectable in the scalp
ders (158–161). Roldan et al. (162) observed the development EEG unless there is a local bone defect in the midtemporal
of mu rhythm-like activity in the course of Hatha yoga exer- region where it has been termed (together with coexisting cen-
cises. This activity was called chi rhythm. It was believed to be tral mu rhythm in the vicinity) “breach rhythm” by Cobb et al.
a specific rolandic pattern of 11 to 17/sec waves and theta (142), who regarded the temporal activity as an abnormal
frequencies. Covello et al. (163) felt that several entities of mu rhythm. There is now good reason to presume that the rhyth-
rhythm exist, even in one subject, and that their underlying mical temporal lobe activity, which is clearly independent from
physiologic mechanisms may differ. Personal impressions the posterior alpha and rolandic mu rhythm, constitutes a
lend some support to this idea. Earlier observations (130) led physiologic rhythm.
to the conclusion that even the slightest degree of lessened The function of this rhythm is still debatable, however. Its
vigilance (i.e., earliest drowsiness) was incompatible with presence as an independent temporal alpha rhythm has been
mu rhythm; this view has been supported by Schoppenhorst demonstrated with the use of magnetoencephalography in
et al. (132). However, there have been some personal observa- healthy adults by Hari (personal communication, 1990, 1991),
tions of patients demonstrating their best stretches of central who felt that this rhythm is strongly related to the cortical audi-
mu rhythm in the light drowsy state after posterior alpha tory function (“auditory alpha rhythm”). Personal data
dropout. Yamada and Kooi (164) even described mu rhythm (170–173) do not provide good evidence for a relationship to
in sleep (stages 1 and 2, as well as rapid eye movement [REM] auditory function. In some individuals, cognitive activities
sleep) in 20 individuals (19 patients with various complaints). result in blocking responses of the third rhythm. Rhythmical
The concept of Covello et al. (163) is certainly in need of fur- activity in the alpha and upper theta range over the anterior
ther study. temporal–midtemporal region in conventional scalp EEGs can
be found in patients with cerebrovascular problems (175) and
Clinical Significance other types of temporal lobe changes, but this abnormal
Pure rolandic mu rhythm as such is categorically within normal rhythm can be differentiated from the physiologic “third
limits. In some cases, however, spiky discharges may be present rhythm” (173).
192 Part II ■ Normal EEG

Beta Rhythm ularly abundant in quantity and of rather high voltage to be


The Greek letter beta stands for the frequency band above termed abnormal. Even such fast records are often considered
13/sec. In customary EEG recording, frequencies above 70/sec only slightly abnormal unless they occur in unresponsive
are considerably attenuated due to filter effects. This estab- patients, where such fast activity may represent a severe abnor-
lishes a natural upper limit of discernible EEG activity some- mality. This view is essentially congruent with the study of
where between 50 and 100/sec. The upper limit of the beta Drake (189).
band is either considered open-ended or defined as 30/sec
(with gamma frequencies above). Rhythmical beta activity is Spatial Distribution
encountered chiefly over the frontal and central regions. A In certain individuals, the posterior alpha rhythm is unusually
central beta rhythm is related to the rolandic mu rhythm and fast and its frequency exceeds the upper limit of the alpha
can be blocked by motor activity or tactile stimulation, as rhythm (13/sec). Such a 14 to 15/sec rhythm shows a good
described above. The amplitude of beta activity seldom blocking response to eye opening and enhancement with eye
exceeds 30 µV. Beta activity may be locally enhanced over closure; it may be considered a fast equivalent of the alpha
bone defects and shows considerable increase (in quantity and rhythm. Gradual acceleration of the alpha frequency may be
voltage) after the administration of barbiturates, some non- caused by hyperthyroidism. Vogel and Götze (190) separated
barbituric sedatives, and minor tranquilizers. With very large the occipital 14 to 19/sec rhythm from the rest of beta activities,
amounts and high amplitudes of beta activity, abnormal pro- which were divided into frontocentral bursts of 20 to 30/sec
portions may be reached; in general, however, fast activity waves, and normal occipital alpha rhythm and diffuse beta
alone is seldom a cogent reason for calling a tracing abnormal. activity generally mixed with alpha frequencies. Vogel
Historically, the discovery of beta activity is closely linked to (191–193) subsequently divided the anterior beta activity into
the first description of the alpha rhythm by Berger (1). His first the 25 to 30/sec activity of the frontal region, occurring in brief
report on the human EEG of 1929 contains the following pas- trains, and the 20 to 25/sec activity of comparatively larger
sage: “The electroencephalogram represents a continuous curve amplitudes occurring for variable periods.
with continuous oscillations in which … one can distinguish Frontal runs of beta activity often exceed 30/sec and reach 35
larger first order waves with an average duration of 90 sigma to 40/sec when the patient is allowed to fall asleep. This activity
(msec) and smaller second order waves of an average duration may be found in unmedicated persons; sleep-inducing medica-
of 35 sigma (msec)” (after Ref. 41). In his second report, Berger tion, which increases the activity in the 18 to 25/sec band, also
(176) pointed out: “For the sake of brevity I shall subsequently augments activities in the faster beta range.
designate the waves of first order as alpha waves, the waves of The region of fastest EEG frequencies and highest levels of
second order as beta waves.” Berger (62) reversed earlier views cerebral blood flow (as well as oxygen and glucose uptake) is the
and pointed out “that the beta waves and not the alpha waves of frontocentral area. This observation has prompted the concept
the EEG are the concomitant phenomena of mental activity.” of “cerebral hyperfrontality” (194). This investigator has
This concept is now provoking renewed interest. pointed out that (a) prefrontal activity constitutes an integral
part of consciousness and (b) abnormal prefrontal activity is
Prevalence of Beta Activity accompanied by various forms of altered consciousness.
The physiologic beta frequencies may be broken down as
Beta activity is found in almost every healthy adult. Simon’s
follows:
(29) statement that beta frequencies have been found in 22% of
normal adults probably applies to more conspicuous forms of 1. Frontal beta: fairly common, may be very fast, no relation-
beta activity. Depth EEG studies and electrocorticograms show ship to physiologic rhythm.
a remarkably large share of the beta band; such recordings have 2. Central beta: partly but not generally the basis of rolandic
been obtained mostly in epileptics or in patients with neuro- mu rhythm often mixed with mu rhythm.
logic or psychiatric problems. Although obtained in patients 3. Posterior beta: often a fast alpha-equivalent, reactive-like
and not in healthy persons, such depth records are quite alpha rhythm.
informative concerning the spatial distribution of fast frequen- 4. Diffuse beta: no linkage with any special physiologic rhythm.
cies. Every scalp recording performed over a bone defect gives
By contrast, “vertex beta” or “fast central midline rhythm”
testimony to the wealth of cortical beta activity.
(195)—vertex activity in the 10 to 25/sec range—appears to be
The prevalence of beta activity in the normal person can also
the expression of polyetiologic focal abnormality in the fronto-
be derived from various studies of normal volunteers
central region.
(30,177–185). The results of these studies have been reviewed
by Kuhlo (186) in his thorough EEG handbook article on beta
rhythms. Later work has been presented by Fortuin and Künkel Other Influences on Beta Activity
(187) and Kozelak and Pedley (188). One of the most commonly encountered influences on the
While Gibbs and Gibbs (42) found varying degrees of fast abundance of beta activity in clinical practice is the use of cen-
activity to be abnormal, depending on age, electroencephalog- tral nervous system (CNS)–acting medications, particularly
raphers have shown a more lenient philosophy toward fast benzodiazepines and barbiturates. This topic is discussed in
tracings over the ensuing decades. Beta activity must be partic- more detail elsewhere in this volume.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 193

It is not surprising that beta activity in its various incar nations slightly contaminated with EEG activity. The definition of
has, over the decades, been associated with various personality Chatrian et al. (18) reads as follows:
traits and psychiatric disorders (42,177,178,183,196–199), but
Low voltage record. A waking record characterized by activity of
rarely with any degree of clinical or statistical certitude (200).
amplitudes not greater than 20 µV over all head regions.
During the entire maturational period (including early
With appropriate instrumental sensitivities this activity can
adulthood), beta activity has been found to be relatively more
be shown to be composed primarily of beta, theta, and, to a
prominent in females than in males (201).
lesser degree, delta waves, with or without alpha activity,
Ultrafast and Ultraslow EEG Activity over the posterior areas.
This definition serves as a solid guideline. One could subdivide
Interest in normal EEG activity within the ultrafast range from
low-voltage records into two or three degrees; occasionally, one
80 to 1000/sec has been surging over the past two decades. The
may see tracings in which 10 µV are not exceeded. Even such
advent of digital EEG recording has flung open the gate to this
“very low voltage records” may be due to desynchronization
new domain of promising work. The study of ultrafast activity
and thus occur in a waking and alert patient or even in a con-
can offer new epileptologic insights (202–205) but also, above
trol subject (Fig. 11.5). Low-voltage records are categorically
all, improved understanding of normal cortical perception,
within broad normal limits of variability and do not represent
motor activity, and, in particular, neurocognitive processes
an abnormality unless the frequency spectrum shows abnor-
(206–209). Similarly, there has been renewed interest in ultra-
mal local or diffuse slowing, asymmetries, or paroxysmal
slow activity, from near zero (DC) up to 0.3/sec. Again, while
events. In a comatose patient, one must infer that the low volt-
activity in this range may improve focal epilepsy diagnosis
age is due to true decline of cerebral activity and not merely
(210), there are also insights to be gained in neurocognitive sci-
caused by desynchronization. Such records must be regarded
ence, motor initiation, and sleep (211).
as extremely abnormal. This indicates that the record reader
The Low -Voltage Record should depend on behavioral observation (patient comatose,
on respirator, etc.). On the other hand, the EEG as such reveals
A below-average voltage output is compatible with perfect CNS
signs of telltale character for the experienced reader, such as
functioning. In such individuals, the small amplitudes are the
presence or absence of eye blinks, types of artifacts, etc., even
result of a lesser degree of synchronization of electrical activity
when he or she must read without any behavioral information.
in the neuronal level. Brainstem reticular formation systems
strongly influence the degree of cortical neuronal synchroniza-
tion; arousal mediated through the ascending mesodiencephalic Prevalence and Clinical Significance
reticular formation has a desynchronizing effect (212), whereas Early studies of low-voltage tracings were carried out by Davis
synchronizing effects are presumed to originate from the pon- and Davis (50), Jasper et al. (217), and Finley (177). Jasper et al.
tine portion of the ascending reticular formation (213,214). (217) used the term flat EEG, which is not reserved for truly iso-
This type of brainstem regulation of the cortical EEG syn- electric records.
chronization is likely to account for numerous fluctuations in
the EEG amplitudes, especially those related to the level of
vigilance. It is not clear whether these reticular effects account
for a low-voltage EEG as a personal trait (with genetic deter-
mination). Under pathologic conditions, low-voltage tracings
in vertebrobasilar artery insufficiency are probably due to
some degree of pontine ischemia acting on the reticular for-
mation. This could also be true for the frequently encountered
low-voltage tracings of chronic alcoholics in advanced stages,
with the possibility of early stages of pontine myelinolysis;
this explanation, however, is but a working hypothesis (215).
True decline of the cerebral EEG voltage output (i.e., low-volt-
age activity independent of desynchronizing mechanisms) is a
grave danger sign of a preterminal state unless it occurs for a
few seconds only, as in certain syncopal attacks. It may also
indicate diffuse chronic cortical degeneration (as described by
Ref. 216).

Definition
The term low-voltage EEG has always proved to be quite
flexible; it is in need of a strict definition. It was pointed out
earlier in this chapter that precise figures concerning ampli- Figure 11.5 Low-voltage record with poor alpha organization, in a
tudes are not truly accurate, because they depend on interelec- patient aged 31 years. Note that recording was done with above-stan-
trode distances and the montage; even an ear electrode may be dard gain. Finer details demonstrable with further gain increase.
194 Part II ■ Normal EEG

Gibbs et al. (179) (also see Refs. 42 and 218) found low-volt- amount of theta activity (178,183) because these slow frequen-
age fast records in 11.6% of 1000 normal adult subjects. The cies tend to linger on through the third decade of life; the com-
study of Adams (219) revealed a prevalence of 8% (in 427 nor- pletely mature aspect of the human EEG cannot be expected
mal individuals); the breakdown into age ranges is most impor- before the age of 25 to 30 years.
tant, because low-voltage tracings were found in 1% between On the basis of power spectra from normal adults, Rugg and
ages 0 and 20 years, 7% between 20 and 39 years, and 11% Dickens (227) presume that alpha and theta activity is gener-
between 40 and 69 years. Pine and Pine (220) reported 7.25% ated by separate mechanisms. Slow (theta and delta) EEG activ-
low-voltage records in 2000 neurologic and psychiatric patients ity has been correlated with cholinergic activities and central
between 17 and 70 years. Vogel and Götze (221) found low- cholinergic pathways (103). The observation of slowing
voltage tracings in 7.09% of 1540 patients. In 7000 patients induced by the action of cholinergic substances on the ascend-
including a large pediatric segment, only 3.6% were found to ing brainstem reticular formation dates back to Rinaldi and
have a low-voltage tracing (222). Himwich (228). Further information is found in the overview
According to Gibbs and Gibbs (42), the prevalence of low- of Riekkinen et al. (229).
voltage tracings increases sharply after the age of 13 years. This
is congruent with the observations of Panzani and Turner Spatially Restricted Theta Rhythms
(223), Adams (219), and Lucioni and Penati (224). Low-voltage With the use of a stress paradigm, Nowak and Marczynski (61)
fast records in children below the age of 10 years are suspect and produced in six of 24 healthy volunteers a rhythmical 5 to 6/sec
clearly abnormal if neither hyperventilation nor non-REM theta response originating from either the occipital region or
(NREM) sleep changes the low-voltage character of the tracing. vertex during a state of maximal alertness.
In adults, persistence of low-voltage activity with hyperventila- A rhythmical 4/sec pattern occurring over the vertex solely
tion and sleep is not categorically abnormal, although this lends in the waking state has been described as “4/sec vertex spindles”
more support to the presumption that such patients have an by Van Huffelen and Magnus (230). This very rare pattern may
organic cerebral disorder. Among neurologic disorders that represent a mild abnormality and is found mainly in adoles-
have been associated with a higher prevalence of low-voltage cents with syncopal attacks and other signs of vasomotor insta-
EEG activity are traumatic brain injury (225), basal ganglia dis- bility. The use of the term spindles appears to be somewhat out
orders (216), and endocrine and psychiatric disorders (219). of place when one considers the slow frequency and the occur-
There is some evidence that genetic factors play an impor- rence in the waking state. Hence, Daoust-Roy (231) proposed
tant role in determining a low-voltage tracing in a healthy per- the term 4/sec vertex rhythm.
son (221). This genetic predisposition, however, obviously does Rhythmical theta activity in the 6 to 7/sec range over the
not manifest itself in infancy or childhood, a time when almost frontal midline region has been correlated with mental activities
all low-voltage records bear the mark of abnormality. With ado- such as problem solving (232–238). In personal studies of men-
lescence (age 13, according to Gibbs and Gibbs Ref. 42), an tal EEG activation, we were unable to produce this pattern (44);
unknown maturational change apparently starts to desynchro- this indicates the technical difficulties in the demonstration of
nize the cortical EEG activity. the task-related frontal theta rhythm. This vexing problem has
been clarified by the work of Takahashi et al. (239), who found
Theta Rhythm that individuals with rhythmical frontal midline 6 to 7/sec activ-
The term theta was introduced by Walter and Dovey (226) and ity in light drowsiness also had the same type of activity during
denotes the frequency range of 4 to 7/sec or 4 to 7.5/sec. This mental tasks: “These two frontal theta rhythms closely resembled
frequency band was a part of the delta range until Walter and each other in frequency (94.6%) and distribution (83.8%)”
Dovey felt that an intermediate band should be established. The (237). One wonders if these tasks become boring enough to pro-
term theta was chosen in order to allude to its presumed thala- duce light drowsiness rather than increased alertness (one is even
mic origin. The intermediate character of the theta band has tempted to utilize the old Pavlovian term internal inhibition, i.e.,
been stressed in the German terminology Zwischenwellen, sleep induction in a certain repetitious experimental setting).
meaning intermediate waves. According to the international
nomenclature, the theta band is the “frequency band from 4
Lambda Waves
to under 8 Hz” and the theta rhythm is the “rhythm with a
frequency of 4 to under 8 Hz” (18). Definition and Historical Aspects
Lambda waves are “sharp transients occurring over the occipi-
Theta Activity in the Waking Adult tal region of the head of waking subjects during visual explo-
The normal adult waking record contains but a small amount ration. Mainly positive relative to other areas. Timelocked to
of theta frequencies and no organized theta rhythm. Theta fre- saccadic eye movements. Amplitude varies, but is generally
quencies and theta rhythms, however, play an important role in below 50 µV” (18).
infancy and childhood, as well as in states of drowsiness and This visually induced occipital activity was described by
sleep (all discussed elsewhere in this volume). Larger contin- Gastaut (240); there was an earlier oral presentation by Evans
gents of theta activity in the waking adult are abnormal and are (EEG Society in London, 1949). Subsequent EEG studies on
caused by various forms of pathology. Studies of young adults lambda waves were presented by Cobb and Pampiglione (241),
such as army personnel and navy pilots have shown a sizable Evans (242,243), and Roth and Green (244). A very extensive
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 195

review of this subject is found in the handbook article of


Chatrian (245).

Prevalence
Lambda waves are unmistakably present in some records (Figs.
11.6 and 11.7) and are not readily demonstrable in others. They
are most prominent in waking patients intently viewing an illu-
minated visual field (245). The prevalence essentially depends
on the thoroughness of the EEG evaluation and the emphasis
placed on the demonstration of this special phenomenon.
According to Tsai and Liu (1965; quoted in Ref. 245), lambda
waves are most frequent between ages 3 and 12 years (82.3%);
their prevalence declines to 72% between 18 and 30 years and
to 36.4% between 31 and 50 years.

Morphology
Lambda waves have been described as biphasic or triphasic; the
most prominent phase is positive. Their form has been
described as triangular or sawtooth shaped. The amplitude is
usually below 20 µV and may exceed 50 µV in some persons.
The overall duration of lambda waves lies between 200 and 300
msec. These waves repeat themselves, usually at intervals from
200 to 500 msec. Marton et al. (246) have demonstrated the
complexities of lambda waves; their predominant positive com-
ponent is preceded and followed by a negative component.

Spatial Distribution Figure 11.7 Prominent occipital lambda waves during eye opening.
Lambda waves are most prominent in (or confined to) the Patient, aged 54 years, looking ahead in the dimly lit laboratory. There
occipital leads. Spread into parietal and posterotemporal areas is no evidence of scanning ocular artifacts. Note instability and fast
is common and, in certain cases, the maximum may be found character of alpha rhythm (1 to 13/ sec and even faster). The bottom part
in areas adjacent to the occipital lobe (243,244). Lambda activ- shows continuation of the upper part.
ity is strictly bilateral synchronous. With the use of depth elec-
trodes, Perez-Borja et al. (98) demonstrated multiple foci of
lambda waves, either in or near the calcarine region or more Precipitating Factors
laterally in the occipital lobes. Voluntary scanning eye movements (exploratory saccades) play
a very important role. It was found that most lambda waves
follow an exploratory eye movement with a latency of 67 to 85
msec (mean, 78 msec) (247). The preceding eye movement has
been subsequently used as a trigger for computer techniques
(143,248–253). Further interesting details on this subject are
found in the handbook article of Chatrian (245).
Lambda waves are best found in brightly lit laboratories and
cannot be elicited in darkness. The size of a given pattern and its
distance and color are further variables. Binocular viewing of a
picture may or may not produce larger lambda waves than
monocular viewing (254,255). Presentation of a new picture
enhances lambda production (245,256). Lambda activity does not
seem to be related to recognition of objects (247). Hypnotic sug-
gestion of a test picture failed to produce a lambda response (255).

Relationships to Alpha Rhythm


and Visual Evoked Responses
There appears to be considerable independence between poste-
rior alpha rhythm (with eyes closed) and lambda waves (with
eyes open); no interaction or complementary action of these
Figure 11.6 Occipital lambda waves while viewing pictures, in a patient two activities has been found. Marked similarities between
aged 15 years. Note ocular artifacts caused by scanning eye movements. lambda waves and visual evoked responses have been reported
196 Part II ■ Normal EEG

by Remond et al. (252) and Lesèvre (257). Relationships usually emerge about 60 to 90 minutes after sleep onset; this is
between lambda waves and occipital photic driving were origi- closer to 90 minutes in sedated sleep. This necessitates unusually
nally presumed by Gastaut (240) and Evans (243) but could not prolonged recording and, in the hustle of a regular laboratory,
be confirmed by Chatrian et al. (258), Perez-Borja et al. (98), blocks one of the recording units for an unduly long period.
and Scott et al. (255). Here the need for nocturnal sleep recording becomes obvi-
ous. There are now a growing number of specialized nocturnal
Pathologic Significance sleep laboratories that offer 24-hour recordings of a given
No correlation between lambda waves and any neurologic or patient. These laboratories have concentrated on (a) nocturnal
psychiatric disorder has been demonstrated. sleep research in healthy volunteers, (b) the exploration of the
sequential nocturnal sleep phases in patients with sleep disor-
Theories on the Basic Neurophysiologic Mechanism ders, and (c) the study of poorly understood (and possible
Lambda waves have been thought to be related to (a) visual epileptic) attacks of strictly nocturnal occurrence.
evoked responses, (b) oculomotor–visual integration, (c) ocu- Specialized sleep laboratories are polygraphically oriented
lomotor potentials, or (d) arousal mechanisms. These theories and use EEG in combination with a variety of other physiologic
have been thoroughly discussed by Chatrian (245). parameters. It is not unusual to find only two channels reserved
Billings (259) has proposed that there are two different for EEG recording. This polygraphic approach has made enor-
occipital lambda waves in the human. The first one is the result mous progress over the past decades but will not be discussed
of the release of peripheral visual inhibition during the braking further in this chapter.
phase of the primary saccade. This wave is transmitted via the The scoring of sleep stages (as it is being done in sleep labo-
faster conducting Y-type fibers of the optic nerve (subserving ratories) is essentially based on the rules laid down by
peripheral vision). The second wave is elicited by the return of Rechtschaffen and Kales (260). These rules have been strongly
normal central vision, during or slightly before the braking criticized and major changes in the nomenclature are being
phase of the secondary corrective saccade. This wave is trans- introduced and are becoming more widely accepted (see the dis-
mitted via the slower conducting X-type fibers of the optic cussion on polysomnography in Chapter 41). Kubicki and his
nerve (subserving central vision). According to Billings, coworkers have been the most consistent proponents of criticism
“lambda waves, long considered to be of rather trivial import by (261–263). Most of this criticism has been directed toward (a)
clinical electroencephalographers, are of considerable func- lack of insufficient EEG information (limitation to central lead)
tional significance in the visual system.” and (b) misleading delta activity criteria resulting in erroneous
scoring. This is particularly true with stage 1 and 2 segments
THE RECORDING OF EEG DURING SLEEP occurring within REM sleep and falsely scored as REM sleep.
The emphasis on sleep sections in routine laboratory work
Sleep EEG recordings have been performed in regular EEG lab- varies greatly and depends on the influence of major centers
oratories for at least 60 years in order to secure information not and schools. Most major American schools of EEG traditionally
available in the usual waking–resting record with such activa- use sleep in routine EEG work to a great extent. Some laborato-
tions as hyperventilation and intermittent photic stimulation. ries try to attempt sleep in every or almost every patient at any
The purpose of these EEG sleep studies is to search for abnor- age. In this author’s laboratory, the proportion of sleep records
malities that may be hidden in the waking state. The more this was consistently around 60%. Debates about the usefulness of
type of combined waking and sleep recording is carried out in sleep can be heated; many electroencephalographers use sleep
the regular EEG laboratory, the greater will be the insight into only as a method to obtain a tracing in uncooperative and oth-
the sleep patterns of normal individuals. erwise unmanageable children. The informative value of short
Nevertheless, the term sleep EEG may be somewhat pre- sleep recordings has been stressed by Gastaut et al. (264,265)
sumptuous when one considers that only a short segment of and Niedermeyer and Höller (266).
sleep can be recorded under daytime recording circumstances. There is no doubt that the overall information derived from
Furthermore, the recorded sleep is basically a nap, either sponta- a wakefulness record is greater and hence clinically more signif-
neous or induced by a sedative, and is thus different from noctur- icant. There are, however, a number of conditions, especially in
nal sleep. In the regular EEG laboratory, one can observe the the domain of epileptic seizure disorders, in which sleep pro-
process of falling asleep during the daytime. The ensuing sleep is vides essential information. (This excludes the sleep disorders
relatively short and, for the most practical purposes, a duration of themselves, which ought to be evaluated in a specialized sleep
10 to 30 minutes will suffice. The result is a tracing that shows the laboratory.) For the sake of this important minority, sleep por-
transition from the waking state to drowsiness and from drowsi- tions should be secured in most patients.
ness (stage 1) to light NREM sleep (stage 2). These stages of falling The drowsy state and light sleep (stage 2) are usually the most
asleep and light sleep are usually the most informative stages for informative phases; for this reason, the sleep-onset portion with
the electroencephalographer who is searching for clinically rele- a length of 5 to 30 minutes in stage 2 sleep may be sufficient. In
vant abnormalities and especially for epileptic discharges not infants and children, a sleep section (nap recording) is almost a
demonstrable in the waking state. In certain cases, deeper sleep necessity, not simply because of easier management of the sleep-
(stage 3 or possible stage 4) may yield additional information; ing child but also because of the needed information. In infants
this may require a more prolonged sleep recording. REM stages and in small and preschool children, the EEG recording often
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 197

cannot start until the child has fallen asleep. In these cases, great 2. Anticonvulsant medication does not have to be discontinued.
efforts must be made to secure a readable waking portion after 3. Use sleep deprivation with other techniques of activation
awakening. (see also Ref. 277).
It can be understood readily that pad electrodes pressed 4. Patients must be kept awake during the test period.
against the scalp by caps or bands are counterproductive in a 5. Expect very few false-positive cases but many false-negative
sleep laboratory. The slight discomfort caused by such elec- cases.
trodes is enough to keep the patient from falling asleep. For this 6. Sleep deprivation is a genuine activation method. Its effi-
reason, electrodes suitable for sleep recording ought to be fixed cacy in provoking abnormal EEG discharges is not due to
to the head by an adhesive. drowsiness.
In some laboratories, the technologist and the apparatus are
The insistence on keeping the patient awake during the
placed in the same room where the patient lies. In the absence
recording may be debatable and its value should be thoroughly
of video monitoring, this setup has advantages such as closer
assessed. There is reason to presume that bilateral synchronous
observation, especially in patients with clinical seizures, over
seizure discharges are triggered by short fluctuations of the
the system of having patient and machine in separate rooms
level of awareness (light drowsiness followed by an arousing
with a window permitting observation of the patient. With this
stimulus; see Ref. 165); for this reason, the significance of short
latter setup, the technician can more effectively request help
and minor drowsy periods should be emphasized rather than
from colleagues or the medical director. Beds or carts are supe-
minimized.
rior to reclining chairs.
As to the search for EEG abnormalities, a differentiation
should be made between patients with primary generalized
SPONTANEOUS SLEEP, SLEEP DEPRIVATION, epilepsy and temporal lobe epilepsy. In the former, the strict
AND SEDATED SLEEP routine suggested by Naitoh and Dement (279) might be highly
effective; these patients should by all means be kept awake dur-
Spontaneous Sleep ing recording (also see Ref. 275). In temporal lobe epilepsies,
Sleep without medication is undoubtedly preferable to sedated the ensuing deep sleep (stage 3) could be more informative.
sleep. In an appropriately darkened room, many children and Rodin et al. (280) have extended the period of sleep loss in
adults fall asleep quite readily. Outpatients from distant areas normal young volunteers to 120 hours of total sleep depriva-
and those who have experienced strenuous travel in the early tion. There was evidence of frequent generalized bursts similar
morning hours also are likely to sleep without medication. to those seen in primary generalized epilepsy. These changes,
however, could not be confirmed by other investigators
Sleep Deprivation (281–284), who used sleep deprivation for periods as long as
Sleep deprivation prior to recording has been suggested over the 264 hours. Welch and Stevens (285) also felt that sleep depriva-
past decade by a number of electroencephalographers. This tion did not activate the EEG of healthy subjects.
method has been described by Bennett (267), Bennett et The suggestion of Gibbs and Gibbs (218) to keep the
al. (268), Mattson et al. (269), Oller-Daurella (270), Pratt et patients awake until after midnight is helpful as a means to
al. (271), Wittenbecker and Kubicki (272), Rumpl et al. (273), obtain sleep more readily the following morning in the labora-
Deisenhammer and Klingler (274), Degen (275), Jovanovic tory. However, this widely used practice almost completely
(276), Kubicki et al. (277), and Klingler et al. (278). It consists of dilutes the basic principle of sleep deprivation. In practice, good
sleep loss for 24 to 36 hours. Its goal is the detection of epileptic sleep recordings in stage 2 are only slightly less informative than
discharges that could be missed otherwise. According to the view tracings obtained after sleep deprivation for 24 hours.
of Pratt et al. (271), a good deal of the activation of paroxysmal Sleep deprivation is a true activator, it is a specific stress
patterns may be ascribable to drowsiness and sleep as such, but imposed on the CNS to activate otherwise hidden epileptic
some other factors specific for sleep deprivation have proved to activity. Sleep recordings in general are done in a physiologic
be potent activators. In the research of Pratt et al., activation of state and should not be listed as activation. The sleep depriva-
epileptic patterns was obtained in 41% of 114 patients. tion stress can be augmented by the effect of anticonvulsants
Sleep deprivation may have drawbacks. Many individuals with sedative effect (phenobarbital or primidone) against
and especially children may be so fatigued that they fall asleep which the patient must fight. According to Klass and Fischer-
almost instantly in the laboratory with rapid evolution of stages Williams (286), the benefits derived from sleep deprivation out-
3 and 4; with the possible exception of temporal lobe epilepsy, weigh the risks of clinical seizures. These authors, however,
these are much less informative electroencephalographically point out that in sleep-deprived persons photic stimulation
than stage 2. This is especially true in cases of generalized requires greater caution. It appears that the emphasis placed on
epilepsies (primary generalized epilepsy, Lennox–Gastaut syn- sleep deprivation in most clinical neurophysiology laboratories
drome) and benign focal spikes in children. has declined over the past two decades.
Naitoh and Dement (279) have summarized the current sta-
tus of sleep deprivation as follows: Sedated Sleep
1. One night of sleep loss is sufficient. Sleep loss can be per- Whenever a sleep recording is desirable and cannot be obtained
formed at home with the help of a family member. naturally, sedated sleep is dictated by necessity. The most
198 Part II ■ Normal EEG

Tabl e 11 . 1 1. Drowsiness: from alpha dropout to vertex waves


2. Light sleep: spindles, vertex waves, K complexes
Guidelines for Sedation to Obtain 3. Deep sleep: much slowing, K complexes, some spindles
EEG Sleep Records 4. Very deep sleep: much slowing, some K complexes
REM sleep: desynchronization with faster frequencies
Recommended sedative: chloral hydrate (oral); chloral The transition from stage to stage may be somewhat imprecise,
hydrate syrup U.S.P. but in general, staging of sleep periods is not difficult for elec-
A. Adult and adolescents above 15 years troencephalographers who are used to reading records in both
Average weight (56–80 kg): 1.75–2.5 g the waking state and sleep.
In the clinical EEG laboratory, most sleepers reach only stage 2,
Below average weight (40–55 kg): 1.25–2 g stage 3 is reached occasionally, and stage 4 seldom reached
Above average weight (above 80 kg): 2.5–3.5 g unless unusually long sleep recording time is allotted or in cases
B. Children (5–14 years) of profound tiredness after sleep loss. REM stages are seldom
seen in the clinical laboratory except in infants, young children,
1.0–2.0 g (depending on weight) and adults with sleep loss. Their occurrence at sleep onset may
C. Infants and small children (4 months to 4 years) denote a primary sleep disorder (especially a narcolepsy–cata-
Below 1/2 year: 0.125–0.25 g plexy complex), but REM sleep onset may also occur in patients
with severely disturbed sleep due to central disturbances such
1/2–1 year: 0.25–0.5 g (max 0.75 g) as delirium tremens or in deep-seated cerebral pathology.
1–4 years: 0.5–1.0 g (max 1.5 g if close to 4 years and above Reviews of the EEG characteristics (294) and the neurobio-
average weight) chemical basis of sleep (295) are recommended sources of fur-
ther information. The current classification scheme has been
Advantages of chloral hydrate: induced fast EEG activity less prominent than slightly modified and is outlined in Chapter 41.
with barbiturates and rapidly metabolized. Side effects (paradoxical excitation
with irrational behavior) rare.
THE “MICROSTRUCTURE” OF SLEEP:
important advice to a novice in this technique is patience; the
CYCLIC ALTERNATING PATTERNS
patient must not be overpowered by strong and rapidly effective Sleep is not solely determined by macrostructural shifts of
sedatives. Therefore, the intravenous and intramuscular routes NREM and REM sleep stages. Evidence of a “microstructure” of
must be avoided; with these the patient’s sleep becomes very sleep has been provided over the past 20 years. Transient activa-
unnatural and contains elements of an early state of general tion phases (“phases transitoires”) were reported by Schieber et
anesthesia. The highly informative stages of drowsiness (stage 1) al. (296). “Cyclic alternating patterns” of sleep were observed by
and light sleep (stage 2) are quickly passed, and the ensuing deep Terzano and his coworkers, first under severely abnormal condi-
sleep yields very little information in most cases. Enormous tions such as Creutzfeldt–Jakob disease (297), later in normal
amounts of fast activity may also obscure important details. individuals (298,299). Cyclic alternating patterns (CAPs) occur
Orally or rectally administered sedatives act more gently and mainly in NREM sleep, are generally associated with transient
slowly. The ensuing sleep contains many or most features of lightening of sleep depth, and are independent of afferent stim-
spontaneous sleep. Most laboratories have the greatest experi- uli, thus constituting modality-independent responses (299,300).
ence with chloral hydrate (Table 11-1). The oral route is stan- The cyclic alternating pattern consists of an A phase of enhance-
dard; rectal administration is usually confined to infants ment of electrical activity and a subsequent B phase characterized
reluctant to take the medication by mouth. by attenuation of EEG activity. Each phase lasts only between
2 and 60 seconds.
Terminology of Sleep Stages
A somewhat different type of periodic activity of light sleep
The discovery of human REM sleep has been attributed to has been demonstrated by Evans (301), who observed alternat-
Aserinsky and Kleitman (287), who were able to demonstrate reg- ing periods of alpha and theta activity with a usual interval of
ularly recurrent periods of altered ocular motility during sleep. about 16 seconds. This activity is thought to be related to inter-
Further insight into the significance of the REM stages prompted mittent arousing mechanisms involving cortex and brainstem
a new terminology of sleep stages that emphasized the dichotomy activating systems (301).
of two basically different types of sleep: non-REM (“slow sleep”)
and REM sleep (also “paradoxical sleep” or “fast sleep”).
This dualistic approach subsequently proved to be appropri- THE STATE OF DROWSINESS
ate because of impressive differences of neurobiochemistry and
neuronal circuitry (288–292). Early Drowsiness
A commonly used classification (essentially following that of The level of vigilance tends to fluctuate, and fleeting brief
Dement and Kleitman Ref. 293) divides sleep into the following episodes of very light drowsiness are very common in waking
stages: records. Boredom, fatigue, and monotony may quickly induce
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 199

such periods in a patient instructed to relax during the test.


Drowsiness is a very interesting state from the electroen-
cephalographer’s viewpoint. It shows particularly marked age-
determined changes; the “hypnagogic” rhythmical 4 to 6/sec
theta activity of late infancy and early childhood represents a
unique feature of that period of life. Later in childhood and, in
many cases, in the declining years of life, the onset of
drowsiness is characterized by greater amounts of slow activity
mingling with the posterior alpha rhythm. An electroen-
cephalographically defined drowsy state does not exist in the
neonate and, in senility, abrupt transitions from wakefulness to
light sleep are again quite common.
In the adult, the onset of drowsiness is characterized by grad-
ual or brisk “alpha dropout” (Fig. 11.8). The alpha waves are
replaced by low-voltage slow activity, mainly in the range of 2 to
7/sec. Very low-voltage 15 to 25/sec activity may be mixed with
the slowing. Thus, the tracing appears to be desynchronized due
to the general voltage decline. This state must be strictly sepa-
rated from periods of enhanced alertness after eye opening or
caused by mental or emotional stress (associated with alpha Figure 11.9 Early drowsiness in a 56-year-old patient with dizziness
blocking). In enhanced alertness, there is also general desyn- and memory loss. Rhythmical 5 to 7/ sec activity is noted over posterior
chronization with low-voltage activity, but the slow frequency regions very shortly after the disappearance of alpha waves. This rhyth-
component is absent and beta frequency clearly predominates, mical theta activity in earliest drowsiness is not categorically abnormal;
mixed with some remnants of alpha. In patients with low-volt- however, incipient cerebrovascular disorder is suspected.
age fast waking records, the onset of early drowsiness may be
quite poorly defined. Alpha dropout of early drowsiness and
immediate slowing in adulthood is a suspect finding (Fig. 11.9). Deepening of drowsiness is associated with enhancement of
slow activity. Trains of 2 to 3/sec and 4 to 7/sec waves reach
medium voltage and may become diffusely predominant. At
this stage, arousing stimuli lead to immediate return of the pos-
terior alpha rhythm, called “paradoxical alpha response” (302).
These stretches of reactivated alpha rhythm are often character-
ized by higher amplitude than the individual’s regular alpha
rhythm. When alpha rhythm is reactivated in deep drowsiness
or in NREM sleep, the maximum is usually frontal rather that
occipital (“short microarousals”).
If a record is dominated by patterns of drowsiness and sleep
and if waking states can be maintained for short periods only,
then there is reason to presume that such a recording is not
within normal limits. In such cases, there is usually a large
amount of diffuse slow (2 to 6/sec) activity preponderant dur-
ing drowsiness and even in early drowsy states. In these individ-
uals, the sequential structure of waking and sleeping is likely to
be disturbed. Cerebrovascular and metabolic problems often
account for such changes. Sleep deprivation must be ruled out
in these cases, but in general healthy sleep-deprived adults are
unlikely to exhibit excessive slowing in states of drowsiness.
A major study of the EEG in drowsiness has been presented by
Santamaria and Chiappa (303). Their work has placed special
emphasis on changes in the alpha rhythm with occurrence of a
centrofrontal alpha and temporal alpha (change in alpha distri-
bution due to early drowsiness) as well as changes in alpha ampli-
tude (diminution as well as increase). These changes of alpha
distribution in early drowsiness do not seem to be the rule. As
Broughton and Hasan (304) have pointed out, only a small por-
tion of their subjects showed anterior diffusion of alpha rhythm
Figure 11.8 Alpha dropout with early drowsiness in normal adult. (along with slowing by 0.5 to 1.5/sec) during drowsiness.
200 Part II ■ Normal EEG

however, that biparietal humps truly denote vertex waves. In the


typical referential montages of Gibbs and Gibbs (42,218),
vertex waves appear most strikingly in parietal leads, because
no midline and rolandic electrodes are placed.
The vertex wave is a compounded potential; a small spike
discharge of positive polarity precedes the large following nega-
tive wave, which is almost always the most prominent feature of
the discharge. Another small positive spiky discharge usually fol-
lows. The complexity of this potential reflects its neurophysio-
logic character as an evoked potential. As a “secondary” evoked
response, it is best compared with the vertex auditory evoked
response that is maximal over the vertex (311), despite the differ-
ent views of Vaughan and Ritter (1970) (see discussion of Storm
van Leeuwen (312). Its principal deflections are found in a
latency range of 50 to 800 msec (313). H. Davis et al. (314) found
a positive deflection P1 at 50 to 60 msec, a negative N1 at about
100 msec, a positive P2 at 170 to 200 msec, and a negative N2 at
about 300 msec. The authors feel that tactile vertex responses
have slightly longer latency and show a more prominent P1.
Vertex waves may occur as an isolated event; their large
amplitude usually towers over the rest of the EEG activity. Quite
frequently, salvos of repetitive vertex waves occur, firing at a
range of about 1/sec or even somewhat faster.
Although the maximum of the vertex waves is almost invari-
Figure 11.10 Vertex wave in deep drowsiness. Enhanced fast activity is ably the vertex itself, their spread may reveal asymmetries that
due to medication (chloral hydrate). are fairly common in children. Pathology seldom selectively
depresses the vertex wave, but such cases may occur (315).
Sharp boundaries must be drawn between vertex waves and
In light drowsiness, the P300 response (standard auditory rolandic spikes. Truly epileptogenic spikes may occur over the
paradigm) increases in latency and decreases in amplitude “but vertex. In practice, however, the differentiation between abnor-
the counts of infrequent tones remained correct nevertheless” mal vertex spikes and the physiologic vertex wave is not diffi-
(305). This important study shed some light on mental func- cult, because the configuration of vertex spikes is more slender
tioning in light drowsiness. On the other hand, EEG coherence and more pointed (also see Ref. 277). Vertex waves may become
studies show altered inter- and intrahemispheric EEG coher- small and inconspicuous in aged individuals and are often
ence in light drowsiness (306). poorly demonstrable in such persons.
Deep Drowsiness Positive Occipital Sharp Transients of Sleep (POSTS)
Vertex Waves Another important physiologic potential of deep drowsiness
The hallmark of deep drowsiness is the appearance of vertex is the POSTS, which become apparent in deep drowsiness and
waves (Fig. 11.10). These waves indicate an altered state of cere-
bral responsivity; vertex waves are secondary evoked potentials
of several modalities, perhaps mostly auditory, that converge
from their cortical projection areas in regions underlying the
vertex electrode. This region constitutes the posterior portion
of the supplementary motor area of the frontal lobe along the
interhemispheric fissure. The vertex waves represent an evoked
event, like K complexes, discussed later.
The term vertex wave was introduced by Liberson (307). On
effect (308), an earlier term, is no longer used. French authors
have used the term vertex spikes (“pointes au vertex”)
(309,310). The catalog of EEG patterns in the Handbook of
Electroencephalography and Clinical Neurophysiology uses the
term vertex sharp wave (3). Other synonyms are v waves and
vertex sharp transients. Surprisingly, Dutertre (3) does not list Figure 11.11 POSTS (positive occipital sharp transients of sleep) in a
the term biparietal hump (42) as a synonym of vertex waves and 34-year-old subject. (From Niedermeyer E, Lentz WJ. Dreaming in non-
discusses this term among “slow waves.” There is no doubt, REM sleep. Waking and Sleeping. 1976;1:49–59.)
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 201

persist during light sleep and deep sleep (Fig. 11.11). This dis- Once sleep has begun, the record shows a mixture of
charge was described first by Gibbs and Gibbs (42) as “positive arousal-related patterns that truly deserve the often overused
spike-like waves in occipital areas”; it was found most com- term background activity. For the electroencephalographer,
monly in adolescents and young and middle-aged adults. Its sleep would be a drab and dull affair if the picture were not
prevalence declines after the age of 70 years (316). enlivened by numerous arousal responses such as spindles, ver-
Similarities to occipital lambda waves were pointed out in tex waves, and K complexes. In addition, positive occipital tran-
several studies (98,258,317,318), and the term lambdoid activity sients of sleep are extremely common and may reflect some
of drowsiness and sleep was used. Hess (319) used the term ongoing visual activities.
monophasic theta in view of the predominant theta frequency of In sleep, the electrophysiologic “roar” of the brain, which
these chiefly rhythmical positive vents, whereas the term occipi- accounts for the alpha rhythm and a variety of other slow and
tal sharp waves of sleep is found in the work of Arfel and Laurette fast activities, mellows and does not muffle event-related pat-
(320). Kugler and Laub (14) used the term lambda waves during terns to the same degree as in the waking state. Events express-
sleep and subsequently proposed the term rho waves (321,322). ing themselves as evoked potentials, such as the vertex wave,
Vignaendra et al. (323) studied the relationship of these poten- tower over the background activity, whereas the same events
tials to the nocturnal sleep cycle, stressing their abundance in must be extracted by special averaging methods from the elec-
stage 2 and 3 sleep and scarcity or absence in REM sleep; these troencephalographic “noise” of the brain in the waking state. In
investigations also introduced the aforementioned term positive the more recent view of the “chaos theory,” however, the EEG
occipital sharp transients of sleep (POSTS). noise should not be regarded as an anarchic state (328).
These potentials are found in about 50% to 80% of a healthy These are but a few words from the vantage point of the EEG
adult population (49% according to Prior and Deacon (317); laboratory. The polygraphic approach of nocturnal or 24-hour
79% according to Brenner et al. (324)). It is, therefore, quite dif- polysomnography is quite different and is presented elsewhere.
ficult to attribute a special significance to this activity. A much The polygraphic approach permits further insight into a variety
lower figure (24%) and a striking preponderance of females of autonomically and neurobiochemically governed mecha-
were reported by Pristasova and Prochazka (325). nisms. On the other hand, the polygraphic approach cannot do
Vignaendra et al. (323) hypothesize that POSTS might rep- full justice to EEG details because of limitations of channels and
resent a “playback” of information to the visual cortex in order data compression due to slow paper speed, so that the clinical
to reexamine visual material collected during the day. A rela- electroencephalographer’s insight into EEG sleep mechanisms
tionship of POSTS to dreaming (true dreams rather than hyp- retains its great value.
nagogic imagery) in NREM sleep (stage 2) was suggested by
Niedermeyer and Lentz (326), whereas Dement (327) strongly Background Activity During Light Sleep
emphasizes the lack of dreaming in this stage. According to Background activity shows considerable interindividual varia-
Chatrian (245), POSTS are unrelated to oculomotor activity as tion. Slow frequencies ranging from 0.75 to 4/sec are usually pre-
well as to visual imagery. In blind or severely amblyopic indi- dominant; their voltage is high with a very prominent occipital
viduals, POSTS fail to materialize (324). peak in small children and gradually falls throughout adoles-
cence and adulthood. The true amount of the slowest frequency
components is not readily assessable because of limitations due
LIGHT SLEEP (STAGE 2 SLEEP) to input capacitance and between-stages capacitance (deter-
mining the time constant and the low-frequency filtering). Very
The Transition into Light Sleep (Stage 2 Sleep) pronounced slowing, especially in frontal leads, is often due to
The distinction between deep drowsiness and light sleep is perspiration artifacts. In adulthood, the voltage of the slow
imprecise and has necessarily been drawn in a somewhat arbi- activity is moderate and may fall to the extent that the record-
trary and artificial manner. In individuals with a well-developed ing must be done with higher sensitivity in order to evaluate the
drowsy state (i.e., in most children, adolescents, and adults), background amplitudes.
there is a very smooth transition from drowsiness to sleep. It is Patients with low-voltage records in the waking state may
customary to regard the appearance of spindle trains in the 12 to also show below-average voltage while asleep. This is particu-
15/sec range as the signal of sleep onset. Not much can be gained larly true when the low-voltage awake tracing is likely to be
by the observation of the patient. Even snoring may occur in related to organic cerebral problems affecting the synchroniza-
what is deep drowsiness from the EEG viewpoint. tion of EEG activity, probably in the pontine level (see Ref.
In EEG practice, montages ought to be suitable for the 213). Patients with vertebrobasilar artery insufficiency infre-
recognition of early sleep patterns. The first runs of spindles quently show such low-voltage patterns during both wakeful-
occur over the vertex with considerable spread into parietal and ness and sleep (165,329), and low-voltage patterns are also
central leads. These areas should be well covered. A circular common in advanced cases of chronic alcoholism (330). On the
montage covering frontopolar, temporal, and occipital areas other hand, the low-voltage waking records of tense and overly
could miss the first minutes of stage 2 sleep. Referential linkages anxious patients change to average voltage output in sleep.
from the vertex and centroparietal areas to the ipsilateral ear are Fast frequencies are commonly in the range of 15 to 30/sec,
quite informative because of the wide interelectrode distance. which lies clearly above the spindle frequency of 12 to 14/sec.
202 Part II ■ Normal EEG

This activity may occur even in unsedated patients with no his-


tory of chronic intake of sedatives. Some individuals show
frequencies in the range of 28 to 40/sec, especially over frontal
areas. Gibbs and Gibbs (42) focused much attention on such
fast patterns (“F1, F2, F3 patterns”) and consider very fast activ-
ity (above 30/sec) an abnormality.
These very fast waves occur in recurrent trains, mainly over
anterior regions. In some individuals, this type of very fast
activity could be drug related, and one wonders if one is deal-
ing with a 2:1 harmonic of the usual sedative-induced fast fre-
quencies (customarily in the 18 to 25/sec range). Relationships
to epileptic changes are not clearly demonstrable (218).
Lorimer (331) found a high prevalence of such fast patterns in
a prison population. Kubicki et al. (262,263) have emphasized
the occurrence of “subvigil beta activity” during short arousal
periods. With the use of the ultrafast frequency range (80 to
1000 Hz), more light will be shed on the function of fast and
ultrafast frequency during sleep.
According to human sleep studies of Marshall et al. (332), a
negative DC shift is associated with the transition from the
waking state to sleep. Luthringer et al. (333) reported a paral-
lelism between slow wave activity and plasma renin levels in
human sleep.

Sleep Spindles
Sleep spindles are also known as sigma activity or sigma waves
(Figs. 11.12 and 11.13). These latter terms were at first interna-
tionally encouraged, for instance by the IFSECN in 1961, Figure 11.13 Light sleep (non-REM stage 2), in a patient aged 40 years.
Well-developed and rather widespread spindle activity, mostly in the
range of 13 to 15/ sec, most prominent in superior frontal, central, and
parietal leads. Also note slow background activity.

but were discouraged in a later glossary from the same federa-


tion (18). In this glossary, spindles are defined as a “group of
rhythmic waves characterized by progressively increasing, then
gradually decreasing amplitude” (see also Ref. 3).
This waxing and waning of amplitudes is exactly what the
term spindle implies: a belly in the middle tapering off to the left
and to the right. Unfortunately, experience teaches us time and
again that this shape of spindle train is not the rule and may be
rather uncommon. As a matter of fact a train of posterior alpha
waves in the waking state may show better spindle shape than
the average sleep spindles. Be this as it may, the fact remains that
the term sleep spindles is in widespread use and should be
retained in order to avoid confusion.
The observation of sleep spindles dates back to the work of
Loomis et al. (334). Gibbs and Gibbs (42) made remarkable
contributions to the description of the wave morphology of
spindles. The first spindle trains show frequencies around
14/sec (from 12.5 to 15.5/sec with a pronounced peak at exactly
14/sec, according to Gibbs and Gibbs). These investigators
described the spatial distribution of spindle trains as primarily
Figure 11.12 Light sleep (non-REM stage 2), in a patient aged 31 years. central, but the use of midline electrodes shows a very definite
Two categories of spindles are shown; spindles of 14 to 15/ sec fre- maximum over the vertex. It is, therefore, worthwhile to include
quency dominate over centroparietal region, whereas less abundant the vertex region in montages when it is anticipated that the
spindles in the 12 to 13/ sec range are found over frontal area. patient will fall asleep.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 203

Gibbs and Gibbs (42) separated the 14/sec spindle activity of literature dealing with spindles that, in reality, are trains of
from the 12/sec and the 10/sec spindles. The 12/sec spindle barbiturate-induced beta activity. Bremer (343) suggested that
train appears a little later with deepening sleep although the barbiturate spindles are the animal counterpart of human alpha
patient is still in stage 2. These spindles range, according to rhythm. Barbiturate spindles have been used as animal models
Gibbs and Gibbs (42), from 11 to 13.5/sec with a pronounced for human alpha rhythm (90,91,344). It must not be forgotten
peak at 12/sec. Their spatial distribution is different; there is that this work is based on drug-induced fast activity Steriade et
accentuation over frontal areas, with an impressive maximum al. (103) have strongly refuted the concept of Andersen and
over frontal midline. Gibbs and Gibbs stress bilaterally inde- Andersson (90,91). Based on experimental work in the cat, it is
pendent occurrence of 12/sec over frontal areas, but such asym- now presumed that the reticular thalamic nucleus of the thala-
metries are not always striking. Both types of spindles may be mus generates spindle activity (103,345).
seen simultaneously in stage 2. With deepening sleep (at the In very unusual cases, spindles may be present even in the
transition from stage 2 to 3), an even slower type of spindle waking state, although in attenuated form; this is true for occa-
(around 10/sec) (42) is commonly seen; it is more widespread sional cases of “extreme spindles” (346). Extremely rare are
with a frontal maximum. These spindles are likely to be a fore- cases of typical (unmitigated) spindle activity—along with ver-
runner of rhythmical activity in the 6 to 10/sec range, which is tex waves and K complexes—in the presence of posterior alpha
commonly found in stage 3. This type of activity should be dif- rhythm in a waking individual (347).
ferentiated from sleep spindles (335).
The evolution of sleep spindles in infancy and childhood is K Complexes
presented elsewhere in this volume. Spindles are most impres- K complexes make their appearance in stage 2 sleep and consti-
sive in childhood and adolescence; their voltage tends to tute an impressive response to arousing stimuli (Fig. 11.14).
become smaller throughout adulthood. In old age, spindles of These compounded potentials were first described by Loomis et
low voltage are very common; this is probably not due to old al. (334); the reason for calling them “K complexes” remains
age alone, but due to cerebrovascular disease. Sleep spindles obscure (there have been reports that the naming was made on
share this decline of voltage with vertex and K complexes as age the spur of the moment without attaching any significance to
advances. Sleep spindles have been reported to be more fre- the letter K). Others assume that “K” stands for “knocking” and
quent in women than in men (336); the same type of sex differ- the ensuing arousal activity in the EEG.
ence was also noticed in K complexes. Davis et al. (348) gave an excellent description of the single
The physiologic basis, nature, and significance of spindles components of the K complex. As to the topographical distri-
are beyond the scope of this chapter. Briefly, Hess (319) felt that bution, the K complex shows a maximum over the vertex, but
spindles cannot be explained simply as “slowed beta activity.” there are also K complexes with an indubitable maximum over
Spindles occur independently from faster, mostly 18 to 25/sec,
activity and both patterns may be seen at the same time. Basic
concepts of spindle activity have been discussed by Jankel and
Niedermeyer (337).
The spatial distribution of spindles indicates predominant
frontal lobe involvement. The vertex regions, where spindles
appear first, reflect activity from the most posterior portion of
the supplementary motor region, or frontal lobe. This indicates
a certain relationship to vertex waves (see above) and most K
complexes (see below) recorded from the vertex region. Vertex
waves as well as K complexes are often combined with spindle
trains. There is evidence that vertex waves and K complexes are
arousal responses, whereas the arousal-induced nature of spin-
dles still requires convincing confirmation, despite the encour-
aging findings of Church et al. (338) (also see Ref. 337).
In depth recordings from epileptic patients, sleep spindles
tend to occur in deep frontal regions (from orbital cortex or
from buried gyri of the inferior or middle frontal gyri) while
the patient is drowsy and not yet asleep, according to scalp EEG
evidence (337,339–342). With thalamic depth recordings in
patients with states of chronic pain, independent thalamic
(ventrobasal complex) spindles may appear in states of very
light drowsiness (38). These findings suggest the presence of
multiple generator areas for spindles in humans.
Sleep spindles are probably found in most mammals (see Figure 11.14 Close-up of a K complex, in a patient aged 17 years. Note
Ref. 337). Unfortunately, the term spindles is used quite loosely midline maximum (especially Cz: vertex) and the three principal com-
in experimental neurophysiology. There has been a sizable bulk ponents, the sharp, slow, and fast components.
204 Part II ■ Normal EEG

the frontal midline. Davis et al. had already noted the existence DEEP SLEEP (STAGE 3)
of central and frontal K complexes, but these authors did not
use midline electrodes. Brazier (349) presumed two distinct This stage is recorded much less often than stage 2 under rou-
generators; these were area 6, corresponding with the vertex, tine EEG laboratory conditions. In this stage, the slow back-
and area 9, corresponding with frontal midline. ground activity moves into the foreground and dominates the
As to the wave morphology, the K complex consists of an ini- picture. Delta frequencies in the 0.75 to 3/sec range are partic-
tial sharp component, followed by a slow component that fuses ularly prominent over the anterior regions. Rhythmical activity
with a superimposed fast component. The sharp component is of lesser voltage and in the 5 to 9/sec range is quite common;
biphasic and not seldom multiphasic. Its shape may closely resem- these frequencies probably do not represent a slow spindle
ble that of an isolated vertex wave, but in general the sharp com- equivalent. Sleep spindles in the 10 to 12/sec and even in the 12
ponent of the K complex shows greater complexity and greater to 14/sec ranges are still present but gradually become less
variation from complex to complex. The sharp component was impressive; they seem to be “suffocated” by the luxuriant
thoroughly investigated by Roth and Green (350). The slow com- growth of high-voltage delta activity.
ponent is represented by a large slow wave that may exceed 1000 A close look at the anterior delta activity reveals interspersed
msec in duration; its length and the degree of prominence greatly sharp transients that may be just rudimentary. This indicates a
depend on the filter setting. Shortening of the time constant all certain relationship to K complexes, which seem to be present
but suppresses the slow component. Superimposed on the slow in unending sequences. Nevertheless, more typical isolated K
component are 12 to 14/sec sleep spindles that represent the fast complexes as response to arousing stimuli can also materialize,
component. K complexes may follow each other in long stretches, showing the typical three components and disrupting the gen-
but this is more common in deeper stages of sleep. In the catalog eral slow pattern (Fig. 11.15).
of EEG signals in the Handbook of Electroencephalography and Anterior delta activity with interspersed minor sharp tran-
Clinical Neurophysiology (3), the K complex is described as having sients may form a pattern that has been described as “mitten
only a slow and a fast component; the initiating sharp component pattern.” This pattern has been amply discussed by Gibbs and
is equated with a vertex wave. Gibbs (218), but there are older descriptions by Leemhuis and
The evoked nature of the K complex was clearly demon- Stamps (360), Lyketsos et al. (361), Winfield and Sparer (362),
strated by Niiyama et al. (351) in healthy young adults. These Halász and Kajtor (363), and Gibbs and Gibbs (364). The
authors confirmed N100 and P200 components preceding the authors have categorized the mitten patterns according to the
K complex (313,352) and feel that even an apparently sponta-
neous K complex is, in reality, induced by an afferent stimulus.
The slow component of the K complex has been thought to
be related to a cognitive process or information processing
(353). This is a persuasive concept that is apt to explain the
basic difference between afferent arousing mechanisms of ver-
tex waves and K complexes. The greater spread of the K com-
plex into the frontal region would further bolster this concept.
The K complex is largest in older children and in early adoles-
cence; the sharp component is particularly impressive at that time.
With advancing age, the K complex shows a decline of voltage and
often degenerates into an insignificant slow potential with tiny
superimposed spindle-like waves. A distinction between sponta-
neous and click-evoked K complexes was made by Halász et al.
(354), who investigated the relationship between these two types.
The K complex shows considerable interindividual variability.
Paiva and Rosa (355) distinguish six morphologic types of K com-
plexes, especially on the basis of variations in the negative compo-
nent. These authors also found differences in the configuration of
K complexes with regard to sleep stages 2, 3, and 4. Their study
also confirmed the maximum of K-complex density in stage 2.
According to Weisz et al. (356), the generation of K complexes
is not controlled by the right hemisphere. Unilateral right-sided
control of arousing mechanisms had been proposed by Heilman
and Van den Abell (357) following studies of unilateral spatial
neglect (358). In this condition, K complexes are present, whereas
spindles are absent (356). It might be interesting to note that both Figure 11.15 Deep sleep (non-REM stage 3), in a patient aged 38 years.
spindles and K complexes are absent in fatal thalamic degenera- There is a greater amount of slow activity than in stage 2. Spindles are
tion with insomnia (359), a condition in which the anterior and still abundant but less prominent than in stage 2. K complexes are
the mediodorsal thalamic nuclei degenerate selectively. noted over anterior areas, forming “mitten” patterns.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 205

shape and duration of the sharp component, which is called the This stage of very deep sleep seems to have considerable
“thumb section,” while the slow component would represent endocrinologic effect because the release of pituitary growth
the “hand section” of the mitten. hormone is limited to stages 3 and 4 with a peak at stage 4 (371).
In a sizable number of healthy persons, there is an “alpha Arousal at this stage can be associated with manifestations of
sleep pattern” that reaches its maximum in stage 3. This pattern sleep disorders (somnambulism, nocturnal terror, or enuresis)
was described first by Scheuler and Stinshoff (335) and in predisposed individuals (372), and can produce marked con-
Scheuler et al. (365); it is characterized by rhythmical 7 to fusion (“sleep drunkenness”) in some patients. On the basis of
11/sec activity of frontal maximum, mostly intermingled with coherence analysis, Banquet (373) presumes that a transient par-
slow activity in the delta range. This pattern shows certain peri- tial interhemispheric disconnection occurs in stage 4.
odicities (366). The alpha sleep pattern must be differentiated
from alpha-delta sleep (367,368), a pattern associated with RAPID EYE MOVEMENT (REM) SLEEP
delta sleep deficit and often noted in patients with fibrositis
(369). By contrast, the alpha sleep pattern has been found to be REM sleep should be assessed with polygraphic methods.
associated with a stable sleep organization (370). Under regular EEG laboratory conditions, the observation of
REM sleep requires a long waiting period, because the first
VERY DEEP SLEEP (STAGE 4) phase of REM does not appear before 60 to 90 minutes after
sleep onset; sedation of the patient may even lengthen this
Stage 4 is of rather little interest in the clinical EEG laboratory, period. A prolonged morning or afternoon recording may
although cases of temporal lobe epilepsy may occasionally induce such REM stages, which are quite common in daytime
withhold their spike activity until stages 3 and 4 are reached; naps (374), provided that such naps are long enough.
this, however, is an exception rather than the rule. In this stage, Whenever the electroencephalographer must search specifi-
slow activity in the delta range becomes even more preponder- cally for REM periods (especially when no regional sleep labora-
ant than in stage 3. Spindles become quite rare but are still tory with facilities for polysomnography is available), stress must
detectable, sometimes only when slow frequencies are filtered be laid on the use of two channels for electrooculography (two
out (327) (Fig. 11.16). additional canthus electrodes, connected to the ears), one channel
for cutaneous electromyography (submental region) and a ther-
mocouple or strain gauge for pneumographic documentation.
In patients with a history of narcolepsy and additional
attacks of cataplexy and/or sleep paralysis, REM sleep may
occur at sleep onset. Even with the regular EEG laboratory rou-
tine of short daytime sleep recordings, however, sleep-onset
REM or a REM stage shortly after falling asleep may occur
unexpectedly (Fig. 11.17). Patients with severely disrupted
sleep–waking cycles, in delirium tremens, or with deep-seated
lesions or disturbances involving the brainstem may occasion-
ally show early REM phases.
The REM stage EEG is of low-voltage activity, polyrhythmic,
and similar to stage 1 (light drowsiness); periods of alpha
waves, slightly slower than in wakefulness, or “sawtooth waves”
in the 2 to 6/sec range appear in short bursts over frontal leads
or vertex (375). These sawtooth-like bursts may occur in con-
junction with ocular movements (376). REM sleep also con-
tains a good deal of alpha band activity, which is attenuated
after REM sleep deprivation (377).
The sudden appearance of totally unexpected REM sleep in
a routine EEG laboratory recording must not escape the elec-
troencephalographer, even though the patient has no additional
oculographic leads (also see Ref. 378). The ocular potentials are
mostly quite impressive and almost always give rise to marked
eye movement artifacts in frontopolar and anterior temporal
leads. Once familiarized with the tempo and shape of these dis-
charges, the electroencephalographer will rarely miss REM
states. Of course, the use of polygraphic documentation makes
the task much easier. When an REM phase occurs directly at
Figure 11.16 Very deep sleep (non-REM stage 4), in a patient aged 41 sleep onset, the transition from drowsiness to REM is not very
years. Pronounced generalized 1.5 to 2/ sec activity with frontal maxi- pronounced in the EEG, whereas the change from deep NREM
mum and occasional traces of spindles. sleep to REM is a very dramatic one.
206 Part II ■ Normal EEG

6. Eckhorn R, Schanze T, Brosch M, et al. Stimulus-specific synchro-


nizations in cat visual cortex: multiple microelectrode and corre-
lation studies from several cortical areas. In: Başar E, Bullock TH,
eds. Induced Rhythms in the Brain. Boston: Birkhäuser;
1992:47–80.
7. Gray CM, Engel AK, König P, et al. Mechanisms underlying the
generation of neuronal oscillations in cat visual cortex. In: Başar E,
Bullock TH, eds. Induced Rhythms in the Brain. Boston:
Birkhäuser; 1992:29–45.
8. Walter WG. The location of cerebral tumors by electroencephalog-
raphy. Lancet. 1936;2:305–308.
9. Knott JR. The theta rhythm. In: Remond A, ed. Handbook of
Electroencephalography and Clinical Neurophysiology. Vol 6A.
Amsterdam: Elsevier; 1976:69–77.
10. Silbert PL, Radhakrishnan K, Johnson J, et al. The significance of
the phi rhythm. Electroencephalogr Clin Neurophysiol.
1995;95:71–76.
11. Belsh JM, Chokroverty S, Barabas G. Posterior rhythmic slow
activity in EEG after eye closure. Electroencephalogr Clin
Neurophysiol. 1983;56:562–568.
12. Laugier H, Liberson WT. Contribution à l’étude de l’EEG humain.
C R Soc Biol (Paris). 1937;125:13–17.
13. Kugler J. Elektroenzephalographie in Klinik und Praxis. 3rd ed.
Stuttgart: Thieme; 1981.
14. Kugler J, Laub M. “Puppet show” theta rhythm. Electroencephalogr
Clin Neurophysiol. 1971;31:532–533 [Abstract].
15. Magnus O, Van der Holst M. Zeta waves: a special type of slow delta
waves. Electroencephalogr Clin Neurophysiol. 1987;67:140–146.
16. Siepman TAM, Cherian PJ, Visser GH. Zeta waves, an unusual EEG
finding in structural brain lesions: report of two patients. Am
Figure 11.17 Unexpected sleep-onset REM episode, recorded without J END Technol. 2004;44:24–29.
special ocular leads in a 26-year-old patient with brittle juvenile dia- 17. Cooper R, Winter AL, Crow JJ, et al. Comparison of subcortical,
betes. Note ocular artifacts and general voltage attenuation with mixed cortical and scalp activity using indwelling electrodes in man.
Electroencephalogr Clin Neurophysiol. 1965;18:217–228.
frequencies.
18. IFSECN. A glossary of terms commonly used by clinical electroen-
cephalographers. Electroencephalogr Clin Neurophysiol. 1974;37:
538–548.
AROUSAL FROM SLEEP 19. Markand OM. Alpha rhythms. J Clin Neurophysiol. 1990;7:163–189.
In adolescents and adults, arousal from sleep is a quick process 20. Petersén I, Eeg-Olofsson O. The development of the electroen-
with almost immediate change from sleep into a waking pat- cephalogram in normal children from the age of 1 through 15
years. Nonparoxysmal activity. Neuropädiatrie. 1971;2:247–304.
tern, usually with well-developed posterior alpha rhythm. One
21. Townsend RE, Lubin A, Naitoh P. Stabilization of alpha frequency
single K complex or a sequence of K complexes may mark the by sinusoidally modulated light. Electroencephalogr Clin
transition. More dramatic rhythmic theta activity during Neurophysiol. 1975;39:515–518.
arousal from sleep can be seen in children and is discussed else- 22. Mulholland T. The concept of attention in the electroencephalo-
where in this volume. graphic rhythm. In: Evans CR, Mulholland T, eds. Attention in
Neurophysiology: An International Conference. London:
REFERENCES Butterworth; 1969:100–127.
23. Mulholland T, Evans CR. An unexpected artifact in the human
1. Berger H. Über das Elektrenkephalogramm des Menschen. Arch electroencephalogram and the orienting of the eyes. Nature
Psychiatr Nervenkr. 1929;87:527–570. (Lond). 1965;207:36–37.
2. Jasper HH, Andrews HL. Electroencephalography. III. Normal dif- 24. Fenwick BBC, Walker S. The effect of eye position on the
ferentiation of occipital and precentral regions in man. Arch alpha rhythm. In: Evans CR, Mulholland T, eds. Attention in
Neurol Psychiatry (Chicago). 1938;39:96–115. Neurophysiology: An International Conference. London: Butterworths;
3. Dutertre F. Catalogue of the main EEG patterns. In: Remond A, ed. 1969:128–141.
Handbook of Electroencephalography and Clinical Neurophysiology. 25. Isokawa M, Komisaruk BR. Convergence of finger tremor and EEG
Vol 11A. Amsterdam: Elsevier; 1977:40–79. rhythm at the alpha frequency induced by rhythmical photic stim-
4. Başar E. Brain natural frequencies are causal factors for resonances ulation. Electroencephalogr Clin Neurophysiol. 1983;55:580–585.
and induced rhythms. In: Başar E, Bullock TH, eds. Induced 26. Storm van Leeuwen W, Bekkering DH. Some results obtained with
Rhythms in the Brain. Boston: Birkhäuser; 1992:425–467. the EEG spectograph. Electroencephalogr Clin Neurophysiol. 1958;
5. Bullock TH. Introduction to induced rhythms: a widespread, het- 10:563.
erogeneous class of oscillations. In: Başar E, Bullock TH, eds. 27. Cobb WA. The normal adult EEG. In: Hill D, Parr G, eds.
Induced Rhythms in the Brain. Boston: Birkhauser; 1992:1–26. Electroencephalography. New York, NY: Macmillan; 1963:232–249.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 207

28. Simonova O, Roth B, Stein J. EEG studies of healthy population— 51. Golla F, Hutton EL, Walter WG. The objective study of mental
normal rhythms of resting recording. Act Unv Carol Med (Praha). imagery. I. Physiological concomitance. Appendix on new method
1967;13:543–551. of electroencephalographic analysis. J Ment Sci. 1943;89:216–223.
29. Simon O. (Simonova, O.) Das Elektroenzephalogramm. Munich: 52. Kuhlo W. The beta rhythms. In: Remond A, ed. Handbook of
Urban & Schwarzenberg; 1977. Electroencephalography and Clinical Neurophysiology. Vol 6A.
30. Brazier MAB, Finesinger JE. Characteristics of the normal elec- Amsterdam: Elsevier; 1976:29–46.
troencephalogram. I. A study of the occipital cortical potentials in 53. Lennox WG, Gibbs FA, Gibbs EL. The brain wave pattern, an
500 normal adults. J Clin Invest. 1944;23:303–311. hereditary trait. Evidence from 74 “normal” pairs of twins. J Hered.
31. Wieneke GH, Deinema CHA, Spoelstra P, et al. Normative spectral 1945;36:233–243.
data on alpha rhythm in male adults. Electroencephalogr Clin 54. Travis LE, Gottlober AB. Do brain waves have individuality?
Neurophysiol. 1980;49:636–645. Science. 1936;84:532–533.
32. Kellaway P, Maulsby RL. The Normative Electroencephalographic 55. Veldhuizen RJ, Jonlman EJ, Poortvliet DCJ. Sex differences in age
Data Reference Library. Final report. Houston, TX: Baylor College regression parameters of healthy adults—normative data and
of Medicine; 1966:50–52. practical implications. Electroencephalogr Clin Neurophysiol. 1993;
33. Kiloh LG, McComas AJ, Osselton JW. Clinical Electroence- 86:377–384.
phalography. 3rd ed. London: Butterworths; 1972. 56. Lemere F. The significance of individual differences in the Berger
34. Matousek M, Okawa M, Petersén I. Inter-hemispheric differ- rhythm. Brain. 1936;59:366–375.
ences in normals and in psychiatric patients with normal 57. Saul LJ, Davis H, Davis PA. Correlations between electroen-
EEG. Electroencephalogr Clin Neurophysiol. 1981;52:33P–34P cephalograms and the psychological organization of the indi-
[Abstract]. vidual. Trans Am Neurol Assoc. 1937;63:167–169.
35. Adrian ED, Matthews BHC. The Berger rhythm, potential changes 58. Lindsley DB. Electrical potentials of the brain in children and
from the occipital lobe in man. Brain. 1934;57:345–359. adults. J Gen Psychol. 1938;19:285–306.
36. Berger H. Über das Elektrenkephalogramm des Menschen. Zehnte 59. Henry CE, Knott JR. A note on the relationship between personal-
Mitteilung (10th report). Arch Psychiatr Nervenkr. 1935;103: ity and the alpha rhythm of the electroencephalogram. J Exp
444–454. Psychol. 1941;28:362–366.
37. Albe-Fessard D. Electrophysiological techniques used to differ- 60. Broadhurst A, Glass A. Relationship of personality measures to the
entiate thalamic nuclei. In: Remond A, ed. Handbook of alpha rhythm of the electroencephalogram. Br J Psychiatry.
Electroencephalography and Clinical Neurophysiology. Vol 10B. 1969;115:199–204.
Amsterdam: Elsevier; 1975:46–58. 61. Nowak SM, Marczynski TJ. Trait anxiety is reflected in EEG alpha
38. Gücer G, Niedermeyer E, Long DM. Thalamic recordings in response to stress. Electroencephalogr Clin Neurophysiol. 1981;52:
patients with chronic pain. J Neurol (Berlin). 1978;219:47–61. 175–191.
39. Lopes da Silva F, Van Lierop THMT, Schrijer CF, et al. Organization 62. Berger H. Über das Elektrenkephalogramm des Menschen.
of thalamic and cortical alpha rhythm: spectra and coherences. Vierzehnte Mitteilung (14th report). Arch Psychiatr Nervenkr.
Electroencephalogr Clin Neurophysiol. 1973;35:627–640. 1938;108:407–431.
40. Berger H. Über das Elektrenkephalogramm des Menschen. Sechste 63. Gastaut H. Correlations between the electroencephalographic and
Mitteilung (6th report). Arch Psychiatr Nervenkr. 1933;99: the psychometric variables (M.M.P.I., Rosenzweig, intelligence
555–574. tests). Electroencephalogr Clin Neurophysiol. 1960;12:226–227.
41. Gloor P. The work of Hans Berger. In: Remond A, ed. Handbook of 64. Henry CE. Electroencephalograms of normal children. Monogr
Electroencephalography and Clinical Neurophysiology. Vol 1A. Soc Res Child Dev, Nat Res Council. Vol XI. Washington, DC; 1944.
Amsterdam: Elsevier; 1971:11–24. 65. Kreezer G. Electrical potentials in the brain in certain types of
42. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 1. mental deficiency. Arch Neurol Psychiatry (Chicago). 1936;36:
Cambridge, MA: Addison-Wesley; 1950. 1206–1214.
43. Könönen M, Partanen JV. Blocking of EEG alpha activity during 66. Shagass C. An attempt to correlate the occipital alpha frequency of
visual performance in healthy adults. A quantitative study. the electroencephalogram with performance on a mental ability
Electroencephalography. 1993;87:164–166. test. J Exp Psychol. 1946;36:88–92.
44. Niedermeyer E, Krauss GL, Peyser CE. The electroencephalogram 67. Knott JR. The alpha rhythm. In: Remond A, ed. Handbook of
and mental activation. Clin Electroencephalogr. 1989;20:215–226. Electroencephalography and Clinical Neurophysiology. Amsterdam:
45. Berger H. Über das Elektrenkephalogramm des Menschen. Dritte Elsevier; 1976:7–29.
Mitteilung (3rd report). Arch Psychiatr Nervenkr. 1931;94:16–60. 68. Gasser T, Von Lucadou-Müller I, Verleger R, et al. Correlating EEG
46. Berger H. Über das Elektrenkephalogramm des Menschen. Vierte and IQ: a new look at an old problem using computerized EEG
Mitteilung (4th report). Arch Psychiatr Nervenkr. 1932;97:6–26. parameters. Electroencephalogr Clin Neurophysiol. 1983;55:493–504.
47. Berger H. Über das Elektrenkephalogramm des Menschen. 69. Klimesch W, Doppelmayr M, Schimke H, et al. Alpha frequency,
Siebente Mitteilung (7th report). Arch Psychiatr Nervenkr. reaction time, and the speed of processing information. J Clin
1933;100:301–320. Neurophysiol. 1996;13:511–518.
48. Berger H. Über das Elektrenkephalogramm des Menschen. 70. Jasper HH, Cruikshank RM. Visual stimulation and the after-
Zwölfte Mitteilung (12th report). Arch Psychiatr Nervenkr. 1937; image as affecting the occipital alpha rhythm. J Gen Psychol.
106:165–187. 1937;17:29–48.
49. Adrian ED. The discovery of Berger. In: Remond A, ed. Handbook 71. Fisch BJ, Lemos MS, Hauser WA. Age-related changes in alpha
of Electroencephalography and Clinical Neurophysiology. Vol 1. rhythm frequency and reactivity. Electroencephalogr Clin
Amsterdam: Elsevier; 1971:5–10. Neurophysiol. 1990;75:S45 [Abstract].
50. Davis H, Davis PA. Active potentials of the brain in normal per- 72. Frank, G. Circadian periodicity in cerebral electrical output: rela-
sons and in normal states of cerebral activity. Arch Neurol tion to body temperature and stage of sleep-wakefulness cycles.
Psychiatry (Chicago). 1936;36:1214–1224. Electroencephalogr Clin Neurophysiol. 1964;17:712 [Abstract].
208 Part II ■ Normal EEG

73. Scheich H. Interval histograms and periodic diurnal changes of 95. Adrian ED, Yamagiwa K. The origin of the Berger rhythm. Brain.
human alpha rhythm. Electroencephalogr Clin Neurophysiol. 1935;58:323–351.
1969;26:442 [Abstract]. 96. Aird RB, Garoutte B. Studies on the “cerebral pacemaker.”
74. Harding GFA, Thompson CRS. EEG rhythms and the internal Neurology (Minneapolis). 1958;8:581–589.
milieu. In: Remond A, ed. Handbook of Electroencephalography and 97. Walter DO, Rhodes JM, Brown D, et al. Comprehensive spectral
Clinical Neurophysiology. Vol 6A. Amsterdam: Elsevier; 1976: analysis of human EEG generators in posterior cerebral areas.
176–194. Electroencephalogr Clin Neurophysiol. 1966;20:224–237.
75. Gundel A. The influence of body temperature on the waking EEG. 98. Perez-Borja C, Chatrian GE, Tyce FA, et al. Electrographic pat-
Electroencephalogr Clin Neurophysiol. 1984;57:33P [Abstract]. terns of the occipital lobes in man: a topographic study based on
76. Dubois M, Sato S, Lees DE, et al. Electroencephalographic changes use of implanted electrodes. Electroencephalogr Clin
during whole body hyperthermia in humans. Electroencephalogr Neurophysiol. 1962;14:171–182.
Clin Neurophysiol. 1980;50:486–495. 99. Remond A. The importance of topographic data in EEG phe-
77. Westmoreland B, Klass OW. A distinctive rhythmic EEG discharge nomena and an electric model to reproduce them. In: Walter DO,
of adults. Electroencephalogr Clin Neurophysiol. 1981;51:186–191. Brazier MAB, eds. Advances in EEG Analysis. Amsterdam:
78. Anand BK, China GS, Singh B. Some aspects of electroencephalo- Elsevier; 1968:29–46.
graphic studies in yogis. Electroencephalogr Clin Neurophysiol. 100. Cooper R, Mundy-Castle AC. Spatial and temporal analysis of
1961;13:452–456. the alpha rhythm: a toposcopic analysis. Electroencephalogr Clin
79. Bagchi BK, Wenger MA. Simultaneous EEG and other recordings Neurophysiol. 1960;12:153–165.
during some yogi practices. Electroencephalogr Clin Neurophysiol. 101. Liske E, Hughes HM, Stowe DE. Cross-correlation of human
1958;10:193 [Abstract]. alpha activity: normative data. Electroencephalogr Clin
80. Gastaut H. Vom Berger-Rhythmus zum Alpha-Kult und zur Neurophysiol. 1967;22:429–436.
Alpha-Kultur. Z EEG-EMG. 1974;5:189–199. 102. Inouye T, Shinosaki K, Yagasaki A. The direction of spread of
81. Nowlis D, Kamiya J. The control of electroencephalographic alpha alpha activity over the scalp. Electroencephalogr Clin Neurophysiol.
rhythms through auditory feedback and the associated mental 1983;55:290–300.
activity. Psychophysiology. 1970;6:476–484. 103. Steriade M, Gloor P, Llinas RR, et al. Basic mechanisms of cere-
82. Brown BB. Recognition of aspects of consciousness through asso- bral rhythmical activities. Electroencephalogr Clin Neurophysiol.
ciations with EEG alpha activity represented by a light signal. 1990;76:481–508.
Psychophysiology. 1970;6:442–451. 104. Maddocks JA, Hodge RS, Rex J. Observations on the occurrence
83. Wallace RK. Physiological effects of transcendental meditation. of precentral activities at alpha frequencies. Electroencephalogr
Science. 1970;167:1751–1754. Clin Neurophysiol. 1951;3:370 [Abstract].
84. Wallace RK, Benson M, Wilson AJ. A wakeful hypometabolic phys- 105. Schütz E, Müller HW. Über ein neues Zeichen zentralnervöser
iologic state. Am J Physiol. 1971;221:795–799. Erregbarkeitssteigerung im Elektroenzephalogramm. Klin
85. Banquet JP. Comparative study on the EEG spectral analysis dur- Wochenschr. 1951;29:22–23.
ing sleep and yoga meditation. In: Proceedings of the First European 106. Gastaut H, Terzian H, Gastaut Y. Étude d’une activité élec-
Congress of Sleep Research. Basel; 1973:389–394. troencéphalographique méconnue: “Le rhythme rolandique en
86. Stigsby B, Rodenberg JC, Moth HB. Electroencephalographic find- arceau.” Mars Méd. 1952;89:296–310.
ings during mantra meditation (transcendental meditation). A 107. Gastaut H. Étude électrocorticographique de la réativité des
controlled qualitative study of experienced meditators. Electro- rhythmes rolandiques. Rev Neurol (Paris). 1952;87:176–182.
encephalogr Clin Neurophysiol. 1981;51:434–442. 108. Gastaut H, Dongier M, Courtois G. On the significance of
87. Dongier M, McCallum WC, Torres F, et al. Psychological and psy- “wicket rhythms” (“rhythmes en arceau”) in psychosomatic
chophysiological states. In: Remond A, ed. Handbook of medicine. Electroencephalogr Clin Neurophysiol. 1954;6:687.
Electroencephalography and Clinical Neurophysiology. Vol 16A. 109. Magnus O. The cerebral alpha-rhythm (“rhythme en arceau”).
Amsterdam: Elsevier; 1976:195–256. Electroencephalogr Clin Neurophysiol. 1954;6:349–350.
88. Scherzer E. Das flache EEG als bioelektrischer Ausdruck der 110. Van der Drift JH, Magnus O. The EEG in cerebral ischemic lesions.
Erwartungsspannung (psychogene Alphareduktion). Psychiatr Correlations with clinical and pathological findings. In: Gastaut H,
Neurol (Basel). 1966;152:207–212. Meyer JS, eds. Cerebral Anoxia and the Electroencephalogram.
89. Bishop GH. The interpretation of cortical potentials. Cold Spring Springfield, IL: Charles C Thomas; 1961:180–196.
Harb Symp Quant Biol. 1936;4:305–319. 111. Kuhlman WN. Functional topography of the human mu rhythm.
90. Andersen P, Andersson SA. Physiological mechanisms of the alpha Electroencephalogr Clin Neurophysiol. 1978;44:83–93.
waves. In: Kellaway P, Petersén I, eds. Clinical Electroencephalography 112. Pfurtscheller G, Aranibar A. Occipital rhythm activity within the
of Children. New York, NY: Grune & Stratton; 1968:31–48. alpha band during conditioned externally paced movement.
91. Andersen P, Andersson SA. Thalamic origin of cortical rhythmic Electroencephalogr Clin Neurophysiol. 1978;45:226–235.
activity. In: Remond A, ed. Handbook of Electroencephalography and 113. Pfurtscheller G, Aranibar A. Änderungen in der spontanen EEG-
Clinical Neurophysiology. Vol 2C. Amsterdam: Elsevier; 1974:90–118. Aktivität vor Willkürbewegungen. Neue Wege bei der
92. Frost J. Physiological bases of normal EEG rhythms. In: Remond Untersuchung der zentralen mu-Aktivität. Z EEG-EMG.
A, ed. Handbook of Electroencephalography and Clinical Neuro- 1978;9:18–23.
physiology. Vol 6A. Amsterdam: Elsevier; 1976:150–160. 114. Pfurtscheller G. Central beta rhythms and sensorimotor activities
93. Lopes da Silva F, Van Lierop THMT, Schrijer CF, et al. Essential dif- in man. Electroencephalogr Clin Neurophysiol. 1981;51: 253–264.
ferences between alpha rhythms and barbiturate spindles: spec- 115. Pfurtscheller G. Event-related EEG desynchronization.
tra and thalamocortical coherences. Electroencephalogr Clin Electroencephalogr Clin Neurophysiol. 1990;75:S117 [Abstract].
Neurophysiol. 1973;35:641–645. 116. Pfurtscheller G. Event-related synchronization (ERS): an electro-
94. Watanabe S. Rhythmicity of EEG and stability of alpha rhythm. physiological correlate of cortical areas at rest. Electroencephalogr
Electroencephalogr Clin Neurophysiol. 1981;52:62P [Abstract]. Clin Neurophysiol. 1992;83:62–69.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 209

117. Pfurtscheller G, Klimesch W. Event-related synchronization and 140. Hess R. Postoperative controls. In: Remond A, ed. Handbook of
desynchronization of alpha and beta waves in a cognitive task. In: Electroencephalography and Clinical Neurophysiology. Vol 14C.
Başar E, Bullock TH, eds. Induced Rhythms in the Brain. Boston, Amsterdam: Elsevier; 1975:56–65.
MA: Birkhäuser; 1992:117–128. 141. Kubicki S, Schoppenhorst M, Sack HJ, et al. Post-traumatischer
118. Pfurtscheller G, Neuper C. Simultaneous EEG 10 Hz desynchro- mu wave Fokus: der Einfluß von Schlaf, Pharmaka und
nization and 40 Hz synchronization during finger movements. motorischer Aktivität. Z EEG-EMG. 1973;4:203–209.
NeuroReport. 1992;3:1057–1060. 142. Cobb WA, Guiloff RJ, Cast J. Breach rhythm: the EEG related to skull
119. Pfurtscheller G, Flotzinger D, Neuper C. Differentiation between defects. Electroencephalogr Clin Neurophysiol. 1979;47:251–271.
finger, toe and tongue movement in man based on 40 Hz EEG. 143. Chatrian GE. Characteristics of unusual EEG patterns: incidence,
Electroencephalogr Clin Neurophysiol. 1994;90:456–460. significance. Electroencephalogr Clin Neurophysiol. 1964;17:
120. Pfurtscheller G, Lopes da Silva FH. Event-related EEG/MEG syn- 471–472.
chronization and desynchronization: basic principles. Clin 144. Chatrian GE. The mu rhythms. In: Remond A, ed. Handbook of
Neurophysiol. 1999;110:1842–1857. Electroencephalography and Clinical Neurophysiology. Vol 6A.
121. Pfurtscheller G, Lopes da Silva F, eds. Event-related desynchro- Amsterdam: Elsevier; 1976: 46–69.
nization. Handbook of Electroencephalography and Clinical 145. Chatrian GE, Petersen MC, Lazarte JA. The blocking of the
Neurophysiology, revised series. Vol 6. Amsterdam: Elsevier; 1999. rolandic wicket rhythm and some central changes related to move-
122. Klass D, Bickford RG. Observations on the rolandic arceau ment. Electroencephalogr Clin Neurophysiol. 1959;11: 497–510.
rhythm. Electroencephalogr Clin Neurophysiol. 1957;9:570. 146. Ciganek L. A contribution to the rolandic “arceau” rhythm.
123. Schnell RG, Klass DW. Further observations on the rolandic Electroencephalogr Clin Neurophysiol. 1959;11:185–186.
arceau rhythm. Electroencephalogr Clin Neurophysiol. 1966;20:95 147. Gastaut H, Naquet R, Gastaut Y. A study of the mu rhythm in
[Abstract]. subjects lacking one or more limbs. Electroencephalogr Clin
124. Beek H. Age and the central rhythm “en arceau.” Neurophysiol. 1965;18:720–721.
Electroencephalogr Clin Neurophysiol. 1958;10:356 [Abstract]. 148. Brechet R, Lecasble R. Reactivity of mu rhythm to flicker.
125. Gastaut H, Bacher F, Bert J, et al. Étude des corrélations entre les Electroencephalogr Clin Neurophysiol. 1965;18:721–722 [Abstract].
variables électroencéphalographiques et psychométriques 149. Koshino Y, Niedermeyer E. Enhancement of rolandic mu rhythm
(M.M.P.I., Rosenweig). Rev Neurol (Paris). 1959;101:376–384. by pattern vision. Electroencephalogr Clin Neurophysiol. 1975;38:
126. Gastaut H, Dongier S, Dongier M. Electroencephalographie et 535–538.
nevroses. Rev Neurol (Paris). 1959;101:435–436. 150. Jasper HH, Penfield W. Electrocorticograms in man: effects of
127. Dongier M, Dongier S. Quelques aspects de l’électroencéphalo- voluntary movement upon the electrical activity of the precentral
gramme des névroses. Évol Psychiat. 1958;1:1–18. gyrus. Arch Psychiatr Nervenkr. 1949;183:163–174.
128. Picard P, Laboureur P, Navranne P. Examen électroencéphalo- 151. Graf M, Niedermeyer E, Schiemann J, et al. Electrocorticography:
graphique de personnel navigeant. Méd Aéronaut. 1955;10: information derived from intra-operative recordings during
53–58. seizure surgery. Clin Electroencephalogr. 1984;15:83–91.
129. Simonova O, Roth B, Stein J. An EEG study of healthy subjects. 152. Van Huffelen A, Poortvliet D, Van der Wulp C. Quantitative elec-
Normal rhythms of the resting record. Electroencephalogr Clin troencephalography in cerebral ischemia. Detection of abnor-
Neurophysiol. 1966;20:279 [Abstract]. malities in “normal” EEGs. In: Pfurtscheller G, Jonkman J, Lopes
130. Niedermeyer E, Koshino Y. My-Rhythmus: vorkommen und da Silva FH, eds. Brain Ischemia: Quantitative EEG and Imaging
klinische Bedeutung. Z EEG-EMG. 1975;6:69–78. Techniques. Amsterdam: Elsevier; 1984:3–28.
131. Stroganova TA, Orekhova EV, Posikera IN. EEG alpha rhythm in 153. Arroyo S, Lesser RP, Gordon B, et al. Functional significance of
infants. Clin Neurophysiol. 1999;110:997–1012. the mu rhythm of human cortex: an electrophysiologic study
132. Schoppenhorst M, Brauer F, Freund G, et al. The significance of with subdural electrodes. Electroencephalogr Clin Neurophysiol.
coherence estimates in determining cerebral alpha and mu activ- 1993;87:76–87.
ities. Electroencephalogr Clin Neurophysiol. 1980;48:25–33. 154. Bostem F, Dongier M, Demaret A, et al. Discussion à propos du
133. Koshino Y, Isaki K. Familial occurrence of the mu rhythm. Clin rhythme mu. Rev Neurol (Paris). 1964;111:335–337.
Electroencephalogr. 1986;17:44–50. 155. Bostem F, Dongier M, Demaret A, et al. Discussion on mu rhythm.
134. Storm van Leeuwen W, Wieneke G, Spoelstra P, et al. Lack of Electroencephalogr Clin Neurophysiol. 1965;18:721 [Abstract].
bilateral coherence of mu-rhythm. Electroencephalogr Clin 156. Gastaut H, Broughton R, Regis H, et al. Quelques données nouvelles
Neurophysiol. 1978;44:140–146. à propos due rhythme mu. Rev Neurol (Paris). 1964;111:331–332.
135. Farnarier C, Mattei JP, Naquet R. The mu rhythm of the vertex 157. Gastaut H, Jus A, Jus C, et al. Étude topographique des réactions
and its reactivity: an infrequent observation. Electroencephalogr électroencéphalographiques conditionées chez l’homme.
Clin Neurophysiol. 1981;52:55P [Abstract]. Electroencephalogr Clin Neurophysiol. 1957;9:1–34.
136. Salmelin R, Hari R. Spatiotemporal characteristics of sensorimo- 158. Finley WW, Smith HA, Etherton MD. Reduction of seizures and
tor neuromagnetic rhythms related to thumb movement. normalization of EEG in a severe epileptic following sensorimo-
Neuroscience. 1994;60:537–550. tor biofeedback training. Biol Psychol. 1975;2:189–203.
137. Salmelin R, Hari R. Characterization of spontaneous MEG 159. Kuhlman WN. EEG feedback training: enhancement of
rhythms in healthy adults. Electroencephalogr Clin Neurophysiol. somatosensory cortical activity. Electroencephalogr Clin
1994;91:237–248. Neurophysiol. 1978;45:290–294.
138. Nashmi R, Mendonça AJ, MacKay WA. EEG rhythms of the sen- 160. Kuhlman WN EEG feedback training of epileptic patients: clin-
sorimotor region during hand movements. Electroencephalogr ical and electroencephalographic analysis. Electroencephalogr
Clin Neurophysiol. 1994;91:456–467. Clin Neurophysiol. 1978;45:699–710.
139. Tannier C, Bentzinger C, Feuerstein J. A case of a lesional unilat- 161. Sterman MB, MacDonald LR, Stone RK. Biofeedback training of
eral mu rhythm: clinical and EEG evolution. Electroencephalogr sensorimotor EEG in man and its effect on epilepsy. Epilepsia.
Clin Neurophysiol. 1983;56:21P [Abstract]. (New York). 1974;15:395–416.
210 Part II ■ Normal EEG

162. Roldan E, Lepicovska V, Dostalek C, et al. Mu-like EEG rhythm de l’examen psychologique chez 309 candidats pilotes a l’aéro-
generation in the course of Hatha-yogi exercises. Electro- nautiques. In: Fischgold H, Gastaut H, eds. Conditionnement et
encephalogr Clin Neurophysiol. 1981;52:13P [Abstract]. RŽactivitŽ en ƒlectroencŽphalographie. Amsterdam: Elsevier;
163. Covello A, DeBarros-Ferreira M, Lairy GC. Étude télémétrique 1957:304–314.
des rhythmes centraux chez l’enfant. Electroencephalogr Clin 184. Roger A, Bert J. Étude des corrélations entre les differents vari-
Neurophysiol. 1975;38:307–319. ables EEG. Rev Neurol (Paris). 1959;101:334–360.
164. Yamada T, Kooi KA. Level of consciousness and the mu rhythm. 185. Vogel F, Fujiya Y. The incidence of some inherited EEG variants
Clin Electroencephalogr. 1975;6:80–88. in normal Japanese and German males. Hum Genet.
165. Niedermeyer E. The Generalized Epilepsies. Springfield, IL: Charles 1969;7:38–42.
C Thomas; 1972. 186. Kuhlo W. Slow posterior activities. In: Remond A, ed. Handbook
166. Niedermeyer E. Focal and generalized seizure discharges in the of Electroencephalography and Clinical Neurophysiology. Vol 6A.
electroencephalogram and their response to diazepam. Int Med Amsterdam: Elsevier; 1976:89–104.
Digest. 1972;7:49–61. 187. Fortuin B, Künkel H. The beta-type EEG in adults.
167. Isch-Treussard C. Paroxysmal arched rhythms in the child. Electroencephalogr Clin Neurophysiol. 1983;56:67P [Abstract].
Electroencephalogr Clin Neurophysiol. 1972;33:354 [Abstract]. 188. Kozelak JW, Pedley TA. Beta and mu rhythms. J Clin
168. Niedermeyer E, Long DM, Hendler NH, et al. Chronic pain and Neurophysiol. 1990;7:191–207.
mu rhythm. Electroencephalogr Clin Neurophysiol. 1982;53:30P 189. Drake ME. Clinical correlates of very fast beta activity in the
[Abstract]. EEG. Clin Electroencephalogr. 1984;15:237–241.
169. Pfurtscheller G. Rolandic mu rhythm and assessment of cerebral 190. Vogel F, Götze W. Statistische Betrachtungen über die Beta-
functions. Am J EEG Technol. 1986;26:19–32. Wellen im EEG des Menschen. Dtsch Z Nervenheilkd.
170. Niedermeyer E. An independent alpha-like rhythm in the tempo- 1962;184:112–136.
ral region. Electroencephalogr Clin Neurophysiol. 1990;75:S103 191. Vogel F. Untersuchungen zur Genetik der Beta Wellen im EEG
[Abstract]. des Menschen. Dtsch Z Nervenheilkd. 1962;184:137–173.
171. Niedermeyer E. Alpha-like rhythmical activity of the temporal 192. Vogel F. Zur genetischen Grundlage fronto-präzentraler Beta
lobe. Clin Electroencephalogr 1990;21:210–224. Wellen in EEG des Menschen. Hum Genet. 1966;2:227–237.
172. Niedermeyer E. Ein unabhängiger alpha-artiger Rhythmus über 193. Vogel F. The genetic basis of the normal human electroen-
dem Temporallappen. Das EEG-Labor. 1990;12:165–173. cephalogram (EEG). Hum Genet. 1970;10:91–114.
173. Niedermeyer E. The “third rhythm”: further observations. Clin 194. Ingvar DH. The concept of “the cerebral hyperfrontality?”
Electroencephalogr. 1991;22:83–96. Electroencephalogr Clin Neurophysiol. 1987;67:25P [Abstract].
174. Niedermeyer E. Alpha rhythms as physiological and abnormal 195. Veilleux M, Westmoreland BF, Sharbrough FW. Fast central mid-
phenomena. Int J Psychophysiol. 1997;26:31–49. line rhythm. Electroencephalogr Clin Neurophysiol. 1988;69:
175. Kendel K, Koufen H. EEG-Veränderungen bei zerebralen 91P–92P [Abstract].
Gefäszinsulten des Hirnstamms. Dtsch Z Nervenheilkd. 1970;197: 196. Remond A, Lesèvre N. Remarques sur l’activité cérébrale des
42–55. sujets normaux. La typologie électroencéphalographique dans
176. Berger H. Über das Elektrenkephalogramm des Menschen. ses rapports avec certains caractérs psychologiques. In: Fischgold
Zweite Mitteilung (2nd report). J Psychol Neurol (Leipzig). H, Gastaut H, eds. Conditionnement et RŽactivitŽ en ƒlec-
1930;40:160–179. troencŽphalographie. Amsterdam: Elsevier; 1957:235–255.
177. Finley K. On the occurrence of rapid frequency potential changes 197. Gastaut H. Confrontation entre les données de l’élec-
in the human electroencephalogram. Am J Psychiatry. 1944;101: troencéphalogramme et des examens psychologiques chez 522
194–200. sujets repartis en trois groupes differents. V. Conclusions
178. Gallais P, Collomb H, Milletto G, et al. Confrontation entre les don- d’ensemble. In: Fischgold H, Gastaut H, eds. Conditionnement et
nées de l’électroencéphalogramme et des examens psychologiques RŽactivitŽ en ÉlectroencŽphalographie. Electroencephalogr Clin
chez 522 sujets repartis en trois groupes differents. II. Confrontation Neurophysiol Suppl. 1957;6:321–338.
des données de l’électroencéphalogramme et de l’examen psy- 198. Cohn R. The influence of emotions on the human electroen-
chologique chez 113 jeunes soldats. In: Fischgold H, Gastaut H, eds. cephalogram. J Nerv Ment Dis. 1946;104:351–357.
Conditionnement et RŽactivitŽ en ƒlectroencŽphalographie. 199. Giannitrappani D, Kayton L. Schizophrenia and EEG spectral
Electroencephalogr Clin Neurophysiol Suppl. 1957;6:294–303. analysis. Electroencephalogr Clin Neurophysiol. 1974;36:377–386.
179. Gibbs FA, Gibbs EL, Lennox WG. Electroencephalographic clas- 200. Gastaut H, Lee MC, Laboureur P. Comparative EEG and psycho-
sification of epileptic patients and control subjects. Arch Neurol metric data for 825 French naval pilots and 511 control subjects
Psychiatry (Chicago). 1943;50:111–128. of the same age. Aerosp Med. 1960;31:547–552.
180. Mundy-Castle AC. Theta and beta rhythm in the electroen- 201. Matsuura M, Yamamoto K, Fukuzawa H, et al. Age development
cephalogram of normal adults. Electroencephalogr Clin and sex differences of various EEG elements in healthy children and
Neurophysiol. 1951;3:477–486. adults. Quantification by a computerized wave form recognition
181. Mundy-Castle AC. An analysis of central responses to photic method. Electroencephalogr Clin Neurophysiol. 1985;60:394–406.
stimulation in normal adults. Electroencephalogr Clin 202. Draguhn A, Traub RD, Bibig A, et al. Ripple (around 200-Hz)
Neurophysiol. 1953;5:1–22. oscillations in temporal structures. J Clin Neurophysiol.
182. Obrist WD. The electroencephalogram of normal aged adults. 2000;17:361–376.
Electroencephalogr Clin Neurophysiol. 1954;6:235–244. 203. Niedermeyer E, Sherman DL. Ultrafast frequencies: a new chal-
183. Picard P, Navaranne P, Laboureur P, et al. Confrontations des lenge for electroencephalography with remarks on ultraslow fre-
données de l’électroencéphalogramme et des examens psy- quencies. Clin Electroencephalogr. 2001;32:119–121.
chologiques chez 522 sujets repartis en trois groupes differents. 204. Niedermeyer E, Sherman DL. Ultrafast EEG frequencies— not to
III. Confrontations des données de l’électroencéphalogramme et be neglected in the future. Am J EEG Technol. 2001;41:192–198.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 211

205. Schiff SJ, Colella D, Jacyna GM, et al. Brain chirps: spectro- 229. Riekkinen P, Buzsaki G, Riekkinen P Jr, et al. The cholinergic sys-
graphic signatures of epileptic seizures. Electroencephalogr Clin tem and EEG slow waves. Electroencephalogr Clin Neurophysiol.
Neurophysiol. 2000;111:953–958. 1991;78:89–96.
206. Curio G. Ain’t no rhythm fast enough: EEG bands beyond beta. 230. Van Huffelen AC, Magnus O. 4 c/sec vertex spindles. Electro-
J Clin Neurophysiol. 2000;17:339–340. encephalogr Clin Neurophysiol. 1973;34:543–546.
207. Curio G. Linking 600-Hz “spike-like” EEG/MEG wavlets (“sigma 231. Daoust-Roy J. A waking 4 Hz vertex rhythm: 4 cps vertex spindles
bursts”) to cellular substrates: concepts and caveats. J Clin revisited. Am J EEG Technol. 1989;29:147–163.
Neurophysiol. 2000;17:377–396. 232. Arellano AP, Schwab RS. Scalp and basal recording during men-
208. Traub RD, Jefferys JGR, Whittington MA. Fast Oscillations in tal activity. In: Proceedings of the First International Congress of
Cortical Circuits. Cambridge, MA: MIT Press; 1999. Psychiatry, Paris, 1950.
209. Ziemann U, Rothwell JC. I-waves in motor cortex. J Clin 233. Brazier MAB, Casby JU. Crosscorrelation and autocorrelation
Neurophysiol. 2000;17:397–405. studies of electroencephalographic potentials. Electroencephalogr
210. Vanhatolo S, Holmes MD, Tallgren P, et al. Very slow EEG responses Clin Neurophysiol. 1952;4:201–211.
lateralize temporal lobe seizures. Neurology. 2003;60:1098–1104. 234. Ishihara T, Yoshii N. Multivariate analytic study of EEG and
211. Vanhatolo S, Tallgren P, Anderson S, et al. DC-EEG recording dis- mntal activity in juvenile delinquents. Electroencephalogr Clin
closes prominent very slow activity patterns in preterm infants. Neurophysiol. 1972;33:71–80.
Clin Neurophysiol. 2002;113:1822–1825. 235. Mizuki Y. Frontal midline theta activity during performance of
212. Moruzzi G, Magoun HW. Brain stem reticular formation and mental tasks. Electroencephalogr Clin Neurophysiol. 1982;54:25P
activation of the EEG. Electroencephalogr Clin Neurophysiol. [Abstract].
1949;1:455–473. 236. Mizuki Y. Frontal lobe: mental functions and EEG. Am J EEG
213. Magnes J, Moruzzi G, Pompeiano O. EEG-synchronizing struc- Technol. 1987;27:91–101.
tures in the lower brain stem. In: Wolstenholme GIW, O’Connor 237. Mizuki Y, Tanaka O, Isozaki H, et al. Periodic appearance of theta
M, eds. The Nature of Sleep. London: Churchill Livingstone; rhythm in the frontal midline during performance of a mental
1961:57–78. task. Electroencephalogr Clin Neurophysiol. 1980;49:345–351.
214. Magni F, Moruzzi G, Rossi GF, et al. EEG arousal following inac- 238. Mizuki Y, Takii O, Nishijima H, et al. The relationship between
tivation of the lower brain stem by selective injection of barbitu- the appearance of frontal midline theta activity (Fm theta) and
rates into the cerebral circulation. Arch Ital Biol. 1959;97:33–46. memory function. Electroencephalogr Clin Neurophysiol.
215. Lawson AM, Niedermeyer E. Central pontine myelinolysis: clini- 1983;56:56P [Abstract].
cal and EEG considerations. Am J Electroneurodiagnostic Technol. 239. Takahashi N, Shinomiya S, Mori D, et al. Frontal midline theta
2002;42:151–153. rhythm in young healthy adults. Clin Electroencephalogr.
216. Rosas HD, Koroshetz WS, Chen YI, et al. Evidence for more wide- 1997;28:49–54.
spread cerebral pathology in early HD. Neurology. 2003;60: 240. Gastaut Y. Un signe électroencéphalographique peu connu: les
1615–1620. pointes occipitales survenant pendant l’ouverture des yeux. Rev
217. Jasper HH, Fitzpatrick CA, Colomon P. Analogies and opposites Neurol (Paris). 1951;84:640–643.
in schizophrenia and epilepsy. Am J Psychiatry. 1939;95:835–851. 241. Cobb WA, Pampiglione G. Occipital sharp waves responsive to
218. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 3. visual stimuli. Electroencephalogr Clin Neurophysiol. 1952;4:110–111.
Reading, MA: Addison–Wesley; 1964. 242. Evans CC. Comments on “occipital sharp waves responsive to
219. Adams A. Studies on flat electroencephalogram in man. visual stimuli.” Electroencephalogr Clin Neurophysiol. 1952;4:111.
Electroencephalogr Clin Neurophysiol. 1959;11:34–41. 243. Evans CC. Spontaneous excitation of the visual cortex and associ-
220. Pine I, Pine HM. Clinical analysis of patients with low voltage ation areas—lambda waves. Electroencephalogr Clin Neurophysiol.
EEG. J Nerv Ment Dis. 1953;117:191–198. 1953;5:69–74.
221. Vogel F, Götze W. Familienuntersuchungen zur Genetik des nor- 244. Roth M, Green J. The lambda wave as a normal physiological
malen Elektroenzephalogramms. Dtsch Z Nervenheilkd. 1959; phenomenon in the human electroencephalogram. Nature
178:668–700. (Lond). 1953;172:864–866.
222. Niedermeyer E. Clinical correlates of flat or low voltage records. 245. Chatrian GE. The lambda waves. In: Remond A, ed. Handbook of
Electroencephalogr Clin Neurophysiol. 1963;15:148 [Abstract]. Electroencephalography and Clinical Neurophysiology. Vol 6A.
223. Panzani R, Turner M. Étude électroencéphalographique de la Amsterdam: Elsevier; 1976:123–149.
maladie asthmatique. Correlations électroclinques-Hypothèses 246. Marton M, Szirtes J, Donauer N, et al. Averaged lambda poten-
pathogéniques. Presse Méd. 1952;3:1826–1828. tials in man and monkey. Electroencephalogr Clin Neurophysiol.
224. Lucioni R, Penati G. Sulla frequencza e sul significato in psichia- 1982;53:14P–15P [Abstract].
tria dei tracciati cosidetti piatti. Riv Neurol. 1966;36:200–208. 247. Green J. Some observations on lambda waves and peripheral stim-
225. Chatrian GE. The low voltage EEG. In: Remond A, ed. Handbook ulation. Electroencephalogr Clin Neurophysiol. 1957;9:691–704.
of Electroencephalography and Clinical Neurophysiology. Vol 6A. 248. Barlow JS. Some statistical characteristics of electrocortical activ-
Amsterdam: Elsevier; 1976:77–89. ity in relation to visual oculomotor tracking in man. Bol Inst
226. Walter WG, Dovey VJ. Electroencephalography in cases of sub- Estud Med Biol (Mex). 1963;21:497–518.
cortical tumour. J Neurol Neurosurg Psychiatry. 1944;7:57–65. 249. Barlow JS. Evoked responses in relation to visual perception and ocu-
227. Rugg MD, Dickens AMJ. Dissociation of alpha and theta activity lomotor reaction times in man. Ann NY Acad Sci. 1964;112:432–467.
as a function of verbal and visuospatial tasks. Electroencephalogr 250. Barlow JS, Ciganek L. Lambda responses in relation to visual
Clin Neurophysiol. 1982;53:201–207. evoked responses in man. Electroencephalogr Clin Neurophysiol.
228. Rinaldi F, Himwich HE. Cholinergic mechanisms involved in 1969;26:183–192.
function of mesodiencephalic activating system. Arch Neurol 251. Remond A, Lesèvre N. Étude de la fonction du regard par
Psychiatry. 1955;73:396–402. l’analyse des potentiels évoqués visuels (lumière, patterns
212 Part II ■ Normal EEG

complexes et mouvements des yeux). In: Dubois Poulsen A, Lairy 272. Wittenbecker H, Kubicki S. Statistische Auswertung von 719
GC, Remond A, eds. La Fonction du Regard. Paris: Colloques de Kurzschlafableitungen nach Schlafentzug. Z EEG-EMG. 1976;8:
l’INSERM; 1971:127–175. 205–209.
252. Remond A, Lesèvre N, Torres F. Étude chronotopographique de 273. Rumpl E, Lorenzi E, Bauer G, et al. Zum diagnostischen Wert des
l’activité occipitale moyenne recueilli sur le scalp chez l’homme EEG nach Schlafentzug. Z EEG-EMG. 1977;8:205–209.
en rélation avec le déplacement du regard (complexe lambda). 274. Deisenhammer E, Klingler D. Das Schlafentzug-EEG bei
Rev Neurol (Paris). 1965;113:193–226. Kranken mit gesicherter und fraglicher Epilepsie sowie bei
253. Scott DF, Bickford RG. Electrophysiologic studies during scan- Kranken ohne epileptische Anfälle. Z EEG-EMG. 1978;9:38–42.
ning and passive eye movements in humans. Science. 275. Degen R. A study of the diagnostic value of waking and sleep EEGs
1967;155:101–102. after sleep deprivation in epileptic patients on anticonvulsive ther-
254. Bickford RG, Klass DW. Eye movements and the electroen- apy. Electroencephalogr Clin Neurophysiol. 1980;49: 577–584.
cephalogram. In: Bender MB, ed. The Oculomotor Systems. New 276. Jovanovic UJ. General considerations of sleep and sleep depriva-
York, NY: Hoeber; 1964:293–302. tion. In: Degen R, Rodin EA, eds. Epilepsy, Sleep and Sleep
255. Scott DF, Groethuysen UC, Bickford RG. Lambda responses in Deprivation. 2nd ed. Amsterdam: Elsevier; 1991:205–215.
the human electroencephalogram. Neurology (Minneapolis). 277. Kubicki S, Scheuler W, Wittenbecher H. Short-term sleep EEG
1967;17:770–778. recordings after partial sleep deprivation as a routine procedure
256. Green J. Lambda waves in the EEG and their relation to some to uncover epileptic phenomena: an evaluation of 719 EEG
visual tasks. Proc EPTA. 1954;5:3. recordings. In: Degen R, Rodin EA, eds. Epilepsy, Sleep and Sleep
257. Lesèvre N. Étude des reponses moyennes récueillies sur la région Deprivation. 2nd ed. Amsterdam: Elsevier; 1991:217–230.
postérieure du scalp chez l’homme au cours de l’exploration 278. Klingler D, Tragner H, Deisenhammer E. The nature of the influ-
visuelle (“complexe lambda”). Psychol Franç. 1967;12:26–36. ence of sleep deprivation on the EEG. In: Degen R, Rodin EA,
258. Chatrian GE, Bickford RG, Petersen MC, et al. Lambda waves eds. Epilepsy, Sleep and Sleep Deprivation. 2nd ed. Amsterdam:
with depth electrodes in humans. In: Ramey ER, O’Doherty DS, Elsevier; 1991:231–233.
eds. Electrical Studies on the Unanesthetized Brain. New York, NY: 279. Naitoh P, Dement W. Sleep deprivation in humans. In: Remond
Hoeber; 1960:291–310. A, ed.-in-chief. Handbook of Electroencephalography and Clinical
259. Billings RJ. The origin of the occipital lambda wave in man. Neurophysiology. Vol 7A. Amsterdam: Elsevier; 1976:146–151.
Electroencephalogr Clin Neurophysiol. 1989;72:95–113. 280. Rodin EA, Luby ED, Gottlieb S Jr. The electroencephalogram
260. Rechtschaffen A, Kales A. A Manual of Standardized during prolonged experimental sleep deprivation. Electro-
Terminology, Techniques, and Scoring System for Sleep Stages of encephalogr Clin Neurophysiol. 1962;14:544–551.
Human Subjects. University of California, LA: Brain Information 281. Heinemann LG. Der Mehrtägige Schlafentzug in der experi-
Service; 1968. mentellen Psychoseforschung: psychopathologie und EEG. Arch
261. Kubicki S, Herrmann WM. The future of computer-assisted Psychiatr Nervenkr. 1966;208:117–197.
investigation of the polysomnogram: sleep-microstructure. J Clin 282. Johnson LC. Psychological and physiological changes following
Neurophysiol. 1996;13:285–294. total sleep deprivation. In: Kales A, ed. Sleep: Physiology and
262. Kubicki St, Meyer Ch, Rohmel J. Dies Schlafspindelperiodik. Z Pathology. Philadelphia: JB Lippincott; 1969:206–220.
EEG-EMG. 1986;17:55–61. 283. Jovanovic UJ, Liebaldt GP, Mühl M, et al. Schlafentzug and seine
263. Kubicki St, Höller L, Pastelak-Price C. Subvigil beta activity: a Begleiterscheinungen. Arch Psychiatr Nervenkr. 1971;124: 183–202.
study of fast EEG patterns in drowsiness. Am J EEG Technol. 284. Naitoh P, Kales A, Kollar EJ, et al. Electroencephalographic
1987;27:15–31. changes after prolonged deprivation of sleep. Electroencephalogr
264. Gastaut H, Gomez-Almanzar M, Taury M. Der provozierte Clin Neurophysiol. 1969;27:2–11.
Mittagsschlaf: eine einfache, erfolgreiche Methode zur 285. Welch LK, Stevens JB. Clinical value of the electroencephalogram
Schlafaktivierung bie Epileptikern. Z EEG-EMG. 1983;14:6–11. following sleep deprivation. Aerosp Med. 1971;42:349–351.
265. Gastaut H, Gomez-Almanzar M, Taury M. The enforced nap: a 286. Klass DW, Fischer-Williams M. Sensory stimulation, sleep and
simple effective method of inducing sleep activation in epileptics. sleep deprivation. In: Remond A, ed.-in-chief. Handbook of
In: Degen R, Niedermeyer E, eds. Epilepsy, Sleep and Sleep Electroencephalography and Clinical Neurophysiology. Vol 3D.
Deprivation. Amsterdam: Elsevier; 1984:75–83. Amsterdam: Elsevier; 1976:5–73.
266. Niedermeyer E, Höller L. Kurzschlaf im EEG-Eine Fundgrube sonst 287. Aserinsky E, Kleitman N. Regularly occurring episodes of eye
übersehener EEG-Abnormitäten. Z EEG-EMG. 1984;15:57–66. mobility and concomitant phenomena during sleep. Science.
267. Bennett DR. Sleep deprivation and major motor convulsions. 1953;118:273–274.
Neurology (Minneapolis). 1963;13:953–958. 288. Dahlström A, Fuxe K. Evidence for the existence of monoamino-
268. Bennett DR, Mattson RH, Ziter FA, et al. Sleep deprivation: neu- containing neurons in the cerebral nervous system. I.
rological and electroencephalographic effects. Aerosp Med. Demonstration of monamines in the cell bodies of brain stem
1964;35:888–890. neuron. Acta Physiol Scand Suppl. 1964;232:1–55.
269. Mattson RH, Pratt KL, Calverley JR. Electroencephalograms of 289. Jouvet M. Biogenic amines and the states of sleep. Science.
epileptics following sleep deprivation. Arch Neurol (Chicago). 1969;163:32–41.
1965;13:310–315. 290. Jouvet M. Some monoaminergic mechanisms controlling sleep
270. Oller-Daurella L. La privation de sommeil comme méthode d’ac- and waking. In: Karczmar AG, Eccles JC, eds. Brain and Human
tivation de l’EEG chez l’épileptique. Rev Neurol (Paris). Behavior. New York, NY: Springer; 1972:131–161.
1966;115:530–535. 291. Jouvet M, Delorme F. Locus coeruleus et sommeil paradoxal. CR
271. Pratt KL, Mattson RH, Weikers NJ, et al. EEG activation of Soc Biol (Paris). 1965;159:895–899.
epileptics following sleep deprivation: a prospective study of 114 292. Ungerstedt U. Stereotaxic mapping of the monoamine pathway
cases. Electroencephalogr Clin Neurophysiol. 1968;24:11–15. in the rat brain. Acta Physiol Scand Suppl. 1971;367:1–48.
Chapter 11 ■ Normal EEG and Sleep: Adults and Elderly 213

293. Dement W, Kleitman N. Cyclic variations in EEG during sleep 315. Niedermeyer E, Pribram HFW. Unilateral suppression of vertex
and their relation to eye movements, body motility and dream- sharp waves in the sleep electroencephalogram (case report).
ing. Electroencephalogr Clin Neurophysiol. 1957;9:673–690. Electroencephalogr Clin Neurophysiol. 1966;20:401–404.
294. Erwin CW, Somerville ER, Radtke RA. A review of electroencephalo- 316. Wright EA, Gilmore RL. Features of the geriatric EEG: age-
graphic features normal of sleep. Clin Neurophysiol. 1984;1:253–274. dependent incidence of POSTS. Electroencephalogr Clin
295. Guilleminault C, Baker TL. Sleep and electroencephalography: Neurophysiol. 1984;58:49P [Abstract].
points of interest and points of controversy. J Clin Neurophysiol. 317. Prior PF, Deacon PA. Spontaneous sleep in healthy subjects in
1984;1:275–291. long-term serial electroencephalograph readings. Electro-
296. Schieber JP, Muzet A, Ferrière PJR. Les phases d’activation tran- encephalogr Clin Neurophysiol. 1969;27:422–424.
sitoire spontanées au cours du sommeil normal chez l’homme. 318. Roth M, Shaw J, Green J. The lambda wave as a normal physio-
Arch Sci Physiol. 1971;25:443–464. logical phenomenon in the human electroencephalogram.
297. Terzano MG, Mancia D, Zacchetti O, et al. The significance of Nature (Lond). 1953;172:864–866.
cyclic EEG changes in Creutzfeldt-Jakob disease: prognostic value 319. Hess R. The electroencephalogram in sleep. Electroencephalogr
of their course in 9 patients. Ital J Neurol Sci. 1981;3:243–254. Clin Neurophysiol. 1964;16:44–55.
298. Terzano MG, Parrino L. The phenomena of cyclic alternating 320. Arfel G, Laurette G. Occipital sharp waves of sleep contrasted
pattern. Clin Neurol Neurosurg. 1987;89:164–167. with lambda waves. Clin Electroencephalogr. 1973;4:16–32.
299. Terzano MG, Parrino L. Functional relationship between micro- 321. Kugler J, Laub M. Lambdoid-Wellen (RHO-Wellen) in Schlaf. In:
and macrostructure of sleep. In: Terzano MG, Halász P, Declerck Jovanovic UJ, ed. The Nature of Sleep. Stuttgart: Fischer; 1973:
AC, eds. Phasic Events and Dynamic Organization of Sleep. New 31–35.
York, NY: Raven Press; 1991:101–119. 322. Kugler J. Lambda waves and rho waves. Electroencephalogr Clin
300. Halász P. Information processing during sleep: introduction. In: Neurophysiol. 1976;40:193–194 [Abstract].
Koella WP, Obal F, Schulz H, et al., eds. Sleep 1986. Stuttgart: 323. Vignaendra V, Matthews RL, Chatrian GE. Positive occipital
Fischer; 1988:77–78. sharp transients of sleep: relationships to nocturnal sleep cycle in
301. Evans BM. Periodic activity in cerebral arousal mechanisms—the man. Electroencephalogr Clin Neurophysiol. 1974;37:239–246.
relationship to sleep and brain damage. Electroencephalogr Clin 324. Brenner RP, Zauel DW, Carlow TJ. Positive occipital sharp transients
Neurophysiol. 1992;83:130–137. of sleep in the blind. Neurology (Minneapolis). 1978;28:609–612.
302. Goldie L, Green JM. Paradoxical blocking and arousal in the 325. Pristasova E, Prochazka M. Bioccipital theta rhythmic and positive
drowsy state. Nature (Lond). 1960;187:952–953. occipital sharp waves of sleep. Electroencephalogr Clin Neurophysiol.
303. Santamaria J, Chiappa KH. The EEG in Drowsiness. New York: 1981;52:1P–2P [Abstract].
Demos; 1987. 326. Niedermeyer E, Lentz WJ. Dreaming in non-REM sleep. Waking
304. Broughton R, Hasan J. Quantitative topographic electroen- Sleeping. 1976;1:49–51.
cephalographic mapping during drowsiness and sleep onset. 327. Dement WC. Some Must Watch While Some Must Sleep. San
J Clin Neurophysiol. 1995;12:372–386. Francisco: San Francisco Book Co; 1976.
305. Koshino Y, Nishio M, Murata T, et al. The influence of light 328. Röschke J, Başar, E. The EEG is not a simple noise: strange attrac-
drowsiness on the latency and amplitude of P300. Clin tors in intracranial structures. In: Başar E, ed. Chaos in Brain
Electroencephalogr. 1993;24:110–113. Function. Berlin: Springer; 1990:49–62.
306. Wada Y, Nanbu Y, Koshino Y, et al. Inter-and intrahemispheric 329. Niedermeyer E. The electroencephalogram and vertebrobasilar
EEG coherence during light drowsiness. Clin Electroencephalogr. artery insufficiency. Neurology (Minneapolis). 1963;13:412–422.
1996;27:24–88. 330. Engel GL, Romano J. Delirium, a syndrome of cerebral insuffi-
307. Liberson WT. Functional electroencephalography in mental dis- ciency. J Chron Dis. 1959;9:260 (Quoted after Kiloh LG, McComas
orders. Dis Nerv Syst. 1945;5:357–364. AJ, Osselton JW. Clinical Electroencephalography. London:
308. Davis PA. Effects of acoustic stimuli on the waking human brain. Butterworth; 1972).
J Neurophysiol. 1939;2:494–499. 331. Lorimer FM. Further clinical observations on exceedingly fast
309. Bancaud J, Bloch V, Paillard J. Contribution E.E.G. à l’étude des activity, a rare electroencephalographic abnormality. Clin
potentiels évoqués chez l’homme au niveau de vertex. Rev Neurol Electroencephalogr. 1970;1:84–91.
(Paris). 1953;89:399–418. 332. Marshall L, Molle M, Schreiber H, et al. Scalp recorded direct
310. Gastaut Y. Les pointes négatives évoquées sur le vertex: leur sig- current potential shifts associated with the transition to sleep in
nification psychophysiologique et pathologique. Rev Neurol man. Electroencephalogr Clin Neurophysiol. 1994;91:346–352.
(Paris). 1953;89:382–389. 333. Luthringer R, Branderberger G, Schaltenbrand N, et al. Slow
311. Kooi KA, Tipton AC, Marshall RE. Polarities and field configura- wave electroencephalic activity parallels renin oscillations during
tion of the vertex components of the human auditory evoked sleep in humans. Electroencephalogr Clin Neurophysiol. 1995;95:
response: a reinterpretation. Electroencephalogr Clin Neuro- 318–322.
physiol. 1971;31:166–169. 334. Loomis AL, Harvey EN, Hobart GA. Distribution of disturbance-
312. Storm van Leeuwen W. Auditory evoked responses (AERs). In: patterns in the human electroencephalogram with special refer-
Remond A, ed.-in-chief. Handbook of Electroencephalography and ence to sleep. J Neurophysiol. 1938;1:413–430.
Clinical Neurophysiology. Vol 8A. Amsterdam: Elsevier; 1975:71–85. 335. Scheuler W, Stinshoff D. Das Alpha-Schlafmuster-eine kaum
313. Weitzman ED, Kremen H. Auditory evoked responses during dif- beachtete EEG-Variante. Z EEG-EMG. 1982;13:34–41.
ferent stages of sleep in man. Electroencephalogr Clin Neuro- 336. Gaillard J-M, Baudat J, Blois R. The effect of sex depressive state
physiol. 1965;18:65–70. and minor tranquilizers on K potentials and spindles during
314. Davis H, Mast T, Yoshie N, et al. The slow response of the human sleep. Clin Neurol Neurosurg. 1987;89:172–173.
cortex to auditory stimuli. Electroencephalogr Clin Neurophysiol. 337. Jankel WR, Niedermeyer E. Sleep spindles. J Clin Neurophysiol.
1966;21:105–113. 1985;2:1–35.
214 Part II ■ Normal EEG

338. Church MW, Johnson LC, Seales DM. Evoked K complexes and 359. Tinuper P, Montagna P, Medori R, et al. The thalamus participates
cardiovascular responses to spindle-synchronous and spindle- in the regulation of the sleep-waking cycle. A clinico-pathological
asynchronous stimulus clicks during NREM sleep. study in fatal familial thalamic degeneration. Electroencephalogr
Electroencephalogr Clin Neurophysiol. 1978;45:443–453. Clin Neurophysiol. 1998;73:117–123.
339. Caderas M, Niedermeyer E, Uematsu S, et al. Sleep spindles 360. Leemhuis AJ, Stamps F. Mitten patterns in electroencephalogram
recorded from deep cerebral structures in man. Clin of patients with Parkinsonism. In: Proceedings of the American
Electroencephalogr. 1982;13:216–225. EEG Society. Atlantic City, NJ; 1949.
340. Niedermeyer E, Jankel WR. Falling asleep: depth EEG and ther- 361. Lyketsos G, Belinson L, Gibbs FA. Electroencephalograms of
mographic observations. Electroencephalogr Clin Neurophysiol. nonepileptic psychotic patients awake and asleep. Arch Neurol
1984;58:8P [Abstract]. Psychiatry (Chicago). 1953;69:707–712.
341. Niedermeyer E, Jankel WR, Uematsu S. Falling asleep: observa- 362. Winfield DL, Sparer PJ. The electroencephalogram in paralysis
tions and thoughts. Am J EEG Technol. 1986;26:166–175. agitans. Dis Nerv Syst. 1954;15:144–120.
342. Zobniw AM, Yarworth S, Niedermeyer E. Depth electroen- 363. Halasz P, Kajtor F. "Mittens"—a new form of electroencephalo-
cephalography. J Electrophysiol Technol. 1975;1:215–240. graphic wave. Ideggyogy Szemle (Hungarian). 1962;15:46–57
343. Bremer F. Cerebral and cerebellar potentials. Physiol Rev. 1958; (quoted after Gibbs FA, Gibbs EL, 1964).
38:357–388. 364. Gibbs FA, Gibbs EL. The mitten pattern: an electroencephalo-
344. Andersen P, Eccles JC. Inhibitory phasing of neuronal discharge. graphic abnormality correlating with psychosis. J Neuro-
Nature (Lond). 1962;196:645–647. psychiatry. 1963;5:6–13.
345. Steriade M, Deschênes M. Intrathalamic and brainstem-thalamic 365. Scheuler W, Stinshoff D, Kubicki St. The alpha sleep pattern.
networks involved in resting and alert stages. In: Bentivoglio M, Differentiation from other sleep patterns and effects of hyp-
Spreafico R, eds. Cellular Thalamic Mechanisms. Amsterdam: notics. Neuropsychobiology. 1983;10:395–399.
Elsevier; 1988:37–62. 366. Scheuler W, Rappelsberger P, Schmatz F, et al. Periodicity analysis
346. Gibbs EL, Gibbs FA. Extreme spindles: correlation of electroen- of sleep EEG in the second and minute ranges—example of appli-
cephalographic sleep pattern with mental retardation. Science. cation in different alpha activities in sleep. Electroencephalogr Clin
1962;138:1106–1107. Neurophysiol. 1990;76:222–234.
347. Niedermeyer E, Singer HS, Folstein SE, et al. Hypersomnia with 367. Hauri P, Hawkins DR. Alpha delta sleep. Electroencephalogr Clin
simultaneous waking and sleep patterns in the electroencephalo- Neurophysiol. 1973;34:233–237.
gram. A case report with neurotransmitter studies. J Neurol 368. McNamara ME. Alpha sleep: a mini review and update. Clin
(Berlin). 1979;221:1–13. Electroencephalogr. 1993;24:192–193.
348. Davis H, Davis PA, Loomis AL, et al. Electrical reactions of human 369. Moldofsky H, Lue FA. The relationship of alpha and delta EEG
brain to auditory stimulation during sleep. J Neurophysiol. 1939; frequencies to pain and mood in “fibrositis” patients treated with
21:500–514. chlorpromazine and I-tryptophan. Electroencephalogr Clin
349. Brazier MAB. The electrical fields at the surface of the head dur- Neurophysiol. 1980;50:71–80.
ing sleep. Electroencephalogr Clin Neurophysiol. 1949;1:195–204. 370. Scheuler W, Kubicki St, Marquardt J, et al. The alpha-sleep-pat-
350. Roth M, Green J. The form, voltage distribution and physiological tern-quantitative analysis and functional aspects. In: Koella WP,
significance of the K complex. Electroencephalogr Clin Neurophysiol. Obal F, Schulz H, et al., eds. Sleep 1986. Stuttgart: Fischer; 1988.
1956;8:385–402. 371. Sassin J, Parkee D, Mace J, et al. Human growth hormone release
351. Niiyama Y, Satoh N, Kutsuzawa O, et al. Electrophysiological evi- relation to slow-wave sleep and sleep-waking cycle. Science.
dence suggesting that sensory stimuli of unknown origin induce 1969;165:513–515.
spontaneous K complex. Electroencephalogr Clin Neurophysiol. 372. Gastaut H, Broughton R. A clinical and polygraphic study of
1996;98:394–400. episodic phenomena during sleep. In: Wortis J, ed. Recent
352. Ujszaszi J, Halász P. Long latency evoked potential components in Advances in Biological Psychiatry. Vol VII. London: Plenum Press;
human slow wave sleep. Electroencephalogr Clin Neurophysiol. 1964:197–221.
1988;69:516–522. 373. Banquet JP. Inter- and intrahemispheric relationships of the EEG
353. Niiyama Y, Fushimi M, Sekine A, et al. K complex evoked in NREM activity during sleep in man. Electroencephalogr Clin Neuro-
sleep is accompanied by a slow negative potential related to cogni- physiol. 1983;55:51–59.
tive process. Electroencephalogr Clin Neurophysiol. 1995;95:27–33. 374. Salzarulo P. Etude é’lectroencéphalographique et polygraphique
354. Halász P, Pal I, Rajna P. Relationship between spontaneous and du sommeil d’aprés-midi chez le sujet normal. Electroencephalogr
evoked K complexes. Electroencephalogr Clin Neurophysiol. Clin Neurophysiol. 1971;30:399–407.
1982;53:7P [Abstract]. 375. Passouant P. EEG and sleep. Electro-clinical semeiology. In:
355. Paiva T, Rosa A. The K complex variability in normal subjects. In: Remond A, ed.-in-chief. Handbook of Electroencephalography
Terzano MG, Halász P, Declerck AC, eds. Phasic Events and and Clinical Neurophysiology. Vol 7A. Amsterdam: Elsevier; 1975:
Organization of Sleep. New York: Raven Press; 1991:167–187. 3–11.
356. Weisz J, Soroker N, Oksenberg A, et al. Effects of hemi-thalamic 376. Schwartz RA. EEG et mouvements oculaires dans le sommeil de
damage on K-complexes evoked by monaural stimuli during nuit. Electroencephalogr Clin Neurophysiol. 1962;14:126–128.
mid-afternoon sleep. Electroencephalogr Clin Neurophysiol. 377. Roth C, Achermann P, Borbely AA. Alpha activity in the human
1995;94:148–150. REM sleep topography and effect of REM sleep deprivation. Clin
357. Heilman KM, Van den Abell T. Right hemispheric dominance for Neurophysiol. 1999;110:632–635.
mediating cerebral activation (quoted after Weisz et al., 1995). 378. Matsuo F. Recognition of REM sleep in standard EEG.
Neuropsychologia. 1979;17(3–4):315–321. Electroencephalogr Clin Neurophysiol. 1981;52:490–493.
358. Heilman KM, Schwartz HD, Watson RT. Hypoarousal in patients
with neglect syndrome and indifference. Neurology.
1978;28:229–232.
CHAPTER

Activation Methods
TAKEO TAKAHASHI AND KEITH H. CHIAPPA 12
Various activation procedures—such as hyperventilation, inter- With regard to the underlying mechanism of HV’s provoca-
mittent photic stimulation, or sleep—may be used to enhance tion of slowing and seizure discharges, Gibbs et al. (5) suggested
preexisting abnormalities and induce abnormal findings in an that diffuse slowing is caused by inadequate compensatory
otherwise normal electroencephalogram (EEG). This chapter vasoconstriction of the cerebrum in response to systemic
discusses common activation methods as well as methods that
hypocapnia. According to Gotoh et al. (6), diffuse slowing is
are less frequently carried out in EEG laboratories. Sleep-
considered to be the direct result of cerebral ischemic anoxia
induced, sleep-deprived, and especially psychotropic drug-
induced effects are not discussed here; they are covered in other resulting from hypocapnic cerebral vasoconstriction. Yamatani
chapters. et al. (7), measuring cerebral blood flow in the right carotid
artery during HV, reported that decreased PCO2 and cerebral
blood flow were the fundamental factors causing EEG slowing.
HYPERVENTILATION Fisch and So (8) summarized the physiologic basis of the EEG
response to HV as the following: alteration of PCO2, rather
Precipitation of seizures by hyperventilation (HV) (1) was than pH or PO2, is the most important factor in producing the
known prior to the discovery of the human EEG. Therefore, EEG response to HV, whereas the most obvious and dramatic
EEG activation by HV has been widely used in almost all clin- physiologic effect of HV is decreased cerebral blood flow.
ical EEG laboratories since its introduction, and Nims et al. (2) Sherwin (9) attributed the diffuse slowing to synchronous
showed the provocative efficacy of generalized synchronous activity in the nonspecific thalamocortical projecting systems,
paroxysmal discharges and of absence seizures produced by which become more active in hypocapnia. Patel and Maulsby
this test. This method consists of deep and regular respiration (10) raised the possibility that hypocapnia induced by HV
at a rate of about 20/min for a period of 2 to 4 minutes. In decreased activity in mesencephalic reticular formation, which
adults, such HV will cause air exchange of 20 to 50 L/min and then caused EEG slowing, just as drowsiness and sleep produce
a drop in partial pressure of carbon dioxide (PCO2) in the EEG slowing. Similar mechanisms to those described above are
range of 4 to 7 mm Hg (3). considered to be important for provoking paroxysmal dis-
An understanding of the time course and magnitude of the charges (11); hyperexcitability of neurons may be induced by
systematic changes in blood gases produced by routine HV in respiratory alkalosis (12). However, the change from the pure
adults is important for clinical interpretations of EEGs. delta response to the appearance of intermixed spikes and clear-
Achenbach-Ng et al. (4) studied these changes in adults and cut spike–wave discharges remains obscure. Interestingly, it has
children and found that during 3 minutes of HV, the PCO2 fell been reported that intravenous administration of diazepam
a mean of 18 mm Hg and the partial pressure of oxygen (PO2) prevented HV-induced spike patterns, though it failed to atten-
rose 7 mm Hg, the former with a near-linear response curve uate the delta response (13).
(see Fig. 12.1). Note that PCO2 reached its nadir 30 seconds The magnitude of the HV response depends on a number of
after the end of the 3 minutes of HV. From this point, after the factors (14). First, vigorous exchange of air can enhance activa-
command to “breathe normally,” PCO2 rose linearly to return tion effects. For the purpose of routine examination with HV,
to resting levels in about 5.5 minutes, more slowly for children. however, it is suggested that the rate of breathing be as close as
As PCO2 rose during this post-HV period, PO2 fell to 25 mm possible to that of the resting rhythm (15 to 20 breaths/min)
Hg below resting values at 5.2 minutes after cessation of HV, (14). Second, age is an important factor. Slow waves appear much
and then rose to resting levels over the following 11.6 minutes more abruptly, are more pronounced, and persist longer in chil-
(Fig. 12.1). Any perturbation of ventilation in the post-HV dren than in adults. The degree and abruptness of the response
period (e.g., talking) prolonged these steps. These findings min- are related directly to age (15). The most dramatic EEG responses
imize the clinical importance of slowing in the EEG that out- to HV usually occur between the ages of 8 and 12 years (16).
lasts the HV period, and correlate with the “rebuildup” of According to Gibbs et al. (5), in children between 3 and 5 years of
post-HV slowing seen in moyamoya disease (see below). age, 97% of epilepsy patients and 70% of normal subjects showed
The characteristic EEG response to HV, most prominent in diffuse slowing with HV; after 20 years of age, more than 40% of
children, consists of a fluctuating increase of bilaterally syn- epilepsy patients showed diffuse slowing, whereas it was seen in
chronous slow activity and slowing of alpha and beta rhythms. less than 10% of normal subjects. The areas most affected by HV
In normal adults, although the slowing is generally not marked, in children are occipitotemporal regions (17,18). Delta waves
there are wide differences among individuals. tend to appear initially in the posterior regions and spread
215
216 Part II ■ Normal EEG

Figure 12.1 Time course and magnitude of absolute PCO2


and PO2 changes with 3 minutes of hyperventilation in nine
normal adult subjects. The error bars are 1 standard devia-
tion. A transcutaneous heated membrane technique was
used for the blood gas measurements.

forward in the younger age group, whereas they tend to appear in Unusually prolonged post-HV high-voltage slowing may be
the frontal regions and spread backward in the older age group seen in patients with syncopal attacks of various etiologies (26).
(19). Third, blood glucose level is also important in determining Buildup of slow waves after the end of HV (“re-buildup”) may
the degree of HV response. In adults, a low blood glucose level be a diagnostic finding in children with moyamoya disease (27).
(less than 80 mg/dL) tends to enhance appearance of slow waves; In a study that addressed three cases of moyamoya disease,
a high level (more than 120 mg/dL) tends to inhibit or prevent Kameyama et al. (28) reported that this EEG re-buildup was
such an effect (20). Delta waves induced by HV can occasionally noted about 5 minutes after HV cessation; moreover, correspon-
be the first indication of pathologic hypoglycemia secondary to ding decreases in cerebral blood flow and PO2 were documented
an islet cell tumor in a patient referred for an EEG (19). Fourth, with it. Such a post-HV hypoxia 5 minutes after cessation of
an erect position as compared to a reclining position enhances HV was also found in a study of nine normal adult subjects (4)
the effect of HV; EEG slowing occurs earlier and with greater (Fig 12.1). Note that it is suggested that HV should be avoided
intensity. This is thought to be a result of relative cerebral anoxia in patients with moyamoya disease (29).
(21). Diffuse slowing induced by HV usually disappears rapidly The American EEG Society Guidelines (AEEGS) Committee
after ceasing HV; it may persist for up to 30 seconds in normal (30) proposed the following minimum technical requirements:
adults, but even further persistence at any age, by itself, is not nec- HV should be used routinely unless other medically justifiable
essarily an abnormality. reasons (e.g., a recent intracranial hemorrhage, significant car-
Aside from diffuse slowing, HV may induce diffuse sharp diopulmonary disease, sickle cell disease or trait, or patient
waves or spike–wave discharges of epileptogenic significance; it inability or unwillingness to cooperate) contraindicate it. It
is particularly effective in eliciting bilaterally synchronous should be performed for a minimum of 3 minutes. At times,
spike–wave discharges in a patient with generalized epilepsies. HV must be performed for a longer period in order to obtain
Dalby (22) found that patients with absence seizures were more adequate EEG activation. The guidelines suggest continued
sensitive to HV than patients with nonabsence seizures, with the recording for at least 1 minute after cessation of overbreathing,
incidence of spike–wave paroxysms being 50% in the former but the slow time course of the recovery of PCO2 levels dis-
and 25% in the latter. During HV, not only absence seizures but cussed above and the findings in the moyamoya patients indi-
also complex partial seizures are induced in epilepsy patients. In cate that at least 6 minutes of post-HV recording would be
general, latent abnormalities are likely to be activated by HV. prudent.
Slow-wave foci associated with localized lesions may also be Regarding the clinical interpretation of EEG changes seen
aggravated; abnormalities in temporal regions are more prone to during HV, a few simple principles should be kept as guidelines,
accentuation than those elsewhere (23). Miley and Forster (24) and any clinical interpretation that exceeds these guidelines
reported that vigorous HV of longer duration induced abnormal should have a firm, rational basis for the variance. These guide-
discharges with or without clinical seizures in 11% of epilepsy lines are: HV-induced slowing may be of any amplitude, loca-
patients with complex partial seizures. Arain et al. (25) have sug- tion, rhythmicity, and time course and should still be
gested that, because of the high incidence of significant clinical considered to be within normal limits unless it is significantly
and EEG events provoked by HV, this activation procedure asymmetrical or contains definite spikes.
should be performed on a daily routine basis in patients in As with most amplitude asymmetry considerations in clini-
epilepsy monitoring units. cal EEG interpretations, there are no precise values available for
Chapter 12 ■ Activation Methods 217

normal limits in HV-induced slowing, but a 2:1 ratio is defi- to have patients open and close their eyes during the stimula-
nitely abnormal and any asymmetry greater than 1.5:1 is prob- tion. In a standardization of screening methods for photosensi-
ably abnormal. HV-induced slowing can persist for several tivity (37), the following IPS frequencies are used: 1, 2, 3, 4, 6, 8,
minutes after the command to breathe normally, as discussed 10, 12, 14, 16, 18, 20, 60, 50, 40, 30, and 25 Hz (8). The three
above, and this persistence or reappearance is only very rarely main EEG changes induced by IPS are explained below.
abnormal. With respect to the duration of photic stimulation in the
Regarding a clinical interpretative declaration stating that clinical EEG laboratory, it should be noted that the technologist
there are HV-induced spikes in the EEG, a very conservative must be alert to stop stimulation immediately on identifying a
approach needs to be employed. As in most other sections of photoparoxysmal response (PPR) since prolonged stimulation
the EEG, the admixture and interaction of different, nonepilep- can trigger a generalized tonic–clonic seizure (GTCS) in a sen-
tiform frequencies can sum to produce a waveform with an sitive patient. Also, in these patients, once a PPR has been defi-
epileptiform appearance that is not a true spike with the patho- nitely identified, further stimulation should not be attempted as
physiologic significance of that cortical event. This is especially these patients may become more sensitive as stimulation con-
true with HV-induced slowing and its superimposed faster fre- tinues or is repeated, and may launch into a seizure with little
quencies, and even more so in childhood since that age has the warning. The additional information gained from repeated
most complex mixture of frequencies and the highest ampli- stimulation is not worth the risk to the patient.
tudes of all of those components, including the HV-induced
slowing. In the pediatric age group both paroxysmal drowsy PHOTIC DRIVING RESPONSE
hypersynchrony and HV-induced bursts of slowing may con-
tain admixed components with a spike-like appearance that are, This is a physiologic response consisting of rhythmic activity
despite their superficial appearance, normal variants. This dif- elicited over the posterior regions of the head by IPS frequen-
ferentiation constitutes the major challenge in the interpreta- cies of about 5 to 30 Hz. The term photic driving response (PDR)
tion of pediatric EEGs. should be limited to activity time-locked to the stimulus and of
In addition, the presence of altered responsiveness in associ- a frequency identical or harmonically related to the stimulus
ation with high-amplitude rhythmic slowing (HIHARS) with frequency (38).
HV is not abnormal (31). In the latter study, 8 of 12 healthy As a rule, PDR is found over posterior regions. In infants,
nonepileptic children (mean age 9.6 years) exhibited impaired PDR can be elicited a few hours after birth (39), but PDR
verbal recall and failed to respond to repeated auditory clicks. remains relatively small up to about 6 years of age (40). In older
Furthermore, Lum et al. (32) studied 77 episodes of HIHARS children, PDR becomes much larger, particularly at low fre-
with loss of awareness from 22 children and 107 absence quencies (23). The amplitude of PDR is usually higher in chil-
seizures during HV from another 22 children; eye opening and dren than in adults and again tends to increase in elderly
eyelid flutter were seen more frequently in absence seizures, people. Regardless of age, however, an exaggerated PDR to low
whereas fidgeting, smiling, and yawning occurred more fre- flash frequencies (0.5 to 3 Hz) usually signifies acute or suba-
quently during HIHARS episodes, and arrest of activity, staring, cute neuronal dysfunction (41); examples include MELAS
and oral and manual automatisms were observed in both (mitochondrial myopathy, encephalopathy, lactic acidosis, and
groups, so that the EEG is the only reliable means of differenti- stroke-like episodes) (8) and the late infantile form of ceroid
ating the two conditions, that is, epileptic versus nonepileptic. lipofuscinosis (42,43).
According to Fisch and So (8), large positive occipital sharp
INTERMITTENT PHOTIC STIMULATION transients of sleep (POSTS) and lambda waves in response to
scanning a complex pattern are predictive of a prominent PDR.
The effect of intermittent photic stimulation (IPS) on the Destructive cortical lesions may cause unilateral PDR depres-
human EEG was first studied by Adrian and Matthews (33). sion, whereas irritative lesions, such as those associated with
They used sinusoidal IPS derived from a constant light source epileptic scars, may produce increased PDR on the side of the
in front of which a disc with cutout sectors was rotated. After lesion (19). Interpretation suggesting abnormal PDR should be
this earliest report, similar instruments were used for more than made carefully because a minor and inconsistent asymmetry of
a decade (34). Walter et al. (35) were the first to report activa- PDR is infrequently seen even in normal subjects; Coull and
tion of paroxysmal discharges by IPS with an electronic strobe Pedley (44) claim that an asymmetry in amplitude only, in the
light. After this pioneering work, the method of IPS using a absence of other EEG changes, should not be viewed as abnor-
strobe light became popular (36). The strobe lamp is placed at mal. Very low voltage or absence of PDR is of little diagnostic
a distance of 20 to 30 cm in front of the subject’s eyes. IPS is significance because some normal persons are not responsive to
commonly presented through closed eyelids, but it can be IPS (41). According to Chiba et al. (45), diffuse PDR could be
administered with the eyes open. Details of IPS use in routine observed in 25% of the patients who were receiving hemodial-
examination vary greatly between EEG laboratories. The fol- ysis treatment.
lowing protocol is suggested by Bickford (19): flashes at fre- It has been reported that PDR elicited by 5-Hz IPS with eyes
quencies of 1, 3, 6, 9, 10, 15, 20, and 30 Hz each are given in closed is similar to that elicited by a red (saturated long-
trains of 5-second duration with eyes open and closed in a wavelength red) 5-Hz flicker stimulation, whereas PDR elicited
room with reduced illumination. It is sometimes advantageous by 5-Hz IPS with eyes open is similar to that elicited by a
218 Part II ■ Normal EEG

5-Hz flickering dot pattern (46). In that case, decreased lumi- changes in response to 5-Hz flicker stimuli in a young patient
nance, such as 20 candela (cd)/m 2, was used. (This luminosity with occipital lobe epilepsy: stimuli using white and red light
is very low compared to that of a stroboscopic IPS, for example, elicited 5- and 10-Hz PDRs, whereas those by dot pattern
Grass photostimulator (intensity 8): 3939 cd/m 2 (47).) (6 mm in diameter; 0.5 cycles/degree [c/deg]) elicited 5-Hz
Furthermore, using a 5-Hz red flicker stimulation and, to a high-amplitude PDRs. Stimuli using dot patterns (0.5 mm:
greater extent, a 5-Hz flickering dot pattern provided by a visual 4.9 c/deg; 1 mm: 2.1 c/deg; 2 mm: 1.5 c/deg; 4 mm: 0.8 c/deg)
stimulator (48), high-amplitude PDR over 50 V was elicited also showed a similar finding as that shown in Figure 12.2. As
more frequently. Such an excitatory effect of a dot pattern or for high-amplitude PDRs observed in elderly individuals, they
other similar patterns on PDR (49) can be demonstrated easily tend to be elicited by stimuli using moderate (2 mm in diame-
by placing the pattern on the strobe light when IPS is given to ter) to large dot patterns (4 and 6 mm in diameter) as demon-
the subject with eyes open (50). strated in Figure 12.3. Namely, when dot patterns with lower
Studies of PDR utilizing 5-Hz red flicker and 5-Hz flickering spatial frequencies are employed, high-amplitude PDR tends to
dot pattern (6 mm in diameter) stimuli by means of visual be elicited in elderly patients (53), whereas its incidence
stimulators (47,51) yielded the following results (50). High- increases in younger patients by use of dot patterns with higher
amplitude PDR is more frequently elicited using either a red spatial frequencies.
flicker stimulation or a flickering dot pattern stimulation, irre- Using pictures such as a Lily, the 5-Hz flickering picture
spective of diagnostic group, than a white flicker stimulation stimulation also elicited a high-amplitude PDR similar to that
with equal luminance, such as 20 cd/m 2. High-amplitude PDRs evoked by a flickering dot pattern (54). These PDRs elicited by
elicited by flickering dot pattern and red flicker stimuli in adult visual stimuli not only to the full field, but also to the center,
patients suggest the presence of an occipital lobe disturbance. periphery, and each hemifield (regional visual stimulation)
Incidence of high-amplitude PDR elicited by a flickering dot provide us with useful clinical information (55). Figure 12.4
pattern stimulation in adult patients is 2.4 times higher in shows results of 5-Hz fundamental PDRs elicited by regional
female patients than in male patients. A study of PDR in adult visual stimulation in a total of 54 adult psychiatric outpatients,
psychiatric outpatients showed that PDR amplitudes increased from whom patients with epilepsy and cerebral vascular disor-
significantly with patient age (52). Figure 12.2 depicts EEG ders were excluded. We made a power spectral analysis on

Figure 12.2 EEG changes in response to 5-Hz white flicker, red flicker, and flickering dot pattern (6 mm in diameter) stim-
uli to the full field (30 30 ) in a 20-year-old woman with occipital lobe epilepsy. Stimulus was provided by use of a
square-type strobe-filter method (92); luminance was maintained at 20 cd/ m 2. Lower tracings show mapping analysis of
photic driving responses (PDRs) elicited by each flicker stimulation.
Chapter 12 ■ Activation Methods 219

Figure 12.3 EEG changes in response to 5-Hz flickering dot pattern (0.5, 2, and 6 mm in diameter) stimuli to the full-
field (30 30 ) in a 70-year-old woman with benign paroxysmal positional vertigo. Stimulus was provided by use of a
square-type strobe-filter method (92); luminance was maintained by 20 cd/ m 2. Lower tracings show mapping analysis of
photic driving responses (PDRs) elicited by each flickering dot pattern.

occipital PDRs elicited by a 5-Hz flickering dot pattern (6 mm


in diameter: 0.5 c/deg) stimulation to the center (11 11 ),
periphery (11 11 to 57 57 ), full field (57 57 ), and
each hemifield with luminance maintained at 10 cd/m 2. The
average PDR amplitude in response to regional flickering dot-
pattern stimuli in 28 patients aged over 45 years was higher
than the average PDR amplitude in 26 patients aged 20 to 44
years. Amplitudes of PDRs elicited by stimuli to the periphery
and lower hemifields were similar to those elicited by a stimu-
lus to the full field. Regarding PDRs elicited by lateral hemifield
stimuli, contralateral PDRs were slightly higher than ipsilateral
ones, but their difference was not statistically significant.
Particular efficiency in eliciting higher PDR by lower hemifield
stimulation compared with other hemifield stimuli might be a
result of its selective stimulus to the visual cortex, which is
located closely beneath electrodes 01 and 02.
Figure 12.4 Photic driving response (PDR) elicited by regional visual A power spectral analysis of PDRs elicited by the above
stimuli in 54 adult psychiatric outpatients. We used an SLS-5100 visual visual stimuli may be used as a new tool for evaluation of drugs
stimulator (Nihon Kohden). Power spectral analysis was made on the influencing the brain (53). Other clinical usefulness of this
record for 5 seconds after each 2-second stimulation. Power spectra are EEG examination is reported in the following discussion. In a
5-, 10-, 15-, 20-, and 25-Hz PDRs; only that of 5-Hz fundamental PDR, patient with amblyopia, a distinct difference of PDRs evoked
which was the greatest, is shown in this figure. The root of each power by stimulation of 5-Hz flickering dot pattern to each eye was
is expressed for convenience to indicate amplitude. (Modified from obtained (56); PDRs evoked by hemifield 5-Hz flickering dot-
Takahashi T. EEG activation by visual stimuli. In: Sato M, Matsuoka H, pattern stimuli enabled EEG diagnosis of a parietal lobe
eds. The Newest Clinical Electroencephalography. Tokyo: Asakura metastatic brain tumor in a patient who had suffered from
Shoten; 1993:323–344, with permission.) breast cancer (57).
220 Part II ■ Normal EEG

PHOTOMYOCLONIC RESPONSE closure, especially immediately after eye closure (75). Patients
(PHOTOMYOGENIC RESPONSE) with PPR are less sensitive to IPS when asleep (76,77).
Generalized PPR may be most pronounced in the frontal, cen-
This is a response to IPS characterized by appearance of brief tral, or occipital regions. Hishikawa et al. (76) classified general-
repetitive muscle spikes over anterior regions of the head. These ized PPRs in photosensitive epilepsy patients into PPRs that
often increase gradually in amplitude as stimulation continues appear first in the occipital area and PPRs that occur simultane-
and cease promptly when the stimulus is withdrawn. The ously over all areas or appear earlier over anterior regions. In the
response is associated frequently with eyelid flutter, a vertical former group, an augmented visual evoked response could be
oscillation of the eyelids and eyeballs; sometimes it is associated obtained. Generalized PPRs in children are usually rhythmic
with discrete jerking, mostly involving musculature of the face and higher in amplitude than those seen in adults. Occipital
and head (38). spikes as a sole response to IPS may not be indicative of epilepsy
Principal features of this response were first described by (78). Unilateral occipital spikes are rarely induced by IPS. These
Gastaut and Rémond (58) and Bickford et al. (59), who intro- patients, as opposed to patients with generalized PPR, often have
duced the term photomyoclonic response (PMR), which differs a history of a local posterior lesion, mostly traumatic. Very
from PPR. Occasionally, a PMR can be seen with a PPR. The rarely, frontal spikes can be provoked by IPS (79).
most effective triggering IPS frequency lies between 12 and 18 Jeavons (80) found a PPR in 2.8% of the patients referred for
Hz (60). The PMR tends to appear in conjunction with muscu- EEG examination, a figure similar to that of Gastaut et al. (61).
lar tension. It occurs less often in children than in adults. The patients, whose seizures are induced by visual stimuli such
Gastaut et al. (61) reported that PMR was found in 0.3% of as viewing a visual pattern, television (81), or eye closure, are
normal subjects, 3% of patients with epilepsy, 13% of patients particularly sensitive to IPS and tend to demonstrate PPR.
with brainstem lesions, and 17% of patients with psychiatric Gastaut et al. reported that the PPR is almost entirely confined
disorders. The incidence of PMR in other studies (62–65), how- to patients with primary subcortical epilepsy and that it occurs
ever, was lower than in that of Gastaut et al., ranging from 0.1% in 40% of cases with absence seizures and in 20% of cases with
to 0.8%. The PMR is considered to be a nonspecific finding that GTCSs. According to Stevens (82), PPR occurred in 53% of that
is not significant for elimination diagnosis of a seizure disorder study’s patients with nonfocal seizures, but in only 3% of that
(66). It is enhanced in early stages of alcohol withdrawal in study’s patients with focal seizures. A similar view has also been
chronic alcoholics (67) or after sudden withdrawal from barbi- expressed by Niedermeyer (11), who states that generalized
turates and related sedatives (68). A symptomatic transient PPRs are more highly associated with primary generalized
PMR secondary to a metabolic disorder (severe hypocalcemia) epilepsy than with incompletely generalized or focal spikes.
can occur (69). However, Guerrini et al. (83) proposed a concept called idio-
pathic photosensitive occipital lobe epilepsy based on detailed
PHOTOPAROXYSMAL RESPONSE analysis of 10 patients with recurrent episodes of visually
(PHOTOCONVULSIVE RESPONSE) induced occipital seizures; all seizures were stimulus (mostly
television and computer screen) related and began with ele-
This is a response to IPS characterized by spike-and-wave and mentary visual symptoms, followed in most patients by a slow
multiple spike-and-wave complexes that are bilaterally syn- clustering of cephalic pain, epigastric discomfort, and vomiting,
chronous, symmetrical, and generalized and that may outlast with either normal or only mildly impaired responsiveness: all
the stimulus by a few seconds. There may be associated impair- of them showed PPR of types 1 to 4 (see below). The ictal events
ment of consciousness and brisk jerks involving musculature of that may accompany PPR are predominantly absence seizures,
the whole body, most predominantly that of the upper extrem- GTCSs, and myoclonic jerks, especially of the eyelids or arms
ities and head (38). Newmark and Penry (66) state that gener- (84). Gambardella et al. (85) reported a 17-year-old girl with
alized slow activity and posterior spikes are not accepted pure photosensitive epilepsy who showed photic-induced
universally as a PPR, but the significance of these discharges epileptic negative myoclonus (PPR was accompanied by loss of
increases if they continue after stimulation is discontinued. postural tone in both arms). They claim that negative
Although PPRs in photosensitive epilepsy patients are elicited myoclonus should be included among the ictal phenomena
by a broad range of IPS frequencies from 1 to 65 Hz (70), the accompanying PPR.
most epileptogenic frequencies are within the range of 15 to 18 Some studies have suggested that presence of PPR may be a
Hz (71). PPR is most frequently induced by 15-Hz IPS with eyes familial trait (73,86,87). Doose et al. (86) reported that PPR
closed and 20-Hz IPS when the eyes are open (72). When IPS is may be regarded as a symptom of susceptibility to convulsions
given with eyes open, careful attention to eye movement and of the centrencephalic type. In a genetic study of photosensi-
position is needed because a directional change from central to tive epilepsy, Waltz et al. (88) classified PPR into four types:
lateral gaze diminishes the effect of IPS in evoking PPR. This type 1, spikes with occipital rhythm; type 2, parieto-occipital
effect is greater than the diminution effect with monocular IPS spikes with a biphasic slow wave; type 3, parieto-occipital
stimulation as compared with binocular IPS stimulation in elic- spikes with a biphasic slow wave and spread to the frontal
iting PPR (73). Regarding levels of ambient light, with high- region; type 4, generalized spikes and waves or polyspikes and
intensity IPS, the effects of normal ambient lighting may be waves. They found that type 4 occurred more often in both
negligible (74). During IPS, the PPR may be induced by eye probands with epilepsy and their siblings than the respective
Chapter 12 ■ Activation Methods 221

controls, suggesting that coincidence of photosensitivity will during 6-month periods identified 118 patients who had a first
appear as higher if only type 4 is considered to be indicative seizure while playing ESGs when they were 7 to 19 years old. Of
of photosensitivity. Photosensitivity is age dependent, being 118 patients, 46 and 25 had definite and probable causal rela-
highest in late childhood and early adolescence; it is more tionships, respectively; 47 patients had no such relationship.
common in females (86). According to a prospective nation- Within that age group, the annual incidence of first seizure trig-
wide study made in Great Britain (89), the annual incidence of gered by playing ESGs (71 patients altogether) was estimated to
cases of epilepsy with type 4 PPR on their first EEG was be 1.5/100,000, representing approximately 3% of all new
approximately 2% of all new cases of epilepsy. When restricted patients with epilepsy in this age range. Of 71 patients, 46
to the age range of 17 to 19 years, the annual incidence rose to showed type 4 PPR, whereas 25 showed types 1 to 3 PPR.
approximately 10% of all new cases of epilepsy presented in Although photosensitivity is thought to play the most important
this age range. Analyzing generalized PPRs found in role in engendering ESG-induced seizures, other circumstances,
128 chronic epilepsy patients, De Graaf et al. (90) reported that acting either singly or in combination, should be taken into con-
there was a significantly higher occurrence in whites (2.7%, 72 sideration (95). Examples include (i) seizure precipitation by
of 2657) as compared with blacks (0.1%, 1 of 848) and subjects specific cognitive activities, decision making, hand movements,
of mixed race (0.9%, 55 of 5958), concluding that genetic etc.; (ii) seizure precipitation by nonspecific emotional factors
rather than environmental factors influence generalized PPR. relating to the subjects’ engagement in games, such as anxiety or
For epileptic seizures triggered by television, photosensitivity excitement; (iii) lowering of the seizure threshold by fatigue or
remains the most common single mechanism (47). Since the sleep deprivation; and (iv) chance occurrence of a spontaneous
first description of space invader epilepsy by Rushton (91), seizure in a person with epilepsy while playing ESGs. Figure 12.5
patients with seizures triggered by electronic screen games illustrates generalized paroxysmal discharges elicited by a video
(ESGs) such as video, console, and computer games have been game and generalized PPRs elicited by flickering visual stimuli
reported (92,93). A nationwide study made in Great Britain (94) in a 13-year-old boy who had an ESG-induced GTCSs.

Figure 12.5 Generalized paroxysmal discharges elicited by a video game (Super Tetris 2 Bombliss) and generalized pho-
toparoxysmal response (PPRs) elicited by flickering stimuli in a 13-year-old photosensitive epilepsy patient. We used a
12-in. television placed 1 m in front of the patient. Full-field (30 30 ) stimuli of a 15-Hz red flicker and a 25-Hz flicker-
ing 1.5 c/ deg dot pattern provided by use of square-type strobe-filter method elicited type 4 PPR. Luminance maintained
at 20 cd/ m 2. This EEG was recorded under drug-free conditions.
222 Part II ■ Normal EEG

Occurrence of a PPR in an otherwise seizure-free individual


is uncommon; these patients tend to have attacks of headache
or psychological problems, such as personality disorder with
hysterical reactions or an anxiety state (60). Incidence of PPR in
nonepileptic, apparently normal subjects is estimated at proba-
bly less than 2% (66).
Reilly and Peters (64) claim that the following criteria are
useful for determining whether or not the patients are epileptic:
prolonged PPR (outlasting the stimulus) suggests probable Figure 12.6 A flash lamp and circular-type strobe filter. Luminance
epilepsy (90%), whereas self-limited discharge is not diagnostic. of direct stroboscopic intermittent photic stimulation (IPS) by use
Jayakar and Chiappa (96) studied 35 patients with PPR (1% of of LS-706A (Nihon Kohden), as shown in the left tra cing, is
all patients seen in an EEG laboratory in a general hospital) and 3,939 20 cd/ m 2. Inserting a vertical grating pattern (middle tracing)
found that 77% had a definite history of epilepsy, 9% a ques- along the filter holder on the front of the flash lamp as shown in the left
tionable history, and 14% had had no seizures. PPRs were pro- tracing greatly reduces luminosity to 30 cd/ m 2 and spatial frequency of
longed more than 100 msec beyond the stimulus in 11 patients each pattern filter is 2 c/ deg. Using this filter, the weakest possible stim-
and self-limited in 24, and the incidence of seizures was not dif- uli of red flicker and pattern flicker ought to be used, taking great care
ferent in the two groups. Furthermore, the PPR prolongation not to induce clinical seizures in photosensitive individuals; one might,
was related to the time at which the stimulus was stopped after for example, start stimuli from lower flicker frequencies and terminate
the appearance of the PPR—shorter durations of stimulation examinations when PPR appear (92).
tended to have longer prolongation.
With respect to the duration of photic stimulation in the clin-
ical EEG laboratory, it should be noted that the technologist must ric pattern) has been found to be more effective than a homoge-
be alert to stop stimulation immediately on identifying a PPR neous field (or white light flicker) in eliciting PPR (75,104,105).
since prolonged stimulation can trigger a GTCS in a sensitive In addition, saturated long-wavelength red (red) facilitates
patient. Also, in these patients, once a PPR has been definitely induction of PPR (106,107). An EEG activation by use of red
identified, further stimulation should not be attempted as these flicker and flickering geometric pattern stimuli, which were
patients may become more sensitive as stimulation continues or given only in “eyes-open” conditions, revealed a higher rate of
is repeated, and may launch into a seizure with little warning. PPR provocation than by ordinary stroboscopic IPS (51). (In the
Especially in adults without previous seizures and without a former method, an SLS-5100 visual stimulator (50) was
family history of seizures, PPR usually suggests a toxic, meta- employed and luminance maintained at 20 cd/m 2.) Additional
bolic, or drug withdrawal state (41,68,97,98). Symmetrical pos- studies (108,109) have disclosed that elicited PPRs were found in
terior high-amplitude spikes can be evoked at slow IPS rates in 18% of epilepsy patients, suggesting that incidence of photosen-
patients with diffuse encephalopathies, such as progressive sitivity in epilepsy patients is 3.6 times higher than that of the
myoclonic epilepsy or Creutzfeldt–Jakob disease (44,99,100). commonly accepted 5% reported by Binnie and Jeavons (110).
Spontaneous epileptiform abnormalities (SEAs) are Figure 12.6 shows a circular-type strobe filter (93,111), which is
reported to occur in up to 65% of patients with a PPR. Gilliam a simple device that is capable of inducing similar activation
and Chiappa (101) examined seizure classifications and SEAs in effects to the SLS-5100 visual stimulator. Using strobe filters,
115 consecutive patients who had a PPR. A PPR was the only visual stimuli (red flicker and flickering geometric pattern, lumi-
epileptiform abnormality in 47 patients (41%), 27 (24%) had nance kept at 30 cd/m 2) are easily presented to patients. Their
focal SEAs, and 41 (36%) had only generalized SEAs. Seventeen stimulus luminance is far less intense than that of a stroboscopic
patients (15%) had partial seizures and 40 (35%) had only gen- IPS. In addition to examining photosensitivity with an ordinary
eralized seizures, and seizure classification was strongly associ- strobe, such low-luminance visual stimuli may be required for
ated with the type of SEA (P 0.0001). Although the PPR is precise examination of photosensitive epilepsy patients.
often presumed to signify primary generalized epilepsy, most Chiappa et al. (112) and Hill et al. (113) used functional
patients with a PPR and focal SEAs have partial seizures. This is magnetic resonance imaging (fMRI) and magnetic resonance
perhaps an indication that the presence of an underlying gener- spectroscopy (MRS) to study metabolic and hemodynamic
alized hyperexcitability, possibly related to a channelopathy (for consequences of photic stimulation–triggered and spontaneous
example), enhances the likelihood that a focal lesion will gener- generalized epileptiform discharges in humans. EEG recordings
ate an electrophysiologic abnormality. were performed in the MR scanner during photic stimulation
Several conditions are known to alter PPR. As described in normal subjects and patients with PPR. There were no fMRI-
above, PPR is often associated with eye closure and is most likely registered hemodynamic abnormalities found in relation to the
to occur immediately after eye closure. This effect may also brief PPR seen in the patients. However, irrespective of the
result from movement of the eyelids or of the eyes themselves. In presence of a PPR, these patients showed four unique findings
addition, eye closure may produce activation by suddenly elim- compared with normal subjects: (i) slightly but significantly
inating the visual pattern and by causing the field of vision to increased lactate levels in the occipital cortex in the resting
become red-hued (102,103). When stimuli are presented with state, (ii) an increased area of visual cortex activation with
the eyes open, a patterned field of vision (or flickering geomet- photic stimulation, (iii) noncontiguous areas of signal attenua-
Chapter 12 ■ Activation Methods 223

tion, simultaneous with the occipital cortex stimulus-induced


increased fMRI signal, most prominent in perirolandic regions,
and (iv) a marked decrement (undershoot) of fMRI signal
intensity immediately after the photic stimulation in the occip-
ital cortex and in the region of the posterior cingulate gyrus.
These findings suggest abnormal interictal metabolism and
increased vascular reactivity in the photosensitive patients.
Along with PPR, another term in use is photoepileptiform
response (PER). This response is elicited by photic stimulation.
According to their topography, they are divided into the follow-
ing three categories (8): (i) anterior dominant or generalized
(bisynchronous and approximately symmetrical); (ii) occipital
dominant (bisynchronous and approximately symmetrical);
and (iii) occipital dominant and localized (unilateral or Figure 12.7 Geometric patterns used for square-type strobe-filter
strongly lateralized). The category 1 PPR is one form of PER method. Each pattern was presented 30 cm before the eyes, resulting in
and is most closely associated with epilepsy. its spatial frequency at 2 c/ deg; pattern contrast is 0.98.

OTHER FORMS OF SENSORY STIMULATION that are arranged geometrically (72). According to Wilkins et al.
(133), to induce paroxysmal discharges, patterns (black and
Visual Stimulation
white stripes of equal width and spacing) must have a spatial
Various visual stimuli other than IPS may provoke PERs. The sim- frequency between 1 and 4 c/deg. Line patterns are more epilep-
ple maneuver of eye closure induces posterior slow-wave tran- togenic than dot patterns (Fig. 12.7). Oscillation of a grating
sients, especially in children (114,115), and may also provoke pattern greatly enhanced provocation of paroxysmal dis-
paroxysmal discharges (116,117). Paroxysmal discharges similar charges; gratings oscillating in a direction orthogonal to the
to PPR seen in photosensitive epilepsy patients may even be lines were the most effective, and the optimum oscillation fre-
induced by slow closure of the eyelids (118). In the act of eye clo- quency was in the range of 15 to 20 Hz (134).
sure carried out in a lighted room, lights off and homogeneous Figure 12.8 is an example of 3-Hz spike-and-wave complexes
visual field (ganzfeld) (119–121) are considered to play significant associated with an absence seizure induced by viewing a grating
roles as visual stimuli in producing paroxysmal discharges. In fact, pattern in a 3.4-year-old girl with severe myoclonic epilepsy in
posterior sharp waves or spikes can be seen occasionally after or at infancy (135). This patient was very sensitive to vertical, hori-
the onset of IPS; these are called off-response and on-response, zontal, and oblique stripes with a wide range of spatial frequen-
respectively (73,122). Spike–wave discharges associated with cies. She was also sensitive to dot patterns, but slightly less so
seizures can rarely be induced by a single-flash light stimulus than to stripes. Interestingly, this girl repeatedly stared at those
(102). Paroxysmal discharges may be evoked by red light alone patterns as if she were enjoying the absence seizures. A dot pat-
(93,119,123). Darkness may also induce paroxysmal discharges tern, however, more frequently evokes lambda waves and rhyth-
(124,125). Similar paroxysmal discharges produced by ganzfeld mic posterior slow activity than a grate pattern (136). Lambda
and total darkness have been reported by Panayiotopoulos waves are more frequently evoked in subjects who show good
(126,127), who called it fixation-off sensitivity (FOS); this unpat- PDRs, especially to low-frequency IPS. Radhakrishnan et al.
terned vision can be obtained either through 10 spherical lenses (137) reported pattern-induced negative occipital potentials
or through underwater goggles covered with semitransparent (PINOP), which are similar to lambda waves but differ in
paper. He claims that FOS is suspected if EEG abnormalities response characteristics with surface negativity. Photosensitive
appear and persist as long as the eyes remain closed and disappear epilepsy patients are also often sensitive to pattern viewing,
when the eyes are opened. In such cases, EEG abnormalities of with an incidence of 5% (102), 22% (105), or 72% (138).
FOS can be elicited by elimination of central vision and fixation; Paroxysmal discharges induced by viewing usually arise from
FOS is related to eyes-closed, not eye-closure EEG abnormalities. the occipital area, suggesting that they may arise primarily in an
According to Panayiotopoulos (126,127), the incidence of FOS- epileptogenic visual cortex (103,130,133). Geometric pattern
related EEG abnormalities appears to be as frequent as that of PPR stimulus occasionally induces fast activity (beta rhythm) (53).
in children younger than 12 years of age; FOS is probably more Pattern viewing may also enhance mu rhythm (139).
frequently associated with benign childhood epilepsy with occip- It has been reported that photosensitive epilepsy patients are
ital paroxysms than with any other epileptic condition. particularly sensitive to full-field visual stimuli of a red flicker
and flickering geometric pattern, when a decreased luminance,
Pattern, Pattern Flicker, and Red Flicker Stimulation such as 10 to 20 cd/m 2, is used as a stimulating light source (108).
Geometric patterns often provoke paroxysmal discharges in Figure 12.9 illustrates EEG changes in response to stroboscopic
epilepsy patients, particularly in photosensitive ones (128–132). IPS, red flicker, and flickering dot pattern stimuli in a photosen-
The patterns that are most effective for activation consist of sitive epilepsy patient. Particular emphasis ought to be given to
closely spaced lines or dots with sharply contrasting interfaces the finding that IPS to eyes open was ineffective, whereas red
224 Part II ■ Normal EEG

Figure 12.8 Generalized 3-Hz spike-and-


wave complex elicited by a horizontal
2.1 c/ deg grating pattern stimulation in a
3.4-year-old girl with severe myoclonic
epilepsy in infancy. Vertical and horizontal
eye movements, electromyogram (EMG) of
the left hand, and electrocardiogram (ECG)
were recorded simultaneously. Downward
and upward arrows indicate the beginning
and the end of a grating pattern stimula-
tion, respectively. In accordance with
occurrence of a generalized paroxysmal
discharge, regular button pressing shown
at the top became absent, suggesting that
she has an absence seizure. (Modified from
Takahashi T, Sasaki M. Pattern sensitivity:
its activation method and clinical EEG find-
ings. Rinsho Nohha (Osaka). 1971;13:
727–732, with permission.)

Figure 12.9 EEG changes in response to stroboscopic IPS, red flicker, and flickering dot pattern stimuli in a 14-year-old
girl with photosensitive epilepsy. By use of Grass PS33-plus photic stimulator (intensity at 8), 15-Hz IPS was given 30 cm
above the eye. Using the strobe-filter method, 15-Hz red flicker and 15-Hz flickering 2 c/ deg dot pattern stimuli to the full
field (25 25 ) were given from 30 cm before the eye; luminance maintained at 20 cd/ m 2.
Chapter 12 ■ Activation Methods 225

however, was less effective. Lateral hemifield stimuli elicited gen-


eralized PPR (equal PPR), but upper and lower hemifield ones
were ineffective in this patient. In terms of PPR etiology, equal
PPRs as shown in Figure 12.11 are thought to be related to
genetic factors, whereas an unequal PPR, in which either left or
right hemifield stimulation elicits a PPR, is indicative of acquired
cerebral disturbance, such as head trauma (109).
Regarding provocation of generalized PPR by visual stimuli
to the center and periphery, a red flicker stimulation to the cen-
ter is more effective than that to the periphery, indicating that
Figure 12.10 Dot pattern filters used for regional flickering dot pattern macular stimulation plays a central role in it. In eliciting PPR by
stimuli by use of square-type strobe-filter method. Diameter of a single flickering dot pattern stimulation to the full field, a dot pattern
dot is 4 mm (0.8 c/ deg), and luminance maintained at 20 cd/ m 2. (See with higher spatial frequency, such as 2.1 to 1.5 c/deg, shows the
color insert.) most significant efficacy; flicker frequencies must be between
20 and 15 Hz in that condition (146). Flickering dot pattern
stimulation to the center too, when a higher frequency dot pat-
flicker and flickering dot pattern stimuli elicited generalized tern (2.1 to 4.9 c/deg) is used, elicits generalized PPR similar to
PPRs, suggesting that stroboscopic IPS to eyes open, with lumi- that induced by red flicker stimulation to the center (147),
nance of 3939 cd/m 2, is too strong to elicit PPR in this patient whereas that to the periphery by use of 0.5 c/deg dot pattern
(140). In addition to hemifield (109,141–144) and quadrant-field elicits generalized PPR with apparent local onset from posterior
(145) stimuli, visual stimulation to the center and periphery by regions (109). Therefore, photosensitive epilepsy patients who
use of the above stimuli can also yield useful information to fur- are sensitive to red flicker and flickering dot pattern stimuli are
ther elucidate PPR (109). As an example, Figure 12.10 shows fil- thought to have hyperexcitable areas not only in the striate
ters used to provide a flickering dot pattern stimulation to the cortex but also in the parastriate cortices. Local PPR in the
center, periphery, and hemifield by the square-type strobe-filter striate cortex elicited by red flicker stimulation would immedi-
method (53,93); similar field-size filters are used in red flicker ately transmit to the nonspecific diffuse projection system.
stimuli. Figure 12.11 demonstrates EEG changes in response to This could engender generalized PPR with their simultaneous
such regional visual stimuli in the same patient shown in Figure occurrence over all areas, as distinguished from those elicited by
12.9. A red flicker stimulation to the center promptly elicited gen- flickering dot pattern stimulation. In the latter, initial local PPR
eralized PPR as shown in the left tracing. That to the periphery, would seem to originate in the striate cortex as well as in the

Figure 12.11 Generalized PPRs elicited


by regional red flicker stimuli in a 14-year-
old girl with photosensitive epilepsy (the
same patient shown in Fig. 12.9).
226 Part II ■ Normal EEG

parastriate cortices. These might produce different generalized cial care in interpretation is needed because arousal secondary
PPR, preceded by spikes over the posterior regions. to auditory stimuli, not auditory stimulus per se, may play a
Of 18 photosensitive patients sensitive to red flicker stimula- role in triggering paroxysmal discharges.
tion (140), all cases were sensitive to at least one of the flicker- Hearing-induced seizures are precipitated by hearing more
ing geometric pattern stimuli. Although this finding suggests or less specific music (159), voices, and other sounds. According
that common neuronal pathways are involved in PPR induction to Rosenow and Lüders (160), those patients usually have auto-
by red flicker and flickering geometric pattern stimuli, two dif- motor seizures or GTCSs and temporal lobe epilepsy; the
ferent visual cortical pathways, namely the parvocellular (P) pathogenesis of auditory-induced seizures most probably
and the magnocellular (M) systems, are presumed to severally involves activation of temporal auditory areas.
play a part in the following (93,148): the P system deals with
color, and V4 plays a central role for perception of both shape Somatosensory Stimulation
and color (149). In human subjects, the region of the lingual When areas of the primary sensorimotor cortex become
and fusiform gyri has been identified as the color center hyperexcitable as a result of disease, contralateral afferent stim-
(150,151). Regarding reversing patterns, Harding and Jeavons uli may trigger focal spikes and partial seizures. Along with tac-
(47) maintain that they are dealing with transient phenomena tile stimulation (161–164), local stimuli by cold wind, rubbing,
and that perhaps the M system provides critical transmission vibration, and stretching a muscle appear to rarely provoke
that is similar to the mechanism of pattern sensitivity. This spikes and seizures as reviewed by Takahashi (53). Tapping
understanding may also hold true for flickering geometric pat- anywhere about the upper torso or head produces myoclonic
tern stimulation (53). From findings obtained by regional movements associated with diffuse spike–wave discharges in
visual stimulation technique, however, the following view some young idiopathic epilepsy patients who are sensitive to
appears to be most likely: generalized PPRs elicited by red afferent stimuli (41). Focal areas that are sensitive to these
flicker and high spatial frequency pattern flicker stimuli to the stimuli are the finger, arm, shoulder, chest, face, head, and feet.
center are mediated by the P system, whereas those elicited by In one patient, partial elementary seizures were evoked by
low spatial frequency pattern flicker stimulation to the periph- tooth brushing (165). O’Brien et al. (166) reported a 23-year-
ery are mediated by the M system (93). old female complex epilepsy patient with seizures precipitated
The above knowledge obtained from EEG diagnosis by low- exclusively by tooth brushing and in whom structural and
luminance visual stimuli provides us important clues for functional imaging demonstrated a right posterior frontal
detailed understanding particularly of seizures induced by focus. Using tactile stimulation comprising tapping of the pal-
stimulative screen images observed through use of TV, video mar tips of the fingers or toes, Langill and Wong (167) investi-
systems, computer, and so on (93). In a study that dealt with (i) gated 304 patients with rolandic discharges. In this stimulus
30 photosensitive epilepsy patients and (ii) 78 epilepsy patients condition, right-hand tapping, for instance, would elicit dis-
with PPR (152), red flicker and pattern flicker stimuli elicited charges over the left central region, whereas tapping of either
type 4 PPR more frequently in (i) than in (ii) subjects, suggest- foot would elicit discharges at the vertex. Such tactile-
ing that both the P-ventral and M-dorsal visual pathways are in enhanced discharges constituted 38.2%; they were regarded as
a sensitive state in photosensitive epilepsy patients. In contrast, benign, age-related phenomena.
pattern flicker stimulation alone tended to induce type 4 PPR in Reviewing published cases of somatosensory reflex epilepsy,
the (ii) subjects, suggesting that only the M-dorsal visual path- Servit (168) stated that seizures are provoked by stimulation of
way is in a sensitive state in epilepsy patients with a PPR. the trigeminal somatosensory region in the majority of patients
(over 80%). Exaggerated responses in such patients can be
Auditory Stimulation obtained by electrical stimulation of peripheral nerves. The
When compared with visual stimuli, activation of paroxysmal amplitude of somatosensory evoked potentials (SEPs) to
discharges by auditory stimuli is extremely rare. Being either median nerve electrical stimulation is markedly enlarged (giant
transient or continuous, paroxysmal discharges provoked by SEPs) in most patients with cortical myoclonus of various eti-
auditory stimuli have been found mainly over temporal areas ology (169–173).
(153–157). Pure tones have been shown by Arellano et al. (153) Mima et al. (174) studied proprioception-related SEPs with
to activate paroxysmal discharges in a psychomotor epilepsy passive flexion movement of the middle finger at the proximal
patient. Prechtl (158) reported that repetitive clicks caused interphalangeal joint in seven cortical myoclonus patients who
aggravation of preexisting temporal lobe abnormalities in 37% had associated giant SEPs. In three of the seven patients, the
of a large group of mixed cases. Stevens (82) found the activat- proprioception-related SEPs were also enlarged. Therefore, the
ing mechanism in 4.5% of temporal lobe epilepsy patients. authors concluded that hyperexcitability of the sensorimotor
These findings suggest that an epileptic disturbance in the pri- cortex in cortical myoclonus is modality-specific; cortical
mary auditory area of superior temporal gyrus (Heschl’s gyrus) excitability is exaggerated to both cutaneous and deep receptor
may chiefly be responsible for auditory stimuli that give rise to inputs in some patients, but only to cutaneous input in others.
such paroxysmal discharges. However, Kiloh et al. (23) showed Figure 12.12 shows an example of unilateral central–parietal
that unilateral central sharp waves could be evoked by random spikes evoked by electrical stimulation of the contralateral
taps. When generalized paroxysmal discharges appear to be median nerve in a patient with benign childhood epilepsy with
induced by auditory stimulation in drowsiness and sleep, spe- centrotemporal spike.
Chapter 12 ■ Activation Methods 227

Figure 12.12 Electrical stimulation of right median nerve evoked


spikes, maximal over left central–parietal areas, in a 6-year-old boy with
Figure 12.13 EEG responses to odorous stimuli in a 29-year-old
benign childhood epilepsy with centrotemporal spike.
woman with epilepsy with generalized tonic–clonic seizures (GTCSs)
on awakening.
Plasmati et al. (175) found increased incidence of giant SEPs
(38%) in 44 benign epilepsy with rolandic spikes (BERS) patients.
According to their further study (176), which investigated 54 or immediately after exposure to perfumed air. We also demon-
BERS patients and 61 age-matched control subjects, short laten- strated EEG changes in response to olfactory stimuli in an adult
cies were normal in all BERS patients, whereas the amplitude of epilepsy patient (79); generalized paroxysmal discharges were
middle latency was increased in a significant percentage of chil- induced by ether and acetic acid. Figure 12.13 shows another
dren with BERS. Giant responses occurred in 25.9% of BERS example of odor sensitivity in a patient with GTCSs on awaken-
patients and in 4.9% of normal subjects. This phenomenon was ing; olfactory stimuli with acetic acid and coffee elicited gener-
presumed to depend on physiologic, functional cortical hyperex- alized paroxysmal discharges. In distinction to other modalities
citability related to maturation processes. They state that the giant of sensory stimuli, repeated olfactory stimuli tend to quickly
responses clearly differ from those observed in myoclonic epilep- become ineffective. On EEG activation, olfactory stimuli using
sies and are likely to be generated in different neuronal pools in vanilla, coffee, and so on, may be desirable because ammonia
the sensorimotor cortex. The similar distribution of these giant and similar pungent substances stimulate the trigeminal nerve.
middle-latency SEPs with tactile evoked EEG spikes suggests a Very little is known about the influence of vestibular stimu-
common generator for these responses. Electrical stimulation of lation on the EEG. Behrman and Wyke (180) reported that
the median nerve on routine EEG examination evoked bilateral 6-Hz activity arising from the temporal area could be
central–parietal spikes in 3% of patients; all of them were epilepsy evoked by caloric stimulation. Focal EEG changes were pro-
patients, the majority of whom showed PPR as well (156). In a duced by rotational stimulation in 9 of 15 epileptics tested
case reported by Goldie and Green (177), seizures elicited by rub- (181); in 4 patients, bilateral 2- to 7-Hz activity arose from the
bing the face could also be precipitated just by thinking about temporoparietal regions. Karbowski (181) performed caloric
rubbing the face or by hypnotic suggestion of cutaneous stroking. stimulations in 62 epilepsy patients (irrigation for 15 seconds
Van Cott et al. (178) reported four adult patients with stimulus- with 100 mL water at 27 C and at 44 C). In 22 cases, an increase
sensitive seizures and myoclonus following severe hypoxic- in, or the appearance of, EEG abnormalities was detected. In
ischemic injury (postanoxic intention myoclonus, also termed two further cases epileptic seizures were triggered.
Lance-Adams Syndrome); their EEGs showed bursts of general-
ized spike and polyspike activity following tactile stimulation Multisensorial and Vegetative Sensory Stimuli
associated with clinical seizures. In a comatose patient with More than 600 patients with hot water epilepsy have been
reversible viral encephalitis, the association between stimulus- reported worldwide; more than 90% of them are from southern
induced periodic epileptiform discharges, arousal EEG responses, India (183). The following factors are presumed to be involved
and limb jerking was observed (179). in its pathophysiology: (i) high temperature, (ii) direct contact
of skin with water, (iii) body part in contact with water, and (iv)
OTHER SENSORY STIMULI mode of bathing. Generalized epileptiform abnormalities were
found in 15% to 22% of patients (184). Other reports have
Regarding EEG activation by olfactory stimulation, very few described both diffuse and focal abnormalities, predominantly
data exist. Stevens (82) tested 61 epileptics with gross olfactory in the temporal lobes. Ictal EEG abnormalities in 10 patients
stimulation. The stimuli consisted of a strong perfume (Tabu) include rhythmic left temporo-occipital spikes (183,185).
and a malodorous H 2S compound. Sixteen out of 61 temporal Seizures induced by eating (eating seizures) are rare.
lobe epilepsy patients demonstrated exaggerated spiking during Rémillard et al. (186,187) proposed two main clinical syn-
228 Part II ■ Normal EEG

dromes of eating seizures, differentiating temporolimbic from ively, following the blinks by 100 to 200 msec. These blink-
extralimbic, usually suprasylvian, seizure onset on clinical, radi- triggered CMTSs appeared almost exclusively while the patient
ologic, and EEG evidence. In addition, there was a subgroup of was awake; it was inferred that the blinking while awake was
patients activated by certain tastes, such as sweets (186). They associated with spikes resulting from abnormal and excessive
argued that the occurrence of eating seizures may be related to excitatory interconnections between the precentral frontal areas
the excitation of a critical mass of epileptogenic cortex by vari- and the epileptogenic zone of the rolandic cortex. In one patient,
ous afferent stimuli, citing roles of the following: taste, mastica- seizures were triggered by blinking when beginning to speak
tion, gastric distension, somatosensory or proprioceptive input, (202). The phi rhythm (referring to posterior rhythmic slow
and subcortical structures (187). waves occurring after eye closure) is an unusual age-related phe-
Seizures of “abdominal epilepsy” have reportedly been nomenon seen mostly in young patients. According to Silbert
induced by vegetative sensory stimuli, such as enemas and et al. (203), who defined it as a minimum of three consecutive
injection of Prostigmin (116); such EEGs are characterized by monomorphic posterior delta waves occurring within 2 seconds
appearance of generalized spike–wave complexes, occasionally of eye closure on at least two occasions, the phi rhythm has no
associated with temporal and hemispheric foci. significance for diagnosis of structural cerebral lesions or
epilepsy. In epilepsy patients with this rhythm, however, the
Unexpected Startling Stimuli seizure disorder is more likely to be generalized than partial. In
One may rarely encounter patients with startle epilepsy epilepsy patients, spike–wave discharges can also be induced not
(163,164,188). Startle-induced seizures are focal or generalized only by closing eyes (116,117,199,204–206), but also by opening
tonic or atonic seizures, not always associated with loss of con- of the eyes (114) and performing conjugate eye movements
sciousness (189). Unexpected startling, mostly loud noises, and (103,114,207). However, closing eyes is the most common effec-
other unexpected somatosensory stimuli, such as sudden move- tive technique (114).
ments, are common precipitating factors; very rarely, visual Paroxysmal discharges can also be provoked by voluntary
stimuli are capable of startling. Those EEGs are usually accom- eye closure and even by opening the eyes in darkness (103,119,
panied by a temporary desynchronization followed by a gener- 199,200,204), indicating that spike–wave discharges may be
alized or focal seizure pattern (116). The interictal awake EEG evoked without the influence of visual stimuli. Generalized and
shows frontal, central, or multifocal epileptiform discharges in focal paroxysmal discharges induced by such eye movements
the majority of patients (190–192). Based on studies by animal are usually maximal over the frontal areas (103). Sensory affer-
models and clinical observations (189,193), startle epilepsy has ents arising from contraction of the orbicularis oculi muscle
been classified as “proprioceptive epilepsy” (194). From a clini- (200) and proprioceptive impulses from the ocular muscles
cal-EEG study of 22 children with startle-provoked epilepti- (207) are thought to play a role in the mechanism of provoca-
form seizures (SPESs), considering the high diversity of SPES tion of seizure discharges by closing eyes and by conjugate eye
patients, Tibussek et al. (195) infer that a common underlying movements. In such epilepsy patients, similar paroxysmal dis-
pathophysiologic mechanism is unlikely. charges to those described above may also be induced by ocular
pressure (205) or passive lid closure in darkness (208). In an
Transcranial Magnetic Stimulation epilepsy patient, right occipital spikes and complex partial
Hufnagel et al. (196) used the method of transcranial magnetic seizures could be induced by convergence (209).
stimulation (TMS) to induce paroxysmal discharge; positive The above findings are most often seen in photosensitive
findings were obtained in 12 of 13 patients. Schuler et al. (197) epilepsy patients (66). Figure 12.14 is an example of generalized
statistically compared activation effects of paroxysmal discharge spike–wave discharges with an anterior maximum induced by
with those by TMS and HV in 10 patients with drug-resistant eye closure in darkness in comparison with that induced by a
partial epilepsy. TMS caused an activation of the focal paroxys- red flicker stimulation in a photosensitive epilepsy patient.
mal discharges only in three patients, whereas HV produced it in Figure 12.15 shows another example of generalized spike–wave
six cases; TMS even caused significant reduction of paroxysmal discharges induced by eye closure in darkness in a photosensi-
discharge in two patients. These results led them to conclude that tive epilepsy patient; similar spike–wave discharges were also
TMS was not better than HV. To contrast to low-frequency ( 1 induced by passive lid closure and ocular pressure in darkness.
Hz) TMS studies, Tassinari et al. (198) reported results obtained
by high-frequency ( 1 Hz) TMS (repetitive TMS [rTMS]) in Limb and Trunk Movements
more than 60 epilepsy patients; rTMS-induced seizures were Just as the rolandic mu rhythm is blocked by contralateral move-
found in 2 of 10 patients with progressive myoclonic epilepsy and ments, suppression of central spikes is also commonly observed
in 1 of 4 patients with epilepsia partialis continua. with contralateral motor activities, such as clenching of the fist
(13). Alternatively, active or passive movements of the limbs may
OTHER FORMS OF ACTIVATION provoke focal motor seizures and focal paroxysmal discharges in
a rare reflex epilepsy patient (210). Forster (119) reported a
Eye Movements patient who had clonic seizures of the right hand; seizures
Blinking is known to evoke spike–wave discharges (199,200). invariably occurred with the use of the right hand, such as with
Nadkarni et al. (201) reported that a 6-year-old boy’s blinking writing or finger tapping, even occurring when the patient
triggered central midtemporal spikes (CMTSs) occurring reflex- imagined movement of the hand. Rarely, seizure occurred with
Chapter 12 ■ Activation Methods 229

Figure 12.14 Generalized paroxysmal discharges induced


by a 15-Hz red flicker stimulation and eye closure in a dark
room in a 24-year-old woman with photosensitive epilepsy.
A red flicker stimulus was provided by a visual stimulator
(48); luminance maintained at 20 cd/ m 2.

use of the left hand. These seizures were associated with focal All descriptions cited below have only recently been grouped
spikes over the left motor areas. together as praxis-induced seizures (214): epilepsia arith-
Movement after various periods of rest may also induce metices, drawing-induced seizures, chess and card epilepsy,
seizures, including tonic seizure and paroxysmal choreoatheto- seizures during card games and draughts, reflex epilepsy evoked
sis (119,211,212). In such patients, in addition to startle or sud- by decision making or by specific psychic activity, seizures
denness, the motor movement itself and the proprioceptive evoked by playing with Rubik’s cube, seizures induced by think-
feedback secondary to the movements are thought to be ing, and reflex epilepsy with response to games of chance, cal-
involved in precipitating seizures (119,194). culations, and spatial decisions.
By use of a method of neuropsychological EEG activation
Neuropsychological EEG Activation (NPA), Matsuoka et al. (219) reported results obtained from
Reflex epilepsies are divided into a simple and a complex group 480 Japanese patients with different types of epilepsy. NPA tasks
according to the triggering mechanism. The former comprises consist of reading, speaking, writing, written arithmetic calcu-
seizure precipitation by simple sensory stimuli and by move- lation, mental arithmetic calculation, and spatial construction.
ments, whereas the latter involves complex mental processes, They provoked epileptic discharges in 38 patients (7.9%) and
such as reading (213–215), and a variety of other cognitive were accompanied by myoclonic seizures in 15 patients,
tasks. Regarding seizures elicited by cognitive tasks, the term absence seizures in 8, and simple partial seizures in 1. Among
praxis-induced seizures (216–218) has recently become popular. the cognitive tasks, mental activities mainly associated with the

Figure 12.15 Generalized paroxysmal discharges


induced by eye closure, passive lid closure, and ocular
pressure in an 18-year-old man with photosensitive
epilepsy. Simultaneous recordings of horizontal and
vertical eye movements are shown (bottom).
230 Part II ■ Normal EEG

use of the hands, that is, writing (68.4%), written calculation than that of “frontal midline theta” being recorded maximally at
(55.3%), and spatial construction (63.2%), provoked the most the frontal midline (223).
discharges, followed by mental calculation (7.9%) and reading
(5.3%). As for precipitating events, action programming–type Emotional Changes
activities were thought to be the most crucial in 32 out of the It has long been thought that certain emotional states may be
38 patients (84.2%), followed by thinking-type activities in influential either directly or indirectly in precipitating seizures
4 patients (10.5%). Regarding classification of epilepsies, (224,225). Stevens (226) used emotional activation of the EEG
seizure-precipitating mental activities were almost exclusively in 30 epilepsy patients, 8 with GTCSs, and 22 with complex par-
(in 36 out of the 38 patients) related to idiopathic generalized tial seizures. Stimuli consisted of a stressful interview that
epilepsies (IGEs), and were rarely (in only 2 out of the 38 elicited strong feelings of anger, sorrow, embarrassment, mirth,
patients) related to temporal lobe epilepsy. In IGE patients, and pleasure. Two thirds of GTCS patients showed no abnormal
provocative effects of NPA were related to myoclonic seizures changes, while three quarters of complex partial seizure
rather than absence or GTCSs. These results suggest that NPA is patients had abnormal findings. Abnormal changes not
a useful tool for examining the relationship between cognitive observed in previous tracings were seen in nearly half of the
function and epileptic seizures. complex partial seizure group, but in only one patient in the
Shown in Figure 12.16 are paroxysmal discharges induced by GTCS group. Similar emotional activation of EEG patterns has
written arithmetic in a juvenile myoclonic epilepsy (JME) been reported by Small et al. (227).
patient; the following new question (right tracing) provoked
more intensified paroxysmal discharges than those by the first Hypoglycemia
one (left tracing). The right tracing of Figure 12.17 illustrates Provocation of convulsive seizures and spike discharges by
bilateral paroxysmal discharges elicited by the Uchida–Kraepelin hypoglycemia, especially during insulin shock treatment of
test (continuous one-digit addition) in a JME patient who had schizophrenia (228), has been well documented (229);
recurrent myoclonic seizures during video games; he was non- spike–wave discharges are usually observed when the patient
photosensitive (220). Those shown on the right appear to be has “muscle twitches.” Convulsions and EEG abnormalities
similar to generalized paroxysmal discharges evoked by a video also occur in patients with spontaneous episodes of hyperin-
game (the left tracing), suggesting that seizures induced by video sulinism secondary to islet cell tumors of the pancreas; the
games are not caused by visual stimuli, but are instead produced same finding may be observed in rare patients with nonpan-
by combined factors of hand movements and calculation, which creatic tumors associated with hypoglycemia. Hypoglycemia
would be an example of praxis. of nonpancreatic origin (e.g., ketogenic diet) occasionally
During mental work requiring concentration of attention, facilitates generalized paroxysmal activity (11). Therefore,
some subjects show theta-activity bursts in the frontal midline EEG activation by fasting may be informative in generalized
(221). However, magnetoencephalography analysis (222) dis- epilepsy patients if no paroxysmal discharges could be found
closed that frontal midline theta-burst activities elicited by sim- on routine EEG examination. Attempts have also been made
ilar tasks in fact distribute widely in the frontal cortices of both to induce paroxysmal activity through hypoglycemia pro-
cerebral hemispheres. Therefore, the authors claim that an duced by tolbutamide (230,231). Hypoglycemic activation
expression of frontal mental theta waves is more appropriate may also by a useful technique for predicting the presence of

Figure 12.16 Generalized paroxysmal


discharges induced by written arithmetic in
a 25-year-old man with juvenile myoclonic
epilepsy. The left paroxysmal discharge in
the left tracing appeared 10 seconds after
calculation was started. As shown in the
right tracing, another question elicited
intensified paroxysmal discharges that
appeared 2 seconds after a calculation was
started; simultaneous video-EEG monitor-
ing revealed that myoclonic seizures of
both arms were associated with it.
Chapter 12 ■ Activation Methods 231

Figure 12.17 Paroxysmal discharges elicited by video game and Uchida–Kraepelin test in a 16-year-old boy with JME.
The left tracing shows generalized paroxysmal discharges associated with myoclonic seizures of upper extremities; they
occurred 3 minutes after the game (Puyo Puyo) started. Uchida–Kraepelin test at 2 and 3 minutes elicited bilateral parox-
ysmal discharges associated with myoclonic seizures of both hands.

pathology in patients considered for anterior temporal lobec- operative electrocorticographic and depth electrode recording
tomy (232). (239). These methods, however, have not been widely used.

Antiepileptic Drug Withdrawal Procedure for


PHARMACOLOGIC ACTIVATION
Long-Term Monitoring
Drugs Before epilepsy surgery, antiepileptic drug (AED) withdrawal in
Alpha-chloralose or alpha-chloralose-scopolamine has occa- the video-EEG monitoring environment is currently being car-
sionally been used for EEG activation in epilepsy patients with ried out for the purpose of increasing the probability of inter-
normal routine EEG (233,234). The adult patient was given a ictal and ictal abnormalities occurring during the record. The
preparation consisting of scopolamine hydrobromide 0.5 mg most important risk of AED withdrawal is the development of
and alpha chloralose 500 mg, in capsule form, by mouth (233). status epilepticus (240,241). Such complications of AED with-
Of 30 epilepsy patients, 22 showed activation efficacies. drawal, however, can be prevented if patients are under the care
Intravenous tripelennamine (Pyribenzamine) is also known to of experienced staff (8).
activate focal or diffuse paroxysmal discharges (235). In addi-
tion, it has been reported that intravenous administration of Metrazol and Megimide
some antiparkinsonism agents, such as 1 mg of scopolamine Pentylenetetrazol (Metrazol) has been used therapeutically to
hydrobromide (236), 10 to 20 mg of procyclidine hydrochloride induce convulsions in schizophrenia patients (242). Ziskind et
(237), or 10 mg of methixene hydrochloride (238), is capable of al. (243) demonstrated the value of Metrazol in triggering
provoking focal paroxysmal discharges, especially in temporal paroxysmal discharges. A series of similar studies followed:
lobe epilepsy patients. Intravenous alfentanil hydrochloride, Kaufman et al. (244), Ziskind and Bercel (245), Cure et al. (246),
which is a short-duration opioid analgesic, has been demon- Jasper and Courtois (247), and Bancaud (248). The effectiveness
strated to be effective in inducing temporal lobe spikes for intra- of a given amount of Metrazol depends to a large extent on the
232 Part II ■ Normal EEG

rapidity of its injection as well as the total amount in relation to et al. (257) provides us important clues for future EEG studies:
body weight (247). These authors recommended dilution of a performing standard awake EEG in patients with JME, they
10% solution of Metrazol in normal saline so that 1 cc of the obtained a significantly greater rate of detection of generalized
mixture contains 1 mg/kg of body weight. One milliliter is then epileptiform abnormalities in the morning than in an after-
given thereafter every 30 seconds. noon session.
Among epilepsy patients, those with bilaterally synchronous
EEG disturbances were more sensitive than those with focal ACKNOWLEDGMENTS
cortical seizures (246). On the other hand, Ajmone-Marsan and
Ralston (249) showed activation of various types of focal The authors wish to thank Professor Y. Tsukahara for his discus-
seizures by this method. They reported that the epileptogenic sions of the manuscript. They are also very grateful to Mr. B.A.
supplementary motor area appears to be very sensitive to Fast for manuscript preparation assistance.
Metrazol stimulation.
Gastaut (250) described an activation method combined REFERENCES
with Metrazol and IPS (see Ref. (53)). He pointed out that this
test differentiates between epilepsies involving a subcortical 1. Foerster O. Hyperventilationsepilepsie. Zbl Ges Neurol Psychiatr.
diencephalic mechanism and those of cortical origin. A low 1924;38:289–293.
threshold to IPS was found in idiopathic (absence seizure group 2. Nims LF, Gibbs EL, Lennox WG, et al. Adjustment of acid–base
balance of patients with petit mal epilepsy to overventilation. Arch
in particular) and myoclonic epilepsies. Normal or elevated
Neurol Psychiatry. 1940;43:262–269.
thresholds were found in the focal cortical group. Although this 3. Morrice JKW. Slow wave production in the EEG, with reference
method was more sensitive than activation by Metrazol alone, a to hyperpnoea, carbon dioxide and autonomic balance.
high incidence of low-threshold responses (5 mg/kg or less) in Electroencephalogr Clin Neurophysiol. 1956;8:49–72.
a number of other conditions such as schizophrenia and hyste- 4. Achenbach-Ng J, Siao TCP, Marvroudakis N, et al. Effects of rou-
ria, and even normal subjects, has been observed. tine hyperventilation on PCO2 and PO2 in normal subjects: impli-
Bemegride (Megimide) was first investigated as a barbiturate cations for EEG interpretations. J Clin Neurophysiol. 1994;11:
antagonist (251). Megimide was then found to have a convul- 220–225.
sant action similar to that of Metrazol (252,253). 5. Gibbs FA, Gibbs EL, Lennox WG. Electroencephalographic response
Despite numerous studies, especially between 1946 and 1970 to overventilation and its relation to age. J Pediatr. 1943;23:497–505.
(254), neither of the above tests is presently considered to be a 6. Gotoh F, Meyer JS, Takagi Y. Cerebral effects of hyperventilation in
man. Arch Neurol. 1965;12:410–423.
satisfactory method for distinguishing between potential
7. Yamatani M, Konishi T, Murakami M, et al. Hyperventilation activa-
epileptic and nonepileptic individuals. Furthermore, the risk of tion on EEG recording in childhood. Epilepsia. 1994;35:1199–1203.
failure in seizure control with resection of a focus that was 8. Fisch BC, So EL. Activation methods. In: Ebersole JS, Pedley TA,
localized by chemically (Megimide) induced seizures is two eds. Current Practice of Clinical Electroencephalography. 3rd ed.
times higher than that with resection of a spontaneous seizure Philadelphia, PA: Lippincott Williams & Wilkins; 2003:246–270.
focus (255). Therefore, drugs are no longer used to induce 9. Sherwin I. Hyperventilation: mode of action and application in
seizures for localization in epilepsy surgery (8). electroencephalography. Am J EEG Technol. 1984;24:201–211.
10. Patel VM, Maulsby RL. How hyperventilation alters the EEG: a
Re-enactment of the Triggering Situation review of controversial viewpoints emphasizing neurophysiologi-
When a fixed set of ambient circumstances is associated with cal mechanisms. J Clin Neurophysiol. 1987;4:101–120.
convulsive or nonconvulsive paroxysmal attacks, re-enactment 11. Niedermeyer E. The Generalized Epilepsies. Springfield, IL: Charles
C. Thomas; 1972.
of the situation, if possible, is desirable for reaching a diagnosis.
12. Esquivel E, Chaussai M, Plouin P, et al. Physical exercise and
Fariello et al. (256) reported that re-enactment determined the voluntary hyperventilation in childhood absence epilepsy.
diagnosis in 30 of 32 patients with paroxysmal disorders. The Electroencephalogr Clin Neurophysiol. 1991;79:127–132.
authors claim that an appropriately executed re-enactment 13. Niedermeyer E. Focal and generalized seizure discharges in the
should entail polygraphic recording of at least EEG, electrocar- electroencephalogram and their response to intravenous diazepam.
diogram, and respiration. Int Med Dig. 1972;7:49–61.
14. Bostem F. Hyperventilation. In: Remond A, ed. Handbook of
Electroencephalography and Clinical Neurophysiology. Vol 3D.
CONCLUDING REMARKS Amsterdam: Elsevier; 1976:74–88.
The greater the number of different activation methods used in 15. Ziegler DK, Hassanein RS, Dick AR. Effect of age and depth of
EEG evaluation of an epilepsy patient, the greater the chance of hyperventilation on a quantitative electroencephalographic
response. Clin Electroencephalogr. 1975;6:184–190.
obtaining abnormal findings. From a practical point of view,
16. Petersen I, Eeg-Olofsson O. The development of the electroen-
however, desirable activations are those methods that can be cephalogram in normal children from age 1 through 15 years—
carried out easily, in a short time, with affordable equipment, nonparoxysmal activity. Neuropaediatrie. 1971:2:247–304.
and without undesirable side effects for patients. Special meth- 17. Kellaway P. An orderly approach to visual analysis: the parameters
ods of activation are needed in epilepsy patients for whom of the normal EEG in adults and children. In: Klass DW, Daly DD,
seizures tend to appear under certain conditions while awake. eds. Current Practice of Clinical Electroencephalography. New York:
Lastly, considering the methods while awake, a report by Labate Raven Press; 1979:69–147.
Chapter 12 ■ Activation Methods 233

18. Petersen I, Eeg-Olofsson O. The development of the electroen- 40. Walter VJ, Walter WG. The effect of physical stimuli on the EEG.
cephalogram in normal children from the age of 1 through 15 years: Electroencephalogr Clin Neurophysiol. 1951;2(suppl):60–68.
non-paroxysmal activity. Neuropadiatrie. 1971;2:247–304. 41. Kooi KA, Tucker PR, Marshall RE. Fundamentals of Electro-
19. Bickford RG. Activation procedures and special electrodes. In: encephalography. Hagerstown, MD: Harper & Row; 1978.
Klass DW, Daly DD, eds. Current Practice of Clinical Electro- 42. Pampiglione G, Harden A. So-called neuronal ceroid lipofusci-
encephalography. New York: Raven Press; 1979:269–305. nosis: neurophysiological studies in 60 children. J Neurol
20. Davis H, Wallace WM. Factors affecting changes produced in elec- Neurosurg Psychiatry. 1977;40:323–330.
troencephalograms by standardized hyperventilation. Arch Neurol 43. Pinsard N, Livet MO, Saint-Jean M. A case of cerebral lipidosis
Psychiatry. 1942;47:606–625. with an atypical presentation. Rev Electroencephalogr Neurophysiol
21. Billinger TW, Frank GS. Effects of posture on EEG slowing during Clin. 1978;8:175–179.
hyperventilation. Am J EEG Technol. 1969;9:22–27. 44. Coull BM, Pedley TA. Intermittent photic stimulation: clinical use-
22. Dalby MA. Epilepsy and 3 per second spike and wave rhythms. fulness of non-convulsive responses. Electroencephalogr Clin
Acta Neurol Scand. 1969;40(suppl):1–183. Neurophysiol. 1978;44:353–363.
23. Kiloh LG, McComas AJ, Osselton JW. Clinical Electro- 45. Chiba T, Kitawaki M, Aoki Y, et al. Hemodialysis and electroen-
encephalography. 3rd ed. London: Butterworths; 1972. cephalogram: through a long term observation. Rinsho Noha
24. Miley CE, Forster FM. Activation of partial complex seizures by (Osaka). 1982;24:195–201.
hyperventilation. Arch Neurol. 1977;34:371–373. 46. Takahashi T, Tsukahara Y. Influence of red light and pattern on
25. Arain AM, Arbogast PG, Abou-Khalil BW. Utility of daily super- photic driving. Tohoku J Exp Med. 1979;127:45–52.
vised hyperventilation during long-term video-EEG monitoring. 47. Harding GFA, Jeavons PM. Photosensitive Epilepsy. New ed.
J Clin Neurophysiol. 2009;26(1):17–20. London: Mac Keith Press; 1994.
26. Engel J Jr. A practical guide for routine EEG studies in epilepsy. 48. Tsukahara Y, Takahashi T. Visual stimulator for EEG activation.
J Clin Neurophysiol. 1984;1:109–142. Electroencephalogr Clin Neurophysiol. 1973;35:333–335.
27. Kodama N, Aoki Y, Hiraga H, et al. Electroencephalographic findings 49. Tsukahara Y, Takahashi T. Pattern-evoked high-amplitude photic
in children with moyamoya disease. Arch Neurol. 1979;36:16–19. driving in epileptic patients. In: Ito M, ed. Integrative Control
28. Kameyama M, Shirane R, Tsurumi Y, et al. Evaluation of cerebral Functions of the Brain. Vol 2. Tokyo: Kodansha Scientific; 1979:
blood flow and metabolism in childhood moyamoya disease: an 90–92.
investigation into “re-build-up” on EEG by positron CT. Child 50. Takahashi T. Activation methods. In: Niedermeyer E, Lopes da
Nerv Syst. 1986;2:130–133. Silva F, eds. Electroencephalography: Basic Principles, Clinical
29. Diamini N, Goyal S, Jarosz J, et al. Paroxysmal episodes, “re-build up” Applications and Related Fields. 3rd ed. Baltimore, MD: Williams &
phenomenon and moyamoya disease. Epileptic Disord. 2009;11(4): Wilkins; 1993:241–262.
324–328. 51. Takahashi T, Tsukahara Y, Kaneda S. EEG activation by use of stro-
30. AEEGS Guidelines Committee. Guideline one: minimum techni- boscope and visual stimulator SLS-5100. Tohoku J Exp Med. 1980:
cal requirements for performing clinical electroencephalography. 130:403–409.
J Clin Neurophysiol. 1994;11:4–5. 52. Takahashi T, Kataoka K, Tsukahara Y. Power spectral analysis of
31. Epstein MA, Duchowny M, Jayakar P, et al. Altered responsive- photic driving elicited by flickering dot pattern and red flicker
ness during hyperventilation-induced EEG slowing: a non- stimuli in adult psychiatric outpatients—with special reference to
epileptic phenomenon in normal children. Epilepsia. 1994;35(6): age and gender. Tohoku J Exp Med. 1988;156:165–173.
1204–1207. 53. Takahashi T. Activation methods. In: Niedermeyer E, Lopes da
32. Lum LM, Connolly MB, Farrell K, et al. Hyperventilation-induced Silva F, eds. Electroencephalography: Basic Principles, Clinical
high-amplitude rhythmic slowing with altered awareness: a video- Applications and Related Fields. 4th ed. Baltimore, MD: Williams &
EEG comparison with absence seizures. Epilepsia. 2002;43(1): Wilkins; 1999;261–284.
1372–1378. 54. Takahashi T. Photic Driving Response elicited by Low-Luminance
33. Adrian ED, Matthews BHC. The Berger rhythm: potential changes Visual Stimulation: An Atlas. Sendai, Japan: Tohoku University
from the occipital lobes in man. Brain. 1934;57:355–385. Press; 2005.
34. Bickford RG, Daly D, Keith HM. Convulsive effects of light stimu- 55. Takahashi T. EEG activation by visual stimuli. In: Sato M,
lation in children. Am J Dis Child. 1953;86:170–183. Matsuoka H, eds. The Newest Clinical Electroencephalography.
35. Walter WG, Dovey VJ, Shipton H. Analysis of the electrical Tokyo: Asakura Shoten; 1993:323–344.
response of the human cortex to photic stimulation. Nature. 1946; 56. Takahashi T, Fukushi SM, Suzuki T. Diagnosis of amblyopia on the
158:540–541. basis of the photic drivings evoked by flickering dot pattern. J
36. Kasteleijn-Nolst Trenité DGA. Reflex seizures induced by intermit- Sendai City Hosp. 1981;2:21–26.
tent light stimulation. In: Zifkin BG, Andermann F, Beaumanoir A, 57. Takahashi T, Tomioka H. Photic driving evoked by hemi-field
et al., eds. Reflex Epilepsies and Reflex Seizures: Advances in flickering dot pattern stimulation in a patient with brain tumor.
Neurology. Vol 75. Philadelphia, PA: Lippincott-Raven; 1998:99–121. Electroencephalogr Clin Neurophysiol. 1985;61:381–384.
37. Kasteleijn-Nolst Trenité DGA, Binnie CD, Harding GFA, et al. Photic 58. Gastaut H, Rémond A. L’activation de l’électroencéphalogramme
stimulation: standardization of screening methods. Epilepsia. dans les affections cérébrales non épileptogènes (vers une neuro-
1999;40(suppl 4):75–79. physiolgie clinique). Rev Neurol. 1949;81:594–598.
38. Chatrian GE, Bergamini L, Dondey M, et al. A glossary of terms 59. Bickford RG, Sem-Jacobsen GW, White PT, et al. Some observa-
most commonly used by clinical electroencephalographers. tions on the mechanism of photic and photometrazol activation.
Electroencephalogr Clin Neurophysiol. 1974;37:538–548. Electroencephalogr Clin Neurophysiol. 1952;4:275–282.
39. Ellingson RJ. Cortical electrical responses to visual stimulation in 60. Niedermeyer E, Fineyre F, Riley T, et al. Myoclonus and the elec-
the human infant. Electroencephalogr Clin Neurophysiol. 1960;12: troencephalogram: a review. Clin Electroencephalogr. 1979;10:
633–677. 75–95.
234 Part II ■ Normal EEG

61. Gastaut H, Trevisan C, Naquet R. Diagnostic value of electroen- 83. Guerrini R, Dravet C, Genton P, et al. Idiopathic photosensitive
cephalographic abnormalities provoked by intermittent photic occipital lobe epilepsy. Epilepsia. 1995;36:883–891.
stimulation. Electroencephalogr Clin Neurophysiol. 1958;10:194–195 84. Kasteleijn-Nolst Trenité DGA, Binnie CD, Meinardi H.
[abstract]. Photosensitive patients: symptoms and signs during intermittent
62. Kooi K, Thomas MH, Mortenson FN. Photoconvulsive and photo- photic stimulation and their relation to seizures in daily life.
myoclonic responses in adults: an appraisal of their clinical signif- J Neurol Neurosurg Psychiatry. 1987;560:1436–1549.
icance. Neurology. 1960;10:1051–1058. 85. Gambardella A, Aguglia U, Oliveri RL, et al. Photic-induced
63. Melsen S. The value of photic stimulation in the diagnosis of epileptic negative myoclonus: a case report. Epilepsia. 1996;37:
epilepsy. J Nerv Ment Dis. 1959;128:508–519. 492–494.
64. Reilly EW, Peters JF. Relationship of some varieties of electroen- 86. Doose H, Gerken H, Hien-Volpel KF, et al. Genetics of photosen-
cephalographic photosensitivity to clinical convulsive disorders. sitive epilepsy. Neuropaediatrie. 1969;1:56–73.
Neurology. 1973;23:1040–1057. 87. Takahashi T. Photoconvulsive response and disposition. Folia
65. Small JG. Photoconvulsive and photomyoclonic responses in psychi- Psychiatr Neurol Jpn. 1976;30:349–356.
atric patients. Electroencephalogr Clin Neurophysiol. 1971;2:78–88. 88. Waltz S, Christen H-J, Doose H. The different patterns of pho-
66. Newmark ME, Penry JK. Photosensitivity and Epilepsy: A Review. toparoxysmal response—a genetic study. Electroencephalogr Clin
New York: Raven Press; 1979. Neurophysiol. 1992;83:138–145.
67. Victor M. The role of alcohol in the production of seizures. In: 89. Quirk JA, Fish DR, Smith SJM, et al. Incidence of photosensitive
Niedermeyer E, ed. Modern Problems of Pharmacopsychiatry— epilepsy: a prospective national study. Electroencephalogr Clin
Epilepsy. Vol 4. Basel: Karger; 1970:185–199. Neurophysiol. 1995;95:260–267.
68. Wikler A, Essig CF. Withdrawal seizures following chronic intoxi- 90. De Graaf AS, Lombard CJ, Classen DA. Influence of ethnic and
cation with barbiturates and other sedative drugs. In: Niedermeyer geographic factors on the classic photoparoxysmal response in
E, ed. Modern Problems of Pharmacopsychiatry— Epilepsy. Vol 4. the electroencephalogram of epilepsy patients. Epilepsia. 1995;36:
Basel: Karger; 1970:170–184. 219–223.
69. Bax-aan de Stegge BM, van Putten MJAM. A patient with a transient 91. Rushton DN. Space invader epilepsy. Lancet. 1981;1:501.
photomyogenic response. Clin Neurophysiol. 2010;121:118–120. 92. Kasteleijn-Nolst Trenité DGA, Martins da Silva A, Ricci S, et al.
70. Harding GFA, Harding PF. Televised material and photosensitive Video games are exciting: a European study of video game-
epilepsy. Epilepsia. 1999;40(suppl 4):65–69. induced seizures and epilepsy. Epileptic Disord. 2002;4:121–128.
71. Topalkara K, Alarcon G, Binnie CD. Effects of flash frequency and 93. Takahashi T. Photosensitive Epilepsy: EEG Diagnosis by Low-
repetition of intermittent photic stimulation on photoparoxysmal Luminance Visual Stimuli and Preventive Measures. Tokyo: Igaku-
responses. Seizure. 1998;7:249–255. Shoin Publication Service; 2002.
72. Klass DW, Fischer-Williams M. Sensory stimulation, sleep and sleep 94. Quirk JA, Fish DR, Smith SJM, et al. First seizures associated with
deprivation. In: Remond A, ed. Handbook of Electroencephalography playing electronic screen games: a community-based study in
and Clinical Neurophysiology. Vol 3D. Amsterdam: Elsevier; Great Britain. Ann Neurol. 1995;37:733–737.
1976:5–73. 95. Binnie CD, Harding GFA, Richens A, et al. Video games and
73. Jeavons PM, Harding GFA. Photosensitive Epilepsy: A Review of the epileptic seizures—a consensus statement. Seizure. 1994;3:
Literature and a Study of 460 Patients. London: Heinemann; 1975. 245–246.
74. Van Egmond P, Binnie, CD, Veldhuizen R. The effect of back- 96. Jayakar P, Chiappa KH. Clinical correlations of photoparoxysmal
ground illumination on sensitivity to intermittent photic stimula- responses. Electroencephalogr Clin Neurophysiol. 1990;75:251–254.
tion. Electroencephalogr Clin. Neurophysiol. 1980;48:599–601. 97. Solomon S, Fine D. The precipitation of seizures by photic stim-
75. Panayiotopoulos CP. Effectiveness of photic stimulation on various ulation in a patient with hypoparathyroidism. J Nerv Ment Dis.
eye-states in photosensitive epilepsy. J Neurol Sci. 1974;23: 1960;130:253–260.
165–173. 98. Victor M, Brausch C. The role of abstinence in the genesis of
76. Hishikawa Y, Yamamoto J, Furuya E, et al. Photosensitive epilepsy: alcoholic epilepsy. Epilepsia. 1967;8:1–20.
relationships between the visual evoked responses and the epilep- 99. Gastaut H. Introduction to the study of organic generalized
tiform discharges induced by intermittent photic stimulation. epilepsies. In: Gastaut H, Jasper H, Bancaud J, et al., eds. The
Electroencephalogr Clin Neurophysiol. 1967;23:320–334. Physiopathogenesis of the Epilepsies. Springfield, IL: Charles C.
77. Sato S, Dreifuss FE, Penry JK. Photic sensitivity of children with Thomas; 1969:147–157.
absence seizures in slow wave sleep. Electroencephalogr Clin 100. Lee RG, Blair RDG. Evolution of EEG and visual evoked response
Neurophysiol. 1975;39:479–489. changes in Jakob–Creutzfeldt disease. Electroencephalogr Clin
78. Maheshwari MC, Jeavons PM. The clinical significance of occipi- Neurophysiol. 1973;35:133–142.
tal spikes as a sole response to intermittent photic stimulation. 101. Gilliam FG, Chiappa KH. Significance of spontaneous epilepti-
Electroencephalogr Clin Neurophysiol. 1975;39:93–95. form abnormalities associated with a photoparoxysmal response.
79. Takahashi T. Activation methods. In: Niedermeyer E, Lopes da Neurology. 1995;45(3):453–455.
Silva F, eds. Electroencephalography: Basic Principles, Clinical 102. Bickford RG, Klass DW. Sensory precipitation and reflex mecha-
Applications and Related Fields. Baltimore, MD/Munich: Urban & nisms. In: Jasper HH, Ward AA, Pope A, eds. Basic Mechanisms of
Schwarzenberg; 1982:179–195. the Epilepsies. Boston, MA: Little, Brown and Company;
80. Jeavons PM. The use of photic stimulation in clinical electroen- 1969:543–564.
cephalography. Proc Electrophysiol Technol Assoc. 1969;16:225–240. 103. Takahashi T, Tsukahara Y. Generalized paroxysmal discharges
81. Wilkins AJ, Darby CE, Binnie CD, et al. Television epilepsy: the role induced by visual stimuli and eye movements. Tohoku J Exp Med.
of pattern. Electroencephalogr Clin Neurophysiol. 1979;47:163–171. 1975;115:1–10.
82. Stevens JR. Control and peripheral factors in epileptic discharge: 104. Jeavons PM, Harding GFA, Panayiotopoulos CP, et al. The
clinical studies. Arch Neurol. 1962;7:330–338. effect of geometric patterns combined with intermittent photic
Chapter 12 ■ Activation Methods 235

stimulation in photosensitive epilepsy. Electroencephalogr Clin 128. Bickford RG, Klass DW. Stimulus factors in the mechanism of tel-
Neurophysiol. 1972;33:221–224. evision induced seizures. Trans Am Neurol Assoc. 1962;87:176–178.
105. Takahashi T, Tsukahara Y. Photoconvulsive response induced by 129. Binnie CD, Wilkins AJ. Visually induced seizures not caused by
use of “visual stimulator”. Tohoku J Exp Med. 1980;130:273–281. flicker (intermittent light stimulation). In: Zifkin BG,
106. Takahashi T, Tsukahara Y. Influence of color on the photoconvulsive Andermann F, Beaumanoir A, et al., eds. Reflex Epilepsies and
response. Electroencephalogr Clin Neurophysiol. 1976;41:124–136. Reflex Seizures: Advances in Neurology. Vol 75. Philadelphia, PA:
107. Takahashi T, Tsukahara Y. Usefulness of blue sunglasses in pho- Lippincott-Raven; 1998:123–138.
tosensitive epilepsy. Epilepsia. 1992;33:517–521. 130. Chatrian GE, Lettich E, Miller LH, et al. Pattern-sensitive
108. Takahashi T. Techniques of intermittent photic stimulation and epilepsy. 1. An electrographic study of its mechanisms. Epilepsia.
paroxysmal responses. Am J EEG Technol. 1989;29:205–218. 1970;11:125–149.
109. Takahashi T. Pathophysiological mechanisms of photosensitivity 131. Darby CE, Wilkins AJ, Binnie CD, et al. Routine testing for pat-
in IGEs. In: Malafosse A, Genton P, Hirsch E, et al., eds. Idiopathic tern sensitivity. J Electrophysiol Technol. 1980;6:202–210.
Generalized Epilepsies: Clinical, Experimental and Genetic Aspects. 132. Klass DW. Pattern activation of seizures. In: Lüders HO,
London: John Libbey; 1994:305–315. Noachtar S, eds. Epileptic Seizures: Pathophysiology and Clinical
110. Binnie CD, Jeavons PM. Photosensitive epilepsies. In: Roger J, Semiology. New York: Churchill Livingstone; 2000:598–608.
Bureau M, Dravet C, et al., eds. Epileptic Syndromes in Infancy, 133. Wilkins AJ, Binnie CD, Darby CE. Visually-induced seizures. Prog
Childhood and Adolescence. 2nd ed. London: John Libbey; Neurobiol. 1980;15:85–117.
1992:299–305. 134. Binnie CD, Findlay J, Wilkins AJ. Mechanisms of epileptogenesis
111. Takahashi T. EEG diagnosis of photosensitive seizures and its in photosensitive epilepsy implied by the effects of moving pat-
preventive measures. Jpn J Clin Neurophysiol. 2000;28:236–245. terns. Electroencephalogr Clin Neurophysiol. 1985;61:1–6.
112. Chiappa KH, Hill RA, Huang-Hellinger F, et al. Photosensitive 135. Takahashi T, Sasaki M. Pattern sensitivity: its activation method
epilepsy studied by functional magnetic resonance imaging and and clinical EEG findings. Rinsho Nohha (Osaka). 1971;13:
magnetic resonance spectroscopy. Epilepsia. 1999;40(suppl 4):3–7. 727–732.
113. Hill RA, Chiappa KH, Huang-Hellinger F, et al.. Hemodynamic 136. Takahashi T. Study of visual epilepsy. 4. Pattern activation and
and metabolic aspects of photosensitive epilepsy revealed by clinical EEG findings. Clin Neurol (Tokyo). 1974;14:479–488.
functional magnetic resonance imaging and magnetic resonance 137. Radhakrishnan K, Sunku AJ, Donat JF, et al. Pattern-induced
spectroscopy. Epilepsia. 1999;40:912–920. negative occipital potentials (PINOP). J Clin Neurophysiol. 2007;
114. Takahashi T. EEG activation by movement of the eyelids and 24:277–280.
eyes. Rinsho Noha (Osaka). 1976;18:334–344. 138. Stefansson SB, Darby CE, Wilkins AJ, et al. Television epilepsy
115. Westmoreland BF, Sharbrough FW. Posterior slow wave tran- and pattern sensitivity. Br Med J. 1977;2:88–90.
sients associated with eye blinks in children. Am J EEG Technol. 139. Koshino Y, Niedermeyer E. Enhancement of rolandic mu rhythm
1975;15:14–19. by pattern vision. Electroencephalogr Clin Neurophysiol. 1975;38:
116. Gastaut H, Tassinari CA. Triggering mechanisms in epilepsy: the 535–538.
electro-clinical point of view. Epilepsia. 1966;7:85–138. 140. Takahashi T, Nakasato N, Yokoyama H, et al. Low-luminance
117. Green JB. Seizures on closing the eyes. Neurology. 1968;18:391–396. visual stimuli compared with stroboscopic IPS in eliciting PPR in
118. Darby CE, De Korte RA, Binnie CD, et al. The self-induction of photosensitive patients. Epilepsia. 1999;40(suppl 4):44–49.
epileptic seizures by eye closure. Epilepsia. 1980;21:31–42. 141. Soso MJ, Lettich E, Belgum JH. Pattern-sensitive epilepsy. II.
119. Forster FM. Reflex Epilepsy, Behavioral Therapy and Conditional Effects of pattern orientation and hemifield stimulation.
Reflexes. Springfield, IL: Charles C. Thomas; 1977. Epilepsia. 1980;21:313–323.
120. Gumnit RJ, Niedermeyer E, Spreen O. Seizure activity uniquely 142. Takahashi T. Lateral hemifield flickering pattern stimulation in a
inhibited by patterned vision. Arch Neurol. 1965;13:363–368. patient with pattern-sensitive epilepsy. Epilepsia. 1983;24:548–556.
121. Takahashi T. Influence on EEGs of gazing at a fixation point and 143. Takahashi T. Hemifield red flicker stimulation in a patient with
of ganzfeld. Brain Nerve (Tokyo). 1976;28:95–103. pattern-sensitive epilepsy. Epilepsia. 1984;25:223–228.
122. Colon EJ, Vingerhoets HM, Notermans SLH, et al. Off-response: 144. Wilkins AJ, Binnie CD, Darby CE. Interhemispheric differences
its clinical incidence in very young children. Electroencephalogr in photosensitive epilepsy. I. Pattern sensitivity thresholds.
Clin Neurophysiol. 1979;46:601–604. Electroencephalogr Clin Neurophysiol. 1981;52:461–468.
123. Takahashi T, Tsukahara Y. EEG activation by red color. Igaku No 145. Takahashi T. Hemi- and quadrant-field visual stimuli in EEG
Ayumi (Tokyo). 1972;83:25–26. diagnosis of photosensitive epilepsy. In: Kimura J, Shibasaki H,
124. Lugaresi E, Cirignotta F, Montagna P. Occipital lobe epilepsy with eds. Recent Advances in Clinical Neurophysiology. Amsterdam:
scotosensitive seizures: the role of central vision. Epilepsia. Elsevier; 1996:223–228.
1984;25:115–120. 146. Takahashi T, Tsukahara Y. Photoparoxysmal response elicited by
125. Panayiotopoulos CP. Conversion of photosensitive to scotosensi- flickering dot pattern stimulation and its optimal spatial fre-
tive epilepsy: report of a case. Neurology. 1979;29:1550–1554. quency of provocation. Electroencephalogr Clin Neurophysiol.
126. Panayiotopoulos CP. With comments and contributions from 1998;106:40–43.
Binnie, C.D., and Takahashi, T. Fixation-off-sensitive epilepsies: 147. Takahashi T, Tsukahara Y. Photoparoxysmal response elicited by
clinical and EEG characteristics. In: Wolf P, ed. Epileptic Seizures flickering dot pattern stimulation to the center and periphery.
and Syndromes. London: John Libbey; 1994:55–66. Clin Neurophysiol. 2000;111:1968–1973.
127. Panayiotopoulos CP. Fixation-off sensitive epilepsies: clinical and 148. Takahashi T, Tsukahara Y. Photoparoxysmal response-evoked by
EEG characteristics. In: Zifkin BG, Andermann F, Beaumanoir regional visual stimuli in a photosensitive epilepsy patient. In:
A, et al., eds. Reflex Epilepsies and Reflex Seizures: Advances in Koga Y, Nagata K, Hirata K, eds. Pan-Pacific Conference on Brain
Neurology. Vol 75. Philadelphia, PA: Lippincott-Raven; 1998: Topography. Amsterdam: Elsevier; 1997:507–511.
139–157. 149. Zeki S. A Vision of the Brain. Oxford: Blackwell Scientific; 1993.
236 Part II ■ Normal EEG

150. Allison T, Begleiter A, McCarthy G, et al. Electrophysiological 169. Berkovic SF, So NK, Andermann F. Progressive myoclonus
studies of color processing in human visual cortex. Electro- epilepsies: clinical and neurophysiological diagnosis. J Clin
encephalogr Clin Neurophysiol. 1993;88:343–355. Neurophysiol. 1991;8:261–274.
151. Lueck CJ, Zeki S, Friston KJ, et al. The colour centre in the cere- 170. Dawson GD. Investigations on a patient subject to myoclonic
bral cortex of man. Nature. 1989;340:386–389. seizures after sensory stimulation. J Neurol Neurosurg Psychiatry.
152. Takahashi T. EEG Atlas of Photosensitive Epilepsy Studied Using 1947;10:141–162.
Low-Luminance Visual Stimulation: An Atlas. Sendai, Japan: 171. Halliday AM. The electrophysiological study of myoclonus in
Tohoku University Press; 2008. man. Brain. 1967;90:241–284.
153. Arellano AP, Schwab RS, Casby JU. Sonic activation. 172. Shibasaki H, Yamashita Y, Neshige R, et al. Pathogenesis of giant
Electroencephalogr Clin Neurophysiol. 1950;2:215–217. somatosensory evoked potentials in progressive myoclonic
154. Gastaut HJ, Benoit PH, Vigouroux M, et al. Potentiels évoqés par epilepsy. Brain. 1985;108:225–240.
des stimuli auditifs sur la région temporale de certains épilep- 173. Sutton GG, Mayer RF. Focal reflex myoclonus. J Neurol Neurosurg
tiques. Electroencephalogr Clin Neurophysiol. 1954;6:557–564. Psychiatry. 1974;37:207–217.
155. Peet RM, Daly DD, Bickoford RG. Some clinical and electroen- 174. Mima T, Terada K, Ikeda A, et al. Afferent mechanism of cortical
cephalographic responses to sound stimulation in epileptic myoclonus studied by proprioception-related SEPs. Electro-
patients. Trans Am Neurol Assoc. 1952;7:215–217. encephalogr Clin Neurophysiol. 1997;104:51–59.
156. Takahashi T. EEG responses to various stimuli. In: Shimazono Y, 175. Plasmati R, Michelucci R, Forti A, et al. The neurophysiological
Kitamura K, Otomo E, eds. Atlas of Clinical EEG. Tokyo: Bunkodo; features of benign partial epilepsy with rolandic spikes. In: Degan
1974:157–193. R, Dreifuss FE, eds. Benign Localized and Generalized Epilepsies of
157. Vigouroux M, Benoit P, Gastaut H. Evoked potentials by auditory Early Childhood [Epilepsy Res. suppl. 6]. Amsterdam: Elsevier;
stimuli in the temporal region in an epileptic patient. 1992:45–48.
Electroencephalogr Clin Neurophysiol. 1954;6:163–164. 176. Plasmati R, Pastorelli F, Tassinari CA. Giant midlatency somatosen-
158. Prechtl HFR. Provocation of electroencephalographic changes in sory evoked potentials in benign epilepsy with rolandic spikes. In:
the temporal region by intermittent acoustic stimuli. Lüders HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology and
Electroencephalogr Clin Neurophysiol. 1959;11:511–519. Clinical Semiology. New York: Churchill Livingstone; 2000:62–68.
159. Wieser HG, Hungerbueler H, Siegel AM, et al. Musicogenic 177. Goldie L, Green JN. A study of the psychological factors in a case
epilepsy: review of the literature and case report with ictal single of sensory reflex epilepsy. Brain. 1959;82:505–524.
photon emission computed tomography. Epilepsia. 1997;38: 178. Van Cott AC, Blatt I, Brenner RP. Stimulus-sensitive seizures in
200–207. postanoxic coma. Epilepsia. 1996;37:868–874.
160. Rosenow F, Lüders HO. Hearing-induced seizures. In: Lüders 179. Koutroumanidis M, Tsatsou K, Bonakis A, et al. Stimulus-
HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology and induced bilateral central periodic discharges, cortical myoclonus
Clinical Semiology. Philadelphia, PA: Churchill Livingstone; and arousal responses in mild reversible coma. Clin Neurophysiol.
2000:580–584. 2008;119:2459–2464.
161. De Marco P, Negrin P. Parietal focal spikes evoked by contralat- 180. Behrman S, Wyke BD. Vestibulogenic seizures: a consideration of
eral tactile somatotopic stimulation in four non-epileptic sub- vertiginous seizures, with particular reference to convulsions
jects. Electroencephalogr Clin Neurophysiol. 1973;34:308–312. produced by stimulation of labyrinthine receptors. Brain.
162. De Marco P, Tassinari CA. Extreme somatosensory evoked 1958;81:529–541.
potential (ESEP): an EEG sign forecasting the possible occur- 181. Munter M, Goetze W, Krokowski G. Telemetrische EEG.
rence of seizures in children. Epilepsia. 1981;22:569–575. Untersuchungen ratatorischer Vestibulatisreizung. Dtsch Z
163. Deonna T. Reflex seizures with somatosensory precipitation: Nervenheilkd. 1964;186:137–148.
clinical and electroencephalographic patterns and differential 182. Karbowski K. Epileptic seizures induced by vestibular and audi-
diagnosis, with emphasis on reflex myoclonic epilepsy of infancy. tory stimuli. In: Beaumanoir A, Gastaut H, Naquet R, eds. Reflex
In: Zifkin BG, Andermann F, Beaumanoir A, et al., eds. Reflex Seizures and Reflex Epilepsies. Geneva: Editions Médicine &
Epilepsies and Reflex Seizures: Advances in Neurology. Vol 75. Hygiène; 1989:255–260.
Philadelphia, PA: Lippincott-Raven; 1998:193–206. 183. Acharya JN, Kotagal P. Activation of seizures by hot water. In:
164. Vignal J-P, Biraben A, Chauvel PY, et al. Reflex partial seizures of Lüders HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology
sensorimotor cortex (including cortical reflex myoclonus and and Clinical Semiology. New York: Churchill Livingstone;
startle epilepsy). In: Zifkin BG, Andermann F, Beaumanoir A, 2000:615–627.
et al., eds. Reflex Epilepsies and Reflex Seizures: Advances in 184. Mani KS, Mani AJ, Ramesh CK. Hot-water epilepsy—a peculiar
Neurology. Vol 75. Philadelphia, PA: Lippincott-Raven; 1998: type of reflex epilepsy: clinical and EEG features in 108 cases.
207–226. Trans Am Neurol Assoc. 1975;99:224–226.
165. Holmes GL, Blair S, Eisenberg E, et al. Tooth-brushing-induced 185. Lisovoski F, Prier S, Koskas P, et al. Hot-water epilepsy in an adult:
epilepsy. Epilepsia. 1982;23:657–661. ictal EEG, MRI and SPECT features. Seizure. 1992;1:203–206.
166. O’Brien TJ, Hogan RE, Sedal L, et al. Tooth-brushing epilepsy: a 186. Rémillard GM, Andermann F, Zifkin BG, et al. Eating epilepsy: a
report of a case with structural and functional imaging and elec- study of ten surgically treated patients suggests the presence of
trophysiology demonstrating a right frontal focus. Epilepsia. two separate syndromes. In: Beaumanoir A, Gastaut H, Naquet
1996;37:694–697. R, eds. Reflex Seizures and Reflex Epilepsies. Geneva: Editions
167. Langill L, Wong PKW. Tactile-evoked rolandic discharges: a Médecine & Hygiène; 1989:289–300.
benign finding? Epilepsia. 2003;44:221–227. 187. Rémillard GM, Zifkin BG, Andermann F. Seizures induced by
168. Servit Z. The application of reflex theory in the interpretation of eating. In: Zifkin BG, Andermann F, Beaumanoir A, et al., eds.
the clinical picture, genesis and treatment of epilepsy. Epilepsia. Reflex Epilepsies and Reflex Seizures: Advances in Neurology.
1962;3:209–228. Vol 75. Philadelphia, PA: Lippincott-Raven; 1998:227–240.
Chapter 12 ■ Activation Methods 237

188. Alajouanine T, Gastaut H. La syncinésie-sursaut et l’épilep- 212. Lishman WA, Symonds CP, Whitty CW, et al. Seizures induced by
sitesursaut è déclanchement sensorial ou sensitif inopiné. Les movement. Brain. 1962;85:93–108.
faits anatomocliniques (15 observations). Rev Neurol. 1955;93: 213. Radhakrishnan K, Silbert PL, Klass DW. Reading epilepsy: an
29–41. appraisal of 20 patients diagnosed at the Mayo Clinic, Rochester,
189. Rosenow F, Lüders HO. Startle-induced seizures. In: Lüders HO, Minnesota, between 1949 and 1989, and delineation of the
Noachtar S, eds. Epileptic Seizures: Pathophysiology and Clinical epileptic syndrome. Brain. 1995;118:75–89.
Semiology. Philadelphia, PA: Churchill Livingstone; 2000:585–592. 214. Wolf P. Activation of seizures by reading and praxis. In: Lüders
190. Aguglia U, Tinuper P, Gastaut H. Startle-induced epileptic HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology and
seizures. Epilepsia. 1984;25:712–720. Clinical Semiology. New York: Churchill Livingstone; 2000:
191. Manford MR, Fish DR, Shorvon SD. Startle provoked epileptic 609–614.
seizures: features in 19 patients. J Neurol Neurosurg Psychiatry. 215. Mayer T, Wolf P. Perioral reflex myoclonia in reading epilepsy
1996;61:151–156. and juvenile myoclonic epilepsy. In: Wolf P, Inoue Y, Jifkin B, eds.
192. Saenz-Lope E, Herranz FJ, Masdeu JC. Startle epilepsy: a clinical Reflex Epilepsies: Progress in Understanding. Esher, UK: John
study. Ann Neurol. 1984;16:78–81. Libbey Eurotext; 2004:57–70.
193. Bancaud J, Talairach J, Lamarche M, et al. Hypothèses neuro- 216. Goossens LAZ, Andermann F, Andermann E, et al. Reflex seizures
physiopathologiques sur l’épilepsie-sursault chez l’homme. Rev induced by calculation, card or board games, and spatial tasks: a
Neurol. 1975;131:559–571. review of 25 patients and delineation of the epileptic syndrome.
194. Loiseau P, Duché B. Seizures induced by movement. In: Neurology. 1990;40:1171–1176.
Beaumanoir A, Gastaut H, Naquet R, eds. Reflex Seizures and 217. Inoue Y, Seino M, Kubota H, et al. Epilepsy with praxis-induced
Reflex Epilepsies. Geneva: Editions Médicine & Hygiène; 1989: seizures. In: Wolf P, ed. Epileptic Seizures and Syndromes. London:
109–114. John Libbey; 1994:81–92.
195. Tibussek D, Wohlrab G, Boltshauser E, et al. Proven startle- 218. Inoue Y, Zifkin B. Praxis induction and thinking induction: one
provoked epileptic seizures in childhood: semiologic and electro- or two mechanisms? A controversy. In: Wolf P, Inoue Y, Jifkin B,
physiologic variability. Epilepsia. 2006;47:1050–1058. eds. Reflex Epilepsies: Progress in Understanding. Esher, UK: John
196. Hufnagel A, Elger CE, Durwen HF, et al. Activation of the epilep- Libbey Eurotext; 2004:41–55.
tic focus by transcranial magnetic stimulation of the human 219. Matsuoka H, Takahashi T, Sasaki M, et al. Neuropsychological
brain. Ann Neurol. 1990;27:49–60. EEG activation in patients with epilepsy. Brain. 2000;123:
197. Schuler P, Claus D, Stefan H. Hyperventilation and transcranial 318–330.
magnetic stimulation: two methods of activation of epileptiform 220. Takahashi Y, Shigematsu H, Kubota H, et al. Nonphotosensitive
EEG activity in comparison. J Clin Neurophysiol. 1993;10:111–115. video game-induced partial seizures. Epilepsia. 1995;36:837–841.
198. Tassinari CA, Cincotta M, Zaccara G, et al. Transcranial magnetic 221. Ishihara T, Yoshii N. Multivariate analytic study of EEG and men-
stimulation and epilepsy. Clin Neurophysiol. 2003;114:777–798. tal activity in juvenile delinquents. Electroencephalogr Clin
199. Green JB. Self-induced seizures. Arch Neurol. 1966;15:579–586. Neurophysiol. 1972;33:71–80.
200. Vignaendra V, Ghee LH, Lee LC, et al. Epileptic discharges trig- 222. Sasaki K, Tsujimoto T, Nishikawa S, et al. Frontal mental theta
gered by blinking and eye closure. Electroencephalogr Clin wave recorded simultaneously with magnetoencephalography
Neurophysiol. 1976;40:491–498. and electroencephalography. Neurosci Res. 1996;26:79–81.
201. Nadkarni MA, Postolache V, Gold A, et al. Central mid-temporal 223. Yamaguchi Y. Frontal midline theta activity. In: Yamaguchi N,
spikes triggered by blinking. Electroencephalogr Clin Neurophysiol. Fujisawa K, eds. Recent Advances in EEG and EMG Data
1994;90:36–39. Processing. Amsterdam: Elsevier; 1981:391–396.
202. Terzano MG, Parrino L, Manzoni GC, et al. Seizures triggered by 224. Guey J, Bureau M, Dravet C, et al. A study of the rhythm of petit
blinking when beginning to speak. Arch Neurol. 1983;40:103–106. mal absences in children in relation to prevailing situations. The
203. Silbert PL, Radhakrishnan K, Johnson J, et al. The significance of use of EEG telemetry during psychological examinations, school
the phi rhythm. Electroencephalogr Clin Neurophysiol. 1995;95: exercises and periods of inactivity. Epilepsia. 1969;10:441–451.
71–76. 225. Temkin NR, Davis GR. Stress as a risk factor for seizures among
204. Lewis JA. Eye closure as a motor trigger for seizures. Neurology. adults with epilepsy. Epilepsia. 1984;25:450–456.
1972;22:1145–1150. 226. Stevens JR. Emotional activation of the electroencephalogram in
205. Robinson LJ. Induction of seizures by closing of the eyes, or by patients with convulsive disorders. J Nerv Ment Dis. 1959;128:
ocular pressure, in a patient with epilepsy. J Nerv Ment Dis. 339–351.
1939;90:333–336. 227. Small JG, Stevens JR, Milstein V. Electroclinical correlates of
206. Tieber E. Anfallmuster bei Augenschluss. Bericht über drei Fälle emotional activation. J Nerv Ment Dis. 1964;138:146–155.
in einer Familie. Neuropaediatrie. 1972;3:305–312. 228. Sakel M. Neue Behandlugsmethode der Schizophrenie. Wien:
207. Shanzer S, Aprial R, Atkin A. Seizures induced by eye deviation. Moritz Perthes; 1935.
Arch Neurol. 1965;13:621–626. 229. Poire R. Hypoglycemic epilepsy: clinical, electrographic and bio-
208. Toma S, Mano T, Shiozawa Z. Seizure discharges induced by eye logical study during induced hypoglycemia in man. In: Gastaut
closure. Clin Neurol (Tokyo). 1975;15:340–346. H, Jasper HH, Bancaud J, et al., eds. The Physiopathogenesis of the
209. Vignaendra V, Lim CL. Epileptic discharges triggered by eye con- Epilepsies. Springfield, IL: Charles C. Thomas; 1969:75–110.
vergence. Neurology. 1978;28:589–591. 230. Green JB. The activation of electroencephalographic abnormali-
210. Arseni C, Stoica I, Serbanescu T. Electro-clinical investigations on ties by tolbutamide-induced hypoglycemia. Neurology. 1963;13:
the role of proprioceptive stimuli in the onset and arrest of con- 192–200.
vulsive epileptic paroxysms. Epilepsia. 1967;8:162–170. 231. Mosowich A. The role of oral hyperglycaemiants as activators in
211. Hudgins RL, Corbin KB. An uncommon seizure disorder: famil- epilepsy. In: Chorobski J, ed. Neurological Problems. Oxford:
ial paroxysmal choreoathetosis. Brain. 1966;89:199–204. Pergamon; 1967:117–130.
238 Part II ■ Normal EEG

232. Sperling MR. Hypoglycemic activation of focal abnormalities in 246. Cure C, Rasmussen T, Jasper HH. Activation of seizures and elec-
the EEG of patients considered for temporal lobectomy. troencephalographic disturbances in epileptic and in control sub-
Electroencephalogr Clin Neurophysiol. 1984;58:506–512. jects with “Metrazol”. Arch Neurol Psychiatry. 1948;59:691–717.
233. Bercel NA. Experience with a combination of scopolamine and 247. Jasper H, Courtois G. A practical method for uniform activation
alpha chloralose (S.A.C.) in activating normal EEG of epileptics. with intravenous Metrazol. Electroencephalogr Clin Neurophysiol.
Electroencephalogr Clin Neurophysiol. 1953;5:297–304. 1953;5:443–444.
234. Verdeaux G, Verdeaux J, Marty R. L’activation des élec- 248. Bancaud J. EEG activation by Metrazol and Megimide in the diag-
troencéphalogrammes par le chloralose. Electroencephalogr Clin nosis of epilepsy. In: Remond A, ed. Handbook of Electro-
Neurophysiol. 1954;6:19–28. encephalography and Clinical Neurophysiology. Amsterdam:
235. King G, Weeks SD. Pyribenzamine activation of the electroen- Elsevier; 1976:105–120.
cephalogram. Electroencephalogr Clin Neurophysiol. 1965;18: 249. Ajmone-Marsan C, Ralston BL. The Epileptic Seizure: Its
503–507. Functional Morphology and Diagnostic Significance. Springfield,
236. Vas CJ, Exley KA, Parsonage MJ. An appraisal of hyoscine as an IL: Charles C. Thomas; 1957.
EEG activation agent. Electroencephalogr Clin Neurophysiol. 1967; 250. Gastaut H. Combined photic and Metrazol activation of the
22:373–377. brain. Electroencephalogr Clin Neurophysiol. 1950;2:249–261.
237. Vas CJ, Exley KA, Parsonage MJ. Activation of the EEG by procy- 251. Shulman A, Shaw F, Cass N, et al. A new treatment of barbiturates
clidine hydrochloride in temporal lobe epilepsy. Epilepsia. intoxication. Br Med J. 1955;1:1238–1244.
1967;8:241–251. 252. Delay J, Schuller E, Drossopoulo G, et al. Un nouvel activant des
238. Vas CJ, Exley KA, Parsonage MJ. Methixene hydrochloride as an électroencéphalogrammes: l’imide de l’acide éthyl-méthyl glu-
EEG activating agent. Epilepsia. 1967;8:252–259. tarique (NP 13), ou Mégimide. Rev Neurol. 1956;94:315–318.
239. Cascino GD, So EL, Sharbrough FW, et al. Alfentanil-induced 253. Drossopoulo G, Gastaut H, Verdeaux G, et al. Comparison of
epileptiform activity in patients with partial epilepsy. J Clin EEG “activation” by pentamethylenetetrazol (Metrazol) and
Neurophysiol. 1993;10:520–525. bemegride (Megimide). Electroencephalogr Clin Neurophysiol.
240. Fagan K, Lee S. Prolonged confusion following convulsion due to 1956;8:710–711.
generalized nonconvulsive status epilepticus. Neurology. 1990;40: 254. Ajmone-Marsan C. Pentylenetetrazol: historical notes and com-
1689–1694. ments on its electroencephalographic activation properties. In:
241. Simon R. Physiologic consequences of status epilepticus. Lüders HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology
Epilepsia. 1985;26(suppl 1):S58–S66. and Clinical Semiology. New York: Churchill Livingstone;
242. von Meduna I. Versuch über die biologische Beeinflussung des 2000:563–569.
Ablaufs der Schizophrenie. I. Campher und Cardiazol Krämpfe. 255. Wieser HG, Bancaud J, Talairach J, et al. Comparative value of
Zbl Ges Neurol Psychiatr. 1935;152:235–262. spontaneous and chemically and electrically induced seizures in
243. Ziskind E, Sjaardema H, Bercel NA. Minimal electroencephalo- establishing the lateralization of temporal lobe seizures.
graphic response to Metrazol as a method for measuring the con- Epilepsia. 1979;20:47–59.
vulsive threshold for use in human beings. Science. 1946;104: 256. Fariello RG, Booker HE, Chun RWM, et al. Reenactment of the
462–463. triggering situation for the diagnosis of epilepsy. Neurology. 1983;
244. Kaufman IC, Marshall C, Walker AE. Metrazol activated elec- 33:878–884.
troencephalography. Res Publ Assoc Nerv Ment Dis. 1946;26: 257. Labate A, Ambrisio R, Gambardella A, et al. Usefulness of a
476–486. morning routine EEG recording in patients with juvenile
245. Ziskind E, Bercel NA. Preconvulsive paroxysmal electroen- myoclonic epilepsy. Epilepsy Res. 2007;77:17–21.
cephalographic changes after Metrazol injection. Res Publ Assoc
Res Nerv Ment Dis. 1946;26:487–501.
CHAPTER

Artifacts of Recording
BARBARA DWORETZKY, SUSAN HERMAN, AND WILLIAM O. TATUM IV 13
R ecording electric activity from the brain is subject
to extracerebral interference or “artifact” that is present
in essentially every electroencephalogram (EEG) (1).
Artifacts may be derived from a variety of sources. They can
waveform morphologies and electric fields and therefore can be
fairly easily identified, nonphysiologic artifacts show a wide
variety of morphologies that can either distort or obscure nor-
mal EEG activity. In the most severe cases, artifact may render
obscure the cortically generated signal to a variable degree and the recording uninterpretable. When artifacts result from
render it impossible to interpret or lead to misinterpretation of equipment malfunction, the EEG machine should be immedi-
the EEG as abnormal and possibly “epileptic” in origin. Failure ately tested to ensure there are no electric safety hazards. In the
to recognize such potentials as artifact is one of the leading rea- digital age of EEG recording, it is easier to know with certainty
sons for inappropriate use of antiepileptic medications, and a the generators of EEG contaminants by examining the patient
common reason for patient referral to an epilepsy center (2,3). relative to the recording, and subsequently successfully elimi-
The ideal EEG reflects purely cerebral activity and negligible nating the artifact that is identified (proof of principle).
extracerebral electric activity. It is performed by a qualified EEG Illustrations of artifacts are provided to enhance the principles
technologist who ideally creates an artifact-free recording in this chapter.
within the confines of a technically adequate recording on a
cooperative patient in a quiet and controlled environment. This GENERAL PRINCIPLES
ideal EEG is pragmatically unachievable but the principles
behind such a recording form the basis for recognizing the The recognition of EEG artifact is an acquired skill. It relies on
“contaminants” or “noise” that exist in the locations where EEG maintaining a low threshold to expect artifact in every epoch of
is performed. EEG interpretation is an acquired visual art, and the recording. For certain ubiquitous artifacts such as eye
the identification of common artifacts is often a matter of movements, noticing diminished amplitudes, slower frequen-
“automatic” recognition by the reader. cies, or changes in the direction of the dipoles, for example, may
To minimize confusion generated by artifact, approved suggest a state change (i.e., drowsiness, rapid eye movement
guidelines for electrode placement are essential (4,5). With the [REM] sleep). A marked asymmetry in eye movements may
ubiquitous use of digital EEG synchronized with video, newly suggest asymmetric electrode placement, enucleation (pros-
discovered rhythmic artifacts are being identified on a regular thetic eye), or a frontal skull defect (6).
basis in special care units (SCUs). Offline reformatting of the There are some general principles that can be applied when
EEG is not only possible, but also desirable, placing the respon- confronting possible EEG artifact. Artifact caused by loose elec-
sibility of recognition, identification, and elimination of EEG trodes can be distinguished from normal activity by its some-
artifact on the physician interpreting the EEG as well as the times characteristic shape, positive charge, restriction to a single
technologist. The burgeoning field of epilepsy and ICU moni- electrode, and apparent superimposition on normal cerebral
toring with video correlation has led to easy confirmation of
artifact sources and has produced many “art(ifact) collectors.”
Some artifacts are extremely common and desirable for routine Tabl e 1 3 . 1
recording purposes. For example, eye movements can help
define the normal stages of sleep. The foundation for recording EEG Artifact
principles in EEG may be subdivided into artifacts that are
physiologic and those that are nonphysiologic (Table 13.1). Physiologic Nonphysiologic
Physiologic artifacts include those generated by the patient’s Eye movement Electrode and lead
biologic activity with electric potentials that are generated near
the head (muscles, eyes, heart), or by patient movement. These Electromyographic Instrumental
may require camouflaging or ignoring the extracerebral source Glossokinetic Environmental
since elimination is not always possible. Nonphysiologic arti- Electrocardiographic Internal electric stimulators
facts are generated anywhere in the EEG recording system from
the electrode–scalp interface to the instrument itself or from Sweat
the environment, including from devices near or within the Patient movement
patient. While many biologic artifacts show characteristic

239
240 Part II ■ Normal EEG

activity. Because the artifact is usually limited to a single elec- reference electrode, ground, or preamplifier should be
trode, it will be seen only in channels containing that electrode checked for integrity.
and will not show a “field” to neighboring electrodes. The elimination of artifact from the EEG once it is recog-
Reformatting into various montages, particularly referential nized involves identifying the source. Troubleshooting artifact
montages, may help to identify a discharge as artifactual. involves checking the electrodes (including the ground and
Because some electrode artifacts may be difficult to recognize impedances), the apparatus, the patient (if still connected),
and may mimic abnormal patterns, any electrode showing high and the ambient activity. Artifact, most apparent in a loose or
impedance or frequent pops should be reapplied or replaced. In dry electrode, is easily eliminated by regelling or replacing the
some cases, high impedance or loose electrodes show severe electrode during the study. Eliminating the artifact by chang-
artifact with minor patient movements. These electrodes ing a lead or turning off an external device can confirm the
should be reapplied as well. Confirming that electrodes are source. The technologist should annotate the recording when
placed and labeled correctly in the proper headbox jack should activity generated by the patient or the external environment
be accomplished when expected waveforms appear in the is interfering with the quality of the study. EEGs that run con-
wrong channel. tinuously over days such as epilepsy monitoring, ambulatory
Neurosurgical procedures and certain developmental dis- EEG, or ICU monitoring studies may contain large amounts
orders disrupt normal cortical architecture and can yield pos- of artifact. This may not allow proper artifact elimination as
itive discharges or ones with unusual fields (7) and thus the technologist is not present for the majority of the record-
mimic electrode artifact. Skull asymmetries and edema ing. However, even when a technologist is on site, trou-
require documentation to properly reflect the differing ampli- bleshooting is not always simple or successful. It may be
tudes seen on the recording. Breach rhythms that emerge fol- necessary to camouflage rather than eliminate artifact.
lowing craniotomy may become “spikier” with Artifacts can mimic nearly any abnormal pattern (8), so it is
superimposition of myogenic potentials (Fig. 13.1). Rhythmic critical that every attempt to discover the origin of the artifact
or periodic activity on an EEG recording should be matched be undertaken.
with known rhythmic physiologic (i.e., cardiac, respirations)
or nonphysiologic activity (i.e., drips, pumps, machines) to ARTIFACTS OF PHYSIOLOGIC ORIGIN
uncover the source. If the waveforms appear in more than one
nonadjacent brain region at once (nonanatomic pattern), the Physiologic sources of artifacts, apparent on every EEG record-
activity is likely to be due to artifact. Additionally, if the wave- ing, are generated from the biologic properties of the patient
forms appear in all leads and are unusual in appearance, the and the many sources within the body that can act as electric

Figure 13.1 Myogenic potentials (arrows) superimposed on a breach rhythm in the left parasagittal chain can
appear “spikier.”
Chapter 13 ■ Artifacts of Recording 241

dipoles. The most common and important of these artifacts are two channels help identify potentials that are in phase with
produced by the eyes, the muscles of the head and face, and the cerebral, and out of phase with extracerebral, ocular sources
heart. Other important physiologic generators of artifact (9). Eye movement artifact is important for the determination
include the tongue or glossokinetic artifact, respiration, and of the normal waking background as eye opening blocks the
potentials generated in the skin by sweat glands. In addition, the alpha rhythm and eye closure produces alpha. The artifact is
patient’s own movements, especially if rhythmic, may be a produced by the Bell’s phenomenon as the eyeballs roll upward
source of confusion on an EEG. Placing an extra electrode on or with the closure of the eyes, producing an upward movement
near a moving body part or utilizing video allows the reader to of the positive dipole created by the cornea. Eye movements
confirm the nonbrain origin. can be quite rapid with fluttering eyelids occurring at rates up
to 13 Hz and appear abnormal (Fig. 13.3) especially if they are
Eye Movement Artifact asymmetric (Fig. 13.4)! They can also be rather slow in the
Eye movement artifacts are seen in virtually every awake and delta range mimicking abnormal patterns such as frontal inter-
conscious individual during routine EEG. Most eyeball artifact mittent rhythmic delta activity (FIRDA; Fig. 13.5). With hori-
is easy to recognize by its anterior location and its bilateral, zontal eye movements, the artifact is generated over the lateral
synchronized appearance. When the eyes move, the electric eye leads, F7 and F8 (Fig. 13.6). It can be quite striking as in the
dipole between the positively charged cornea and the nega- case of lateral gaze nystagmus, generally noted more from the
tively charged retina also moves, yielding a large potential side of the fast phase (10). The characteristic deflection that
depending on the direction and rate of the movement. In gen- includes a rapid rise and more gradual fall with the corrective
eral, it is symmetric as long as lead placement is measured well, movement is quite easy to recognize. The side of the positive
the patient is not missing an eyeball (Fig. 13.2), both retinas phase reversal of the rapid rise indicates the direction of the
are functional, and the frontal bones are intact (6). Eye move- fast component of the nystagmus. With leftward eye move-
ment monitors placed above and below the eye using one to ment, the F7 electrode has positive reversals at the same time

Figure 13.2 Lateral eye movements are noted asymmetrically only from the right due to an enucleated left eyeball
(prosthetic eye).
242 Part II ■ Normal EEG

Figure 13.3 Eye blinks can be so rapid that they appear abnormal. Note the bifrontal symmetric 9-Hz activity that
represents eye fluttering, elicited with eye closure (arrows).

Figure 13.4 Unilateral eye fluttering is noted in a patient with a detached retina on the right.
Chapter 13 ■ Artifacts of Recording 243

Figure 13.5 Slower appearing eye blinks (2.5 to 3 Hz) intermixed with EMG appear similar to 3-Hz spike-and-wave
activity. Eye leads show out-of-phase activity (arrow), confirming origin from eye movements and distinguishing
from epileptiform activity and frontal intermittent rhythmic delta activity.

Figure 13.6 Rapid lateral eye movements demonstrated on an awake patient (gray bar). Note the eye leads LUC
and RLC, showing the out-of-phase activity. The arrow points to a lateral rectus spike.
244 Part II ■ Normal EEG

Figure 13.7 Left-beating nystagmus is identified by the F7 electrode (longitudinal bipolar montage, top two chan-
nels of the third electrode chain) demonstrating positive reversals at the same time that the F8 electrode reveals neg-
ative reversals (gray bar). Most of the eye movements are preceded by lateral rectus spikes.

that the F8 electrode reveals negative reversals (Fig. 13.7). bursts of fast activity (Fig. 13.9). Technologists can ask the
Additionally, eye movements are crucial to correctly identify patient to open the mouth that relaxes the jaw and diminishes
the stages of sleep such as the slow roving subdelta in the lat- this artifact. Frontalis muscle contraction during periocular
eral eye leads representing drowsy eyeballs during stage Ia and movement may elicit sustained or individual motor unit
REM artifact also seen in F7 and F8 lateral eye leads during potentials that can appear like “railroad tracks” (Fig. 13.10).
REM sleep (Fig. 13.8). Lateral rectus muscle spikes representing motor unit potentials
recorded from the lateral orbit may simulate small interictal
Electroretinogram (ERG) discharges (i.e., F7/F8) (Fig. 13.11). During intermittent photic
During photic stimulation, light from the flash stimulus may stimulation, eye flutter artifact at frequencies of 6 Hz may
produce artifact in the frontopolar leads (Fp1/Fp2). This arti- mimic generalized spike-and-wave when coupled with myo-
fact can be mistaken for photic driving due to the entrainment genic potentials and create a “pseudo-photoparoxysmal
with the stimuli, but is not located occipitally. The source may response” (Fig. 13.12) when muscle spikes are time-locked to
be the retina (ERG) or from a nonphysiologic source such as a the flash frequency.
frontopolar electrode with high impedance creating a photo-
cell. Covering the eye with a towel will block the input to the Glossokinetic Artifact
retina (ERG) and this should not be confused with the photo- The tongue is a very large muscle centrally located inside the
electric effect (11). head. Similar to the eye, the tongue creates a bioelectric dipole
with the tip of the tongue negative relative to the root (11).
Muscle Artifact During oropharyngeal motion, a direct current (DC) potential
Muscle is another commonly observed artifact in the EEG. It is is produced that is often diffusely seen with frontal and tempo-
high-frequency activity noted to be very “spiky” but too fast to ral predominance (11). Cephalad–caudad motions may be
be an epileptic discharge. It is observed more while the patient produced by tongue movements involuntarily while speaking
is awake, and may obscure critical portions of the recording, or when swallowing (Fig. 13.13) producing delta frequency
for example, during hypermotor seizures. The frontalis and waves that may mimic FIRDA distinguishable by the
temporalis muscles are principal sites of myogenic artifact. superimposed muscle artifact. Similar artifact may be
Frontalis muscle becomes most involved with forced eye clo- produced when patients are asked to say “lilt” or “ta ta ta,”
sure, and photic stimulation (10). Temporalis muscles become identifying similar patterns as artifact more conclusively
active with jaw clenching, chewing, or bruxism appearing as (Fig. 13.14). Horizontal tongue movements (i.e., tongue in
Chapter 13 ■ Artifacts of Recording 245

Figure 13.8 This longitudinal bipolar montage demonstrates slow roving eye movements in the lateral eye leads
(F7 and F8) signifying drowsiness. Eye monitors are LUC (left upper canthus) and RLC (right lower canthus). The
alpha rhythm attenuates (vertical arrow and line), and slow lateral eye movements appear. There is also EKG arti-
fact (arrowheads) and lateral rectus spikes (horizontal arrow).

Figure 13.9 Bursts of myogenic artifact in an alternating pattern are consistent with bruxism.
246 Part II ■ Normal EEG

Figure 13.10 The left frontopolar electrode (Fp1) (arrows) demonstrates a motor unit firing that appears like “rail-
road tracks.”

Figure 13.11 Lateral rectus spikes (arrows) are noted bilaterally in the frontopolar leads. Lateral eye movements
show out-of-phase deflections at F7 and F8 (gray bar).
Chapter 13 ■ Artifacts of Recording 247

Figure 13.12 Photomyogenic artifact is noted in the frontal (not occipital) eye leads during intermittent photic
stimulation (stimulus artifact is not displayed). This should not be mistaken for photoparoxysmal response.

Figure 13.13 Diffuse anterior predominant delta during waking state (eye blinks, muscle) corresponds to drinking
(glossokinetic artifact).
248 Part II ■ Normal EEG

Figure 13.14 Bifrontal 2- to 4-Hz square-shaped waveforms representing lingual (glossokinetic) artifact. Note that
these are in phase in eye channels, so cannot represent eye movements.

cheek) can create unilateral artifact on the EEG that may be waveforms predominately in the temporal derivations of the left
confusing. Validation is possible through application of tongue hemisphere due to the vector created by the electric conduction of
movement monitors with electrodes placed above and below the heart. Opposite polarities of the R-wave from the EKG may be
the mouth over the cheek and chin (10). Using a bipolar mon- seen at the left (negative) and right (positive) ear electrodes. EKG
tage, positive phase reversals are evident with tongue move- can be readily identifiable as the source because of the regularity
ment in a similar fashion to those recorded during eye and time-locked waves synchronous with the EKG. Importantly,
movements. when an electrode is positioned over an artery, the blood pulsing
through the vessel moves it regularly to produce a periodic and
Electrocardiogram (EKG) Artifact smooth waveform time-locked to the cardiac rhythm appearing
While many artifacts “contaminate” the EEG, some (i.e., the EKG) approximately 200 msec after the QRS complex (Fig. 13.17)
are essential for interpreting physiologic functions that may occur (10–12). Pulse artifact can be eliminated by moving the electrode
during the recording session. The heart is another key source of off the artery.
EEG artifact and is variably present on recordings depending on
the montage used and the size of the patient. Referential montages Sweat
often accentuate EKG artifact (Fig. 13.15), especially using ipsilat- Scalp perspiration will also produce artifact by creating
eral ear reference with the larger intra-electrode distances. Linked unwanted electric connections between electrodes. Perspiration
ears montage can reduce the artifact (Fig. 13.16). Overweight artifact appears as very low–frequency ( 0.5 Hz) low-amplitude
patients with short stocky necks as well as babies may be predis- undulating waves. Changes in the DC electrode potential from
posed to EKG artifact because the dipole is situated closer to the perspiration may result in an unstable baseline (sweat sway)
recording electrodes and better able to transmit the current (10). and crossing of tracings in adjacent channels (Fig. 13.18).
This may be reduced by altering head position. Recording the Perspiration artifacts are usually seen in multiple adjacent
EKG during routine EEG is essential to enable recognition of the channels or over the entire scalp and poses a risk for bridging
cardiac–cerebral relationship. Channel I of the EKG is approxi- between electrodes (see also the section “Electrode and Input
mated with electrodes linking the left and right chest using bipo- Lead Artifacts”). They can be reduced somewhat by increasing
lar recording to identify the QRS. EKG interference may be noted the low-frequency filter, and by drying the scalp and reapplying
as a small spike at 1/sec that in isolation mimics an interictal spike electrodes, or by cooling the patient with a fan.
or in succession and on a low-amplitude record mimics periodic
lateralized or generalized periodic epileptiform discharges Patient Movements
(PLEDs or GPEDs). Bipolar montages may reveal apparent inter- Patient movements cause the leads or electrodes to move, and
ictal epileptiform discharges (IEDs) that are “focal” diphasic thus can provide a large source of “physiologic” artifact on
Chapter 13 ■ Artifacts of Recording 249

Figure 13.15 Bipolar recording shows prominent EKG artifact time-locked to the QRS complex, maximal in ampli-
tude over both temporal–occipital regions (arrows).

recordings. This is especially true for the awake and ambulatory display the time-locked behavior with EEG usually leads to
patient (13), but is also quite notable in those who are agitated accurate interpretation. Technical guidelines for recording in
and confused. Synchronous video has been inordinately helpful the EMU have been developed by the American Clinical
in distinguishing the varieties of patient movements when a Neurophysiology Society (15). For further discussion, see
technologist is not present for the recording. Regular appearing Chapter 37. Long-term monitoring provides a greater likeli-
patient movements such as tremor may manifest at different hood of deterioration of the patient–electrode interface with
frequencies and amplitudes such as the 6-Hz tremor of drying out of the electrolytic gel or nonadherence of the elec-
Parkinson disease (Fig. 13.19) or scratching near an electrode. trodes to promote artifact that may override interpretation dur-
During bipolar recording, extra adjacent electrodes placed on ing behavioral events. In the EMU, approximately 20% to 25%
the suspected limb are helpful to confirm tremor artifact but of patients with episodes will be manifesting psychogenic
they may be readily apparent in the EKG lead. Sudden or rapid nonepileptic attacks (PNEA) (16). These are described in more
patient movements such as shivering (Fig. 13.20) can mimic the detail in Chapter 33. The importance of recognizing behavioral
paroxysmal quality of an electrographic seizure, but usually do sources in addition to historic information is crucial to recog-
not show progression. Additional artifact that leads to regular nizing nonepileptic events especially if there is “pseudo-evolu-
movement of the head or body from large pulses through the tion” simulating an ictal recording (Fig. 13.22) (4).
aorta or ballistocardiographic artifact gives rise to delayed
pulses that can appear more generalized but still linked to the
EKG (14). During electrocerebral inactivity recordings at sensi- ARTIFACT OF NONPHYSIOLOGIC ORIGIN
tivities of 2 V/mm, ballistocardiographic artifact and pulse
artifact may be more prominent with small scalp arteries caus- Electrode and Input Lead Artifacts
ing repetitive movements of electrodes. Stabilizing the head Electrode artifacts are one of the most common types of
with towels under the neck often eliminates this artifact. nonphysiologic artifact. Any unusual waveform that is
Artifact due to respiration is also generated by the movement of restricted to a single electrode should be considered to be arti-
the body or the head (Fig. 13.21). This can be seen during factual (17,18). The scalp–electrode interface and its impor-
hyperventilation over posterior head regions. A technologist tance in accurate recording of cerebral electric activity are
can observe the breathing and annotate the recording to indi- described in Chapters 5, 6, and 8. Disruptions of this interface
cate where inhalation and exhalation occur (11). In the epilepsy result in several types of artifact. Poor electrode contact or
monitoring unit (EMU), the technologist is away from the “loose” electrodes are a frequent cause of artifact. The charac-
patient but continuous video and the ability to transmit and teristic electrode “pop” occurs when an electrode moves and the
250 Part II ■ Normal EEG

Figure 13.16 A: EKG artifact is accentuated when using an ipsilateral ear reference. B: EKG artifact is substantially
diminished by linking the ears.

electric double layer is disturbed, creating a DC potential simi- also help to determine whether the activity is artifactual.
lar to discharging a capacitor (19). These electrode movements Similar pops can be seen with drying of electrode gel or paste,
may be minute and not well seen by visual observation. Pops poor connection of the electrode to the electrode wire, broken
are sudden positive (or less commonly negative) discharges lead wires, or faulty connection between electrode pin and the
and usually show an initial high-voltage very steep deflection pin jack. If reapplication of the electrode does not fix the arti-
followed by an exponential decay caused by the amplifier’s low- fact, the electrode should be examined for corrosion at these
frequency filter. The shape of the “pop” often resembles that of contact points.
the calibration waveform, with “mirror images” of the discharge In some cases, poor or fluctuating contact between elec-
seen in bipolar channels with the electrode in common trode and scalp results in continuous low-amplitude rhythmic
(Fig. 13.23). Pops may be single or repetitive. When an elec- or semirhythmic slow activity, simulating focal slowing or
trode shows repeated pops, the impedance should be measured seizures (Fig. 13.24) (6). Again, this can be distinguished from
and the electrode reapplied. Recording from an additional elec- pathologic brain activity by restriction to a single electrode
trode placed close to the electrode with suspected artifact may contact. Similar artifacts can be caused by drying of electrolyte
Chapter 13 ■ Artifacts of Recording 251

Figure 13.17 Pulse artifact is noted at O1 time-locked to the EKG but occurring 250 msec after the QRS.

Figure 13.18 Subdelta activity is noted with crossing baselines consistent with “sweat sway.”
252 Part II ■ Normal EEG

Figure 13.19 Tremor artifact (6 Hz) picked up over the left temporal chain (and EKG lead) due to motor unit fir-
ing and rhythmic movement artifact.

Figure 13.20 Shiver and body jerks causing occipital “pops” appear as part of a nonepileptic psychogenic event.
Chapter 13 ■ Artifacts of Recording 253

Figure 13.21 Respiration artifact occurring every 3 seconds, subtle on the left side of the page, and enhanced on
the right side by increasing the gain. Artifact is caused by rocking head movements on posterior electrodes.

Figure 13.22 Patient using a pumice stone to sand the calluses of her left foot. Subtle back and forth motion of
the head/ body renders this artifact nearly indistinguishable from seizure without the help of video.
254 Part II ■ Normal EEG

Figure 13.23 Electrode pop is noted over the P4 electrode, most prominent in the area of skull breach, which
evolves into faster, almost ictal-appearing discharges.

Figure 13.24 Electrode pop imitating a very localized seizure. The Fp1 electrode is not well placed or has air-dried
paste. Rhythmic 5-Hz activity remains quite focal but then slows to 4 Hz almost “evolving.”
Chapter 13 ■ Artifacts of Recording 255

between electrode and scalp or by movement of the electrode fore becomes the sum of the activity from the two electrodes,
leads. Changing impedances from movement of the electrode leading to spuriously low-amplitude recordings. Sometimes
can cause rhythmic artifact (see also the section “Artifacts of activity in the channel containing the salt bridge can appear
Physiologic Origin”: respiration, hyperventilation, tremor, flat or even isoelectric. This type of artifact is more common
pulse artifact). Since EEG instruments use differential ampli- when using closely spaced electrodes, as in the International
fiers, each amplifier channel receives input from two elec- 10-10 modified system or when using the full 10-20 place-
trodes, and the impedances must match reasonably well. ment in neonates.
Guidelines for minimum standards suggest that individual Bimetal potentials can occur when dissimilar metals make
electrodes must have impedances of less than 5000 (5). contact, either at the electrode–wire interface or between the
Mismatched impedance between electrodes will cause the EEG electrode pin and jack box receptacle. Dental fillings with dis-
amplifiers to work improperly, causing distortion of the EEG similar metals may also cause spiky artifacts when the metals
signal in channels containing those electrodes (20). High touch during chewing and speaking.
impedance electrodes can cause falsely low or high amplitudes Finally, high impedance electrodes can cause photovoltaic or
and loss of low-frequency components of EEG. In addition, photoelectric artifact during intermittent photic stimulation.
channels with mismatched electrode impedances are more sus- Each flash causes a photochemical reaction in the electrode.
ceptible to environmental artifacts, particularly 60-Hz artifact This is seen as brief spike-like transients in the high impedance
(21). Digital EEG machines utilize a single machine or refer- electrodes simultaneously with the flash (Fig. 13.26).
ence electrode, and all other montages are reformatted based Photoelectric artifact can be confirmed by its ability to be
on this referential recording. The impedance of the machine blocked by covering the involved electrode.
reference electrode is therefore of critical importance, and Proper electrode application and maintenance is the best
problems can result in technically limited studies that interfere way to reduce electrode artifact. Electrodes should be cleaned
with interpretation. well and inspected regularly to look for signs of corrosion
Ground lead recordings occur when a very high imped- between the electrode and conducting wire, damaged insula-
ance or disconnected electrode exceeds the input impedance tion, or broken lead wires. Faulty electrodes should be dis-
of the recording system (22). In this case, the ground elec- carded. If silver chloride electrodes are used, they should be
trode is substituted for the poor contact electrode, sometimes rechlorided on a regular schedule. Good electrode contact
leading to bizarre waveforms (Fig. 13.25). Salt bridges occur should be ensured by adequate preparation of the skin to
when electrode paste or gel is smeared between two electrode remove surface oils. Electrode impedance should be measured
locations. EEG activity recorded from each electrode there- and verified to be less than 5000 (5). Extra electrode paste

Figure 13.25 Ground lead recording. Very high impedance electrode at O1 causes ground electrode (at Fpz) to
become input 2 of channels including O1. Therefore, T5-O1 effectively becomes T5-Fpz, resulting in “upside down”
eye blinks (arrows) in this channel.
256 Part II ■ Normal EEG

Figure 13.26 The T4 electrode acts as a “photocell” detecting the 9-Hz photic stimulus artifact. There is also
increased 60-Hz artifact in this high impedance channel when the photic stimulator is active.

between electrodes should be removed. Covering the electrode In analog EEG machines, each amplifier had multiple
with collodion or EC2 paste prevents small movements of the switchable components that could malfunction, causing
electrode and slows drying of the electrode paste. Electrode excessive noise, abnormal transients, or loss of signal (22).
wires should be bunched together to prevent movement and Electrode selection switches were used to determine which
excessive tension on electrodes. Occasionally, the leads are two electrodes were input to each amplifier. Switch malfunc-
placed into the incorrect jack inside the headbox and can yield tions and incorrect electrode selection were common sources
unusual fields (Fig. 13.27). of artifact. Similar errors could occur in sensitivity, low-
frequency filter, and high-frequency filter selectors. In digital
Instrumental Artifacts EEG machines, amplifiers are hardwired, with each channel
Malfunction of almost any part of the EEG machine can made up of a hardwired input 1 and machine reference as
cause artifact. Improved design and construction of EEG input 2, reducing the likelihood of switch problems. EEG is
machines, as well as the nearly universal use of digital tech- acquired with filters wide open and digital filters are applied
nology, has reduced the likelihood of equipment malfunc- without altering the original signal. Software errors in sensi-
tion, but this still occurs frequently enough that tivity, filters, or montages, however, can still lead to spurious
electroencephalographers (EEGer) and EEG technologists displays.
must have a thorough understanding of the most common Failure of components common to all channels often causes
technical artifacts. lack of power or 60-Hz artifact. Excessive 60-Hz artifact can be
Modern amplifiers are small, low-power, single-chip multi- caused by a faulty or absent ground connection to the patient,
channel devices with solid-state integrated circuits. Amplifier or defects in the power supply or other parts of the instrument
“noise” is caused by thermal agitation of electrons in the (Fig. 13.28A). Portable equipment is more susceptible to
amplifier circuits. Noise contains components at all frequen- mechanical wear and tear, such as vibration or jolting of
cies and therefore increases with the bandwidth of the ampli- machines. Loose wiring or loosening of circuit board connec-
fier. Noise levels should be less than 2 V measured peak to tions is a common source of artifact, causing loss of signal or
peak (5). This noise is typically only problematic when intermittent failures.
recording at sensitivities of 1 to 2 V/mm, such as used for Digital artifacts include aliasing (inadequate sampling rate),
electrocerebral inactivity recordings (22). A malfunctioning errors in analog to digital conversion, skew errors, multiplexing
amplifier component (capacitor, resistor, transistor) can pro- artifacts, and blocking (23). These are discussed in detail in
duce larger amplitude noise, which is added to and distorts Chapter 7. Aliasing is caused by sampling at a rate less
cerebral activity. than twice the highest frequency present in the signal, and can
Chapter 13 ■ Artifacts of Recording 257

Figure 13.27 A: Classic left temporal discharges appear as bizarre bilateral sharp waves with multiple phase rever-
sals and temporal regions show bizarre alpha and theta activity. This is due to lead T3 connected into T4 jack in the
jack box (and vice versa). B: After correction of lead error.

introduce spurious slower frequencies into the EEG. some environmental artifacts, such as movement of people and
Multiplexing artifacts occur when the samples from multiple objects around the patient (25).
amplifiers are switched. Blocking can occur when the amplifier
is saturated, producing a squared-off waveform at the limits of Mains Power Supply (Alternating Current Artifact)
amplifier resolution. The most common source of electric interference is the alter-
Finally, incorrect equipment display can cause artifacts nating mains power supply, at 60 Hz in the United States and
(11). In analog machines, damage to or misalignment of pen 50 Hz in Europe. Electric interference from mains power sup-
recorders can result in apparent asynchrony of simultaneous plies can be either electrostatic or electromagnetic. In a prop-
discharges, while displacement from zero baseline leads to dis- erly designed modern neurophysiology lab, power lines are
torted morphology of waveforms. Incorrect pressure at the pen adequately grounded and severe electric interference is rarely
tip can also distort waveforms: if pressure of the pen on paper problematic. If necessary, interference can be reduced by shield-
is too great (overdamping), the pen drags and acts as a high- ing power cables (with outer metal braid connected to ground),
frequency filter, while underdamping results in pen overshoot. by using a shielded room for recordings, or by moving the
Malfunction of paper drive mechanisms can lead to variations patient farther from the source of interference in the room (22).
in chart speed, causing waveforms to appear longer or shorter Environmental artifacts are more common in areas outside the
than actual brain activity. In digital displays, using monitors EEG lab (“hostile environments”), particularly intensive care
with inadequate display resolution can cause aliasing, intro- units and operating rooms, where other medical equipment can
ducing spurious slow frequencies into the display that resolve interfere with EEG recordings.
when a screen resolution is adequate to display all recorded Sixty-hertz alternating current can cause interference via
samples (24). electrostatic effects. Power lines will couple to other nearby
conductors (including the patient, technologist, electrode
Environmental Artifacts wires, metal bed frames, etc.) and induce a small alternating
A wide variety of artifacts arise from objects and electric devices potential of opposite polarity to the mains potential (22). While
around the patient. The vast array of morphologies and origins small, this potential can be very large in comparison to the volt-
can make identification of these artifacts difficult. Typically, age of EEG activity. Connecting the patient to ground (earth)
artifactual waveforms from external devices show very different will reduce this potential. Artifact can also be reduced keeping
morphologies from normal cerebral activity, and may often be all electrode wires and cables short, bundling electrode wires as
identified as artifactual even if the exact source cannot be a group, and unplugging unnecessary equipment. A notch filter
found. Simultaneous video recording may also help to identify can be used to remove excessive 60-Hz artifact; however, it will
258 Part II ■ Normal EEG

Figure 13.28 A: Sixty-hertz artifact (electrostatic artifact) obscuring most of the record because of the lack of a ground
electrode. B: More typical appearance of 60-Hz artifact in a high impedance O1 electrode. C: Same segment of EEG as
B with 60-Hz notch filter on.

remove signals of interest around 60 Hz and so should not be current. Common hospital and EEG lab sources of interference
used routinely (Fig. 13.28B and C). Other electric equipment include photic stimulators, portable air pumps, monitoring
near the patient can cause electromagnetic interference from devices, lights and lamps, fans, air conditioning, pneumatic
current flowing through cables or from transformers and boots, heating and cooling blankets, electric beds, intravenous
motors. The intensity of the magnetic flux is proportional to infusion pumps, feeding delivery systems, elevators, computers,
Chapter 13 ■ Artifacts of Recording 259

Figure 13.29 Twenty-hertz artifact (a harmonic of 60 Hz) caused by a cooling blanket obscures nearly all of
the EEG.

telephones, diathermy, and x-ray equipment. Several of these coupling or transfer of energy between adjacent cables. This is
are discussed in more detail below. The electric field generated usually readily identified as artifact because of widespread
by the other equipment is inductively coupled to the electrode distribution and unusual electric fields. Rhythmic artifacts
wires, usually producing an out-of-phase harmonic of 60-Hz can be induced by movement of electrode wires during pat-
activity (Fig. 13.29). The harmonics will not be eliminated by ting (burping or soothing infants) or chest percussion therapy
the notch filter. Electromagnetic artifact can be reduced by (i) (Fig. 13.30) (28). Similar rhythmic or repetitive artifact may
ensuring that all equipment has a shielded metal case connected be produced by electrode wire or patient movement from ven-
to ground, (ii) plugging all equipment into the same connec- tilators, percussive anti-bedsore beds (Fig. 13.31), intra-
tion point, and (iii) ensuring that power cables do not run in arterial balloon pumps, and oscillatory ventilators. The shape
parallel and are remote from electrode wires and cables. of the artifact depends on the frequency of activation of the
Sixty-hertz artifact is present in small amounts in all EEG device, and is usually a slow wave or a mixture of frequencies
recordings, but is usually of low amplitude because amplifier superimposed on a slow wave, and is typically stereotyped or
common mode rejection removes most of the artifact. When monomorphic. Percussive beds or oscillatory ventilators cause
there are high or mismatched impedance electrodes, the ability artifact in the theta or alpha frequency range, usually best seen
of the amplifier to cancel out noise is reduced, and 60-Hz arti- in posterior head electrodes or electrodes in contact with the
fact will appear in channels including the high impedance elec- bed. An extra lead can be connected to the ventilator tubing to
trodes (26,27). This artifact is often a handy “detector” for high reveal the source.
impedance electrodes during EEG recordings. These electrodes Movement of people or objects around the patient may
should be reapplied as soon as possible. If 60-Hz artifact is seen induce electrostatic artifacts, even if the wires themselves do not
in all channels, this usually indicates a problem with the move. Examples can be seen with movements of nurses and EEG
ground electrode or the ground of the EEG machine, and technologists around the bed, movement of clothing or bedding
should be investigated promptly to ensure there is no electric (especially synthetic fabrics), swaying of tubing near the patient,
safety hazard (6). or movement of water in tubes near the electrode wires or input
cable (22). These waveforms typically are high voltage, with
Electrostatic Artifacts complex morphologies and nonphysiologic fields. A common
Electrostatic forces may induce currents in electrode wires or source of electrostatic artifact is from movement of water in res-
cables. Movement of the electrode wires themselves will cause piratory tubing in intubated patients. Water movement results
focal or widespread slow waves at the frequency of the in high-voltage slow waves with superimposed fast components
moving wires. Using short electrode wires, bundling the wires (Fig. 13.32), and usually occurs with a widespread field over the
in plastic tubing, or wrapping them with gauze will decrease frontal regions or the side of the head nearest to the respiratory
this type of artifact. Movement of the input cable between tubing (29). Similarly, electrostatic charges on drops of fluid in
electrode jack box and the amplifier can also cause capacitive intravenous drips may cause spike-like potentials coinciding
260 Part II ■ Normal EEG

Figure 13.30 Rhythmic appearing waves are noted over the right side from patting the chest while the right side
of the head is on the bed.

with the drops of the infusion (Fig. 13.33) (30). Piezoelectricity may also be produced by continuous veno-venous hemofiltra-
is an electric charge generated by the mechanical deformation of tion and dialysis machines (32,33). These artifacts arise from a
polymeric materials. Artifact from electric infusers may be combination of static and piezoelectric currents in pumps rotat-
caused by this static electricity generation between polyvinyl ing between 50 and 600 rpm, causing currents to flow into the
chloride tubing and the pump roller head (31). Unusual artifacts patient (and then to electrodes) via fluid in the tubing.

Figure 13.31 Rhythmic artifact generated from an anti-bedsore percussion bed.


Chapter 13 ■ Artifacts of Recording 261

Figure 13.32 Ventilator artifact noted periodically with superimposed faster waves from water vibrating inside the
tubing.

Figure 13.33 Electrostatic charges generate spike-like artifact from intravenous drips (arrows).
262 Part II ■ Normal EEG

Figure 13.34 Rhythmic activity


over the right temporal region looks
like a partial seizure until the dialy-
sis machine is turned off demon-
strating machine artifact.

The technologist should try to minimize movements bizarre intermittent artifact. Microwave ovens near the EEG
around the patient and note any changes in the EEG caused by lab may produce radio frequency interference. Televisions and
such movements. In addition, the technologist should include computer screens can cause high-frequency interference at the
notations about equipment connected to the patient, with an screen refresh rate when they are in close proximity to
attempt to correlate unusual EEG transients with equipment the patient. In the operating room, the electrocautery devices
function (e.g., respirator function or IV drops). Briefly turn- generate high-voltage high-frequency signals that travel
ing off a machine during the EEG can confirm the origin of through the body to the recording electrodes. The EEG
the rhythmic artifact such as from a dialysis machine machine should be turned off during use of coagulation.
(Fig. 13.34). Electrodes or transducers near the presumed site However, high-frequency cautery in adjacent rooms may also
of artifact generation may aid in identification of artifact produce interference (36).
sources. Any equipment with a motor may produce artifact due to
the switching magnetic field within the motor. This activity can
Electromagnetic Artifacts be constant or intermittent. Infusion motor artifact is generated
High-frequency interference from nearby television and radio by automatic programmable intravenous infusion pumps (30).
stations is rare but difficult to eliminate. Artifact can be seen in This typically is characterized by very brief spiky transients,
all channels. Hospital paging systems and walkie-talkies using which are stereotyped and repeat at regular intervals (37). Slow
radio frequency carriers may also cause artifact. Noncellular components may also be present.
telephones may cause ringing artifacts, related to the voltages Line isolation monitors, commonly present in the intensive
that develop in the telephone lines as the telephone rings care unit and operating room, scan electric outlets for leakage
(Fig. 13.35) (34). Portable and cell phones can produce artifact currents. They generate low-voltage artifact at a particular fre-
even when not being used (35). The effect of cell phones quency, which may be noticeable at increased sensitivity record-
depends on distance, operating frequency, and communication ings in comatose or anesthetized patients (38).
technology. Cell phones may cause repetitive sharp and slow-
wave complexes with a nonphysiologic distribution. These can Electric Stimulators
be widespread but may be of higher voltage in electrodes near Pacemakers produce very spiky, usually high-voltage artifact
the cell phone. Many of these are eliminated by the EEG (Fig. 13.36). These spikes are best seen in the EKG channel, but
machine’s antialiasing high-frequency filter but may cause may occasionally show a field extending to EEG electrodes.
Chapter 13 ■ Artifacts of Recording 263

Figure 13.35 Two-second burst of 30 cycle/ sec telephone artifact (electromagnetic artifact, vertical arrow) dif-
fusely, but highest amplitude in high impedance electrode Fp2. This leads to eye movements and EMG as the phone
is answered, obscuring a second “ring artifact.”

High-frequency artifacts can occur with deep brain stimulators, significantly distort the processed data (40). Therefore, the raw
the vagus nerve stimulator, spinal cord stimulators, and abdom- EEG should be available for comparison whenever quantitative
inal electrostimulators. Again, these are usually best seen in the results are analyzed, and careful attention should be paid to
EKG channel (38). The vagus nerve stimulator produces a scalp quality of data acquisition (41). Several commonly used meth-
negative evoked potential with very high amplitude ods to remove artifacts prior to digital processing are described
(Fig. 13.37). This potential is generated in the neck, and can be in the following sections.
recorded with electrodes over the stimulating electrodes in
the neck and sometimes in the EKG (39). Use of Band Pass Filters
If the analysis is restricted to certain frequency bands, an auto-
mated algorithm can be designed to only analyze activity in this
ARTIFACT DETECTION AND REJECTION
frequency band. For example, a 1- to 20-Hz band pass may be
Artifacts are usually easily recognized by experienced EEGer. The used to remove muscle artifact (42). This method is not very
processes of visual analysis, remontaging, and digital filtering useful for analysis of the entire bandwidth of EEG, as artifacts
allow identification of most physiologic and nonphysiologic arti- can occur at any frequency. Even for very narrow frequency
facts. Reviewing an epoch of EEG in a different montage may bands, there may be significant artifact remaining after band
allow the EEGer to determine that a particular waveform does not pass filtering. The process of filtering may significantly alter the
have a physiologic field, and thus be more certain of its artifactual appearance of EEG and make subsequent identification of arti-
nature. Digital filters can be applied and removed multiple times, facts more difficult.
and can significantly improve interpretation of EEG contami-
nated by artifacts by allowing specific frequencies to be removed Manual Rejection of Artifact Segments
from the digital display. These filters may distort normal or abnor- In this case, a technologist or EEGer visually reviews the entire
mal EEG activity, such as epileptiform discharges, so the ability to EEG recording and marks segments with artifacts. This is a
remove the filters and view the “raw” EEG is essential. reliable method, and may detect some artifact that would be
Processing of EEG data to remove artifacts before digital missed by automated techniques. It is time-consuming, how-
analysis (e.g., automatic spike or seizure detection, compressed ever, and reader fatigue may become problematic for long or
spectral analysis) is more difficult. Unrecognized artifacts can multichannel recordings. Subtle or brief artifacts may not be
264 Part II ■ Normal EEG

Figure 13.36 Pacemaker artifact best noted in the EKG lead but seen diffusely on EEG leads.

Figure 13.37 VNS artifact.


Chapter 13 ■ Artifacts of Recording 265

identified, and different readers may have different thresholds Patient manipulation through suctioning, dressing changes,
for rejection. This method is only possible for offline (not and movement during routine nursing activities or ICU proce-
real-time) digital analysis. Once the artifact segments are dures (i.e., central lines, cardiopulmonary rescucitation (CPR))
identified, they are completely eliminated from the analysis, produces copious artifacts and it can be helpful to train nursing
including any intervening normal EEG that may have been of staff to annotate the record for events that occur at the patient’s
interest. bedside. However, the EEG technologist and EEGer are both
crucial to identifying and recognizing sources as well as serving
Automatic Rejection of Artifact Segments to “troubleshoot” and eliminate artifact that may lead to limited
This technique rejects short segments of EEG if the segment or false interpretations. The American Board of Technologists
exceeds predefined thresholds. These thresholds can be simple in EEG and evoked potentials has established baseline stan-
analysis of the EEG channels themselves such as amplitude, dards for technological recording of EEG, and the American
numbers of zero crossings, or 60-Hz artifact. If a segment Board of Clinical Neurophysiology holds similar standards for
shows very high amplitude, it is eliminated. Some techniques EEG interpretation to help guide proper interpretation of the
use other special electrodes to identify artifact signals, such as EEG in the presence of artifact. Technologists and EEGer
electrooculogram, EMG, EKG, or accelerometers (19). If the remain challenged to recognize the source of each and every
signal in these channels exceeds a threshold, the segment of artifact that might jeopardize interpretation of the recording
EEG will be rejected. This technique can be used online or and therefore conservative interpretation is warranted when-
offline. These automatic rejection techniques will not identify ever doubt exists. Furthermore, even when recognition and
all possible artifacts, especially for ambulatory patients or in identification of the source of artifact is accomplished, elimina-
electrically hostile environments. Again, the entire EEG seg- tion is not always possible. Digital EEG has ushered in a new era
ment is rejected if the threshold is exceeded, so some useful for clinical neurophysiologists. Yet, principles of noncerebral
EEG may be eliminated. waveforms, the electric properties, electric fields, and recording
techniques still require an orderly approach for the successful
Automated Subtraction of Artifact visual analysis of artifact on the EEG. The systematic approach
of recognition, source identification, and elimination of artifact
These methods aim to identify artifact and to remove only the is an important process to reduce the chance for misinterpreta-
artifact from the recording, leaving a “clean” EEG for digital tion of the EEG and limit the potential for adverse clinical con-
analysis (19). Simultaneously recorded EOG or EKG can sequences.
be subtracted from an EEG channel using a computer. The
algorithm must be adapted for each individual patient. These
techniques may result in distortion of the EEG signal, as REFERENCES
the EOG also contains some normal brain signals that will
1. Aurlien H, Gjerde IO, Aarseth JH, et al. EEG background activity
be subtracted. described by a large computerized database. Clin Neurophysiol.
Source decomposition techniques aim to decompose EEG 2004;115(3):665–673.
signals into individual components that represent EEG and 2. Krauss GL, Abdallah A, Lesser R, et al. Clinical and EEG features of
others that represent artifact. Once the artifact component is patients with EEG wicket rhythms misdiagnosed with epilepsy.
identified, it is removed, and the remaining signal is recom- Neurology. 2005;64(11):1879–1883.
posed. Examples of these methods include spatial filtering (43), 3. Benbadis SR, Tatum WO. Overinterpretation of EEGs and misdi-
principal component analysis, and independent component agnosis of epilepsy. J Clin Neurophysiol. 2003;20(1):42–44.
analysis (44). Wavelet-based and neural network algorithms 4. Tatum WO. Normal EEG. In: Tatum WO, Husain A, Benbadis SR,
(45) allow adaptation of this method to a wider variety or arti- et al., eds. Handbook of EEG Interpretation. New York, NY: Demos;
fact types. These techniques are discussed in full in Chapters 56 2008:1–50.
5. Society ACN. Guideline one: minimum technical requirements for
and 58.
performing clinical electroencephalography [online]. J Clin
Neurophysiol. 1994;11(1):2–5.
CONCLUDING STATEMENTS 6. Fisch BJ. Fisch and Spehlmann’s EEG Primer: Basic Principles of
Digital and Analog EEG. 3rd ed. Amsterdam: Elsevier; 1999:631.
Numerous artifacts are encountered during routine or special 7. Alarcon G, Binnie CD, Garcia Seoane JJ, et al. Mechanisms
EEG recordings. While some artifacts are critical for interpreta- involved in the propagation of interictal epileptiform discharges in
tion of the EEG (i.e., sleep staging), many are not useful and partial epilepsy. Electroencephalogr Clin Neurophysiol Suppl.
1999;50:259–278.
serve to “contaminate” the recording and interfere with the
8. Klass DW, Westmoreland BF. Electroencephalography: general prin-
interpretation. Technological advances for artifact identifica- ciples and adult electroencephalograms. In: Daube J, ed. Clinical
tion and rejection have improved the ability to eliminate the Neurophysiology. Philadelphia, PA: F.A. Davis; 1996:73–103.
vast artifact present in longer term monitoring of EEG such as 9. Matsuo F, Shaner EE. EEG artifact due to a metallic prosthetic eye.
in the ICU and EMU (46). These special care neurology units Am J EEG Technol. 1975;15:75–78.
may house patients with skull asymmetries from craniotomies 10. Klem GH. Artifacts. In: Ebersole JS, Pedley TA, eds. Current
and scalp edema requiring documentation to properly reflect Practice of Clinical Electroencephalography. Philadelphia, PA:
markedly asymmetric amplitudes on the recording (Fig. 13.14). Lippincott Williams & Wilkins; 2003:271–287.
266 Part II ■ Normal EEG

11. Tyner FS, Knott JR, Mayer W Jr. Fundamentals of EEG Technology: 29. MacGillivray BB. Section IV: artefacts, faults, and fault-finding. In:
Basic Concepts and Methods. New York, NY: Raven Press; Remond A, ed. Handbook of Electroencephalography and Clinical
1983:280–311. Neurophysiology. Amsterdam: Elsevier; 1974:88–102.
12. Brittenham Dorothea M. Artifacts. In: Daly DD, Pedley TA, eds. 30. Lininger AW, Volow MR, Gianturco DT. Intravenous infusion
Current Practice of Clinical Electroencephalography. New York, NY: motor artifact. Am J EEG Technol. 1981;21(4):167–173.
Raven Press; 1990:85–105. 31. Khambatta HJ, Stone JG, Wald A, et al. Electrocardiographic artifacts
13. Ebersole JS, Bridgers SL, Silva CG. Differentiation of epileptiform during cardiopulmonary bypass. Anesth Analg. 1990;71(1):88–91.
abnormalities from normal transients and artifacts on ambulatory 32. McGrath B, Columb M. Renal replacement therapy causing ECG
cassette EEG. Am J EEG Technol. 1983;23:113–125. artefact mimicking atrial flutter. Br J Intensive Care. 2004; 14:49–52.
14. Stern JM, Engel Jr J. Artifacts. In: Stern JM, Engel J Jr, eds. Atlas of 33. Young B, Osvath L, Jones D, et al. A novel EEG artifact in the inten-
EEG Patterns. Philadelphia, PA: Lippincott Williams & Wilkins; sive care unit. J Clin Neurophysiol. 2002;19(5):484–486.
2005:55–86. 34. Saunders MG. Artifacts: activity of noncerebral origin in the EEG.
15. Guideline twelve: guidelines for long-term monitoring for In: Klass DW, Daly DD, eds. Current Practice of Clinical
epilepsy. J Clin Neurophysiol. 2008;25(3):170–180. Electroencephalography. New York, NY: Raven Press; 1979:37–68.
16. Benbadis S, Agrawal V, Tatum W. How many patients with psy- 35. Sethi PK, Sethi NK, Torgovnick J. Mobile phone artifact. Clin
chogenic nonepileptic seizures also have epilepsy? Neurology. Neurophysiol. 2006;117(8):1876–1878.
2001;57(5):915–917. 36. Nuwer M. Basic electrophysiology: evoked potentials and signal
17. Geddes LA. Bioelectrodes, part I. Am J EEG Technol. 1973;13: processing. In: Nuwer M, ed. Evoked Potential Monitoring in the
195–203. Operating Room. New York, NY: Raven Press; 1986:5–49.
18. Geddes LA. Bioelectrodes, part III. Am J EEG Technol. 1973;15: 37. Egol AB, Guntupalli KK. Intravenous infusion device artifact in
99–106. the EEG—confusion in the diagnosis of electrocerebral silence.
19. Barlow J. Artifact processing (rejection and minimization) in EEG Intensive Care Med. 1983;9(1):29–32.
data processing. In: Lopes da Silva F, van Leeuwen W, Remond A, 38. Patel SI, Souter MJ. Equipment-related electrocardiographic arti-
eds. Clinical Applications of Computer Analysis of EEG and Other facts: causes, characteristics, consequences, and correction.
Neurophysiological Signals. Handbook of EEG. Amsterdam: Anesthesiology. 2008;108(1):138–148.
Elsevier; 1986:15–64. 39. Hammond EJ, Uthman BM, Reid SA, et al. Vagus nerve stimula-
20. Gordon M. Artifacts created by imbalanced electrode impedance. tion in humans: neurophysiological studies and electrophysiolog-
Am J EEG Technol. 1980;20:149–160. ical monitoring. Epilepsia. 1990;31(suppl 2):S51–S59.
21. Ferree TC, Luu P, Russell GS, et al. Scalp electrode impedance, 40. Nuwer MR. Quantitative EEG: I. Techniques and problems of fre-
infection risk, and EEG data quality. Clin Neurophysiol. 2001; quency analysis and topographic mapping. J Clin Neurophysiol.
112(3):536–544. 1988;5(1):1–43.
22. Cooper R, Wosselton J, Shaw JC. EEG Technology. 3rd ed. London: 41. Anderer P, Roberts S, Schlogl A, et al. Artifact processing in com-
Butterworth-Heinemann; 1990. puterized analysis of sleep EEG—a review. Neuropsychobiology.
23. Blum D. Computer-based electroencephalography: technical 1999;40(3):150–157.
basics, basis for new applications, and potential pitfalls. 42. O’Donnell RD, Berkhout J, Adey WR. Contamination of scalp
Electroencephalogr Clin Neurophysiol. 1998;106(2):118–126. EEG spectrum during contraction of cranio-facial muscles.
24. Epstein CM. Aliasing in the visual EEG: a potential pitfall of video Electroencephalogr Clin Neurophysiol. 1974;37(2):145–151.
display technology. Clin Neurophysiol. 2003;114(10):1974–1976. 43. Ille N, Berg P, Scherg M. Artifact correction of the ongoing EEG
25. Hirsch LJ, Kull L. Continuous EEG monitoring in the intensive using spatial filters based on artifact and brain signal topogra-
care unit. Am J Electroneurodiagn Technol. 2004;44(3):137–158. phies. J Clin Neurophysiol. 2002;19(2):113–124.
26. Ford RG. A practical guide to EEG recording technique. Am J EEG 44. Astolfi L, Cincotti F, Mattia D, et al. Removal of ocular artifacts for
Technol. 1981;21:79–101. high resolution EEG studies: a simulation study. Conf Proc IEEE
27. Seaba PJ. Differential amplifiers and their limitations. Am J EEG Eng Med Biol Soc. 2006;1:976–979.
Technol. 1984;24(1):11–23. 45. Robert C, Gaudy JF, Limoge A. Electroencephalogram processing
28. Sethi NK, Torgovnick J, Sethi PK. Rhythmic artifact of physiother- using neural networks. Clin Neurophysiol. 2002;113(5):694–701.
apy in intensive care unit EEG recordings. J Clin Neurophysiol. 46. Young GB, Campbell VC. EEG monitoring in the intensive care
2008;25(1):62. unit: pitfalls and caveats. J Clin Neurophysiol. 1999;16(1):40–45.
Part III Clinical EEG: General
CHAPTER

Patterns of Unclear Significance


JONATHAN CHARLES EDWARDS AND EKREM KUTLUAY 14
T FAST ALPHA VARIANT/ SLOW
he subject of patterns of unclear significance encom-
passes a selection of electroencephalographic findings ALPHA VARIANT
that may look abnormal, but usually are not. These vari-
ants can be challenging, and may commonly lead to misinter- At times, harmonics or subharmonics of the background
pretation. These variants can be mainly grouped into two types: rhythm are seen. When this occurs, the resultant appearance is
variant rhythms and variant transients. Some of these findings of a rhythm that is twice as fast or half as fast as the awake back-
will also be discussed in the chapters on the normal EEG ground. When the appearance is of a rhythm that is twice the
(Chapters 9 and 10). frequency of the awake background, it is referred to as a “fast
The difficulties of interpretation posed by these variants arise alpha variant” (1,2). A “slow alpha variant” has an apparent fre-
for several reasons. These patterns possess features that are sim- quency that is about half that of the awake background (1–3).
ilar in some ways to abnormal patterns. Beginning with variant These harmonics will often have a notched appearance, as if
rhythms, as a general rule, a few common features help distin- many of the waves are simply being cut in half, but only partly
guish these rhythms from definitely abnormal findings: first, the so. Slow and fast alpha variants are reactive to eye opening and
variant rhythms are, for the most part, monomorphic. The term closure (Fig. 14.1).
“monomorphic” simply means that each of the waves in the Fast alpha variant is easy to detect, since awake posterior
rhythm looks distinctly like the other waves in the rhythm. The background rhythms in the mid to upper beta range would
waves have a characteristic shape, or “morphology,” that repeats. be quite unusual. True beta rhythms are most often seen in
Second, most of the variants have an arch-shaped appearance, the frontal, central, and parietal regions. When a posterior
although some of the normal rhythms may have notches in some background rhythm of 16, 18, or 20 is seen, it is easy to think
or all of the waves. Third, and most importantly, the variant of fast alpha variant. However, slow alpha variant is more
rhythms for the most part do not evolve. challenging, and may easily be misinterpreted as occipital
One important source of misinterpretation of these patterns slowing. This tendency to misread slow alpha variant as
may simply be that many of these patterns are uncommon. A sig- abnormal stems mostly from the fact that slowing is such an
nificant number of clinicians who read EEGs in clinical practice easily recognized abnormality, and all electroencephalogra-
may have only 2 to 3 months of closely supervised EEG training. phers look for signs of slowing. As a general helpful rule it is
The chances of seeing a rare pattern are quite low during this good to remember that any time one sees occipital slowing,
short training. Without extensive preparation in the fundamen- one must think of slow alpha variant first, before calling the
tals of electroencephalography and extensive supervised experi- record abnormal. One should look to find a sample of nor-
ence, misinterpretation of a rare pattern would not be surprising. mal awake background within the record. If the slowing is
Finally, some confusion may arise from the fact that many of approximately half the frequency of the patient’s awake back-
these patterns have changed names since first being described. ground, then the slowing is probably just a slow alpha vari-
In some cases, the nomenclature has changed numerous times. ant. An additional clue can be small notches in some of the
For some of patterns, numerous names are still concurrently in “slow waves,” bearing in mind that the notches can be quite
use today. subtle (Fig. 14.2).
A particular challenge in assessing the true prevalence and
clinical significance of these patterns is the tendency toward ALPHA SQUEAK
subject bias. Most patients who undergo EEG are doing so
because of neurological symptoms, and many of these patients Immediately on eye closure, the awake background is some-
have a clinical history suggestive of epilepsy. While a few stud- times initially faster, and of lower voltage. Over approximately
ies have looked at these findings in a general population, most 0.5 to 2 seconds, the background slows, and increases in ampli-
have reviewed their presence in a clinical population. Since EEG tude to become the normal awake background. This initial
is used to help diagnose epilepsy or other neurological condi- “speeding up” of the background immediately on eye closure is
tions, the true predictive value of these findings may be biased referred to as an “alpha squeak,” and is a normal finding
(Tables 14-1 and 14-2). (Fig. 14.3) (2,4–6).

267
Tabl e 1 4 . 1

Summary of Characteristics for Patterns of Unclear Significance

Finding Peak Age State Location Frequency Duration Characteristics


Slow or fast alpha Children and Awake, eyes closed Occipital Half or double Brief or lasting Sinusoidal, archiform,
variant adults the posterior many seconds notched, flat-topped
dominant rhythm
Alpha squeak Children and Awake, immediately Occipital Initially fast (beta) Typically Sinusoidal,
adults on eye closure slowing rapidly to 0.5–2 seconds spindle-shaped
the posterior
dominant rhythm
Rhythmic of Younger to Drowsiness, light Midtemporal, 5–7 Hz Several seconds, Arched, notched,
midtemporal middle-aged sleep at times or up to a flat-topped
theta bursts of adults with spread to minute or more Notche may
drowsiness parasagittal or give the
(“RMTD”) occipital– waves a sharply
temporal region contoured
Bilateral or appearance
independent
over the right
or left side
Midline theta Children and Common in Midline (CZ, 4–7 Hz Brief or Arched, notched,
rhythm (“Cigánek adults drowsiness FZ), parasagittal lasting many flat-topped
rhythm”) and during seconds
mental activation.
Less common
in resting
wakefulness
Subclinical Adults, mostly Drowsiness and Maximal in May begin with Brief or Sharply contoured,
rhythmic age 40s–80s resting wakefulness temporal and delta frequency, lasting minutes archiform,
electrographic (mean 60s) (rare cases parietal regions but then notched, flat-topped
discharge in reported in sleep) Usually bilateral, increases to
adults (SREDA) but may be 4–7 Hz
asymmetric

268
14- and 6-Hz Childhood and Drowsiness, Posterior 14 or 6 Hz 0.5–1 Archiform,
positive bursts adolescence light sleep temporal second comb-like,
low amplitude
6-Hz spike Children and Drowsiness, Highest 5–6 Hz 1–2 seconds Usually
and wave adults light sleep amplitude low-amplitude
bursts over fronto- ( 25 V) and
central short-duration
regions ( 30 msec) spike
component
Benign sporadic Mainly in Drowsiness, Anterior and Sporadic Usually 50 msec Mono- or diphasic
sleep spikes adults. Also light sleep midtemporal spike with steep
reported in descending arm
children and
adolescents
Wickets Adults Awake, sleep Temporal 6–11 Hz if Average of Archiform,
regions they occur 2–4-second monophasic
in runs bursts in
sleep
Frontal arousal Children Awakening, arousal Frontal 6.5–8.5 Hz Up to 13 Monomorphic
rhythm from sleep seconds

269
270 Part III ■ Clinical EEG: General

Tabl e 1 4 . 2 to the occipital–temporal region. Like mu rhythm, RMTD may be


seen bilaterally or may be independent over the right or left side.
Nomenclature Over Several Decades Like most rhythmic variants, it is typically arch-shaped, and is often
notched. These notches may give the waves a somewhat sharply
Other Terminologies contoured appearance, which may be mistaken for a sharp wave. At
EEG Pattern Used Previously times, the notched arches may have a flat-topped appearance
Rhythmic midtemporal Psychomotor variant (7) (1,7,8). RMTD may occur in runs that last several seconds, or even
theta bursts of drowsiness Rhythmic midtemporal up to a minute or more. However, like other normal rhythms,
(RMTD) discharge (8) RMTD can be clearly distinguished from seizure activity, because
Rhythmic theta bursts it is monomorphic, monorhythmic, and does not evolve.
of drowsiness (1) The reported overall prevalence of RMTD is quite low, rang-
Rhythmic temporal theta ing from 0.1% to 2% of records (9–13). The prevalence appears
bursts of drowsiness to be lower in age-matched normal controls than it is in EEGs
performed in neurological patients (9). While some studies (8)
Midline theta rhythm Theta-discharges in the have reported that a significant number of patients who are
middle line (15) found to have RMTD may have epilepsy, the majority of
Electrographic theta patients with this finding will not. RMTD has been reported in
discharges in the the presence of a structural abnormality (14). The overall clin-
midline (18) ical significance of this finding is unclear. It is most commonly
Cigánek rhythm considered a normal variant (Fig. 14.4).
Rhythmical midline
theta (23)
Subclinical rhythmic Paroxysmal discharge of the MIDLINE THETA RHYTHM
EEG discharge of adults temporo-parieto-occipital (CIGÁNEK RHYTHM)
(SREDA) junction (24)
14- and 6-Hz positive 14- and 6-Hz positive spikes Midline theta rhythm was first described by Cigánek, and is still
bursts (32), Ctenoids (33) commonly referred to as “Cigánek rhythm” (15). As is the case
with many variant rhythms, the midline theta rhythm was orig-
6-Hz spike and wave Spike and wave phantom (43) inally thought to be indicative of epilepsy, but was later found
bursts to be present in normal subjects also. Cigánek’s 1961 report is
Benign sporadic sleep Small sharp spikes (31), actually titled “Theta-discharges in the middle line—EEG
spikes Benign epileptiform symptom of temporal lobe epilepsy.” Midline theta is seen dur-
transients of sleep (50) ing wakefulness and drowsiness. Similar to RMTD, the midline
Wickets Wicket spikes (58) theta rhythm is typically a 4- to 7-Hz arch-shaped or sinusoidal
rhythm that may at times be notched or flat-topped (16). The
Frontal arousal rhythm Frontal arousal rhythm (63) main difference in appearance from RMTD is the location
(typically maximum over CZ), and presence in wakefulness.
The reported prevalence of midline theta rhythm has been
RHYTHMIC MIDTEMPORAL THETA highly variable, ranging from 2% to 35% of subjects (17,18). This
BURSTS OF DROWSINESS wide disparity is in part accounted for by subject selection.
Maulsby included the presence of midline theta during drowsiness
First described by Gibbs and Gibbs, this pattern has been given sev- and determined that it was present in 35% of normal adults. Many
eral names over nearly six decades (7). This pattern’s earlier name others have reported midline theta during mental tasks in a high
“psychomotor variant” is still commonly used. “Psychomotor” is number of patients (19–22). With the exclusion of midline theta
an older term for a complex partial seizure, typically of temporal rhythm that is seen only during drowsiness of mental activation,
lobe origin. Rhythmic midtemporal theta bursts of drowsiness the prevalence has been reported to be low in normal controls, and
(RMTD) has commonly been referred to as “psychomotor variant,” higher in patients with epilepsy. A more recent study (23) sug-
because it was felt to have a strong resemblance to a temporal lobe gested that when drowsiness and mental activation are excluded,
seizure. Subsequent names have included: “rhythmic midtemporal midline theta rhythm was common (26% overall) in their patients
discharge” (8), “rhythmic theta bursts of drowsiness,” and “rhyth- with epilepsy, and uncommon (0/54) in controls. Interestingly, in
mic temporal theta bursts of drowsiness” (1). the same series, and using the same exclusion criteria, midline
The name RMTD is widely accepted, and quite clear in listing theta rhythm was seen much more frequently in frontal lobe
the most distinctive characteristics. RMTD is seen mostly during epilepsy (48.1%) than in temporal lobe epilepsy (3.7%).
drowsiness and light sleep, although it may rarely be seen The clinical significance of midline theta rhythm is not univer-
during wakefulness. It occurs in bursts or trains of rhythmic 5- to sally agreed upon (16,23). It is typically regarded as a non specific
7-Hz activity. RMTD is maximal in the midtemporal leads, and finding, and is generally read as normal. Midline theta rhythm
may at times spread to the parasagittal regions, or more posteriorly during the awake recording and without mental tasks may
Chapter 14 ■ Patterns of Unclear Significance 271

Figure 14.1 Fast alpha variant.

Figure 14.2 Slow alpha variant.


272 Part III ■ Clinical EEG: General

Figure 14.3 Alpha squeak.

Figure 14.4 Rhythmic midtemporal theta bursts of drowsiness.


Chapter 14 ■ Patterns of Unclear Significance 273

Figure 14.5 Midline theta rhythm.

warrant further study. A potential practical challenge and con- This particular pattern has a distinctive feature that is differ-
founding variable would be in actually knowing what a subject is ent from other non-epileptic variants and that makes it especially
doing mentally in an awake, at rest recording, and ruling out that difficult to recognize and easy to misdiagnose. The SREDA pat-
particular mental tasks are taking place. Most evidence suggests tern may initially evolve. SREDA may begin with a single sharp
that the presence of midline theta rhythm during drowsiness or transient or a series of sharp transients, initially at delta frequen-
during mental task is not an abnormality (Fig. 14.5). cies, and then increasing to theta, before becoming a well-defined
4- to 7-Hz rhythm. A smaller number of cases remain in the 2- to
SREDA (SUBCLINICAL RHYTHMIC 4-Hz frequency range. After the initial period in which the
rhythm is established, however, SREDA does not continue to
ELECTROGRAPHIC DISCHARGE IN ADULTS)
evolve, despite the fact that SREDA may be seen in prolonged
SREDA is a fairly rare pattern that can be easily misinterpreted. It runs of 20 seconds to several minutes. The rhythm ends abruptly
is perhaps the most challenging pattern among those that are in about half of patients, and in the other cases, the SREDA grad-
considered normal variants. First described in 1961 by Naquet et ually dissipates and merges with ongoing normal background
al., this pattern was originally thought to be associated with rhythms. Unlike seizures, SREDA is not followed by “postictal”
hypoxia (24). Westmoreland and Klass described 65 patients who slowing, and no clinical changes accompany SREDA.
had 142 EEG recordings, and provided the term “Subclinical While this pattern may easily be mistaken for an ictal pat-
Rhythmic EEG Discharge in Adults.” In this series, SREDA was tern, evidence suggests that this pattern has no clear association
seen in patients aged 42 to 80, with an average age of 61 (25). with seizures (13,25,28,29). In a large series, there was not only
The pattern is seen mostly during relaxed awake or drowsy a low incidence of clinical history of seizures or subsequent
states, although SREDA can be seen during sleep (25,26). The development of seizures, but also a “uniform lack of any clinical
typical appearance of SREDA is a diffuse, sharply contoured, accompaniment during the bursts, even with the involvement of
theta rhythm that is usually maximal in the temporal and both hemispheres and persistence of several minutes” (25). One
parietal regions. While SREDA is usually bilateral, it may be study of ictal single photon emission computer tomography
asymmetric. An interesting feature of SREDA is that it often demonstrated no significant changes in uptake during the
occurs in long runs, such as 15 seconds to a minute or more bursts, further suggesting that the pattern is not ictal (30).
(25). The appearance on visual inspection is of a monomorphic
rhythm. However, a more recent detailed digital study using fre- 14- AND 6-HZ POSITIVE BURSTS
quency spectral analysis and Laplacian montages suggests that
SREDA is “composed of a complex mixture of multiple rapidly Like many other variants, this unique EEG phenomenon was
shifting frequencies predominantly in the theta range, which show also described by Gibbs and Gibbs (31,32). Although it was pre-
poor spatial and temporal resolution” (Fig. 14.6) (27). viously called 14- and 6-Hz positive spikes, the current
274 Part III ■ Clinical EEG: General

Figure 14.6 Subclinical electrographic discharges of adults (SREDA).

preferred term is 14- and 6-Hz positive bursts. Lombroso et al. positive bursts were the most common pattern of uncertain sig-
also proposed the name “ctenoids” due to their appearance and nificance in their series of 100 asymptomatic adults (39).
complex morphology resembling a comb (ktenos in Greek Several reports were published in the literature correlating
translates to “comb”) (33). the existing of this pattern with certain symptoms and illnesses.
The typical bursts last between 0.5 and 1 second and usually Wide variety of psychiatric conditions and neurovegetative
occur during light sleep or drowsiness (34–36). However, a few symptoms were subjected to research (33,34,40). Epilepsy was
cases with occurrence in REM sleep have also been reported also thought to be more frequent in patients with 14- and 6-Hz
(37). The bursts can be unilateral or bilateral but usually show positive bursts (41). However, most of the patients with behav-
a shifting predominance if enough bursts are recorded in the ioral symptoms and organic CNS impairment were children or
same patient (34–36). They are usually small in amplitude; adolescents, which is the age of peak incidence for this phe-
therefore, referential or long-distance electrode montages nomenon (42). One study suggests that a majority of patients
reveal better waveforms (34–36). Although the laterality of with seizures and 14- and 6-Hz positive spikes also showed
these bursts can appear to shift, the maximum amplitude is other epileptiform abnormalities in their EEGs (41).
expressed over the posterior temporal head regions (34–36). This controversial pattern seems to be an age-related phe-
There seems to be strong age predilection. This activity ini- nomenon. Although there are arguments regarding a patholog-
tially starts to appear during early childhood and reaches its ical significance, the reported incidence of up to 58% in normal
peak during adolescence (34–37). The coexistence of 14- and 6- teenagers and 12% in healthy adults makes this relatively hard
Hz positive bursts and 6-Hz spike and wave bursts was also to prove (Fig. 14.7).
reported. Silverman hypothesized that a maturational sequence
might be responsible for this co-occurrence, and that most of SIX-HZ SPIKE AND WAVE BURSTS
the 6-Hz spike and wave bursts are part of the 14- and 6-Hz pos-
itive bursts (38). The 14- and 6-Hz positive bursts are still seen Six-Hz spike and wave bursts were first described in 1950 as
during adulthood but the incidence decreases with advanced age “wave and spike phantom” (32,43). The first collected cases
(34). Lombroso et al. reported highest incidence of 58% in the revealed a unique EEG pattern of 1-second bursts of 5- to 6-Hz
literature in teenage boys between the ages of 13 and 16 years spike and waves (43). The average amplitude of the discharge
(33). In adults, the incidence is much lower. An incidence as low was low, around 25 V or less, with the spike component less
as 4% was seen in a group of psychiatric patients (38), whereas than 30-msec duration (43–45). This pattern can be seen in
a study of EEG in asymptomatic normal adults revealed a rate both children and adults, with amplitude usually higher over
around 12% (39). In fact, the latter claimed that 14- and 6-Hz the fronto-central regions (43,44). These bursts were mostly
Chapter 14 ■ Patterns of Unclear Significance 275

Figure 14.7 Fourteen- and 6-Hz positive bursts.

seen during drowsiness and light sleep but they were also seen (47). Current consensus is that this EEG pattern is of unclear
during full wakefulness (43). Bursts are usually diffuse but may clinical significance; however, high amplitude of the spikes,
show anterior or posterior predominance. Marshall suggested slower rate than 6 Hz, and persistence of the discharges in
the comparison linkage that measures the potential difference deeper stages of sleep are more likely associated with seizures
between analogous scalp areas for better viewing rather than (Fig. 14.8) (35,49).
traditional bipolar or referential montages (43).
Average incidence of 6-Hz spike and wave bursts is BENIGN SPORADIC SLEEP SPIKES
between 1% and 2.5% of all EEG recordings (13,43,44,46,47).
Several authors tried to link this controversial pattern to Small sharp spikes (SSS), the term originally described by Gibbs
specific complaints, ranging from simple fainting to vegeta- and Gibbs, are also known as benign epileptiform transients of
tive or dysautonomic pathologies, and from psychiatric dis- sleep (BETS) or benign sporadic sleep spikes (BSSS) (32,34,50).
turbances to seizures (42,43,44,48). However, occurrence in However, the latter became a more preferred term recently. As
normal people and absence of clinical symptoms during the indicated by the name SSS, these are short in duration and low in
bursts make it difficult to label these discharges as a specific amplitude. Although they are usually less than 50 msec and less
indicator of any certain disease (34,48). As mentioned earlier, than 50 V, duration can be slightly longer and amplitude can be
Silverman suggested that there is relationship as a matura- slightly higher depending on the recording circumstances and
tional sequence and waveform spectrum between 6-Hz spike montage used (34–36). The shape is rather simple and consists of
and wave bursts and 14- and 6-Hz positive bursts (38). He mono- or diphasic spike with steep descending arm (34,35). An
claimed that most of the 6-Hz spike and wave bursts are part aftergoing slow wave is not prominent and usually is lower in
of the 14- and 6-Hz positive bursts and transition between amplitude than the spike component (34,36,50). Background
these two types may lead to occurrence of both patterns in activity at the region of BSSS is not disrupted (34,35). These dis-
the same person (38). charges are best seen during drowsiness and light sleep (stage I
On investigating more than 60,000 EEGs over 30 years, and II) and are best displayed over anterior and midtemporal or
Hughes proposed two distinct subtypes of this pattern (47). ear electrodes (34,35). Although they have been reported in chil-
The features of these two variants were summarized by dren and adolescents, BSSS are mainly seen in adults (35,51).
acronyms WHAM (Wake, High amplitude, Anterior location, The origin and cortical location of the BSSS have been the
Male gender) and FOLD (Female gender, Occipital location, subject of a few investigations. Recording with depth electrodes
Low amplitude, Drowsiness) (47). Hughes indicated that these in two patients revealed similarity to surface findings (52).
two variants are more extreme forms, and also suggested that Invasively recorded spikes had widespread distribution and
the more components of the WHAM form that exist in a sin- similar simple morphological appearance (53). They occurred
gle person, the more likely is the association with seizures as single sporadic transients without disrupting the ongoing
276 Part III ■ Clinical EEG: General

Figure 14.8 Six-Hz spike and wave discharges.

background activity; however, the amplitude was higher than WICKETS


surface recordings (52). Another study with subdural electrodes
showed that BSSS were more widespread than on-surface Wickets are archiform waveforms first described as wicket
recordings, and were mainly seen over the posterior mesial tem- spikes by Reiher and Lebel in 1977 (58). They are simple
poral area (53). monophasic waveforms with surface negativity (58). Initial
BSSS are generally not thought to be related to any certain descriptions revealed that wicket spikes are found in both
disease, and are accepted as a variant of normal EEG activity awake and sleep EEGs (58). Because of the intermixed back-
(35,50,54). However, over the years, several reports tried to ground activities during the awake state, they are better recog-
find a connection between BSSS and seizures or certain psy- nized during sleep (58,59). They are best seen during initial
chiatric conditions. BSSS were thought to be a sign of epilep- stages of sleep but also reported during REM sleep (58,60).
togenicity. Different authors reported higher incidence of Their location remains unchanged during different stages of
BSSS in patients with seizures particularly in patients of sleep (60). Wickets can occur as isolated “spikes” or they may
younger age (51,55). One study indicated that the highest come in runs (34,36,58). If they occur in runs, the usual fre-
incidence is seen in patients younger than 20 years of age, with quency is between 6 and 11 Hz. The amplitude of a wicket
a gradual decrease with advanced age leading to complete dis- spike may range between 60 and 210 V and is maximally
appearance after age 80 years (55). Still other studies claimed expressed over the temporal regions (34,36,58). Although they
a relationship between BSSS and certain psychiatric condi- shift sides, often one side is more dominant than the other
tions. Positive correlations were reported with manic depres- (58).
sive psychosis and psychotic depressive episodes as well as Wickets are usually seen in adults (58). They are almost
during acute mania (56,57). exclusively reported in adults 30 years or older; however, cases
BSSS were first described about half a century ago. They are as young as 20 years of age have also been reported (12,36,58).
very common EEG findings seen with up to 25% incidence in Wickets are reported to be an uncommon EEG pattern. Their
the normal population (36,50). Although several studies incidence in large EEG series has been reported to be less than
reported increased frequency of BSSS in certain psychiatric 1%, ranging between 0.03% and 0.96% (12,13,58). In adults 30
conditions and seizures because of their high incidence in nor- years and older this incidence may rise up to 2.9% (58).
mal population, we believe this peculiar EEG activity is of Wickets are considered to be a normal variant. In Reiher and
unknown clinical significance with no statistical relationship to Lebel’s original description, wickets occurred four times more
a certain disease (Fig. 14.9). in patients with no history of seizures than in those with a
Chapter 14 ■ Patterns of Unclear Significance 277

Figure 14.9 Benign sporadic sleep spikes (BSSS).

history of seizures (58). An experienced electroencephalogra- Twenty-five years after the initial description of this rare and
pher can differentiate wickets from temporal sharp waves easily unusual EEG pattern, Hughes and Daaboul discussed their find-
by the lack of aftergoing slow waves and the absence of distor- ings in 50 cases (64). The incidence of FAR was 0.22% in their
tion of the background activities (36,49,58). However, one can series of 22,500 patients seen over an 8-year period. The charac-
also over-read or misinterpret wickets as epileptiform sharp teristics of the pattern were similar to those described before.
waves, especially if they occur in isolation (61,62). Clues men- While FAR was the only atypical pattern in 58% of these patients,
tioned above, as well as observation of runs of wicket spikes nearly half (42%) also showed other abnormalities, including
somewhere else in the same EEG, should alert the reader that focal and generalized epileptiform activities. Clinically, 70% of the
this is a non- epileptiform variant (Fig. 14.10) (61). children had a seizure disorder, and 56% had both seizure and
cognitive/behavior disorders. Only 6% of the reported patients
FRONTAL AROUSAL RHYTHM (FAR) had neither seizure nor cognitive/behavior disorders. However,
the incidence of cognitive/behavior disorders was not statistically
This unique EEG pattern was first described by White and different from their control group whose EEG did not show a FAR
Tharp in 1974 although the pattern was first recognized in 1969 pattern. Their conclusion was that FAR is related to seizures. To
(63). In their original report, the authors described 8 cases out support this view, a recent case report described a 6-year-old boy
of 4780 EEGs reviewed over a 4-year period. This group of eight with mental retardation and possible seizure disorder (65). A typ-
children consisted of four males and four females with a mean ical FAR pattern was seen during few episodes associated with eye
age of 6.6 years, ranging from 2 to 14 years. Their initial diag- fluttering followed by chewing, increased inspiration, and upper
noses were mild cerebral dysfunction and/or seizure disorder. lip quivering. These episodes were all recorded during arousal
Representative EEG samples showed normal awake and sleep from sleep and there were no clinical changes seen when the
patterns. However, during arousal from stage II sleep, a 6.5- to patient was awakened without the FAR pattern present.
8.5-Hz rhythm was seen over the frontal regions involving Few illustrative reports are available in the literature. The
mainly F3 and F4 electrodes with no or little spread to adjacent incidence of FAR in general population is unknown. The gen-
regions. The overall amplitude of this activity was between 30 eral consensus is that FAR is more of a non specific EEG pattern
and 150 V, with duration of up to 13 seconds. This pattern was with unknown clinical significance (2,35). However, the
originally described in children with evidence of minimal cere- reported case series as well as demonstration of FAR as an ictal
bral dysfunction and seizures but authors noted that the inci- pattern in a reported case warrants careful review for other
dence in a normal population was unknown at the time of the interictal activity, and detailed questioning for possible seizure-
publication (63). like activity (Fig. 14.11).
278 Part III ■ Clinical EEG: General

Figure 14.10 Wickets.

Figure 14.11 Frontal arousal rhythm (FAR) (courtesy of Dr William Tatum IV).
Chapter 14 ■ Patterns of Unclear Significance 279

REFERENCES 23. Beleza P, Bilgin Ö, Noachtar S. Interictal rhythmical midline theta


differentiates frontal from temporal lobe epilepsies. Epilepsia.
1. Chatrian GE, Bergamini L, Dondey M, et al. A glossary of terms 2009;50:550–555.
most commonly used by clinical electroencephalographers. 24. Naquet R, Louard C, Rhodes J, et al. A propos de certaines
Electroencephalogr Clin Neurophysiol. 1974;37:538–548. décharges paroxystiques du carrefour temporo-pariéto-occipital.
2. Westmoreland BF, Klass DW. Unusual EEG patterns. J Clin Leur activation par l’hypoxie. Rev Neurol. 1961;105:203–207.
Neurophysiol. 1990;7:209–228. 25. Westmoreland BF, Klass DW. A distinctive rhythmic EEG dis-
3. Goodwin JE. The significance of alpha variants in the EEG, and charge of adults. Electroencephalogr Clin Neurophysiol. 1981;51:
their relationship to an epileptiform syndrome. Am J Psychiatry. 186–191.
1947;104:369–379. 26. Fleming WE, Avidan A, Malow BA. Subclinical rhythmic electro-
4. Bekkering D, Kamp A, Storm van Leeuwen W, et al. Example of the graphic discharge of adults (SREDA) in REM sleep. Sleep Med.
use of the spectrograph and the magnetograph—the squeak phe- 2004;5:77–81.
nomenon. Electroencephalogr Clin Neurophysiol. 1956;8:721. 27. O’Brien TJ, Sharbrough FW, Westmoreland BF, et al. Subclinical
5. Storm van Leeuwen W, Bekkering D. Some results obtained with rhythmic electrographic discharges of adult (SREDA) revisited: a
the EEG spectrograph. Electroencephalogr Clin Neurophysiol. study using digital EEG analysis. J Clin Neurophysiol. 1998;15:
1958;10:563–570. 493–501.
6. Storm van Leeuwen W, Kamp A, Kniper J. Concerning the 28. Begum T, Ikeda A, Takahashi J, et al. Clinical outcome in patients
“squeak” phenomenon of the alpha rhythm. Electroencephalogr with SREDA (subclinical rhythmic EEG discharge of adults).
Clin Neurophysiol. 1960;12:244. Intern Med. 2006;45:141–144.
7. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. 1st ed. 29. Westmoreland BF, Klass DW. Unusual variants of subclinical
Cambridge, MA: Lew A. Cummings; 1941:55, 109. rhythmic electroencephalographic discharge of adults.
8. Lipman IJ, Hughes JR. Rhythmic midtemporal discharges: an Electroencephalogr Clin Neurophysiol. 1997;102:1–4.
electro-clinical study. Electroencephalogr Clin Neurophysiol. 1969; 30. Thomas P, Migneco O, Darcourt J, et al. Single photon emission
27:43–47. computed tomography study of subclinical rhythmic electro-
9. Gibbs FA, Rich CL, Gibbs EL. Psychomotor-variant type of seizure graphic discharge in adults. Electroencephalogr Clin Neurophysiol.
discharge. Neurology. 1963;13:991–998. 1992;83:223–227.
10. Gibbs EL, Gibbs FA. Psychomotor-variant type of paroxysmal 31. Gibbs FA, Gibbs EL. Fourteen and six per second positive spikes.
cerebral dysrhythmia. Clin Electroencephalogr. 1989;20:147–152. Electroencephalogr Clin Neurophysiol. 1963;15:553–558.
11. Maulsby RL. EEG patterns of uncertain diagnostic significance. In: 32. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 3.
Klass DW, Daly DD, eds. Current Practice of Electroencephalography. Reading, MA: Addison Wesley; 1964.
New York: Raven Press; 1979:411–419. 33. Lombroso CT, Chwartz IH, Clark DM, et al. Ctenoids in healthy
12. Radhakrishnan K, Santoshkumar B, Venugopal A. Prevalence of adults. Controlled study of 14- and 6-per-second positive spiking.
benign epileptiform variants observed in an EEG laboratory from Neurology. 1966;16:1152–1158.
South India. Clin Neurophysiol. 1999;110:280–285. 34. Klass DW, Westmoreland BF. Nonepileptogenic epileptiform elec-
13. Santoshkumar B, Chong JJ, Blume WT, et al. Prevalence of benign troencephalographic activity. Ann Neurol. 1985;18:627–635.
epileptiform variants. Clin Neurophysiol. 2009;120:856–861. 35. Ebersole JS, Pedley TA. Current Practice of Clinical Electroence-
14. Hennessy MJ, Koutroumanidis M, Hughes E, et al. Psychomotor phalography. 3rd ed. Philadelphia, PA: Lippincott Williams &
EEG variant of Gibbs: an association with underlying structural Wilkins; 2003.
pathology. Clin Neurophysiol. 2001;112:686–687. 36. Tatum WO, Husain AM, Benbadis SR, et al. Normal adult EEG and
15. Cigánek L. Theta-discharges in the middle line —EEG symptom of patterns of uncertain significance. J Clin Neurophysiol. 2006;23:
temporal lobe epilepsy. Electroencephalogr Clin Neurophysiol. 1961; 194–207.
13:669–673. 37. Beun AM, van Emde Boas W, Dekker E. Sharp transients in the
16. Westmoreland BF, Klass DW. Midline theta rhythm. Arch Neurol. sleep EEG of healthy adults: a possible pitfall in the diagnostic
1986;43:139–141. assessment of seizure disorders. Electroencephalogr Clin
17. Maulsby R. The Normative Electroencephalographic Data Reference Neurophysiol. 1998;106:44–51.
Library. Final Report, Contract NAS 9-1200. Washington, DC: 38. Silverman D. Phantom spike-waves and the fourteen and six per
National Aeronautics and Space Administration; 1968.
^
second positive spike pattern: a consideration of their relationship.
18. Mokrán V, Cigánek L, Kabátnik Z. Electrographic theta discharges Electroencephalogr Clin Neurophysiol. 1967;23:207–213.
in the midline. Eur Neurol. 1971;5:288–293. 39. Jabbari B, Russo MB, Russo ML. Electroencephalogram of asymp-
19. Gevins A, Smith ME, McEvoy L, et al. High-resolution EEG map- tomatic adults. Clin Neurophysiol. 2000;111:102–105.
ping of cortical activation related to working memory: effects of 40. Boutros N, Fristad M, Abdollohian A. The fourteen and six posi-
task difficulty, type of processing, and practice. Cereb Cortex. tive spikes and attention-deficit hyperactivity disorder. Biol
1997;7:374–385. Psychiatry. 1998;44:298–301.
20. Okada S, Urakami Y. Midline theta rhythm revisited. Clin 41. Hughes JR, Cayaffa JJ. Positive spikes revisited—in the adult. Clin
Electroencephalogr. 1993;24:6–12. Elecroencephalogr. 1978;9:52–59.
21. Takahashi N, Shinomiya S, Mori D, et al. Frontal midline theta 42. Small JG, Sharpley P, Small IF. Positive spikes, spike-wave phan-
rhythm in young healthy adults. Clin Electroencephalogr. 1997; toms, and psychomotor variants. A survey of these EEG patterns
28:49–54. in psychiatric patients. Arch Gen Psychiat. 1968;18:232–238.
22. Inanaga K. Frontal midline theta rhythm and mental activity. 43. Marshall C. Some clinical correlates of the wave and spike phan-
Psychiatry Clin Neurosci. 1998;52:555–566. tom. Electroencaphalogr Clin Neurophysiol. 1955;7:633–636.
280 Part III ■ Clinical EEG: General

44. Tharp BR, Arsenal E. The 6-per-second spike and wave complex. 55. Hughes JR, Gruener G. Small sharp spikes revisited: further data
The wave and spike phantom. Arch Neurol. 1966;15:533–537. on this controversial pattern. Clin Electroencephalogr. 1984;15:
45. Tharp BR. The six per second spike and wave complex (the wave 208–213.
and spike phantom). Electroencephalogr Clin Neurophysiol. 1967; 56. Small JG. Small sharp spikes: EEG signals of psychiatric signifi-
23:291. cance. Electroencephalogr Clin Neurophysiol. 1970;28:417–422.
46. Olson SF, Hughes JR. The clinical symptomatology associated with 57. Inui K, Motomura E, Okushima R, et al. Electroencephalographic
the 6 c/sec spike and wave complex. Epilepsia. 1970;11:383–393. findings in patients with DSM-IV mood disorder, schizophrenia,
47. Hughes JR. Two forms of the 6/sec spike and wave complex. and other psychotic disorders. Biol Psychiatry. 1998;43:69–75.
Electroencephalogr Clin Neurophysiol. 1980;48:535–550. 58. Reiher J, Lebel M. Wicket spikes: clinical correlates of a previously
48. Thomas JE, Klass DW. Six-per-second spike-and-wave pattern in undescribed EEG pattern. Can J Neurol Sci. 1977;4:39–47.
the electroencephalogram. Neurology. 1968;18:587–593. 59. Waveform Window #11. Wicket Spikes. Am J Electroneurodiagn
49. Westmoreland BF. Epileptiform electroencephalographic patterns. Technol. 2008;48:52–55.
Mayo Clin Proc. 1996;71:501–511. 60. Gelisse P, Kuate C, Coubes P, et al. Wicket spikes during rapid eye
50. White JC, Langston JW, Pedley TA. Benign epileptiform transients movement sleep. J Clin Neurophysiol. 2003;20:345–350.
of sleep. Clarification of the small sharp spike controversy. 61. Benbadis SR, Tatum WO. Overinterpretation of EEGs and misdi-
Neurology. 1977;27:1061–1068. agnosis of epilepsy. J Clin Neurophysiol. 2003;20:42–44.
51. Saito F, Fukushima Y, Kubota S. Small sharp spikes: possible rela- 62. Krauss GL, Abdallah A, Lesser R, et al. Clinical and EEG features of
tionship to epilepsy. Clin Electroencephalogr. 1987;18:114–119. patients with EEG wicket rhythms misdiagnosed with epilepsy.
52. Westmoreland BF, Reiher J, Klass DW. Recording of small sharp Neurology. 2005;64:1879–1883.
spikes with depth electroencephalography. Epilepsia. 1979;20: 63. White JC, Tharp BR. An arousal pattern in children with organic
599–606. cerebral dysfunction. Electroencephalogr Clin Neurophysiol.
53. McLachlan RS, Lubus N. Cortical location of benign paroxysmal 1974;37:265–268.
rhythms in the electrocorticogram. Can J Neurol Sci. 2002;29: 64. Hughes JR, Daaboul Y. The frontal arousal rhythm. Clin
154–158. Electroencephalogr. 1999;30:16–20.
54. Reiher J, Klass DW. “Small sharp spikes” (SSS): electroencephalo- 65. Hughes JR. The frontal arousal rhythm (FAR) is an ictal pattern: a
graphic characteristics and clinical significance. Electroencephalogr case report. Clin Electroencephalogr. 2003;34:13–14.
Clin Neurophysiol. 1970;28:94.
CHAPTER
EEG of Degenerative Disorders
of the Central Nervous System
JOHN GAITANIS
15
INTRODUCTION of one or more of these enzymes. Undegraded material accumu-
lates, leading to cellular and organ dysfunction. Most of these
Degenerative disorders of the central nervous system encom- conditions exhibit autosomal recessive inheritance.
pass a wide range of genetic and metabolic conditions, which
result in progressive neurologic disability. Senile dementia can GM2 Gangliosidoses/ Tay–Sachs Disease
be included under this heading, but is discussed elsewhere in The infantile form of Tay–Sachs disease is named for the British
the text. Metabolic encephalopathies are distinct from chronic, ophthalmologist Warren Tay, who, in 1881, described the
degenerative conditions in that they often result from systemic cherry-red spot, and the American neurologist Bernard Sachs,
disorders and are commonly reversible. Given the wide array of who reported on the clinical features and the enlarged pyrami-
etiologies for degenerative diseases, they can be categorized in dal neurons of the condition. The biochemical understanding
multiple ways. The most common classification system is to came about when Ernst Klenk found a novel group of glycol-
separate these disorders based on the type of cellular organelle ipids in the brains of these patients, which was later discovered
that is primarily involved, as is done in this chapter. Special to be GM2 ganglioside. Its accumulation in neurons results
attention is also given to the predominant neuroanatomical from a deficiency of the lysosomal enzyme hexosaminidase A.
structures involved, since this allows for an EEG to neu- The hexosaminidase enzyme is comprised of alpha and beta
roanatomical correlation (Table 15-1). subunits. Hexosaminidase A consists of one alpha and one beta
subunit and hexosaminidase B is composed of two beta sub-
LYSOSOMAL DISORDERS units. In Tay–Sachs disease, there is loss of the alpha subunit
resulting in absence of hexosaminidase A.
Lysosomes are membrane-bound organelles that function as the The result is a neurologically devastating disease that begins
digestive system of the cell. They contain enzymes that break in the first months of life. The initial symptom is excessive star-
down macromolecules originating either internally or exter- tle myoclonus to noise, sound, or touch. The startle does not
nally. Lysosomal storage disorders occur when there is deficiency attenuate with subsequent exposures, distinguishing it from a
normal Moro response. The myoclonic movements consist of
arm and leg extension. As the disease progresses, motor, visual,
Tabl e 1 5 . 1
and intellectual impairments develop. The affected infant
Disorders affecting primarily cortex develops axial hypotonia with appendicular spasticity and
Tay–Sachs disease increased reflexes. Motor skills, such as head control, rolling,
Neuronal ceroid lipofuscinosis and purposeful hand movements, are lost. Visual loss ensues as
Mucopolysaccharidosis lipid accumulates in the retinal ganglion cells, resulting in a
Sialidosis whitish discoloration around the fovea—the characteristic
Rett syndrome cherry-red spot. With intraneuronal accumulations of GM2
Menkes disease ganglioside, macrocephaly develops. The children experience
Alpers–Hunttenlocher syndrome progressive cognitive impairment and seizures. Feeding and
Disorders affecting primarily white matter autonomic disturbances develop. Between 3 and 5 years of age,
Metachromatic leukodystrophy the patient succumbs to cachexia or aspiration pneumonia.
Globoid leukodystrophy The disease affects multiple regions of the brain. The basal
X-linked adrenoleukodystrophy ganglia and cerebral white matter show low-density signal on
Neonatal adrenoleukodystrophy CT in early stages and enlargement of the caudate nucleus later
Disorders affecting primarily basal ganglia in the disease course (1). Early into the disease, the interictal
Leigh syndrome EEG may be normal. By age 1, there is a rapid and progressive
Neurodegeneration with brain iron accumulation deterioration of the EEG (2). Paroxysmal features are not
Infantile neuroaxonal dystrophy prominent, but during myoclonic seizures, widespread spike
Wilson disease and sharp waves are seen (3). A progressive decline in voltage
occurs in later stages of the disease, likely resulting from contin-

281
282 Part III ■ Clinical EEG: General

ued neuronal loss. The electroretinogram remains normal even abnormalities, ataxia, and pyramidal or extrapyramidal signs
in advanced stages (2,4), since only the ganglion cell layer of the (10). These disorders are inherited in an autosomal recessive
retina is affected. Progressive loss of the VEP occurs between 9 pattern and there are at least seven genes responsible for the
and 15 months (2). clinical phenotypes.
Sandhoff disease, resulting from mutations of the beta sub- MRI findings in INCL include cerebral atrophy, callosal
unit of hexosaminidase A, is phenotypically similar to thinning, T2-weighted hyperintensities in the thalami, and
Tay–Sachs disease, the only differences being the presence cerebellar atrophy. After 4 years of age, the atrophy is severe and
of hepatosplenomegaly, cardiomyopathy, or N-acetylglu- the entire white matter shows abnormally high signal intensity.
cosamine-containing oligosaccharides in the urine of patients SPECT studies show progressive cerebral and cerebellar hypop-
with Sandhoff disease. The AB variant occurs when there is a erfusion with relative sparing of the basal ganglia. LINCL and
deficiency of the GM2 activator, which is necessary for hydrol- JNCL reveal lesser degrees of atrophy (11).
ysis of GM2 gangliosides by hexosaminidase A. Its phenotype The EEG findings reflect the predominance of cortical
is indistinguishable from the infantile form of Tay–Sachs (5). abnormalities on MRI and SPECT. In INCL, the EEG initially
Late-onset variants of Tay–Sachs also exist, and can manifest in shows a lack of attenuation to eye opening and a disappear-
childhood, adolescence, or adulthood. They may result in a ance of sleep spindles. The background activity gradually
wide constellation of symptoms including spastic paraparesis, attenuates and becomes flat by 3 years of age. ERGs are
ataxia with cerebellar atrophy, seizures, or psychiatric symp- unrecordable by age 3 and VEPs are unrecordable by 4 years.
toms. Psychosis and depression can even be the initial manifes- There is progressive attenuation of cortical somatosensory
tation in adult patients (6). EEG changes in late-onset GM2 potentials (12). In LINCL, seizures are the presenting symp-
gangliosidosis are variable and unrelated to age or enzyme tom, and the EEG reveals occipital spike waves in response to
defect (2). photic stimulation at 1 to 2 Hz. The ERG is abnormal at pres-
entation and extinguishes in time. VEPs are enhanced, but
GM1 Gangliosidosis diminish in the final stages of the disease (13). In JNCL, the
GM1 gangliosidosis results from a deficiency of -galactosi- EEG shows disorganization and spike- and slow-wave com-
dase, which is responsible for cleavage of the terminal galac- plexes. ERG and VEPs are abnormal early into the course of
tose of GM1. The result is neuronal storage of the disease. The EEG abnormalities of ANCL are nonspecific
monosialoganglioside GM1. Histopathology reveals a marked (14). In one case series, the frequency of background slowing
decrease in the number of oligodendrocytes and myelin in all types of NCL was 94.6% and epileptiform discharges
sheaths. As a result, spasticity, tonic spasms, and pyramidal were seen in 81.1% (15). Slow-frequency photic stimulation
signs predominate. As in Tay–Sachs disease, a cherry-red spot elicited a response in 5 out of 22 patients, a giant SSEP was
is seen and the ERG remains normal (2). The EEG also shows seen in 7 of 25 patients, and slow or absent waveforms were
a progressive deterioration as it does in GM2 patients. EEGs seen in 9 of 25 patients on VEP (15).
universally reveal abnormal slowing, which worsens as the
disease advances. By 2 to 3 years of age, 4 to 5 cycle/sec rhyth- Mucopolysaccharidoses
mic activity is seen in the temporal regions. Paroxysmal dis- The mucopolysaccharidoses (MPSs) are a heterogeneous
charges, however, are not commonly observed in GM1 group of disorders caused by deficiency of a lysosomal enzyme
patients (7). involved in the degradation of glycosaminoglycans (16). As
glycosaminoglycans accumulate in lysosomes, cell and organ
Neuronal Ceroid Lipofuscinoses dysfunction result. Seven types exist. The age of onset and
The neuronal ceroid lipofuscinoses (NCL) are a group of neu- severity vary, but most exhibit a chronic, progressive course
rodegenerative conditions caused by intralysosomal accumu- characterized by multisystemic involvement, coarse facies,
lation of ceroid and lipofuscin (8). Originally described as a organomegaly, and bony defects (dysostosis multiplex) (16).
form of “amaurotic familial idiocy,” the group of disorders The more severe forms (MPS I, MPS II, and MPS VIII) result
was renamed NCL by Zeman to better distinguish them from in progressive mental retardation, whereas patients with MPS
gangliosidoses (9). Clinically, the disease group is character- IV and MPS VI have normal cognition, even when somatic
ized by cognitive decline, progressive myoclonic epilepsy, and manifestations are severe (16). The only variant without
motor regression. Some forms also cause visual failure (8). somatic symptoms is MPS III, of which there are four sub-
The four originally described phenotypes are organized types. MPS III presents between 1 and 6 years of age with
according to age of onset: infantile (INCL), late infantile delayed psychomotor development and behavioral problems.
(LINCL), juvenile (JNCL), and adult neuronal ceroid lipofus- Progressive dementia, sleeplessness, and epilepsy follow (17).
cinosis (ANCL). Patients with INCL are normal at birth, and In MPS III, the EEG is normal in half of patients, but shows
exhibit developmental delays or myoclonic seizures between 6 diffuse slowing in the rest (18). Low-amplitude 12- to 15-Hz
and 24 months of age. In LINCL, patients experience develop- activity is seen intermixed with generalized delta frequencies
mental regression and epilepsy between 2 and 4 years. JNCL (19). These findings correspond to the severe cortical involve-
develops between 4 and 10 years with visual loss being the ini- ment seen pathologically (19). VEPs and BAEPs are almost
tial symptom and epilepsy following shortly afterwards. always normal (18). Enzyme replacement therapy and
ANCL appears sometime around 30 years of age and presents hematopoietic stem cell transplantation offer potential benefit
with progressive myoclonic epilepsy, dementia, behavioral in these conditions (20,21).
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 283

Sialidosis Types I and II (Neuraminidase (NPD-A), infants present in the first few weeks or months of life
Deficiency) with failure to thrive and hepatomegaly. Psychomotor regression
In sialidosis, a deficiency of the enzyme neuraminidase leads to follows, with head lag, inability to sit, and loss of interest in sur-
progressive storage of sialidated glycopeptides and oligosaccha- roundings. In time, rigidity and opisthotonus predominate. Death
rides. Sialidosis bears clinical and histopathologic resemblance occurs in the second or third year. Startle myoclonus, like that seen
to the mucopolysaccharidoses, in that patients exhibit mild in Tay–Sachs, is not observed and there are no coarse features, like
Hurler-like facies, skeletal dysplasia, and psychomotor retarda- those present in mucopolysaccharidosis. In Niemann–Pick type B
tion. Sialidosis is divided into types I and II (22). Type I has a (NPD-B), visceral involvement predominates without significant
later age of onset and patients exhibit little if any dysmorphol- neurologic impairment (31). An intermediate form between
ogy. Visual failure, cherry-red spots, ataxia, and myoclonus are NPD-A and NPD-B may also exist with retinal degeneration and
common features (23,24). Type II patients have an earlier age of slowed nerve conduction presenting in adolescence or adulthood
onset and demonstrate coarse facies, hepatosplenomegaly, and (30). In Niemann–Pick type C (NPD-C), patients present between
skeletal anomalies (dysostosis multiplex and spinal deformities) ages 3 and 8 years with ataxia, vertical supranuclear gaze palsy, and
(25). MRI findings include atrophy of the cerebral hemispheres, hepatosplenomegaly. In time, they experience dementia, dystonia,
cerebellum, and pontine region (26). and seizures (32). Cataplexy occurs late in the disease course (33).
EEG findings in sialidosis types I and II are indistinguishable Niemann–Pick type D (NPD-D) occurs in French Canadian
(27). In type I, the background is low voltage and rhythmic patients who share a common founder mutation (34).
spikes are observed over the vertex, which increase in frequency In late childhood or adult-onset NPD-C, the EEG may reveal
during sleep (27). Spike-wave discharges precede myoclonic generalized slowing (Fig. 15.1) (35). Late into the course of
jerking in both types I and II (28,29). VEPs demonstrate low NPD-C, abnormally high-voltage and diffuse alpha activity is
amplitudes and prolonged latencies in type I (28), while SEPs seen and is enhanced by intermittent photic stimulation. In a
exhibit giant potentials in both conditions (28,29). patient with myoclonus, EEG–EMG frequency analysis demon-
strated a cortical origin for the myoclonus (36).
Niemann–Pick Disease
Niemann–Pick disease occurs when a deficiency of the enzyme Gaucher Disease
acid sphingomyelinase (ASM) causes accumulation of sphin- Gaucher disease is the most common lysosomal storage
gomyelin within cells of the monocyte–macrophage system (30). disorder. It results from a deficiency of the lysosomal enzyme
Four phenotypic subtypes exist. In Niemann–Pick disease type A beta-glucocerebrosidase, resulting in accumulation of

Figure 15.1 EEG in a 10-year-old boy with Niemann–Pick type C. Note the frontal slowing with intermixed sharp
waves bilaterally.
284 Part III ■ Clinical EEG: General

glucocerebroside in the mononuclear phagocyte system (37). slowing of the background activity (48). As the clinical severity
The characteristic features include hepatosplenomegaly, bony of the disease progresses, so does the degree of slowing. Later
disease, and neurologic decline. Three phenotypic subtypes into the course of the illness, as the patients develop epileptic
exist. Type I has minimal if any neurologic impairment. Bulbar seizures, focal spike waves appear. In the final stages of the dis-
symptoms predominate in type II patients (38). In type III ease course, hypersynchronous activity is seen (49). Both the
patients, ocular motor apraxia is often the initial finding. clinical and EEG features of the disease can improve following
The pathologic hallmark of the disorder is the Gaucher cell, bone marrow transplantation (50).
which is an enlarged macrophage with cytoplasmic linear inclu-
sions resulting from excessive lysosomal accumulation of gluco- Globoid Leukodystrophy (Krabbe Disease)
cerebroside (38). Enzyme replacement therapy with recombinant Krabbe disease (KD) results from a deficiency of the lysosomal
glucocerebrosidase (Cerazyme) is the mainstay of treatment in enzyme galactosylceramidase. Severe loss of myelin occurs and
Gaucher disease (39), but is not effective for the neurologic globoid cells are seen in the CNS white matter. Early infantile
symptoms since it does not cross the blood–brain barrier. KD accounts for 90% of all cases and presents in the first 6
MRI in type II disease is often normal, but cerebral atrophy is months of life (51) with irritability and rapidly progressive
commonly observed in type III patients (40). BAERs and SSEPs rigidity and tonic spasms. MRI of the brain shows symmetric
can demonstrate abnormalities, even when no lesions are signal abnormalities in the periventricular regions of the poste-
detectable by MRI (41). EEGs show rhythmical sharp waves (6 to rior cerebral hemispheres. Peripheral nerves are also affected,
10/sec), polyspikes, and spike-wave discharges in type III patients but no visceromegaly is observed. The remaining 10% of
(42). These features are diffuse, but have a posterior predomi- patients have late-infantile or juvenile KD, which presents with
nance. EEGs are commonly normal in type II patients. Reduced ataxia and later exhibits dystonia and visual failure (52).
amplitude can also be an EEG feature of some patients (41). EEGs are abnormal in a majority of the early infantile cases,
but the opposite is true for late-infantile and juvenile-onset
Fabry Disease patients. Abnormalities consist of slowing without epileptiform
Fabry disease is the only X-linked lysosomal disorder; the oth- activity in 46%, epileptiform activity without slowing in 15%,
ers are inherited in an autosomal recessive fashion. Hence, the and both slowing and epileptiform features in 38% of patients
disease affects men more severely than it does women (43). It (53). Likewise, BAEPs are abnormal in 88% of the early infan-
results from a deficiency of alpha-galactosidase A. This leads to tile patients and only 40% of the late-onset cases. The most fre-
storage of glycosphingolipids in a variety of tissues, producing quent BAEP abnormality is absence of waves III and V (53).
cellular dysfunction, inflammation, and fibrosis. Symptoms
begin in early childhood with peripheral neuropathy and burn- PEROXISOMAL DISORDERS
ing pain of the hands and feet, which can remain disabling
throughout life (43). Hypohydrosis, nausea, and postprandial Peroxisomal disorders are a group of heterogeneous conditions
diarrhea are also seen in childhood. By adulthood, proteinuria that share dysfunction of peroxisomal function. Peroxisomes are
and renal failure develop. Left ventricular hypertrophy and bound by a single membrane and contain a fine granular matrix.
coronary artery disease are also common. Angiokeratoma They are histologically identified by the presence of catalase.
develop on the skin. Neurologically, cryptogenic strokes occur. They are present in all human tissues except erythrocytes. They
Enzyme replacement therapy can clear microvascular endothe- carry out over 40 metabolic functions, including beta-oxidation
lial deposits from the kidneys, heart, and skin, reversing the of very-long-chain fatty acids (VLCFA) and biosynthesis of plas-
pathogenesis (44). MRI scans reveal gray and white matter malogen and cholesterol. Some of those reactions can occur in
lesions. The anterior circulation generally appears normal, but other organelles, but many are exclusive to peroxisomes.
tortuous basilar arteries are common (45). EEGs can reveal
either focal or diffuse slowing (46). X-linked Adrenoleukodystrophy
X-linked adrenoleukodystrophy (X-ALD) is the most common
Metachromatic Leukodystrophy of the peroxisomal disorders. It is characterized by accumula-
Metachromatic leukodystrophy (MLD) is characterized by an tion of VLCFA in the brain and adrenal tissues (54). VLCFA are
inability to degrade sulfated glycolipids due to a deficiency of also detected in fibroblasts, blood cells, and plasma, allowing
the lysosomal enzyme arylsulfatase A. The disorder affects cen- for identification of the condition (55). X-ALD affects the CNS
tral and peripheral myelin. There is widespread loss of myeli- or adrenal glands and can have diverse presentations. The cere-
nated oligodendroglia in the CNS and segmental demyelination bral form presents between 4 and 8 years of age with symptoms
of peripheral nerves (47). Clinically, it presents with motor mimicking ADHD. Psychomotor regression and cortical blind-
regression and ataxia at around 2 years of age (47) and leads to ness follow and seizures are observed late into the disease
a decerebrate state some years after the initial onset of symp- course. MRI shows lesions of the parieto-occipital white matter
toms. Epileptic seizures can occur late into the course of the ill- with contrast enhancement at the leading edge (56). The mean
ness. A late-onset form exists and presents in the third decade interval between the first symptom and an apparent vegetative
with psychiatric symptoms. state is 2.4 years (48). Adrenomyeloneuropathy is a variant that
Earlier into the course of MLD, EEGs are often normal. If affects primarily the spinal cord and does not present until the
abnormalities are seen at presentation, they consist of mild third decade. Since X-ALD is an X-linked condition, it presents
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 285

earlier and with greater severity in males, but heterozygous 3000 or so genes are required to make a mitochondrion reside in
women can be symptomatic. the nuclear DNA. Inheritance patterns are therefore varied with
During early stages of the disease, EEGs are normal in about both maternal and Mendelian patterns observed.
half of patients and show posterior slowing in the remainder. The
location of the slowing corresponds to the region of MRI abnor- Leigh Syndrome
malities (48). As the disease progresses, the slowing becomes In 1951, Leigh described a case of subacute necrotizing
more widespread and paroxysmal discharges are observed (48). encephalomyelopathy in a 7-month-old infant. The patient had
In one reported case, the ictal EEG during a tonic seizure revealed symmetric lesions of the thalamus, midbrain, pons, medulla,
an electrodecrement of the background activity (48). and posterior columns of the spinal cord (63). Leigh syndrome
results from defects in cytochrome oxidase or the pyruvate dehy-
Zellweger Syndrome drogenase complex. Inheritance patterns can be maternal, X-
Zellweger syndrome (ZWS), also known as cerebrohepatorenal linked, or autosomal recessive. Patients presenting prior to 12
syndrome, results from deficiencies of VLCFA beta-oxidation, months are likely to exhibit psychomotor retardation, vomiting,
phytanic acid oxidation, and plasmalogen synthesis. As a result, weight loss, and weakness. In patients presenting after 18
plasma levels of VLCFAs and phytanic acid are high, whereas months, symptoms include coma, ophthalmopareis, or nystag-
erythrocyte concentrations of plasmalogen are reduced. ZWS mus (64). Neuroimaging reveals abnormal signal in the basal
presents in infancy with craniofacial dysmorphism (high fore- ganglia and brain stem with sparing of the mamillary bodies.
head, large anterior fontanelle, separated cranial sutures, There is a strong predilection for the putamen; absence of puta-
hypoplastic supraorbital ridges, and epicanthal folds) and minal involvement should call the diagnosis into question (65).
hepatomegaly (57). Neurologic findings include hypotonia The interictal EEG can reveal focal or multifocal epilepti-
with absent reflexes, profound developmental delays, seizures, form activity, which becomes more frequent during sleep.
and impaired vision and hearing (57). MRI reveals both corti- During partial seizures, the ictal EEG reveals posterior or hemi-
cal (polymicrogyria and pachygyria) and white matter (delayed spheric background attenuation or ictal fast activity (66).
myelination) abnormalities (58).
Interictal EEGs in ZWS show bilateral independent multifo- MELAS Syndrome (Mitochondrial Myopathy,
cal spike waves, predominantly in the frontal motor cortex (59). Encephalopathy, Lactic Acidosis, and Stroke)
Continuous negative vertex sharp and spike waves are com- MELAS is characterized by mitochondrial myopathy,
monly observed. They were found in 9 of 11 patients in one encephalopathy, lactic acidosis, and recurrent stroke-like
study (60) and seen in a single case in a separate report (61). episodes. Many patients also exhibit seizures, migraines, corti-
This finding, observed during wakefulness and sleep, is pathog- cal blindness, short stature, or ragged-red fibers on muscle
nomonic for ZWS (61). BAEPs and VEPs can have absent or biopsy (67). The mechanism of stroke in this condition remains
delayed responses (60). controversial. Some implicate a mitochondrial angiopathy,
resulting in small vessel occlusion, and others postulate that the
Neonatal Adrenoleukodystrophy impairment of oxidative metabolism results in lactic acidosis
Neonatal adrenoleukodystrophy (NALD) is a disorder of perox- and brain injury (68). Imaging studies support the later
ysomal biosynthesis resulting from impaired ATPases that are hypothesis (68,69). MRI abnormalities are most pronounced in
necessary for importing proteins into peroxisomes. NALD pres- the parietal–occipital regions (69).
ents at birth with facial dysmorphism (midface hypoplasia) Likewise, interictal EEGs reveal diffuse sharp waves with an
(57). Hepatomegaly and adrenal cortical atrophy can also be occipital predominance. The epileptiform abnormalities can be
observed. Neurologically, patients present with hypotonia, optic enhanced with photic stimulation (66). PLEDs can also be seen
nerve atrophy, and seizures (62). Psychomotor regression over the affected occipital lobe (70). In one report of 11 patients
occurs in early childhood. MRI reveals a severe deficiency of with a syndrome of overlapping MERRF–MELAS phenotype,
white matter in addition to heterotopias and polymicrogyria. EEG abnormalities correlated with the severity of the clinical
Interictal EEGs show high-voltage slow waves and bilateral phenotype. In milder cases, they included disorganization of
independent multifocal spike waves. Later in the disease course, the background activity and focal or generalized delta–theta
background suppression can be observed (59). waves (Fig. 15.2). More severely affected patients exhibited
bursts of generalized spike or polyspike and slow-wave com-
MITOCHONDRIAL DISORDERS plexes (71). MELAS patients can develop epilepsia partialis con-
tinua. EEGs at those times reveal pseudoperiodic spike waves in
Mitochondrial diseases are a heterogeneous group of conditions the contralateral temporal–occipital regions (72).
that result from dysfunction of the mitochondrial respiratory
chain. The mitochondria’s main function is generating ATP to MERRF Syndrome (Myoclonus, Epilepsy,
power the cells they reside in. Because mitochondria are present Ragged-red Fibers)
in every cell type, with the sole exception of erythrocytes, mito- MERRF is characterized by myoclonic epilepsy and myopathy
chondrial dysfunction can have wide-ranging effects. The (with ragged-red fibers on muscle biopsy). Dementia, ataxia,
mitochondrial genome contains 37 genes, of which 13 encode and generalized tonic–clonic seizures are also observed. It
structural proteins for the respiratory chain. The remaining results from an A-to-G mutation at nucleotide 8344 in the
286 Part III ■ Clinical EEG: General

Figure 15.2 EEG in a 13-year-old boy with MELAS syndrome and seizures. Note the continuous semirhythmic right
occipital slowing.

mitochondrial DNA (73). The mutation impairs mitochondrial generalized, but the occipital regions are most commonly
protein synthesis and thus impairs oxidative phosphorylation. involved (75). Periodic (approximately 1 Hz) epileptiform dis-
MERRF is transmitted by maternal inheritance, but the clinical charges can be seen (76) (Fig. 15.3) and high-amplitude slow-
phenotype varies, even within the same family. ing with polyspikes is characteristic for the condition (77).
In one series, EEG abnormalities were seen in six of nine
patients. They included slowing of the background activity and PROGRESSIVE MYOCLONIC EPILEPSIES
generalized epileptiform discharges (15). Polyspike- and spike-
and slow-wave complexes are enhanced by photic stimulation The progressive myoclonic epilepsies are a group of unrelated
and may occur in conjunction with myoclonic jerking (66). disorders that share myoclonic epilepsy in common.
Generalized tonic–clonic, atypical absence, and simple and
Alpers–Huttenlocher Syndrome (Diffuse complex partial seizures can be seen in these conditions. They
Degeneration of Cerebral Gray Matter With are generally refractory to medical management and worsen
Hepatic Cirrhosis) over time. Cognitive impairment and ataxia are common
In 1931, Alpers reported the clinical features and neuropathology among these conditions. MERRF is usually included among the
of a 4-month-old girl with intractable generalized epilepsy (74). progressive myoclonic epilepsies, but is discussed in the section
He termed the condition “diffuse progressive degeneration of the “Mitochondrial Disorders.”
gray matter of the cerebrum.” The neuropathology is character-
ized by extensive brain atrophy with loss of cerebral neurons. Lafora Disease
Psychomotor retardation and intractable epilepsy are universally Lafora disease (LD) generally presents between 12 and 17
seen. Liver failure is common but sometimes absent. The syn- years of age (78). Generalized tonic–clonic seizures are the
drome is caused by mutations of the nuclear gene encoding the presenting symptom in 71% (15). Cognitive decline, visual
catalytic subunit of mitochondrial polymerase gamma (POLG). impairment, occipital seizures, and myoclonic seizures are
also observed. The occipital seizures can cause transient
Mitochondrial DNA Depletion Syndrome blindness, visual hallucinations, or a photoconvulsive
The EEG reveals slow or absent posteriorly dominant rhythms response. Myoclonus worsens as the disease progresses.
(75). Interictal discharges can be focal, multifocal, or Continuous generalized myoclonus can result in early wheel-
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 287

Figure 15.3 EEG in a 10-month-old boy with Alpers–Hunttenlocher syndrome. Note the periodic 1-Hz sharp waves
over the right frontal region.

chair dependency. Most patients die from neurologic compli- partial seizures can also increase over the course of the disease.
cations within 10 years of disease onset (78). It is inherited in These symptoms are accompanied by ataxia, tremor, and
an autosomal recessive fashion. The pathologic hallmark of dysarthria. Cognitive dysfunction and visual impairment are
LD is the presence of Lafora bodies in neurons, liver, and mus- absent or mild (15). Although the condition clinically resembles
cle. Lafora bodies stain positive with periodic-acid Schiff and LD, Lafora bodies are not seen in ULD. It is inherited in an
have a dense core with a less dense periphery. EEGs in LD autosomal recessive fashion and results from an unstable
reveal background slowing in 97.4% of patients and general- expansion of a 12-nucleotide repeat within the CSTB gene on
ized epileptiform discharges in 84.2% (15). Focal and multifo- chromosome 21q22.3 (81).
cal discharges can be observed. Photosensitivity with a fast The EEG reveals generalized epileptiform discharges (15)
stimulus frequency is seen in 25%. EEG changes can precede with a frequency of 3 to 5/sec. Photic stimulation elicits gener-
clinical symptoms by up to 6 years (79). As the disease pro- alized spikes and polyspikes. As the disease progresses, so does
gresses, the background rhythm slows and the alpha rhythm the frequency of the epileptic discharges (80).
and sleep morphologies are lost (78). Generalized irregular
spike-wave discharges with an occipital predominance domi- Severe Myoclonic Epilepsy of Infancy
nate the background activity. Unlike many primary general- (Dravet Syndrome)
ized epilepsies, epileptiform discharges diminish during sleep Severe myoclonic epilepsy of infancy (SMEI) presents in the
in LD. In one case series, giant SSEPs were seen in 24 of first year of life with febrile seizures (82). The seizures are gen-
31 patients and VEPs revealed prolonged or absent waveforms eralized or focal. Status epilepticus in the setting of fever is
in 12 of 31 patients (15). common (82). The early age of onset helps distinguish it from
other forms of progressive myoclonic epilepsy. It is character-
Unverricht–Lundberg Disease ized by generalized tonic, clonic, tonic–clonic, absence, or
Unverricht–Lundberg disease (ULD) presents between 6 and 15 myoclonic seizures, which are often refractory to medications.
years of age with stimulus-sensitive myoclonus or generalized A ketogenic diet provides benefit in some patients (82). Ataxia
tonic–clonic seizures (80). The myoclonus may be focal, multi- and psychomotor regression are common, although cognitive
focal, or generalized. It progresses over the course of the illness development is normal prior to 1 year. The condition results
and can be disabling. Tonic–clonic, absence, and complex from a mutation of the sodium channel gene, SCN1A, which
288 Part III ■ Clinical EEG: General

also causes generalized epilepsy with febrile seizures plus in the first days of life. All of the infants had urine with the
(GEFS ) syndrome (83). SMEI may therefore represent a vari- smell of maple syrup (87). The condition, now termed maple
ant of GEFS . syrup urine disease, is caused by deficiency of branched chain
During the first 2 years of life, EEGs are normal (82). After alpha-ketoacid dehydrogenase complex. This results in an ele-
the second year, generalized spike and polyspike waves are vation of branched chain amino acids in the blood and urine.
observed (82). The discharges increase in frequency during Five clinical phenotypes of the condition exist (88). In the clas-
sleep. Photic stimulation elicits discharges in about one fourth sic form, symptoms develop in the first week of life. Seizures are
of patients (82). common and coma and death can result if not treated early.
Initially, neuroimaging is normal, but does reveal generalized
AMINOACIDEMIAS AND edema in severe cases (89).
ORGANIC ACIDEMIAS During acute metabolic decompensation, EEG reveals
spikes, polyspikes, spike-wave complexes, triphasic waves,
Amino and organic acidemias are conditions that cause an severe slowing, and bursts of periodic suppression (90). A char-
abnormal build-up of amino acids in the blood. acteristic comb-like pattern of rolandic sharp waves is seen on
Aminoacidemias occur when there are elevated blood levels of EEG between the second and third weeks and may persist after
any amino acid, whereas organic acidemias refer specifically to the acute metabolic decompensation has passed (90). Spike-
elevated levels of branched chain amino acids. These conditions wave discharges are seen with photic stimuli in just over half of
are generally diagnosed in infancy. They present with acute patients (90).
metabolic decompensation, during which somnolence, hypoto-
nia, vomiting, and seizures are observed. Multisystemic involve- Homocystinuria
ment is common. Untreated, these conditions can result in Homocystinuria is a metabolic disorder caused by a cystathio-
coma and death. Treatment can include hemofiltration, dietary nine [beta]-synthase gene deficiency, resulting in increased
restriction of particular nutrients or a class of nutrients, and serum levels of homocysteine and methionine. Patients have
specific supplements designed to remove toxic metabolites. skeletal changes resembling Marfan syndrome. Cognitive
Late-onset forms also exist and may present without overt delays, psychiatric disturbances, strokes, and extrapyramidal
metabolic crisis. These conditions are diagnosed by chromatog- findings can result. Seizures occur in 21% of patients and are
raphy or mass spectroscopy. Laboratory investigations are best usually generalized tonic–clonic (91).
performed during acute episodes. Del Giudice et al. (92) reported EEG findings in 19 patients.
Ten of those patients had abnormalities, which generally con-
Phenylketonuria sisted of background slowing. Mid-temporal paroxysmal activ-
Phenylketonuria results from deficient activity of phenylalanine ity was seen in two patients. Centrotemporal spikes, increasing
hydroxylase (PAH), the enzyme that converts phenylalanine to in drowsiness and sleep, can occur and may mimic benign
tyrosine. As a result, phenylalanine accumulates and is excreted childhood epilepsy with centrotemporal spikes (BECTS) (93).
in the urine in large amounts as phenylketones. Phenylalanine
is neurotoxic and untreated patients develop mental retarda- Propionic Acidemia
tion. Restricting phenylalanine in the diet can normalize Propionic acidemia results from a deficiency of the mitochon-
plasma phenylalanine levels and improve outcome. If instituted drial enzyme propionyl-CoA carboxylase, which converts pro-
in infancy, before the patient is symptomatic, mental retarda- pionyl-CoA to D-methylmalonyl CoA. Biotin is a cofactor for
tion can be averted. For this reason, newborn screening has this reaction. Most patients present as neonates with an acute
been mandatory since the 1960s. metabolic crisis with hypotonia, lethargy, or seizures. The
Some infants exhibit EEG abnormalities at the time of diag- seizures may result from NMDA receptor-mediated mecha-
nosis, consisting of focal or multifocal spike and sharp waves nisms. Later onset patients present with developmental delays
(84). Many of those same patients will have normal EEGs 1 year without metabolic decompensation, acute encephalopathy, or
after initiating dietary restriction of phenylalanine. Overall, episodic ketoacidosis (94). Acute metabolic crises can be treated
patients who begin treatment sooner are more likely to have with total protein restriction and hemofiltration (peritoneal
normal EEGs (85). Some patients with EEG abnormalities in dialysis and exchange transfusion). Carnitine supplementation
infancy will develop abnormalities later in childhood, even and antibiotics (to reduce intestinal production of propionic
though they remain on a restricted diet. EEG abnormalities in acid by normal gut flora) are also used. Sass et al. (94) reported
such patients are indicative of higher phenylalanine levels (gen- EEG findings in 32 patients. Of those, nine showed moderate
erally greater than 20 mg/dL) (86). Older patients and those who abnormalities and six were clearly abnormal. The remaining 17
initiate treatment later in childhood are more likely to exhibit patients had normal EEGs. A separate case report (95) revealed
abnormalities on EEG (86). Focal epileptic discharges are the disorganized background activity at the time of acute metabolic
most common abnormality observed (86). decompensation.

Maple Syrup Urine Disease Methylmalonic Acidemia


In 1954, Menkes described four siblings with feeding difficulty, Methylmalonic acidemia (MMA) is a heterogeneous group of
irregular respirations, hypertonia, and opisthotonus presenting conditions characterized by accumulation of methylmalonic
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 289

acid and its byproducts. It results from impairments of methyl- seen with a combined regimen of dextromethorphan and
malonate or cobalamin (vitamin B12) metabolism. Hence, sodium benzoate (103).
some patients respond to cobalamin supplementation and oth-
ers do not. Patients with defects of cobalamin metabolism have Hartnup Disease
simultaneous homocystinuria. Hartnup disease is an autosomal dominant disorder of amino
MMA typically presents in the first weeks of life with vomit- acid transport within the renal tubules and intestinal tract. It
ing, lethargy, hypotonia, and seizures. Later onset cases also results in malabsorption of neutral monoamino monocar-
exist and may present with hypoglycemia, myopathy, or throm- boxylic amino acids and aminoaciduria. Symptoms include pel-
bosis (due to homocystinuria). In one case report of an infant lagroid dermatitis, photosensitivity, ataxia, and neuropsy-
with MMA and tonic seizures, EEG showed polyspike bursts chiatric abnormalities. EEG can reveal epileptiform features
(96). Another report described sharp transients superimposed (104), but is normal in most cases.
on a low-voltage and discontinuous tracing with long flat peri-
ods. Following treatment with hydroxycobalamin (vitamin Lowe Syndrome (Oculocerebrorenal Syndrome)
B12), blood homocysteine levels declined and a reduction was Lowe syndrome was originally described in 1952 by Lowe et al.
seen in the length of the interburst periods (97). (105), who reported the histories of three unrelated male
infants with congenital cataracts, glaucoma, developmental
Isovaleric Acidemia retardation, hyporeflexia with low musculature, osteopenia,
Isovaleric acidemia is caused by a deficiency of the mitochon- decreased ammonia production, and a “peculiar, high-pitched,
drial enzyme isovaleryl-CoA dehydrogenase, resulting in accu- irritating cry.” Affected children also exhibit hyperactivity,
mulation of isovaleryl-CoA derivatives. It generally presents in pseudotumor cerebri, ophthalmoplegia, or hypoglycemia. The
infancy with poor feeding, vomiting, obtundation, and seizures. condition is X-linked and is caused by mutations of the OCRL1
The affected infants have a characteristic odor, which resembles gene on chromosome Xq26. OCRL1 encodes a protein product
“dirty socks” and is best appreciated in sweat and cerumen. that plays a role in cytoskeletal remodeling and cellular traffick-
Patients presenting after the newborn period exhibit failure to ing. Lowe syndrome has a high mortality rate in the first
thrive, developmental delays, or mental retardation. The initial months from metabolic derangements and metabolic acidosis.
therapeutic approach is to decrease leucine intake and promote Most patients will survive into the second or third decade, but
an anabolic state with increased calories. EEG may reveal low- will develop chronic renal disease. MRI reveals small cyst-like
voltage activity (less than 20 V) with bursts of delta slowing or changes in the periventricular white matter and H-MRS
sharp waves (98). In an experimental animal model, infusion of demonstrates prominent myoinositol peaks indicating gliosis
isovaleric acid causes slowing on EEG, which worsens as the (106,107). EEG shows an irregular, poorly sustained posteriorly
concentration increases (99). dominant rhythm and bursts of high voltage slowing.
Independent bitemporal spike- and slow-wave discharges are
Glycine Encephalopathy (Nonketotic seen (108). Generalized tonic–clonic, tonic, atonic, and com-
Hyperglycinemia) plex partial seizures have been described. In one report of a
Glycine encephalopathy is caused by dysfunction of the mul- patient with atonic seizures, the ictal EEG revealed secondarily
timeric glycine cleavage enzyme system, resulting in accumu- generalized epileptiform discharges (109).
lation of glycine in all body tissues, including the brain. The
disorder presents in the first week of life with apnea, lethargy, INBORN ERRORS OF UREA SYNTHESIS
hypotonia, myoclonus, seizures, and feeding difficulty (100).
Brain imaging is normal in half of cases. When abnormalities Urea cycle disorders occur when there is deficiency of one of the
are seen, they include agenesis of the corpus callosum, pro- enzymes in the cycle that converts nitrogen to urea. There are
gressive atrophy, or delayed myelination (100). Diagnosis is six enzymes involved: carbamyl phosphate synthetase I (CPSI),
confirmed by the detection of elevated cerebral spinal fluid ornithine transcarbamylase (OTC), arginosuccinate synthetase
(CSF) glycine levels and an elevation of the CSF-to-plasma (ASS), argininosuccinate lyase (ASL), N-acetyl glutamate syn-
glycine ratio (101). Classic neonatal-onset patients have ratios thetase (NAGS), and arginase. Dysfunction of any of the
greater than 0.2, but atypical, later onset cases also exist, and enzymes, with the exception of arginase, results in hyperam-
have ratios of roughly 0.09 (101). Atypical presentations can monemia and metabolic decompensation in infancy. Newborns
include seizures, developmental delays, and movement disor- appear well in the first 24 to 48 hours of life. After the infant
ders outside of the neonatal period. In classical neonatal pre- begins taking protein from formula or breast milk, vomiting,
sentations, EEG demonstrates a burst suppression pattern or lethargy, irritability, and poor feeding develop (110). Cerebral
hypsarrhythmia (100). In later onset variants, the EEG can edema results in hyperventilation and metabolic alkalosis
reveal generalized spike and slow waves (102). Since glycine (111). Half of the affected patients have seizures.
can stimulate NMDA glutamate receptors, treatment with Patients with partial enzyme deficiencies have later onset
NMDA antagonists (dextromethorphan and ketamine) has presentations. This occurs most commonly in female carries of
been tried. Sodium benzoate decreases CSF glycine levels and OTC, since OTC is an X-linked condition. Late-onset patients
can further improve seizure control and alertness (103). In exhibit developmental delays, seizures, psychiatric disorders,
one case, clinical and electrophysiologic improvements were and chronic vomiting (112). Following high protein loads or
290 Part III ■ Clinical EEG: General

catabolic stress, headache, vomiting, and ataxia develop. The including lack of head control, inability to sit or walk, and poor
diagnosis is suspected when an elevated plasma ammonia level eye contact. Myoclonic seizures occur. Dysmorphism (microg-
is seen in conjunction with a normal blood glucose and a nor- nathia, a prominent forehead, and low-set ears) is sometimes
mal anion gap. The diagnosis is confirmed by performing observed (121). INAD is distinguished from PKAN by the ear-
enzyme analysis from liver, fibroblasts, or red blood cells. lier age of onset and greater pyramidal signs (spasticity and
In one study of four infants with urea cycle disorders hyperreflexia) (122). Like PKAN, however, INAD is associated
(113), all of the patients demonstrated abnormalities on at with high iron levels in the globus pallidus on MRI (123).
least one EEG. The most common findings included multifo- Cerebellar atrophy is also observed. The two conditions are
cal spike- and spike- and slow-wave discharges. Clancy and genetically distinct; INAD is caused by mutations of the
Chung (114) reported burst suppression in cases of neonatal PLA2G6 gene. Death usually occurs by age 10.
citrullinemia. They observed that the length of the EEG inter- Interictal EEG reveals high-voltage fast rhythms (16 to 22
burst interval correlated with serum levels of ammonia, sug- Hz) in sleep and wakefulness. Diffuse slow spike-wave com-
gesting that hyperammonemia may directly contribute to plexes are also observed (124). In a 4-year-old boy with tonic
encephalopathy. Nagata et al. (115) noted that EEG abnor- spasms, the ictal EEG revealed diffuse, high-voltage, irregular
malities were more often observed in patients with CT abnor- sharp and slow-wave complexes followed by desynchronization
malities, and that patients with a normal CT were less likely (124). Visual evoked potentials are abnormal and EMG demon-
to exhibit EEG findings. strates chronic degeneration.

Wilson Disease (Hepatolenticular Degeneration)


DISORDERS OF HEAVY METAL In Wilson disease, a defect of copper transport and excretion
ACCUMULATION leads to copper accumulation in the liver. As cirrhosis develops,
copper leaks into plasma and accumulates in the brain, kidneys,
Neurodegeneration w ith Brain Iron Accumulation and cornea where it results in injury. The earliest pathology in
Neurodegeneration with brain iron accumulation (NBIA) is a Wilson disease occurs in the liver; the initial deposition of
collection of phenotypically similar conditions, which share in hepatic copper is clinically silent.
common brain iron deposition. Included within this heading There is wide variability in the age of disease onset, but the
are pantothenate kinase-associated neurodegeneration mean is 16 years (125). Younger patients tend to present with
(PKAN), infantile neuroaxonal dystrophy (INAD), neurofer- hepatic disease and older patients are more likely to exhibit
ritinopathy, and aceruloplasminemia. Patients present with neuropsychiatric symptoms at presentation (126). In one
posture and gait abnormalities, bradykinesia, and rigidity review of 282 patients (124), 69.1% presented with neurologic
(116). MRI distinguishes these disorders, but all share in com- symptoms. Of those, the predominant features were parkin-
mon a hypointense globus pallidus on T2 sequencing (117). sonism (62.3%), dystonia (35.4%), cerebellar dysfunction
More definitive confirmation can be accomplished through (28%), pyramidal signs (16%), chorea (9%), athetosis (2.2%),
genetic testing. myoclonus (3.4%), and behavioral abnormalities (16%). All of
PKAN is caused by a mutation of the pantothenate kinase 2 the patients with neurologic symptoms had Kayser–Fleischer
gene (PANK2). It is phenotypically classified into classical and rings on ophthalmologic assessment. Serum ceruloplasmin was
atypical forms. Classical PKAN presents around age 3 with decreased in 93% and 24-hour copper excretion was increased
clumsiness and gait abnormalities. Dystonia begins in the lower in 70%. MRI reveals bilateral T2 hyperintensities within the
extremities. Dysarthria, spasticity, visual impairment, and basal ganglia and thalamus as well as mild to moderate atrophy.
behavioral problems follow. Pigmentary retinopathy is com- EEG changes in Wilson disease include a reduction in alpha
mon (116). MRI reveals T2 hyperintense signal surrounded by frequency (17.6%), and increased beta (14.7%), theta (20.5%),
a rim of hypointensity in the globus pallidus, creating the “eye- or delta (2.9%) activity. Focal epileptiform discharges are seen
of-the-tiger” sign, which is pathognomonic for the condition in 11.7% of patients. In one series of 23 patients, only 3 exhib-
(116). Atypical presentations have a later onset and are charac- ited EEG abnormalities (127), while in a literature review of 80
terized by psychiatric symptoms and movement disorders. The patients, over half showed EEG abnormalities (128). In general,
EEG in PKAN is normal in some cases (118,119). Observed the degree of EEG abnormality parallels the severity of clinical
abnormalities can include low voltage slowing and fast activity involvement. The EEG may improve during or after treatment
(119). Swaiman et al. (119) reported a single patient with (128). Prolonged latencies and abnormal waveforms are
seizures and bursts of generalized slowing and sharp waves on observed with visual and auditory evoked potentials.
EEG. Bilateral multifocal spikes and excessive posterior slowing
were also noted. Menkes Disease (Kinky Hair Disease)
Menkes disease (MD) is a fatal X-linked condition caused by a
Infantile Neuroaxonal Dystrophy mutation of the copper-transport gene ATP7A, which is neces-
(Seitelberger Disease) sary for copper excretion from enterocytes. Copper deficiency
INAD is characterized pathologically by axonal swelling and results in impaired function of copper-dependent enzymes.
spheroid bodies in the central nervous system. Onset is between The neurologic symptoms result mainly from deficiency of
6 months and 2 years. Patients present with neuromotor delay cytochrome c oxidase. The clinical features can therefore
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 291

resemble those in Leigh disease. Dysfunction of lysyl oxidase OTHER METABOLIC CONDITIONS
leads to connective tissue abnormalities and predisposes
patients to arterial aneurysms and intracranial hemorrhage. Glut-1 Deficiency Syndrome (De Vivo Syndrome)
Tyrosinase deficiency accounts for hypopigmentation of hair Glut-1 is expressed on vascular endothelial cells and is the
and skin. Patients typically present at 2 to 3 months of age with major transporter of glucose into the central nervous system.
loss of developmental milestones, truncal hypotonia, failure to Dysfunction of Glut-1 results in low CSF glucose levels in the
thrive, temperature instability, and epilepsy. The physical exam- absence of hypoglycemia. Patients present in infancy with pro-
ination is notable for abnormally kinky hair, which is shorter gressive microcephaly, developmental delays, and seizures
and sparser over the sides and back. Skin and hair are hypopig- (133). Atypical forms exhibit ataxia or mental retardation with-
mented. Minor dysmorphology, including sagging cheeks, a out epilepsy.
depressed nasal bridge, and high arched palate are noted. MRI Seizure types include generalized tonic, clonic, myoclonic,
reveals intracranial vessel tortuosity, white matter changes, and atonic, and atypical absence. In infancy, the EEG reveals multi-
ischemic lesions of the deep gray matter (129). Treatment with focal spike-wave discharges, which localize to the temporal and
copper replacement may improve clinical outcomes if started occipital regions. With maturation, generalized 2.5- to 4-Hz
early in the course of the disease (130). spike- and slow-wave discharges are observed (133). The
Bahi-Buisson et al. (131) classified three stages of MD based seizures can worsen with phenobarbital, since it inhibits Glut-1
on seizure types and EEG findings. In early MD, patients function. The ketogenic diet is the only effective therapy since it
develop focal clonic status epilepticus, which is accompanied on allows the brain to utilize ketone bodies as a fuel source.
EEG by slow spike waves in the posterior head regions. Interictal
EEG reveals multifocal polymorphic slow waves. During the Pyridoxine-Dependent Seizures
intermediate stage of the disease, infantile spasms occur and a Pyridoxine-dependent seizures (PDS) are a rare autosomal
modified hypsarrhythmic pattern is seen on EEG. In late-stage recessive condition, presenting with neonatal epilepsy. A variety
MD, patients exhibit multifocal seizures, tonic spasms, and of seizure types, including infantile spasms, is observed. The
myoclonus. EEG reveals multifocal high-amplitude activity seizures are resistant to traditional anticonvulsants, but are well
mixed with irregular slow waves (Fig. 15.4) (131). The degree of controlled by a daily pharmacologic dose of pyridoxine (vitamin
EEG abnormality relates in part to serum copper levels (132). B6). Patients with PDS require pyridoxine supplementation

Figure 15.4 EEG in a 9-month-old boy with Menkes disease. Note the right temporal delta slowing and spike-
and slow-wave discharges.
292 Part III ■ Clinical EEG: General

throughout life. In a related condition, pyridoxine-responsive reveal EEG abnormalities. In one report of 18 patients with SCA,
seizures, patients are able to successfully wean off of pyridoxine temporal theta and delta intrusions were noted (139). Epilepsy
without seizure recurrence. PDS is caused by deficiency of the is common in SCA10, and a case series of 18 patients found EEG
enzyme -aminoadipic semialdehyde ( -AASA) dehydroge- abnormalities in all 18 (140). The most common finding was
nase, which is encoded by the antiquitin gene (ALDH7A1). - slow and disorganized background activity. A case report of a
AASA acts within the lysine catabolism pathway (134). Elevated father and son with complex partial seizures and SCA2 noted
levels of -AASA and pipecolic acid (PA) result, and can serve as temporal epileptiform discharges and slowing in both patients
biomarkers for the condition. (141). Patients with DRPLA also develop epilepsy, and may
In PDS, the interictal EEG is abnormal in both the awake experience complex partial, myoclonic, atypical absence, and
and asleep states. The background is discontinuous with a sup- generalized tonic–clonic seizures. In 63.6% of patients with
pression burst-like pattern. Spike and polyspike waves are seen DRPLA, the EEG shows epileptiform features. Photosensitivity
over the central and temporal regions. Continuous high-voltage was observed in 27.3%, and those patients with a photoparoxys-
rhythmic delta slowing occurs (135). Administration of pyri- mal response also had progressive myoclonic epilepsy (142).
doxine (100 to 500 mg intravenously) results in rapid normal-
ization of the EEG, but should be given under close supervision Friedreich Ataxia
since it may result in apnea and bradycardia. Folinic acid sup- Friedreich ataxia (FRDA1) is an autosomal recessive trinu-
plementation and lysine restriction may also play a role in treat- cleotide repeat disorder of the frataxin gene on chromosome
ment of PDS. 9p. The frataxin gene acts to regulate mitochondrial iron con-
tent. Iron overload results in damage to respiratory chain com-
Folinic Acid-Responsive Seizures plexes I, II, and III through generation of oxygen free radicals
Folinic acid-responsive seizures present with myoclonic, clonic, (143). FRDA1 therefore has features of a mitochondrial disor-
or subtle (apnea and irritability) seizures in infancy. The condi- der. It presents prior to adolescence with progressive cerebellar
tion is confirmed by two characteristic peaks of CSF dysfunction and hypoactive knee and ankle reflexes. Patients
monoamine metabolic analysis. The identity of the peaks also exhibit dysarthria, nystagmus, impaired position and
remains unknown. MRI reveals white matter abnormalities and vibratory sensation, and a Babinski sign. Cardiac dysfunction
atrophy. EEG demonstrates a discontinuous background pat- (subaortic stenosis and hypertrophic cardiomyopathy) and dia-
tern with multifocal spike or sharp waves (135). In one report betes are common medical complications and one half of
of two patients with folinic acid-responsive seizures, both were patients die of heart failure (144).
found to have increased CSF -AASA and PA (137). Those No qualitative EEG pattern is seen in FRDA1 (144). EEG also
patients were subsequently found to have mutations of the fails to provide prognostic information, since there is no rela-
ALDH7A1 gene, indicating that folinic acid-responsive seizures tionship between the degree of EEG abnormality and the sever-
and PDS may be the same condition. ity of the disease.

CEREBELLAR DISORDERS GENETIC CONDITIONS


Spinocerebellar Ataxias Rett Syndrome
Spinocerebellar ataxias (SCAs) are a heterogeneous group of The clinical features of Rett syndrome were defined by a con-
progressive cerebellar syndromes with varying combinations of sensus panel held in 2001 (146). Necessary criteria for the diag-
ataxia, dysmetria, and oculomotor deficits (138). Additional nosis include female sex, normal development through the first
clinical findings include pigmentary retinopathy, extrapyrami- 6 months, deceleration of head growth, psychomotor regres-
dal movement disorders (parkinsonism, dystonia, and chorea), sion, loss of purposeful hand movements, and hand-wringing
pyramidal signs, dementia, seizures, and peripheral neuropathy stereotypies. Pathologically, Rett syndrome is characterized by
(138). MRI reveals cerebellar, olivopontocerebellar, or global reduced dendritic arborization. Seventy percent of patients
brain atrophy. The SCAs are genetically heterogeneous autoso- who meet clinical criteria carry a mutation of the X-linked
mal dominant conditions. A gene-based classification scheme is methyl CpG-binding protein (MECP2) gene (147), which is
used, in which the numbering system represents the order of critical for postnatal neuronal maturation. The X-linked cyclin-
gene discovery (SCA 1 to 8, SCA 10 to 23, and SCA 25). Genes dependent kinase-like 5 (CDKL5) gene can also result in a Rett
for fibroblast growth factor 14 (FGF14) and dentatorubral-pal- syndrome (RTT)-like phenotype. Patients with CDKL5 muta-
lidoluysian atrophy (DRPLA) are also included within the fam- tions are less likely to exhibit normal development in the first
ily of SCAs. Many forms result from trinucleotide repeat year and more likely to have early onset intractable epilepsy,
disorders. Anticipation, progressively worsening phenotypes of infantile spasms, and severe hypotonia (148).
subsequent generations within the same family, is observed. EEG criteria for Rett syndrome have been proposed (149,150)
The age of onset and response to symptomatic treatment differ and match the clinical stages of the disease (151). Stage I occurs
among the various types of SCA (138). between 6 and 18 months of age and is characterized by develop-
EEG findings in SCA vary among the different types. Variants mental arrest and decelerating head growth. The EEG is normal
that are commonly associated with epilepsy are more likely to or slightly slow and epileptiform activity is absent. In stage II, the
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 293

patient exhibits further developmental deterioration with loss of chromosome. Five percent to 10% of cases result from a muta-
hand skills, severe dementia, and seizures. On EEG, there is slow- tion of the maternal UBE3A gene. Three percent to 5% have an
ing or loss of the occipitally dominant rhythm and central spike abnormality of DNA methylation in the maternal copy of the
waves are seen during sleep (Fig. 15.5). During stage III, patients gene. Two percent to 3% of patients inherited both copies of
stabilize, but continue to have psychomotor delay and epilepsy. chromosome 15 from the father and none from the mother
The EEG reveals multifocal epileptiform discharges and general- and, therefore, have no functional copy of UBE3A from the
ized spike and slow waves with an absence of the posteriorly mother (152). Abnormalities of the same region on 15q11-q13
dominant rhythm. Stage IV occurs after 8 years of age and also result in Prader–Willi syndrome. The phenotype is deter-
involves decreasing mobility and loss of ambulation. Seizures mined by the parental origin of the chromosome 15 defect with
become less prominent. The EEG may reveal continuous, gener- maternal abnormalities resulting in Angelman syndrome and
alized, slow spike-wave activity in sleep. Rhythmic frontal theta paternal abnormalities leading to Prader–Willi. Hence, these
activity can be seen in wakefulness and drowsiness. conditions illustrate the phenomenon of genomic imprinting.
CDKL5 patients more commonly have normal interictal Ninety percent of patients with Angelman syndrome have
EEGs. In a study of 20 CDKL5 patients, 15 had a normal inter- seizures. Epilepsy is generally more severe in patients with a
ictal EEGs at onset and the other 5 revealed only background deletion of the 15q11-q13 region, which includes encoding
slowing (148). In CDKL5 patients with intractable seizures, information for GABAa receptor subunits. Epilepsy begins
EEG abnormalities include high-amplitude delta waves with between 1 and 3 years in most patients. Seizure types include
bursts of pseudoperiodic high-amplitude spikes, polyspikes, or epileptic spasms, atypical absence, myoclonic, tonic, atonic, and
spike and slow waves (148). tonic–clonic. Multiple seizure types occur in approximately half
of patients with a 15q11-q13 deletion. The EEG resembles hyp-
Angelman Syndrome sarrhythmia and demonstrates runs of rhythmic 2- to 3-Hz
Angelman syndrome is characterized by developmental delay, high-amplitude activity (greater than 300 mV) over the frontal
speech and motor impairment, ataxia, epilepsy, and an appar- regions. Multifocal spikes or sharp waves can be intermixed
ently happy demeanor (152). It is caused by a lack of expression (153). This pattern is distinguished from hypsarrhythmia by a
of the UBE3A gene resulting from abnormalities of chromo- lack of fragmentation during sleep. Rhythmic runs of slow
some 15q11-q13. Approximately 70% of Angelman syndrome spike and slow waves occur and may indicate nonconvulsive
cases result from a de novo 15q11-q13 deletion of the maternal status epilepticus (152) (Fig. 15.6). This pattern can mimic the

Figure 15.5 EEG in a 3-year-old girl with Rett syndrome. Note the bilateral central spike waves.
294 Part III ■ Clinical EEG: General

Figure 15.6 EEG in a 3-year-old boy with Angelman syndrome. Note the continuous, rhythmic, bifrontally predominant
delta slowing with intermixed sharp features.

changes seen in Lennox–Gastaut syndrome, but the clinical decline, termed senile myoclonic epilepsy, occurs late into the
phenotype helps distinguish between these conditions. Tremor condition. Myoclonus is most common in the early morning
is common and correlates with 4- to 10-Hz electromyographic hours. The EEG reveals generalized spike-wave discharges asso-
bursts, but no EEG changes are seen, indicating that the tremor ciated with bilateral myoclonic jerks, whereas segmental jerks
is not due to seizure activity (154). Likewise, bouts of laughter can occur without accompanying EEG changes (157). Spectral
do not have any accompanying EEG changes and are unlikely to analysis of EEG reveals reduced alpha power in the posterior
represent gelastic seizures (152). cortical regions and diffuse EEG slowing (158).

Dow n Syndrome (Trisomy 21)


Down syndrome is a chromosomal disorder caused by all or REFERENCES
part of an extra 21st chromosome. It is characterized by intel- 1. Fukumizu M, Yoshikawa H, Takashima S, et al. Tay Sachs disease:
lectual impairment, short stature, and facial dysmorphism. progression of changes on neuroimaging in four cases.
Hypotonia is seen on the neurologic examination. Older Neuroradiology. 1992;34:483–486.
patients develop Alzheimer-type dementia. Other medical fea- 2. Pampiglione G, Harden A. Neurophysiological investigations in
tures include congenital heart disease, obstructive sleep apnea, GM1 and GM2 gangliosidoses. Neuropediatrics. 1984;15:74–84.
frequent bouts of otitis media, and thyroid dysfunction. 3. Kellaway P, Petersen I, eds. Clinical Electroencephalography in
Epileptic seizures are more prevalent in patients with Down Children. New York and London: Grune and Stratton; 1968:287–306.
syndrome than in the general population. All major seizure 4. Cooperhaver RM, Goodman G. The electroretinogram in infan-
tile, late infantile, and juvenile amaurotic familial infancy. Arch
types have been reported, but infantile spasms are particularly
Ophthalmol. 1960;63:559–566.
common (155). The EEG shows hypsarrhythmia, which can 5. Sakuraba H, Itoh K, Shimmoto M, et al. GM2 gangliosidosis AB
resolve following administration of diazepam. Many patients go variant: clinical and biochemical studies of a Japanese patient.
on to develop other seizure types following infantile spasms, Neurology. 1999;52:372.
including myoclonic, atonic, tonic–clonic, or absence (156). 6. Zaroff CM, Neudorfer O, Morrison C, et al. Neuropsychological
Older Down syndrome patients have increased risks of epilepsy assessment of patients with late onset GM2 gangliosidosis.
and dementia. Myoclonic epilepsy associated with cognitive Neurology. 2004;62:2283.
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 295

7. Harden A, Martinovic Z, Pampiglione G. Neurophysiological 30. Wasserstein MP, Aron A, Brodie SE, et al. Acid sphingomyelinase
studies in GM1 gangliosidosis. Ital J Neurol Sci. 1982;3:201–206. deficiency: prevalence and characterization of an intermediate
8. Cooper JD. Progress towards understanding the neurobiology of phenotype of Niemann–Pick disease. J Pediatr. 2006;149:554–559.
Batten disease or neuronal ceroid lipofuscinosis. Curr Opin Neurol. 31. Kolodney EH. Niemann–Pick disease. Curr Opin Hematol.
2003;16:121. 2000;7:48.
9. Vinken PJ, Bruyn GW, eds. Handbook of Neurology. Vol 10. 32. Vanier MT, Millat G. Niemann–Pick disease type C. Clin Genet.
Amsterdam: North Holland Publishers; 1970:588–679. 2003;64:269–281.
10. Wisniewski K. Neuronal ceroid-lipofuscinoses. Gene Rev. 2006: 33. Vankova J, Stepanova I, Jech R, et al. Sleep disturbances and
1–26. hypocretin deficiency in Niemann–Pick disease type C. Sleep.
11. Groebel HH, Mole SE, Lake BD, eds. The Neuronal Ceroid 2003;15:427–430.
Lipofuscinoses (Batten Disease). Amsterdam: IOS Press; 1999:16–36. 34. Greer WL, Riddell DC, Murty S, et al. Linkage disequilibrium
12. Santavuori P, Lauronen L, Kirveskari E, et al. Neuronal ceroid lipo- mapping of the Nova Scotia variant of Niemann–Pick disease. Clin
fuscinoses in childhood. J Neurol Sci. 2000;21:s35–s41. Genet. 1999;55:248–255.
13. Groebel HH, Mole SE, Lake BD, eds. The Neuronal Ceroid 35. Sevin M, Lesca G, Baumann N, et al. The adult form of
Lipofuscinoses (Batten Disease). Amsterdam: IOS Press; 1999:37–54. Niemann–Pick disease type C. Brain. 2007;130:120–133.
14. Goebel H, Wisniewski KE. Current state of clinical and morpho- 36. Canafoglia L, Bugiani M, Uziel G, et al. Rhythmic cortical myoclonus
logical features of human NCL. Brain Pathol. 2004;14:61–69. in Niemann–Pick disease type C. Mov Disord. 2006;21:1453–1436.
15. Sinha S, Satishchandra P, Gayathri N, et al. Progressive myoclonic 37. Chen M, Wang J. Gaucher disease: review of the literature. Arch
epilepsy: a clinical, electrophysiological and pathological study Pathol Lab Med. 2008;132:851–853.
from South India. J Neurol Sci. 2007;252:16–23. 38. Zhao H, Grabowski GA. Gaucher disease: perspectives on a proto-
16. Muenzer J. The mucopolysaccharidoses: a heterogeneous group of type lysosomal disease. Cell Mol Life Sci. 2002;59:694–707.
disorders with variable pediatric presentations. J Pediatr. 2004; 39. Brady RO. Enzyme replacement for lysosomal diseases. Annu Rev
I44:s27–s34. Med. 2006;57:283–296.
17. Ruijter GJG, Valstar MJ, van de Kamp JM, et al. Clinical and 40. Chang YC, Huang CC, Chen CY, et al. MRI in acute neuropathic
genetic spectrum of Sanfilippo type C (MPS IIIC) disease in the Gaucher’s disease. Neuroradiology. 2000;42:48–50.
Netherlands. Mol Genet Metab. 2008;93:104–111. 41. Miyata R, Watanabe A, Hasegawa T, et al. Neurophysiological
18. Husain AM, Escolar ML, Kurtzberg J. Neurological assessment analysis in an 18-month-old girl with Gaucher’s disease type 2. No
of mucopolysaccharidosis III. Clin Neurophysiol. 2006;117: To Hattatsu. 2006;38:289–293.
2059–2063. 42. Nishimura R, Omos-Lau N, Ajmone-Marsan C, et al.
19. Kriel RL, Hauser WA, Sung JH, et al. Arch Neurol. 1978;35:838–843. Electroencephalographic findings in Gaucher disease. Neurology.
20. Tolar J, Gewal SS, Bjoraker KJ, et al. Combination of enzyme 1980;30:152–159.
replacement and hematopoietic stem cell transplantation as therapy 43. Zarate YA, Hopkin RJ. Fabry’s disease. Lancet. 2008;372:1427–1435.
for Hurler syndrome. Bone Marrow Transplant. 2008;41:531–535. 44. Eng CM, Guffon N, Wilcox WR. Safety and efficacy of recombi-
21. Wrait JE, Clarke LA, Beck M, et al. Enzyme replacement therapy nant human alpha-galactosidase A-replacement therapy in Fabry’s
for mucopolysaccharidosis I: a randomized, double-blinded, disease. N Engl J Med. 2001;345:9–16.
placebo-controlled, multinational study of recombinant human 45. Low M, Nicholls K, Turbridy N, et al. Neurology of Fabry’s disease.
alpha-L-iduronidase (laronidase). J Pediatr. 2004;144:581–588. Int Med J. 2007;37:436–447.
22. Lowden JA, O’Brien JS. Sialidosis: a review of human neu- 46. Lou HOC, Reske-Nielsen E. The central nervous system in Fabry’s
raminidase deficiency. Am J Hum Genet. 1979;31:1–18. disease. Arch Neurol. 1971;25:351–359.
23. Thomas PK, Abrams JD, Swallow D, et al. Sialidosis type I: cherry- 47. Gieselmann V, Franken S, Klein D, et al. Metachromatic leukodys-
red spot myoclonus syndrome with sialidase deficiency and altered trophy: consequences of sulphatide accumulation. Acta Paediatr
electrophoretic mobilities of some enzymes known to be glyco- Suppl. 2003;443:74–79.
proteins. J Neurol Neurosurg Psychiat. 1979;42:873–880. 48. Wang PJ, Hwu WL, Shen YZ. Epileptic seizures and electroen-
24. Swallow DM, Evans L, Stewart G, et al. Sialidosis type I: cherry red cephalographic evolution in genetic leukodystrophies. J Clin
spot-myoclonus syndrome with sialidase deficiency and altered Neurophysiol. 2001;18:25–32.
electrophoretic mobility of some enzymes known to be glycopro- 49. Eggers C, Lederer H, Scheffner D. EEG changes in the course of
teins. Ann Hum Genet. 1979;43:27–35. progressive cerebral disorders in children. Monatsschr Kinderheilkd.
25. Kelly TE, Graetz G. Isolated acid neuraminidase deficiency: a dis- 1977;125:8–15.
tinct lysosomal storage disease. Am J Med Genet. 1977;1:31–46. 50. Solders G, Celsing G, Hagenfeldt L, et al. Improved peripheral
26. Palmeri S, Villanova M, Malandrini A, et al. Type I sialidosis: a nerve conduction, EEG, and verbal IQ after bone marrow trans-
clinical, biochemical, and neuroradiological study. Eur Neurol. plantation for adult metachromatic leukodystrophy. Bone Marrow
2000;43:88–94. Transplant. 1998;22:1119–1122.
27. Engel J, Rapin I, Giblin DR. Electrophysiological studies in two 51. Scriver CR, Beaudet AL, Sly WS, et al., eds. The Metabolic and
patients with cherry red spot-myoclonus syndrome. Epilepsia. Molecular Bases of Inherited Disease. 8th ed. New York: McGraw-
1977;18:73–87. Hill; 2001:3669–3693.
28. Louboutin JP, Nogues B, Caillaud C, et al. Multimodality evoked 52. Lyon F, Hagberg B, Evrand P, et al. Symptomatology of late onset
potentials and EEG in a case of cherry red spot-myoclonus syn- Krabbe’s leukodystrophy: the European experience. Dev Neurosci.
drome and alpha-neuraminidase deficiency (sialidosis type I). Eur 1991;13:240.
Neurol. 1995;35:175–177. 53. Husain AM, Altuwaijri M, Aldosari M. Krabbe disease: neurophys-
29. Tobimatsu S, Fukui R, Shibasaki H, et al. Electrophysiological iologic studies and MRI correlations. Neurology. 204;63:4.
studies of myoclonus in sialidosis type 2. Electroencephalogr Clin 54. Igarashi M, Schaumburg HH, Powers J, et al. Fatty acid abnormal-
Neurophysiol. 1985;60:16–22. ity in adrenoleukodystrophy. J Neurochem. 1976;26:851–860.
296 Part III ■ Clinical EEG: General

55. Moser HW, Moser AB, Frayer KK, et al. Adrenoleukodystrophy: 80. Koskiniemi M, Donner M, Majuri H, et al. Progressive
increased plasma content of saturated very long chain fatty acids. myoclonus epilepsy. A clinical and histopathological study. Acta
Neurology. 1981;31:1241–1249. Neurol Scand. 1974;50:307–332.
56. Kumar AJ, Rosenbaum AE, Naidu S, et al. Adrenoleukodystrophy: 81. Pennacchio LA, Lehesjoki AE, Stone NE, et al. Mutations in the
correlating MR imaging with CT. Radiology. 1987;165:496. gene encoding cystatin B in progressive myoclonus epilepsy
57. Percy AK, Rutledge SL. Adrenoleukodystrophy and related disor- (EPM1). Science. 1996;271:1731–1734.
ders. Ment Retard Dev Disabil Res Rev. 2001;7:179–189. 82. Caraballo RH, Fejerman N. Dravet syndrome: a study of 53
58. Barkovich AJ, Peck WW. Am J Neuroradiol. 1997;18:1163–1170. patients. Epilepsy Res. 2006;70s:S231–S238.
59. Takahashi Y, Suzuki Y, Kumazaki K, et al. Epilepsy in peroxisomal 83. Del-Favero CL, Ceulemans J, Ceulemans B, et al. De novo muta-
diseases. Epilepsia. 1997;38:182–188. tions in the sodium-channel gene SCN1A cause severe myoclonic
60. Govaerts L, Colon E, Rottevell J, et al. A neurophysiological study epilepsy of infancy. Am J Hum Genet. 2001;68:1327–1332.
of children with the cerebro-hepato-renal syndrome of Zellweger. 84. Blaskovics M, Engel R, Podosin RL, et al. EEG pattern in
Neuropediatrics. 1985;16:185–190. phenylketonuria under early initiated dietary treatment. Arch
61. Panjan DP, Meglic NP, Neubauer D. A case of Zellweger syndrome Pediatr Adolesc Med. 1981;135:802–808.
with extensive MRI abnormalities and unusual EEG findings. Clin. 85. Mina I, Hirotaka Y, Misao O. EEG pattern in phenylketonuria.
Electroencephalogr. 2001;32:28–31. J Jpn Pediatr Soc. 2004;108:1366–1371.
62. Chang YC, Huang CC, Huang SC, et al. Neonatal adrenoleukodys- 86. Gross PT, Berlow S, Schuett VE, et al. EEG in phenylketonuria.
trophy presenting with seizure at birth: a case report and review of Arch Neurol. 1981;38:122–126.
the literature. Pediatr Neurol. 2008;38:137–139. 87. Menkes JH, Hurst PL, Craig JM. A new syndrome: progressive
63. Leigh D. Subacute necrotizing encephalomyelopathy in an infant. familial infantile cerebral dysfunction associated with an unusual
J Neurol Neurosurg Psychiatry. 1951;14:216–221. urinary substance. Pediatrics. 1954;14:462.
64. Pincus JH. Subacute necrotizing encephalomyelopathy (Leigh’s 88. Scriver CR, Beaudet AL, Sly WS, et al., eds. The Metabolic and
disease): a consideration for clinical features and etiology. Dev Molecular Bases of Inherited Disease. Vol II. New York: McGraw-
Med Child Neurol. 1971;14:78–87. Hill; 2001.
65. Medina L, Chi TL, DeVivo DC, et al. MR findings in patients with 89. Brismar J, Aqeel A, Brismar G, et al. Maple syrup urine disease:
subacute necrotizing encephalopathy (Leigh syndrome). Am findings on CT and MR scans of the brain in 10 infants. Am
J Neuroradiol. 1989;11:379–384. J Neurorad. 1990;11:1219.
66. Canafoglia L, Franceschetti S, Antozzi C, et al. Epileptic phenotypes 90. Korein J, Sansaricq C, Kalmijn M, et al. Maple syrup urine disease:
associated with mitochondrial disorders. Neurology. 2001;56: clinical, EEG, and plasma amino acid correlations with a theoretical
1340–1346. mechanism of acute neurotoxicity. Int J Neurosci. 1994;79:21–45.
67. Pavlakis SG, Phillips PC, DiMauro S, et al. Mitochondrial myopa- 91. Mudd SH, Skovby F, Levy HL, et al. The natural history of homo-
thy, encephalopathy, lactic acidosis, and strokelike episodes: a dis- cystinuria due to cystathionine beta-synthase deficiency. Am
tinctive clinical syndrome. Ann Neurol. 1984;16:481–488. J Hum Genet. 1985;37:1–3.
68. Stoquart-ElSankari S, Lehmann P, Perin B, et al. MRI and diffu- 92. Del Giudice E, Striano S, Andria G. Electroencephalographic
sion-weighted imaging follow-up of a stroke-like event in a patient abnormalities in homocystinuria due to cystathionine synthase
with MELAS. J Neurol. 2008;255:1593–1595. deficiency. Clin Neurol Neurosurg. 1983;85:165–168.
69. Ito H, Mori K, Harada M, et al. Serial brain imaging analysis of 93. Buoni S, diBartolo RM, Molinelli M, et al. Atypical BECTS and
stroke-like episodes in MELAS. Brain Dev. 2008;30:483–488. homocystinuria. Neurology. 2003;61:1129–1131.
70. Bell AJ. MELAS syndrome: overview and case study. Am 94. Sass JO, Hofmann M, Skladal D, et al. Proprionic acidemia revis-
J Electroneurodiagn Technol. 1985;35:83–91. ited: a workshop report. Clin Pediatr. 2004;43:837–843.
71. Serra G, Piccinnu R, Tondi M, et al. Clinical and EEG findings in 95. Akman I, Imamoglu S, Demirkol M, et al. Neonatal onset propri-
eleven patients affected by mitochondrial encephalomyopathy onic acidemia without acidosis: a case report. Turk J Pediatr.
with MERRF–MELAS overlap. Brain Dev. 1996;18:185–191. 2002;44:339–342.
72. Ribacoba R, Salas-Puig J, Gonzalez C, et al. Characteristics of status 96. Aikoha H, Sasakia M, Sugaia K, et al. Effective immunoglobulin
epilepticus in MELAS. Analysis of four cases. Neurologia. 2006;21: therapy for brief tonic seizures in methylmalonic acidemia. Brain
1–11. Dev. 1997;19:502–505.
73. Shoffner JM, Wallace DC. Mitochondrial genetics: principles and 97. Bellieni CV, Ferrari F, DeFelice C, et al. EEG in assessing hydrox-
practice [editorial]. Am J Hum Genet. 1992;51:1179–1186. ycobalamin therapy in neonatal methylmalonic aciduria with
74. Alpers BJ. Diffuse progressive degeneration of gray matter of cere- homocystinuria. Biol Neonate. 2000;78:327–330.
brum. Arch Neurol Psychiatry. 1931;25:469–505. 98. Gilbert-Barness E, Barness LA. Isovaleric acidemia with promye-
75. Hunter M, Mackay MT, Peters H, et al. Alper’s syndrome with locytic myeloproliferative syndrome. Pediatr Dev Pathol. 1999;2:
POLG mutations: clinical, EEG, and radiological features. J Clin 286–291.
Neurosci. 2008;15:341. 99. Teychenne PF, Walters I, Claveria LE, et al. The encephalopathic
76. Joshi CN, Greenberg CR, Mhanni AA, et al. Ketogenic diet in action of five-carbon-atom fatty acids in the rabbit. Clin Sci Mol
Alpers–Hutternlocher syndrome. Pediatr Neurol. 2009;40:314–316. Med. 1976;50:463–472.
77. Boyd SG, Harden A, Egger J, et al. Progressive neuronal degenera- 100. Hoover-Fong JE, Shah S, Van Hove JL, et al. Natural history of non-
tion of childhood with liver disease (“Alpers’ disease”): character- ketotic hyperglycinemia in 65 patients. Neurology. 2004;63:1847.
istic neurophysiological features. Neuropediatrics. 1986;17:75–80. 101. Applegarth DA, Toone JR. Nonketotic hyperglycinemia (glycine
78. Minassian BA. Lafora’s disease: towards a clinical, pathologic, and encephalopathy): laboratory diagnosis. Mol Genet Metab. 2001;
molecular synthesis. Pediatr Neurol. 2001;25:21–29. 74:139–146.
79. Acharya JN, Satishchandra P, Shankar SK. Familial progressive 102. Flusser H, Kormann SH, Sato K, et al. Mild glycine encephalopa-
myoclonus epilepsy: clinical and electrophysiologic observations. thy (NKH) in a large kindred due to a silent exonic GLDC splice
Epilepsia. 1995;36:429–434. mutation. Neurology. 2005;64:1426–1430.
Chapter 15 ■ EEG of Degenerative Disorders of the Central Nervous System 297

103. Hamosh A, McDonald JW, Valle D, et al. Dextromethorphan and 125. Taly AB, Meenakshi-Sundaram S, Sinha S, et al. Wilson disease:
high-dose benzoate therapy for nonketotic hyperglycinemia in description of 282 patients evaluated over 3 decades. Medicine
an infant. J Pediatr. 1992;121:131–135. (Baltimore). 2007;86:112–121.
104. Amladi TS, Kohil M. Hartnup disease. Indian J Dermatol Venereol 126. Saito T. Presenting symptoms and natural history of Wilson dis-
Leprol. 1994;60:105–107. ease. Eur J Pediatr. 1987;146:261–265.
105. Lowe CU, Terrey M, MacLachan EA. Organic aciduria, decreased 127. Giagheddu M, Tamburini G, Piga M, et al. Comparison of MRI,
renal ammonia production, hydrophthamos[hydrophthalamos], EEG, EPs, and ECD-SPECT in Wilson’s disease. Acta Neurol
and mental retardation: a clinical entity. Am J Dis Child. 1952; Scand. 2001;103:71–81.
83:164–184. 128. Heller GL, Kooi KA. The electroencephalogram in hepatolentic-
106. Schneider JF, Boltshauser E, Neuhaus TJ, et al. MRI and proton ular degeneration (Wilson’s disease). Electroencephalogr Clinical
spectroscopy in Lowe syndrome. Neuropediatrics. 2001;32: Neurophysiol. 1962;14:520–526.
45–48. 129. Hsich GE, Robertson RL, Irons M, et al. Cerebral infarction in
107. Sener RN. Lowe syndrome: proton MR spectroscopy and diffu- Menkes disease. Pediatr Neurol. 2000;23:425–428.
sion MR imaging. J Neuroradiol. 2004;31:238–240. 130. Kaler SG, Holmes CS, Goldstein DS, et al. Neonatal diagnosis and
108. Ono J, Harada K, Mano T, et al. MRI findings and neurologic treatment of Menkes disease. N Engl J Med. 2008;358:605–614.
manifestation in Lowe oculocerebrorenal syndrome. Pediatr 131. Bahi-Buisson N, Kaminska A, Nabbout R, et al. Epilepsy in Menkes
Neurol. 1996;14:162–164. disease: analysis of clinical stages. Epilepsia. 2006;47:380–386.
109. Erdogan F, Ismailogullari S, Soyuer I, et al. Different seizure types 132. White SR, Reese K, Sato S, et al. Spectrum of EEG findings in
and skin lesions in oculocerebrorenal syndrome of Lowe. J Child Menkes disease. Electroencephalogr Clin Neurophysiol. 1993;87:
Neurol. 2007;22:427–431. 57–61.
110. Leonard JV, Morris AA. Urea cycle disorders. Semin Neonatol. 133. Wang D, Pascual JM, Yang H, et al. Glut-1 deficiency syndrome:
2002;7:27. clinical, genetic, and therapeutic aspects. Ann Neurol. 2005;
111. Butterworth RF. Effects of hyperammonaemia on brain function. 57(1):111–118.
J Inherit Metab Dis. 1998;21(suppl 1):6. 134. Bennett CL, Chen Y, Hahn S, et al. Prevalence of ALDH7A1
112. Maestri NE, Lord C, Glynn M, et al. The phenotype of ostensibly mutations in 18 North American pyridoxine-dependent seizure
healthy women who are carriers for ornithine transcarbamylase (PDS) patients. Epilepsia. 2009;50:1167–1175.
deficiency. Medicine (Baltimore). 1998;77:389. 135. Nabbout R, Soufflet C, Plouin P, et al. Pyridoxine dependent
113. Verma NP, Hart ZH, Kooi KA. Electroencephalographic findings epilepsy: a suggestive electroclinical pattern. Arch Dis Child Fetal
in urea-cycle disorders. Electroencephalogr Clin Neurophysiol. Neonatal Ed. 1999;81:F125–F129.
1984;57:105–112. 136. Torres OA, Miller VS, Buist NMR, et al. Folinic acid-responsive
114. Clancy RR, Chung HJ. EEG changes during recovery from acute neonatal seizures. J Child Neurol. 1999;14:529–532.
severe neonatal citrullinemia. Electroencephalogr Clin Neurophysiol. 137. Gallagher RC, Van Hove JL, Scharer G, et al. Folinic acid-respon-
1991;78:222–227. sive seizures are identical to pyridoxine-dependent epilepsy. Ann
115. Nagata N, Matsuda I, Matsuura T, et al. Retrospective survey of Neurol. 2009;65:550–556.
urea cycle disorders: part 2. Neurological outcome in forty-nine 138. Manto MU. The wide spectrum of spinocerebellar ataxia (SCAs).
Japanese patients with urea cycle enzymopathies. Am J Med Genet. Cerebellum. 2005;4:2–6.
1991;40:477–481. 139. Sack G, Gruss B, Lossner J, et al. EEG studies of spinocerebellar
116. Hayflick SJ. Neurodegeneration with brain iron accumulation: from ataxia. Psychiatr Neurol Med Psychol (Leipz). 1977;29:521–528.
genes to pathogenesis. Semin Pediatr Neurol. 2006;13:182–185. 140. Rasmussen A, Matsuura T, Ruano L, et al. Clinical and genetic
117. McNeill A, Birchall D, Hayflick SJ, et al. T2 and FSE MRI distin- analysis of four Mexican families with spinocerebellar ataxia type
guishes four subtypes of neurodegeneration with brain iron 10. Ann Neurol. 2001;50:234–239.
accumulation. Neurology. 2008;70:1614. 141. Tan NCK, Zhou Y, Tan ASC, et al. Spinocerebellar ataxia type 2
118. Cossu G, Melis M, Floris G, et al. Hallervorden Spatz syndrome with focal epilepsy—an unusual association. Ann Acad Med
(pantothenate kinase associated neurodegeneration) in two Singapore. 2004;33:103–106.
Sardinian brother with homozygous mutation in PANK 2 gene. 142. Inazuki G, Baba K, Naito H. Electroencephalographic findings of
J Neurol. 2002;249:1599–1600. hereditary dentatorubral-pallidoluysian atrophy (DRPLA). Jpn J
119. Remond A, ed. Handbook of Electroencephalography and Clinical Psychiatry Neurol. 1989;43:213–220.
Neurophysiology. Vol 15A. Amsterdam: Elsevier; 162–191. 143. Rötig A, de Lonlay P, Chretien D, et al. Aconitase and mitochon-
120. Swaiman K, Smith SA, Trock GL, et al. Sea blue histiocytes, lym- drial iron–sulphur protein deficiency in Friedreich ataxia. Nat
phocytic cytosomes, movement disorder. 59Fe uptake in basal Genet. 1997;17:215–217.
ganglia: Hallervorden-Spatz disease or ceroid storage disease 144. Hewer RL. Study of fatal cases of Friedreich’s ataxia. Br Med J.
with abnormal isotope scan? Neurology. 1983;33:301–305. 1968;3:649–652.
121. Seven M, Ozkilic A, Yuksel A. Dysmorphic face in two siblings 145. Bourchard RW, Bouchard JP, Bouchard R, et al. Electro -
with infantile neuroaxonal dystrophy. Genet Counsel. 2002;13: encephalographic findings in Friedreich’s ataxia and autosomal
465–473 recessive spastic ataxia of Charlevoix-Saguenay (ARSACS). Can J
122. Hortnagel K, Nardocci N, Zorzi G, et al. Infantile neuroaxonal Neurol Sci. 1979;6:191–194.
dystrophy and pantothenate kinase-associated neurodegenera- 146. Hagberg B, Hanefeld F, Percy A, et al. An update on clinically appli-
tion: locus heterogeneity. Neurology. 2004;63:922–924. cable diagnostic criteria in Rett syndrome: comments to Rett
123. Morgan NV, Westaway SK, Morton JEV, et al. PLA2G6, encoding Syndrome Clinical Criteria Consensus Panel Ssatellite to European
a phospholipase A2, is mutated in neurodegenerative disorders Paediatric Neurology Society meeting, Baden Baden, Germany,
with high brain iron. Nat Genet. 2006;38:752–754. September 11, 2001. Eur J Paediatr Neurol. 2002;6:293–297.
124. Wakai S, Asanuma H, Hayasaka H, et al. Ictal video-EEG analysis 147. Percy A, Lane J. Rett syndrome: clinical and molecular update.
of infantile neuroaxonal dystrophy. Epilepsia. 1994;35:823–826. Curr Opin Pediatr. 2004;16:670–677.
298 Part III ■ Clinical EEG: General

148. Bahi-Buisson N, Nectoux J, Rosas-Vargas H, et al. Key clinical 154. Stecker MM, Myers SM. Reserpine responsive myoclonus and
features to identify girls with CDKL5 mutations. Brain. 2008;131: hyperpyrexia in a patient with Angelman syndrome. Clin Neurol
2647–2661. Neurosurg. 2003;105:183–187.
149. Glaze DG, Frost JD Jr, Zoghbi HY, et al. Rett’s syndrome: correla- 155. Stafstrom CE, Patxot OF, Gilmore HE, et al. Seizures in children
tion of electroencephalographic characteristic with clinical stag- with Down syndrome: etiology, characteristics and outcome. Dev
ing. Arch Neurol. 1987;44:1053–1056. Med Child Neurol. 1991;33:191–200.
150. Glaze DG. Neurophysiology of Rett syndrome. Ment Retard Dev 156. Silva ML, Cieuta C, Guerrini R, et al. Early clinical and EEG fea-
Disabil Rev. 2002;8:66–71. tures of infantile spasms in Down syndrome. Epilepsia. 1996;37:
151. Hagberg B, Witt-Engerstrom I. Rett syndrome: a suggested stag- 977–982.
ing system for describing impairment profile with increasing age 157. De Simone R, Daquin G, Genton P. Senile myoclonic epilepsy in
towards adolescence. Am J Med Genet. 1986;26:47–59. Down syndrome: a video and EEG presentation of two cases.
152. Pelc K, Boyd SG, Cheron G, et al. Epilepsy in Angelman syn- Epileptic Disord. 2006;8:223–227.
drome. Seizure. 2008;17:211–217. 158. Kaneko WM, Phillips EL, Riley EP, et al. EEG findings in fetal alco-
153. Valente KD, Andrade JQ, Grossmann RM, et al. Angelman syn- hol syndrome and Down syndrome children. Electroencephalogr
drome: difficulties in EEG pattern recognition and possible mis- Clin Neurophysiol. 1996;98:20–28.
interpretations. Epilepsia. 2003;44:1051–1063.
CHAPTER
The EEG in Congenital Malformations of
Cortical Development, Neurocutaneous
Disorders, Cerebral Palsy, Autism/ Mental
16
Retardation, and ADHD/ Learning
Disabilities of Childhood
MARY REPOLE ANDRIOLA

T
his chapter reviews numerous disorders of infancy and the EEG abnormalities localized to the area of the shunt itself
childhood considered congenital with a known genetic and were first noted after surgery, though asynchrony of sleep
or familial basis or acquired by insults to the developing spindles was noted in both groups (3). Bartoshesky et al. (4)
brain. These include various structural abnormalities, cerebral found in studying a group of 111 children with meningomye-
palsy, autism, mental retardation, attention-deficit hyperactiv- locele that 24 out of 98 children with shunted hydrocephalus
ity disorder (ADHD), and learning disabilities. had seizures. More of these children had other brain malfor-
mations besides Arnold Chiari, shunt infections, and perhaps
CONGENITAL MALFORMATIONS shunt revisions and these children were more likely to be
OF CORTICAL DEVELOPMENT developmentally delayed than the rest (4). Saukkonen et al.
(5) following 168 shunted hydrocephalic children found
Anencephaly is a severe anomaly with absence of the cerebrum seizures in 22% of children prior to surgery in addition to
and superior skull. These infants survive only a very brief time; abnormal EEGs. Some of these patients presented with
thus, EEG studies are not usually obtained for practical and seizures as newborns and some had infantile spasms with
technical reasons. hypsarrhythmic EEGs but there was no specific localization or
Hydranencephaly refers to nearly complete absence of the EEG pattern otherwise. The subsequent occurrence of
cerebral hemispheres that is thought most likely due to bilat- seizures, 25.6% after shunting for a total of 47.6% over a long
eral cerebrovascular occlusions or intrauterine infections with follow-up period with a mean of 8.9 years, they found related
cytomegalovirus (CMV) or toxoplasmosis after the fourth to the lack of use of prophylactic antiepileptic medication
month of gestation. The infant is often initially normo- causing them to recommend treatment in those hydrocephalic
cephalic and nondysmorphic. The EEG is usually attenuated children prior to shunting with abnormal EEGs with epilepti-
(1) but Velasco et al. reported a hydrancephalic patient with form discharges. They did not find a relationship of EEG
seizures considered consistent with the Lennox Gastaut syn- abnormalities to the site of the shunt, number of revisions,
drome, though the typical slow spike-wave pattern was not nor infections, but they did find the development of seizures
seen (2). related to the occurrence of the slit ventricle syndrome (5). A
Hydrocephalus refers to a dilatation of the ventricular sys- similar incidence of seizures (48.5%) was reported by Noetzel
tem due to a variety of underlying brain structural anomalies and Blake (6) in 68 children with congenital hydrocephalus
or secondarily acquired from bleeding or infection. It is often not associated with myelomeningocele. Brain malformations,
treated by a neurosurgical procedure of shunting, typically developmental impairment, and spike, spike-wave, or slow-
with a catheter from the right lateral ventricle to the peri- wave paroxysmal EEG activity at seizure onset focal or diffuse
toneal cavity. The underlying abnormalities of brain struc- correlated with intractable seizures while age of shunting,
ture, the presence of the shunt (the burr hole as a revisions, or infections did not. The few children without
craniotomy), and at times complications of the shunt with mental retardation were able to come off their medication
infection, hemorrhage, and multiple revisions can give rise to after 3 years seizure free (6). Seizures may also occur in these
EEG abnormalities and seizures. Ines and Markand (3) children as a symptom of shunt malfunction (7). Faillace and
reviewed the EEGs of 92 hydrocephalic patients and found a Canady described the various EEG findings in 9 out of 15
higher incidence of EEG abnormalities in the shunted (95%) patients with seizure activity in the presence of shunt mal-
as compared to the nonshunted patients (47%), with a 65.4% function. These included diffuse dysfunction and focal spike
versus 18.2% incidence of seizures in the two groups. Many of activity (Fig. 16.1) (8).

299
300 Part III ■ Clinical EEG: General

Figure 16.1 A 37-year-old woman with hydrocephalus, Arnold Chiari II, and myelodysplasia. Patient has had multiple
shunts and revisions—initially on the right, now on the left. History of seizures as a child. EEG: slowing and higher volt-
age on the right plus independent sharp activity from either hemisphere.

Holoprosencephaly (HPE) is a syndrome of congenital brain closed. The gray matter is abnormal, often composed of
malformations with incomplete separation of the hemispheres polymicrogyri or heterotopic gray matter. This congenital dis-
and basal ganglia of varying severity often associated with midline order of brain development occurring at the end of the first and
facial dsymorphisms, hypothalamic pituitary dysfunction, the beginning of the second trimester may be associated with
developmental delay, abnormal EEGs, and epilepsy in approxi- other cerebral anomalies, particularly agenesis of the corpus
mately half the patients. A dorsal cyst may be present produc- callosum, absence of the septum pellucidum, polymicrogyria,
ing hydrocephalus that requires shunting. The disorder has and gray matter heterotopias, neurologic and developmental
multiple causes: teratogenic and chromosomal including tri- impairment, epilepsy, and EEG abnormalities. Packard et al.
somy 13 and 15 and the sonic hedgehog mutation (SHH), SIX, (13) reported on 47 children with all types of which 57% had
TGIG, and ZIC2. This can be considered an abnormality of seizures, one third of which were difficult to control. The most
neural induction in the ventrodorsal plane (9). There are four common seizure type was complex partial (70%). Though
types: alobar, semilobar, lobar, and middle interhemispheric seizures presented earlier and were harder to control in the
variant (10,11). Clegg et al. (12) reviewed the literature and bilateral open lip type, there was no correlation of seizure type
reported on 32 children, approximately half of whom had or occurrence with the type of schizencephaly. Of the 27
epilepsy with various forms of the disorder. Of the 20 patients patients with seizures, EEGs were unavailable in 1, normal in 3,
who had EEGs the abnormalities noted were hypersynchronous contained spike and sharp wave discharges or sharp waves
theta during waking and sleep, prominent spindle-like beta localizing to the cleft in 15, multifocal discharges in 5, and dif-
during drowsing and sleep, multiple epileptiform discharges, fuse or focal slowing in 3. The neurodevelopmental outcome of
and periods of attenuation. Posterior gradient flattening was these children was dependent on the extent of cortex involved.
seen in one child with a dorsal cyst associated with alobar HPE They often have motor and language impairment (13). The
(Figs. 16.2 and 16.3) (12). EEGs in nine patients with schizencephaly were reviewed by
Schizencephaly refers to a full-thickness gray matter-lined Granata et al. (14). Focal epileptiform discharges that correlated
cleft of a hemisphere that may be unilateral or bilateral, open with their clinical seizures and the location of the cleft were the
(communication between ventricle and subarachnoid space) or most common findings. Their six patients with unilateral clefts
Chapter 16 ■ The EEG in Congenital Malformations 301

Figure 16.2 A 10-year-old girl with semi-


lobar holoprosencephaly. The patient has
marked developmental retardation and at
times appears to have myoclonic spasms
involving her upper trunk but her parents
prefer not having her on medication. Her
EEG is without age-appropriate waking
and sleeping patterns and frequent high-
voltage paroxysmal synchronous notched
delta discharges as illustrated.

had IQs in the normal range, hemiparesis, and incompletely Hemimegalencephaly refers to congenital hypertrophy of
controlled partial seizures that were postural at onset followed one cerebral hemisphere with abnormal chaotic cytoarchitec-
by turning or complex partial but did not generalize (Fig. 16.4) ture of gray and white matter with an ipsilateral enlarged ven-
(14). A report of two cases by Iannetti et al. (15) raises the pos- tricle. Clinically, the patients have hemiparesis, mental
sibility of congenital CMV infection causing schizencephaly. retardation, and seizures. The EEG data in 12 patients reported
These patients were negative for a mutation in the homeobox by Paladin et al. (16) found partial seizures in all patients and 3
gene EMX2 that has been found in many patients with schizen- also had infantile spasms. In addition, they also reviewed the lit-
cephaly (15). erature. Three EEG patterns were observed, tended not to

Figure 16.3 An 8-year-old girl with the


middle interhemispheric variant of holo-
prosencephaly. She has mild facial dys-
morphism, developmental delay, and
symptomatic epilepsy. Her EEG reveals
(A) asynchrony and multifocal spikes, (B)
and marked activation by sleep consistent
with ESES. (C) MRI demonstrates fused
frontal lobes, absent septum pellucidum,
and falx. (D) MRI shows agenesis of the
body of the corpus callosum with radia-
tion of the gyri.(continued)

A
302 Part III ■ Clinical EEG: General

C D

Figure 16.3 (continued)

change over time, and were correlated with the patient’s clinical frontal by 2 years of age. Though seizure onset occurred
history: before 6 months, these children learned to walk and talk and
could attend special schools.
1. Triphasic spike complexes of large amplitude maximum in
3. Suppression burst occurred at birth or within the first few
the abnormal hemisphere that appeared early as did seizures.
months of life. This pattern seemed to respond to steroid ther-
No significant developmental milestones were achieved and
apy with a relatively favorable outcome in two patients (16).
one patient died at age 7 days.
2. Isolated “alpha-like” activity appeared at approximately Aicardi syndrome as first described in 1965 occurs in girls with
6 months of age in the central area and extended to the severe mental retardation and ocular abnormalities who present
Chapter 16 ■ The EEG in Congenital Malformations 303

Figure 16.4 A 39-year-old man with incompletely controlled partial epilepsy and right hemiparesis secondary to left pari-
etal open lip schizencephaly who nevertheless holds a job and is married. EEG findings of low-voltage left temporal sharp
waves and underlying slowing.

with infantile spasms (17). Their brains have a thin unlayered cor- centrotemporal. Some of the patients are retarded or develop-
tex with diffuse polymicrogyria with fused molecular layer, nodu- mentally delayed. Gastaut et al. proposed that the seizures were
lar heterotopias in the periventricular region and centrum related to the disconnection of the corpus callosum (22).
semiovale, and agenesis of the corpus callosum. The patients may Intrauterine infections with viruses under the acronym
also have chorioretinal lacunae, coloboma, and vertebral and TORCH (toxoplasmosis, rubella, CMV, and herpes simplex
costal abnormalities. They usually have a shortened life span. The virus) plus syphilis can cause some of the congenital anomalies
EEG findings are that of completely asynchronous activity reviewed previously in this section. Infants with infections close
between the hemispheres consistent with agenesis of the corpus to the time of delivery may have systemic symptoms. Sequelae
callosum plus multifocal discharges on a burst suppression pat- include chorioretinitis, sensorineural hearing loss, hydro-
tern. There is also absence of recognizable stage II sleep activity cephalus, microcephaly, developmental retardation, various neu-
such as spindles (18). There are more than 100 cases reported (19). rologic deficits, and seizures with abnormal EEGs (23).
Agenesis of the corpus callosum may be seen as a total or A number of teratogens are known to adversely affect the
partial phenomenon with the severity of clinical and EEG find- fetus. Fetal exposure to alcohol is a now-known preventable ter-
ings often related to other brain anomalies. Lacey (20) reviewed atogen causing fetal alcohol syndrome (FAS) with microcephaly
40 children prospectively followed for up to 15 years. More than (as part of the intrauterine growth retardation), developmental
half of the patients were retarded and half had seizures. EEGs retardation, and irritability and restlessness in infants.
were done in all patients and abnormalities were consistent Craniofacial anomalies are also reported (24). A study by
with the clinical picture from hypsarrhythmia in the seven Kaneko et al. (25) comparing the EEG findings in 18 matched
patients with infantile spasms to background slowing and mul- children with the FAS and Down syndrome versus normal con-
tifocal and generalized discharges in the patients with retarda- trols using power spectra analysis found significant reductions
tion and symptomatic epilepsy (Fig. 16.5) (20). Agenesis of the in the mean power of alpha frequencies for both clinical groups.
corpus callosum can also be caused by metabolic disorders (21). While Down syndrome children had diffuse slowing, FAS chil-
When a lipoma occurs in the corpus callosum, epilepsy, usu- dren had decreased posterior alpha especially on the left (25).
ally partial and difficult to control, and an abnormal EEG, begins Another common modern teratogen inflicting harm on the
in childhood. The EEG abnormality may appear temporal or fetal brain is cocaine. Infants exposed to cocaine have been
304 Part III ■ Clinical EEG: General

has a hemiparesis, developmental delay, and epilepsy. Ho et al.


(31) describe the seizure localization in 14 patients with poren-
cephaly and intractable seizures. The porencephaly was in the dis-
tribution of the middle cerebral artery in eight, posterior cerebral
in three, internal carotid in one, and multiple vessels in two. The
majority of patients had complex partial seizures. Scalp EEGs
demonstrated abnormal background with focal slowing over the
cyst in four patients. Eight patients had interictal discharges noted
anterior temporally (one bilateral) and four extratemporally. Ictal
monitoring confirmed temporal localization as did one intracra-
nial monitoring. A total of 13 patients had hippocampal forma-
tion atrophy ipsilateral to the side of the lesion, 2 of whom
underwent surgery and became seizure free (31).
Arachnoid cysts are intracranial congenital lesions in which
redundant infoldings of the arachnoid membrane appears to
trap cerebral spinal fluid into a cyst. These are often found
Figure 16.5 A 13-month-old female infant with agenesis of the corpus “incidentally” when an imaging study is obtained post-trauma
callosum, microcephaly, congenital cataracts, symptomatic intractable or as an evaluation for another diagnosis such as epilepsy. There
epilepsy, and severe developmental delay. The patient has a brother has been a question raised as to whether or not these cysts play
with the same disorder. Her EEG is markedly abnormal with lack of age- a causative role in a patient’s epilepsy and therefore should be
appropriate background rhythms, asynchrony, and multifocal spikes. treated surgically. The consensus appears to be that cyst
Hemispherical asynchrony is the electrographic hallmark of this disor- removal is not likely to reduce seizures, but instead these
der. Her EEG at 5 months when she presented with infantile spasms patients need to be treated with medication even if epileptiform
had a hypsarrhythmic pattern. activity is noted in the region of the cyst (32,33).
Infantile nonprovoked epilepsy after the neonatal period is
often related to cortical dysplasias. The pathogeneses and the
pathology of malformations of cortical development are
reported to have smaller head circumferences from birth reviewed by Barkovich et al. (34). Focal cortical dysplasia refers
through at least 2 years of age, and developmental delay when to a localized area of abnormal cortical structure that may be
evaluated at 2 years (26). A report of 10 children with intrauter- appreciated now preoperatively by a combination of improved
ine exposure to cocaine and other street drugs found significant imaging and EEG studies. Patients tend to have intractable
intracranial and ophthalmologic anomalies including absence epilepsy with focal interictal discharges in about half the cases.
of the corpus callosum, schizencephaly, hydrancephaly, poren- They may be neurologically normal or impaired and may be
cephaly, septo-optic dysplasia, and optic atrophy (27). Half of candidates for seizure surgery.
the patients had seizures. Many of these malformations and Tassi et al. in a review of histologic sections from a large
their associated EEG findings have been described above. epilepsy surgery series correlated the clinical, EEG, MRI, and
Congenital blindness may be associated with cerebral mal- neuropath findings in patients with cortical dysplasia. They sep-
formations, developmental retardation, or be purely on an ocu- arate the pathology into the following three subtypes: (i) archi-
lar basis. The EEG of a blind child usually lacks occipital alpha tectural dysplasia with disruption of cortical lamination and
while central rhythms are preserved. Occipital spikes may be ectopic neurons in the white matter, (ii) cytoarchitectural dyspla-
seen in the absence of clinical seizures. These findings tend to sia with abnormal cortical lamination plus giant neurofilament-
decrease with age (28). enriched neurons, and (iii) (Taylor type) cortical dysplasia,
We should note, however, that the earlier theory that visual abnormal cortical layering plus giant dysmorphic neurons, and
deprivation can cause electrical abnormalities over the visual balloon cells. Seizure frequency was more severe with the
cortex was not supported by some data, namely by the study of increased severity of the pathology. The epileptogenic zone was
Jan and Wong (29) who investigated 104 children with ocular mainly extratemporal and the best seizure free rate after surgery
visual loss. In this study although most totally blind children’s was in the Taylor type (35). Krsek et al. (37) reviewing the results
records lacked alpha, five had spontaneously appearing and dis- of 200 children who underwent seizure surgery for cortical dys-
appearing alpha rhythm. plasia used the more complex classification system proposed in
A more recent study (30) showed a significant reduction of 2004 by Palmini et al. (36) that has more subgroups and corre-
alpha power in the blind compared to the sighted controls over lated history, clinical findings, EEG, MRI, neuropsychological,
parieto-occipital recording sites and concluded that brain surgical variables, and outcome data. Mild malformations of cor-
structures, associated with the generation of posterior alpha tical development and focal cortical dysplasia type I were more
rhythms in sighted adults, might be either reorganized or atro- frequently associated with perinatal risk factors. Ictal EEGs were
phied following blindness from birth. more frequently precisely localized in the mild cortical dysplasia
Congenital porencephaly is residua of an infarct that may have and the focal cortical dysplasia II group b (37). Kim et al. (38)
occurred during the pre- or perinatal period. The patient usually report on 166 consecutive adult patients at the time of surgery
Chapter 16 ■ The EEG in Congenital Malformations 305

followed over a 10-year span. Forty additional patients were lateral hippocampal hypoplasia. Three patients were either
excluded as they also had hippocampal sclerosis. Scalp video EEG autistic or had mild mental retardation (Fig. 16.7). Most
monitoring, interictal (42.7%), and ictal (76.5%) correctly local- patients had epilepsy with focal seizures difficult to control and
ized to the resected lobe, while the MRI had a detection rate of the three who did not yet were under 15 years. Background
42.2% that was greatest the more severe the cortical dysplasia rhythms were normal as were stage II sleep patterns. Photic
(38). For both series, the best surgical outcome was related to the driving was bilateral except in the unilateral cases where it was
completeness of the resection (37,38). ipsilateral. Multifocal epileptiform activity was seen interictally
Congenital abnormalities of brain structure now known to even in the unilateral patients in addition to bilateral asynchro-
be or likely to be on a chromosomal (genetic basis) such as neu- nous abnormalities though in slow-wave sleep, bilaterally dif-
ronal migrational disorders often lead to EEG abnormalities fuse bursts of polyspikes were seen. Three ictal patterns are also
and symptomatic epilepsy. Examples are periventricular nodu- described all partial onset. There is some evidence that seizure
lar heterotopia (PNH), subcortical band heterotopia (SBH), activity may arise from the heterotopic nodules (40,41). The
and lissencephaly (34). patients may have hippocampal sclerosis; however, surgery to
PNH are subependymal nodules of gray matter along the remove the hippocampus does not achieve seizure control (41).
superior lateral walls of the lateral ventricles from the frontal Double cortex syndrome or SBH is characterized by a sub-
horns to the trigones. Bilateral PNH occurs in females as an X- cortical band of gray matter and an abnormal DCX gene (XLIS)
linked dominant (Xq28) usually with mild epilepsy onset in the in most cases (34). Mosaic mutations of the LISI gene that is
second decade and normal to borderline IQ. The interictal EEG usually the responsible gene in severe lissencephaly can be
is normal or there are temporal discharges (Fig. 16.6). When the found in patients with posteriorly prominent SBH (42). These
periventricular nodules are seen in association with other brain patients, more commonly female, are retarded with usually
malformations, the patients usually are males, have a younger intractable epilepsy. The EEG pattern most typically seen is that
age at presentation, mental retardation, more difficult to con- of hypsarrhythmia with clinical infantile spasms followed by a
trol seizures (often atonic), and EEGs with focal and bisynchro- Lennox Gastaut pattern though multifocal spikes may be noted
nous discharges (39). (Figs. 16.8–16.10) (43,44). In a 13-year-old female patient who
Battaglia et al. (40) discuss the spectrum of PNH in 54 had generalized tonic and grand mal seizures and the DCX gene
patients including the various types with known genetic point mutation by DNA analysis, a persistent Lennox Gastaut slow
mutations of the FLNI gene in bilateral, symmetrical familial spike-wave pattern was seen on EEG associated with SBH on
occurring mostly in females and familial patients with bilateral MRI and increased PET FDG uptake in the band (45).
PNH not related to the gene and sporadic cases. Unilateral PNH Mutations of the X-linked gene DCX (XLIS) usually cause SBH
is usually found in the posterior paratrigonal region of the lat- in females and lissencephaly in males (46).
eral ventricle but can extend into white matter and cortical tis- Lissencephaly that means a smooth cerebral surface is also
sue. In this study, 12 out of 14 were females with 4 patients caused by deletions involving the L1S1 gene on chromosome
having abnormal overlying neocortical areas and 8 having ipsi- 17 p13.3 (34). This can be isolated or part of a syndrome such

Figure 16.6 A 12-year-old girl with


developmental delay, partial-onset
seizures, and bilateral frontal and lateral
periventricular heterotopic gray matter
nodules. EEG: right mid-temporal–central
sharp and slow waves with wide field acti-
vated by drowsiness.
306 Part III ■ Clinical EEG: General

Figure 16.7 An 8-year-old girl with


macrocephaly, developmental delay/ autis-
tic spectrum disorder, and partial-onset
symptomatic epilepsy with head turning
and eye deviation to the left, lip smacking,
and confusion. A heterotopic gray matter
lesion close to the right posterior lateral
ventricle is noted on MRI while the EEG
demonstrates focal slowing from the right
posterior quadrant.

Figure 16.8 A 3-year-old girl who presented at 5 months with infantile spasms that responded to ACTH treatment. She
then developed prolonged complex partial seizures at 2.5 years. Initial MRI was reported as normal; however, subsequent
MRI revealed double cortex syndrome. EEG at 5 months—high-voltage bursts of polyspike and sharp wave discharges are
noted in both the waking and sleeping states. These bursts are often bilateral and are followed by attenuation. There are
also sharp wave transients seen in a multifocal manner. Often, when the patient has these discharges, she shows no clin-
ical change. At other times when she has these discharges in the waking state, she may stop and blink, she may have a
head drop, or she may have an actual infantile spasm.
Chapter 16 ■ The EEG in Congenital Malformations 307

Figure 16.9 Polymorphic predominantly delta activity, occasionally sharp contoured is seen bilaterally but maximal in
voltage in right temporal region. These transients were associated with two clinical episodes of staring and unresponsive-
ness. Right temporal sharp and slow waves are seen throughout with phase reversals at T4 and T6.

as the Miller–Dieker syndrome. The patients are usually excessively folded cortex without the normal six-layer struc-
severely retarded, initially hypotonic, and then spastic. Most ture is seen in children with developmental delay and seizures
have intractable seizures and abnormal EEGs with diffuse fast that may be intractable. EEGs may show multifocal spikes.
rhythms of high amplitude though diffuse slow is also seen in Guerrini et al. described nine patients with multilobar polymi-
the first year of life (47). A study of 17 patients in the crogyri and electrical status epilepticus during sleep (ESES)
Netherlands by de Rijk van Andel et al. (48) with serial EEG with infrequent focal motor seizures but eight out of nine had
recordings found high-amplitude beta at times cycling with atonic drop attacks. These patients however who ranged from
sharp- and slow-wave complexes especially when the patients severely mentally retarded to one patient with normal cogni-
had more seizures. Other patterns included high-voltage slow tive function outgrew their ESES and were either seizure free
and diffuse rhythmic high-voltage alpha and beta patterns off or on medication except for two patients with infrequent
(48). Sébire et al. (49) reported 43 children with macrogyri or seizures (52).
agyri (lissencephaly type I). Rapid rhythms of high-voltage
and high-amplitude delta to theta complexes were seen in the NEUROCUTANEOUS DISORDERS
latter group (49).
Patients with bilateral perisylvian polymicrogyria syn- Neurofibromatosis I (NFI) is a neurocutaneous disorder associ-
dromes have pseudobulbar palsy, mental retardation, and usu- ated with skin, eye, brain, and systemic lesions. Seizures and asso-
ally symptomatic epilepsy with most typically the Lennox ciated EEG abnormalities occur in a minority of patients. Young
Gastaut syndrome clinically and the pattern on EEG. MRI et al. reviewing their experience in an NF clinic over a 7-year
reveals bilateral perisylvian microgyri while pathologically period found 5% of the patients had seizures diagnosed mainly in
small prominent convolutions are seen without proper cortical childhood (53). Korf et al. (54) found 22 out of 359 persons with
lamination (50). A more widespread bilateral generalized NFI followed in a children’s hospital over a 10-year period, to have
polymicrogyria syndrome was reported by Chang et al. (51) one or more seizures, with onset ranging from newborn to over
and there are other regional polymicrogyri syndromes. Thin 20 years. There was a variety of seizure types including febrile
308 Part III ■ Clinical EEG: General

of tubers and the tuber burden, the more likely an infant is to


have infantile spasms and to be cognitively impaired. This
makes it even more imperative that they are treated early with
vigabatrin, perhaps over ACTH and even surgery on occasion
(57,61–63). Major et al. (64) studying the epileptogenicity of
the tubers and the surrounding cortex in three patients with TS
undergoing seizure surgery using subdural grids on the tubers
and cortex and depth electrodes into the tubers found signifi-
cant epileptiform activity in the surrounding tissue while the
tuber was cold. Patient seizures were markedly reduced after the
tuber and local tissue were removed (64,65).
Sturge Weber syndrome is a neurocutaneous syndrome
with leptomeningeal angiomatosis most commonly in the
occipital, parietal region, facial cutaneous vascular malforma-
tion in the ophthalmic division especially involving the skin
around the eye and eye lid, glaucoma, and hemianopsia. This is
presumably based on an embryogenic error in the formation of
the cephalic venous microvasculature that occurs at 5 to 8
weeks gestation. Venous stasis occurs leading to ischemia of the
underlying cortex with laminar cortical necrosis and calcifica-
tion. Symptomatic partial epilepsy that is often progressive
Figure 16.10 MRI: there is a bilateral and symmetrical mantle of tis- occurs in 75% to 90% of patients by age 3 with the initial
sue within the white matter bilaterally that follows gray matter signal seizures often associated with fever. The severity of the epilepsy
on all sequences (arrows) and is consistent with band heterotopias has not been directly related to prognosis but instead 50% to
(double cortex syndrome). 75% of the patients have mental retardation, ADHD, and
recurrent stroke-like episodes. Depending on the size of the
malformation, 25% to 60% develop a hemiparesis and a vascu-
seizures and one infant with infantile spasms but in none did the lar-type headache occurs in about half (65). The initial EEG
seizures appear to correlate with a structural lesion in the brain. may reveal attenuation ipsilateral to the vascular malformation
EEG findings also varied depending on the seizure type from nor- and then subsequent focal and then bilateral epileptiform
mal to focal to generalized discharges (54). Kulkantrakorn and activity (66).
Geller obtained similar results in their retrospective study of 499 Incontinentia pigmenti (Bloch–Sulzberger syndrome) is a
NFI patients (55), while Vivarelli et al. (56) reported a correlation rare neurocutaneous disorder that is X-linked (Xq28) occur-
of seizures in their patients with structural lesions seen on imag- ring predominantly in females as it is lethal in the male. The
ing in 64%. A small percentage (7%) of their 198 patients with disorder involves the skin with a rash that may present at birth
NFI was found on retrospective analysis to have epilepsy (56). on the flexor surface of the limbs and trunk, eventually leading
Tuberous sclerosis (TS) is an autosomal dominant disorder to first linear hyperpigmentation and then hypopigmentation.
that involves the skin, eyes, multiple organs, and the brain. It is Abnormalities of the hair, teeth, and eyes with macular lesions,
a dominant trait with variable low penetrance, but more than retinal detachments, and strabismus are reported. Focal
50% of cases are de novo mutations. There are two genes seizures with onset in the newborn period, abnormal EEGs
involved, TSC1 (hamartin 9q34) and TSC2 (tuberin 16p13). with focal epileptiform discharges, and residual neurologic
More severe neurologic involvement is reported with the latter deficits including developmental retardation and hemiparesis
(57). The patients may have abnormal EEGs often presenting can occur. Microvascular infarcts noted on MRI in the periven-
with seizures in the first year of life as infantile spasms with tricular white matter appear to begin as early as the first week
hypsarrhythmia or atypical hypsarrhythmia or partial seizures of life (Fig. 16.12) (67).
with the subsequent development of the Lennox Gastaut syn- Hypomelanosis of Ito (HI) or incontinentia pigmenti achro-
drome (58). The ultimate cognitive, behavioral (autistic spec- mans is a neuroectodermal disorder characterized by large
trum), and developmental outcome is directly related to the hypopigmented areas that are whorled with various neuronal
earlier onset of epilepsy and the severity of the abnormality of migrational abnormalities such as hemimegencephaly, pachy-
the EEG and proportional to the control of the epilepsy gyria, cortical dysplasia, gray matter heterotopias, gray/white
(Fig. 16.11) (59). matter hamartomas plus abnormal white matter, and neuronal
Seizures and neurobehavioral deficits appear directly related morphology. Mental retardation occurs in the majority, and
to the cerebral tubers (number and tuber burden) that are pres- symptomatic epilepsy with focal EEG abnormalities occurs in
ent in the majority of patients and are characterized by a loss of approximately half the patients. Placantonakis et al. reported
the normal cortical six-layer structure and dysmorphic neu- two patients with hemimegencephaly, HI, and intractable
rons, astrocytes, and giant cells that have a pathologic appear- seizures with focal discharges who successfully underwent
ance similar to cortical dysplasia (60). The greater the number seizure surgery (68).
Chapter 16 ■ The EEG in Congenital Malformations 309

Figure 16.11 A 5-year-old girl with intractable partial-onset symptomatic epilepsy secondary to tuberous sclerosis. EEG:
prominent beta is seen with the patient in the sedated sleeping state. Slow and sharp activity is seen from multiple foci
including left anterior temporal and left parietal regions.

Linear nevus sebaceous syndrome is a neurocutaneous dis- mal neonatal EEGs such as a burst suppression pattern in
order with epidermal sebaceous facial or scalp nevus associated infants who have suffered hypoxic ischemic encephalopathy are
with many of the cerebral malformations reviewed in the sec- associated with a poor outcome for mortality and morbidity as
tion “Congenital Malformations of Cortical Development,” reported by Sinclair et al. (Fig. 16.13) (71).
neurologic deficits, mental retardation, and symptomatic Cerebral palsy by definition “is an umbrella term covering a
epilepsy in more than half the patients and EEG abnormalities. group of nonprogressive but often changing motor impairment
Seizures may present in infancy as infantile spasms with hypsar- syndromes secondary to lesions or anomalies of the brain aris-
rhythmic EEG or as focal seizures. Other epileptic syndromes ing in early stages of its development” (72). The insult or brain
may develop and epilepsy is usually intractable (69). anomaly may occur prenatal, perinatal, or postpartum. There is
no specific etiology. The severity may vary, but there is no pro-
CEREBRAL PALSY gression or improvement, though motor manifestations may
change with age. There are common clinical features such as
Neonatal seizures have a high association with subsequent spasticity, but these are not universal and comorbidities occur
seizures and developmental disabilities including cerebral palsy including abnormal EEGs and symptomatic epilepsy
and developmental delay. Guillet and Kwon found no statistical (Fig. 16.14). Motor findings may be unilateral (hemiplegic) or
difference in outcome in infants discharged home after neona- bilateral (para- or quadriplegic) and abnormal tone may occur
tal seizures on prophylactic phenobarbital or not; 24% had as hypotonia or abnormal movements (choreoathetotic).
seizures, 19% of their patients developed cerebral palsy, and Abnormal EEGs are seen in patients with cerebral palsy without
54% were developmentally delayed (70). As phenobarbital has clinical seizures on no medication, though this may only be for
been shown harmful to newborn brain in the experimental ani- a point in time. The more abnormal the brain is, structurally,
mal model, concern has been expressed regarding treatment, the more vulnerable it is to electrographic disturbances and the
but it is clear that neonatal seizures are also harmful and that clinical manifestation of epilepsy. Russman and Ashwal
neonates with seizures have a poor prognosis. Severely abnor- reviewed studies including 1918 children with cerebral palsy
310 Part III ■ Clinical EEG: General

Figure 16.12 A 6-year-old girl with typical skin lesions of incontentia pigmenti, a disorder inherited
from her mother. She had the onset of symptomatic epilepsy at 3 months of age. Right focal motor
seizures remain difficult to control, her development has been delayed, and she has developed a left
hemiparesis. EEG demonstrates poorly organized background rhythms for age and multifocal spikes
seen in waking and sleeping states.

Figure 16.13 A 3-day-old full-term infant with hypoxic ischemic encephalopathy secondary to pro-
lapsed cord with Apgars of 1, 1, and 4. Seizures developed within a few hours of life and were treated
with phenobarbital and fosphenytoin. He is on the ventilator in a coma with a burst suppression EEG
pattern.
Chapter 16 ■ The EEG in Congenital Malformations 311

Figure 16.14 A 6-year-old boy with cerebral palsy left hemiparesis, developmental delay, and partial
symptomatic epilepsy. His MRI shows areas of tissue loss with gliosis in the inferior posterior right
frontal lobe most likely the sequela of a prior infarct. EEG findings: persistent epileptiform activity as
spike and slow waves from the right centroparietal region activated by sleep.

and found an average of 43% with epilepsy. Severity was related tion, and therapeutic approaches. She writes “EEGs are indi-
to the type of cerebral palsy, onset of seizures as neonate, num- cated for children in whom epilepsy is suspected but it should
ber of seizure medications, intractability, abnormalities on be kept in mind that nonepileptic staring spells are much more
imaging studies, and lower intelligence (73). When periventric- common than absence seizures” (78). A study by Kagan-
ular leukomalacia is present, these children are likely to have Kushnir et al. to establish guidelines for the use of screening
cerebral palsy, cognitive impairment, visual difficulties, and EEGs in autism spectrum disorders to discover subclinical
symptomatic intractable epilepsy usually of multifocal onset epileptiform activity reviewed 25 studies and resulted in a con-
and multiple seizure types (74). clusion that there is insufficient evidence to recommend for or
Ulegyria is scaring of the cortex with thinning and small against routine screening EEGs in autistic patients (79). It has
mushroom-shaped gyri and atrophy at the base of the convolu- been recommended to obtain overnight EEG recordings to cap-
tion and better preserved apex. This is frequently associated ture prolonged sleep in children even without known seizures
with epilepsy and is considered related to perinatal ischemia. but with language or autistic regression, fluctuating deficits or
Kuchukhidze et al. reported on a group of 25 patients with the mute children (76).
typical MRI findings and the comorbidities of cerebral palsy Frank epileptiform abnormalities seen on EEGs in the autis-
and neurologic disabilities and the possibility of seizure surgery tic children such as “rolandic” spikes or benign epileptiform
for their pharmacoresistant seizures (75). Usui et al. (76) car- discharges with centrotemporal spikes (BECTs) have been
ried out seizure surgery on 10 patients with posterior cortex treated with antiepileptic drugs, high-dose steroids, high-dose
epilepsy secondary to ulegyria. Nine had the classic perinatal Valium before bed, and surgery even in patients without appar-
history of hypoxia. Engel’s class I outcome was achieved in ent clinical seizures. There are no evidence-based guidelines
seven (76). available. Approximately one third of individuals with autism
have seizures and these often present after early childhood in
AUTISM/ MENTAL RETARDATION the second and third decades (Fig. 16.15).
Less than 10% of children with autistic symptoms have a
Autism is now considered a developmental disorder with its known etiology such as in the case of Rett syndrome that is
specific features listed in the Diagnostic and Statistical Manual included in the autistic spectrum. Some children with
of Mental Disorders, 4th Edition (DSM-IV) (77). Rapin Angelman, fragile X (FXS), and Down syndromes and TS also
reviewed the disorder’s broad clinical manifestations, evalua- have autistic symptoms.
312 Part III ■ Clinical EEG: General

Figure 16.15 A 17-year-old boy with autism and onset of seizures. Waking EEG was normal. Sleeping
EEG: left temporal spike- and slow-wave transients with underlying slow waves. His seizures were
responsive to Lamictal.

Canitano et al. (80) studied 46 consecutive children who met whom they sequenced the gene SYNGAP1, 2 had seizures and
the DSM-IV criteria for autism with waking and chloral abnormal EEGs “with bioccipital spikes during intermittent
hydrate-induced sleep. Epileptic abnormalities were seen in light stimulation” (82).
35% with only 6 (13%) out of the 16 patients having epilepsy of There are many etiologies though mental retardation is more
various categories. Autism with regression was found in common in boys in part thought to be related to mutations in X-
approximately half and 20% showed epileptiform abnormali- linked genes first described with the FXS. The clinical description
ties on EEG, but only one had seizures. The children with of X-linked mental retardation by Martin and Bell in 1943 (83)
epilepsy were reported as “severely retarded” (80). was followed by a fragile site on the X chromosome found in four
Problems also arise in distinguishing the moderate to severely boys and an unaffected female by Lubs in 1969 (84). Subsequent
retarded child in whom the development of language and social improvement of the culture method by the 1980s allowed the
function is impaired merely due to retardation without neces- identification of what is now considered the most common chro-
sarily attributing another diagnosis or disorder such as autism. mosomal inherited form of mental retardation and developmen-
Mental retardation refers to a significant impairment in cogni- tal delay for which genetic testing is possible. FXS is due to an
tive and general adaptive abilities manifested during the develop- expansion of a trinucleotide (CGG) repeat of the FMR1 gene on
mental period. Retardation may be mild (IQ 50 to 70), moderate the X chromosome. It is the lack of messenger RNA causing lack
(IQ 30 to 50), or severe (IQ 30) and the more severe the more of FMRP protein that causes the syndrome.
likely there are other associated neurologic signs and symptoms It is much more common in males who are dysmorphic with
including seizures and abnormal EEGs. In a population-based large ears, and who in adolescence develop macroorchidism.
study, Steffenburg et al. (81) studied epilepsy in retarded children Their developmental delay is greatest in the area of speech/lan-
6 to 13 years of age. The more severe the retardation, the younger guage and they are often hyperactive. Some are diagnosed as
the age of seizure onset. There were a variety of seizures and some autistic. Approximately 20% will have usually infrequent
children had more than one type. Approximately half had fre- seizures easy to control as was my experience with a boy I fol-
quent, daily to weekly seizures. This group also included children lowed in the 1970s initially with developmental delay and
with cerebral palsy and other neurologic impairment (81). hyperactivity. At age 9, he experienced a brief seizure in his
Nonsyndromic autosomal mental retardation has been iden- sleep with a typical EEG with centrotemporal spikes noted in
tified in patients with various genetic synaptic abnormalities sleep. His family preferred not to treat. His next and last seizure
representing de novo truncating mutations (K138X, R579X, occurred 4 years later. Another son was born much more lan-
and L81RFsX22). Of the 3 nondysmorphic children found out guage impaired but he never experienced a seizure. They both
of a group of 94 mentally retarded patients by Hamdan et al. in were subsequently diagnosed with FXS in the 1980s.
Chapter 16 ■ The EEG in Congenital Malformations 313

Musumeci et al. reported on 192 patients with FXS of whom developmental delay, seizures, and microcephaly in the first
18% had epilepsy mostly focal with a pattern of benign epilepsy year of life. Children have an abnormal EEG with diffuse sharp
with centrotemporal spikes (85). Berry-Kravis (86) reviewed data and slow waves of 2 to 4 Hz, laugh frequently, are overexcitable,
of 136 patients with the FXS and found 11% had seizures easily and extremely active (91). The disorder is related to an abnor-
to control. Twenty of 120 without seizures had EEGs. The most mality of the ubiquitin ligase gene UBE3A located at 15q11.2
common EEG abnormality was centrotemporal spikes. Those (92). In certain brain regions, this gene has monoallelic expres-
with this abnormality had no seizures after childhood (86). sion from maternal chromosome 15. Involvement of three
Rett syndrome was described in 1966 by Rett (87) and is the GABAa receptor subunit genes may explain the severe epilepsy.
second most common cause of X-linked mental retardation now The EEG pattern is typical of a Lennox Gastaut-type pattern
known to be caused by MECP2 gene. It occurs predominantly in with continuous slow spike-wave activity. The EEG patterns are
females who have progressive retardation, microcephaly, and considered characteristic and able to lead to the correct diagno-
characteristic loss of meaningful use of their hands but with sis. Minassian et al. (93) studied the prolonged video EEGs of
stereotypical hand movements. Wark (88) studied the EEG in 14 20 patients with the various pheno- and genotypes of
patients, 8 of whom had simultaneous videos. In one younger Angelman syndrome. The majority (nine) had maternally
patient, the hand movements had a 1:1 time locked relationship inherited 15q11-13 deletions (Class I) with severe intractable
with “rolandic” spikes perhaps eliciting the spikes while the older epilepsy: atypical absences, myoclonus, atonic seizures, and
patient’s spikes were less frequent with the hand movements. flexor spasms. Four had uniparental disomy (Class II): two had
Focal central spike-wave discharges were more common in the myoclonic seizures. Five had methylation imprinting abnor-
younger patients while rhythmic spike-wave discharges probably malities (Class III): one had mild epilepsy and another frequent
ictal were more common in the older (Fig. 16.16) (88) absences and myoclonus, tonic, and atonic seizures. Two had
A patient with Rett syndrome confirmed by genetic analysis mutations in the UBE3A gene (Class IV), with infrequent atyp-
as having a nonsense mutation in the MECP2 gene had deteri- ical absence, myoclonus, and tonic seizures, and in 20% a
oration of her seizure control at age 6. While her EEG was genetic cause could not be identified. They found that “all had
“rolandic” at age 2, her EEG at age 6 was considered typical of diffuse bifrontally dominant high amplitude 1–3 Hz notched or
the “typical” EEG described for Angelman syndrome (89). triphasic or polyphasic slow waves or slow and sharp waves.”
Angelman syndrome described by Angelman consists of The youngest (2 years) Class I had hypsarrhythmia while older
severe mental retardation, microcephaly, ataxic gait, and tremu- patients with eyes closed had alpha background and a 15-year-
lousness jerky movements described as “puppet-like” (90). old had hypnagogic hypersynchrony an immature pattern for
Presenting symptoms are failure to thrive, feeding problems, drowsing at this age (93).

Figure 16.16 A 9-year-old girl with autistic spectrum and hand wringing is found to have Rett syn-
drome. EEG: independent centrotemporal spikes right greater than left activated by sleep.
314 Part III ■ Clinical EEG: General

Korff et al. (94) looked for notched delta at a large tertiary Thirty (83%) had seizures, 43% febrile as the initial seizure.
care children’s hospital and found retrospectively 1.1% of video Ninety-two percent of the adults had seizures: myoclonic, atyp-
EEGs had this pattern. The specificity for Angelman was 38% ical absences. Sixty-six percent of the 150 EEGs contained
with the youngest 14 months of age. Seven of the 14 patients triphasic anterior delta in 83% of patients. This was found prior
who had Angelman syndrome features and notched delta did to the diagnoses in almost half. The authors note that high-
not appear to have genetic testing for this disorder even though amplitude rhythmic 4- to 6-Hz theta up to 12 years in retarded
they were returning for outpatient visits to a tertiary care cen- patients should suggest Angelman syndrome (Fig. 16.17) (96).
ter (94). This chapter highlights how an EEG finding can alert Guerrini et al. studied the almost continuous rhythmic
to a syndrome diagnosis, though of course it is not necessarily myoclonus involving hands and face accompanied by rhythmic
pathonemonic for the syndrome. 5- to 10-Hz EEG activity in 11 patients with Angelman syn-
Valente et al. (95) reviewed 26 patients with Angelman syn- drome and concluded that this is related to a unique pattern of
drome with 70 EEGs and 15 videos. The major abnormality was fast-bursting cortical myoclonus (97).
a delta pattern seen in 22 including a hypsarrhythmic variant in Down syndrome (trisomy 21) is the most common of the
addition to the notched delta. Other less-specific abnormalities autosomal trisomy syndromes. The children have a characteristic
such as theta seen diffusely or posteriorly and posterior dis- facies, mental retardation from mild to severe, and at times dis-
charges were described (95). Laan et al. (96) follow the evolu- orders of other organ systems. Seizures such as infantile spasms
tion of epilepsy and EEG findings in Angelman syndrome in 36 or the Lennox Gastuat syndrome can occur in the more severely
patients (15q11–13 deletion) over a 1 to 39 years follow-up. impaired but the incidence of epilepsy is low (Fig. 16.18) (98). A

Figure 16.17 A 12-year-old girl with Angelman syndrome and seizures. EEG is lacking age-appropriate background and
has high-voltage 2- to 3-Hz delta with notching or spike activity that is diffuse and often not symmetrical.
Chapter 16 ■ The EEG in Congenital Malformations 315

Figure 16.18 A 28-year-old man with


Down syndrome and moderate mental
retardation who has intractable drop
seizures associated with a Lennox Gastaut
EEG pattern. EEG: note poor development
of age-appropriate waking rhythms and
slow spike-wave discharges with intermit-
tent attenuations.

study of 18 matched Down syndrome patients with normal con- Objections have been raised regarding the true scientific
trols and FAS found slower activity but 7 out of 8 kept their eyes validity of this diagnosis as “the most common neurobehavioral
open tending to block age-appropriate alpha (25). The EEG in condition of childhood” not based on the disorder as a disease,
patients with Down syndrome can be quite normal, especially in but on a group of symptoms secondary to a result of the behav-
childhood to young adulthood, if they can cooperate and are not ioral manifestations of a host of psychological, psychiatric, and
markedly cognitively impaired (Fig. 16.19). educational difficulties. There is a tendency to use apparent
Mental/developmental retardation may be caused by a num- response to medication with stimulants or atomoxetine as a
ber of endocrine and metabolic disorders that at this time in “proof” of the diagnosis (101).
many countries are screened for at the time of birth such as Specific EEG studies regarding ADD/ADHD have been
hypothyroidism. One still has to be aware of neonates present- unrewarding. Recent literature regarding EEG and ADHD has
ing with myoclonus and burst suppression EEGs due to nonke- focused on quantitative EEG (QEEG). A meta-analysis of these
totic hyperglycinemia, or other inborn errors such as in the numerous studies culled down to nine observed increased theta
urea cycle, or an infant with infantile spasms with a hypsar- and decreased beta power compared to controls. As these inves-
rhythmic EEG having B6 deficiency or dependency. Other rare tigators observed this change in theta/beta ratio may arise with
disorders such as glucose transporter type I deficiency syn- other conditions, a prospective study is indicated especially for
drome can present with intractable seizures early in life with control of many aspects such as mental state, alertness, and
severely abnormal EEGs. It is beyond the scope of this chapter medication (102).
to review these entities (99). Epileptiform abnormalities in the EEGs of children with
ADD/ADHD have been described in the reports of Hughes et
ADD/ ADHD al. (103) and Richer et al. (104). Hughes reported on all
ADHD children without a clinical history of seizures, retarda-
ADD/ADHD is a controversial diagnosis with guidelines avail- tion, or psychiatric symptoms referred to a children’s hospital
able from the American Academy of Pediatrics and specific pediatric neurology clinic and found 30.1% had definite
diagnostic criteria under DSM-IV (77). Previous terms now “non- controversial” spike activity. The EEGs usually included
discarded were minimal brain damage or dysfunction (MBD). sleep with focal spikes more common than generalized.
Patients may also have learning disabilities including dyslexia or Richer’s group reported epileptiform activity in twice as many
behavioral disorders (oppositional defiant disorder) and psy- children with ADHD as controls 1.3% versus 0.6% mostly
chiatric disorders such as bipolar disorder. The diagnosis con- elicited by hyperventilation or photic stimulation. Only 3 of
sists of a list of symptoms noted by parents and teachers with 21 went on to develop epilepsy and they concluded there is no
onset prior to age 7. These are mainly inattention, impulsivity, clinical utility in using EEG to diagnosis epilepsy in children
and hyperactivity. These symptoms are more common in boys, with ADHD. Of course children with inattention could be
are frequently familial, and have a maturational aspect though having absence seizures and a careful history is always warran -
some patients never seem to outgrow the disorder (100). ted (Fig. 16.20).
316 Part III ■ Clinical EEG: General

Figure 16.19 A 17-year-old man with Down syndrome who had an episode of loss of consciousness. Patient attends spe-
cial education and is only mildly retarded. His EEG is normal.

Figure 16.20 A 6-year-old boy with


ADHD has no history of seizures, though
he is said to be a restless sleeper. His EEG
shows right centrotemporal spikes in
sleep.
Chapter 16 ■ The EEG in Congenital Malformations 317

Figure 16.21 A 6-year-old girl with a


recent seizure out of sleep after going to
sleep several hours later than usual. Her
10-year-old sister has the same EEG pat-
tern and clinical history. They are in regu-
lar classes in school doing well and are on
no medications. EEG: abundant independ-
ent left centrotemporal spikes and sharp
waves at times reflected to the right hemi-
sphere activated by sleep.

A higher than expected occurrence of ADD/ADHD has been 8. Faillace WJ, Canady AI. Cerebrospinal fluid shunt malfunction
described in children with epilepsy. In a group of children with signaled by new or recurrent seizures. Childs Nerv Syst. 1990;6(1):
benign epilepsies and otherwise negative neurologic history, 37–40.
but with 25 males to 15 females, 70% had ADHD by DSM-IV 9. Sarnat HB, Flores-Sarnat L. Neuropathologic research strategies in
holoprosencephaly. J Child Neurol. 2001;16(12):918–931.
criteria with it more common in the females (105).
10. Patterson MC. Holoprosencephaly: the face predicts the brain; the
There have been no specific EEG findings currently noted in
image predicts its function. Neurology. 2002;59(12):1833–1834.
children with dyslexia or any of the learning disabilities. There 11. Lewis AJ, Simon EM, Barkovich AJ, et al. Middle interhemispheric
is controversy regarding the epileptic or spike burden in chil- variant of holoprosencephaly: a distinct cliniconeuroradiologic
dren with epilepsy, even the “benign” asymptomatic types with subtype. Neurology. 2002;59(12):1860–1865.
or without treatment who do not perform as well scholastically 12. Clegg NJ, Gerace KL, Sparagana SP, et al. Holoprosencephaly: a
or on at least some neuropsychological tests as their siblings or review. Am J EEG Technol. 2002;42:59–72.
peer group (Fig. 16.21) (106,107). 13. Packard AM, Miller VS, Delgado MR. Schizencephaly: correlations
of clinical and radiologic features. Neurology. 1997;48(5):
1427–1434.
REFERENCES 14. Granata T, Battaglia G, D’Incerti L, et al. Schizencephaly: neurora-
1. McAbee GN, Chan A, Erde EL. Prolonged survival with hydranen- diologic and epileptologic findings. Epilepsia. 1996;37(12):
cephaly: report of two patients and literature review. Pediatr 1185–1193.
Neurol. 2000;23(1):80–84. 15. Iannetti P, Nigro G, Spalice A, et al. Cytomegalovirus infection and
2. Velasco M, Velasco F, Gardea G, et al. Polygraphic characterization schizencephaly: case reports. Ann Neurol. 1998;43(1):123–127.
of the sleep–epilepsy patterns in a hydranencephalic child with 16. Paladin F, Chiron C, Dulac O, et al. Electroencephalographic
severe generalized seizures of the Lennox–Gastaut syndrome. Arch aspects of hemimegalencephaly. Dev Med Child Neurol.
Med Res. 1997;28(2):297–302. 1989;31(3):377–383.
3. Ines DF, Markand ON. Epileptic seizures and abnormal electroen- 17. Aicardi J, Lefebvre J, Lerique-Koechlin A. A new syndrome: spasms
cephalographic findings in hydrocephalus and their relation to the in flexion, callosal agenesis, ocular abnormalities. Electroencephalogr
shunting procedures. Electroencephalogr Clin Neurophysiol. Clin Neurophysiol. 1965;19:609–610.
1977;42(6):761–768. 18. Fariello RG, Chun RW, Doro JM, et al. EEG recognition of
4. Bartoshesky LE, Haller J, Scott RM, et al. Seizures in children with Aicardi’s syndrome. Arch Neurol. 1977;34(9):563–566.
meningomyelocele. Am J Dis Child. 1985;139(4):400–402. 19. Erenberg G. Aicardi’s syndrome: report of an autopsied case and
5. Saukkonen AL, Serlo W, von Wendt L. Epilepsy in hydrocephalic review of the literature. Cleve Clin Q. 1983;50(3):341–345.
children. Acta Paediatr Scand. 1990;79(2):212–218. 20. Lacey DJ. Agenesis of the corpus callosum. Clinical features in 40
6. Noetzel MJ, Blake JN. Seizures in children with congenital hydro- children. Am J Dis Child. 1985;139(9):953–955.
cephalus: long-term outcome. Neurology. 1992;42(7):1277–1281. 21. Kolodny EH. Agenesis of the corpus callosum: a marker for inher-
7. Hack CH, Enrile BG, Donat JF, et al. Seizures in relation to shunt ited metabolic disease? Neurology. 1989;39(6):847–848.
dysfunction in children with meningomyelocele. J Pediatr. 22. Gastaut H, Regis H, Gastaut JL, et al. Lipomas of the corpus callo-
1990;116(1):57–60. sum and epilepsy. Neurology. 1980;30(2):132–138.
318 Part III ■ Clinical EEG: General

23. Bale JF Jr, Murph JR. Congenital infections and the nervous sys- 45. Fong J, Wu G, Wyllie E, et al. Interictal hypermetabolic subcortical
tem. Pediatr Clin North Am. 1992;39(4):669–690. band on brain FDG–PET in doublecortin mutation. Neurology.
24. Jones KL, Smith DW. Recognition of the fetal alcohol syndrome in 2008;71(7):535.
early infancy. Lancet. 1973;2(7836):999–1001. 46. Dobyns WB, Andermann E, Andermann F, et al. X-linked mal-
25. Kaneko WM, Phillips EL, Riley EP, et al. EEG findings in fetal alco- formations of neuronal migration. Neurology. 1996;47(2):
hol syndrome and Down syndrome children. Electroencephalogr 331–339.
Clin Neurophysiol. 1996;98(1):20–28. 47. Gastaut H, Pinsard N, Raybaud C, et al. Lissencephaly
26. Chasnoff IJ, Griffith DR, Freier C, et al. Cocaine/polydrug use in (agyria–pachygyria): clinical findings and serial EEG studies. Dev
pregnancy: two-year follow-up. Pediatrics. 1992;89(2):284–289. Med Child Neurol. 1987;29(2):167–180.
27. Dominguez R, Aguirre Vila-Coro A, Slopis JM, et al. Brain and 48. de Rijk van Andel JF, Arts WF, de Weerd AW. EEG in type I
ocular abnormalities in infants with in utero exposure to cocaine lissencephaly [letter to the editor]. Dev Med Child Neurol.
and other street drugs. Am J Dis Child. 1991;145(6):688–695. 1988;30(1):126–127.
28. Lairy GC. The evolution of the EEG from birth to adulthood. In: 49. Sébire G, Goutieres F, Tardieu M, et al. Extensive macrogyri or no
Handbook of Electroencephalography and Clinical Neurophysiology. visible gyri: distinct clinical, electroencephalographic, and genetic
Vol 6B. Amsterdam: Elsevier; 1973:3–117. features according to different imaging patterns. Neurology.
29. Jan JE, Wong PKH. Behaviour of the alpha rhythm in electroen- 1995;45(6):1105–1111.
cephalograms of visually impaired children. Dev Med Child 50. Kuzniecky R, Andermann F, Guerrini R. Congenital bilateral peri-
Neurol. 1988;30(4):444–450. sylvian syndrome: study of 31 patients. The CBPS Multicenter
30. Kriegseis A, Hennighausen E, Rösler F, et al. Reduced EEG alpha Collaborative Study. Lancet. 1993;341(8845):608–612.
activity over parieto-occipital brain areas in congenitally blind 51. Chang BS, Piao X, Giannini C, et al. Bilateral generalized polymi-
adults. Clin Neurophysiol. 2006;117(7):1560–1573. crogyria (BGP): a distinct syndrome of cortical malformation.
31. Ho SS, Kuzniecky RI, Gilliam F, et al. Congenital porencephaly and Neurology. 2004;62(10):1722–1728.
hippocampal sclerosis. Clinical features and epileptic spectrum. 52. Guerrini R, Genton P, Bureau M, et al. Multilobar polymicrogyria,
Neurology. 1997;49(5):1382–1388. intractable drop attack seizures, and sleep-related electrical status
32. Gandy SE, Heier LA. Clinical and magnetic resonance features of epilepticus. Neurology. 1998;51(2):504–512.
primary intracranial arachnoid cysts. Ann Neurol. 1987;21(4): 53. Young H, Hyman S, North K. Neurofibromatosis 1: clinical review
342–348. and exceptions to the rules. J Child Neurol. 2002;17(8):613–621
33. Arroyo S, Santamaria J. What is the relationship between arach- [discussion 27–29, 46–51].
noid cysts and seizure foci? Epilepsia. 1997;38(10):1098–1102. 54. Korf BR, Carrazana E, Holmes GL. Patterns of seizures observed in
34. Barkovich AJ, Kuzniecky RI, Jackson GD, et al. Classification sys- association with neurofibromatosis 1. Epilepsia. 1993;34(4):
tem for malformations of cortical development: update 2001. 616–620.
Neurology. 2001;57(12):2168–2178. 55. Kulkantrakorn K, Geller TJ. Seizures in neurofibromatosis 1.
35. Tassi L, Colombo N, Garbelli R, et al. Focal cortical dysplasia: neu- Pediatr Neurol. 1998;19(5):347–350.
ropathological subtypes, EEG, neuroimaging and surgical out- 56. Vivarelli R, Grosso S, Calabrese F, et al. Epilepsy in neurofibro-
come. Brain. 2002;125(Pt 8):1719–1732. matosis 1. J Child Neurol. 2003;18(5):338–342.
36. Palmini A, Najm I, Avanzini G, et al. Terminology and classification 57. Crino PB, Nathanson KL, Henske EP. The tuberous sclerosis com-
of the cortical dysplasias. Neurology. 2004;62(6 suppl 3):S2–S8. plex. N Engl J Med. 2006;355(13):1345–1356.
37. Krsek P, Maton B, Korman B, et al. Different features of 58. Pampiglione G, Moynahan EJ. The tuberous sclerosis syndrome:
histopathological subtypes of pediatric focal cortical dysplasia. clinical and EEG studies in 100 children. J Neurol Neurosurg
Ann Neurol. 2008;63(6):758–769. Psychiatry. 1976;39(7):666–673.
38. Kim DW, Lee SK, Chu K, et al. Predictors of surgical outcome and 59. Muzykewicz DA, Costello DJ, Halpern EF, et al. Infantile spasms in
pathologic considerations in focal cortical dysplasia. Neurology. tuberous sclerosis complex: prognostic utility of EEG. Epilepsia.
2009;72(3):211–216. 2008;50(2):290–296.
39. d’Orsi G, Tinuper P, Bisulli F, et al. Clinical features and long term 60. Mackay MT, Becker LE, Chuang SH, et al. Malformations of corti-
outcome of epilepsy in periventricular nodular heterotopia. cal development with balloon cells: clinical and radiologic corre-
Simple compared with plus forms. J Neurol Neurosurg Psychiatry. lates. Neurology. 2003;60(4):580–587.
2004;75(6):873–878. 61. Jarrar RG, Buchhalter JR, Raffel C. Long-term outcome of epilepsy
40. Battaglia G, Chiapparini L, Franceschetti S, et al. Periventricular surgery in patients with tuberous sclerosis. Neurology. 2004;62(3):
nodular heterotopia: classification, epileptic history, and genesis of 479–481.
epileptic discharges. Epilepsia. 2006;47(1):86–97. 62. Jansen FE, Braams O, Vincken KL, et al. Overlapping neurologic
41. Sisodiya SM, Free SL, Thom M, et al. Evidence for nodular epilep- and cognitive phenotypes in patients with TSC1 or TSC2 muta-
togenicity and gender differences in periventricular nodular het- tions. Neurology. 2008;70(12):908–915.
erotopia. Neurology. 1999;52(2):336–341. 63. Jansen FE, Vincken KL, Algra A, et al. Cognitive impairment in
42. Sicca F, Kelemen A, Genton P, et al. Mosaic mutations of the LIS1 tuberous sclerosis complex is a multifactorial condition.
gene cause subcortical band heterotopia. Neurology. 2003;61(8): Neurology. 2008;70(12):916–923.
1042–1046. 64. Major P, Rakowski S, Simon MV, et al. Are cortical tubers epilepto-
43. Mai R, Tassi L, Cossu M, et al. A neuropathological, stereo-EEG, genic? Evidence from electrocorticography. Epilepsia. 2009;50(1):
and MRI study of subcortical band heterotopia. Neurology. 147–154.
2003;60(11):1834–1838. 65. Thomas-Sohl KA, Vaslow DF, Maria BL. Sturge–Weber syndrome:
44. Ono J, Mano T, Andermann E, et al. Band heterotopia or double a review. Pediatr Neurol. 2004;30(5):303–310.
cortex in a male: bridging structures suggest abnormality of the 66. Comi AM. Advances in Sturge–Weber syndrome. Curr Opin
radial glial guide system. Neurology. 1997;48(6):1701–1703. Neurol. 2006;19(2):124–128.
Chapter 16 ■ The EEG in Congenital Malformations 319

67. Hennel SJ, Ekert PG, Volpe JJ, et al. Insights into the pathogenesis 87. Rett A. On a unusual brain atrophy syndrome in hyperammone-
of cerebral lesions in incontinentia pigmenti. Pediatr Neurol. mia in childhood. Wien Med Wochenschr. 1966;116(37):723–726.
2003;29(2):148–150. 88. Wark J. The electroencephalogram in Rett syndrome: correlation
68. Placantonakis DG, Ney G, Edgar M, et al. Neurosurgical manage- with hand movements. J Child Neurol. 1993;8(1):101–105.
ment of medically intractable epilepsy associated with hypome- 89. Laan LA, Vein AA. A Rett patient with a typical Angelman EEG.
lanosis of Ito. Epilepsia. 2005;46(2):329–331. Epilepsia. 2002;43(12):1590–1592.
69. Menascu S, Donner EJ. Linear nevus sebaceous syndrome: case 90. Angelman H. “Puppet children”. A report of three cases. Dev Med
reports and review of the literature. Pediatr Neurol. 2008;38(3): Child Neurol. 1965;7:681–688.
207–210. 91. Zori RT, Hendrickson J, Woolven S, et al. Angelman syndrome:
70. Guillet R, Kwon J. Seizure recurrence and developmental clinical profile. J Child Neurol. 1992;7(3):270–280.
disabilities after neonatal seizures: outcomes are unrelated to 92. Williams CA. Neurological aspects of the Angelman syndrome.
use of phenobarbital prophylaxis. J Child Neurol. 2007;22(4): Brain Dev. 2005;27(2):88–94.
389–395. 93. Minassian BA, DeLorey TM, Olsen RW, et al. Angelman syn-
71. Sinclair DB, Campbell M, Byrne P, et al. EEG and long-term out- drome: correlations between epilepsy phenotypes and genotypes.
come of term infants with neonatal hypoxic-ischemic Ann Neurol. 1998;43(4):485–493.
encephalopathy. Clin Neurophysiol. 1999;110(4):655–659. 94. Korff CM, Kelley KR, Nordli DR Jr. Notched delta, phenotype, and
72. Mutch L, Alberman E, Hagberg B, et al. Cerebral palsy epidemiol- Angelman syndrome. J Clin Neurophysiol. 2005;22(4):238–243.
ogy: where are we now and where are we going? Dev Med Child 95. Valente KD, Andrade JQ, Grossmann RM, et al. Angelman syn-
Neurol. 1992;34(6):547–551. drome: difficulties in EEG pattern recognition and possible mis-
73. Russman BS, Ashwal S. Evaluation of the child with cerebral palsy. interpretations. Epilepsia. 2003;44(8):1051–1063.
Semin Pediatr Neurol. 2004;11(1):47–57. 96. Laan LA, Renier WO, Arts WF, et al. Evolution of epilepsy and
74. Gurses C, Gross DW, Andermann F, et al. Periventricular leukoma- EEG findings in Angelman syndrome. Epilepsia. 1997;38(2):
lacia and epilepsy: incidence and seizure pattern. Neurol. 195–199.
1999;52(2):341–345. 97. Guerrini R, De Lorey TM, Bonanni P, et al. Cortical myoclonus
75. Kuchukhidze G, Unterberger I, Dobesberger J, et al. Electroclinical in Angelman syndrome. Ann Neurol. 1996;40(1):39–48.
and imaging findings in ulegyria and epilepsy: a study on 25 98. Stafstrom CE, Patxot OF, Gilmore HE, et al. Seizures in children
patients. J Neurol Neurosurg Psychiatry. 2008;79(5):547–552. with Down syndrome: etiology, characteristics and outcome. Dev
76. Usui N, Mihara T, Baba K, et al. Posterior cortex epilepsy second- Med Child Neurol. 1991;33(3):191–200.
ary to ulegyria: is it a surgically remediable syndrome? Epilepsia. 99. Wolf NI, Bast T, Surtees R. Epilepsy in inborn errors of metabo-
2008;49(12):1998–2007. lism. Epileptic Disord. 2005;7(2):67–81.
77. American Psychiatric Association. Diagnostic and Statistical 100. Wolraich ML. Attention-deficit hyperactivity disorder. Semin
Manual of Mental Disorders. 4th ed. Washington, DC: American Pediatr Neurol. 2006;13(4):279–285.
Psychiatric Association; 1994. 101. Furman L. What is attention-deficit hyperactivity disorder
78. Rapin I. Autism. N Engl J Med. 1997;337(2):97–104. (ADHD)? J Child Neurol. 2005;20(12):994–1002.
79. Kagan-Kushnir T, Roberts SW, Snead OC 3rd. Screening elec- 102. Snyder SM, Hall JR. A meta-analysis of quantitative EEG power
troencephalograms in autism spectrum disorders: evidence-based associated with attention-deficit hyperactivity disorder. J Clin
guideline. J Child Neurol. 2005;20(3):197–206. Neurophysiol. 2006;23(5):440–455.
80. Canitano R, Luchetti A, Zappella M. Epilepsy, electroencephalo- 103. Hughes JR, DeLeo AJ, Melyn MA. The electroencephalogram in
graphic abnormalities, and regression in children with autism. J attention deficit-hyperactivity disorder: emphasis on epilepti-
Child Neurol. 2005;20(1):27–31. form discharges. Epilepsy Behav. 2000;1(4):271–277.
81. Steffenburg U, Hagberg G, Kyllerman M. Characteristics of 104. Richer LP, Shevell MI, Rosenblatt BR. Epileptiform abnormalities
seizures in a population-based series of mentally retarded children in children with attention-deficit-hyperactivity disorder. Pediatr
with active epilepsy. Epilepsia. 1996;37(9):850–856. Neurol. 2002;26(2):125–129.
82. Hamdan FF, Gauthier J, Spiegelman D, et al. Mutations in SYN- 105. Zelnik N, Sa’adi L, Silman-Stolar Z, et al. Seizure control and
GAP1 in autosomal nonsyndromic mental retardation. N Engl J educational outcome in childhood-onset epilepsy. J Child Neurol.
Med. 2009;360(6):599–605. 2001;16(11):820–824.
83. Martin JP, Bell J. A pedigree of mental defect showing sex linkage. 106. Riva D, Vago C, Franceschetti S, et al. Intellectual and language
J Neurol Neurosurg Psychiatry. 1943;6:154–156. findings and their relationship to EEG characteristics in benign
84. Lubs HA. A marker X chromosome. Am J Hum Genet. 1969;21(3): childhood epilepsy with centrotemporal spikes. Epilepsy Behav.
231–244. 2007;10(2):278–285.
85. Musumeci SA, Hagerman RJ, Ferri R, et al. Epilepsy and EEG find- 107. Nicolai J, van der Linden I, Arends JB, et al. EEG characteristics
ings in males with fragile X syndrome. Epilepsia. 1999;40(8): related to educational impairments in children with benign
1092–1099. childhood epilepsy with centrotemporal spikes. Epilepsia.
86. Berry-Kravis E. Epilepsy in fragile X syndrome. Dev Med Child 2007;48(11):2093–2100.
Neurol. 2002;44(11):724–728.
CHAPTER
Brain Tumors and Other
Space-Occupying Lesions
ADAM L. HARTMAN AND RONALD P. LESSER
17
GENERAL OVERVIEW amic lesions or lesions causing cerebral edema; generalized
slowing was seen after lesions were made in the midbrain
Brain tumors are a leading cause for new-onset seizures in the tegmental area. Solid tumors generally are not epileptogenic in
adults and many patients still present with seizures as their first themselves; it is the infiltrated or surrounding nontumor tissue
manifestation of a tumor (Fig. 17.1). Although EEG at one time that causes the seizures (2,3). Notable exceptions include devel-
was important for diagnosing brain tumors and other space- opmental tumors such as hypothalamic hamartomas (HH; dis-
occupying lesions, this is now more commonly done using cussed in the section “Hypothalamic Hamartoma”). EEG
imaging studies, such as computed tomography (CT) and mag- findings in most nondevelopmental tumors likely are caused by
netic resonance imaging (MRI) (Fig. 17.2). Clinical neurophys- displacement of normal tissue (either by the mass itself or by
iology still is important in managing these patients, including altered flow of cerebrospinal fluid), physical disruption of
the use of electrocorticography (ECoG) during testing to iden- normal neuronal circuitry, ischemia, or hemosiderin deposition
tify eloquent cortex during resection surgeries, application of (4,5), gap-junction proteins (6), and alterations in neurotrans-
evoked potentials in assessing the location of sensorimotor cor- mitter pathophysiology, such as vesicular glutamate trans-
tex or the extent of tumor involvement, and the application of porters (7), both ionotropic and metabotropic glutamate
magnetoencephalography, for both magnetic source imaging receptor subunits (8), and GABA receptors (9). Data on
(e.g., in localizing spike-generating zones) and functional map- patients with brain tumors provide some of the most convinc-
ping. These topics will be discussed in this chapter. ing arguments for secondary epileptogenesis (wherein an
The EEG findings discussed in this chapter are not specific epileptogenic region gives rise to a secondary epileptogenic
for tumor type and also can occur with other space-occupying focus via strong connectivity between the two regions) (10).
lesions (hence, their inclusion in this chapter). In a series of
classical experiments Gloor et al. (1) found that focal delta-
range slowing was seen over well-circumscribed lesions in the Epidemiology
white matter (with no apparent change in faster frequency The prevalence of seizures depends in part on type of tumor:
activity) or after a focal thalamic lesion, while diffuse hemi- for example, anywhere from 66% to 90% of patients with
spheric slowing was seen after larger thalamic and/or hypothal- low-grade astrocytomas have seizures at some point (11).

Figure 17.1 EEG from a 4-year-old male who presented to Pediatric Epilepsy Clinic with a 2-month history of episodes of
staring and fumbling with the fingers that lasted about 30 seconds. The EEG showed a burst of slowing in the left hemi-
sphere (left figure, fourth second) and runs of periodically repeating high-voltage rhythmical sharp activity over the left
hemisphere region during sleep (right figure). A longitudinal bipolar montage is shown.

321
322 Part III ■ Clinical EEG: General

Figure 17.2 MRI of the patient from Figure 17.1.


Axial sections (FLAIR and T2-weighted sequences)
at the level of the temporal lobes show cystic
changes, swelling, and infiltration of normal brain
parenchyma in the left anterior mesial temporal
lobe. The patient had an MRI soon after his clinic
appointment, underwent a near-total resection of a
WHO grade II astrocytoma, and has remained
seizure-free since surgery.

Seizures are more common in patients with low-grade (vs. LOCALIZATION-RELATED ISSUES
high-grade) tumors (12). They are the presenting symptom in
up to 76% of children with benign astrocytic and oligoden- Hemispheric Tumors
drocytic tumors of the cerebral hemispheres (13). Seizures Cortical tumors produce abnormal EEGs 96% of the time,
occurred in 40% of children with hemispheric brain tumors including focal delta activity (correctly localized) (Figs. 17.1 and
(with the temporal lobe being most commonly involved), 17.2); a minority causes focal dysrhythmia, but EEGs can be
while none with thalamic tumors had seizures (14). There was falsely localizing (Figs. 17.3 and 17.4) (20). EEGs are abnormal
a focal component to the majority of the seizures in that in up to 87% of children with benign astrocytic and oligoden-
series, although nearly 25% of the patients had only general- drocytic tumors of the cerebral hemispheres (13). EEG findings
ized seizures. Seizures can occur in children with supratento- are generally related to location of the tumor and rapidity of
rial or infratentorial tumors, although they are more growth. Most (4,21), but not all (22), studies associate rapidly
commonly the presenting sign in hemispheric tumors (15).
Conversely, only up to 11% of patients with a “possible
seizure” have brain tumors diagnosed (16).

Historical Perspectives on the Utility of EEG


Before the advent of modern imaging, EEG was a critical com-
ponent in the evaluation of patients with suspected masses,
along with pneumoencephalography, ventriculography, and
angiography. In fact, serial EEGs were somewhat useful in dis-
tinguishing strokes (where EEG abnormalities resolved over
time) from tumors (where EEG abnormalities became pro-
gressively worse) (4). Postoperative EEGs were used to moni-
tor tumor recurrence, which was heralded by focal arrhythmic
slow activity at the former tumor site; a recent study found
that EEG slowing indicated that postoperative seizure control
was less likely (17). Much of the literature on EEG in tumors
and other space-occupying lesions is older and studies were
performed without the modern standards of statistical rigor.
The classification of tumors also has evolved over time, so we Figure 17.3 EEG from the end of a seizure in a 79-year-old male with
attempted to be as nonspecific as possible in describing tumor a right frontal astrocytoma. Phase reversals are present in the right
type in older studies, while using current tumor nomenclature frontal, temporal, and parietal leads. Also, there is a prominent phase
for more recent studies. One final caveat is that many reversal in the left parietal region (channels 12 and 13). However, with
observers have noted that the routine EEG can be normal or the transverse montage used, the channels with the most prominent
even misleading in terms of lateralization (14,15,18,19), thus activity only indicate where the differences between the recording elec-
limiting the utility of this test in some instances. In fact, nor- trodes are the greatest. These are not necessarily were the actual ampli-
mal frequency activity may be seen ipsilateral to a tumor (18). tudes are greatest.
Chapter 17 ■ Brain Tumors and Other Space-Occupying Lesions 323

growing tumors with very slow delta-range activity, and more sharp waves occurred in half of these patients. In over 90% of
slowly growing tumors with arrhythmic theta-range activity temporal gliomas, slowing was continuous (27). A more recent
with occasional intermixed epileptiform spikes and sharp series (which included prolonged video–EEG monitoring)
waves. It should be noted that up to one third of patients with noted temporal epileptiform discharges and focal temporal
brain tumors have an epileptogenic focus remote from the slowing as the most common finding with temporal lobe
tumor site, which may give rise to some confusion in localiza- tumors, although a few patients also showed bilateral, general-
tion (10). Meningiomas and other slow-growing or midline ized, or false localization (28).
tumors may be difficult to localize, although hyperventilation
may reveal a focus of slowing not seen otherwise (20,23). Focal Posterior Fossa and “Deep” Tumors
theta-range activity can occur with any type of tumor and one Because most tumors in children over 1 year of age are infraten-
series noted it in 75% of patients with hemispheric tumors torial, it is important to consider posterior fossa and so-called
(18). In that study, focal delta-range activity was most common “deep” (e.g., sellar, chiasmal, and diencephalic) tumors separately.
in patients with glioblastoma multiforme, although it also Although most of these patients have had an imaging study, the
occurred with meningeal tumors. Activity in the delta range presenting symptoms can be so nonspecific (e.g., altered mental
was less frequent with astrocytomas, where focal sharp activity status) that an EEG might be obtained for other reasons in the
was more common. Focal epileptiform activity on the EEG course of a patient’s treatment (e.g., to rule out nonconvulsive
appeared to be correlated generally with clinical seizures in status epilepticus). Therefore, it is important to understand
patients with astrocytomas, but clinical seizures were not as EEG findings in this context so that an appropriate interpreta-
common among patients with meningeal tumors, even though tion of EEG patterns can be made. One found that over three
they also had a fair amount of focal epileptiform activity on fourth of records was abnormal in children with posterior fossa
EEG, seizures were not as common. Newmark et al. found that tumors, with likelihood of abnormality decreasing with age
epileptiform activity was unrelated to focal slowing in patients (from below 10 to over 50 years) (23). However, two other stud-
with gliomas (22). Patients with multiple metastases also may ies found EEGs to be normal in 19% (15) and 24% (19) of chil-
have a good deal of epileptiform activity in their EEGs; over dren. Location within the posterior fossa may be important.
90% of patients with metastatic disease have abnormal EEGs EEGs were abnormal in only 30% of patients with brainstem
including epileptiform activity (4), delta-range activity, and tumors, while over 80% of EEGs were abnormal in those with
dysrhythmias (20).
Recording from the cortical surface shows similar findings.
While focal delta-range activity and/or background attenuation
may be seen with some hemispheric tumors, focal slowing can
be seen from an area literally centimeters away from the actual
tumor (24). Those authors suggested that one potentially help-
ful localizing sign is resistance to afterdischarge occurrence,
even at high current intensities, during cortical stimulation.
Focal delta- and theta-range activity correlates with white
matter involvement of tumors (1,22). This includes tumors
involving both gray and white matter as well as those involving
white matter only. Rhythmic delta activity may indicate
involvement of the thalamus, although involvement of deep
frontal white matter may produce similar findings (22).
Attenuation of background activity is seen in patients with
high-grade gliomas that have thalamic involvement (22) but
also may be seen in regions with extensive peritumoral edema
(25), although this is a nonspecific finding. Irregular delta-
range activity is unrelated to peritumoral edema (22) and the
degree of EEG abnormality does not appear to correlate with
degree of edema (25).
Tumor was present in 18% of series of 282 patients showing
typical periodic lateralized epileptiform discharges (PLEDs) (26).
Frontal tumors tend to produce the slow patterns noted pre-
viously, at times showing frontal intermittent rhythmic dis-
charges (FIRDA). Parietal and occipital tumors may affect the Figure 17.4 EEG from a 56-year-old female with a burst of high-volt-
posterior basic rhythm, but also may show abnormalities in age sharply contoured delta-range activity that was bilateral (longitudi-
more anterior regions, possibly leading to false localization nal bipolar recordings, channels 1 to 8) but maximal over the left
(Fig. 17.4). Temporal tumors can show a unilateral abnormality hemisphere (channels 11 to 16). Her underlying pathology was a left
up to 89% of the time (Fig. 17.1) (27). Focal delta activity or parietal astrocytoma.
324 Part III ■ Clinical EEG: General

cerebellar or fourth ventricle tumors (19). In that series, 27% epileptiform spikes have deep sources in the neighborhood of
had posterior rhythmic delta waves, 32% had generalized bilat- the hamartoma, with later spread to cortical areas (36). A
eral bursts of rhythmic slowing, 51% had posterior arrhythmic subsequent study from the same group of a patient with HH
delta waves, and 11% had rhythmic theta or delta waves on ver- and gelastic epilepsy using simultaneous EEG and fMRI record-
tex or anterior quadrants. ings of several seizures indicated that the epileptic activity
Both Bickford and Martinius et al. noted the suppression of appeared to originate in the area around the tumor and propa-
abnormal rhythmic delta-range activity on arousal in patients gate through the left fornix to the temporal lobe, and later
with deep tumors (19,20). Because these patterns may be pro- through the cingulate fasciculus to the left frontal lobe (37).
duced by tumors distant from where the abnormal EEG pat-
terns are recorded, they are sometimes referred to as ELECTROPHYSIOLOGY IN SURGICAL
“projected” rhythms. EEG may not lateralize, or may falsely lat- NEURO-ONCOLOGY
eralize, posterior fossa tumors (19). EEG abnormalities (poste-
rior slow activity) were more common in patients with Electrophysiology retains a vital role in the surgical manage-
posterior fossa tumors and evidence of increased intracranial ment of tumors and other space-occupying lesions. One of the
pressure (i.e., evidence of third ventricle dilation) (19). most challenging surgical decisions is balancing the desire to
Arrhythmic delta activity is more common in patients with resect as much tumor as possible with the desire to leave intact
more rapid progression of symptoms, possibly due to rapid tissues important to motor, sensory, or language-related func-
expansion of tumor size (19). Nearly half the EEGs of patients tions. Certain masses, particularly tumors with a tendency to
with tumors of the cerebellopontine angle (CPA) were normal infiltrate otherwise normal tissue (e.g., glioblastoma multi-
in one series (23). forme), pose a real dilemma for the surgeon and oncology
team, requiring decisions that may leave the patient either with
Sellar Tumors more residual tumor or without functions critical to optimal
EEG abnormalities in tumors of the sellar region include tem- daily living. Neurophysiology can offer guidance in defining
poral lobe abnormalities, unilateral delta-range activity, and two important regions: the peritumoral epileptogenic zone and
bitemporal dysrhythmia. It is important to remember this in functional cortex (i.e., areas that typically are critical to motor,
the differential diagnosis of temporal lobe abnormalities. In sensory, or language function and whose resection would lead
tumors that compressed the third ventricle, generalized slowing to significant deficits). Simple lesionectomies also are appropri-
was noted, and degree of compression was the only factor that ate in the setting of certain well-defined lesions (e.g., caver-
correlated with abnormalities in the EEG (29). In that series, nomas) or those near noneloquent cortex. Data from some
EEG abnormalities did not predict tumor type (29). series (38,39) suggest lesionectomies may be adequate in some
cases, although selection of candidates for this approach has not
Hypothalamic Hamartoma been thoroughly studied; success in these cases suggests that
HH is a developmental malformation that frequently presents disruption of an epileptogenic network may suffice to suppress
with seizures, including infantile spasms. Other associated con- seizure activity (40).
ditions may include precocious puberty and behavioral prob- One application of electrophysiology in the operating room
lems. An excellent recent review of the topic is available (30). In is in the resection of tumors that are associated with epilepsy,
children, gelastic seizures are most typical at onset, although such as certain astrocytomas, dysembroplastic neuroepithelial
this may not be the case in adults, who tend to present with par- tumors (DNET), gangliogliomas, and gangliocytomas. These
tial-onset seizures (31). The other main cause of gelastic tumors are associated with epileptogenic tissue at their margins,
epilepsy is temporal or frontal lobe epilepsy (32). The EEG in and although the tumors themselves are not typically aggressive,
HH initially may be normal, similar to other deep-seated there can be epileptogenic tissue adjacent to the tumor (41).
masses, although over time it may evolve through the appear- Intraoperative ECoG using standard carbon-tipped electrodes
ance of focal (partial onset), and then generalized seizures, con- or electrodes designed for subdural recordings can help define
sistent with the clinical semiology (30). There may be associated the extent of the epileptogenic zone, even beyond the tumor
autonomic features (33). Focal slowing or epileptiform activity margin (41). In this series, seizure control was achieved in one
over frontal and/or temporal head regions may be the initial patient after epileptogenic cortex was resected, even though not
appearance on EEG, although this evolves into bilateral spike all the tumor was removed, although this may have been an
wave over time if untreated (30). Eventually, a pattern consist- exceptional case. Additionally, the authors point out this strategy
ing of generalized slow spike wave, paroxysmal fast, and elec- may be more useful in children than adults. Given tumor recur-
trodecremental patterns can occur, reminiscent of the EEG in rence rates in some series (42), some suggest that monitoring be
Lennox Gastaut syndrome (34). There is a possibility that the done routinely in cases involving tissue near motor, sensory, or
hamartoma in some way generates abnormal activity that prop- language areas. However, extraoperative recordings from
agates through the cortex, leading to the various seizure types patients with subdural electrodes show that epileptiform activ-
noted in HH (30,35). In four patients with HH presenting ity can be intermittent, and so might be missed during the lim-
epileptic manifestations and displaying interictal spikes over ited recording time available in the operating room.
the frontal and temporal areas, EEG source analysis, based on Cortical mapping of motor, sensory, or language cortex
scalp recordings (32 electrodes), was able to estimate that the can be done intraoperatively (43), or outside the operating
Chapter 17 ■ Brain Tumors and Other Space-Occupying Lesions 325

room (44), depending on the goals of the surgery. Each tech- potentials did not change (48). Also, phase reversal of
nique has its advantages and applications. Intraoperative somatosensory evoked potentials (SEPs) across the rolandic
mapping may reduce the risks of inserting and maintaining sulcus helps the surgeon locate, and therefore avoid, the pri-
subdural electrodes (such as infection and bleeding) and is mary sensorimotor cortex (49,50). Motor cortex stimulation,
less expensive, but extraoperative mapping allows for more and recording of evoked electromyographic changes peripher-
time to better define the regions important for specific func- ally, is another way to locate motor cortex (50). Responses to
tions, while minimizing operative time when tissue is motor cortex stimulation are sensitive to anesthesia however,
removed and minimizing the chances of a stimulation- and this restricts its use in some circumstances.
induced intraoperative seizure (40). If seizures are not a sig-
nificant concern, then intraoperative mapping is more Posterior Fossa Surgery
straightforward, since only EEG slowing or suppression need Electrophysiologic are no longer a primary method for locating
be sought. If intractable epilepsy is the major concern, we tend posterior fossa tumors, but they can help to determine the rela-
to consider the case operation as an epilepsy (rather than tionship of the mass to auditory pathways (51). BAEPs fre-
tumor) evaluation and consider extraoperative EEG recording quently are lost with CPA tumors, due to effects on either the
and mapping, but others have good results with intraoperative peripheral or brainstem portions of the auditory pathway
mapping (44–46). Mapping using evoked potentials is dis- (52,53). Possible changes include decreased amplitude and
cussed later. The technical aspects of extraoperative mapping increased latency (51–54). As would be expected, BAEPs are not
are discussed elsewhere in this book (Chapter 55). as sensitive as MRI in patients with small CPA tumors (55), or
Invasive recordings from patients with tumors have rein- tumors that do not impinge on the auditory pathway (56,57).
forced some findings from scalp recordings but also have In 40 patients with acoustic neuromas, Tanaka et al. (58)
identified a number of additional interesting phenomena. reported that 6 patients with normal ABRs using standard test-
Patients with bilateral interictal findings can have unilateral ing methods had abnormalities when the clicks were given at
ictal onsets and good postoperative outcomes (28). In one high stimulus rates.
series, ECoG during resection of gangliogliomas showed high- BAEPs can be used alone or together with other modalities
voltage slowing in the area of the tumor (47). Another series to assess the effects of tumors on the brainstem, or to monitor
using stereo-EEG in patients with low-grade tumors showed during surgery (51). For example, some have used recordings of
activity less than 10 V in white matter that was infiltrated the blink or masseter reflex (53,59). During posterior fossa sur-
with tumor or only affected by edema in some cases, while gery, other cranial nerves, including III, IV, V, VI, VII, IX, X, XI,
high-voltage slowing was noted in some patients with simi- and XII, have been monitored, along with muscles innervated
larly affected gray matter, or in peritumoral gliosis (some by these nerves, and along with SEPs (51,60). BAEPs along with
specimens also had normal activity) (2). Solid tumors always EMG monitoring can help insure safe microvascular decom-
showed electrical depression. Intraoperative mapping has pression procedures in the posterior fossa. When SEPs are used
demonstrated acute changes in local motor maps right after to monitor during posterior fossa surgery, it should be kept in
tumor resection (46). mind that the usually recorded SEP responses to peripheral
nerves are generated above and below, but not within, the
brainstem (61).
EVOKED POTENTIALS IN THE TREATMENT
OF BRAIN TUMORS Magnetoencephalography
Nakasato et al. (62) evaluated cortical auditory function in 14
Electrophysiologic techniques such as evoked potentials were patients with temporal lobe tumors, using an MRI-linked
once used to help diagnose and locate mass lesions. This has whole-head MEG system. The authors suggested that the MEG
largely been replaced by imaging. However, electrophysiology, system could be used to evaluate cortical auditory function
used as a test of function, retains an important role in the man- noninvasively before and after surgical treatment of temporal
agement of mass lesions, particularly in the operating room. lobe tumors. An example of the usefulness of MEG in the pre-
The techniques are not specific to the management of tumors, operative evaluation of tumors using somatosensory evoked
but instead have been developed to help insure that surgical fields (SEFs) is presented in Chapter 42 and illustrated in
procedures are performed as safely as possible. Figure 42.27.

Cortical and Subcortical Event-related Potentials


Preoperative evoked potentials can be used to document Moore et al. (63) evaluated 33 survivors of childhood cancer
whether sensory pathways are intact prior to, or at the start of, with P300, motor reaction time, and neuropsychiatric testing.
surgery. During surgery, changes in response can signal to the There were abnormalities in P300 and in neuropsychiatric
surgeon that the surgery might be impinging on the monitored scores of patients who had received CRT. The authors suggested
pathway. The surgeon can then modify the surgical approach that cognitive changes in this group might be due to white mat-
and extent of surgical resection. However, a change in responses ter damage. As with other evoked potentials, changes in P300
may be irreversible once seen. Moreover, on occasion there can are nonspecific with respect to etiology, but may correlate with
be postoperative deficits even though intraoperative evoked overall cognitive status.
326 Part III ■ Clinical EEG: General

EEG COHERENCE rhage may be due to nonconvulsive status epilepticus, or to a


postictal state; in either case, an EEG may be obtained.
EEG coherence, a measure of the association between signals in Intracranial hemorrhages may include epidural hematomas (a
a specified frequency band from different brain regions, has true neurosurgical emergency), subdural hematomas (both
been used in both surface and invasive recordings. One study acute and chronic), intraparenchymal hemorrhages, and intra-
that used surface recordings found decreased coherence ventricular hemorrhages. As with tumors, EEG findings
between the cortical region containing a brain lesion, and other depend heavily on the location, extent, and chronicity of the
portions of the same hemisphere (64). Another study examined hemorrhage.
the use of coherence to assess the central sulcus and sensorimo-
tor area during tumor resections in 10 patients (65). The central Acute Subdural Hematoma
sulcus was a region of low coherence, whereas high coherence Slowing (including slow posterior basic rhythm and paroxys-
was noted in the region of the brain tumor. In contrast, coher- mal slow activity) and epileptiform discharges have been
ence measures were not helpful in the temporal lobe. Further noted in patients with acute subdural hematomas (Fig. 17.5)
work would be needed to resolve the discrepancies between (72–75). Case reports have mentioned PLEDs as one EEG
these results. finding in acute subdural hematoma (76), although they have
been seen after subdural hematomas have been evacuated, as
EEG IN BRAIN ABSCESS well (77). EEG abnormalities may not be related to the size of
the hematoma, among other factors (73). One study suggested
Despite the hope expressed in the last edition of this text that that acute and subacute hematomas are associated with
brain abscess would become a rare diagnosis, the surge in changes in EEG amplitude, while chronic hematomas were
patients with acquired immune deficiency syndrome, increased associated with changes in frequency (i.e., increased slow
numbers of patients on immunomodulators or immunosup- activity) (78).
pression after organ transplants or the diagnosis of autoim-
mune disease, and increased recognition of congenital Chronic Subdural Hematoma
immunodeficiencies have led to a resurgence of this condition In a study of patients with unilateral chronic subdural
(Chapter 15, p. 314 of the 5th edition). More recent case reports hematomas and herniation who underwent xenon/CT (to
and series of patients with brain abscesses mention both epilep- assess cerebral blood flow) and electroencephalograms,
tiform activity (66,67) and PLEDs (68). One series showed that increased flow was noted in areas where alpha- and beta-range
14 of 45 patients had focal EEG abnormalities (not specified activity predominated, while blood flow was decreased in
further), while an additional 21 had either regional or hemi- areas where slower frequencies predominated (79).
spheric disturbances; of 3 normal EEGs, 2 patients had cerebel- Furthermore, electrical activity correlated with thalamic
lar abscesses (69). (rather than cortical) blood flow, leading the authors to con-
Michel et al. (70) studied 13 patients with brain abscess and clude that thalamus is the source of the abnormal EEG activ-
found slowing of background activity in the hemisphere with ity in this population. Patients with chronic subdural
the abscess in 6 cases and bilateral generalized slowing in hematomas also can have normal EEGs; this was the case for
another 4 (3 were normal), continuous and nonreactive poly- eight of nine patients recorded 8 months to nearly 4 years
morphic delta-range activity in 12 patients with frontal inter- after the initial insult (80).
mittent delta-range activity in 5 patients, and pseudoperiodic
sharp waves in 2 patients. The EEG localized the abscess in
61.5% of their cases. Focal arrhythmic delta-range activity was
the most consistent finding in another series of 13 patients (71).
Importantly, although EEG was localizing in most cases, it was
falsely localizing in one patient with a cerebellar abscess, per-
haps of the connections between each cerebellar hemisphere
and the contralateral cerebral hemisphere. Epileptiform activity
occurred in four cases. Asymmetry in distribution of beta-
range activity helped lateralize the hemisphere with the abscess
in seven. In summary, EEG may be useful in localizing cortical
abscesses in many cases, but may be misleading or even normal
in others.

EEG IN SUBDURAL HEMATOMA


Just as in tumors, the typical first diagnostic study obtained in
most patients with intracranial hemorrhages is either a CT Figure 17.5 EEG from a 76-year-old male showing delta-range slowing
scan or, less commonly, an MRI. Nonetheless, persistent altered over the right hemisphere. A right frontotemporoparietal subdural
mental status after evacuation or management of a hemor- hematoma and left temporofrontal hygroma were evacuated at surgery.
Chapter 17 ■ Brain Tumors and Other Space-Occupying Lesions 327

EEG AND EVOKED POTENTIALS IN NORMAL combination therapy using cisplatin, vinblastine, and
PRESSURE HYDROCEPHALUS bleomycin in a patient with an ovarian germ cell tumor with
posterior leukoencephalopathy associated with slowing in an
The EEG in patients with normal pressure hydrocephalus (NPH, area of involvement noted on MRI (94);
i.e., idiopathic) can be normal or can show unilateral or bilateral combination therapy for a gastrointestinal stromal tumor using
intermittent slowing (81). In one series of 14 patients with NPH, 5-fluorouracil, adriamycin, and mitomycin-C (95);
half had EEGs with bursts of bilateral monorhythmic slow activ- high-dose 5-fluorouracil and leucovorin (associated with
ity. This activity became more noticeable in four of five patients hyperammonemia, and lactic acidosis (96);
who had serial EEGs, while a similar number had improvements busulfan, given with clonazpam for seizure prophylaxis in chil-
in the EEG after shunting operations (82). This suggests that dren with various types of tumors, with the EEG slowing
EEG changes may be useful for tracking progression of illness in focal slowing (97);
some patients with NPH, but another paper suggested that NPH IL-2 administration with bilateral slowing in the frontal
patients do not have specific patterns on their EEG (83). regions (98);
interleukin-2 for various malignancies or HIV infection with
bursts of frontal intermittent rhythmic delta activity (99);
EEG CHANGES DUE TO ONCOLOGICAL interferon-alpha in adults (100–102) and children (103);
AGENTS ifosfamide and mesna (104,105).
CNS toxicity has been associated with many chemotherapy Epileptiform activity or seizures have been reported with:
agents. Electroencephalographic changes include slowing,
intravenous high-dose methotrexate infusion (106);
epileptiform activity, and voltage suppression. One difficulty in
chlorambucil (diffuse spike-wave activity) in two of nine chil-
interpreting these findings is that patients often are receiving
dren with nephrotic syndrome (107);
multiple agents, plus radiotherapy. Also, more than one finding
intravenous vincristine infusions (a patient with unilateral
can occur in a given patient. Finally, these are patients that often
seizures) (108);
are systemically ill both from their primary illness and from
infusion of paclitaxel (109);
treatments. Therefore, a second difficulty lies in determining
busulfan and cyclophosphamide conditioning regimen prior to
whether a given medication represents a primary or secondary
bone marrow transplant in patients with leukemia
cause of the EEG changes.
(110,111);
Diffuse slowing has been reported in children with systemic
interleukin-2 for various malignancies or HIV infection (with
tumors (including acute lymphoblastic leukemia [ALL], Ewing
periodic epileptiform discharges) (99);
sarcoma, non-Hodgkin lymphoma, reticulum cell sarcoma,
interferon-alpha (100,102) and children (103);
malignant chest wall tumor, and ovarian stroma cell carcinoma)
ifosfamide and mesna (104,105), including nonconvulsive sta-
treated with agents including methotrexate, vincristine, leucov-
tus epilepticus (112).
orin, L-asparaginase, adriamycin, actinomycin, 5-fluorouracil,
cyclophosphamide, cytosine arabinoside, and VP-12-213 (84). EEG attenuation has been reported with:
Diffuse or generalized slowing occurred in less than 20% of
high-dose methotrexate, vincristine in combination with other
children receiving combinations of intrathecal methotrexate
cytotoxic agents, and L-asparaginase (84);
alone, cranial radiation, and triple intrathecal therapy (consist-
inadvertent intrathecal vincristine in a patient with lym-
ing of methotrexate, cytarabine, and prednisone) for B-lineage
phoblastic leukemia; this patient had EEG slowing that
ALL without CNS leukemia (85). Another study concluded that
evolved into “alpha coma” and voltage attenuation after the
vincristine and L-asparaginase were most likely to be associated
event (113);
with EEG background slowing in a group of 13 children receiv-
interferon-alpha and -gamma (114).
ing combination therapy for ALL (86). The same agents were
associated with transient EEG slowing in 79 children treated for
ALL (87). The significance of these EEG changes is unclear, as REFERENCES
EEG abnormalities in long-term survivors of the protocol in the
1. Gloor P, Ball G, Schaul N. Brain lesions that produce delta waves
latter study did not correlate with the presence or degree of
in the EEG. Neurology. 1977;27:326–333.
abnormalities in their exams or neuroimaging (88). 2. Munari C, Musolino A, Daumas-Duport C, et al. Correlation
Other studies report slowing (generalized in distribution between stereo-EEG, CT-scan and stereotactic biopsy data in
unless otherwise described in the following list) with: epileptic patients with low-grade gliomas. Appl Neurophysiol.
1985;48:448–453.
methotrexate (89); intrathecal methotrexate and cytosine ara-
3. Aronica E, Boer K, Becker A, et al. Gene expression profile analysis of
binoside with intravenous cyclophosphamide, vincristine,
epilepsy-associated gangliogliomas. Neuroscience. 2008;151:272–292.
methotrexate, ifosfamide, and prednisolone (90); procar- 4. Klass DW, Daly DD. Electroencephalography in patients with
bazine, CCNU, and vincristine for recurrent gliomas (91); brain tumor. Med Clin North Am. 1960;44:1041–1051.
paclitaxel (92) although one series showed no adverse effects 5. Fish DR. How do tumors cause epilepsy? In: Kotagal P, Lüders HO,
on the EEG in 28 patients with ovarian carcinoma treated eds. The Epilepsies: Etiologies and Prevention. San Diego: Academic
with a paclitaxel–carboplatin regimen (93); Press; 1999:301–314.
328 Part III ■ Clinical EEG: General

6. Aronica E, Gorter JA, Jansen GH, et al. Expression of connexin 43 26. Reyes P, Jameson HD. Clinical correlation of periodic lateralized
and connexin 32 gap-junction proteins in epilepsy-associated brain epileptiform discharges in a general hospital. Electroencephalogr
tumors and in the perilesional epileptic cortex. Acta Neuropathol. Clin Neurophysiol. 1979;46:3P.
2001;101:449–459. 27. Fischer-Williams M, Dike GL. Brain tumors and other space-
7. Alonso-Nanclares L, De FJ. Vesicular glutamate transporter 1 occupying lesions. In: Niedermeyer E, Lopes da Silva F, eds.
immunostaining in the normal and epileptic human cerebral cor- Electroencephalography: Basic Principles, Clinical Applications, and
tex. Neuroscience. 2005;134:59–68. Related Fields. Philadelphia: Lippincott Williams & Wilkins;
8. Aronica E, Yankaya B, Jansen GH, et al. Ionotropic and metabotropic 2005:305–321.
glutamate receptor protein expression in glioneuronal tumours from 28. Zaatreh MM, Firlik KS, Spencer DD, et al. Temporal lobe tumoral
patients with intractable epilepsy. Neuropathol Appl Neurobiol. 2001; epilepsy: characteristics and predictors of surgical outcome.
27:223–237. Neurology. 2003;61:636–641.
9. Richardson MP, Hammers A, Brooks DJ, et al. Benzodiazepine- 29. Nau HE, Bock WJ, Clar HE. Electroencephalographic investiga-
GABA(A) receptor binding is very low in dysembryoplastic tions in sellar tumours, with special regard to different methods of
neuroepithelial tumor: a PET study. Epilepsia. 2001;42: operative treatment. Acta Neurochir (Wien). 1978;44:207–214.
1327–1334. 30. Harvey AS, Freeman JL. Epilepsy in hypothalamic hamartoma:
10. Morrell F, de Toldedo-Morrell L. Secondary epileptogenesis and clinical and EEG features. Semin Pediatr Neurol. 2007;14:60–64.
brain tumors. In: Kotagal P, Lüders HO, eds. The Epilepsies: 31. Mullatti N, Selway R, Nashef L, et al. The clinical spectrum of
Etiologies and Prevention. San Diego: Academic Press, 1999: epilepsy in children and adults with hypothalamic hamartoma.
357–363. Epilepsia. 2003;44:1310–1319.
11. Laws ER Jr, Taylor WF, Clifton MB, et al. Neurosurgical manage- 32. Arroyo S, Lesser RP, Gordon B, et al. Mirth, laughter and gelastic
ment of low-grade astrocytoma of the cerebral hemispheres. seizures. Brain. 1993;116(Pt 4):757–780.
J Neurosurg. 1984;61:665–673. 33. Cerullo A, Tinuper P, Provini F, et al. Autonomic and hormonal
12. Lynam LM, Lyons MK, Drazkowski JF, et al. Frequency of seizures ictal changes in gelastic seizures from hypothalamic hamartomas.
in patients with newly diagnosed brain tumors: a retrospective Electroencephalogr Clin Neurophysiol. 1998;107:317–322.
review. Clin Neurol Neurosurg. 2007;109:634–638. 34. Berkovic SF, Andermann F, Melanson D, et al. Hypothalamic
13. Hirsch JF, Sainte RC, Pierre-Kahn A, et al. Benign astrocytic and hamartomas and ictal laughter: evolution of a characteristic
oligodendrocytic tumors of the cerebral hemispheres in children. epileptic syndrome and diagnostic value of magnetic resonance
J Neurosurg. 1989;70:568–572. imaging. Ann Neurol. 1988;23:429–439.
14. Low NL, Correll JW, Hammill JF. Tumors of the cerebral hemi- 35. Kuzniecky R, Guthrie B, Mountz J, et al. Intrinsic epileptogenesis
spheres in children. Arch Neurol. 1965;13:547–554. of hypothalamic hamartomas in gelastic epilepsy. Ann Neurol.
15. Backus RE, Millichap JG. The seizure as a manifestation of 1997;42:60–67.
intracranial tumor in childhood. Pediatrics. 1962;29:978–984. 36. Leal AJ, Passao V, Calado E, et al. Interictal spike EEG source analy-
16. Forsgren L, Bucht G, Eriksson S, et al. Incidence and clinical char- sis in hypothalamic hamartoma epilepsy. Clin Neurophysiol.
acterization of unprovoked seizures in adults: a prospective popu- 2002;113:1961–1969.
lation-based study. Epilepsia. 1996;37:224–229. 37. Leal AJ, Monteiro JP, Secca MF, et al. Functional brain mapping of
17. Khan RB, Hunt DL, Boop FA, et al. Seizures in children with pri- ictal activity in gelastic epilepsy associated with hypothalamic
mary brain tumors: incidence and long-term outcome. Epilepsy hamartoma: a case report. Epilepsia. 2009;50:1624–1631.
Res. 2005;64:85–91. 38. Cascino GD, Kelly PJ, Sharbrough FW, et al. Long-term follow-up
18. Kershman J, Conde A, Gibson WC. Electroencephalography in dif- of stereotactic lesionectomy in partial epilepsy: predictive factors
ferential diagnosis of supratentorial tumors. Arch Neurol and electroencephalographic results. Epilepsia. 1992;33:639–644.
Psychiatry. 1949;62:255–268. 39. Packer RJ, Sutton LN, Patel KM, et al. Seizure control following
19. Martinius J, Matthes A, Lombroso CT. Electroencephalographic tumor surgery for childhood cortical low-grade gliomas. J
features in posterior fossa tumors in children. Electroencephalogr Neurosurg. 1994;80:998–1003.
Clin Neurophysiol. 1968;25:128–139. 40. Van Ness PC. Pros and cons of lesionectomy as treatment for par-
20. Bickford RG. Electroencephalographic diagnosis of brain tumors. tial epilepsy. In: Kotagal P, Lüders HO, eds. The Epilepsies: Etiologies
Am J Surg. 1957;93:946–951. and Prevention. San Diego: Academic Press, 1999:391–397.
21. Kirstein L. The occurrence of sharp waves, spikes and fast activity 41. Berger MS, Ghatan S, Geyer JR, et al. Seizure outcome in children
in supratentorial tumours. Electroencephalogr Clin Neurophysiol. with hemispheric tumors and associated intractable epilepsy: the
1953;5:33–40. role of tumor removal combined with seizure foci resection.
22. Newmark ME, Theodore WH, Sato S, et al. EEG, transmission Pediatr Neurosurg. 1991;17:185–191.
computed tomography, and positron emission tomography with 42. Ebeling U, Fischer M, Kothbauer K. Surgery of astrocytomas in the
fluorodeoxyglucose 18F. Their use in adults with gliomas. Arch motor and premotor cortex under local anesthesia: report of 11
Neurol. 1983;40:607–610. cases. Minim Invasive Neurosurg. 1995;38:51–59.
23. Daly D, Whelan JL, Bickford RG, et al. The electroencephalogram 43. Berger MS, Ojemann GA. Intraoperative brain mapping tech-
in cases of tumors of the posterior fossa and third ventricle. niques in neuro-oncology. Stereotact Funct Neurosurg. 1992;58(1–4):
Electroencephalogr Clin Neurophysiol. 1953;5:203–216. 153–161.
24. Morris HH III, Luders H, Hahn JF, et al. Neurophysiological tech- 44. Goldring S. A method for surgical management of focal epilepsy,
niques as an aid to surgical treatment of primary brain tumors. especially as it relates to children. J Neurosurg. 1978;49:344–356.
Ann Neurol. 1986;19:559–567. 45. Duffau H, Peggy Gatignol ST, Mandonnet E, et al. Intraoperative
25. Gastaut JL, Michel B. The impact of cranial computerized tomog- subcortical stimulation mapping of language pathways in a con-
raphy on electroencephalography. Electroencephalogr Clin secutive series of 115 patients with grade II glioma in the left dom-
Neurophysiol Suppl. 1978;34:123–132. inant hemisphere. J Neurosurg. 2008;109:461–471.
Chapter 17 ■ Brain Tumors and Other Space-Occupying Lesions 329

46. Duffau H. Acute functional reorganisation of the human motor 66. Aebi C, Kaufmann F, Schaad UB. Brain abscess in childhood—
cortex during resection of central lesions: a study using intraoper- long-term experiences. Eur J Pediatr. 1991;150:282–286.
ative brain mapping. J Neurol Neurosurg Psychiatry. 2001;70: 67. Ma JS, Chen PY, Lau YJ, et al. Brain abscess caused by Salmonella
506–513. enterica subspecies houtenae in a patient with chronic granuloma-
47. Pilcher WH, Silbergeld DL, Berger MS, et al. Intraoperative tous disease. J Microbiol Immunol Infect. 2003;36:282–284.
electrocorticography during tumor resection: impact on seizure 68. Gurer G, Yemisci M, Saygi S, et al. Structural lesions in periodic
outcome in patients with gangliogliomas. J Neurosurg. 1993;78: lateralized epileptiform discharges (PLEDs). Clin EEG Neurosci.
891–902. 2004;35:88–93.
48. Lesser RP, Raudzens P, Luders H, et al. Postoperative neurological 69. Beller AJ, Sahar A, Praiss I. Brain abscess. Review of 89 cases
deficits may occur despite unchanged intraoperative somatosen- over a period of 30 years. J Neurol Neurosurg Psychiatry. 1973;36:
sory evoked potentials. Ann Neurol. 1986;19:22–25. 757–768.
49. Lueders H, Lesser RP, Hahn J, et al. Cortical somatosensory evoked 70. Michel B, Gastaut JL, Bianchi L. Electroencephalographic cranial
potentials in response to hand stimulation. J Neurosurg. 1983;58: computerized tomographic correlations in brain abscess.
885–894. Electroencephalogr Clin Neurophysiol. 1979;46:256–273.
50. Cedzich C, Taniguchi M, Schafer S, et al. Somatosensory evoked 71. Vignadndra V, Ghee LT, Chawla J. EEG in brain abscess: its value in
potential phase reversal and direct motor cortex stimulation dur- localization compared to other diagnostic tests. Electroencephalogr
ing surgery in and around the central region. Neurosurgery. 1996; Clin Neurophysiol. 1975;38:611–622.
38:962–970. 72. Gutierrez-Luque AG, Maccarty CS, Klass DW. Head injury with
51. Cheek JC. Posterior fossa intraoperative monitoring. J Clin suspected subdural hematoma. Effect on EEG. Arch Neurol. 1966;
Neurophysiol. 1993;10:412–424. 15:437–443.
52. Nurlu G, Bavbek M, Colak A, et al. Brainstem auditory evoked 73. Jaffe R, Librot IE, Bender MB. Serial EEG studies in unoperated
potentials and blink reflexes in patients with pontocerebellar angle subdural hematoma. Arch Neurol. 1968;19:325–330.
tumors. Neurosurg Rev. 1994;17:253–260. 74. Poole EW. Some aspects of electroencephalographic disturbances
53. Csecsei GI, Klug N, Szekely G Jr, et al. Multimodality electroneu- following head injury. J Clin Pathol Suppl (R Coll Pathol)
rophysiological findings in intra-axial and extra-axial lesions of 1970;4:187–201.
the brain stem. Acta Neurochir (Wien). 1995;137:48–53. 75. Jones SC, Bamford JM, Heath J, et al. Multiple forms of epileptic
54. Stanton SG, Cashman MZ. Auditory brainstem response. A com- attack secondary to a small chronic subdural haematoma. BMJ.
parison of different interpretation strategies for detection of cere- 1989;299:439–441.
bellopontine angle tumors. Scand Audiol. 1996;25:109–120. 76. Chu NS. Acute subdural hematoma and the periodic lateralized
55. Naessens B, Gordts F, Clement PA, et al. Re-evaluation of the ABR in epileptiform discharges. Clin Electroencephalogr. 1979;10:145–150.
the diagnosis of CPA tumors in the MRI-era. Acta Otorhinolaryngol 77. Westmoreland BF. Periodic lateralized epileptiform discharges
Belg. 1996;50:99–102. after evacuation of subdural hematomas. J Clin Neurophysiol.
56. Khatib ZA, Heideman RL, Kovnar EH, et al. Predominance of pilo- 2001;18:20–24.
cytic histology in dorsally exophytic brain stem tumors. Pediatr 78. Ibrahim MM, Elian M. A reappraisal of the value of the EEG in
Neurosurg. 1994;20:2–10. subdural haematoma. A report on 86 patients, with a follow-up
57. Baran JA, Catherwood KP, Musiek FE. “Negative” ABR findings in study. J Neurol. 1974;207:117–128.
an individual with a large brainstem tumor: hit or miss? J Am Acad 79. Tanaka A, Kimura M, Yoshinaga S, et al. Quantitative electroen-
Audiol. 1995;6:211–216. cephalographic correlates of cerebral blood flow in patients with
58. Tanaka H, Komatsuzaki A, Hentona H. Usefulness of auditory chronic subdural hematomas. Surg Neurol. 1998;50:235–240.
brainstem responses at high stimulus rates in the diagnosis of 80. Lusins J, Jaffe R, Bender MB. Unoperated subdural hematomas.
acoustic neuroma. ORL J Otorhinolaryngol Relat Spec. 1996;58: Long-term follow-up study by brain scan and electroencephalog-
224–228. raphy. J Neurosurg. 1976;44:601–607.
59. Broggi G, Scaioli V, Brock S, et al. Neurophysiological monitoring 81. Brown DG, Goldensohn ES. The electroencephalogram in normal
of cranial nerves during posterior fossa surgery. Acta Neurochir pressure hydrocephalus. Arch Neurol. 1973;29:70–71.
Suppl. 1995;64:35–39. 82. Hashi K, Nishimura S, Kondo A, et al. The EEG in normal pressure
60. Eisner W, Schmid UD, Reulen HJ, et al. The mapping and contin- hydrocephalus. Acta Neurochir (Wien). 1976;33:23–35.
uous monitoring of the intrinsic motor nuclei during brain stem 83. Petersen RC, Mokri B, Laws ER Jr. Surgical treatment of idiopathic
surgery. Neurosurgery. 1995;37:255–265. hydrocephalus in elderly patients. Neurology. 1985;35:307–311.
61. Lesser RP, Lueders H, Hahn J, et al. Early somatosensory potentials 84. Schaffler L, Imbach P, Rudeberg A, et al. Conventional and spectral
evoked by median nerve stimulation: intraoperative monitoring. EEG analysis in children treated with cytotoxic agents. Eur J
Neurology. 1981;31:1519–1523. Cancer Clin Oncol. 1982;18:827–832.
62. Nakasato N, Kumabe T, Kanno A, et al. Neuromagnetic evaluation 85. Lo Nigro L, Di CA, Schiliro G. Acute neurotoxicity in children with
of cortical auditory function in patients with temporal lobe tumors. B-lineage acute lymphoblastic leukemia (B-ALL) treated with
J Neurosurg. 1997;86:610–618. intermediate risk protocols. Med Pediatr Oncol. 2000;35:449–455.
63. Moore BD III, Copeland DR, Ried H, et al. Neurophysiological 86. Lichter-Konecki U, Benninger C, Brandeis WE, et al. Changes in
basis of cognitive deficits in long-term survivors of childhood can- the EEG background activity of children with acute lymphoblastic
cer. Arch Neurol. 1992;49:809–817. leukemia during cytotoxic therapy. Pediatr Hematol Oncol.
64. Harmony T, Marosi E, Fernandez T, et al. EEG coherences in 1987;4:77–85.
patients with brain lesions. Int J Neurosci. 1994;74:203–226. 87. Korinthenberg R, Igel B. Prospective neurophysiological study in
65. Towle VL, Syed I, Berger C, et al. Identification of the sensory/ children treated for acute lymphoblastic leukaemia: serial EEG
motor area and pathologic regions using ECoG coherence. during treatment and long-term follow up with evoked potentials.
Electroencephalogr Clin Neurophysiol. 1998;106:30–39. Eur J Pediatr. 1990;150:127–131.
330 Part III ■ Clinical EEG: General

88. Ueberall MA, Skirl G, Strassburg HM, et al. Neurophysiological 101. Rohatiner AZ, Prior PF, Burton AC, et al. Central nervous system
findings in long-term survivors of acute lymphoblastic leukaemia toxicity of interferon. Br J Cancer. 1983;47:419–422.
in childhood treated with the BFM protocol 81 SR-A/B. Eur J 102. Suter CC, Westmoreland BF, Sharbrough FW, et al. Electro-
Pediatr. 1997;156:727–733. encephalographic abnormalities in interferon encephalopathy: a
89. Ochs J, Parvey LS, Mulhern R. Prospective study of central nerv- preliminary report. Mayo Clin Proc. 1984;59:847–850.
ous system changes in children with acute lymphoblastic 103. Miller VS, Zwiener RJ, Fielman BA. Interferon-associated refrac-
leukemia receiving two different methods of central nervous sys- tory status epilepticus. Pediatrics. 1994;93:511–512.
tem prophylaxis. Neurotoxicology. 1986;7:217–226. 104. Meanwell CA, Blake AE, Kelly KA, et al. Prediction of ifos-
90. Geissler RG, Bergmann L, Hacker H, et al. Severe central nervous famide/mesna associated encephalopathy. Eur J Cancer Clin
adverse effect of intrathecal chemotherapy in a 16-yr-old patient Oncol. 1986;22:815–819.
with Burkitt’s type lymphoma. Eur J Haematol. 1997;58:286–288. 105. Pratt CB, Green AA, Horowitz ME, et al. Central nervous system
91. Postma TJ, van Groeningen CJ, Witjes RJ, et al. Neurotoxicity of toxicity following the treatment of pediatric patients with ifos-
combination chemotherapy with procarbazine, CCNU and vin- famide/mesna. J Clin Oncol. 1986;4:1253–1261.
cristine (PCV) for recurrent glioma. J Neurooncol. 1998;38:69–75. 106. Kubo M, Azuma E, Arai S, et al. Transient encephalopathy
92. Perry JR, Warner E. Transient encephalopathy after paclitaxel following a single exposure of high-dose methotrexate in a child
(taxol) infusion. Neurology. 1996;46:1596–1599. with acute lymphoblastic leukemia. Pediatr Hematol Oncol.
93. Mayerhofer K, Bodner K, Saletu B, et al. Paclitaxel–carboplatin 1992;9:157–165.
chemotherapy does not cause encephalopathy in patients with 107. Ichida F, Konishi T, Asada R, et al. Chlorambucil central nerv-
ovarian cancer: a prospective EEG mapping study in 28 patients. ous toxicity: a significant side effect of chlorambucil therapy in
Anticancer Res. 2001;21:803–808. childhood nephrotic syndrome. Eur J Pediatr. 1985;144:
94. Sueblinvong T, Noophun P, Pataradool K, et al. Posterior 283–286.
leukoencephalopathy following cisplatin, bleomycin and vinblas- 108. Dallera F, Gamoletti R, Costa P. Unilateral seizures following vin-
tine therapy for germ cell tumor of the ovary. J Obstet Gynaecol cristine intravenous injection. Tumori. 1984;70:243–244.
Res. 2002;28:99–103. 109. McGuire WP, Rowinsky EK, Rosenshein NB, et al. Taxol: a
95. Ki SS, Jeong JM, Kim SH, et al. A case of neurotoxicity following unique antineoplastic agent with significant activity in advanced
5-fluorouracil-based chemotherapy. Korean J Intern Med. 2002; ovarian epithelial neoplasms. Ann Intern Med. 1989;111:
17:73–77. 273–279.
96. Yeh KH, Cheng AL. High-dose 5-fluorouracil infusional therapy 110. Meloni G, Raucci U, Pinto RM, et al. Pretransplant conditioning
is associated with hyperammonaemia, lactic acidosis and with busulfan and cyclophosphamide in acute leukemia patients:
encephalopathy. Br J Cancer. 1997;75:464–465. neurological and electroencephalographic prospective study. Ann
97. Vassal G, Deroussent A, Hartmann O, et al. Dose-dependent neu- Oncol. 1992;3:145–148.
rotoxicity of high-dose busulfan in children: a clinical and phar- 111. La MC, Mondini S, Guarino M, et al. Busulfan neurotoxicity and
macological study. Cancer Res. 1990;50:6203–6207. EEG abnormalities: a case report. Neurol Sci. 2004;25:95–97.
98. Pace A, Pietrangeli A, Bove L, et al. Neurotoxicity of antitumoral 112. Wengs WJ, Talwar D, Bernard J. Ifosfamide-induced nonconvul-
IL-2 therapy: evoked cognitive potentials and brain mapping. Ital sive status epilepticus. Arch Neurol. 1993;50:1104–1105.
J Neurol Sci. 1994;15:341–346. 113. Bleck TP, Jacobsen J. Prolonged survival following the inadver-
99. Karp BI, Yang JC, Khorsand M, et al. Multiple cerebral lesions tent intrathecal administration of vincristine: clinical and
complicating therapy with interleukin-2. Neurology. 1996;47: electrophysiologic analyses. Clin Neuropharmacol. 1991;
417–424. 14:457–462.
100. Smedley H, Katrak M, Sikora K, et al. Neurological effects of 114. Born J, Spath-Schwalbe E, Pietrowsky R, et al. Neurophysiological
recombinant human interferon. Br Med J (Clin Res Ed). 1983; effects of recombinant interferon-gamma and -alpha in man.
286:262–264. Clin Physiol Biochem. 1989;7:119–127.
CHAPTER
The EEG in Inflammatory
CNS Conditions
ROBERT L. BEACH, HELEN BARKAN, AND EDGAR DEPERALTA
18
ACKNOWLEDGMENTS with 1- to 70-Hz bandpass filter, and with the notch filter on,
with about 13 seconds per page.
This chapter addresses the electroencephalogram in infection-
related and immune-mediated CNS inflammatory conditions.
This distinction is somewhat artificial in itself, since most CNS PART 1: INFECTIOUS CONDITIONS
inflammation is immune-mediated, either primarily or second-
arily. Previous versions of this chapter were written by the sec- Meningitis
ond author’s most esteemed mentor, Dr. Barbara Westmoreland. Infection and inflammation of the meninges can have bacterial,
The current version is an update, which reflects significantly her fungal, parasitic, autoimmune, and malignancy-related patho-
work and wisdom. Since the last edition, the information on the genesis, or it can be a secondary to a brain abscess or empyema
etiology of several immune-mediated CNS inflammatory with contiguous spread of infection. In the latter case, lateraliz-
processes has progressed significantly, and their associated elec- ing EEG findings can be anticipated. In the rest of the condi-
troencephalographic findings are reviewed here. Some previous tions, the EEG may range from completely normal to severely
topics are covered in other chapters of this volume. abnormal including focal- or generalized-onset seizures and
potentially epileptogenic discharges. However, in the majority
of these conditions, only diffuse slow-wave abnormalities of
INTRODUCTION
variable severity will be seen. There is objective evidence (1), as
The range of EEG abnormalities in inflammatory and infectious cited in the previous version of this chapter (2), that:
disorders was the subject of great interest in the 1950s and Although the electroencephalographic findings are not essential
1960s, but has not received as much attention since then. The for making the specific diagnosis of meningitis, the EEG and
main reason for this lack of engagement is the improvement in particularly serial recordings are helpful in following the course
diagnostic imaging modalities, and the resultant recognition of of the disease, detecting the development of complications or
the limitations of the use of EEG in the diagnosis, treatment, and relapse, and indicating the presence of sequelae of residual brain
follow-up of most of those conditions, with a few notable excep- damage.
tions. The EEG of an inflamed brain can be completely normal,
can reveal minor generalized or lateralized abnormalities, or Purulent Meningitis
may show severe focal or global slowing, and may also contain a Meningococcal, streptococcal, and staphylococcal meningitis
variety of potentially epileptogenic abnormalities or frank elec- are usually community-acquired, with pneumococcal meningi-
trographic seizures of focal, lateralized, or generalized onset. tis being an infrequent complication of pneumonia, otitis
Hence, many believe that most of those EEG abnormalities are media, mastoiditis, severe alcoholism, and CSF leakage after
nonspecific, and of low to moderate diagnostic and prognostic ENT procedures. Staphylococcal meningitis may complicate
value, except for a few “signature” EEG patterns corresponding postoperative course of cranial surgery.
to specific conditions and even to specific pathogens. More Diffuse moderate to very severe and nonspecific slow-wave
EEG-based research directed specifically at validating the prog- abnormalities are typical for the EEG in those conditions (Fig.
nostic and diagnostic value of the EEG in infectious and inflam- 18.1) (3). The slowing is frequently prominent bifrontally, resem-
matory CNS conditions is clearly needed. In the digital EEG age, bling FIRDA (frontal intermittent rhythmic delta activity) possi-
and with sophisticated analysis software, sensitive feature recog- bly associated with increased intracranial pressure (Fig. 18.2).
nition should allow the revitalization of this pursuit. Studies of initial EEG evaluation of patients with purulent
The majority of EEG changes in inflammatory and infec- bacterial meningitis suggest that about one half of the patients
tious CNS disease processes represent the “final common path- have slow-wave abnormalities or electrographic seizures (3).
way” of the electrographic activity of the inflamed cortex in If only community-acquired disease is considered, about
either primary or secondary inflammation. Nevertheless, it is 20% of the patients will have clinical seizures, and a small num-
important to recognize both the specific and the nonspecific ber of those have clinical and electrographic status epilepticus
patterns, and to use them as aid to diagnosis, to gauge the (4). The patients with seizures have mortality of almost three
response to treatment, and for prognostic purposes as well. times higher than those without the seizures. Focal radi-
For consistency, all EEG tracings in the figures illustrating ographic lesions are found in one third of patients with menin-
the text below were captured at the amplitude of 5 V/mm, gitis and in-hospital seizures.

331
332 Part III ■ Clinical EEG: General

Figure 18.1 Awake EEG of a 70-year-old man with community-acquired staphylococcal meningitis showing diffuse mode -
rately severe delta slowing.

Figure 18.2 EEG in an obtunded 63-year-old man with cervical MRSA abscess and secondary meningitis, showing rhyth-
mic bifrontal slowing consistent with FIRDA.
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 333

Other EEG abnormalities in bacterial meningitis also have Meningitis due to cystocercosis, aspergillosis, nocardiosis,
been described, such as triphasic waves and FIRDA, although echinococcosis, mycormycosis, toxoplasmosis, and candidiasis
both are more common for meningitis of other etiologies. is associated with focal slow-wave abnormalities (2), again
An attempt to predict the bacterial or viral nature of the reflecting the patchy nature of meningeal pathology due to
pathogen from the EEG abnormalities suggests that moderately infection by those organisms. No organism-specific patterns are
severe slowing in the delta range is much more likely in bacter- evident, however.
ial meningitis (5).
In neonates with meningitis, with group B streptococcus and Viral Meningitis
E. coli being the most common pathogens, the presence of EEG Viral, otherwise known as “aseptic,” meningitis may not result
abnormalities and/or electrographic and clinical seizures was in any EEG abnormalities, or may result in mild to moderate
directly correlated with prognosis, and associated with poor generalized slowing. In one series of 62 patients, 52% had
outcome (Fig. 18.3). Neonatal convulsive disorders of multiple abnormal EEGs with slow-wave abnormalities (9). Other inves-
etiologies are covered in a separate chapter of this volume. tigators found that only 28% out of 82 patients (10), or in yet
Brain abscess and subdural empyema can be either cause or other studies, one third of patients with CSF-confirmed aseptic
sequelae of bacterial meningitis. The pathogens are often mixed meningitis had EEG abnormalities. Thus, a normal EEG with
mouth flora, including anaerobes, staphylococci, and gram- inflammatory CSF findings strongly suggests viral nature of the
negative commensal organisms. The EEG findings in such cases pathogen—or early stage of CNS involvement.
often reflect a focal lesion—with moderately severe slowing, Practically any infectious agent can cause mild to moderate
potentially epileptogenic abnormalities, and clinical and/or transient meningoencephalitis with variable EEG changes.
electrographic seizures. The focal findings may manifest as Parenchymal involvement is assumed, since biopsy is rarely
polymorphic delta slowing, suppression of activity over the area available, and thus will be discussed in the section “Encephalitis
of suppuration, or, infrequently, lateralized polyphasic poten- with Nonspecific EEG Patterns.” Rarely a purely encephalitic
tially epileptogenic abnormalities such as sharp waves or peri- process, such as HSV encephalitis, can cause focal meningeal
odic lateralized epileptiform discharges (PLEDs). irritation and a clinical and laboratory picture consistent with
aseptic or chemical meningitis.
Basal Meningitis
Basal meningitis due to mycobacteria, Listeria, or spirochetes Mollaret Meningitis
(syphilis and borreliosis) overlaps with the entity called “granu- The agent of this recurrent, lymphocytic meningitis with rela-
lomatous meningitis,” or “chronic meningitis,” and also with tively benign CSF findings and clinical course was thought to
meningoencephalidites described for the same pathogens. The be herpes simplex type II virus, which has been recovered from
separate use of those terms is imprecise, since biopsy is obtained the CSF in some cases. However, some suggest the term should
only in rare cases, and the degree of chronicity varies widely. be restricted to idiopathic cases, as several other infectious and
EEG in basal meningitis ranges from completely normal to autoimmune etiologies as well as CSF leaks and shunts have
severely abnormal, and in addition to varied degrees of general- been implicated in cases of recurrent meningitis (11–14). EEG
ized slowing, hemispheric asymmetries, and sharp paroxysmal in Mollaret meningitis is usually normal; however, diffuse
activity that is either focal or generalized, triphasic waves are not moderately severe slowing can be seen, correlated with the
uncommon (6), as well as FIRDA similar to that in Figure 18.2 clinical symptoms of headache and confusion. Seizures are
(7), which may reflect the underlying increase in intracranial unusual.
pressure. Because of the association of those pathogens with
cerebral vessel vasculitis, it is rare to see normal EEG in those Carcinomatous Meningitis
patients. In one series of 19 patients, 11 had EEG abnormalities. Involvement of the meninges in the neoplastic process is not an
Tuberculous meningitis in another series was associated with uncommon terminal manifestation of systemic cancer. The
similar EEG abnormalities in 24 out of 32 patients. EEG in these patients is practically never normal, with moder-
Focal epileptic discharges and electrographic seizures are ate or very severe slowing that correlates with the clinical sever-
also common in tuberculous meningitis (8), particularly in ity of the encephalopathic state. Focal abnormalities such as
children, and reflect the patchy focal manner of meningeal PLEDs, sharp waves, and spikes are also common. Generalized
inflammation. triphasic periodic waves have also been described.

Fungal and Parasitic Meningitis Meningoencephalitis


These pathogens are relatively uncommon in a healthy host. Meningoencephalitis is a general term, describing the inflam-
Immunosuppressed patients with cryptococcal, cystericercal, mation of both the meninges and the brain parenchyma due to
candidial, or other meningitis—with the inflammation that can toxic, metabolic, infectious, or autoimmune etiologies, with
be isolated to the meninges, but usually also involving the brain many of them overlapping on many occasions. Demyelinating
parenchyma—are typically very ill, and their EEG pattern is disease, connective tissue diseases, and malignancy-related
that of a severe encephalopathy, with polymorphic slowing. encephalitides are discussed later in this chapter. This section
Focal seizures and status epilepticus are common in those will concentrate on infectious and autoimmune etiologies
patients as well as in those with basal meningitis. exclusive of the above-mentioned.
334 Part III ■ Clinical EEG: General

Figure 18.3 EEG in a 7-week-old female born at 32 weeks, who has group B streptococcus meningitis, reveals interictal
diffuse suppression of activity with intermittent focal sharp waves (A), and independent left- and right-sided PLEDs

Similar to the EEG in meningitis, the encephalitic brain can have a normal EEG. The EEG abnormalities range from diffuse
produce a normal or an almost normal study, or display pro- slowing to electrographic seizures, and are of the two
found abnormalities. The same groups of pathogens that cause varieties—disease-specific and diagnostically vague. There are
meningitis, bacterial, viral, parasitic, and fungal, also com- three of the former variety and hundreds of the latter.
monly cause meningoencephalitis. Dissimilar to viral meningi- Nevertheless, the EEG is of value in both cases, since disease-
tis, the patients with viral encephalitis of any etiology rarely specific patterns represent common conditions, and the
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 335

Figure 18.3 (continued) (B and C) with no clinical accompaniment; electrographic seizures with the subtle correlate of
contralateral eye deviation were also seen.

absence of a specific pattern may in itself be diagnostic. The rapidly evolving into pseudoperiodic to periodic PLEDs, 0.3 to
evolution of encephalitic EEG from the prodromal to acute 1 Hz in frequency, and di- or triphasic broad or tight giant
phase of the disease and to recovery is similar to that in menin- sharp waves. These may be bilateral (biPEDs), bifrontal, bitem-
gitis, and may provide useful information for prognosis, com- poral, or even generalized (GPEDs). On occasion, PLEDs are
plications, and potential sequelae. focal with a small field in a specific location, such as the ante-
rior temporal region (Fig. 18.4).
Encephalitis with Disease-Specific EEG Patterns Temporally, PLEDs and/or focal sharp complexes reveal
Only three neurologic infections have a corresponding pathog- themselves within days to a week after the onset of symptoms,
but they may persist for several weeks into the illness, and may
nomic EEG signature: herpes simplex encephalitis, most common
be correlated with the response to treatment.
variants of prion disease, and subacute sclerosing panencephalitis
In addition, seizure discharges are common over the most
(SSPE), although the latter is a very rare diagnostic entity in the
affected areas, consisting of rhythmic activity or polyspike
Western world, where measles vaccination is a norm.
bursts. Clinical correlate may or may not occur, but confusion
Herpes Simplex (HSV-I) Encephalitis This common cata- and dysphasia are likely if the dominant hemisphere is affected.
strophic CNS illness strikes healthy people of all ages, but is Obtundation and coma are not uncommon, especially when
more common in patients over 50 years. HSV-1 has a predilec- EEG abnormalities are severe and widespread. When clinical
tion for the temporal lobes and for the frontotemporal areas, as motor seizures do occur, the frequency of clonic limb activity
compared to its cousin HSV-2, the presumed causative agent of may not match the frequency of PLEDs, which may be signifi-
Mollaret meningitis, which does not usually affect the cantly slower than the clonus.
parenchyma. The classical presentation of intermittent fever, In refractory or fatal cases, a burst suppression pattern may
confusion, and seizures with CSF lymphocytosis and positive be seen, with generalized EEG attenuation following a burst of
PCR for the virus is atypical in clinical practice: the diagnosis is PLEDs or polyspikes, which indicates a grim prognosis. In non-
best made based on any combination of clinical, radiographic, fatal HSV encephalitis, the strikingly abnormal EEG recovers
laboratory, and EEG findings compatible with this condition. slowly to almost normal on serial examinations. The PLEDs
Since this is one of the few treatable encephalitides, initial over- become infrequent, intermittent, and are replaced by lateralized
diagnosis is preferable in the case of HSV-1 encephalitis. slow-wave abnormalities. Clinical improvement often precedes
The initial EEG abnormalities (1,15) consist of polymorphic electrographic normalization.
delta slowing, often more prominent over the most involved Possibly because of the associated severe encephalopathy,
temporal lobe, with focal sharp complexes appearing and other, seemingly atypical patterns are frequently seen in HSV
336 Part III ■ Clinical EEG: General

Figure 18.4 EEG from a 62-year-old man with CSF-positive HSV encephalitis and MRI abnormalities maximal in the right
frontotemporal head region reveals initially right temporal slowing and isolated sharp waves and spikes (A), and with symp-
tomatic progression of the disease, pseudoperiodic PLEDs followed by suppression over the entire right hemisphere (B).

encephalitis, including generalized triphasic waves or atypical stroke or a hemorrhage is common, and radiologic correlation
triphasics, with posterior-to-anterior phase shift, multifocal is crucial.
sharp waves, and severe polymorphic delta slowing.
Because the most common reason for PLEDs is a vascular Prion Diseases Creutzfeldt–Jakob disease (CJD), variant CJD or
insult, dual etiology needs to be considered as part of the differ- “mad cow disease,” the exceedingly rare fatal familial insomnia
ential diagnosis. The coexistence of HSV-1 encephalitis and (a.k.a. Gerstmann–Straussler–Scheinker disease), and kuru are
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 337

the four well-described prion disorders, which can be familial, Subacute Sclerosing Panencephalitis SSPE is almost an irrele-
sporadic, or infectious in etiology. The pathogen is an abnor- vant condition in the Western world, where measles vaccina-
mally folding isoform of a native CNS protein, known as prion tions are routine. However, given an active antivaccination
protein. The most common variant of classic CJD is sporadic minority movement in the developed countries, and the fact
rather than genetic, although many inherited polymorphisms that measles is endemic in some underdeveloped countries, it is
predispose to the disease. Presenting with the clinical manifes- important to include SSPE in this discussion.
tations of a rapidly progressive dementia in a person over 60 SSPE occurs as a late complication in children who survived
years, it is often misdiagnosed at early stages as depression, psy- a measles infection, and clinically resembles CJD, with rapid
chosis, or another psychiatric illness. Spontaneous myoclonus intellectual decline, anorexia, spasticity, immobility, myoclonus,
and stimulus-related myoclonus come in later stages of the dis- choreiform movements, and seizures.
ease, as well as severe generalized spasticity, rhizus sardonicus, Typical periodic giant complexes are present in the EEG,
obtundation, and death. There are no known cases of recovery. with the frequent clinical accompaniment of jerks or whole-
The initial EEG abnormalities are nonspecific and mild, usu- body spasms.
ally consisting of generalized slowing and disorganization of The pattern, initially described by Radermecker and Poser
the background (16,17). However, subclinical, that is, noncon- (26), consists of high-voltage repetitive polyphasic and sharp-
vulsive status epilepticus (NCSE) or epilepsia partialis continua and slow-wave complexes 0.5 to 2 seconds in duration, occur-
(EPC) have been repeatedly described as initial presenting find- ring multiple times in 1 minute.
ings in CJD (18–23). Over weeks to months, focal di- or tripha- Multiple typical and atypical electrographic presentations of
sic sharp discharges appear, with an after-coming slow SSPE have been described (27–31). The typical pattern described
component. These are often generalized or bifrontal, occasion- by Radermecker, correlated with historical and clinical findings,
ally more prominent over one region or hemisphere, without a is definitive for the diagnosis, while atypical EEG patterns of sta-
clinical correlate. As the disease progresses, these become more tus epilepticus, bifrontal spikes, or other generalized abnormali-
persistent, severe, and eventually relentless (17). ties as described above may also be useful to the diagnosis.
The pathognomic pattern of CJD in the EEG consists of con-
tinuous generalized, bisynchronous periodic stereotypic sharp Encephalitis with Nonspecific EEG Patterns
waves, occurring in intervals of 0.5 to 1 second with duration of A multitude of infectious and autoimmune inflammatory
200 to 400 msec. Initially those abnormalities are lateralized; processes, with the two coexisting, affect the brain parenchyma,
however, they become generalized as disease progresses. causing disease clinically ranging from mild to catastrophic.
Atypical patterns may be seen, with bifrontal sharp waves that Common infections in pediatric and adult world, if investigated
are pseudoperiodic, or even with multifocal spike–wave dis- with a CSF and EEG, reveal abnormalities in both, with diffuse
charges. CSF 14-3-3 protein and MRI abnormalities on DWI slowing in the EEG roughly correlated with the onset and the
and later T2 images in cortex and basal ganglia are usually pos- resolution of symptoms. There are hundreds of reports of mild-
itive in both typical and atypical cases (Fig. 18.5) (24). to-moderate-to-fatal encephalitis associated with common
Myoclonic jerks maximal in the upper body may occur in pathogens such as coxsackie virus, adenovirus, mycoplasma,
close temporal association with generalized sharp wave com- rickettsia, mumps, rubella, measles, influenza A and B, and
plexes, or they may have no EEG correlate. Startle myoclonus or many others. None of those have an EEG pattern of significant
provoked myoclonus is seen on occasion. Occasionally, external diagnostic value. Almost all of these pathogens described as
stimuli can drive the rate of both the myoclonus and the corre- capable of causing meningitis actually cause meningoen-
sponding sharp wave discharges. cephalitis. Bacterial, viral, tuberculous, spirochetal, parasitic, or
In the advanced stages of the disease, a burst suppression fungal encephalitis typically has generalized severe polymor-
and then a diffuse suppression pattern is evident, with eventual phic slowing in the delta range on the EEG, but focal slowing is
isoelectric EEG heralding death. also possible in granulomatous or parasitic disease.
Atypical prion disease variants in younger populations or Viral encephalitis caused by enteroviridae, arboviridae, and
related to animal consumption have been described, where the echoviridae, with vectors ranging from ticks to mosquitoes,
pathognomic CJD pattern on the EEG is absent. Insufficient with frequently undetermined etiology, results in EEG abnor-
information exists to comment on the EEG abnormalities in malities ranging from mild to severe slowing to electrocerebral
kuru, “mad cow,” and a rare FFI variant of prion disease. silence, and from lateralized potentially epileptogenic dis-
Heidenhain variant of CJD has been described with the pre- charges to seizures and status epilepticus with burst suppres-
dominant occipital abnormalities of the EEG (25). sion pattern or with constant seizures (Fig. 18.6). The acute
Generalized sharp wave discharges and multifocal or EEG is rarely highly prognostic unless it contains a pattern of
bifrontal pseudoperiodic or periodic polyphasic discharges burst suppression, which is always ominous, and the long-term
with a clinical correlate of a rapidly progressive dementia prognosis, even with fair for survival, puts the patient at lifelong
should alert the practitioner to the possibility of a prion disease; risk of intractable multifocal seizures. Bilateral mesial temporal
however, other infectious or vascular or toxic–metabolic etiolo- sclerosis is not an uncommon complication of viral encephali-
gies should always be considered. Typical diffusion-weighted tis with or without an identified pathogen (Fig. 18.7).
MRI changes may be of help, but the diagnosis is made on a Borrelia burgdorferi-related encephalitis has gotten some
combination of longitudinal EEG and clinical findings. recent attention (32,33), although it is not different than other
338 Part III ■ Clinical EEG: General

Figure 18.5 EEG of a 61-year-old woman with tremor and confusion, with MRI findings suggesting CJD (abnormal T2
signal in the caudate and lentiform nuclei), reveals a progression over 2 weeks from disorganization and slowing with occa-
sional pseudoperiodic sharp waves (A) to continuous periodic sharp wave discharges that are lateralized with a wide field
(B), without a clinical correlate.

secondary autoimmune CNS inflammatory processes, and is diffuse or focal slowing. West Nile encephalitis generally presents
usually mild and self-limited clinically, with nonspecific with widespread electrographic abnormalities similar to other
encephalopathic changes in the EEG infrequently noted during viral meningoencephalitis (34), though anterior or temporal pre-
the course of the disease. dominant slowing has been reported (35). The range of EEG
Hepatitis, particularly hepatitis A, may cause CNS inflamma- abnormalities is wide, from none to status epilepticus and/or
tion and an encephalitic presentation with EEG features of burst suppression pattern that is always ominous (36–38).
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 339

Figure 18.6 Typical nonspecific EEG pattern shown in A, in a 25-year-old woman with encephalitis NOS without a known
pathogen, contracted during a summer vacation. It shows generalized severe slowing and multifocal spikes, some with a
narrow field, and some nearly generalized. Another typical nonspecific EEG pattern, B, is from a 17-year-old male with
CSF-negative, presumably tick-borne encephalitis with a devastating initial course of coma, choreiform movements, status
myoclonus, and wasting. The EEG during clinically comatose state demonstrates generalized moderately severe delta slow-
ing and suppression.
340 Part III ■ Clinical EEG: General

Figure 18.7 This awake-state EEG is from a 60-year-old man with neuroborreliosis, and demonstrates mostly generalized
suppression.

Other encephalitides of viral or presumed viral etiologies PART 2: IMMUNE-MEDIATED


also offer no specific EEG patterns of value for diagnosis INFLAMMATORY CNS CONDITIONS
(39–41).
To summarize, specific electroencephalographic diagnosis In the conditions discussed in Part 1, the brain is the primary
for most viral or bacterial or fungal or parasitic etiologies of or secondary target of attack by an infection or a presumed
encephalitis does not exist; however, serial EEG recordings pro- infection. In immune-mediated conditions, no CNS infection
vide a good basis for prognostication (42). The amazing thing can be identified, and other organ systems may be the princi-
about viral encephalitis is that a severely abnormal EEG may pal site of the inflammation. In cerebral demyelinating disor-
recover in days or weeks, and the EEG recovery is often corre- ders and Rasmussen encephalitis the principal site of the
lated with the clinical outcome. inflammation appears to be the brain. In immune-mediated
conditions the EEG abnormalities may be diffuse or lesion-
HIV-Related Encephalitis specific. In lupus and antiphospholipid antibody diseases,
Clinically or electrographically, AIDS-related encephalitis is not cerebral inflammation is partly secondary to systemic activa-
different from other viral encephalitides; however, there is evi- tion of pro-inflammatory cytokines, but the effects on vascula-
dence of early CNS involvement and poor prognosis in some ture may also result in ischemia that then accounts for the focal
studies (43,44). HIV, “the great imitator,” may present with nature of some EEG abnormalities observed clinically. In para-
meningoencephalitis at early, middle, or late stages of the dis- neoplastic CNS injury, although the tumor may be the primary
ease, without the evidence for secondary opportunistic CNS site of immune-mediated attack, the brain is the site of the
infection (45). No disease-related or diagnostic EEG pattern most damage. Some of the same immunogenic CNS antigens
specific for HIV meningoencephalitis has been reported, though also may be attacked in the absence of a neoplasm, and result
multiple studies confirmed that a variety of generalized and in similar damage.
focal abnormalities in the EEG may be seen, which are different
for the acute and chronic phases of the disease (43,44). In the Idiopathic Inflammatory Demyelinating Disorders
early acute phase, the EEG may be normal or may show the non- Acute disseminated encephalomyelopathy (ADEM) is an
specific pattern of diffuse slowing, while in the late stages, severe umbrella diagnosis for a generally monophasic cerebral
delta slowing and even suppression can be seen. Encephalitides demyelination event, with a range of manifestations from mild
caused by opportunistic infections in AIDS, such as Toxoplasma to very severe. A prodromal event such an upper respiratory
gondii, Cytomegalovirus, and Candida, are discussed above, and infection, recent vaccination, or a flu-like illness is frequently
may show focal EEG abnormalities corresponding to the under- identified, as are recent trauma and surgery. The Marburg vari-
lying lesions, as well as generalized slowing (45). ant of fulminant multiple sclerosis (MS), acute hemorrhagic
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 341

leukoencephalitis (Hurst disease), tumefactive MS (Balo prevent or arrest the devastating course of the disease. Most
disease), and Schilder disease in children are related conditions. patients are hemiparetic by early adolescence, have uncontrol-
Although white matter is primarily affected in those condi- lable hemiconvulsions and severely reduced function by the late
tions, the inflammation may affect both normal cortical func- teens, and require full-time or inpatient care by their twenties.
tion and neuronal excitability, and result in EEG abnormalities Late EEG findings reveal frequent unilateral seizures with a
(46,47) that are varied and nonspecific, with both diffuse and wide field, and bilaterally abnormal background (Fig. 18.8).
focal delta slowing typical for deep white matter lesions. There
may also be focal potentially epileptogenic discharges in tume- Hashimoto Encephalopathy
factive MS and ADEM (47). With recovery the EEG usually nor- Hashimoto encephalopathy (or encephalitis) (HE) or steroid-
malizes, somewhat proportional to the extent of recovery from responsive encephalopathy associated with autoimmune thy-
the initial event. roiditis (SREAT) is a serious but treatable condition that is
In chronic and late-stage MS, EEG is usually abnormal, with probably underdiagnosed, partly because it has a broad range of
diffuse slowing reflecting the degree of encephalopathy, how- clinical presentations. The encephalopathy usually presents with
ever, without a clear correlation to radiographic plaque load the subacute or abrupt onset of confusional state or alteration of
(48). Although overall only 6.5% of MS patients in a recent the level of consciousness, and is frequently accompanied by the
study had epilepsy, the percentage was significantly higher in development of focal or generalized seizures. The most frequent
progressive MS patients. The interictal EEG of MS patients with clinical symptoms in a recent meta-analysis (54) were seizures
epilepsy may reveal epileptiform abnormalities in up to two (80%), confusion (52%), headache (40%), hallucinations
third of these patients (49). MS plaques effectively disconnect (32%), and ataxia (36%). Tremor, myoclonus, stroke-like events
cortical areas, potentially leading to decreased inhibitory trans- (including transient aphasia), and rapid or gradually progressive
mission and ictogenesis. cognitive and behavioral decline are also seen (55,56).
Several thyroid-related autoantibodies have been found in
Rasmussen Encephalitis serum and CSF of patients with HT, reacting against thyroglob-
This progressive epileptic encephalopathy of uncertain etiology ulin, microsomal antigens, thyroid peroxidase, colloid-related
and pathogenesis, involving one hemisphere, could be dis- cell surface antigen, the thyroid-stimulating hormone (TSH)
cussed with the rest of the epileptic encephalopathies. These are receptor, as well as thyroxin and triiodothyroxine (57). In addi-
childhood disorders characterized by cognitive, speech, hemi- tion, patients have cell-mediated immunity directed against
motor, and functional deterioration, and with progressive EEG thyroid antigens (58). High titers of antithyroid antibodies in
changes. However, traditionally Rasmussen disease has been blood and high CSF protein without an accompanying pleocy-
discussed in this section. It is likely that immune-mediated CNS tosis are often seen (59), but not reliably. A recent study found
inflammation is part of the etiopathogenesis of other epileptic that CSF IgG from HE patients specifically recognized two iso-
encephalopathies. In the case of Rasmussen encephalitis, a pri- forms of dimethylargininase-I (DDAHI), and aldehyde reduc-
mary role for the immune attack was proposed (50), but fails to tase-I (AKRIAI) (60).
explain the disease adequately (51) as common CNS antigenic A report of a patient with HE who died of an unrelated con-
responses are seen in many other CNS conditions. dition suggests a peculiar brain stem-dominated vasculitis
Autoantibodies against glutamate receptors, once thought to characterized by T-lymphocytic infiltrates of leptomeningeal
be specific for Rasmussen encephalitis, are found in other focal venules (58). Similar findings were seen in a case in our practice
epilepsies, ischemic infarcts, transient ischemic attacks (TIAs), (RL Beach, personal observations). In another case autopsy
spinocerebellar atrophy, systemic lupus erythematosus (SLE), showed dense lymphocytic infiltrates of the wall of the
and paraneoplastic encephalopathies, and are thus not useful in parenchymal arterioles and venules (61).
the diagnosis of Rasmussen encephalitis. They are a biomarker The Mayo group reported mild to severe generalized slowing
for brain injury particular to many encephalopathies, and may on the EEG corresponding to the severity of the clinical
be of importance in neurologic dysfunction in various condi- encephalopathy, as well as triphasic waves, epileptiform
tions. Immune suppression as a therapy in Rasmussen abnormalities, photomyogenic, and photoparoxysmal
encephalitis may slow the progression of cerebral degeneration responses (62). Others have also found focal or lateralized slow-
and functional decline, but has limited, and transient, effects on ing and epileptiform discharges (Fig. 18.9), and seizures, in
the clinical or electrographic seizures, or on the progression and addition to generalized slowing with high-voltage, delta, bipha-
evolvement of EEG abnormalities (52). sic, and triphasic waves (55,63). When follow-up EEGs have
EEG abnormalities in both glutamate receptor antibody pos- been reported, the EEG findings improved or disappeared with
itive and negative diseases initially consist of lateralized slow- steroid treatment.
ing, followed in months to years by the emergence of potentially Electroencephalography, brain MRI, cerebrospinal fluid
epileptogenic spikes and sharp wave discharges, often associ- examination, and serological tests are useful diagnostic tools,
ated with clinical and electrographic seizures evolving into but none is yet diagnostic in itself. The encephalopathy usually
the syndrome of EPC. Focal electrical status epilepticus responds well to corticosteroid therapy, although additional
undetectable on standard scalp EEG has been demonstrated in immunosuppressive agents are sometimes used (64). The long-
Rasmussen encephalitis (53). An early hemispherectomy may term prognosis generally appears to be favorable.
342 Part III ■ Clinical EEG: General

Figure 18.8 These EEG tracings from a 24-year-old man with late sequelae of Rasmussen encephalitis and epilepsia par-
tialis continua (EPC) illustrate the variety of interictal and ictal abnormalities observed at any time of sampling the EEG.
Interictally (A), only a moderately severe slowing and a lack of normal background is noted: PLED-like pattern without a
clinical correlate is also common (B).

Limbic Encephalitis relies on the clinical picture combined with the demonstration of
The classic syndrome of limbic encephalitis includes subacute cog- MRI and EEG abnormalities in the temporal lobes, and presence of
nitive impairment with behavioral changes, hallucination, sleep inflammatory changes: in the cerebrospinal fluid (66), on imaging
disturbance, and temporal lobe seizures (56,65). The diagnosis (67,68), and rarely pathologically (69–71). Once considered an
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 343

Figure 18.8 (continued) Epilepsia partialis continua in Rasmussen syndrome (EPC) may present as morphologically differ-
ent seizure patterns that may have no clinical accompaniment (C); or may be accompanied by rhythmic movements (D).

extremely rare disorder, almost always related to cancer, and refrac- Regardless whether limbic encephalitis is paraneoplastic or
tory to treatment, limbic encephalitis is now regarded as a relatively an autoimmune response to normal antigens, the clinical
frequent disorder, often unrelated to cancer, associated with and radiologic findings at presentation of these patients are
immune reaction to neuronal surface antigens, that may respond indistinguishable (56). The electroencephalographic findings
to immunotherapy (65,72). are nonspecific, may be similar to those seen in a variety of
344 Part III ■ Clinical EEG: General

Figure 18.9 This EEG shows interictal slowing and sharp waves in a patient with Hashimoto thyroiditis and subacute
encephalopathy. She had C lymphocytosis and increased protein, as well as dramatically elevated thyroid peroxidase and
thyroglobulin antibodies. She improved dramatically with a short course of high-dose solumedrol. Several years later she
relapsed and had similar findings and course.

encephalopathies or associated with other etiologic factors that been described (66,73,77). Epileptiform EEG abnormalities
produce temporal lobe seizures. These EEG findings vary from in temporal lobe were seen in 50% to 60% of patients with
normal to severely abnormal. paraneoplastic limbic encephalitis.
The background activity varies from normal to variable Electrographic seizures have onset over the temporal region
slowing, which may be focal or generalized (66,68,73). Focal (Fig. 18.9) and may secondarily generalize (66,78). The clinical
slowing occurs mostly in the temporal region (66–68) and may deterioration of patient’s mental status correlates with worsen-
be more prominent on the left (67,71). It may also be frontal or ing of EEG findings. Epileptogenic potential may start focally,
unilateral hemispheric. Patients with generalized slowing gen- and then evolve into a more diffuse process, eventually involv-
erally had clinical and MRI evidence of more widespread cen- ing the entire brain as the patient’s condition worsens. This may
tral nervous system disease, than patients with focal slowing become equivalent to EPC. This may be associated with focal or
(55). In addition to generalized slowing, triphasic waves and diffuse pathologic abnormalities (Fig. 18.10) (73,77).
FIRDA have been described (68). In one study, electroen- A very rapidly progressive nonconvulsive status epilepticus,
cephalography was reported to be the most sensitive investiga- starting from focal pseudoperiodic spike and wave discharges,
tion, with generalized or focal slowing as the most commonly extending to the contralateral hemisphere within a week and
observed abnormality (66). becoming continuous sharp waves a day later, despite
Epileptiform abnormalities that occur mostly in the tem- antiepileptic medications was reported in one patient, without
poral region, where they usually are unilateral, bilateral, any imaging abnormalities (79). Seizures in limbic encephalitis
bilateral independent, or multifocal independent, but they are frequently refractory to antiepileptic medications and may
may also be extratemporal, multifocal, or generalized require surgical intervention (74,76,80–82).
(66,68,75,76). The spikes or sharp waves are always most
prominent over the anteriorly situated temporal scalp elec- Antiphospholipid Antibody Syndrome
trodes. Focal epileptiform activity is strongly associated with The antiphospholipid syndrome (APS) is defined by the pres-
clinical seizure and with MRI signal abnormalities in the ence of laboratory markers such as antiphospholipid antibody
medial temporal regions (66). PLEDs or bilateral epilepti- syndrome (APLA) and circulating anticoagulant (CAC) and
form discharges (BIPLEDs), predominantly involving the clinical manifestations such as recurrent thrombosis and
temporal region, but also parietal and frontal regions, have abortions (83).
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 345

Figure 18.10 This EEG shows a focal-onset, left temporal lobe seizure in a patient with antibodies to voltage potassium
channels in the absence of any neoplasm. He presented with intermittent and subacute progressive confusion and MRI
showed left greater than right temporal lobe increased signal on FLAIR. Interictal epileptiform discharges and subtle slow-
ing were also seen.

Among patients with neuropsychiatric symptoms, EEG The electroencephalographic findings in BD are nonspecific
abnormalities are common and correlate with the presence of and vary from normal to severely abnormal. Diffuse and focal
APLA even in the absence of brain abnormalities on the MRI. slowing in the delta and theta range occurs in patients with BD
The main EEG abnormalities are bursts of bitemporal irregular (89–91). Focal slowing is seen over frontal, temporal, or fron-
slowing, consisting of a mixture of theta and delta waves, often totemporal regions (89,92). They are usually associated with
with accentuation on the left during wakefulness. Epileptogenic pathologic abnormalities on imaging studies (89,92). Abnormal
potentials such as temporal sharp waves and generalized epilep- EEG findings are found in up to 80% of patients with neuro-
tiform abnormalities can also be seen (84). Among patients with logic involvement (93), and up to one third of patients without
dementia or memory problems, the EEG may show diffuse or any neurologic symptoms (93,94).
bitemporal slowing and rarely a focal abnormality (77,83,84). EEG findings ranging from normal to severely abnormal
There is a relationship between antiphospholipid antibodies may be seen in patients with subclinical neurologic involve-
and patients with epilepsy (83,85–87). The presence of ment. Abnormal findings include focal slowing in the temporal
antiphospholipid antibodies is associated with recurrent or parieto-occipital regions, spike waves in the temporal, occip-
seizures in patients with refractory focal epilepsy, such as tem- ital, or frontal region, polyspikes, multifocal spikes, generalized
poral lobe epilepsy and extratemporal lobe epilepsy including spikes, polyspike–wave complexes, secondary bilateral syn-
multifocal epilepsy (87). chrony, slow spike waves, 6-Hz spike waves, polyspike bursts,
photoparoxysmal response, photomyogenic response, and theta
Behcet Disease and delta waves (94).
Behcet disease (BD) is a multisystem disease of unknown Neurologic involvement in BD varies and represents one of
etiopathogenesis characterized by recurrent oral and genital the most severe BD manifestations (92). The reported fre-
aphthous ulcers and uveitis with various clinical manifestations quency of seizures varies from 0% to 27% (89,90,95–97).
due to central nervous involvement. Neurologic involvement Epileptic seizures occur as an initial neurologic presentation
has been classified into two major forms: the intra-axial neuro- (28,33) or as a sole neurologic manifestation of BD (92). They
Behcet syndrome (NBS), which is the inflammation of the also occur during exacerbations and chronic course of BD (89).
small veins of the parenchyma in the central nervous system, Seizures have been reported to be provoked by fever, medica-
and the extra-axial NBS that is the thrombosis of the cerebral tions (INH, penicillin, and interferon), or withdrawal of
venous sinus (88). antiepileptic medications (89).
346 Part III ■ Clinical EEG: General

Seizures may be simple partial, complex partial, myoclonic pathologic EEG changes than those suffering from a form of the
jerks, generalized tonic–clonic seizures epilepsialis partialis con- disease with symptoms only in the joints. There is no clear cor-
tinua (EPC) (85), or convulsive status epilepticus (89–92,97,98). relation between EEG changes and the duration of the disease or
Focal epileptic activity over temporal and frontal temporal age at onset (113). The pathologic EEG abnormalities are pre-
regions has been reported (89). In one study, generalized sumed to be caused by primary cerebral process connected with
tonic–clonic convulsions accompanied by focal motor seizures the disease itself, probably vasculitis (113,114).
were the predominant type, and seizures were associated with
high mortality rate (89). Motor seizures, including epilepsia par- Sjogren Syndrome
tialis continua, are associated with a poor prognosis (82). Other Sjogren syndrome is an autoimmune disease that may be pri-
EEG findings reported include PLEDs (99,100). mary or secondary to another connective tissue disease charac-
There are conflicting findings regarding the abnormalities in terized by mononuclear infiltration and destruction of salivary
the CSF and the seizure frequency and severity (89,101). EEG and lachrymal glands leading to xerostomia and xerophthalmia.
abnormalities, however, are correlated with CSF pleocytosis CNS involvement occurs in up to 68%, cognitive involvement
(92), and the presence of (but not reliably the nature or location in 11%, and seizures in 8.5% of patients with Sjogren syndrome
of) clinical symptoms and progression of NBD (102). (115). Various types of seizures occur in Sjogren syndrome
including partial, temporal mostly and parietal, and generalized
Primary Angiitis of the Central Nervous System seizures (115).
Primary angiitis of the central nervous system (PACNS) is rare
multisystem vasculitis that primarily involves the central nerv- Systemic Lupus Erythematosus
ous system. Electroencephalography is a highly sensitive test in Abnormal electrographic findings have been reported in up to
the diagnostic of PACNS (103,104). The EEG abnormalities 87% of SLE patients (116). These are mostly nonspecific gener-
tend to present early in the course of the disease, often being the alized slowing in theta and delta ranges (117–119). In one
sole laboratory abnormality in the initial evaluation (104). The study, paroxysmal focal changes and abnormalities in the back-
lack of specificity compromises EEG’s usefulness in confirming ground were the most common findings (120). Patients with-
the diagnosis. The most common abnormality is diffuse slow- out neurologic deficits have abnormalities in the EEG,
ing, but at times it may be focal (103–105). suggesting the presence of subclinical involvement of central
Focal symptoms occur in 50% or more of patients, and they nervous system (120). However, there is no significant associa-
reliably occur in the setting of diffuse cortical dysfunction as tion between EEG abnormalities and cerebral atrophy, and the
evidenced by encephalopathy and/or diffuse EEG changes EEG abnormalities are not correlated with functional deficits in
(104). Seizures have been noted in 10% to 25% of cases of the neuropsychologic tests. Patients with EEG abnormalities
PACNS (103,104). Clear electrographic seizures are uncommon have significantly lower complement factor C4, more pro-
(96). Partial motor seizures related to focal cerebral vasculitis in nounced thrombocytopenia, and significantly higher titer of
the temporal and parietal lobes, progressing to generalized anticardiolipin IgG than those with normal EEG (119).
tonic–clonic seizures that responded to steroid treatment, have Unilateral and bilateral, focal and more widespread abnor-
been reported (106,107). malities including delta and theta slowing and sharp waves have
been reported; the left hemisphere, particularly the left tempo-
Rheumatoid Arthritis ral leads, is the most common abnormality (116,117,119).
Cerebral involvement associated with rheumatoid arthritis Routine and quantitative EEG analyses also show theta and
(RA) is rare but well documented. This includes vasculitis, cere- delta slowing predominantly affecting the left hemisphere in
bral pachymeningitis, leptomeningitis caused by fibrinoid infil- patients with neuropsychiatric manifestations (121).
tration and rheumatoid nodule, choroid plexus infiltration, or Among SLE patients with headache, symptomatic headache
cerebral complications induced by hyperviscosity associated is associated with frequent paroxysmal changes and idiopathic
with high titers of rheumatoid factors presenting as headache is associated with background changes. Idiopathic
encephalopathy or seizures and responding to immunomodu- headache shows more cerebral pathology on imaging study
latory treatment (108–111). including multiple perfusion deficits on SPECT and hyperin-
The morning symptoms of pain, weakness, and fatigue in tense white matter lesions in the bilateral spheres on MRI (120).
RA may relate to a nonrestorative sleep disorder associated with Epileptic seizures occur in 8% to 37% of cases (116,123–125).
prominent alpha rhythm EEG anomaly, a presumed arousal Seizures occur mostly after the onset of SLE, and acute sympto-
state occurring within sleep, during NREM sleep stages 2, 3, and matic seizures are related to stroke and antiphospholipid anti-
4 (112). bodies. Most patients with a single epileptic seizure have normal
In juvenile rheumatoid arthritis (JRA), various EEG abnor- EEG findings. Those with recurrent epileptic seizures generally
malities are seen including slowing of activity that may be con- have abnormal EEGs, with interictal epileptiform abnormalities
tinual or episodic, abundant beta activity, focal-asymmetrical predominant in the temporal lobes (125).
disturbances that could be either paroxysmal or nonparoxysmal, Generalized tonic–clonic and complex partial seizures are
and bilateral paroxysmal disturbances including photic-induced the most common type of seizures. Other seizure types include
paroxysm. Patients with positive antinuclear antibodies or vis- simple partial and secondary generalized (126). Complex par-
ceral symptoms like pericarditis and/or myocarditis have fewer tial status epilepticus has been reported with a prolonged
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 347

period of confusion and EEG revealed continuous rhythmic • Some more specific patterns—PLEDs or FIRDA—may be
slow waves and triphasic waves (127); resolution of the EEG associated with treatable conditions that are otherwise not
abnormalities and improvement of clinical findings were noted suspected, for example, early HSV encephalitis without fever
after a benzodiazepine injection. or CSF abnormalities, or basal meningitis with developing
hydrocephalus.
Sydenham Chorea • Gross hemispheric asymmetries uncovered by the EEG in
Sydenham chorea is an immune-mediated movement disorder cases where imaging is delayed or equivocal may be diagnos-
associated with acute rheumatic fever, seen mainly in children tic for a lateralized process such as a brain abscess or a sub-
and adolescents. Three types of EEG abnormalities were dural empyema, that is a neurosurgical emergency.
described in 17 of 20 children 10 to 16 years of age, with rheu-
Of no less importance than the diagnosis is the information
matic chorea (128). Seven patients had increased amount of
that the EEG offers about the severity and extent of CNS disease,
posterior slowing, five patients had disruption of the alpha
which is crucial for monitoring the response to treatment.
rhythm and diffuse, monomorphic slowing, and five patients
Developing a library of EEG markers and patterns that may
had bursts of 2- to 4-Hz posterior delta. No correlation between
accurately predict prognosis in inflammatory and infectious
the EEG abnormalities and severity of the chorea, or between
CNS disease is a project for the future. With the maturation of
localization of the EEG abnormalities and the distribution of
quantitative and digital EEG techniques there should be a
movements, was found. Others have found EEG slowing pro-
renewed interest in pursuing this. Despite the nonspecificity of
portional to the severity of the movement disorder (129,130),
the brain response to inflammation, certain features may emerge
and rarely epileptiform abnormalities have been observed
that are helpful in predicting the residual damage to the brain.
(129). Lateralized suppression of sleep spindles without
In conclusion, the EEG in inflammatory and infectious dis-
improvement with the disease regression has also been reported
orders has significant diagnostic and prognostic value, and the
(130). Others report the EEG may improve with the clinical
pathognomic as well as nonspecific pathologic EEG patterns
state, or show a delayed response (131).
need to be recognized by the electroencephalographer.
Other Autoimmune Encephalitides
Kawasaki disease, postinfectious and postimmunization REFERENCES
encephalitis that does not have the imaging findings of ADEM,
1. Radermecker FJ. Infections and Inflammatory Reactions, Allergy
as well as Vogt–Koyanagi–Harada disease—all may present with
and Allergic Reactions; Degenerative Diseases. Amsterdam: Elsevier
transient meningoencephalitis of varied clinical severity. The Scientific Publishing Co; 1997.
EEG pattern in those is nonspecific, reflecting the diffuse 2. Westmoreland BF. The EEG in cerebral inflammatory processes.
encephalopathic state of the patient. Seizures or focal abnor- In: Niedermeyer E, Lopes da Silva F, eds. Niedermeyer’s Textbook of
malities are possible but uncommon. Electroencephalography: Basic Principles, Clinical Applications, and
Related Fields. 5th ed. Philadelphia: Lippincott, Williams and
Wilkins; 2005:323–338.
SUMMARY 3. Kepa L, Oczko-Grzesik B, Warakomska I, et al. Badania EEG u
Quoting Dr. Westmoreland from the previous edition of this chorych z ropnymi, bakteryjnymi zapaleniami opon i mozgu-
przeglad publikacji i obserwacje wlasne [EEG examination in
chapter (2):
patients with purulent, bacterial meningoencephalitis—
Although the EEG shows a variety of patterns with various literature review and own observations]. Wiad Lek. 2006;
inflammatory processes, the EEG can help to (a) confirm and 59(7–8):458–462.
indicate the degree and extent of CNS involvement by the 4. Zoons E, Weisfelt M, de Gans J, et al. Seizures in adults with bacte-
inflammatory process; (b) make the diagnosis of herpes simplex rial meningitis. Neurology. 2008;70(22 pt 2):2109–2115.
encephalitis, Jacob–Creutzfeldt disease, and SSPE when the 5. Bartel P, Schutte CM, Becker P, et al. Discrimination between viral
characteristic electroencephalographic pattern of these diseases and nonviral meningitis by visually analyzed and quantitative elec-
is present; (c) indicate the presence of a focal lesion; (d) moni- troencephalography. Clin Electroencephalogr. 1999;30(2):35–38.
tor the course of the disease process; and (e) detect the develop- 6. Konno S, Sugimoto H, Nemoto H, et al. Triphasic waves in a
ment of complications and sequelae. patient with tuberculous meningitis. J Neurol Sci. 2010;
291:114–117.
In addition to her succinct summary of the usefulness of the 7. Kalita J, Misra UK, Das BK. SPECT changes and their correlation
EEG in infectious and inflammatory disorders, we want to with EEG changes in tuberculous meningitis. Electromyogr Clin
emphasize the adjuvant diagnostic value of the EEG. Despite Neurophysiol. 2002;42(1):39–44.
the great range of possible EEG patterns at presentation and 8. Patwari AK, Aneja S, Ravi RN, et al. Convulsions in tuberculous
their significant overlap, the initial diagnostic value of the EEG meningitis. J Trop Pediatr. 1996;42(2):91–97.
should not be overlooked because: 9. Wang RJ, Wang DX, Wang JW, et al. Analysis of 62 adult patients
with viral meningitis. Zhonghua Shi Yan He Lin Chuang Bing Du
• Many of the above-described inflammatory and infectious Xue Za Zhi. 2009;23(3):218–220.
conditions can present with NCSE, which is a life- threatening 10. Pollak L, Klein C, Schiffer J, et al. Electroencephalographic abnor-
neurologic emergency requiring immediate aggressive treat- malities in aseptic meningitis and noninfectious headache. A com-
ment, and EEG remains the one modality to diagnose NCSE. parative study. Headache. 2001;41(1):79–83.
348 Part III ■ Clinical EEG: General

11. Capouya JD, Berman DM, Dumois JA. Mollaret’s meningitis due 31. Westmoreland BF, Sharbrough FW, Donat JR. Stimulus-induced
to human herpesvirus 6 in an adolescent. Clin Pediatr. EEG complexes and motor spasms in subacute sclerosing panen-
2006;45(9):861–863. cephalitis. Neurology. 1979;29(8):1154–1157.
12. Gao B, Yang J, Zhuang S, et al. Mollaret meningitis associated with 32. Eriksson B, Wictor L. EEG with triphasic waves in Borrelia
an intraspinal epidermoid cyst. Pediatrics. 2007;120(1):e220–e224. burgdorferi meningoencephalitis. Acta Neurol Scand. 2007;116(2):
13. Mikdashi J, Kennedy S, Krumholz A. Recurrent benign lympho- 133–136.
cytic (Mollaret) meningitis in systemic lupus erythematosus. 33. Kerling F, Blumcke I, Stefan H. Pitfalls in diagnosing limbic
Neurologist. 2008;14(1):43–45. encephalitis—a case report. Acta Neurol Scand. 2008;118(5):
14. Prandota J. Mollaret meningitis may be caused by reactivation of 339–342.
latent cerebral toxoplasmosis. Int J Neurosci. 2009;119(10): 34. Klein C, Kimiagar I, Pollak L, et al. Neurological features of West
1655–1692. Nile virus infection during the 2000 outbreak in a regional hospi-
15. Cobb W. The periodic events of subacute sclerosing leucoen- tal in Israel. J Neurol Sci. 2002;200(1–2):63–66.
cephalitis. Electroencephalogr Clin Neurophysiol. 1966;21(3): 35. Gandelman-Marton R, Kimiagar I, Itzhaki A, et al.
278–294. Electroencephalography findings in adult patients with West Nile
16. Wieser HG, Schwarz U, Blattler T, et al. Serial EEG findings in spo- virus-associated meningitis and meningoencephalitis. Clin Infect
radic and iatrogenic Creutzfeldt–Jakob disease. Clin Neurophysiol. Dis. 2003;37(11):1573–1578.
2004;115(11):2467–2478. 36. Bagic A, Boudreau EA, Greenfield J, et al. Electro-clinical evolution
17. Wieser HG, Schindler K, Zumsteg D. EEG in Creutzfeldt–Jakob of refractory non-convulsive status epilepticus caused by West Nile
disease [see comment]. Clin Neurophysiol. 2006;117(5):935–951. virus encephalitis. Epileptic Disord 2007;9(1):98–103.
18. Cohen D, Kutluay E, Edwards J, et al. Sporadic Creutzfeldt–Jakob 37. DellaBadia J Jr, Jaffe SL, Singh J, et al. An occipital lobe epilepto-
disease presenting with nonconvulsive status epilepticus. Epilepsy genic focus in a patient with West Nile encephalitis. Eur J Neurol.
Behav. 2004;5(5):792–796. 2004;11(2):111–113.
19. Fernandez-Torre JL. Creutzfeldt–Jakob disease presenting as 38. Rodriguez AJ, Westmoreland BF. Electroencephalographic charac-
refractory nonconvulsive status epilepticus. J Intensive Care Med. teristics of patients infected with West Nile virus. J Clin
2005;20(3):180–181. Neurophysiol. 2007;24(5):386–389.
20. Fernandez-Torre JL. Nonconvulsive status epilepticus in 39. Misra UK, Kalita J, Syam UK, et al. Neurological manifestations of
Creutzfeldt–Jakob disease. Clin Neurophysiol. 2006;117(8): dengue virus infection. J Neurol Sci. 2006;244(1–2):117–122.
1879–1880. 40. Nishiya K, Taira K, Kuroki S. A case of secondary meningoen-
21. Parry J, Tuch P, Knezevic W, et al. Creutzfeldt–Jakob syndrome cephalitis associated with mycoplasma pneumoniae infection.
presenting as epilepsia partialis continua. J Clin Neurosci. Kansenshogaku Zasshi. 2001;75(3):209–212.
2001;8(3):266–268. 41. Okamoto K, Fukuda M, Shigemi R, et al. Two cases of acute
22. Rossetti AO, Dunand M. Creutzfeldt–Jakob disease: evolution encephalitis/encephalopathy associated with adenovirus type 3
from nonconvulsive status epilepticus, through SIRPIDs, to gener- infection. No to Hattatsu. 2004;36(6):487–491.
alized periodic discharges. Clin Neurophysiol. 2007;118(11): 42. Misra UK, Kalita J. Seizures in encephalitis: predictors and out-
2533–2536. come. Seizure. 2009;18(8):583–587.
23. Shapiro JM, Shujaat A, Wang J, et al. Creutzfeldt–Jakob disease 43. Bernad PG. The neurological and electroencephalographic
presenting as refractory nonconvulsive status epilepticus. changes in AIDS. Clin Electroencephalogr. 1991;22(2):65–70.
J Intensive Care Med. 2004;19(6):345–348. 44. Tinuper P, de Carolis P, Galeotti M, et al. Electroencephalogram
24. Lanska DJ. Diagnosis of Creutzfeldt–Jakob disease: effect of clini- and HIV infection: a prospective study in 100 patients. Clin
cal criteria on incidence estimates. Analysis of EEG and CSF Electroencephalogr. 1990;21(3):145–150.
14-3-3 proteins as aids to the diagnosis of Creutzfeldt–Jakob dis- 45. Villar Del Saz S, Sued O, Falco V, et al. Acute meningoencephalitis due
ease. Neurology. 2001;56(10):1422–1423. to human immunodeficiency virus type 1 infection in 13 patients:
25. Keyrouz SG, Labib BT, Sethi R. MRI and EEG findings in clinical description and follow-up. J Neurovirol. 2008;26:1–6.
Heidenhain variant of Creutzfeldt–Jakob disease. Neurology. 46. Chabolla DR, Moore JL, Westmoreland BF. Periodic lateralized
2006;67(2):333. epileptiform discharges in multiple sclerosis. Electroencephalogr
26. Radermecker J, Poser CM. The significance of repetitive parox- Clin Neurophysiol. 1996;98(1):5–8.
ysmal electroencephalographic patterns. Their specificity in 47. Levic ZM. Electroencephalographic studies in multiple sclerosis.
subacute sclerosing leukoencephalitis. World Neurol. 1960;1: Specific changes in benign multiple sclerosis. Electroencephalogr
422–433. Clin Neurophysiol. 1978;44(4):471–478.
27. Caselli RJ, Scheithauer BW, O’Duffy JD, et al. Chronic inflamma- 48. Striano P, Orefice G, Brescia Morra V, et al. Epileptic seizures in
tory meningoencephalitis should not be mistaken for Alzheimer’s multiple sclerosis: clinical and EEG correlations. Neurol Sci.
disease. Mayo Clin Proc. 1993;68(9):846–853. 2003;24(5):322–328.
28. Kister I, Inglese M, Laxer RM, et al. Neurologic manifestations of 49. Martinez-Juarez IE, Lopez-Meza E, Gonzalez-Aragon Mdel C,
localized scleroderma: a case report and literature review. et al. Epilepsy and multiple sclerosis: increased risk among pro-
Neurology. 2008;71(19):1538–1545. gressive forms. Epilepsy Res. 2009;84(2–3):250–253.
29. Schott JM. Ischemic bilateral hippocampal dysfunction during 50. Rogers SW, Andrews PI, Gahring LC, et al. Autoantibodies to glu-
transient global amnesia. Neurology. 2008;70(20):1940–1941. tamate receptor GluR3 in Rasmussen’s encephalitis. Science.
30. Smith JB, Westmoreland BF, Reagan TJ, et al. A distinctive clinical 1994;265(5172):648–651.
EEG profile in herpes simplex encephalitis. Mayo Clin Proc. 51. Pleasure D. Diagnostic and pathogenic significance of glutamate
1975;50(8):469–474. receptor autoantibodies. Arch Neurol. 2008;65(5):589–592.
Chapter 18 ■ The EEG in Inflammatory CNS Conditions 349

52. Bien CG, Schramm J. Treatment of Rasmussen encephalitis half a 73. Gultekin SH, Rosenfeld MR, Voltz R, et al. Paraneoplastic limbic
century after its initial description: promising prospects and a encephalitis: neurological symptoms, immunological findings and
dilemma. Epilepsy Res. 2009;86(2–3):101–112. tumour association in 50 patients. Brain. 2000;123(pt 7):1481–1494.
53. Fattouch J, Di Bonaventura C, Di Gennaro G, et al. Electrical sta- 74. Fadul CE, Stommel EW, Dragnev KH, et al. Focal paraneoplastic
tus epilepticus “invisible” to surface EEG in late-onset Rasmussen limbic encephalitis presenting as orgasmic epilepsy. J Neurooncol.
encephalitis. Epileptic Disord. 2008;10(3):219–222. 2005;72(2):195–198.
54. Alink J, de Vries TW. Unexplained seizures, confusion or halluci- 75. Alamowitch S, Graus F, Uchuya M, et al. Limbic encephalitis and
nations: think Hashimoto encephalopathy. Acta Paediatr. small cell lung cancer. Clinical and immunological features. Brain.
2008;97(4):451–453. 1997;120(pt 6):923–928.
55. Rodriguez AJ, Jicha GA, Steeves TD, et al. EEG changes in a patient 76. Jacobs DA, Fung KM, Cook NM, et al. Complex partial status
with steroid-responsive encephalopathy associated with antibod- epilepticus associated with anti-Hu paraneoplastic syndrome.
ies to thyroperoxidase (SREAT, Hashimoto’s encephalopathy). J Neurol Sci. 2003;213(1–2):77–82.
J Clin Neurophysiol. 2006;23(4):371–373. 77. Shavit YB, Graus F, Probst A, et al. Epilepsia partialis continua:
56. Vernino S, Geschwind M, Boeve B. Autoimmune encephalo- a new manifestation of anti-Hu-associated paraneoplastic
pathies. Neurologist. 2007;13(3):140–147. encephalomyelitis. Ann Neurol. 1999;45(2):255–258.
57. Ferracci F, Moretto G, Candeago RM, et al. Antithyroid antibodies 78. Thieben MJ, Lennon VA, Boeve BF, et al. Potentially reversible
in the CSF: their role in the pathogenesis of Hashimoto’s autoimmune limbic encephalitis with neuronal potassium channel
encephalopathy. Neurology. 2003;60(4):712–714. antibody. Neurology. 2004;62(7):1177–1182.
58. Nolte KW, Unbehaun A, Sieker H, et al. Hashimoto encephalopa- 79. Espay AJ, Kumar V, Sarpel G. Anti-Hu-associated paraneoplastic
thy: a brainstem vasculitis? Neurology. 2000;54(3):769–770. limbic encephalitis presenting as rapidly progressive non-convul-
59. Mocellin R, Walterfang M, Velakoulis D. Hashimoto’s encephalo- sive status epilepticus. J Neurol. Sci. 2006;246(1–2):149–152.
pathy: epidemiology, pathogenesis and management. CNS Drugs. 80. Mut M, Schiff D, Dalmau J. Paraneoplastic recurrent multifocal
2007;21(10):799–811. encephalitis presenting with epilepsia partialis continua.
60. Gini B, Lovato L, Cianti R, et al. Novel autoantigens recognized by J Neurooncol. 2005;72(1):63–66.
CSF IgG from Hashimoto’s encephalitis revealed by a proteomic 81. Nahab F, Heller A, Laroche SM. Focal cortical resection for com-
approach. J Neuroimmunol. 2008;196(1–2):153–158. plex partial status epilepticus due to a paraneoplastic encephalitis.
61. Striano P, Pagliuca M, Andreone V, et al. Unfavourable outcome of Neurologist. 2008;14(1):56–59.
Hashimoto encephalopathy due to status epilepticus. One autopsy 82. Porta-Etessam J, Ruiz-Morales J, Millan JM, et al. Epilepsia par-
case. J Neurol. 2006;253(2):248–249. tialis continua and frontal features as a debut of anti-Hu paraneo-
62. Schauble B, Castillo PR, Boeve BF, et al. EEG findings in steroid- plastic encephalomyelitis with focal frontal encephalitis. Eur JNeurol.
responsive encephalopathy associated with autoimmune thyroidi- 2001;8(4):359–360.
tis. Clin Neurophysiol. 2003;114(1):32–37. 83. Chapman J, Abu-Katash M, Inzelberg R, et al. Prevalence and clin-
63. Sporis D, Habek M, Mubrin Z, et al. Psychosis and EEG abnormal- ical features of dementia associated with the antiphospholipid
ities as manifestations of Hashimoto encephalopathy. Cogn Behav syndrome and circulating anticoagulants. J Neurol Sci. 2002;
Neurol. 2007;20(2):138–140. 203–204:81–84.
64. Marshall GA, Doyle JJ. Long-term treatment of Hashimoto’s 84. Lampropoulos CE, Koutroumanidis M, Reynolds PP, et al.
encephalopathy. J Neuropsychiatry Clin Neurosci. 2006;18(1):14–20. Electroencephalography in the assessment of neuropsychiatric
65. Tuzun E, Dalmau J. Limbic encephalitis and variants: classifica- manifestations in antiphospholipid syndrome and systemic lupus
tion, diagnosis and treatment. Neurologist. 2007;13(5):261–271. erythematosus. Arthritis Rheum. 2005;52(3):841–846.
66. Lawn ND, Westmorel BF, Kiely MJ, et al. Clinical, magnetic reso- 85. Cimaz R, Romeo A, Scarano A, et al. Prevalence of anti-
nance imaging, and electroencephalographic findings in paraneo- cardiolipin, anti-beta2 glycoprotein I, and anti-prothrombin anti-
plastic limbic encephalitis. Mayo Clinic Proc. 2003;78(11): bodies in young patients with epilepsy. Epilepsia. 2002;43(1):
1363–1368. 52–59.
67. Dirr LY, Elsted AD, Donofrio PD, et al. Evolution of brain MRI 86. Verrot D, San-Marco M, Dravet C, et al. Prevalence and significa-
abnormalities in limbic encephalitis. Neurology. 1990;40(8): tion of antinuclear and anticardiolipin antibodies in patients with
1304–1306. epilepsy. Am J Med. 1997;103(1):33–37.
68. Tan KM, Lennon VA, Klein CJ, et al. Clinical spectrum of voltage- 87. Liimatainen S, Peltola M, Fallah M, et al. The high prevalence of
gated potassium channel autoimmunity. Neurology. 2008;70(20): antiphospholipid antibodies in refractory focal epilepsy is related
1883–1890. to recurrent seizures. Eur J Neurol. 2009;16(1):134–141.
69. Corsellis JA, Goldberg GJ, Norton AR. “Limbic Encephalitis” and 88. Siva A, Altintas A, Saip S. Behcet’s syndrome and the nervous
its association with carcinoma. Brain. 1968;91(3):481–496. system. Curr Opin Neurol. 2004;17(3):347–357.
70. Bakheit AM, KennedyPG, Behan PO. Paraneoplastic limbic 89. Aykutlu E, Baykan B, Serdaroglu P, et al. Epileptic seizures in
encephalitis: clinico-pathological correlations. J Neurol Neurosurg Behcet disease. Epilepsia. 2002;43(8):832–835.
Psychiatry. 1990;53(12):1084–1088. 90. Joseph FG, Scolding NJ. Neuro-Behcet’s disease in caucasians:
71. Dalmau J, Graus F, Rosenblum MK, et al. Anti-Hu--associated a study of 22 patients. Eur J Neurol. 2007;14(2):174–180.
paraneoplastic encephalomyelitis/sensory neuronopathy. A 91. Aktekin B, Dogan EA, Oguz Y, et al. Epilepsia partialis continua in
Clinical Study of 71 Patients. Medicine. 1992;71(2):59–72. a patient with Behcet’s disease. Clin Neurol Neurosurg. 2006;
72. Graus F, Saiz A, Lai M, et al. Neuronal surface antigen antibodies 108(4):392–395.
in limbic encephalitis: clinical-immunologic associations. 92. Chroni E, Monastirli A, Polychronopoulos P, et al. Epileptic
Neurology. 2008;71(12):930–936. seizures as the sole manifestation of neuro-Behcet’s disease:
350 Part III ■ Clinical EEG: General

complete control under interferon-alpha treatment. Seizure. 113. Lang H, Anttila R, Svekus A, et al. EEG findings in juvenile
2008;17(8):744–747. rheumatoid arthritis and other connective tissue diseases in chil-
93. Midorikawa T. Electroencephalographic studies on Behcet’s dren. Acta Paediatr Scand. 1974;63(3):373–380.
disease—with special reference to the electroencephalogram of 114. Sillanpaa M, Lang AH, Kalimo H. Natural history of juvenile
neuro-Behcet’s disease. Rinsho Shinkeigaku. 1975;15(11): rheumatoid arthritis. A follow-up study of a case with special ref-
860–869. erence to clinical, electroencephalographic and neuropathologi-
94. Tunc T, Ortapamuk H, Naldoken S, et al. Subclinical neurological cal findings. Acta Paediatr Scand. 1978;67(4):537–541.
involvement in Behcet’s disease. Neurol India. 2006;54(4):408–411. 115. Delalande S, de Seze J, Fauchais AL, et al. Neurologic manifesta-
95. Lannuzel A, Lamaury I, Charpentier D, et al. Neurological mani- tions in primary Sjogren syndrome: a study of 82 patients.
festations of Behcet’s disease in a Caribbean population: clinical Medicine (Baltimore). 2004;83(5):280–291.
and imaging findings. J Neurol. 2002;249(4):410–418. 116. Glanz BI, Schur PH, Khoshbin S. EEG abnormalities in systemic
96. Al-Araji A, Sharquie K, Al-Rawi Z. Prevalence and patterns of lupus erythematosus. Clin Electroencephalogr. 1998;29(3):128–131.
neurological involvement in Behcet’s disease: a prospective study 117. Gibson T, Myers AR. Nervous system involvement in systemic
from Iraq. J Neurol Neurosurg Psychiatry. 2003;74(5):608–613. lupus erythematosus. Ann Rheum Dis. 1975;35(5):398–406.
97. Akman-Demir G, Baykan-Kurt B, Serdaroglu P, et al. Seven-year 118. Abel T, Gladman DD, Urowitz MB. Neuropsychiatric lupus.
follow-up of neurologic involvement in Behcet syndrome. Arch J Rheumatol. 1980;7(3):325–333.
Neurol. 1996;53(7):691–694. 119. Waterloo K, Omdal R, Jacobsen EA, et al. Cerebral computed
98. Mead S, Kidd D, Good C, et al. Behcet’s syndrome may present tomography and electroencephalography compared with neu-
with partial seizures. J Neurol Neurosurg Psychiatry. 2000;68(3): ropsychological findings in systemic lupus erythematosus.
392–393. J Neurol. 1999;246(8):706–711.
99. Pourmand R, Markand ON, Cook JA. Periodic lateralized EEG 120. Gora MK, Dobrzynska L, Mierzejewska E, et al. EEG examination
abnormality in a case of Neuro-Behcet syndrome. Clin in patients with systemic lupus erythematosus. Wiad Lek. 2003;
Electroencephalogr. 1984;15(2):122–124. 56(5–6):220–226.
100. Kihara M, Takahashi M, Mitsui Y, et al. A case of neuro-Behcet’s 121. Glanz BI, Laoprasert P, Schur PH, et al. Lateralized EEG findings
encephalitis with pleds as distinct from herpes simplex in patients with neuropsychiatric manifestations of systemic
encephalitis: a differential diagnosis. Funct Neurol. 1996;11(2–3): lupus erythematosus. Clin Electroencephalogr. 2001;32(1):14–19.
99–103. 122. Gora M, Dobrzynska L, Koseda-Dragan M. Headache in patients
101. Campanile S, Amore M, Cerisoli M, et al. Neurological complica- with systemic lupus erythematosus: analysis of clinical EEG and
tions in Behcet’s disease. Riv Patol Nerv Ment. 1984;105(5):229–237. neuroimaging studies. Pol Arch Med Wewn. 2003;109(5):
102. Matsumoto K. Correlation between EEG and clinicopathological 497–502.
change in neuro-Behcet’s syndrome. Folia Psychiatr Neurol Jpn. 123. Jennekens FG, Kater L. The central nervous system in systemic
1984;38(1):65–79. lupus erythematosus. Part 1. Clinical syndromes: a literature
103. Calabrese LH, Mallek JA. Primary angiitis of the central nervous investigation. Rheumatology (Oxford). 2002;41(6):605–618.
system. Report of 8 new cases, review of the literature, and proposal 124. Ainiala H, Loukkola J, Peltola J, et al. The prevalence of neuropsy-
for diagnostic criteria. Medicine (Baltimore). 1988;67(1):20–39. chiatric syndromes in systemic lupus erythematosus. Neurology.
104. Vollmer TL, Guarnaccia J, Harrington W, et al. Idiopathic granu- 2001;57(3):496–500.
lomatous angiitis of the central nervous system. Diagnostic chal- 125. Appenzeller S, Cendes F, Costallat LT. Epileptic seizures in sys-
lenges. Arch Neurol. 1993;50(9):925–930. temic lupus erythematosus. Neurology. 2004;63(10):1808–1812.
105. Carandang CG, Grant AL. Delirium and isolated angiitis of the 126. Gonzalez-Duarte A, Cantu-Brito CG, Ruano-Calderon L, et al.
central nervous system: a case report and review. CNS Spectr. Clinical description of seizures in patients with systemic lupus
2008;13(3):209–213. erythematosus. Eur Neurol. 2008;59(6):320–323.
106. Berger JR, Romano J, Menkin M, et al. Benign focal cerebral vas- 127. Tsuji M, Tanaka H, Yamakawa M, et al. A case of systemic lupus
culitis: case report. Neurology. 1995;45(9):1731–1734. erythematosus with complex partial status epilepticus. Epileptic
107. Ilhan A, Budak F. Primary angiitis of the central nervous system: Disord. 2005;7(3):249–251.
unusual clinical presentation. Int J Angiol. 2000;9(1):23–26. 128. Terzano MG, Camillo Manzoni G, Mancia D, et al. Valutazione
108. Gobernado JM, Leiva C, Rabano J, et al. Recovery from rheuma- degli aspetti E.E.G. in rapporto con i parametri clinico-evolutivi
toid cerebral vasculitis. J Neurol Neurosurg Psychiatry. 1984; nella corea reumatica [Evaluation of the EEG aspects of rheu-
47(4):410–413. matic chorea as related to the clinico-evolutive parameters of the
109. Kim RC, Collins G. The neuropathology of rheumatoid disease. disease]. Riv Neurol. 1979;49(6):451–470.
Hum Pathol. 1981;12(1):5–15. 129. Johnson DA, Klass DW, Millichap JG. Electroencephalogram in
110. Neamtu L, Belmont M, Miller DC, et al. Rheumatoid disease of Sydenham’s chorea. Arch Neurol. 1964;10:21–27.
the CNS with meningeal vasculitis presenting with a seizure. 130. Usher SJ, Jasper HH. The etiology of Sydenham’s chorea.
Neurology. 2001;56(6):814–815. Electroencephalographic studies. CMAJ. 1941;44:365–371.
111. Zolcinski M, Bazan-Socha S, Zwolinska G, et al. Central nervous 131. Ganji S, Duncan MC, Frazier E. Sydenham’s chorea: clinical, EEG,
system involvement as a major manifestation of rheumatoid CT scan, and evoked potential studies. Clin Electroencephalogr.
arthritis. Rheumatol Int. 2008;28(3):281–283. 1988;19(3):114–122.
112. Moldofsky H, Lue FA, Smythe HA. Alpha EEG sleep and morn- 132. Lavy S, Lavy R, Brand A. Neurological and electroencephalo-
ing symptoms in rheumatoid arthritis. J Rheumatol. 1983; graphic abnormalities in rheumatic fever. Acta Neurol Scand.
10(3):373–379. 1964;40:76–88.
CHAPTER

Cerebrovascular Diseases and EEG


BARBARA TETTENBORN, ERNST NIEDERMEYER, AND DONALD L. SCHOMER 19
ROLE OF EEG IN DIAGNOSIS OF frequencies. Considerable localizing value has been ascribed to
CEREBROVASCULAR DISEASE local depression of the background activity, whereas more exten-
sive depression is regarded as somewhat less informative (7–9).
Computed tomography (CT) and magnetic resonance imaging These authors believe that preservation of faster background
(MRI) are the major diagnostic tools in cerebrovascular disease activities, especially in subacute forms of middle cerebral artery
considering the morphology is a vascular lesion. The major role (MCA) ischemia, is indicative of considerable neuronal survival
of electroencephalography (EEG) is the evaluation of functional in the infarcted zone and hence indicative of a good prognosis.
disturbances caused by cerebrovascular disease, mainly the detec- Recent studies by Hilker et al. (10) could also indicate the absence
tion of epileptogenic foci. Cerebrovascular events are acute, often of delta activity and presence of theta and fast beta frequencies
catastrophic events that naturally have strong influences on the within the EEG focus in stroke patients as predictor of a benign
EEG. The tempo of evolution is an important factor. Acute vas- course, whereas diffuse slowing and focal slow delta frequency
cular slow foci may be much more impressive than focal slowing may point toward a more malignant course.
caused by a slowly growing neoplasm. On the other hand, small Even considering new MRI techniques, including functional
deep vascular lesions are often too far distant from the cortex and MRI, EEG still has its place in diagnostic workup of stroke
may escape EEG detection, whereas deep vascular lesions in the patients. Not only is MRI more expensive, but it also can be some-
brainstem and cerebellum reveal themselves only as secondary what difficult to perform in the acutely ill stroke patient, whereas
phenomena in the cerebral hemispheres. In an acute stroke, a EEG can easily be done and repeated in practically every patient.
massive and highly impressive EEG focus may be present before
a CT scan can demonstrate the lesion (1). MRI has been found to HYPERVENTILATION
be more precise and more informative than CT in the morpho-
logic evaluation of stroke. The EEG is an indicator of abnormal Hyperventilation as a routine procedure in clinical EEG can
physiologic function; the function of the involved central nerv- activate otherwise latent EEG abnormalities in patients with
ous system tissue breaks down before the structure shows its suf- cerebrovascular disorders. It is probably the hypoxic effect of
fering. It has been shown experimentally (2) as well as clinically hyperventilation that produces these changes. This activation
with the use of CT scan (3) that edema alone does not account can give rise to focal EEG slowing in cases of moyamoya disease
for delta activity in EEG. In the modern view, the factor of (11). Kraaier et al. (12) investigated hyperventilation-induced
ischemic tolerance of the brain has been critically analyzed (4) changes with quantitative EEG and considered this method a
along with the demonstration of a large number of biomolecular model for transient ischemia in normal subjects.
substances presumed to be involved in this complex process.
The role of EEG as an indicator of disturbed function, locally RELEVANCE OF ACTIVATION
as well as diffusely, in the event of a stroke has not always been METHODS TO THE ASSESSMENT
duly appreciated. Decades ago, Hachinski and Norris (5) pointed OF CEREBROVASCULAR DISORDERS
out in their study on acute stroke that “electroencephalography is
not much help in the diagnosis of stroke, especially when CT At present, the role of these tests is rather limited. The fact
scanning is available.” But apposed to that, even in our days, remains, however, that these tests have given new insights into
including MRI techniques, it has to be stated that the evaluation hemodynamic and neurophysiologic mechanisms underlying
of the degree and extension of cerebral dysfunction may be just the EEG activity. An interesting overview of the cerebral hemo-
as important as the demonstration of local or diffuse morpho- dynamic evaluation has been presented by Carney et al. (13)
logic changes. An experienced electroencephalographer might be with the EEG aspects of this work being aptly described by
able to provide valuable information about the regional and gen- Anderson et al. (14).
eral functional or dysfunctional state in a stroke. An imaginative
approach to the EEG evaluation of strokes has been proposed by ACUTE STROKE DETECTION
Velho-Groneberg (6). Accordingly, the EEG reflects dynamic
processes that occur in the wake of a stroke. In the emergency evaluation of acute stroke, the progression
In addition to local or more diffuse slow activity, the EEG from transient ischemia to infarction is a dynamic process, which
also shows a more or less serious decline of physiologic patterns is difficult to evaluate based solely on clinical grounds and
such as posterior alpha rhythm and intermixed slower or faster anatomic imaging. Advances in CT and MRI scanning, including

351
352 Part III ■ Clinical EEG: General

diffusion-weighted MRI (DWI), are providing new insights into thrombotic, embolic, or hemorrhagic. Important pathogenic
the pathophysiology of cerebral ischemia. Perfusion measures, mechanisms include cerebral edema and the syndrome of inap-
such as perfusion-weighted imaging, may be able to estimate propriate antidiuretic hormone, which leads to reduction of cere-
infarcted tissue with information about blood flow levels, but bral sodium, potassium, and chloride content, altered brain
these measures may be less sensitive to dynamic changes in tissue energy metabolism, and neurotransmitter amino acid uptake (5).
viability than the EEG. Within seconds, the EEG reflects changes Cardiac factors, poststroke depression, and drug effects also may
in cerebral blood flow and metabolism. Both human and animal lead to poststroke deterioration. In evolving and deteriorating
evidence indicates that the ischemic cascade is rapid, with strokes, the ischemic penumbra plays a major role (19). In dete-
marked changes occurring over seconds and minutes, and irre- riorating strokes, the EEG slowing tends to become more diffuse.
versible changes occurring within a few hours. DWI and EEG It also may reflect drug effects (sedatives), but it is apt to remain
detect pathophysiologic responses to reduced blood flow around notably unaltered in the case of poststroke depression.
the same range, between 30 and 40 mL/100 g/min (15), suggest-
ing that they are comparable in their sensitivity to the stage of
ischemic gradient. The accuracy of EEG scalp recordings, how- EEG IN HEMISPHERIC STROKE
ever, may be limited by adequate sampling of the electrical field. The most common cause of a hemispheric ischemic lesion is
Biophysical simulations have indicated for many years that the atherosclerotic disease of the internal carotid artery or MCA.
spatial frequency of the scalp EEG requires higher sampling Size of infarct and clinical symptoms relate to localization and to
densities to avoid artificial smoothing of the scalp potential collateral circulation. An infarct may encompass the entire terri-
field caused by undersampling (16,17). Luu et al. (18), in their tory supplied by carotid artery, followed in a few days by severe
study on acute stroke patients, found within 8 to 36 hours after edema, raised intracranial pressure, transtentorial herniation,
symptom onset using a 128-channel sensor montage, that an and even death. At the other extreme, occlusion of carotid artery
accurate description of stroke-related topographic EEG changes may occur without obvious clinical deficits, because of adequate
is dependent on adequate spatial sampling density. Accurate collateral circulation. In acute stages, EEG is usually the most
description of the spatial distribution of the stroke-related EEG sensitive laboratory index. Massive hemispheric infarct due to
was achieved only with the 64- and 128-channel EEG. As the carotid occlusion causes marked EEG abnormalities (20–22):
recording density decreases to 32 channels, the distribution of the
scalp EEG spectra is distorted, potentially resulting in mislocal- • Widespread arrhythmic delta activity occurs over the
ization of the affected region. Their results demonstrated that the involved hemisphere, most prominently in temporal or fron-
EEG acquired with the conventional 19-channel montage is clin- totemporal regions (Fig. 19.1). In patients with smaller hemi-
ically useful, if the data are reviewed by an expert, but localizing spheric infarcts, EEG from uninvolved hemisphere usually
information is limited by the available data. Nevertheless, for remains normal.
daily clinical routine 128-channel EEG recording for every stroke • Ipsilateral alpha rhythm may be absent or of lower amplitude
patient is not realistic. Therefore, studies on the value of conven- and slower frequency. Reactivity of alpha rhythm may dimin-
tional 19-channel montage using the international 10-20 system ish or disappear (Fig. 19.2). Rolandic beta activity and sleep
are currently being done. Conventional EEG recordings could be spindles may also disappear.
useful in predicting the subgroup of acute stroke patients who are • Extensive infarcts may markedly decrease or “suppress” elec-
predisposed to develop epileptic seizures in the follow-up. trocerebral activity, normal and abnormal, over the affected
In patients with large MCA infarction, space-occupying brain hemisphere during the first few days. In patients with smaller
edema may lead to a malignant course with up to 80% mortal- hemispheric infarcts, EEG from uninvolved hemisphere usu-
ity using conservative treatment. As interventional treatment ally remains normal. With extensive infarcts causing cerebral
strategies must be started before the deterioration occurs, pre- edema and displacement of midline structures, variable
dictors of a malignant course are necessary. Hilker et al. (10) degrees of slow activity appear in contralateral hemicranium,
investigated the value of EEG within 24 hours after the onset of often with bilateral intermittent rhythmic 2 to 3 per second
stroke in 25 patients suffering a large MCA infarction. Their delta activity (IRDA) (Fig. 19.3), which may be marked in the
findings indicated that the absence of delta activity and the pres- frontal region (FIRDA, frontal intermittent rhythmic delta
ence of theta and fast beta frequencies within the focus predict a activity) (Fig. 19.4). In children, this rhythmic delta activity
benign course, whereas diffuse generalized slowing and slow may be most marked occipitally (OIRDA, occipital intermit-
delta activity in the ischemic hemisphere may point toward a tent rhythmic delta activity). Marked diffuse EEG changes
malignant course. Therefore, EEG can deliver useful informa- may make lateralization difficult; also, massive infarcts caus-
tion to select those patients who develop malignant edema. ing ipsilateral voltage depression and prominent contralateral
slow activity can lead to erroneous lateralization.
• With acute hemispheric infarctions, particularly embolic,
DETERIORATING STROKES
epileptiform activity occurs commonly and tends to be peri-
Various complications may interfere with recovery from a stroke. odic (periodic lateralized epileptiform discharges [PLEDs])
The term “deteriorating stroke” has been used by Hachinski and (Fig. 19.5). Such patients are usually obtunded and have focal
Norris (5) and includes progressively evolving strokes, recurrent seizures with corresponding neurologic deficits. PLEDs and
strokes, and also spatially extending strokes. Their causes may be clinical manifestations usually resolve in 1 to 2 weeks.
Chapter 19 ■ Cerebrovascular Diseases and EEG 353

50 l V

50 l V

Figure 19.2 Recent acute cerebrovascular ischemia (3 days earlier)


Figure 19.1 Age 57 years. Acute cerebrovascular event on the day due to right middle cerebral artery thrombosis and good CT scan evi-
before this record was obtained. There is marked left frontotemporal dence of infarction in the corresponding territory. Acute left hemiplegia.
polymorphic delta activity. Also note alpha depression and loss of detail Age 64 years. Patient awake; right-sided alpha diminished and a large
over left posterior quadrant. The right hemisphere and especially the zone of mixed 3 to 6 per second activity involving most of the right
right frontal area show some degree of delta activity. hemisphere.

During the first days after infarction, EEG abnormalities are abnormalities; in general, their findings paralleled the findings of
most prominent, with extent and voltage of slow activity progres- Kayser-Gatchalian and Neuendörfer (20). Although unusual in
sively declining. Few prospective studies have evaluated EEGs seri- the acute phase of the infarction, epileptiform discharges can
ally in a carefully defined population. Kayser-Gatchalian and evolve with time, possibly heralding postinfarction seizures.
Neuendörfer (20) did EEGs on 79 patients with infarcts in carotid In patients with large MCA infarction, space-occupying brain
distribution. Initial EEGs were done within 48 hours after infarc- edema may lead to a malignant course with up to 80% mortal-
tion and again on days 5, 10, and 22. They visually classified EEG ity under conservative treatment. As interventional treatment
abnormalities into “background slowing” and “focal changes,” strategies must be started before the deterioration occurs, pre-
with each group being divided by defined criteria into the follow- dictors of a malignant course are necessary. Burghaus et al. (24)
ing categories: “none,” “mild,” “moderate,” or “severe.” In the initial reported the results of a study on early EEG within 24 hours
EEGs, “background slowing” reliably predicted state of conscious- after the onset of stroke in 25 patients suffering a large MCA
ness: only 1 of 14 patients with severe or moderate slowing was infarct (12 patients with a malignant and 13 with a nonmalig-
alert, whereas 33 of 44 patients without slowing were alert. Focal nant course). EEG analysis was performed according to well-
changes paralleled motor deficits: 39 of 51 patients with moderate established indicators for focal as well as global changes. Their
or severe focal changes had severe paresis or hemiplegia, whereas results indicated that the absence of delta activity and the pres-
only 7 of 17 patients without focal changes had severe weakness. ence of theta and fast beta frequencies within the focus predict a
Background slowing in the initial EEG predicted outcome on day benign course (P 0.05), whereas diffuse generalized slowing
22: only 2 of 14 patients with moderate or severe slowing and slow delta activity in the ischemic hemisphere may point to
improved, whereas 38 of 44 patients without slowing improved. a malignant course.
Persistent focal changes paralleled persistent severe hemiparesis or Infarction due to occlusion of the anterior cerebral artery is
hemiplegia. In a smaller series of 15 patients, Sainio et al. (23) rare. EEG findings consist of (i) ipsilateral frontal delta activity
compared visual with spectral analysis. Results of visual analysis (either arrhythmic or rhythmic, or both) and (ii) depression of
equaled spectral analysis, except for a lower incidence of focal anterior beta activity.
354 Part III ■ Clinical EEG: General

Figure 19.3 A 79-year-old male patient presented with acute onset of aphasia, right-sided hemiparesis, and gait instability.
CT scan 3 hours after onset of symptoms without evidence of acute infarction. EEG showed posterior 9 per second alpha
rhythm on the right, suppressed on the left. Generalized intermittent temporoparietal polymorphic theta/ delta slowing (IRDA).

Occlusion of the posterior cerebral artery may result in (i) setting of multifocal or diffuse cerebral injury, such as anoxia,
infarction of the mesial temporal lobe with arrhythmic delta and herald a less favorable prognosis with higher mortality.
activity over the posterior head region and (ii) marked disor- Approximately 80% to 90% of patients with PLEDs experience
ganization or absence of alpha rhythm. clinical seizure activity, primarily focal motor seizures. In 1991,
Reiher et al. (28) described the entity “PLEDs plus,” character-
PLEDS: CLINICAL CORRELATES ized by PLEDs admixed with high-frequency, low-voltage
“polyspike” rhythms. These have an even stronger correlation
PLEDs are a relatively uncommon electroencephalographic with clinical seizures and status epilepticus. PLEDs are generally
pattern characterized by lateralized or focal periodic or near- not considered an ictal pattern, though this has been reported,
periodic spike, spike-wave, or sharp wave complexes present and remains a matter of ongoing debate (29–31).
throughout most or all of the recording (Fig. 19.5B). Chatrian
et al. introduced the term PLEDs in 1964 (25), though the phe- EEG IN VERTEBROBASILAR ISCHEMIC
nomenon was first described in 1952 by Echlin et al. (26). PLEDs DISEASE
occur in all age groups, from infants to adults. They are usually
seen transiently in the setting of an acute destructive cerebral Ischemia in the vertebrobasilar territory includes infarction of
lesion and occur early in the course of patient illness. They can brainstem, diencephalon, or cerebellum with varying neurologic
be seen less commonly with systemic disturbances and a remote deficits depending on specific artery involved and extent and
cerebral lesion. Rare chronic PLEDs, persisting for a period of location of the infarct. Disparity between extensive neurologic
3 months to more than 20 years, have been reported in patients deficits and EEG changes characterize brainstem and also most
with chronic brain lesions and associated partial seizure disor- cerebellar infarcts (Figs. 19.6 and 19.7). Less than one third of
ders. Bilateral, independently occurring PLEDs (BiPLEDs) were patients with limited infarcts to the brainstem show even slightly
recognized by Chatrian in 1964 (25) and formally characterized altered EEGs (32). Basilar artery occlusion infarcting ventral pons
in 1981 by de la Paz and Brenner (27). They are seen in the but sparing tegmentum causes a distinctive clinical picture
Chapter 19 ■ Cerebrovascular Diseases and EEG 355

Figure 19.4 A 76-year-old female patient presented with fluctuating aphasia, hemianopia, and right hemiataxia.
Symptoms lasted only a few hours. MRI revealed no acute infarction but signs of a chronic microangiopathic encephalopa-
thy with multiple subcortical infarctions. EEG showed intermittent theta slowing in the left temporal region and intermit-
tent frontal rhythmic delta activity (FIRDA).
Figure 19.5 A,B: A 71-year-old female with acute onset left-sided hemiparesis. MRI revealed paraventricular right-sided
ischemia. EEG showed regional slowing on the right side, predominantly temporal. Here also periodic lateralized epilepti-
form discharges (PLEDS) with maximum over T4 are noticed.
Chapter 19 ■ Cerebrovascular Diseases and EEG 357

Figure 19.6 A 59-year-old male with sudden onset of vertigo, headache, and general weakness. MRI revealed extended
cerebellar infarction. Etiology was cardioembolic with known atrial fibrillation under insufficient anticoagulation. EEG is
inconspicuous with normal posterior alpha activity.
358 Part III ■ Clinical EEG: General

Figure 19.7 A 69-year-old male with sudden onset of nausea, dizziness, and weakness of right arm. MRI shows infarc-
tion in the territory of the superior cerebellar artery. EEG is normal.
Chapter 19 ■ Cerebrovascular Diseases and EEG 359

known as “deefferented” or “locked-in” syndrome (33). Patients of the brain has confirmed that patients with leukoaraiosis and
are alert but are mute and tetraplegic, with the only preserved seizures have a more severe decline of regional cerebral blood
motor acts being blinks and vertical eye movements. In these flow and metabolic rate of oxygen in the whole cortex compared
patients, EEG is normal (34,35); the alpha rhythm reacts to vari- to those without seizures (43) and to those with late-onset symp-
ous stimuli, including intermittent photic stimulation that pro- tomatic seizures (44). Patients with extended chronic subcortical
duces photic responses. Patients with bilateral pontine infarcts infarctions can show generalized intermittent delta activity on
involving tegmentum show striking dissociation between clinical EEG recording (Fig. 19.8).
and EEG findings (25,36). Despite coma and extensive bilateral
motor deficits, EEGs may show activity of alpha frequency. EEG IN CEREBRAL ISCHEMIC
However, this alpha activity fails to react to various stimuli such
DISEASE DUE TO EMBOLISM
as intermittent photic stimulation; hence, it is abnormal. Because
of the association of coma and alpha activity, the term “alpha All forms of cerebral emboli can cause a variety of clinical and
coma” has been applied to such patients (37). A similar associa- EEG changes. Already decades ago, Lhermitte and his coworkers
tion also occurs in patients rendered comatose by cardiopul- demonstrated that a surprisingly large number of strokes
monary arrest (38) or drug overdose (39). believed to be thrombotic are due to emboli of cardiac origin
At more rostral level, infarcts of midbrain and diencephalon (45–47). Because of new imaging techniques and more insight
cause not only coma but also EEG alterations. Bilaterally syn- into pathophysiology of cerebral ischemia, it is well known that
chronous and symmetrical theta–delta activity, sometimes maxi- about 40% of all cerebral ischemic events are embolic in origin.
mal over frontal regions, replaces normal rhythms. Nevertheless, Emboli can either arise from thrombotic material within the
these relatively mild changes, compared with the depth of coma larger brain supplying arteries (common carotid or internal
and extent of neurologic deficits, should alert EEGers to an carotid arteries, vertebral arteries) or they can be cardiac in ori-
intrinsic brainstem lesion. Thrombosis of rostral portion of basi- gin. Aortic arch plaque material may also cause cerebral ischemia.
lar artery can also cause infarcts of occipital lobes. EEGs show Embolism of thrombotic material and gas and fat emboli may
posterior arrhythmic delta activity with loss of alpha rhythm, give rise to clinically and electroencephalographically similar pic-
unilaterally or bilaterally. tures (Fig. 19.9A,B). In embolism, however, the extremely acute
Jouvet (40) has correlated sites of brainstem lesions with shock-like effect may trigger a chain of vasomotor responses in
alterations in consciousness and changes in EEG activity. He adjacent vascular territories. These responses either hasten the
has concluded the following: catastrophic events or resolve quickly so that recovery may evolve
• Lesions of medulla oblongata and caudal pons alter usually with unexpected speed.
neither consciousness nor EEG. Rasheva (48) and Rasheva et al. (49) noted clinical epileptic
• Extensive lesions of upper pons and lower mesencephalon seizures in 10.1% and paroxysmal EEG discharges (spikes and
involving more than one half of tegmentum usually cause sharp waves) in 57.1% of a large population of 328 patients
coma. EEGs may show alpha activity that does not react to with embolic stroke.
external stimuli. Although vital in maintaining conscious- The importance of cerebral gas embolism during and after
ness, mesopontine regions seemingly contribute little to gen- cardiac surgery is widely known. According to Naquet and
esis of EEG activity. Vigouroux (50), gas emboli may produce EEG changes, similar
• Lesions of rostral brainstem and caudal diencephalon cause to those seen in watershed infarction, to be discussed in the fol-
not only deep coma but also diffuse, nonreactive theta and lowing section. In fat embolism, Müller and Klingler (51)
delta activity. reported marked delta–theta activity of frontotemporal accen-
tuation. Similar findings were reported by Scherzer (52).
In bilateral paramedian thalamic infarction, triphasic waves Experimental cerebral embolism lends itself to the study of
can appear on electroencephalogram at acute stage (41). immediate EEG effects. These are dramatic and instantaneous.
Triphasic waves are nonspecific EEG findings occurring in both In the rabbit, Hegedüs et al. (53) demonstrated abrupt flatten-
metabolic and nonmetabolic conditions. The triphasic waves in ing of the affected hemisphere after embolization of the MCA;
bilateral paramedian thalamic infarction might be related to there was even some voltage decrease contralaterally (Fig. 19.3).
level of consciousness and not necessarily predictive of poor This clearly shows that the effects of a major embolism extend
prognosis. beyond the involved arterial territory. But in general, the EEG
cannot effectively assist in the differentiation of thrombotic and
EEG IN SUBCORTICAL ARTERIOSCLEROTIC embolic cerebral ischemia.
ENCEPHALOPATHY
WATERSHED ISCHEMIA
The incidence of epileptic seizures in subcortical vascular
encephalopathy is more related to a higher rate of lacunae within Ischemic states usually pertain to the territory of the involved
the subcortical white matter rather than in the basal ganglia and artery, but another mechanism has become clinically impor-
brainstem. The higher incidence of epilepsy in this group can be tant. This is the concept of “extraterritorial ischemia” that gives
explained by diaschisis resulting in suppression of the inhibitory rise to “watershed infarctions” (“boundary infarctions”) along
cortical activity (42). Positron emission tomographic examination the borders of the major arterial territories, between middle,
360 Part III ■ Clinical EEG: General

Figure 19.8 A 79-year-old female patient presented with a first primary focal, secondary generalized epileptic seizure to
the emergency unit. MRI revealed extended chronic subcortical infarctions without evidence of an acute infarction. EEG
showed marked left polymorphic delta activity and generalized intermittent rhythmic delta activity (IRDA).

posterior, and anterior cerebral arteries. This mechanism is with consistent lateralization to the affected hemisphere. Against
based on a decline in the systemic circulation. This is a drop in this slow and disorganized background of rather moderate volt-
blood pressure (BP) or cardiac output, usually combined with age, there are consistently repetitive large sharp waves or spikes,
a diseased and plaque-ridden cerebral vasculature. It is a logical often compounded and consisting of several subcomponents
and convincing concept when it is assumed that boundary areas and phases, usually over the posterotemporal–occipital–parietal
between the major cerebral arterial territories are most likely to region of the hemisphere that harbors the lesion. They are called
suffer from ischemia. The work of Zülch and Behrend (54) PLEDS. This periodic spike discharge is rhythmically or quasi-
clearly demonstrates the distinction between the watershed rhythmically repetitive at a rate of approximately 1 per second.
type and the classical obstructive ischemic infarction. The discharge may become quite widespread, even reaching into
Acute watershed-type ischemia is most commonly found in the other hemisphere. Occasionally, the periodic spikes show a
elderly patients with a history of cerebral arteriosclerosis and superior frontal maximum.
chronic cardiovascular problems. A variety of illnesses may trig- At this stage of repetitive spike activity, contralateral focal
ger the acute cerebral ischemia. Acute infection, dehydration, motor twitching (fingers or hand, face, abdomen, leg, foot, or
uncontrolled diabetes mellitus, cardiovascular decompensation, toes) is the common accompaniment of the periodic spikes, but
severe anemia, recent craniocerebral trauma, extracerebral such PLEDs often occur without accompanying motor activity.
embolism, liver disease with portocaval encephalopathy, and Further work on these discharges was presented by Alajouanine
severe arterial hypertension are events that can facilitate the et al. (55), Barolin et al. (56), Chatrian et al. (25), Markand and
occurrence of watershed ischemia (50). Clinically, a contralateral Daly (57), and Pohlmann-Eden et al. (29,30). These latter authors
hemiparetic deficit is usually present, but it may not be as pro- feel that PLEDs indicate focal hyperexcitability in the penumbra
nounced as in primary capsular ischemia or hemorrhage. zone of an ischemic stroke. Multifocal epileptic seizures of partial
The EEG is slow with a mixed delta–theta range frequency character may also evolve in watershed ischemia, along with
and severely disorganized. These changes are essentially diffuse independent EEG foci (58).
Figure 19.9 A 22-year-old male. Skiing accident with closed tibial and fibular fracture. Twenty-four hours later, the patient
complained about total loss of vision. MRI showed multifocal tiny high intensity areas on diffusion-weighted images in con-
cordance with the diagnosis of fat embolism. A: EEG shows generalized delta activity. B: 4 days later, a posterior alpha rhythm
in the 8 to 10 per second range has been restored; 3 to 6 per second theta–delta activity centro-parieto-temporo-occipital, more
marked on the right side.
362 Part III ■ Clinical EEG: General

EEG IN TRANSIENT ISCHEMIC ATTACKS always be checked whether the patient is able to recall the episode
of focal disturbance completely, as more than two thirds of
Transient ischemic attacks (TIAs) are defined as a temporary, patients with an inhibitory seizure will have difficulties to
clinically completely reversible event of focal, nonconvulsive, describe their temporary deficit clearly.
neurologic dysfunction secondary to a failure of sufficient Early EEG should always be performed in patients with a
blood supply to a distinct area of the brain, retina, or other transient episode of focal neurologic disturbance, as abnormal-
nervous tissue. Most TIAs last less than 15 minutes, but the old ities can indicate the possibility of a seizure, whereas in TIA a
purely clinical standard definition of TIA was that symptoms normal EEG should be expected (63–66).
and signs have to be resolved within 24 hours (59,60).
According to this definition DWI demonstrated acute perma-
nent ischemic lesions in approximately 30% of TIA patients, EEG IN INTRACRANIAL VENOUS
climbing to 50% when DWI is combined with perfusion- THROMBOSIS
weighted imaging (61). A new proposed definition by the TIA
In the era of antibiotic therapy, episodes of infectious intracra-
Working Group is that TIA is a brief episode of neurologic dys-
nial venous thrombosis and thrombophlebitis have become
function caused by focal brain or retinal ischemia with clinical
rare. Complicated ear infections (transverse sinus) or facial car-
symptoms typically lasting less than 1 hour, most often some
buncles (cavernous sinus) occasionally cause such a progression
minutes, without evidence of acute infarction on modern neu-
of infectious material into the venous system of the cranial cav-
roimaging (61,62).
ity. Complications of abortion are another cause; widespread
In some patients, TIA presents with the symptomatology
cortical venous thrombosis may develop intrapartum. Reviews
of limb shaking with brief, arrhythmic flailing or jerking
of the field have been presented by Garcin and Pestel (67),
movements of the arm or leg, which can be easily misdiag-
Dubois (68), Huhn (69,70), and Carter (71).
nosed as a seizure disorder. But early diagnosis of a cerebrovas-
EEG changes in cortical venous thrombosis intrapartum can
cular disorder is important for immediate initiation of
be very pronounced, with widespread prominent delta activity;
appropriate secondary stroke prevention including medical
these patients are not necessarily in a state of impaired con-
and interventional or surgical procedures. EEG testing is often
sciousness but show marked changes of higher cortical func-
ordered in a patient with limb shaking TIA because of suspi-
tions, such as aphasia, mutism, or apraxia, and the course of
cion of seizures. Taghavy and Hamer (60) reviewed the records
ensuing recovery may be very slow.
of patients with TIAs over a period of 6 years. In 79 patients
Literature on the EEG in cerebrovenous pathology is scanty.
with clinical TIA, an EEG was performed, in 44.3% demon-
The work of Huhn (69), Lemmi and Little (72), Franco (73),
strating focal abnormal electrical activity after recovery from
and Hubach and Struck (74) has been particularly informative.
TIA corresponding to the affected hemisphere. In only 17.7%
Van der Drift and Kok (9) demonstrated the underlying patho-
of these patients, there was also an appropriate hypodensity in
physiologic mechanisms as well as the correlations of EEG and
cranial CT scan. Failure to consider cerebral hypoperfusion as
venous pathology. Focal slowing and ictal epileptic activity have
a potential cause of focal delta EEG slowing in the presence of
been reported (75).
normal structural imaging studies may contribute to delay in
correct diagnosis.
The above mentioned new TIA definition makes it even ROLE OF EEG IN PROGNOSIS
more difficult to distinguish TIAs from seizures, especially from OF EPILEPTIC SEIZURES AFTER
inhibitory seizures in elderly patients (63). Recognition of ISCHEMIC STROKE
modern diagnostic clues can prompt expedited magnetic reso-
nance angiography (MRA), carotid duplex ultrasound, or The incidence rates for epilepsies have two peaks regarding age:
computed tomography angiography (CTA) studies of the neck one in the first decade of life and a second at higher age with a
and intracranial blood vessels to identify arterial stenoses or steep increase between age 70 and 80 (76). Cerebral ischemia is
occlusions (64). the single most common cause of first seizures and epilepsy in
De Reuck and Van Maele (63) performed a study on TIAs and later life above the age of 60 years, ahead of degenerative disor-
inhibitory seizures in elderly patients and included 25 patients ders, brain tumors and head trauma, accounting for up to one
with inhibitory seizures and 252 patients with TIA over a 7-year third of newly diagnosed seizures in this population. The lead-
study period taking into account the old and new TIA definition. ing type of stroke-related seizures is focal with a high rate of
The clinical characteristics and the EEG findings were compared. secondary generalization (77–86).
Temporary speech disturbance, associated with some partial The frequency of seizures after stroke varies from 5% to
amnesia for the event, was the most common clinical presenta- approximately 40% (81,84–91), but only a minority will develop
tion of an inhibitory seizure. Additionally, specific and nonspe- epilepsy. A number of risk factors such as cortical involvement,
cific postictal EEG abnormalities were observed in the majority infarct size, and stroke severity have been identified as predictors
of inhibitory seizure patients, whereas the EEG was normal in of seizure development (92). Large cortical infarctions in the
more than 90% of TIA patients. Diffuse slowing and intermittent anterior circulation have a high risk of developing seizures (93).
rhythmic delta activities were only observed in patients with Status epilepticus can be a presenting symptom of acute stroke
seizures, not in TIA patients. They concluded that it should and can lead to increased mortality.
Chapter 19 ■ Cerebrovascular Diseases and EEG 363

Based on differences in their presumed pathophysiology, definition of early- and late-onset seizures according to the
seizures after stroke are usually divided into early- and late-onset guidelines of the “International League against Epilepsy” is often
seizures, in a paradigm comparable to posttraumatic epilepsy. neglected. Infarct etiology is not taken into account in most of
The division in early- and late-onset postischemic seizures is the studies because of the lack of duplex sonography and
important because the expected good prognosis of early seizures echocardiography. The quality of neuroimaging techniques with
(77,84,94–96) could not be approved by other authors CT (used in most of the older studies) is not powerful enough to
(85,97,98). Usually, early seizures are defined as those occur- detect clearly infarct morphology. Some studies describing the
ring within 7 days of the stroke; late-onset seizures are usually predictive value of the EEG have been published in the past. As
defined as occurring after day 7 within the first year after the an example, Carrera et al. (99) used a continuous EEG detection
stroke. Results of recent studies revealed early seizures as inde- to describe the incidence of epileptic electric activity, which has
pendent risk factor for late seizures and development of epilepsy no predictive power regarding the risk for poststroke seizures.
(78,99,100). This method is not practicable in the acute phase of stroke. Some
There are no clear guidelines for the treatment of seizures in authors point out that the occurrence of PLEDs has a predictive
stroke and hence the treatment needs to be initiated in the con- value regarding development of poststroke seizures, but this is
text of the individual patient. The presence of comorbid condi- discussed controversially (29,30,81,88).
tions and the use of other drugs also complicate antiepileptic
therapy, and the risk of drug interactions is a particular hazard
in elderly patients on multiple comedications. The optimal tim- EEG AND BRAIN MAPPING TECHNIQUES
ing and type of anticonvulsive treatment remain debated (91). FOR EVALUATING POSTSTROKE RECOVERY
Several findings suggest that the majority of first-generation AND REHABILITATION
anticonvulsive drugs are not the best choice in stroke patients.
Despite the importance of the problem, there are few data on Brain mapping techniques have proven to be vital in under-
the natural history of stroke-related seizures and no good guide- standing the molecular, cellular, and functional mechanisms of
posts to suggest when to initiate anticonvulsant therapy after recovery after stroke (101). Magnetoencephalography (MEG)
stroke. The exact statistical risk of further seizures after a first and EEG techniques work in a completely different manner than
poststroke seizure is not known; therefore, it has to be a case- the image of blood flow–based brain mapping methods. MEG
by-case decision when to start medication. After a single early and EEG detect signals that arise predominantly from the den-
seizure, long-term anticonvulsant therapy is usually not recom- dritic fields of cortical pyramidal neurons. They detect these sig-
mended even though recent studies could not reproduce the pre- nals from a large area on the order of several square centimeters.
viously thought good prognosis of early seizures. There is also With EEG, the tissue between the scalp electrode and the cortex
still debate about treatment after a first late seizure. After a sec- attenuates the strength of the signal and the signal strength
ond seizure long-term anticonvulsant therapy is usually recom- decreases with the square of the distance between them. Because
mended. tissues do not affect the MEG signal, MEG typically enables
Poststroke seizures are in most patients well controlled with better signal source localization than EEG, but the magnetic
a single anticonvulsant drug. The choice of drug is given by the fluctuations drop off as the cube of the distance between the
general recommendations of anticonvulsant medication in source and the detector increases. EEG and MEG techniques
patients with focal seizures. In these mainly elderly patients, may use an event-based approach to identify the characteristics
interactions with other medications and cognitive side effects of brain activity in response to specific behaviors. Strens et al.
have to be considered especially. Whether the new anticonvul- (102) found that the degree of signal synchrony was greater
sants have advantages as compared with the standard medica- between medial and lateral motor areas in stroke patients than
tion with carbamazepine, oxcarbazepine, or valproic acid is still in healthy subjects but reduced with recovery. In another study,
open and under investigation. the degree of reduction in delta-wave signals from perilesional
The presence of seizures after stroke constitutes a challenge for brain tissue correlated with the amount of language recovery
patients, families, and physicians. Limited information is available following rehabilitation (103). The drawback of the EEG
regarding the risk factors of developing poststroke seizures and method clearly is the lack of anatomic information.
the impact of seizures on the clinical outcome including mortal- A number of recent studies report that quantitative elec-
ity, disability, quality of life, and cognition. No guidelines exist for troencephalography (QEEG) measures of cerebral pathophysi-
the treatment of poststroke seizures, particularly the point of time ology in acute or subacute stroke might augment future
at which treatment should be started, and it is not clear whether prognoses regarding patient outcomes (104–107). Finnigan et
treatment influences course of illness and/or long-term prognosis al. (105) reported that QEEG delta power measures in acute
of stroke patients, quality of life, and development of mood stroke and delta/alpha power ratio measures in subacute stroke
changes. New therapeutic approaches and guidelines can only be are both highly correlated with ischemic stroke patients’ out-
developed when the mechanisms and risk factors of poststroke comes assessed via the National Institute of Health Stroke Scale
seizures are better described and understood. (NIHSS). QEEG measures were averaged over all scalp elec-
Several, mostly retrospective, studies dealing with this topic trodes and further a significant correlation between subacute
have methodologic drawbacks. Most of them have small case delta/alpha power ratio and outcome measures was obtained
numbers and mix up ischemic and hemorrhagic strokes. The when the former was computed from a standard, 19-channel
364 Part III ■ Clinical EEG: General

array. Moreover, multinomial logistic regression analyses disease (118,119). Eder et al. (113) introduced a paradigm for
revealed that this QEEG per se enhanced the sensitivity and detecting and mapping PMBS for loosely defined movements
specificity of outcome prediction beyond that afforded by sub- of the upper extremities on a drawing board, which was tested
acute lesion volume defined via perfusion-weighted MRI. These in eight acute stroke patients with mild hemiparesis and eight
outcomes are consistent with QEEG and observations by other normal subjects. A follow-up testing was conducted 3 months
authors acquired from acute ischemic stroke patients, including after the initial recordings. Their results confirmed repro-
those who had deceased in the ensuing days. There seems to be ducibility of PMBS patterns in normal subjects since the repli-
a prognostic value of poststroke shift of scalp delta power max- cation of the recording has only a negligible effect on the
ima from the ipsilateral to the contralateral hemisphere indicat- lateralization. The side of hemiparesis in acute stroke patients
ing substantial worsening of cerebral pathophysiology (107). A could be distinguished on the basis of quantitative measure of
standard 19-electrode array appears adequate for the detection lateralization. The follow-up testing in three recovered stroke
of this marker, and routine usage of a standard electrode assay patients revealed a trend of changes in the lateralization toward
with fewer electrodes renders bedside EEG monitoring more the contralateral side of movement, an indication that this tech-
feasible than does a high-density array. The potential limita- nique is an option for evaluation of therapy-induced changes in
tions of such EEG monitoring are possible contamination of cortical movement organization (113).
QEEG indices by artifacts such as those due to patient move-
ment and/or by sleep- or drowsiness-related EEG activity. EEG IN POSTSTROKE HYPERSOMNIA
QEEG measures, computed promptly, perhaps automatically,
might help guide future clinical therapeutic decisions. In The incidence of stroke is about 300 to 600 per 100,000 per year
patients with large MCA infarctions, where decompressive sur- (Sudlow and Warlow, 1997). The frequency of hypersomnia
gery is being contemplated, such QEEG observations, in con- after stroke is essentially unknown but may be as high as 20%
cert with other investigations like MRI, may assist in more to 40% (120,121). In a consecutive series of 100 patients with
timely decisions to operate. ischemic stroke, 22 patients reported at 1 month after stroke
Recovery from stroke can vary greatly among patients even excessive daytime sleepiness and/or increase in sleep needs as
though they may have identical clinical symptoms. Several compared with prestroke situation (122). Patients with and
attempts have been made to determine neuroplastic changes on without poststroke hypersomnia were similar in age, gender,
the basis of electroencephalograms, which are readily available Epworth sleepiness score, and estimated sleep need hours
in clinical settings. In many studies, movement-related poten- before stroke. Patients with poststroke hypersomnia had a more
tials (MRPs) were studied in connection with stroke rehabilita- severe stroke and worse short-term outcome. The less well-
tion. MRPs constitute a waveform that is obtained by taking the defined symptom “fatigue” may be found even in a higher per-
“average” EEG, in terms of onset of movement or myoelectrical centage of poststroke patients (123).
activity of the prime mover (108,109). Many studies demon- The semiologic spectrum of poststroke hypersomnia is wide
strated topographical alterations of some MRP components and varies according to stroke topography (Bassetti, 2005; 122).
during the recovery period after a stroke (110–112). The analy- In deep (subcortical) hemispheric and particularly paramedian
sis of MRP, however, presents methodologic difficulties because thalamic lesions, hypersomnia may correspond to a so-called
of its low signal to noise ratio. Adequate recording of MRP presleep behavior, during which patients yawn, stretch, close their
requires careful repetition of many trials in which the elec- eyes, curl up, and assume a normal sleeping posture, while com-
tromyographic envelope of the investigated prime mover is plaining of a constant sleep urge. Some of these patients are able
similar, and subjects need to be trained to perform similar to control this behavior when stimulated or given explicit, active
movements. These practical issues led Eder and colleagues tasks to perform. In some patients with deep frontal, thalamic, or
(113) to consider more stable EEG phenomena as criteria to midbrain lesions, hypersomnia evolves to extreme apathy with
mark stroke recovery. Some prior findings indicate that event- lack of spontaneity and initiative, poverty of movement, a condi-
related desynchronization (ERD) might achieve this purpose tion for which the term “akinetic mutism” was coined. A contin-
(114). The ERD of the central mu-rhythm was studied exten- uum exists between hypersomnia, athymormia, akinetic mutism,
sively in normal subjects (115). Subsequent normative studies and fatigue. In some patients, episodes of hypersomnia, mutism,
have outlined that the symmetry of the mu-rhythm amplitude and akinesia alternate with episodes of insomnia, psychomotor
had, in comparison with ERD, a greater discriminative power agitation, or confusional state (120–122).
between normal subjects and stroke patients (116). Results of The most severe and persistent forms of hypersomnia are
more recent studies point to the existence of two relatively inde- observed in association with lesions in the paramedian thala-
pendent ERD generating systems within the central region of mus, midbrain, or upper pons. The paramedian thalamic stroke
each hemisphere with beta rebound following movement, also syndrome represents the most striking form of hypersomnia
referred to as postmovement beta event–related synchroniza- following focal brain damage. Hypersomnia can also be seen
tion (PMBS) (117). These beta oscillations are dominant on the with unilateral lesions but rarely persists, in these cases, for
contralateral side of movement and are found in the first sec- more than a few months.
ond after termination of movement. Several studies showed Less commonly hypersomnia may complicate lesions in the
that lateralization of PMBS depended on factors such as hand- caudate, striatum, lower pons, medial medulla, and cerebral
edness, age, and pathologic conditions, for example, Parkinson hemispheres (with or without mass effect) (121,122). The degree
Chapter 19 ■ Cerebrovascular Diseases and EEG 365

and duration of hypersomnia over time in these patients are less The EEG may show surprisingly little changes in this serious
severe than in patients with paramedian thalamic stroke. cerebral disorder. There is reason to presume that the neuronal
Hypersomnia following focal brain damage is usually con- oxygenation does not reach critically low values in spite of the
sidered to reflect a decreased arousal secondary to the dysfunc- fact that the cerebral blood flow is diminished in this condition
tion of the wakefulness maintaining neuronal systems (133). In the case of epileptic convulsions, EEG changes are
(ascending reticular activating system, ARAS). The occurrence usually limited to the period immediately before, during, and
of hypersomnia following cortical or striatal strokes without after the attacks. This is akin to the acute encephalopathy of
mass effect supports the assumption of a role of these struc- eclampsia gravidarum, where EEG abnormalities are barely
tures in the maintenance of arousal and more generally in detectable in the preeclamptic state and are practically limited
sleep–wake regulation (124). to the grand mal convulsions themselves.
The most striking example of increased sleep is seen in However, there have been reports of very pronounced EEG
patients with bilateral paramedian thalamic stroke. Arpa et al. abnormalities in hypertensive encephalopathy. Aguglia et al.
(125) reported a 44-year-old man with right lateral-tegmental (134) described severe parieto-occipital changes with promi-
pontine hematoma and severe hypersomnia, in whom long- nent paroxysmal features. After acute onset of hypertensive
term EEG monitoring showed increased amounts of sleep rang- encephalopathy (BP of 300/140 and age 70), Benna et al. (135)
ing from 11 to 15 hours per day during the first 3 months after observed generalized spikes and polyspikes followed by a slow
stroke. The relative amounts of slow wave sleep (4% to 11% of wave. With gradual improvement of hypertension, the paroxys-
total sleep time) and rapid eye movement (REM) sleep (8% to mal pattern lingered on. Figure 19.10 shows marked diffuse
10%) were slightly above normal values. Bastiju et al. (126) slowing in a patient with hypertensive encephalopathy.
described a patient with severe hypersomnia due to bilateral
thalamomesencephalic stroke with an initial sleep behavior EEG IN INTRACEREBRAL BLEEDING
over 18 hours per day. Eight months after stroke, hypersomnia
had regressed clinically to about 12 hours per day. By EEG cri- This form of stroke occurs mainly in hypertensive individuals;
teria, sleep was similarly present over about 12 hours per day, high BP in combination with vascular disease (hyalinosis) may
with an increase in both slow wave sleep (30% of total sleep) lead to a massive rupture. Occasionally, intracerebral hemor-
and REM sleep (22%). rhages are caused by aneurysms, arteriovenous malformations,
or alteration of vessels inside a cerebral neoplasm. The most fre-
SLEEP EEG AFTER HEMISPHERIC STROKE quently involved vessel is the lenticulostriate artery, which is a
small deep branch of the MCA. Its nearness to the internal cap-
Changes in sleep EEG patterns after hemispheric stroke have sule explains the devastating effects on the motor outflow.
been described in studies for the last three decades (127–129). It is likely that subclinical seizures are common after
Müller et al. (128) found a lower sleep efficacy, as well as lower intracranial hemorrhage. Subclinical seizure activity has gained
amounts of slow wave sleep and REM sleep in patients with attention from an increased understanding of the potential
acute hemispheric stroke. Gottselig et al. (129) documented a harmful effects of clinical seizure activity (136). Investigations
reduced spindle activity on the side of the lesion in patients with in the most extreme case of seizure activity, status epilepticus,
extrathalamic stroke. Sleep efficiency, preserved spindle activity, demonstrate that prolonged seizure activity can cause neuronal
and amount of REM sleep in the acute phase of stroke have been death. Animal experiments of convulsive status epilepticus and
shown to be associated with a favorable outcome (130,131). some models of nonconvulsive status epilepticus show that
Siccoli et al. (2008) investigated the hypothesis of a link between prolonged seizure activity causes immediate necrotic cell death
sleep and cognitive functions, particularly memory and atten- and delayed apoptotic cell death, primarily from activation of
tion, after stroke in 11 patients with first-ever hemispheric NMDA-mediated glutamatergic excitotoxic pathways. It is rea-
ischemic stroke within 8 days after symptoms onset and in 9 of sonable to consider that briefer periods of seizure activity cause
them at least 3 months after stroke. In the acute stroke phase, a cell death and this is true in some animal models of limbic
correlation between attention and amounts of slow wave sleep, seizures. However, there are few data demonstrating that dis-
REM sleep, and sleep efficiency was found. crete seizures cause cell death in humans so caution should be
exercised in directly extrapolating this to subclinical seizures.
HYPERTENSIVE ENCEPHALOPATHY The utility of prolonged EEG monitoring in the intensive care
unit has been investigated for various conditions. A decrease in
In this condition, severe arterial hypertension gives rise to cere- relative alpha power may predict vasospasm and delayed cerebral
bral edematous changes and hence to signs of intracranial ischemia after subarachnoid hemorrhage (137,138).
hypertension. Headache may reach an excruciating degree, Claassen et al. (138) performed a retrospective study on 102
especially in the morning, augmented by coughing and strain- consecutive patients with intracerebral hemorrhage who under-
ing and sometimes accompanied by vomiting. Transient attacks went continuous electroencephalographic monitoring.
of blindness may occur, and generalized as well as focal epilep- Convulsive seizures occurred in 19%, another 13% of patients had
tic seizures may materialize. Papilledema with retinal arteriolar subclinical, purely electroencephalographic seizures, and 5% had
spasms and hemorrhages is common. The condition may both electroencephalographic and clinical seizures. They found an
progress into somnolence, delirium, stupor, and coma (132). increased risk of seizures if the hemorrhage reached the cortex or
Figure 19.10 A 71-year-old female with acute loss of consciousness. CT scan revealed a right-sided thalamic hemorrhage.
A: EEG showed right hemispheric rhythmic bi- and triphasic waves also showing on the left, but not synchronously.
B: After 4 days only slight improvement of EEG changes.
Chapter 19 ■ Cerebrovascular Diseases and EEG 367

if it expanded by 30% in the first 24 hours. Periodic epileptiform LOWER BRAINSTEM HEMORRHAGE
discharges were more frequently seen in hemorrhages closer to the
cortex. Midline shift and poor outcome were not more frequent in These uncommon strokes almost always give rise to coma and
patients with seizures when compared with those without respiratory anomalies (33), sometimes progressing to acute
seizures. By contrast, periodic discharges were independently respiratory failure (143). The pupils are usually very small.
associated with poor outcome. There was a trend toward worse Quadriplegia is very common; bouts of decerebrate posturing
outcome for patients with electroencephalographic seizures, but also occur. The EEG may show the striking finding of a well-
seizures did not remain a predictor in multivariate analysis. preserved posterior alpha rhythm that cannot be blocked by
Among those who had seizures during monitoring, 94% of various modalities of stimuli (144–146), but in exceptional
seizures were detected within the first 72 hours. This implies that cases, the reactivity of the alpha rhythm may persist (147).
routinely providing continuous EEG monitoring for 3 days would Other patients show diffuse low-voltage tracings as seen in
frequently detect subclinical seizures. But the retrospective nature cases of lower brainstem infarction (see under “Basilar Artery
of this study limited it to those 13% of all intracranial hemorrhage Thrombosis”).
admissions for whom continuous EEG was requested (136).
There may have been a selection bias toward those who had some
CEREBELLAR HEMORRHAGE
reason to suspect seizure activity. In another prospective study,
seizures occurred in 28% of patients after intracerebral hemor- Acute cerebellar hemorrhage rapidly leads to coma after a few
rhage, suggesting that they are frequent (139). The true incidence minutes of headaches, dizziness, or vertigo. Cerebellar edema
of subclinical seizures after intracerebral hemorrhage awaits a rapidly produces a pressure cone (tonsillar herniation) with
prospective consecutive case series of continuous EEG monitor- fatal outcome. The rapidity of the evolution usually precludes
ing. It could be clinically important to find subclinical seizures if EEG studies. A remarkable study of 22 cases was carried out by
they increase mortality or cause neuronal injury. But, Claassen et Rasheva et al. (49); it demonstrated high-voltage delta activity,
al. (138) have not found seizures to be associated with poor out- more over the contralateral cerebral hemisphere. In patients
come. Obvious seizures do not present a problem; they are usually with cerebellar stroke, movement-related cortical potentials
treated with anticonvulsants. It seems reasonable to treat subclin- were found to be depressed (148).
ical seizures due to intracranial hemorrhage using routine meth-
ods, that is, oral anticonvulsants, but caution should be exercised
in treating aggressively, such as with continuous intravenous SUBARACHNOID HEMORRHAGE
propofol or midazolam. Aggressive therapies can cause severe (INTRACRANIAL ANEURYSMS,
iatrogenic complications, whereas damage from seizures is likely ARTERIOVENOUS MALFORMATIONS,
to be mild. This is unlike status epilepticus where the damage is AND OTHER CAUSES)
significant and risk of mortality is high. It is likely that subclinical
seizures are common after intracranial hemorrhage but paroxys- The extremely dramatic symptomatology of a subarachnoid
mal EEG activity suspicious for seizures has unknown clinical sig- hemorrhage with acute meningeal signs does not need special
nificance. Prolonged EEG monitoring is expensive and presents discussion. Cranial nerve deficits are quite common. In more
logistical problems. At present, the significance of subclinical severe cases, there is evidence of cerebral involvement with
seizures after intracerebral hemorrhage and the need of suppress- impairment of consciousness and, in about 10% to 20% of the
ing them is not known (136). cases, epileptic seizures.
The EEG shows diffuse changes, disorganization, disruption
THALAMIC HEMORRHAGE of the posterior alpha rhythm, and excessive slow activity.
Occasionally, lateralization of slow activity may be indicative of
Thalamic bleeding usually causes temporary loss of conscious- the primarily involved hemisphere (149–151). With total
ness. Larger hemorrhages may give rise to ipsilateral delta activ- dependence on arteriographic demonstration of the causative
ity (Fig. 19.10A,B). According to Jasper and Van Buren (140), lesion and partial dependence on refined CT scan methods,
there is a reduction of alpha rhythm in the case of anteroventral there is no practical need for an EEG in the search for the cor-
thalamic damage. Alpha enhancement may be noted in patients rect localization. The EEG, however, remains a valuable indica-
with posterior thalamic lesions. Lack of sleep spindles has been tor of the general state of cerebral functioning (152,153).
reported (141). Delayed P3 responses were reported by Onofrj The patient’s general condition seriously deteriorates when
et al. (142). subarachnoid bleeding is followed by vasospasm. The patho-
genesis of such widespread vasospasm is poorly understood
MIDBRAIN HEMORRHAGE (154–156). According to Kiloh et al. (157), marked delta foci
occur in areas of massive vasospasm. Such secondary lesions are
Mesencephalic bleeding without extension into the pons is sometimes associated with sharp activity (9). Prolonged
most likely to give rise to Parinaud syndrome; it is seldom fatal. vasospasm and a decrease of cerebral blood flow may lead to
The EEG often shows diffuse activity in the upper theta range dementia and diffuse cerebral atrophy. The area of massive
(7 per second, according to Ref. 9). vasospasm correlates with the area of local EEG slowing (158).
368 Part III ■ Clinical EEG: General

Ruptured bifurcation aneurysms (circle of Willis and vicin- syndromes with occlusion and focal cerebral deficit (168). The
ity) are the most common cause of subarachnoid hemor- EEG in CNS complication of lupus erythematosus is deter-
rhage. Margerison et al. (159) carried out extensive EEG mined by the severity of clinical neurologic deficits and may
studies in 70 patients with ruptured aneurysms in a search for range from normal to markedly abnormal findings of either
valid localizing signs. Massive bleeding from a ruptured focal or diffuse character.
aneurysm into the cerebral parenchyma is prognostically omi- EEG studies of systemic lupus erythematosus and other
nous and can be fatal within less than 1 hour. The EEG pat- collagen or connective tissue diseases such as periarteritis
tern of intracerebral hematomas resulting from such bleeding nodosa, scleroderma, giant cell arteritis, or rheumatoid arthri-
was studied by Van der Drift (8). Regional polymorphic delta tis have been scanty (169–171; see also Ref. 172). According to
activity was found to be more pronounced than in acute MCA Finn et al. (173), the EEG is useful in the early detection of
ischemia. this disease. Kogeorgos and Scott (174) studied 74 eases; EEG
In survivors of subarachnoid hemorrhages from aneurysms, abnormalities were present in 79%. Gottwald and Sturm
epileptic seizures have been found in 12.5% (160); marked EEG (175) reported abnormal records in 77% of 40 patients. The
changes with spikes are also found (157). Scott and Cabral EEG abnormalities consist of focal or diffuse slowing, spikes
(161) stressed the frequent occurrence of epileptic seizures fol- or sharp waves of focal or multifocal character, and “dysrhyth-
lowing intracranial aneurysm surgery (clipping, wrapping). mic phenomena.”
Survivors of subarachnoid bleeding from an aneurysm of In Wegener’s granulomatosis, CNS involvement is less com-
the anterior communicating artery sometimes develop severe mon than in lupus erythematosus. EEG abnormalities may
personality changes and dementia. Their state of apathy and occur; metabolic (renal) causes have to be ruled out (176). In
loss of drive is not associated with typical EEG changes. Severe morphea (localized scleroderma), epileptic seizures are fairly
frontal and hypothalamic (deep perforating branches of ante- common (25%), and both epileptic and nonepileptic EEG
rior cerebral artery) lesions might account for this lamentable abnormalities are three times as common as in age-matched
clinical picture. control population (177).
Arteriovenous malformations are the cause of subarachnoid In Sjögren syndrome, the CNS may be affected by an aseptic
hemorrhages that are much less severe and life threatening than meningoencephalitis (178). Cognitive disturbances and
is bleeding from a ruptured aneurysm. These malformations dementia may also be caused by vasculitis that is treatable with
may also give rise to focal or generalized epileptic seizures that corticosteroids in high dosages (179). These authors also found
usually start between ages 10 and 35. The EEG of patients with diffuse EEG abnormalities in a patient with vasculitis.
arteriovenous malformations has been described as abnormal
in all observed cases (162). The interpretation of the abnormal-
ities is beset with difficulties, because a sizable number of cases OTHER TYPES OF VASCULAR DISEASE
may show an “erroneous” lateralization with slowing or sharp In thromboangiitis obliterans cerebri, temporal focal delta activ-
activity over the “wrong” hemisphere (163–166). As impractical ity may be found over areas of recent cortical damage, but these
as such findings may be, their pathophysiologic substratum is changes disappear quickly; the EEG is probably of little diag-
not devoid of clinical interest, since the misleading lateraliza- nostic value in this disease (180). EEG findings are also
tion is determined by intracerebral steal phenomena. In the regarded as noncontributory in patients with moyamoya disease
writer’s personal experience, even a very large arteriovenous (181). According to Aoki et al. (11), a reappearance of the
malformation may be found without any EEG abnormality. hyperventilation-induced buildup of slow activity approxi-
This evidently indicates the lack of consistent hypoxic impair- mately 20 to 60 seconds after the activation is a typical finding
ment of the cortical function. in moyamoya disease. This phenomenon has been studied more
Subarachnoid bleeding may occur without demonstrable extensively by Ohyama et al. (182). Granulomatous angiitis
cause. In some cases, the source of bleeding consists of aneurys- (183,184) is a progressive and fatal disease. Prominent diffuse
matic widening of a bacterially infected artery (arteritis leading or local slowing of the EEG is a common finding.
to a “mycotic” aneurysm). Bacterial endocarditis is usually the In sickle cell anemia, neurologic complications may occur
cause of this type of vascular pathology. During treatment with occasionally. Impairment of consciousness, convulsions, and
endovascular embolization, EEG monitoring may detect clini- neurologic deficits may prevail for several days or weeks.
cal hazards (167). Marked EEG changes are noted (172,185). Focal slowing was
found to be more impressive than the corresponding neuro-
CEREBROVASCULAR INVOLVEMENT logic symptomatology, with considerable attenuation of the
IN CONNECTIVE TISSUE DISEASES EEG changes during sleep (186).
Neurologic and electroencephalographic changes due to
Systemic lupus erythematosus gives rise to neurologic signs and blood diseases will be discussed under the heading of metabolic
symptoms in 25% to 35% of the patients (168). This is due to encephalopathies and brain tumors in neurooncology. Briefly,
small patchy vessel involvement. Convulsions and psychotic intracerebral hematomas are not uncommon in hemophilia,
episodes are the most common CNS changes. About 2% to 5% of thrombocytopenia, and leukemia. In such cases, marked focal
the patients with lupus erythematosus develop cerebrovascular or diffuse slowing is found in the EEG.
Chapter 19 ■ Cerebrovascular Diseases and EEG 369

CEREBRAL MANIFESTATIONS EEG in Syncope


OF CARDIAC DISEASES Clinically, it can be difficult to differentiate syncope from an
epileptic seizure. Syncope is defined as transient loss of con-
Basic Considerations sciousness and of postural control. Pathophysiologically, it is
Cardiac diseases and dysfunctions can impair the brain in vari- based on global cerebral hypoxia as a result of transient hypoper-
ous ways with a wide variety of pathologic changes. In other fusion of the brain. Patients with syncope usually fall to the
words, only a part of the CNS lesions and disturbances caused ground with the onset of unconsciousness. In experimental stud-
by cardiac problems falls into the category of cerebrovascular ies, the frequency of tonic or myoclonic muscle activity during
disorder. Cerebrovascular phenomena such as arterial syncope ranges between 12% and 100% with most studies find-
embolism are inseparably associated with bacterial infections ing the frequency between 70% and 90%. Thus, convulsive syn-
such as cerebritis and brain abscess. In other cases, one is deal- cope is the rule rather than the exception, which can be explained
ing with hypoxic–anoxic cerebral pathology, especially in com- by the fact that convulsions are an intrinsic component of the
plications of open heart surgery. brain’s response to hypoxia. The most important criterion to dif-
ferentiate syncopal and epileptic convulsions is their specific phe-
nomenology. In contrast to epileptic muscle activity, syncopal
Congenital Heart Disease myoclonic movements are arrhythmic. Mostly, they are multi-
Neurologic disturbances are quite frequent in patients with form rather than generalized; that is, they occur asynchronously
congenital heart disease. According to Tyler and Clark (187), in different regions of the body. Only in rare cases do they last
neurologic deficits occur in 25%, blackout spells in 12%, and longer than 30 seconds. Tonic phenomena are significantly less
convulsions in 6% of the cases. It is therefore no surprise that frequent in syncope and are usually less pronounced. A tonic
abnormal EEG tracings are fairly common in congenital heart extension of the body with flexion of the arms is more common
disease (188–194). Brain abscess is a well-known complication in profound cerebral hypoxia, for example, due to prolonged
of congenital heart disease, especially in infancy and early child- asystole, or in syncope in children, such as breath-holding spells.
hood. Focal polymorphic delta activity is usually very promi- For the differentiation of syncope and epileptic seizures, post-
nent in these abscesses. This type of brain abscess is ictal phenomena are of particular importance. After syncope,
unassociated with endocarditis and dissemination of infected patients are fully reorientated within very few seconds, and even
emboli. Diffuse cerebral hypoxia may give rise to impairment of after prolonged attacks lasting 1 to 2 minutes, the postictal con-
consciousness and marked diffuse slowing in the EEG. Focal fusion or drowsiness lasts rarely longer than 30 seconds. Any
slowing may also occur. Such hypoxic lesion may gradually longer-lasting disorientation suggests an epileptic seizure. But it
become epileptogenic (194,195). must be kept in mind that partial seizures of frontal lobe origin
Cerebral hypoxia may develop in cyanotic as well as in non- are frequently associated with a fast reorientation.
cyanotic forms of congenital heart disease, but it is more fre- In rare cases, there is an interaction between syncopal and
quently found in the former. epileptic mechanisms within a single attack. On the one hand,
this means that an epileptic seizure can emerge from syncope.
On the other hand, in certain forms of epileptic seizures, espe-
Acquired Heart Disease cially in complex partial seizures of temporal lobe origin, car-
CNS complications of acute or subacute bacterial endocarditis diac arrhythmias can arise, which can then lead to syncope. If
are essentially infectious diseases and will be discussed under the medical history suggests epileptic and syncopal phenomena
that heading. In cardiac decompensation, a variety of causes and within the same attack, the diagnosis can only be made by
mechanisms eventually leads to cerebral hypoxia with slowing means of ictal EEG and ECG recordings.
and terminal flattening of the EEG (172,196,197). EEG abnor- The EEG during syncope shows generalized high-amplitude
malities are common in chronic bronchopneumopathy with cor slow-wave activity at the onset of unconsciousness, followed by
pulmonale. These changes may (198), or may not parallel the flattening of the EEG, and subsequently slow waves again before
degree of impaired consciousness (199). the normal background activity returns (Brenner, 1997). This
Paroxysmal tachycardia may be associated with transient sequence is independent of the mechanism of syncope and of
EEG abnormalities (200,201), especially when the attack leads the clinical presentation as convulsive or nonconvulsive syn-
to impairment of consciousness. Walsh et al. (202) stressed cope as it represents the common final path in terms of global
paroxysmal cerebral discharges in conjunction with paroxysmal cerebral hypoxia.
atrial tachycardia, which was considered secondary to the cere- The significance of EEG in the diagnosis of syncope is often
bral events. overestimated. It could be shown that in unselected patients with
The Adams–Stokes syndrome is usually associated with a per- syncope, the recording of a postictal EEG was not helpful.
manent heart block and a very slow pulse rate. The electroen- Epileptiform patterns in an interictal EEG recording can indeed
cephalographer is advised to use EEG monitoring throughout corroborate a diagnosis of epilepsy. However, additional syncopal
the recording. During the attack, slow activity appears after a attacks are not excluded. On the other hand, even in chronic
few seconds of asystolia with progressive amplitude of delta epilepsy epileptiform patterns can be absent during interictal EEG
waves (203) (see also Chapter 31). recordings, thus also preventing a definite classification of the
370 Part III ■ Clinical EEG: General

attacks. Also, episodic seizures induced by alcohol or benzodi- 19. Fisher M, Garcia JH. Evolving stroke and the ischemic penumbra.
azepine withdrawal are usually not associated with epileptiform Neurology. 1996;47:884–888.
patterns in the interictal EEG. Therefore, EEG recordings are not 20. Kayser-Gatchalian MC, Neundörfer B. The prognostic value of
recommended as standard tests alter syncope and should be EEG in ischaemic cerebral insults. Electroencephalogr Clin
Neurophysiol. 1980;49:608–617.
reserved for patients with positive history for epileptic seizures.
21. Paddison RM, Ferris GS. The electroencephalograms in cerebral
vascular disease. Electroencephalogr Clin Neurophysiol. 1961;13:
REFERENCES 99–110.
22. Van der Drift JHA, Magnus O. The EEG in cerebral ischemic lesions.
1. Keilson MJ, Miller AE, Drexler ED. Early EEG and CTT scanning Correlations with clinical and pathological findings. In: Gastaut H,
in stroke—a comparative study [abstract]. Electroencephalogr Clin Meyer JS, eds. Cerebral Annoxia and the Electroencephalogram.
Neurophysiol. 1985;61:21P. Springfield. IL: Thomas; 1961:180–196.
2. Gloor P, Ball G, Schaul N. Brain lesions that produce delta waves 23. Sainio K, Stenberg D, Kesimäki I, et al. Visual and spectral EEG
in the EEG. Neurology (Minneapolis). 1977;27:326–333. analysis in the evaluation of the outcome in patients with ischemic
3. Gastaut JL, Michel B, Sabet Hassan S, et al. Electroencephalography brain infarction. Electroencephalogr Clin Neurophysiol. 1983;56:
in brain edema (127 cases of brain tumor investigated by cranial 117–124.
computerized tomography). Electroencephalogr Clin Neurophysiol. 24. Burghaus L, Hilker R, Dohmen C, et al. Early electroencephalogra-
1979;46:239–255. phy in acute ischemic stroke: prediction of a malignant course?
4. Chen J, Simon R. Ischemic tolerance in the brain. Neurology. Clin Neurol Neurosurg. 2007;109:45–49.
1997;48:306–311. 25. Chatrian GE, Shaw GM, Leffman H. The significance of periodic
5. Hachinski V, Norris JW. The deteriorating stroke.. In: The Acute lateralized epileptiform discharges: an electrographic clinical and
Stroke. Philadelphia, PA: FA Davis (Quoted by Hachinski and pathological study. Electroencephalogr Clin Neurophysiol. 1964;17:
Norris, 1985). 177–193.
6. Velho-Groneberg P. The EEG: an important (and often under- 26. Echlin FA, Arnett V, Zoll J. Paroxysmal high voltage discharges
rated) tool in the diagnosis of stroke. Am J EEG Technol. 1986; from isolated and partially isolated human and animal cerebral
26:213–224. cortex. Electroencephalogr Clin Neurophysiol. 1952;4:147–164.
7. Kok NKD, Van der Drift JHA. The significance of the EEG for local- 27. de la Paz D, Brenner RP. Bilateral independent periodic lateralized
ization and prognosis of supratentorial ischemic disease. A clinical epileptiform discharges. Arch Neurol. 1981;38:713–715.
pathological study. Preliminary report. Rev EEG Neurophysiol. 1971; 28. Reiher J, Rivest J, Grand’Mason F. Periodic lateralized epileptiform
1:295–307. discharges with transitional rhythmic discharges. Electroencephalogr
8. Van der Drift JHA. The EEG in cerebrovascular disease. In: Vinken Clin Neurophysiol. 1991;78:12–17.
PJ, Bruyn GW, eds. Handbook of Clinical Neurology. Vol 11. 29. Pohlmann-Eden B, Hoch DB, Cochius JL, et al. Periodic lateralized
Amsterdam: North-Holland; 1972:267–291. epileptiform discharges: a critical review. J Clin Neurophysiol. 1996;
9. Van der Drift JHA, Kok NKD. The EEG in cerebrovascular disorders 13:519–530.
in relations to pathology. In: A. Remond, Ed-in-chief. Handbook 30. Pohlmann-Eden B, Hoch OB, Cochius JI, et al. Stroke and
of Electroencephalography and Clinical Neurophysiology. Vol 14A. epilepsy: critical review of the literature. Part I: Epidemiology and
Amsterdam: Elsevier; 1972:12–30, 47–64. risk factors. Cerebrovasc Dis. 1996;6:332–338.
10. Hilker BL, Dohmen C, Bosche B, et al. Early electroencephalogra- 31. Fitzpatrick W, Lowry N. PLEDS: clinical correlates. Can J Neurol
phy in acute ischemic stroke: prediction of a malignant course? Sci. 2007;34:443–450.
Clin Neurol Neurosurg. 2007;109:45–49. 32. Friedlander WJ. Electroencephalographic changes in acute brain
11. Aoki Y, Hiraga H, Ichijo S. EEG of moyamoya disease [abstract]. stem vascular lesions. Neurology (Minneapolis). 1959;9:24–34.
Electroencephalogr Clin Neurophysiol. 1977;43:490. 33. Plum F, Posner JB. Diagnosis of Stupor and Coma. Philadelphia, PA:
12. Kraaier V, Van Huffelen AC, Wieneke GH. Changes in quantitative Davis; 1966.
EEG and blood flow velocity due to standardized hyperventilation. A 34. Hawkes CH, Bryan-Smyth L. The electroencephalogram in the
model for transient ischemia in normal subjects. Electroencephalogr “locked-in” syndrome. Neurology. 1974;24:1015–1018.
Clin Neurophysiol. 1988;70:377–387. 35. Markand ON. Electroencephalogram in the “locked-in” syndrome.
13. Carney AL, Anderson EM, Burns E. Cerebral hemodynamic eval- Electroencephalography. 1976;40:529–534.
uation. In: Carney AL, Anderson EM, eds. Diagnosis and Treatment 36. Chase TN, Moretti L, Prensky EL. Clinical and electroencephalo-
of Brain Ischemia. New York, NY: Raven Press; 1981:335–359. graphic manifestations of vascular lesions of the pons. Neurology
14. Anderson EM, Carney AL, Page L. EEG in the vascular laboratory. (Minneapolis). 1968;18:357–365.
In: Carney AL, Anderson EM, eds. Diagnosis and Treatment of 37. Westmoreland BF, Klass DW, Sharbrough FW, et al. “Alpha coma”:
Brain Ischemia. New York, NY: Raven Press; 1981:325–334. electroencephalographic, clinical, pathological and etiological cor-
15. Hossmann KA. Viability thresholds and the penumbra of focal relation. Arch Neurol. 1975;32:713–718.
ischemia. Ann Neurol. 1994;36:557–565. 38. Vignaendra V, Wilkus RJ, Copass MK, et al. Electroencephalographic
16. Genvins A, Le J, Brickett P, et al. Seeing through the skull: advanced rhythms of alpha frequency in comatose patients after cardiopul-
EEG use MRIs to accurately measure cortical activity from the monary arrest. Neurology. 1974;24:582–588.
scalp. Brain Topogr. 1991;4:125–131. 39. Carroll WM, Mastaglia FL. Alpha and beta coma in drug intoxica-
17. Jackel RA, Harner RN. Computed EEG topography in acute stroke. tions uncomplicated by cerebral hypoxia. Electroencephalogr Clin
Neurophysiol Clin. 1989;19:185–197. Neurophysiol. 1979;46:95–105.
18. Luu P, Tucker DM, Englander R, et al. Localizing acute stroke- 40. Jouvet M. Coma and other disorders of consciousness. In: Vinken
related EEG changes: assessing the effects of spatial undersampling. PJ, Bruyn GW, eds. Handbook of Neurology. Vol 12. Amsterdam:
J Clin Neurophysiol. 2001;18:302–317. North-Holland; 1969:64–79.
Chapter 19 ■ Cerebrovascular Diseases and EEG 371

41. Kenangil G, Orken DN, Yalcin D, et al. Triphasic EEG pattern in 62. Albers GW, Caplan LR, Easton JD, et al. TIA Working Group: tran-
bilateral paramedian thalamic infarction. Clin EEG Neurosci. 2008; sient ischemic attack—proposal for a new definition. N Engl J
39:185–190. Med. 2002;347:1713–1716.
42. Schreiner A, Pohlmann-Eden B, Schwartz A, et al. Epileptic 63. De Reuck J, Van Maele G. Transient ischemic attacks and inhibitory
seizures in subcortical vascular encephalopathy. J Neurol Sci. 1995; seizures in elderly patients. Eur Neurol. 2009;62:344–348.
130:171–177. 64. Bearden S, Uthman B. Cerebral hemodynamic compromise asso-
43. De Reuck J, Decoo D, Boon P, et al. Late-onset epileptic seizures in ciated with limb shaking TIA and focal EEG slowing. Am J
patients with leukoaraiosis: a positron emission tomographic Electroneurodiagnostic Technol. 2009;49:225–243.
study. Eur Neurol. 1996;36:20–24. 65. Enge S, Lechner H, Logar C, et al. Clinical value of EEG in transient
44. De Reuck J, Santens P, Decoo D, et al. Positron emission tomo- ischemic attacks. In: Lechner H, Aranibar A, eds. EEG and Clinical
graphic study of late-onset cryptogenic symptomatic seizures. Clin Neurophysiology. Amsterdam: Excerpta Medica; 1980:173–180.
Neurol Neurosurg. 1995;97:208–212. 66. Silva A, Gross PT. Use of EEG in the evaluation of TIAs [abstract].
45. Lhermitte F, Gautier JC, Derouesne C. Anatomopathologie et Electroencephalogr Clin Neurophysiol. 1983;56:13P.
physiopathologie des stenoses carotidiennes. Rev Neurol (Paris). 67. Garcin R, Pestel M. Thrombophlebites Cerebrales. Paris: Masson;
1966;115:641. 1949.
46. Lhermitte F, Guiraud B. Ischemic accidents in the middle cerebral 68. Dubois J. Les thrombophlebites cerebrales post-partum. Gynaecol
artery. Arch Neurol. 1968;19:248. Obstet. 1956;55:478.
47. Lhermitte F, Gautier JC. Sites of cerebral arterial occlusions. In: 69. Huhn A. Die Hirnvenen- und Sinusthrombose. Fortschr Neurol.
Williams D, ed. Modern Trends in Neurology. London: Butterworths; 1957;25:440.
1975:123–140. 70. Huhn A. Die venöse Abflußstörung. In: Quandt J, ed. Die zere-
48. Rasheva M. Epileptic seizures in the acute stage of embolic stroke bralen/Durchblutungsstörungen des Erwachsenenalters. Berlin: VEB
[abstract]. Electroencephalogr Clin Neurophysiol. 1981;52:78P. Verlag Volk und Gesundheit; 1969:773–800.
49. Rasheva M, Stamenov E, Todorova P, et al. Dynamics of electrical 71. Carter AB. Clinical aspects of infarction. In: Vinken PJ, Bruyn GW,
activity of the brain in cerebellar haemorrhage [abstract]. eds. Handbook of Neurology. Amsterdam: North-Holland; 1972:
Electroencephalogr Clin Neurophysiol. 1981;52:78P. 292–326.
50. Naquet R, Vigouroux RA. EEG in cerebral gas emboli from cardiac 72. Lemmi H, Little SC. Occlusion of intracranial venous structures. A
surgery. In: Remond A, Ed-in-chief. Handbook of Electroen - consideration of the clinical and electroencephalographic find-
cephalography and Clinical Neurophysiology. Vol 14A. Amsterdam: ings. Arch Neurol. 1960;3:252–266.
Elsevier; 1972:38–44. 73. Franco PJ. Electroencephalography in venous disturbances of the
51. Müller HR, Klingler M. The electroencephalogram in cerebral fat brain. Rev Neuro-Psiquiatr. 1961;24:447–452.
embolism. Electroencephalogr Clin Neurophysiol. 1965;18:278–386. 74. Hubach H, Struck G. Zur Korrelation von EEG und patho-mor-
52. Scherzer E. EEG—Veränderungen bei zerebraler Fettembolie. phologischen Befunden cerebraler Gefäßprozesse. Arch Psychiatr
Psychiatr Neurol. 1967;153:337. Nervenkr. 1965;206:641.
53. Hegedüs K, Fekete I, Tury F, et al. Experimental focal cerebral 75. Griewing B, Mertins L, Lütcke A, et al. Das Elektroenzephalogramm
ischaemia in rabbits. J Neurol (Berl). 1985;232:223–230. in der Verlaufsbeobachtung und Diagnostik von Thrombosen des
54. Zülch KJ, Behrend RCH. The pathogenesis and topography of Hirnnervensystems. EEG-Labor. 1991;13:103–117.
anoxia and ischemia of the brain in man. In: Gastaut H, Meyer JS, 76. Hauser W, Annegers J, Kurland L. Incidence of epilepsy and unpro-
eds. Cerebral Anoxia and the Electroencephalogram. Springfield, IL: voked seizures in Rochester, Minnesota: 1935–1984. Epilepsia. 1993;
Thomas; 1961:144–163. 34:453–468.
55. Alajouanine T, Lecasble R, Remond A. Elements graphiques parox- 77. Berges S, Moulin T, Berger E, et al. Seizures and epilepsy following
ystiques lents de survenue periodiques, Correlations electroclin- strokes: recurrence factors. Eur Neurol. 2000;43:3–8.
iques. Rev Neurol (Paris). 1955;93:477–478. 78. Ferro JM, Pinto F. Poststroke epilepsy: epidemiology, pathophysi-
56. Barolin C, Scherzer E, Naquet R, et al. Etude electroclinique des etats ology and management. Drugs Aging. 2004;21:639–653.
de mal epileptiques survenant chez les apolectiques. Rev Neurol 79. Kilpatrick CJ, Davis SM, Tress BM, et al. Epileptic seizures in acute
(Paris). 1962;107:242–243. stroke. Arch Neurol. 1990;47:157–160.
57. Markand ON, Daly DD. Pseudoperiodic lateralized paroxysmal 80. Lamy C, Domigo V, Semah F, et al. Early and late seizures after
discharges in the electroencephalogram. Neurology (Minneapolis). cryptogenic ischemic stroke in young adults. Neurology. 2003;60:
1971;21:975–981. 400–404.
58. Matsuo F. Multifocal epileptic seizures associated with acute cere- 81. Lossius MI, Rønning OM, Slapø GD, et al. Poststroke epilepsy:
bral ischemia [abstract]. Electroencephalogr Clin Neurophysiol. 1985; occurrence and predictors—a long-term prospective controlled
61:25P. study (Akershus Stroke Study). Epilepsia. 2005;46:1246–1251.
59. Millikan CH. Transient cerebral ischemia: definition and natural 82. Niedermeyer E. Über Epilepsie im höheren Lebensalter. Arch
history. In: McDowell FH, Sonnenblick H, Lesch M, eds. Current Psychiatr Nervenkr. 1958;197:248–262.
Concepts in Cerebrovascular Disease. New York, NY: Grune and 83. Shinton RA, Gill JS, Melnick SC, et al. The frequency, characteristics
Stratton; 1980:1–6. and prognosis of epileptic seizures at the onset of stroke. J Neurol
60. Taghavy A, Hamer H. Parenchymal “damage” in transient ischemic Neurosurg Psychiatr. 1988;51:273–276.
attacks (TIAs) and prolonged reversible ischemic neurological 84. Bladin C, Alexandrov A, Bellavance A, et al. Seizures after stroke: a
deficits (PRINDs): the role of cranial CT and EEG. Int J Neurosci. prospective multicenter study. Arch Neurol. 2000;57:1617–1622.
1992;66:251–261. 85. Burn J, Dennis M, Bamford J, et al. Epileptic seizures after first
61. Castle J, Mlynash M, Lee K, et al. 2010. Agreement regarding diag- stroke. Br Med J. 1997;315:1582–1587.
nosis of transient ischemic attack fairly low among stroke-trained 86. Gupta S, Naheedy M, Elias D, et al. Post-infarction seizures: a clin-
neurologists. Stroke. 2010;41(7):1367–1370. ical study. Stroke. 1988;19:1477–1481.
372 Part III ■ Clinical EEG: General

87. Holmes GL. The electroencephalogram as a predictor of seizures 109. Gilden L, Vaughan HG, Costa LD. Summated human EEG poten-
following cerebral infarction. Clin Electroencephalogr. 1980;11: tials with voluntary movement. Electroencephalogr Clin
834. Neurophysiol. 1966;20:433–438.
88. Kammersgaard LP, Olsen T. Poststroke epilepsy in the 110. Green JB, Bialy Y, Sora E, et al. High-resolution EEG in poststroke
Copenhagen stroke study: incidence and predictors. J Stroke hemiparesis can identify ipsilateral generators during motor
Cerebrovasc Dis. 2005;14:210–214. tasks. Stroke. 1999;30:2659–2665.
89. Kelly KM. Poststroke seizures and epilepsy: clinical studies and 111. Platz T, Kim IH, Pintschovius H, et al. Multimodal EEG analysis
animal models. Epilepsy Curr. 2002;2:173–177. in man suggests impairment-specific changes in movement-
90. Lancman ME, Golimstok A, Norscini J, et al. Risk factors for related electric brain activity after stroke. Brain. 2000;123:
developing seizures after a stroke. Epilepsia. 1993;34:141–143. 2475–2490.
91. Tettenborn B. Häufigkeit und Behandlung von zerebralen 112. Verleger R, Adam S, Rose M, et al. Control of hand movements
Anfällen und Epilepsie nach Schlaganfall. Arch Neurol Psychiatr. after striatocapsular stroke: high-resolution temporal analysis of
2004;155:169–172. the function of ipsilateral activation. Clin Neurophysiol.
92. Tettenborn B, Vitek L, Grabow T, et al. Carotid stenosis is a risk 2003;114:1468–1476.
factor for recurrent seizures after cerebral ischemia. Epilepsia. 113. Eder CF, Skoic D, Covickovic-Sternic N, et al. Symmetry of post-
1999;40(suppl 7):98. movement beta-ERS and motor recovery from stroke: a low-
93. De Reuck J, De Groote L, Van Maele G, et al. The cortical involve- resolution EEG pilot study. Eur J Neurol. 2006;13:1312–1323.
ment of territorial infarcts as a risk factor for stroke-related 114. Pfurtscheller G, Aranibar A. Event-related desynchronization
seizures. Cerebrovasc Dis. 2008;25:100–106. detected by power measurements of scalp EEG. Electroencephalogr
94. De Reuck J, Goethals M, Claeys I, et al. EEG findings after a cere- Clin Neurophysiol. 1977;42:817–826.
bral territorial infarct in patients who develop early- and late- 115. Pfurtscheller G, Aranibar A. Changes in central EEG activity in
onset seizures. Eur Neurol. 2006;55:209–213. relation to voluntary movement. I. Normal subjects. Prog Brain
95. De Reuck J, Sieben A, Van Maele G. Characteristics and outcomes Res. 1980;54:225–231.
of patients with seizures according to the time of onset in rela- 116. Kopruner V, Pfurtscheller G. Multiparametric asymmetry score
tion to stroke. Eur Neurol. 2008;59:225–228. (MAS)—distinction between normal and ischaemic brains.
96. Kotila M, Waltimo O. Epilepsy after stroke. Epilepsia. 1992;33: Electroencephalogr Clin Neurophysiol. 1984;57:343–346.
495–498. 117. Andrew C, Pfurtscheller G. Lack of bilateral coherence of post-
97. De Reuck J, Goethals M, Vonck K, et al. Clinical predictors of movement central beta oscillations in the human electroen-
late-onset seizures and epilepsy in patients with cerebrovascular cephalogram. Neurosci Lett. 1999;273:89–92.
disease. Eur Neurol. 2005;54:68–72. 118. Stancak A Jr, Pfurtscheller G. Effects of handedness on move-
98. Jacoby A, Snape D, Baker GA. Determinants of quality of life in ment-related changes of central beta rhythms. J Clin Neurophysiol.
patients with epilepsy. Neurol Clin. 2009;27:843–863. 1997;14:419–428.
99. Carrera E, Michel P, Despland PA, et al. Continuous assessment 119. Pfurtscheller G, Pichler-Zalaudek K, Ortmayr B, et al.
of electrical epileptic activity in acute stroke. Neurology. 2006;67: Postmovement beta synchronization in patients with Parkinson’s
99–104. disease. J Clin Neurophysiol. 1998;15:243–250.
100. Kilpatrick CJ, Davis SM, Hopper JL, et al. Early seizures after 120. Bassetti CL. Sleep and stroke. In: Kryger MH, Roth T, Dement
stroke. Risk of late seizures. Arch Neurol. 1992;49:509–511. WC, eds. Principles and Practice of Sleep Medicine. 4th ed.
101. Eliassen JC, Boespflug EL, Lamy M, et al. Brain-mapping tech- Philadelphia, PA: Elsevier Saunders; 2005:811–830.
niques for evaluating poststroke recovery and rehabilitation: a 121. Bassetti CL. Sleep and stroke. Semin Neurol. 2005;25:19–32.
review. Top Stroke Rehabil. 2008;15:427–450. 122. Bassetti CL, Valko P. Poststroke hypersomnia. Sleep Med Clin.
102. Strens LH, Asselman P, Pogosyan A, et al. Corticocortical cou- 2006;1:139–155.
pling in chronic stroke: its relevance to recovery. Neurology. 2004; 123. Carlsson GE, Moller A, Blomstrand C. Consequences of mild
63:475–484. stroke in persons 75 years—a 1 year follow-up. Cerebrovasc Dis.
103. Meinzer M, Elbert T, Wienbruch C, et al. Intensive language 2003;16:383–388.
training enhances brain plasticity in chronic aphasia. BMC Biol. 124. Braun AR, Balkin TJ, Wesensten NJ, et al. Regional cerebral blood
2004;2:20. flow throughout the sleep–wake cycle. Brain. 1997;120:1173–1197.
104. Cuspineda E, Machado C, Aubert E, et al. Predicting outcome in 125. Arpa J, Rodriguez-Albarino R, Izal E, et al. Hypersomnia after
acute stroke: a comparison between QEEG and Canadian neuro- segmenta pontine hematoma: case report. Neurologia. 1995;10:
logical scale. Clin Electroencephalogr. 2003;34:1–4. 140–144.
105. Finnigan SP, Rose SE, Chalk JB. Rapid EEG changes indicate 126. Bastiju H, Nighoghossian N, Salord F, et al. Mesodiencephalic
reperfusion after tissue plasminogen activator injection in acute infarct with hypersomnia: sleep recording in two cases. J Sleep
ischaemic stroke. Clin Neurophysiol. 2006;117:2338–2339. Res. 1994;3(suppl 1):16.
106. Finnigan SP, Rose SE, Walsh M, et al. Quantitative EEG indices of 127. Hachinski VC, Mamelak M, Norris JW. Clinical recovery and sleep
sub-acute ischaemic stroke correlate with clinical outcomes. Clin architecture degradation. Can J Neurol Sci. 1990;17:332–335.
Neurophysiol. 2007;118:2525–2532. 128. Müller C, Ackermann P, Bischof M, et al. Visual and spectral
107. Finnigan SP, Rose SE, Chalk JB. Contralateral hemisphere delta analysis of sleep EEG in acute hemispheric stroke. Eur Neurol.
EEG in acute stroke patients precedes worsening of symptoms 2002;48:164–171.
and death. Clin Neurophysiol. 2008;119:1689–1695. 129. Gottselig JM, Bassetti CL, Ackermann P. Power and coherence of
108. Deecke L, Scheid P, Kornhuber HH. Distribution of readiness sleep spindle frequency activity following hemispheric stroke.
potential, pre-motion positivity, and motor potential of the Brain. 2002;125:373–383.
human cerebral cortex preceding voluntary finger movements. 130. Giubilei F, Iannilli M, Vitale A. Sleep patterns in acute ischemic
Exp Brain Res. 1969;7:158–168. stroke. Acta Neurol Scand. 1992;86:567–571.
Chapter 19 ■ Cerebrovascular Diseases and EEG 373

131. Vock J, Ackermann P, Bischof M. Evolution of sleep and sleep 153. Labar DR, Fisch BJ, Fink ME, et al. Quantitative EEG following
EEG after hemispheric stroke. Brain. 2002;125:373–383. subarachnoid hemorrhage: a comparison of methods [abstract].
132. Toole JF, Patel AN. Cerebrovascular Disorders. 2nd ed. New York, Electroencephalogr Clin Neurophysiol. 1989;72:2P–3P.
NY: McGraw-Hill; 1974. 154. Echlin F. EEG changes following spasms of the basilar and verte-
133. Heine G. Vergleichende Untersuchung über Hirndurchblutungs- bral arteries produced by experimental subarachnoid hemorrhage
größe und Hirnstrombild beim Menschen. Verh Dtsch Ges [abstract]. Electroencephalogr Clin Neurophysiol. 1965;19:319.
Kreislaufforsch. 1953;19:196–210. 155. Levitt P. The effects of subarachnoid blood on the electrocor-
134. Aguglia U, Tinuper P, Farnarier G, et al. 1984. Electroencephalo- ticogram of the cat. In: Proceedings of the American EEG Society,
graphic and anatomo-clinical evidences of posterior cerebral Hans Berger Award Lecture; Washington, DC; September 1970.
damage to hypertensive encephalopathy. Clin Electroencephalogr. 156. Heros RC, Zervas NT, Varsos V. Cerebral vasospasm after sub-
15:53–60. arachnoid hemorrhage: an update. Ann Neurol. 1983;14:599–608.
135. Benna P, Bergamini L, Tarenzi L, et al. Hypertensive encephalopa- 157. Kiloh LG, McComas AJ, Osselton JW. Clinical Electroencephalo-
thy: association with unusual EEG changes [abstract]. Electroen- graphy. 3rd ed. London: Butterworths; 1972.
cephalogr Clin Neurophysiol. 1984;58:74P. 158. Rumpl E, Bauer G, Stampfei G. Der Angiospasmus bei der
136. Fountain NB. Is it time for routine EEG monitoring after intrac- Subarachnoidalblutung als wichtige Ursache für fokale Verän -
erebral hemorrhage? Neurology. 2007;69:1312–1313. derungen im EEG Z. EEG-EMG. 1977;8:200–204.
137. Vespa PM, Nuwer MR, Juhasz C. Early detection of vasospasm 159. Margerison JH, Binnie CD, McCaul LR. Electroencephalographic
after acute subarachnoid hemorrhage using continuous EEG ICU signs employed in the location of ruptured intracranial arterial
monitoring. Electroencephalogr Clin Neurophysiol. 1997;103: aneurysms. Electroencephalogr Clin Neurophysiol. 1970;28:
607–615. 296–306.
138. Claassen J, Jetté N, Chum F, et al. Electrographic seizures and 160. Walton JN. The electroencephalographic sequelae of sponta-
periodic discharges after intracerebral hemorrhage. Neurology. neous subarachnoid haemorrhage. Electroencephalogr Clin
2007;69:1356–1365. Neurophysiol. 1953;5:41.
139. Vespa PM, O’Phelan K, Shah M, et al. Acute seizures after intrac- 161. Scott DF, Cabral R. Development and prevention of seizures after
erebral hemorrhage. Neurology. 2003;60:1441–1446. neurosurgical procedures including ruptured cerebral
140. Jasper HH, Van Buren J. Interrelationship between cortex and aneurysms. In: Proceedings of the American Epilepsy Society. New
subcortical structures: clinical electroencephalographic studies. York; December 3, 1975.
Electroencephalogr Clin Neurophysiol. 1953;4:168–188. 162. Husby J, Norlen G, Petersen Z. Electroencephalographic find-
141. Hirose G, Saeki M, Kosoegawa H, et al. Delta waves in the EEGs ings in intracranial arterial and arteriovenous aneurysms and
of patients with intracerebral hemorrhage. Arch Neurol. 1981; subarachnoid hemorrhages. Acta Psychiatr Scand. 1953;28:
38:170–175. 387–400.
142. Onofrj M, Curatola L, Malatesta P, et al. Delayed P3 event-related 163. Groethuysen UC, Bickford RG, Svien HJ. The EEG in arteriove-
potentials (ERPs) in thalamic hemorrhage. Electroencephalogr nous anomalies of the brain. Arch Neurol Psychiatry. 1955;74:
Clin Neurophysiol. 1992;83:52–61. 506–513.
143. Steegman AT. Primary pontile hemorrhage. J Nerv Ment Dis. 164. Mosmans PCM, Jonkman EV. The significance of the collateral
1951;114:35–65. vascular system of the brain in hunt and steal syndromes. Clin
144. Loeb C, Poggio GF. Electroencephalogram in a case with pon- Neurol Neurosurg. 1980;82:145–156.
tomesencephalic haemorrhage. Electroencephalogr Clin Neuro- 165. Yeh HS, Kashiwagi S, Tew J, et al. Surgical management of epilepsy
physiol. 1953;5:295–296. with cerebral arteriovenous malformations. J Neurosurg. 1990;
145. Loeb C, Rosadini G, Poggio GF. Electroencephalograms during 72:216–223.
coma. Neurology. 1959;9:610–618. 166. Walton A. Pre- and postoperative findings in an arteriovenous
146. Neundörfer BL, Meyer-Wahl L, Meyer JG. Alpha-EEG und aneurysm. A case study. Am J EEG Technol. 1994;34:55–65.
Bewußtlosigkeit. Ein kasuistischer Beitrag zur lokaldiagnostis- 167. Paiva T, Campos J, Baeta E, et al. EEG monitoring during
chen Bedeutung des Alpha-EEG beim bewußtlosen Patienten. Z endovascular embolization of cerebral arteriovenous malforma-
EEG-EMG. 1974;5:106–114. tions. Electroencephalogr Clin Neurophysiol. 1995;95:3–13.
147. Radermecker J. Severe acute necrosis of the pons with long survival: 168. Aita JA. Neurologic Manifestations of General Diseases.
electroclinical symptoms and absence of cerebral lesions [abstract]. Springfield, IL: Thomas; 1975.
Electroencephalogr Clin Neurophysiol. 1967;23:281–282. 169. Taylor RM, Pacella BC. Electroencephalograms in scleroderma.
148. Gerloff C, Altenmüller E, Dichgans J. Disintegration and reorgan- J Nerv Ment Dis. 1949;107:42–47.
ization of cortical motor processing in two patients with cerebel- 170. Krump JE. Modifications electroencephalographiques des mal-
lar stroke. Electroencephalogr Clin Neurophysiol. 1996;98:59–68. adies du collagene. Rev Neurol (Paris). 1955;92:593–607.
149. Roseman E, Bloom BM, Schmidt RP. The electroencephalo- 171. Stein J, Stava Z. Electroencefalogram u sklerodermie. Czechoslov
gram in intracranial aneurysms. Neurology (Minneapolis). 1951;1: Derm. 1961;36:501–512 (Quoted after Gibbs and Gibbs, 1964).
25–38. 172. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 3. 2nd
150. Christian W. Klinische Elektroenzephalographie. 2nd ed. Stuttgart: ed. Reading, MA: Addison-Wesley; 1964.
Thieme; 1975. 173. Finn R, Kamaldeen S, Moona G, et al. The EEG in systemic lupus
151. Daly DD, Markand ON. Focal brain lesions. In: Duly DD, Pedley erythematosus. Electroencephalogr Clin Neurophysiol. 1980;49:91P.
TA, eds. Current Practice of Clinical Electroencephalography. 174. Kogeorgos J, Scott DF. Neuropsychiatric and EEG features in 74
2nd ed. New York, NY: Raven Press; 1990:335–370. cases of systemic lupus erythematosus with cerebral involvement
152. Logar C, Enge S, Körner EM, et al. The value of EEG in subarach- [abstract]. Electroencephalogr Clin Neurophysiol. 1982;53:1P.
noidal haemorrhagia. EEG EMG Z Elektroenzephalogr Elektromyogr 175. Gottwald W, Sturm U. EEG findings in lupus erythematosus.
Verwandte Geb. 1982;13:68–72. Electroencephalogr Clin Neurophysiol. 1984;57:34P–35P.
374 Part III ■ Clinical EEG: General

176. Molaie M, Fariello RG. EEG detection of cerebral involvement in Cerebral Anoxia and the Electroencephalogram. Springfield, IL:
Wegener granulomatosis [abstract]. Electroencephalogr Clin Thomas; 1961:578–589.
Neurophysiol. 1981;51:37P. 191. Beaussart M, Dupuis D, Dupuis R. Electroencephalographic stud-
177. Hwang P, Andermann F, Metrakos K, et al. The seizures of ies of cyanotic congenital heart disease. In: Gastaut H, Meyer JS,
morphea [abstract]. Electroencephalogr Clin Neurophysiol. 1981; eds. Cerebral Anoxia and the Electroencephalogram. Springfield, IL:
52:47P. Thomas; 1961.
178. Alexander EL, Alexander GE. Aseptic meningoencephalitis in 192. Lesny I, Bor I, Vlach V. EEG changes in children suffering from con-
primary Sjögren syndrome. Neurology. 1983;33:593–598. genital heart disease: influence of O2 inhalation. Electroencephalogr
179. Caselli RJ, Scheithauer BW, Bowles CA, et al. The treatable Clin Neurophysiol. 1961;13:173–179.
dementia of Sjögren syndrome. Ann Neurol. 1991;30:98–101. 193. Bekeny G, Horänyi B, Peter A, et al. Elektroenzephalographische
180. Bernsmeier A, Held K. Thromboangiitis obliterans cerebri. In: Untersuchungen bei kongenitalen Herzfehlern. In: Jenenser EEG-
Vinken PJ, Bruyn GW, eds. Handbook of Neurology. Vol 11. Symposion. 30 Jahre Elektroenzephalographie. Berlin: VER Verlag
Amsterdam: North-Holland; 1972:292–326; Vol 12: 384–397. Volk und Gesundheit; 1963:194–200.
181. Nishimoto A, Takeuchi S. Moyamoya disease. Abnormal arterial 194. Kalyanaraman K, Niedermeyer E, Rowe R, et al. The electroen-
networks in the cerebral basal region. In: Vinken PJ, Bruyn GW, cephalogram in congenital heart disease. Arch Neurol (Chicago).
eds. Handbook of Neurology. Vol 12. Amsterdam: North-Holland; 1968;18:98–106.
1972:352–383. 195. Niedermeyer E. The EEG in cardiac diseases. In: Remond A, Ed.-
182. Ohyama H, Niizuma H, Kinjo T, et al. Changes of EEG and tissue in-chief. Handbook of Electroencephalography and Clinical
pCO2 with hyperventilation in children with Moyamoya disease Neurophysiology. Vol 14A. Amsterdam: Elsevier; 1972:65–67.
[abstract]. Electroencephalogr Clin Neurophysiol. 1983;56:55P. 196. Obrist WD, Bissell LF. The electroencephalogram of aged
183. Cravioto H, Feigin I. Non-infectious granulomatous angiitis with patients with cardiac and cerebral vascular disease. J Gerontol.
a predilection for the nervous system. Neurology (Minneapolis). 1955;10:315–330.
1959;9:599–609. 197. Krump JE. Die Lebenswandlung des Elektroenzephalogramms
184. Kolodny EH, Reibeiz JJ, Caviness VS, et al. Granulomatous bei Herz-und Kreislaufkranken. Verh Dtsch Ges Kreislaufforsch.
angiitis of the central nervous system. Arch Neurol. 1968;19: 1958;24:258–265.
510–524. 198. Davidson LAG, Jefferson JM. Electroencephalographic studies in
185. Hill FS, Hughes JG, Davis BC. Electroencephalographic findings respiratory failure. Br Med J. 1959;2:296.
in sickle cell anemia. Pediatrics. 1950;6:277–285. 199. Goulon M, Picidalo JJ, Christophe M, et al. Clinical, electroen-
186. Neidengard L, Niedermeyer E. The electroencephalogram in neuro- cephalographic and biological correlations in 34 cases of chronic
logical complications of sickle cell anemia (SS-hemoglobinopathy). bronchopneumopathy with asphyxia. In: Gastaut H, Meyer JS,
Clin Electroencephalogr. 1975;6:68–74. eds. Cerebral Anoxia and the Electroencephalogram. Springfield,
187. Tyler HR, Clark DB. Incidence of neurological complications in IL: Thomas; 1961:565–577.
congenital heart disease. Arch Neurol Psychiatry. 1957;77:17–22. 200. Subbotnik SI, Feinberg YS, Spielberg PJ. Electroencephalography
188. Losev JE. Kocenke funkcionajnogo softojanija kory golovnogo in paroxysmal tachycardia. Electroencephalogr Clin Neurophysiol.
mozga ditej a vrozdenymi serdca osnovanij elektroencefalo- 1955;7:577–584.
graficeskich issledovanij. Vop Okhrany Materin Dets. 1958;34: 201. Tucker JS, Yoe RH. Simultaneous EEG-EKG recording: study of a
66–75 (Quoted after Lesny et al., 1961). case with complete heart block and ventricular tachycardia.
189. Shev EE, Robinson SJ. Electroencephalographic findings associ- Electroencephalogr Clin Neurophysiol. 1956;8:129–132.
ated with congenital heart disease. Preliminary report. Electroen- 202. Walsh G, Masland W, Goldensohn ES. Paroxysmal cerebral dis-
cephalogr Clin Neurophysiol. 1958;10:253–258. charge associated with atrial tachycardia [abstract]. Electroen-
190. Shev EE, Robinson SJ. Electroencephalographic findings associ- cephalogr Clin Neurophysiol. 1968;24:187.
ated with congenital heart disease. In: Gastaut H, Meyer JS, eds. 203. Broser F. Die cerebralen vegetativen Anfalle. Berlin: Springer; 1958.
CHAPTER

Dementia and EEG


CORNELIS JAN STAM 20
INTRODUCTION positive arguments in favor of specific diseases causing demen-
tia such as AD, frontotemporal lobar dementia (FTLD), vascular
Only a few years after his first report on the human electroen- dementia (VaD), and so on (11,12). In a recent review on the
cephalogram (EEG), Hans Berger described EEG abnormalities usefulness of EEG and ERPs as biomarkers Jackson and Snyder
in dementia, including an autopsy-proven case of Alzheimer indicate that they anticipate expanded application of quantita-
disease (AD) (1,2). Since then, over 2000 research papers and tive EEG (qEEG) as a reliable and sensitive biomarker of mild
more than 200 reviews have been published on the EEG in cognitive impairment (MCI) and AD (13).
dementia. According to DSM IV criteria, dementia is a syn- In this chapter we will give an overview of the literature on
drome characterized by multiple cognitive deficits that cause EEG in dementia. This overview cannot be complete, in view of
impairment in social or occupational functioning and represent the huge number of published research papers; instead we will
a significant decline from a previous level of functioning (3). present a selection of the most important studies, with an
Dementia has now become a major health problem, especially emphasis on more recent investigations that make use of cur-
in the aged Western population. The prevalence of dementia in rent diagnostic criteria and proper statistical methods. First, the
the Western population in those aged over 65 has been esti- most important EEG changes in normal aging will be discussed.
mated between 3.6% and 10.3% (4). In 50% to 60% of patients This topic is important to gain a proper understanding of the
dementia is due to AD (4). In Europe alone, 5 million people borderline between normal changes due to aging and the earli-
are suffering from dementia (5). est signs of brain pathology in MCI and early AD. EEG changes
In view of the importance of dementia as a health problem in MCI, which is in many respects a transitional phase between
and the large number of studies that have been conducted, one normal aging and early dementia, will be discussed separately.
might expect that the role of the EEG in dementia is now well Next, EEG changes in the major categories of dementia will be
defined. Surprisingly, this turns out not to be the case. Despite described. Emphasis will be given to the most common causes
recent efforts at evidence-based guidelines for the diagnosis of of dementia such as AD, VaD, dementia with Lewy bodies
dementia, opinions on the usefulness of the EEG still vary con- (DLB), and FTLD. Rare causes of dementia that are particularly
siderably. According to some authors, the usefulness of the EEG relevant for EEG such as Creutzfeldt–Jakob disease will also be
is very limited and should be restricted to excluding nonconvul- described. The practical use of the EEG in the work up of
sive status epilepticus or supporting a diagnosis of sporadic patients with cognitive complaints will be considered taking
Creutzfeldt–Jakob disease (6). In an American guideline the into account the objective data from the literature as well as the
EEG is not even considered as an option (7). In contrast, other controversies between various guidelines.
authors have advocated a more widespread use of the EEG in
dementia (8,9). Summarizing the EEG literature up to 1996,
Jonkman reached the following three conclusions: (i) there
EEG CHANGES IN NORMAL AGING
remains a need for a laboratory technique to confirm the diag- Compared to the extensive EEG changes that occur in the ear-
nosis since a 100% accuracy cannot be obtained by clinical liest years of life, reflecting developmental processes in the
methods only; (ii) the sensitivity of the EEG is higher or equal brain, the EEG remains relatively stable after the age of 18.
to the best other laboratory techniques; (iii) all AD patients However, EEG changes do occur with normal aging, especially
should be studied at least once by EEG as well as by MRI. after the age of 60, and it is important to delineate these from
In a recent European guideline, the possibility that the EEG the earliest signs of pathology. EEG changes with normal aging
could be considered not simply as a tool to exclude rare condi- have been described extensively in a classic review by Klass and
tions but also as an adjunct in the diagnosis of specific causes of Brenner (14). A more recent review has been published by
dementia is considered (5). Compared to the 2001 USA guide- Rossini et al. (15). One problem confronting EEG studies in the
line, the 2007 European guideline reflects the tendency to move elderly is the definition of “normal.” On the one hand, “normal”
away from a purely exclusionary approach to dementia diagno- can be defined in a statistical sense as referring to what is most
sis that dominated the older literature (10). Currently, there is a prevalent in a population. A consequence of this approach
tendency to use laboratory investigations such as brain imaging, could be that EEG findings will be biased by the presence of as
cerebrovascular fluid markers, and the EEG not just to exclude yet subclinical pathology in an elderly population. For example,
rare and possibly treatable causes of dementia but also to obtain in a study on 52 normal subjects aged 20 to 91 years,

375
376 Part III ■ Clinical EEG: General

Hartikainen et al. found abnormal EEGs in 21% (16). Such age of 85 years the alpha rhythm may slow to 7 to 8 Hz (6).
findings raise the question whether the EEG is very prone to However, even in the very old the alpha frequency is not
false-positive findings or whether a substantial percentage of expected to drop below 7.5 to 8 Hz (18). In addition to slowing,
subjects were suffering from subclinical pathology. the alpha rhythm in the elderly may be characterized by the
Alternatively, “normal” can be defined as complete absence of presence of a benign rhythmic subharmonic variant of the alpha
disease or “successful” aging. Taken to the extreme, this rhythm (19). This variant is characterized by rhythmic activity
approach might imply that the EEG of even the very old should in the theta or delta band, at a frequency that is a subharmonic
not be different from that of an 18 year old, what is, of course, of the alpha rhythm, with clear reactivity to eye opening. Often
not realistic. In practice it is not so easy to determine which def- traces of normal alpha can be found in the same record as well
inition of “normal” holds; it depends strongly on the inclusion (Fig. 20.1). Reports on alpha rhythm slowing are probably
criteria used in EEG studies of healthy elderly. However, the biased at least to some extent by the presence of subclinical
question what is normal remains important, and we will come pathology. In our view slowing of the alpha rhythm below 8 Hz,
back to it when appropriate. Here we will discuss age-related even in the very old, is more likely to reflect pathology than nor-
changes in the major EEG rhythms, as well as some borderline mal aging. In a single subject, slowing of the alpha peak fre-
phenomena such as focal temporal slow activity. quency over time by more than 1 Hz is also a sign of pathology.
The physiologic alpha rhythm of awake, healthy adults is Several studies have addressed changes in the amount and
characterized by a frequency range between 8 and 13 Hz, a pos- amplitude of the alpha rhythm with normal aging. A reduced
terior temporal and occipital dominance and a clear reactivity to percentage time alpha and reduced alpha reactivity were
eye opening. The characteristics of the alpha rhythm, and in par- reported by Markand (20). A reduction of alpha band power was
ticular the alpha peak frequency, are very stable properties also described by Hatikainen et al. (16). According to Babiloni et
within a person and have a strong genetic basis. Several of these al. the power decrease in healthy elderly subjects may be limited
characteristics have been reported to change with normal aging, to the lower alpha band of 8 to 10.5 Hz (21). The lower alpha
most notably the alpha peak frequency. A slowing of the alpha band has been associated with attentional processes, whereas the
frequency was described already in 1954 by Obrist (17). After the upper alpha band may be related to different types of cognitive

Figure 20.1 Male, 55 years. Alternation of normal alpha rhythm on the left and subharmonic slow variant of alpha rhythm
on the right. This can be a normal finding in elderly subjects. Calibration mark: vertical, 50 V; Horizontal, 1 second. Filter
settings: time constant 1 second. Low-pass filter, 35 Hz. Source derivation.
Chapter 20 ■ Dementia and EEG 377

processes, memory, and perceptual performance, but in different of BTTE. At present it cannot be excluded that focal temporal
ways and with different reactivities (22). In addition to a theta and delta may be very sensitive early biomarkers of tem-
decrease in power, the spatial distribution of EEG rhythms poral lobe dysfunction, either of vascular or degenerative ori-
becomes more uniform in the elderly (23). Even though the gin. The significant association between temporal focal
power of the alpha rhythm may decrease with aging, it should be abnormalities and MCI in a large cohort of memory clinic
stressed that this is a fairly nonspecific finding. The so-called patients would favor such a view (30). What would be helpful is
low-voltage EEG with amplitudes less than 20 V can be a nor- a simpler classification scheme of temporal abnormalities in the
mal variant (14). An exception is the low-voltage EEG in elderly, allowing for different grades of abnormalities and not
Huntington disease, which will be discussed later in this chapter. just a classification as benign and malignant.
Compared to the alpha rhythm, much less is known about Another phenomenon that can be encountered infrequently
changes in other rhythms with aging. The mu rhythm may be in the EEG of healthy elderly subjects is a type of rhythmic,
less prevalent in elderly (24). Slowing of the mu rhythm has not anteriorly dominant delta that occurs at the transition between
been the subject of separate studies but is probably in line with wakefulness and sleep. This activity resembles frontal intermit-
alpha rhythm changes. Differences in peak frequency between tent rhythmic delta activity (FIRDA) and has been referred to as
mu and alpha of more than 1 Hz should probably be considered “sleep onset FIRDA” (31). However, it should be stressed that
to be abnormal. Of interest, the alpha rhythm in AD can be FIRDA in the EEG of adults is always abnormal, whereas sleep
slowed with relative sparing of the mu rhythm. The amount of onset FIRDA is considered to be a benign variant. A proper
beta decreases in the elderly (16). Surprisingly, several reports evaluation depends on the context, including the state of wake-
indicate that elderly women have more beta than men (16,25). fulness as well as the presence of other EEG abnormalities.
This finding is suggestive of postmenopausal hormonal Pathologic FIRDA is unlikely to occur in the context of an oth-
changes, but there is no clear evidence for this conjecture. erwise entirely normal EEG. Also related to drowsiness is the
Quantitative analysis has shown that absolute power in delta phenomenon of “subclinical rhythmic EEG discharge of adults”
and theta bands decreases with normal aging (16). In the eldest (SREDA) (32). SREDA is characterized by paroxysms of rhyth-
group, delta power was related to subtle cognitive deficits and mic, sometimes sharply formed activity, mainly in the theta
cholinergic dysfunction. band. The paroxysms have a sudden onset and end, and the
A controversial issue is the interpretation of focal abnormal- activity does not show the evolution of amplitude or frequency
ities in the anterior temporal areas in the EEG of the elderly. that is typical of epileptic seizures. The distribution is either
Such abnormalities have been reported by several authors unilateral or bilateral, predominantly in the temporal areas.
(26–28). These abnormalities are highly prevalent, occurring in SREDA is not associated with any kind of brain pathology and
over one third of subjects over 60 years, often with a preference is considered to be an entirely benign variant.
for the left temporal areas (27). Intermittent temporal slowing
has been related to white matter hyperintensities on MRI (26). MILD COGNITIVE IMPAIRMENT
White matter hyperintensities in healthy elderly have also been
associated with decreased EEG coherence and cognitive abnor- A major trend in current studies on diagnosis in dementia is the
malities in another study (29). In a study in 27 healthy elderly emphasis on early diagnosis. Patients seek medical attention at
subjects with a mean age of 78 years (range: 65 to 83 years) a stage when there are complaints, possibly objective cognitive
Visser et al. describe temporal theta or delta in 16 subjects deficits, but not sufficient criteria for a diagnosis of dementia.
(59%) (28). These temporal EEG abnormalities were associated There is an urgent need for tools to be able to either reassure
with impaired word fluency and ventricular widening on CT. these patients that their complaints are innocent or to antici-
Klass and Brenner discuss the possible clinical significance of pate the advent of full-blown dementia reliably and take appro-
these temporal slow waves extensively (14). They propose to priate actions. The introduction of the concept of MCI reflects
distinguish between more malignant and more benign forms of these current trends. MCI is a condition characterized by objec-
temporal slow waves. They introduce the concept of “benign tive memory disturbances in the absence of other cognitive
temporal transients of the elderly” (BTTE) and give nine crite- deficits (33). Importantly, the objective cognitive deficits are
ria that have to be fulfilled for temporal slow waves in order to insufficient to interfere with social or occupational functioning.
be classified as BTTE. If the criteria are not fulfilled, the tempo- The original concept of MCI was based on memory complaints
ral slow waves should be considered pathologic and may indi- and could be called amnestic MCI. More recently, the concept
cate temporal, possibly vascular pathology. of MCI has been expanded to include nonamnestic as well as
The introduction of the concept of BTTE is a laudable multidomain types of MCI as well (12). In all cases, MCI is con-
attempt to reduce the number of false-positive findings in EEGs sidered to be a transitional phase between normal aging and
of elderly which would be likely to occur if all focal temporal dementia, characterized by complaints, objective deficits, but
slow waves would be classified as abnormal. However, the crite- no interference with daily functioning. MCI patients have a
ria for BTTE are somewhat complex and elaborate, and no clearly increased risk to develop dementia. The risk of develop-
information is available on their intra- and interrater reliability. ing dementia is 15% per year for a patient with MCI, compared
Also, it is not clear whether the association between focal tem- to 1% to 2% for the healthy elderly population (12). Although
poral theta or delta, cognitive dysfunction and structural MCI is clearly a risk factor for dementia, not all patients with
abnormalities does not hold for the supposedly benign category MCI become demented. One major challenge is to identify
378 Part III ■ Clinical EEG: General

those patients who will not go on to develop dementia at an sion in MCI (51). In the study of van der Hiele et al. increased
early stage. This is one of the important objectives of EEG stud- theta activity during eyes closed and less alpha reactivity during
ies in this field. eyes open and memory activation at baseline indicated lower
EEG changes in MCI subjects compared to healthy elderly global cognitive and executive performance at follow-up (52).
subjects are subtle but have been described in many studies. Prichep et al. showed that the EEG might even predict future
Several authors have reported an increase in theta power in MCI cognitive decline in subjects who have subjective memory com-
compared to healthy controls (34–37). Increased theta, but not plaints but do not even fulfill criteria for MCI (53). Baseline
alpha or beta, power correlates with hippocampal volume loss EEG in 44 normal elderly subjects with subjective complaints
(34,38). Jiang et al. found increased alpha and beta band power could predict future cognitive decline during an impressive 7-
in addition to increased theta power in MCI (39). EEG power year follow-up. Decliners were characterized by increased theta
was negatively correlated with MMSE scores. In the study of power, slowing of the mean frequency, and abnormal func-
Babiloni et al. the strength of delta and alpha 1 sources was tional connectivity especially in the right hemisphere in their
decreased in MCI subjects, and these changes also correlated baseline EEGs.
with MMSE changes (40). Surprisingly, lower alpha 1 source To conclude, EEG changes can be demonstrated in MCI, and
power was negatively associated with vascular abnormalities on some of these changes may correlate with structural changes
MRI (41). Liedorp et al. reported a significant association (notably hippocampal atrophy), cognitive deficits, and an
between focal abnormalities and MCI (30). In contrast, a com- increased risk of conversion to dementia. The most consistent
bination of focal and diffuse abnormalities showed a negative finding seems to be an increase in theta power, in particular in
association with MCI and rather favored a diagnosis of AD, VaD, the anterior temporal areas. Studies during cognitive tasks or
or DLB. employing measures of functional connectivity show less con-
Some studies have attempted to increase the sensitivity by sistent results, possibly due to unknown compensation effects.
recording the EEG during tasks or by considering measures of Many authors claim that the EEG can predict conversion to
functional connectivity. The notion of “functional connectiv- dementia in MCI, but there is no agreement on which EEG
ity” refers to statistical interdependencies between measures of measure is most important. In addition to increased theta
brain activity (such as EEG, MEG, and BOLD) recorded over power, slowing of the alpha rhythm and decreased alpha power
different brain regions; such correlation might reflect func- may be important. So far, many of these studies are mainly con-
tional interactions taking place between those brain regions cerned with predictions at the group level. It remains to be
(42). Pijnenburg et al. showed that during a working memory shown that reliable predictions can be made at the level of indi-
task the synchronization likelihood, a general measure of cor- vidual patients in prospective studies.
relations between signals, was significantly higher in MCI
compared to the control subjects in the lower alpha band (8 to ALZHEIMER DISEASE
10 Hz) (43). The increased task-related connectivity might
suggest a compensatory mechanism. In another study involv- Alzheimer disease was first described by Alois Alzheimer in 1907
ing working memory tasks, the EEG coherences in all bands (54). It is the most frequent cause of dementia in the aging
were significantly higher in the MCI group in comparison with Western population, accounting for more than 50% to 60% per-
those in the control group (39). MCI patients showed less cent of all cases (4). Neuropathologically, AD is characterized by
alpha band reactivity during a picture memory task (44). neuronal loss, atrophy, deposition of amyloid beta 1–42 protein
Findings with respect to resting state functional connectivity in senile plaques, and neurofibrillary tangles composed of
seem to be less consistent. Some studies showed no difference abnormally phosphorylated tau protein. Different stages can be
between two groups at rest (39,45). In contrast Babiloni did distinguished in the neuropathology of AD, mainly depending
find a reduced frontoparietal alpha 1 band synchronization on the distribution of neurofibrillary tangles, which is limited to
likelihood in AD and MCI subjects, correlating with lower the hippocampus in early stages and spreads out to entorhinal
MMSE scores (46). and association cortex in later phases (55). Although the deposi-
An important question is whether the EEG changes in MCI tion of amyloid is often considered an important hallmark of the
predict conversion to dementia. Huang et al. found decreased disease (56), neurofibrillary tangles may be more closely related
alpha global field power and more anterior located theta, alpha, to disease symptoms. With the advent of new PET tracers it has
and beta sources in progressive as opposed to stable MCI (47). now become possible to trace the neuropathology of AD in vivo
In the study of Jelic et al. alpha and theta relative power and (57,58). 2-(1-(6-[(2-[18F]Fluoroethyl) (methyl)amino]-2-
mean frequency from left temporal occipital regions proved to naphthyl)ethylidene)malononitrile ([18F]FDDNP) is a tracer
be reliable predictors for conversion to AD (48). Progressive that binds to neurofibrillary tangles and beta-amyloid senile
MCI patients displayed lower event-related theta synchroniza- plaques. FDDNP binding has been shown to correlate with cog-
tion than stable MCI (49). In a study in 69 subjects fulfilling cri- nitive disturbances in AD patients (58). In contrast, Pittsburgh
teria for MCI, of whom 24 converted to AD, Rossini et al. Compound-B (PIB) that binds selectively to amyloid did not
showed increased midline coherence as well as stronger delta, correlate with cognitive dysfunction in AD (57). While the diag-
theta, and alpha 1 sources in converters compared to noncon- nostic usefulness of these new PET tracers remains to be eluci-
verters (50). In contrast, Luckhaus et al. reported an association dated, they present a major breakthrough in the direct
between decreased alpha activity at baseline and risk of conver- neuroimaging of the underlying pathology in AD.
Chapter 20 ■ Dementia and EEG 379

Clinically, AD is characterized by early memory loss, in par- by a relatively nonspecific, stage-dependent pattern of diffuse
ticular impaired encoding of new material. Later, cortical func- slowing (Fig. 20.2). In the earliest phase of the disease the EEG
tions become disrupted resulting in aphasia, apraxia, and can be normal, especially in late onset cases (14). However, in
agnosia. Psychiatric and behavioral problems are also common. general, a normal EEG argues against a diagnosis of AD (30).
Average duration of survival after diagnosis in AD is 5 to 8 Power decreases in the beta and alpha band and increases in the
years (59). A diagnosis of AD depends mainly on clinical find- theta and delta band (63–68). Alpha peak frequency decreases
ings, with neuropathologic confirmation as a gold standard. and reactivity of the alpha rhythm to eye opening also dimin-
However, the interrater agreement of a diagnosis of AD is only ishes (68). Increase in theta and decrease in beta power are
moderate with a kappa of 0.63 (60). Currently, the most probably the first and most sensitive EEG changes in AD
important research criteria for AD are the NINCDS-ADRDA (10,66,69,70). The increase in theta may be more outspoken in
criteria (61). According to Waldemar et al. these criteria have a early onset AD (71,72). A slowing of the alpha peak frequency
sensitivity of 81% and a specificity of 70% across studies with occurs only after the increase in theta (70). Later alpha power
postmortem confirmation (5). Even the autopsy findings may decreases and finally in a relatively late stage delta increases. The
not constitute a straightforward gold standard: kappa scores increase in slow activity is most prominent over left temporal
for neuropathologic interrater reliability vary between 0.65 areas (65,73). Furthermore sources of alpha and beta display a
and 0.59 depending on criteria used (62). The modest agree- shift toward more frontal regions in AD (74). An increase in
ment between clinical diagnostic criteria and neuropathologic gamma band power has been reported in AD during rest and
findings illustrates the need for sensitive, specific, and reliable cognitive tasks, but it is questionable whether EEG activity
biomarkers for AD. However, they may also point to more fun- above 20 Hz can be reliably distinguished from muscle artefact
damental limitations in our concepts of disease entities. We (75,76). In the course of the disease, there is a further decrease
will return to this in our discussion of the clinical usefulness of of beta and alpha power, slowing of the alpha peak frequency,
the EEG. and a further increase in theta and delta power (66,77). Focal
EEG changes in AD were already described by Hans Berger (1). abnormalities are not typical and may be related to the frequent
There is general agreement that the EEG in AD is characterized occurrence of vascular pathology in AD. FIRDA is rare and can

Figure 20.2 Male, 87 years. Alzheimer disease. Irregular, mildly slowed alpha rhythm, more pronounced at posterior tem-
poral areas and diminished reactivity to eye closure. Calibration mark: vertical, 50 V; Horizontal, 1 second. Filter settings:
time constant 1 second. Low-pass filter, 35 Hz. Source derivation.
380 Part III ■ Clinical EEG: General

Figure 20.3 Female, 74 years. Mild cognitive impairment. Spike-wave complexes in the left temporal regions. Calibration
mark: vertical, 50 V; Horizontal, 1 second. Filter settings: time constant 1 second. Low-pass filter, 35 Hz. Source derivation.

be found in only 2.9% of EEGs in AD (78). Epileptiform abnor- left temporal region (69). Changes in the delta band are less evi-
malities are also rare, but AD is associated with a significantly dent, with a study reporting decreased coherence both inter-
increased risk of epilepsy, especially in early onset and severe and intrahemispherically (92) while another one (88) noted
AD (79). Temporal epileptiform activity may mimic AD increased coherence between frontal and posterior regions but
(80–82). An example can be found in Figure 20.3. restricted to a few AD patients.
EEG abnormalities pointing to abnormal functional interac- The normal parietal to frontal direction of information flow
tions between brain regions have been reported frequently in in the alpha and beta band is disrupted in AD patients (93).
AD. Clinical symptoms may be due to a considerable extent to Nonlinear measures also showed a reduced functional connec-
disrupted communication between distributed brain areas, jus- tivity in AD (42,45,94,95).
tifying a description of AD as a disconnection syndrome (83). In addition to spectral analysis and assessment of functional
Functional connectivity can be studied by computing linear coherence, several studies have explored advanced types of sig-
(such as coherence) or nonlinear (such as synchronization like- nal analysis in AD. Artificial neural networks are nonlinear clas-
lihood or mutual information) measures of correlation sifiers that can improve the distinction between EEGs recorded
between EEG signals recorded from different brain regions. A in healthy subjects and patients suffering from MCI or AD
decrease of EEG coherence in the alpha band in AD has been (96–98). Nonlinear EEG analysis attempts to reconstruct the
reported in many studies (69,84–90). Nonetheless van der Hiele dynamic processes in neural networks underlying the EEG
et al. reported no changes of alpha band coherence in AD, (99,100). Nonlinear analysis has been applied to the EEG in AD
although these authors found also that theta relative power was in many studies (4,101). Measures such as the correlation
increased in AD patients and this increase was related to dimension, the largest Lyapunov exponent, and the Kolmogorov
decreased performance in all cognitive domains (68). A lower entropy are used to characterize changes in the complexity of
coherence in the beta band has also been described (89,91). brain networks. In AD, several authors have reported a loss of
Several studies agree with respect to a decrease of coherence in complexity of brain dynamics (97,102–111). These changes may
the theta band interhemispherically (69,92) and to a decrease of correlate with the severity of clinical symptoms. However, it
coherence in the alpha band (92), more specifically in the tem- should be stressed that results from nonlinear EEG analysis may
poro–parieto–occipital region (88) and more focused on the be biased by more basic linear properties of the EEG (112).
Chapter 20 ■ Dementia and EEG 381

These properties can be measured more easily with spectral with hippocampal atrophy (131). Lower alpha band reactivity
analysis and coherence and may depend on the frequency band correlates with decreased performance on tests of global cogni-
considered (113,114). More recently, complex brain networks tion, memory, and executive functioning; brain atrophy corre-
have been characterized with graph theory (115,116). While lates with psychomotor speed (68,132). Loss of coherence in the
healthy subjects have brain networks with the so-called small- fast frequencies relates to impaired performance on neuropsy-
world topology, characterized by a combination of high cluster- chologic tests (36,133). More specifically, Dunkin et al. found
ing and short path lengths, AD patients may have more random correlations between beta band coherence between right tem-
brain networks due to loss of critical communication lines porooccipital electrodes (P4, T4, T6) and scores on the Boston
(117–120). Naming task, and a correlation between scores on block design
The diagnosistic accuracy of EEG changes, based on measures and fascicle coherence (defined, for left and right hemisphere,
of EEG slowing or disturbed functional connectivity, fluctuates as long distance coherence between P3/4 and F3/4, F1/2 and
around 80%. Summarizing 16 studies published between 1983 F7/8) (133). Lower beta band (14 to 22 Hz) mean synchroniza-
and 1995, Jonkman reports a total accuracy between 54% and tion likelihood (averaged over all pairs of electrodes) correlates
100% with a median of 81% (8). An accuracy of 100% was with lower MMSE scores in MCI and AD patients (45). Loss of
obtained in several studies of the EEG during random eye interhemispheric coherence in AD correlates with atrophy of
movement (REM) sleep (121–123). In one of the few studies the corpus callosum (89). Loss of global field synchronization
relating EEG abnormalities to the gold standard of neu- (an average measure of coupling between all brain regions) in
ropathology, Robinson et al. showed that 87% of patients with AD correlates with MMSE and CDR scores (91).
an autopsy-proven diagnosis of AD had an abnormal EEG EEG changes in AD can predict to a certain extent the fur-
(124). However, in the matching control group 35% had abnor- ther course of the disease. In general, EEG slowing already pres-
mal EEGs. Strijers et al. reported an accuracy of 84% in distin- ent at the time of initial diagnosis predicts a poor outcome
guishing between controls and AD patients using either EEG or (8,134). Helkala et al. showed that AD patients with more severe
hippocampus atrophy (125). An earlier study reported a sensi- EEG abnormalities as compared to those with relatively normal
tivity of 85% and a specificity of 88% of medial temporal atro- EEGs had a worse prognosis with more severe neurologic
phy for the detection of mild-to-moderate AD: 85% and 88% abnormalities and a greater risk of institutionalization later in
(126). Using visual analysis and a semiquantitative rating, the the disease (135). Claus et al. showed that slowing on qEEG pre-
grand total EEG (GTE) score, Claus et al. related the diagnostic dicts cognitive and functional decline in AD (136). In a later
gain of the EEG to the a priori likelihood of a diagnosis of AD study, the authors showed that mortality in patients with early
(127). They showed that in the case of an abnormal EEG a diag- AD was predicted by a decrease in alpha and beta activity deter-
nostic gain could be obtained when the a priori likelihood of mined by spectral analysis of the EEG (137). Delta and alpha
AD was between 30% and 40%. Few studies have directly com- power could predict loss of activities of daily life and inconti-
pared the accuracy of visual and quantitative analysis nence in the study of Nobili et al. (138).
(128–130). These older studies suggest a comparable accuracy EEG changes in AD may provide some understanding of the
between 75% and 80% for visual and computer analysis. Adler genetic and other factors involved in the pathophysiology of this
et al. reported that left temporal alpha coherence and global disorder apart from their diagnostic and prognostic meaning.
theta power allowed an identification of AD patients with a sen- Purely genetic causes of AD are rare. Genetic changes in genes
sitivity of 87% and a specificity of 77% (69). for amyloid beta precursor protein, presenilin 1, and presenilin
We can conclude that the test characteristics of the EEG do 2 account for less than 5% of AD cases (12). Most cases of AD
not compare unfavorably with those of some other biomarkers are sporadic, but genetic risk factors are important. This is espe-
(12). In addition it should be noted that these results can be cially relevant in the case of the epsilon 4 (e4) allele of the
obtained with relatively simple semiquantitative visual assess- apolipoprotein E (ApoE) gene. The ApoE e4 allele is a genetic
ment of the EEG (127). Furthermore, compared to other labo- risk modifier for AD (139). Considering the fact that ApoE e4 is
ratory studies, the EEG is relatively cheap, generally available, a risk factor for AD and that EEG changes such as increased
and noninvasive. However, EEG studies are confronted with the theta and loss of functional connectivity could be early bio-
same problem as studies of other biomarkers, namely the prob- markers of AD pathology, it is interesting to study the possible
lems related to the lack of a good gold standard. We will come relationship between genetics and EEG markers. Lehtovirta et al.
back to this issue in a discussion of the practical use of the EEG showed that AD patients with ApoE e4 genotype had more pro-
in dementia diagnosis. nounced slow wave activity (140–142). In contrast, Jelic et al.
Both EEG slowing and disturbed functional connectivity could not confirm the relation between ApoE e4 and EEG slow-
can correlate with severity of dementia and cognitive deficits, as ing but did report a decreased functional connectivity in AD
well as structural changes. EEG slowing correlates with ventric- patients with the e4 allele (143). Kramer et al. used the synchro-
ular widening, cortical atrophy, and worse psychometric per- nization likelihood to determine the relation between ApoE
formance in AD (67). An anterior shift of alpha and beta genotype and functional connectivity in subjects with subjective
sources, and stronger sources in the low-frequency bands corre- memory complaints and AD patients (144). In this study sub-
late with severity of dementia (74). Delta power correlates neg- jects with ApoE e4 had higher synchronization in the lower and
atively and alpha 1 power positively with the AD patient’s upper alpha band than subjects without ApoE e4, irrespective
MMSE scores (65). Decreased cortical alpha power correlates of diagnosis. Compared to controls, AD patients had lower
382 Part III ■ Clinical EEG: General

synchronization in the upper alpha and beta band. This study onset, or a relatively strong EEG change after a single test dose
suggests that both the e4 allele as well as AD pathology influence of the drug, may predict long-term response to cholinesterase
functional connectivity, but they do so in opposite directions inhibitors (156,159–161).
and partly in different frequency bands. In a large study involv- Obviously, the cholinergic hypothesis of AD has received a
ing 145 individuals with AD, their unaffected relatives and unre- lot of attention in AD in view of its therapeutical implications.
lated individuals, Ponomareva et al. showed that AD patients However, treatment with cholinesterase inhibitors is only mod-
with the ApoE e4 allele had a stronger loss of alpha power than erately effective in a subset of patients. This suggests that other
e4 noncarriers (145). The resting EEG of nondemented relatives factors than a loss of central cholinergic activity may be impor-
of AD patients showed no differences related to ApoE genotype. tant. For instance, receptors may be downregulated or even dis-
During hyperventilation, however, only e4 allele carriers showed appear from the membranes, and if there is neuronal death this
synchronous high-voltage slow and sharp waves. This excessive will certainly lead to a decrease in targets of cholinergic inputs.
synchronization is somewhat reminiscent of the increased syn- According to Dringenberg other neurotransmitters in addition
chronization reported by Kramer et al. but does not agree with to acetylcholine have to be considered (162). Changes in several
the loss of coherence found by Jelic et al. (143,144). Apparently, different neurotransmitters could perhaps explain the global
the relation between ApoE genotype and EEG is complex and cortical hyperexcitability of the cortex in AD while the brain is
depends on the study conditions, the EEG measure investigated, at rest (163). In the earliest phase of AD there may be no evi-
and the presence or absence of AD pathology. dence of cholinergic dysfunction (164). In MCI the cholinergic
In contrast, the relation between cholinergic loss and EEG system may even by upregulated, possibly as a compensatory
changes in AD seems to be more straightforward. There is con- mechanism (143). A relation between EEG changes and amy-
siderable evidence for a loss of central cholinergic function in loid beta1–42 is suggested by findings in patients with Down
AD (146,147). This cholinergic loss is presumably due to a loss syndrome. In Down syndrome, an extra chromosome 21 is
of neurons in the nucleus basalis of Meynert, which projects to present. The gene for amyloid precursor protein lies on chro-
medial temporal and neocortical brain areas (146). Impaired mosome 21, and this has been related to the high risk of early
cholinergic function in AD is held responsible for at least some onset of AD in Down patients. In line with this concept, AD in
of the cognitive deficits in this disorder. This concept is known Down is predicted by EEG slowing (165). Furthermore, EEG
as the “cholinergic hypothesis” (148). The cholinergic hypothe- slowing correlates with AD pathology in Down. Another
sis is of considerable interest, since it suggests a possible way to important factor in AD pathology is the abnormally phospho-
treat AD symptoms by activating the central cholinergic system. rylated tau protein. Jelic et al. have reported a correlation
This could be achieved by administration of drugs that inhibit between tau protein in the cerebrospinal fluid and the
the breakdown of acetylcholine by acetylcholine esterase. alpha/delta ratio of the EEG in AD patients (166). Babiloni et al.
Several acetylcholine esterase inhibitors such as tacrine, have shown that homocysteine levels, related to vascular
donepezil, rivastigmine, and galantamine have been used clini- pathology, are related to EEG slowing in mild AD (167). In gen-
cally in AD, with varying results. eral, the simultaneous occurrence of AD and vascular pathol-
Cholinergic loss could explain EEG changes in AD. In an ani- ogy may be more important than has been realized in the past.
mal study, Buzsaki et al. showed that damage to the nucleus According to Schreiter-Gasser et al. the presence of vascular
basalis results in a slowing of the EEG (149). In another animal pathology could magnify the effect of AD pathology on demen-
study, Villa et al. showed that loss of acetylcholine results in an tia severity (168).
increase in low-frequency coupling and a decrease in high-fre-
quency coupling (150). These experimental findings might FRONTOTEMPORAL LOBAR DEMENTIAS
explain why alpha and beta band connectivity are lost in AD,
whereas low-frequency coupling may be spared or even Originally, frontal lobe dementia was synonymous with Pick
increased. Administration of scopolamine, which blocks central disease, a rare degenerative disorder characterized by typical
cholinergic activity, to healthy subjects decreases EEG inter- neuronal inclusions bodies (Pick bodies). Currently, the con-
hemispheric coherence in the delta and beta-1 bands in the rest- cept has been broadened and includes degenerative frontal
ing state. (151). Increase in delta and theta and decrease of alpha dementia with or without Pick bodies, progressive aphasia, and
and beta after scopolamine administration have also been semantic dementia. This spectrum of disorders is referred to as
reported in healthy subjects (152). Scopolamine induces a pat- FTLD. FTLD is characterized by early behavioral and language
tern of memory and cognitive deficits in healthy subjects that problems, frontal-type cognitive dysfunction, and relatively late
resembles the changes in early AD (153). The general slowing of occurrence of memory complaints. In the early phase, FTLD is
the EEG in AD has been interpreted as a cholinergic effect (154). often confused with psychiatric disorders, including personality
If we assume that EEG slowing in AD is due to cholinergic disorders and depression.
loss, we might expect that treatment with drugs that stimulate In view of the frontotemporal dominance of the pathology
the central cholinergic system will reverse the EEG slowing. in FTLD, it can be expected that the EEG and in particular the
Furthermore, EEG changes might predict which patients will posterior alpha rhythm remains normal until late in the disease.
show the best response to treatment. Some evidence for both Several studies have reported normal or only mildly disturbed
conjectures is available. EEG slowing in AD is reversed by treat- EEGs in FTLD (169–172). The EEG in FTLD may show signif-
ment with cholinesterase inhibitors (155–158). EEG slowing at icantly less slowing than in AD (171,173,174). Functional
Chapter 20 ■ Dementia and EEG 383

connectivity, as measured with the synchronization likelihood, authors (78,187–189). Using a semiquantitative visual analysis
is lower in FTLD compared to AD; FTLD values are compara- (GTE) Roks et al. could distinguish between DLB and AD with
ble to nondemented controls (174). EEG coherence in FTLD a sensitivity of 72% and a specificity of 85% at a GTE cutoff of
was also comparable to controls (173). Using qEEG analysis, 9.5. Kai et al. showed increased delta and theta power as well as
Yener et al. reported EEG abnormalities in FTLD patients (175). intrahemispheric coherence in DLB as compared to AD (189).
On the basis of these findings FTLD patients could be distin- AD patients had less beta power mainly in the posterior
guished from both AD patients as well as healthy controls. More regions. In DLB patients treatment with the acetylcholine
recently in a study with neuropathologic confirmation of diag- esterase inhibitor donepezil resulted in a power decrease in
nosis, Chan et al. showed that EEGs of FTLD patients could not delta and theta bands but a similar response was not present in
be distinguished from EEGs of AD patients; however, a very AD patients. Increased delta band power variability, increased
simple visual classification scheme was used in this study and delta coherence, and decreased alpha coherence were demon-
no comparison was made with EEGs of age-matched nonde- strated in DLB compared to AD and elderly controls by
mented controls (176). The discrepancies between the various Andersson et al. (190). Bonanni et al. studied 50 DLB, 50 AD,
studies could be due to variability of the neuropathologic pat- and 40 PDD patients with relatively MCI; diagnosis was con-
tern of degeneration. In a study with neuropathologic confir- firmed in most patients after a 2-year follow-up (187). DLB
mation of the diagnosis, Passant et al. showed that onset before patients had a slower and more variable dominant frequency
65 years is associated with behavioral–mood changes and mild compared to AD patients. Only half of the PDD patients had
EEG abnormalities, while onset after 65 years is related to lan- EEG abnormalities comparable to DLB patients. The authors
guage disturbances and a normal EEG (177). conclude that the EEG may be useful to distinguish between AD
and DLB. This conclusion is in line with some of the other stud-
ies that report more extensive slowing and a higher prevalence
PARKINSON DISEASE WITH of FIRDA in DLB. Furthermore, such a distinction can be made
AND WITHOUT DEMENTIA: with relatively high sensitivity and specificity using simple
DEMENTIA WITH LEWY BODIES visual assessment of the EEG.

Next to AD, Parkinson disease is one of the most prevalent neu-


rodegenerative disorders. Parkinson disease is a multisystem HUNTINGTON DISEASE
disorder characterized by motor symptoms such as akinesia, Huntington disease is a rare neurodegenerative disorder with
rigidity, tremor and postural instability, cognitive and neu- an autosomal-dominant inheritance. It is caused by multiple
ropsychiatric problems. Mild executive cognitive dysfunction CAG repeats of the Huntington gene on chromosome 4. It is
can occur early in the disease, while full-blown dementia is a characterized by severe dementia, psychiatric disturbances, and
frequent complication in the later stages of the disease. A dis- involuntary movements, notably chorea. Neuropathologically
tinction has been made between Parkinson disease with there is conspicuous atrophy of the caudate nucleus. A charac-
dementia (PDD) and DLB, mainly based on the time interval teristic finding in up to 40% of Huntington patients is the
between onset of extrapyramidal and cognitive symptoms; this occurrence of a low-voltage EEG (191). Streletz et al. reported
distinction may be a bit artificial, and in practice considerable increased theta and decreased alpha in Huntington patients
overlap can be expected between PDD and DLB. (192). The EEG may show changes in nondemented mutation
In Parkinson disease without dementia (PD), the EEG is carriers for Huntington disease. van der Hiele et al. found less
probably relatively preserved, at least in the early stages. alpha band reactivity during memory activation in mutation
However, even in nondemented Parkinson patients a correla- carriers compared to nonmutation carriers (193). However,
tion between motor disability and slowing of the background compared to genetic testing the clinical significance of the EEG
activity has been found (178). EEG slowing emerges with pro- in Huntington disease is probably quite limited.
gressive cognitive disturbances (179). In contrast to PD, PDD is
often associated with a slowing and impaired reactivity of the
EEG (178,180,181). As in AD, EEG slowing in PDD is respon- PROGRESSIVE SUPRANUCLEAR PALSY
sive to treatment with rivastigmine (182). AND CORTICOBASAL GANGLIONIC
Extensive slowing of the EEG has also been reported in DLB DEGENERATION
(183). In the study of Liedorp et al. a normal EEG or an EEG
with only focal abnormalities argued strongly against a diagno- Progressive supranuclear palsy (PSP; also called Steele–
sis of DLB, whereas an EEG with both focal and diffuse abnor- Richardson–Olzewski syndrome) is another rare disease char-
malities strongly favored a diagnosis of DLB (30). Of interest, acterized by frontal-type cognitive dysfunction, dementia,
FIRDA may occur frequently in DLB, but not in AD extrapyramidal motor symptoms (in particular axial rigidity),
(78,183,184). In the study of Roks et al., FIRDA was found in and supranuclear vertical gaze palsy. Fowler et al. compared
2.9% of the patients with AD and in 33.3% of the patients with EEG findings in 22 patients with PSP with EEGs of 22 matched
DLB (78). Some authors report comparable slowing of the EEG AD patients (194). PSP patients had EEGs with low voltage,
in DLB and AD (185,186). However, slowing of the EEG back- preserved alpha peak frequency, and modest increase of theta;
ground is more severe in DLB than in AD according to several one patient had FIRDA. Findings in the AD control group were
384 Part III ■ Clinical EEG: General

largely similar. Compared to PSP patients, patients with corti- Leuchter et al., which suggests that connections between areas
cobasal ganglionic degeneration (CBD) had more focal abnor- linked by long corticocortical fibers (see above) would be more
malities, often contralateral to the dominantly affected side and affected in AD (86,87). Quantitative but not visual analysis can
ipsilateral to the atrophy on MRI (195). In this study the preva- distinguish between patients with subcortical VaD with mild or
lence of FIRDA was relatively high in both disorders: 20% in moderate dementia (202). qEEG measures correlated signifi-
CBD and 35.7% in PSP. cantly with MMSE scores in this study. In a large study based
upon visual analysis, VaD was associated with the combination
VASCULAR DEMENTIA of diffuse and focal abnormalities; normal EEGs made a diag-
nosis of VaD less likely (30). Recent reviews agree that the EEG
The diagnosis of VaD depends on three conditions: (i) a can sometimes distinguish VaD from AD, but question the clin-
dementia syndrome; (ii) demonstration of vascular lesions in ical usefulness of this, since other laboratory investigation such
the brain; and (iii) evidence of a causal relation between the as MRI may be more effective in this respect (6,11). Again, the
vascular lesions and the dementia. Although both dementia and significant overlap between AD and VaD especially in the
vascular lesions are common, it is not at all easy to prove they elderly population should be stressed.
are causally related in a single patient. To complicate the issue,
vascular lesions are increasingly considered to be important in
AD as well. The NINDS-AIREN criteria emphasize the impor- DELIRIUM AND METABOLIC/ TOXIC
tance of radiologic evidence for vascular lesions (196). According ENCEPHALOPATHIES
to Holmes et al., these criteria have a sensitivity of 43% and a
specificity of 95% (197). Delirium is a clinical syndrome that can be caused by meta-
The contribution of the EEG to diagnosis and prognosis in bolic, toxic, or infectious encephalopathies, frequently in the
VaD is more limited than in AD. According to Jonkman, the lit- context of pre-existing neurodegenerative brain disorders such
erature up to 1996 shows no consensus on the ability of the as AD. A relation between EEG slowing and disturbed arousal
EEG to distinguish between AD and VaD (8). Several of the in delirious patients was already observed in the classic study by
studies discussed in this review did not use the NINDS-AIREN Romano and Engel (203). The sensitivity of the EEG to brain
criteria and depended on older concepts such as “multi-infarct dysfunction in delirium has been confirmed with quantitative
dementia.” In the studies of Erkinjuntti et al. degree of diffuse analysis (204). Delirious patients showed increased delta and
abnormalities/mean frequency background did not distinguish theta, and decreased alpha as well as a slowing of alpha peak fre-
between AD and MID (198,199). Leuchter et al. investigated quency; these changes were correlated with the severity of cog-
coherence between EEG signals recorded from brain areas nitive disturbances. EEG slowing in delirium is often
linked by two different types of connections: areas linked by nonspecific, making a distinction between delirium and AD
bands of long corticocortical fibers showed greatly diminished difficult; moreover, both conditions frequently occur together.
coherence in subjects with AD but not in MID patients. On the Even so, recent studies have attempted to increase the specificity
contrary areas linked by broad connective networks showed the of the EEG in this respect (205,206). Prolonged activity, consist-
largest decreases in coherence among MID subjects. These find- ing of EEG recording during 3 minutes with eyes open, signifi-
ings can be interpreted as implying that AD is a neocortical cantly improved the ability of the EEG to distinguish between
“disconnection syndrome” in which there is a loss of long cor- delirious patients with or without underlying dementia and
ticocortical tracts, whereas vascular disease (MID) most promi- cognitively unimpaired elderly (205). In another study, delir-
nently affects broad fiber networks that may be affected by ium in dementia was characterized by slower alpha peak fre-
diffuse subcortical vascular damage (86,87). According to quency, loss of alpha power, increased theta and delta; these
Rosen et al. the EEG in multi-infarct dementia displays changes correlated with severity of delirium and cognitive dis-
increased slow activity in combination with relatively preserved turbances (206).
posterior activity, whereas in AD slowing is combined with Other EEG patterns may suggest specific causes of delirium
abnormalities of posterior activity (171). Robinson et al. failed (6,14,207,208). Generalized triphasic waves are often associated
to find EEG differences between AD and VaD (124). with metabolic encephalopathy due to hepatic or renal disease
More recent studies often identify EEG differences between but can also occur in other conditions such as postanoxic states.
VaD and AD. Jeong et al. used nonlinear EEG analysis to com- In metabolic encephalopathies it is especially the rate of change
pare AD and VaD (200). AD patients showed a loss of complex- that determines the EEG abnormalities (6). These triphasic
ity reflected in lower values of the correlation dimension and waves are often responsive to benzodiazepine administration,
largest Lyapunov exponent, whereas complexity was pathologi- but this is not a proof of their epileptic origin. Low-voltage
cally increased in VaD (200). Individual analysis of alpha fre- records with excess fast activity can be found in withdrawal
quency and power could distinguish between AD and VaD in from alcohol or sedative drugs. Focal abnormalities should
the study of Moretti et al. (201). Frontoparietal synchronization always arouse suspicion of underlying structural lesions. In high
likelihood in the alpha 1 band was more reduced in VaD doses antidepressives and antipsychotics, in particular clozap-
patients than in AD in a study of Babiloni et al. (63). This more ine, often produce epileptiform abnormalities in addition to
pronounced loss of long distance connectivity in VaD patients slowing. Nonconvulsive status epilepticus is a rare but impor-
seems to be in disagreement with the coherence findings of tant and treatable cause of delirium; it often arises in elderly
Chapter 20 ■ Dementia and EEG 385

patients after sudden withdrawal of antiepileptic drugs. The Although CJD is a rare cause of dementia, it is considered an
EEG may be normal in alcohol dementia in subjects younger indication for EEG recording in view of the characteristic peri-
than 60 years but can show abnormalities including a low-volt- odic sharp wave complexes (PSWC). The EEG is an essential
age record in older subjects (209). In these subjects slowing of part of the diagnostic process in the WHO diagnostic classifica-
the EEG correlates with cortical atrophy. EEG changes in tion criteria of sporadic CJD (210). According to these criteria,
Wernicke–Korsakov syndrome may vary widely. a definite diagnosis of CJD requires neuropathologic confirma-
tion or demonstration of the abnormal prion protein. Probable
CREUTZFELDT–JAKOB DISEASE CJD requires (i) a rapidly (within 2 years) progressive demen-
tia; (ii) typical PSWC in the EEG and/or 14-3-3 protein in the
Creutzfeldt–Jakob disease (CJD) is a transmissible spongiform cerebrospinal fluid; and (iii) two of the following: (a)
encephalopathy caused by accumulation of abnormal isoform myoclonus, (b) ataxia and visual signs and symptoms, (c)
of host-encoded cellular prion protein. Different forms of CJD extrapyramidal and/or pyramidal signs and symptoms; and (d)
have been described: sporadic, iatrogenic, and variant. The new akinetic mutism. Possible CJD can be diagnosed if the above
variant of CJD is possibly related to bovine spongiform criteria are met with the exception of the EEG or 14-3-3 protein
encephalopathy. Iatrogenic CJD is caused by transmission of abnormalities.
infectious prion protein partials through tissue or medical EEG changes in CJD occur in different stages (211). In the first
instruments. Most forms are sporadic. Other prion disorders stage, the EEG shows nonspecific slowing and sometimes FIRDA.
are Kuru, familial fatal insomnia, and Gertmann–Straussler– In the middle stage, the EEG is characterized by PSWC in about
Scheinker syndrome. Clinically, CJD is characterized by a rap- two third of patients (Fig. 20.4). In the final stage, background
idly progressive dementia with myoclonus. Cerebellar, pyrami- activity between the complexes becomes increasingly flat, and
dal, and extrapyramidal motor symptoms as well as psychiatric finally the complexes may disappear. The PSWC may be localized
symptoms also occur frequently. Early visual symptoms are in the occipital regions in the Heidenhain variant of CJD. The
characteristic for the Heidenhain variant of CJD. PSWC disappear during sleep, sedation, and stimulation. Often a

Figure 20.4 Female, 61 years. Progressive cognitive disturbances, myoclonus, and impaired consciousness. Diagnosis:
Creutzfeldt–Jakob disease. The EEG shows generalized periodic sharp wave complexes (PSWC) with a repetition frequency
of slightly above 1/ sec. Calibration mark: vertical, 50 V; Horizontal, 1 second. Filter settings: time constant 1 second.
Low-pass filter, 35 Hz. Average reference.
386 Part III ■ Clinical EEG: General

clear time locking to clinical myoclonus is absent. If no PSWC are days. Since treatment has to start directly, the EEG does not play
seen after 12 weeks of disease onset, this argues against a diagno- a large role in initial diagnosis. The EEG may provide prognos-
sis of CJD. PSWC are not assumed to occur in variant CJD (212), tic information (6).
but exceptions to this rule have been described (213). In the ini-
tial stages PSWC can occur unilaterally (214). This raises the SUBACUTE SCLEROSING PANENCEPHALITIS
question whether PSWC are distinct from PLEDs (lateralized
periodic discharges or LPD according to the new terminology Subacute sclerosing panencephalitis (SSPE) is a neurodegener-
(215)). According to some authors PSWC and PLEDs may be dif- ative complication of measles infection in childhood (6). The
ferent phenomena, but the distinction seems to be a bit artificial EEG may show characteristic high-voltage generalized com-
(211,216). The detection of EEG abnormalities in suspected CJD plexes occurring in a periodic fashion with very long intervals
can be improved with advanced EEG analysis techniques such as (periodic long interval diffuse discharges [PLIDDs]); these
independent component analysis (ICA) (216,217). complexes have first been described by Radermecker and Poser
Strict criteria for PSWC in CJD were presented by Steinhof (227). The complexes are usually associated with myoclonic
et al. (218). To classify as PSWC the following criteria should be jerks. It should be pointed out that PLIDDs are not fully specific
met: (i) Strictly periodic cerebral potentials, the majority with a for SSPE, since they have also been described in phencyclidine
duration between 100 and 600 msec and an intercomplex inter- intoxication (228,229).
val between 500 and 2000 msec; (ii) generalized and lateralized
complexes accepted; and (iii) at least five repetitive intervals HUMAN IMMUNODEFICIENCY VIRUS
with a duration difference of less than 500 msec to rule out
semiperiodic activity. If these criteria are met, PSWC have a EEGs of 47 patients with the acquired immunodeficiency syn-
sensitivity of 64%, specificity of 91%, a positive predictive value drome (AIDS) or AIDS-related complex (ARC) were described
of 95%, and a negative predictive value of 49% (219). Zerr et al. by Gabuzda et al. (230). EEG abnormalities, related to demen-
reported a specificity of 74%, a positive predictive value of 93%, tia or opportunistic CNS infections, were found in 67% of
and a sensitivity of 66% for PSWC (220). These numbers patients with AIDS and 36% of patients with ARC. No EEG
should be compared to the 14-3-3 protein in the cerebrospinal abnormalities were described in a group of neuropsychologi-
fluid: sensitivity 84% and specificity 94% (220,221). In an cally unimpaired patients infected with HIV (231). In a study by
autopsy series of 201 patients suspected of CJD, van Everbroeck Harrison et al. patients with AIDS had more signs of neurologic
et al. found PSWC in 52% of autopsy-confirmed CJD cases, but dysfunction, cognitive problems, and EEG and MRI abnormali-
also in three patients with AD, one patient with DLB, and one ties than asymptomatic seropositive subjects (232).
patient with VaD (222). In general, autopsy-confirmed studies
indicate that PSWC have a fairly high specificity, but a rather CLINICAL USE OF EEG IN DEMENTIA
low sensitivity for CJD. This could be due to the fact that PSWC
develop later in the disease or may not appear at all in a sub- Following the review of EEG changes in the major categories of
stantial percentage of patients. Genetic forms only display dementia, we return to our initial questions: why are there such
PSWC in 10% of cases (223). Furthermore, PSWC are associ- large differences of opinion concerning the role of the EEG
ated with methionine homozygosity or methionine/valine het- between different dementia guidelines? What is the clinical use-
erozygosity at codon 129 of the prion protein on chromosome fulness of the EEG in dementia, and how should it be used? We
20, while PSWC hardly occur in CJD patients who are homozy- will address these questions and attempt to give some tentative
gotes for valine at codon 129 (6,211). answers.
No completely satisfactory explanation of PSWC in CJD Methodologic problems are one source of controversies
exists. According to Tschampa et al. a reduction of paralbumin- between various studies. Many studies, mainly but not exclu-
positive cells in the thalamic reticular nucleus could be responsi- sively the older ones, suffer from various problems such as
ble for both PSWC and myoclonus in CJD (224). These authors small group size; bad matching of groups in terms of age, gen-
stress that “functional integrity (at least in parts) of the subcor- der, and disease duration; and influence of centrally acting
ticocortical or thalamocortical network is a prerequisite for the drugs. Results may not be representative since they concern
generation of PSWC.” In agreement with this, modeling studies patients referred to secondary or tertiary referral centers for
have shown that typical periodic sharp waves can be reproduced dementia diagnosis. There is a lack of consensus on which EEG
in a model of the alpha rhythm assuming (i) an increased excita- measures to use and insufficient knowledge on the repro-
tion/inhibition balance and (ii) intact but abnormally active cor- ducibility of these measures. Studies that fail to reproduce ear-
ticothalamic loops (225,226). PSWC may reflect pathologic, lier findngs may be underrepresented in the literature due to
highly nonlinear oscillations in corticothalamic networks. publication bias. Inadequate statistical analysis hampers some
studies and could be responsible for the reporting of false-pos-
HERPES ENCEPHALITIS itive findings. Despite these methodologic problems, which are
not unique for EEG research in dementia, many excellent stud-
The EEG is always abnormal in acute encephalitis. Herpes sim- ies have been published, and our knowledge has increased con-
plex encephalitis typically shows periodic discharges, mainly siderably over the last decades. EEG findings in normal aging,
over the temporal lobes, but they may appear only after several MCI, and several major categories of dementia, as well as their
Chapter 20 ■ Dementia and EEG 387

possible pathophysiologic substrate, are now fairly well known. helpful in dementia diagnosis? The answer to this question can
Other problems may therefore be more important in translat- only be tentative since the proper kind of clinical studies simply
ing this knowledge to clinical practice. have not yet been conducted as indicated in the previous section.
One major problem is the lack of a good gold standard (12). First of all it should be stressed that the results of an EEG exam-
It is almost impossible to carry out a laboratory investigation if ination should also be considered within their clinical context,
there is no gold standard to serve as reference. Currently the that is, taking the other relevant information (clinical, neuropsy-
closest one can get to a gold standard in dementia research is a chological, radiologic) into account. Ideally this should be done
combination of clinical diagnosis and neuropathologic confir- in the context of a multidisciplinary meeting of all disciplines
mation. Standards of clinical diagnosis have improved consid- involved, but it may not be easy to do this outside specialized
erably over the past two decades with the introduction of memory clinics. Active participation of the clinical neurophysiol-
guidelines for several of the major diseases causing dementia. ogist in such multidisciplinary teams may greatly enhance the
Similar standards are being introduced for neuropathologic contribution of the EEG to the diagnostic process and may also
diagnosis. However, clinical diagnosis according to well-defined serve as an educational experience for all involved. Second, it is
guidelines correlates only moderately with neuropathologic possible to identify a number of clinical situations where the EEG
diagnosis; furthermore, neuropathologic confirmation is often could contribute to solving a real clinical problem: (i) differenti-
not available and neuropathologic diagnosis itself also has a ating between FTLD and early onset AD. In relative young
limited interrater reproducibility. Under these conditions it is patients the clinical spectrum of both disorders can overlap con-
almost impossible to determine reliably the sensitivity and siderably, AD patients presenting with behavioral problems, and
specificity of a laboratory test such as the EEG in distinguishing FTLD patients also having cognitive complaints. In our view, a
between the normal healthy state and various conditions that clearly abnormal EEG favors a diagnosis of early onset AD and
can give rise to dementia. One solution is to use information on argues strongly against FTLD (174); (ii) differentiating between
the further course of the disease to validate the initial diagnosis. AD and DLB. Again, the differential diagnosis can be difficult in
An excellent example of this can be found in the study of some patients and may have consequences for treatment (for
Bonanni et al. (187). What could also be helpful is to give up the instance, avoidance of neuroleptics in DLB). The EEG is likely to
idea of eternal platonic truths underlying all our observations. be more abnormal in DLB than in AD and may show specific
Concepts such as “Alzheimer disease” or “vascular dementia” or abnormalities such as FIRDA much more often in DLB than in
“dementia with Lewy bodies” are man-made attempts to detect AD (78); (iii) and differential diagnosis of delirium. Here the
regularities in our observations; our concepts can be enriched EEG can differentiate between organic and psychiatric condi-
and may even have to evolve by incorporating all clinical, neu- tions, suggest metabolic encephalopathy or withdrawal syn-
ropathologic, and laboratory information available into consis- dromes, and rule out nonconvulsive status epilepticus; (iv)
tent and useful entities. epilepsy-related cognitive disturbances. Temporal lobe epilepsy
The final and perhaps most important problem of EEG can be associated with cognitive disturbances, in particular mem-
studies in dementia may be that many of them seem to be ory complaints, and can mimic AD (80–82); (v) diagnosis of
addressing the wrong problem. If the purpose of the investiga- Creutzfeldt–Jakob disease according to WHO standards (211).
tion is to describe the pathophysiology of a clinical dementia
syndrome, it makes sense to compare age-matched healthy con-
trols to subjects with a specific disorder causing dementia. REFERENCES
However, if the goal is to determine the clinical usefulness of
the EEG in dementia, such a comparison is useless: distinguish- 1. Berger H. Uber das Elektrenkephalogramm des Menschen. Dritte
ing between a healthy person and a clearly demented patient is Miteilung. Arch Psychiatr Nervenkr. 1931;94:16–60.
2. Berger H. Uber das Elektrenkephalogramm des Menschen. Funfte
a trivial problem from a clinical point of view. Neither the EEG
Miteilung. Arch Psychiatr Nervenkr. 1932;98:231–254.
nor any other laboratory investigation is a test “to see if a 3. American Psychiatric Association. Diagnostic and Statistical
patient is demented.” To be clinically useful, a laboratory test Manual of Mental Sisorders. 4th ed. (DSM IV). Washington, DC:
has to give an answer to a clinical problem, and the answer has APA; 1994.
to have consequences for diagnosis, prognosis, and treatment. 4. Jeong J. EEG dynamics in patients with Alzheimer’s disease. Clin
Ultimately it has to be demonstrated that doing a test, as com- Neurophysiol. 2004;115:1490–1505.
pared to not doing the test, benefits the health and quality of life 5. Waldemar G, Dubois B, Emre M, et al. Recommendations for the
of the patient. Very few EEG studies attempt to address these diagnosis and management of Alzheimer’s disease and other dis-
questions, and this may be a major source of discrepancies orders associated with dementia: EFNS guideline. Eur J Neurol.
between various guidelines. Guidelines that focus upon sensi- 2007;14:e1–e26.
tivity and specificity of comparisons between healthy subjects 6. Smith SJM. EEG in neurological conditions other than epilepsy:
when does it help, what does it add? J Neurol Neurosurg Psychiatry.
and dementia may be far more optimistic in their assessment of
2005;76(suppl II):ii8–ii12.
the usefulness of the EEG than guidelines that stress demon- 7. Knopman DS, DeKosky ST, Cummings JL, et al. Practice parame-
strated clinical usefulness. ter: diagnosis of dementia (an evidence-based review). Report of
In view of these problems, we now have to address our final the Quality Standards Subcommittee of the American Academy of
question: in what conditions can the EEG be expected to be really Neurology. Neurology. 2001;56:1143–1153.
388 Part III ■ Clinical EEG: General

8. Jonkman EJ. The role of the electroencephalogram in the diagno- memory clinic cohort. Dement Geriatr Cogn Disord. 2008;27:
sis of dementia of the Alzheimer type: an attempt at technology 18–23.
assessment. Neurophysiol Clin. 1997;27:211–219. 31. Katz RI, Horowitz GR. Electroencephalogram in the septuagenar-
9. Rosen I. Electroencephalography as a diagnostic tool in dementia. ian: studies in a normal geriatric population. J Am Geriatr Soc.
Dement Geriatr Cogn Disord. 1997;8:110–116. 1982;30:273–275.
10. Boerman RH, Scheltens P, Weinstein HC. Clinical neurophysiol- 32. Westmoreland BF, Klass DW. A distinctive rhythmic EEG dis-
ogy in the diagnosis of Alzheimers disease. Clin Neurol Neurosurg. charge of adults. Electroenceph Clin Neurophysiol. 1981;51:
1994;96:111–118. 186–191.
11. Adamis D, Sahu S, Treloar A. The utility of EEG in dementia: a 33. Petersen RC, Smith GE, Waring SC, et al. Mild cognitive impair-
clinical perspective. Int J Geriatr Psychiatry. 2005;20:1038–1045. ment: clinical characterization and outcome. Arch Neurol.
12. Van der Flier WM, Scheltens P. Use of laboratory and imaging 1999;56:303–308.
investigations in dementia. J Neurol Neurosurg Psychiatry. 34. Grunwald M, Busse F, Hensel A, et al. Correlation between corti-
2005;76(suppl V):v45–v52. cal theta activity and hippocampal volumes in health, mild cogni-
13. Jackson CE, Snyder PJ. Electroencephalography and event-related tive impairment and mild dementia. J Clin Neurophysiol. 2001;18:
potentials as biomarkers of mild cognitive impairment and mild 178–184.
Alzheimer’s disease. Alzheimers Dement. 2008;4(1 suppl 1): 35. Grunwald M, Bussse F, Hensel A, et al. Theta-power differences in
S137–S143. patients with mild cognitive impairment under rest condition
14. Klass DW, Brenner RP. Electroencephalography of the elderly. and during haptic tasks. Alzheimer Dis Assoc Disord. 2002;16:
J Clin Neurophysiol. 1995;12:116–131. 40–48.
15. Rossini PM, Rossi S, Babiloni C, et al. Clinical neurophysiology of 36. Jelic V, Shigeta M, Julin P, et al. Quantitative electroencephalogra-
aging brain: from normal aging to neurodegeneration. Prog phy power and coherence in Alzheimer’s disease and mild cogni-
Neurobiol. 2007;83:375–400. tive impairment. Dementia. 1996;7:314–323.
16. Hartikainen P, Soininen H, Partanen J, et al. Aging and spectral 37. Zappoli R, Versari A, Paganini M, et al. Brain electrical activity
analysis of EEG in normal subjects: a link to memory and CSF (quantitative EEG and bit-mapping neurocognitive CNF compo-
AChE. Acta Neurol Scand. 1992;86:148–155. nents), psychometrics and clinical findings in presenile subjects
17. Obrist WD. The electroencephalogram in normal aged adults. with initial mild cognitive decline or probable Alzheimer-type
Electroenceph Clin Neurophysiol. 1954;6:235–244. dementia. Ital J Neurol Sci. 1995;16:341–376.
18. Hubbard O, Sunde D, Goldensohn ES. The EEG in centenarians. 38. Grunwald M, Hensel A, Wolf H, et al. Does the hippocampal atro-
Electroenceph Clin Neurophysiol. 1976;40:407–417. phy correlate with the cortical theta power in elderly subjects with
19. Aird RB, Gastaut Y. Occipital and posterior electroencephalographic a range of cognitive impairment? J Clin Neurophysiol. 2007;
rhythms. Electroenceph Clin Neurophysiol. 1959;11:637–656. 24:22–26.
20. Markand ON. Alpha rhythms. J Clin Neurophysiol. 1990;7: 39. Jiang ZY. Study on EEG power and coherence in patients with
163–189. mild cognitive impairment during working memory task.
21. Babiloni C, Frisoni G, Steriade M, et al. Frontal white matter vol- J Zhejiang Univ Sci B. 2005;6:1213–1219.
ume and delta EEG sources negatively correlate in awake subjects 40. Babiloni C, Binetti G, Cassetta E, et al. Sources of cortical rhythms
with mild cognitive impairment and Alzheimer’s disease. Clin change as a function of cognitive impairment in pathological
Neurophysiol. 2006;117:1113–1129. aging: a multicenter study. Clin Neurophysiol. 2006;117:252–268.
22. Klimesch W. EEG alpha and theta oscillations reflect cognitive 41. Babiloni C, Frisoni GB, Pievani M, et al. White-matter vascular
and memory performance: a review and analysis. Brain Res Rev. lesions correlate with alpha EEG sources in mild cognitive
1999;29:169–195. impairment. Neuropsychologia. 2008;46:1707–1720.
23. Dustman RE, LaMarche JA, Cohn NB, et al. Power spectral analy- 42. Lee L, Harrison LM, Mechelli A. A report of the functional con-
sis and cortical coupling of EEG for young and old normal adults. nectivity workshop, Dusseldorf 2002. NeuroImage. 2003;19:
Neurobiol Aging. 1985;6:193–198. 457–465.
24. Schnell RG, Klass DW. Further observation son the Rolandic 43. Pijnenburg YA, v d Made Y, van Cappellen van Walsum AM, et al.
arceau rhythm. Electroenceph Clin Neurophysiol. 1966;20:95. EEG synchronization likelihood in mild cognitive impairment
25. Giaquinto S. Nolfe G. The EEG in the normal elderly: a contribu- and Alzheimer’s disease during a working memory task. Clin
tion to the interpretation of aging and dementia. Electroenceph Neurophysiol. 2004;115:1332–1339.
Clin Neurophysiol. 1986;63:540–546. 44. van der Hiele K, Vein AA, Kramer CG, et al. Memory activation
26. Oken BS, Kaye JA. Electrophysiologic function in the healthy, enhances EEG abnormality in mild cognitive impairment.
extremely old. Neurology. 1992;42:519–526. Neurobiol Aging. 2007;28:85–90.
27. Silverman AJ, Busse EW, Barnes RH. Studies in the processes of 45. Stam CJ, van der Made Y, Pijnenburg YAL, et al. EEG synchroniza-
aging: electroencephalographic findings in 400 elderly subjects. tion in mild cognitive impairment and Alzheimer’s disease. Acta
Electroenceph Clin Neurophysiol. 1955;7:67–74. Neurol Scand. 2003;108:90–96.
28. Visser SL, Hooijer C, Jonker C, et al. Anterior temporal focal 46. Babiloni C, Ferri R, Binetti G, et al. Fronto-parietal coupling of
abnormalities in EEG in normal aged subjects; correlations with brain rhythms in mild cognitive impairment: a multicentric EEG
psychopathological and CT brain scan findings. Electroenceph study. Brain Res Bull. 2006;69:63–73.
Clin Neurophysiol. 1987;66:1–7. 47. Huang C, Wahlund L, Dierks T, et al. Discrimination of
29. Cook IA, Leuchter AF, Morgan ML, et al. Cognitive and physio- Alzheimer’s disease and mild cognitive impairment by equivalent
logic correlates of subclinical structural brain disease in elderly EEG sources: a cross-sectional and longitudinal study. Clin
healthy control subjects. Arch Neurol. 2002;59:1612–1620. Neurophysiol. 2000;111:1961–1967.
30. Liedorp M, van der Flier WM, Hoogervorst EL, et al. Associations 48. Jelic V, Johansson S-E, Almkvist O, et al. Quantitative electroen-
between patterns of EEG abnormalities and diagnosis in a large cephalography in mild cognitive impairment: longitudinal
Chapter 20 ■ Dementia and EEG 389

changes and possible prediction of Alzheimer’s disease. Neurobiol 68. van der Hiele K, Vein AA, Reijntjes RH, et al. EEG correlates in the
Aging. 2000;21:533–540. spectrum of cognitive decline. Clin Neurophysiol. 2007;118:
49. Missonnier P, Gold G, Herrmann FR, et al. Decreased theta event- 1931–1939.
related synchronization during working memory activation is 69. Adler G, Brassen S, Jajcevic A. EEG coherence in Alzheimer’s
associated with progressive mild cognitive impairment. Dement dementia. J Neural Transm. 2003;110:1051–1058.
Geriatr Cogn Disord. 2006;22:250–259. 70. Coben LA, Danziger WL, Berg L. Frequency analysis of the rest-
50. Rossini PM, Del Percio C, Pasqualetti P, et al. Conversion from ing awake EEG in mild senile dementia of Alzheimer type.
mild cognitive impairment to Alzheimer’s disease is predicted by Electroencephalogr Clin Neurophysiol. 1983;55:372–380.
sources and coherence of brain electroencephalography rhythms. 71. Schreiter-Gasser U, Gasser T, Ziegler P. Quantitative EEG analysis
Neuroscience. 2006;143:793–803. in early onset Alzheimer’s disease: a controlled study.
51. Luckhaus C, Grass-Kapanke B, Blaeser I, et al. Quantitative EEG Electroenceph Clin Neurophysiol. 1993;86:15–22.
in progressing vs stable mild cognitive impairment (MCI): results 72. Schreiter-Gasser U, Gasser T, Ziegler P. Quantitative EEG analysis
of a 1-year follow-up study. Int J Geriatr Psychiatry. 2008; in early onset Alzheimer’s disease: correlation with severity, clin-
23:1148–1155. ical characteristics, visual EEG and CCT. Electroenceph Clin
52. van der Hiele K, Bollen EL, Vein AA, et al. EEG markers of future Neurophysiol. 1994;90:267–272.
cognitive performance in the elderly. J Clin Neurophysiol. 73. Rice DM, Buchsbaum MS, Starr A, et al. Abnormal slow activity
2008;25:83–89. in left temporal areas in senile dementia of the Alzheimer type. J
53. Prichep LS, John ER, Ferris SH, et al. Prediction of longitudinal Gerontol Med Sci. 1990;45:M145–M151.
cognitive decline in normal elderly with subjective complaints using 74. Dierks T, Ihl R, Frölich L, et al. Dementia of the Alzheimer type:
electrophysiological imaging. Neurobiol Aging. 2006;27:471–481. effects on the spontaneous EEG described by dipole sources.
54. Alzheimer A. Über eine eigenartige Erkrankung der Hirnrinde. Psychiatry Res. 1993;50:151–162.
Allgemeine Zeitschrift für Psychiatrie. 1907;64:146–148. 75. Whitham EM, Pope KJ, Fitzgibbon SP, et al. Scalp electrical
55. Braak H, Braak E. Neuropathological staging of Alzheimer- recording during paralysis: quantitative evidence that EEG fre-
related changes. Acta Neuropathol. 1991;82:239–259. quencies above 20 Hz are contaminated by EMG. Clin
56. Selkoe DJ. Cell biology of the amyloid beta-protein precursor and Neurophysiol. 2007;118:1877–1888.
the mechanism of Alzheimer’s disease. Annu Rev cell Biol. 76. van Deursen JA, Vuurman EF, Verhey FR, et al. Increased EEG
1994;10:373–403. gamma band activity in Alzheimer’s disease and mild cognitive
57. Klunk WE, Engler H, Nordberg A, et al. Imaging brain amyloid in impairment. J Neural Transm. 2008;115:1301–1311.
Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol. 77. Coben LA, Danziger W, Storandt M. A longitudinal EEG study of
2004;55:306–319. mild senile dementia of Alzheimer type: changes at 1 year and at
58. Shoghi-Jadid K, Small GW, Agdeppa ED, et al. Localization of 2.5 years. Electroenceph Clin Neurophysiol. 1985;61:101–112.
neurofibrillary tangles and beta-amyloid plaques in the brains of 78. Roks G, Korf ESC, van der Flier WM, et al. The use of EEG in the
living patients with Alzheimer disease. Am J Geriatr Psychiatry. diagnosis of dementia with Lewy bodies. J Neurol Neurosurg
2002;10:24–35. Psychiatry. 2008;79:377–380.
59. Bracco L, Gallato R, Grigoletto F, et al. Factors affecting course 79. Amatniek JC, Hauser WA, DelCastillo-Castaneda C, et al.
and survival in Alzheimer’s disease. A 9-year longitudinal study. Incidence and predictors of seizures in patients with Alzheimer's
Arch Neurol. 1994;51:1213–1219. disease. Epilepsia. 2006;47:867–872.
60. Farrer LZ, Cupples LA, Blackburn S, et al. Interrater agreement 80. Hogh P, Smith SJ, Scahill RI, et al. Epilepsy presenting as AD: neu-
for diagnosis of Alzheimer’s disease. The mirage study. Neurology. roimaging, electrical features, and response to treatment.
1994;44:652–256. Neurology. 2002;58:298–301.
61. McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of 81. Tombini M, Koch G, Placidi F, et al. Temporal lobe epileptic activ-
Alzheimer’s disease: report of the NINCDS-ADRDA Work Group ity mimicking dementia: a case report. Eur J Neurol. 2005;12:
under the auspices of Department of Health and Human Services 805–806.
Task Force on Alzheimer’s disease. Neurology. 1984;34:939–944. 82. Zeman AZ, Boniface SJ, Hodges JR. Transient epileptic amnesia: a
62. Paulus W, Bancher C, Jellinger K. Interrater reliability in the description of the clinical and neuropsychological features in 10
neuropathologic diagnosis of Alzheimer’s disease. Neurology. cases and a review of the literature. J Neurol Neurosurg Psychiat.
1992;42:329–332. 1998;64:435–443.
63. Babiloni C, Binetti G, Cassetta E, et al. Mapping distributed 83. Cook IA, Leuchter AF. Synaptic dysfunction in Alzheimer’s dis-
sources of cortical rhythms in mild Alzheimer’s disease. A multi- ease: clinical assessment using quantitative EEG. Behav Brain Res.
centric EEG study. Neuroimage. 2004;22:57–67. 1996;78:15–23.
64. Czigler B, Csikós D, Hidasi Z, et al. Quantitative EEG in early 84. Besthorn C, Forstl H, Geiger-Kabisch C, et al. EEG coherence in
Alzheimer’s disease patients—power spectrum and complexity Alzheimer disease. Electroenceph Clin Neurophysiol. 1994;90:242–245.
features. Int J Psychophysiol. 2008;68:75–80. 85. Dunkin JJ, Leuchter AF, Newton TF, et al. Reduced EEG coherence
65. Gianotti LR, Künig G, Lehmann D, et al. Correlation between dis- in dementia: state or trait marker? Biol Psychiatry. 1994;35:
ease severity and brain electric LORETA tomography in 870–879.
Alzheimer’s disease. Clin Neurophysiol. 2007;118:186–196. 86. Leuchter AF, Spar JE, Walter DO, et al. Electroencephalographic
66. Penttilä M, Partanen JV, Soininen H, et al. Quantitative analysis of spectra and coherence in the diagnosis of Alzheimer’s-type and
occipital EEG in different stages of Alzheimer’s disease. multi-infarct dementia. A pilot study. Arch Gen Psychiatry.
Electroenceph Clin Neurophysiol. 1985;60:1–6. 1987;44:993–998.
67. Saletu B, Anderer P, Paulus E, et al. EEG brain mapping in diag- 87. Leuchter AF, Newton TF, Cook AA, et al. Changes in brain func-
nostic and therapeutic assessment of dementia. Alzheimer Dis tional connectivity in Alzheimer-type and multi-infarct demen-
Assoc Disord. 1991;5(suppl 1):S57–S75. tia. Brain. 1992;115:1543–1561.
390 Part III ■ Clinical EEG: General

88. Locatelli T, Cursi M, Liberati D, et al. EEG coherence in 108. Stam CJ, Jelles B, Achtereekte HAM, et al. Investigation of EEG
Alzheimer’s disease. Electroenceph Clin Neurophysiol. 1998; non-linearity in dementia and Parkinson’s disease. Electroenceph
106:229–237. Clin Neurophysiol. 1995;95:309–317.
89. Pogarell O, Teipel SJ, Juckel G, et al. EEG coherence reflects 109. Stam CJ, Jelles B, Achtereekte HAM, et al. Diagnostic usefulness
regional corpus callosum area in Alzheimer’s disease. J Neurol of linear and nonlinear quantitative EEG analysis in Alzheimer’s
Neurosurg Psychiatry. 2005;76:109–111. disease. Clin Electroenceph. 1996;27:69–77.
90. Sloan EP, Fenton GW, Kennedy NS, et al. Neurophysiology and 110. Woyshville MJ, Calabrese JR. Quantification of occipital EEG
SPECT cerebral blood flow patterns in dementia. Electroenceph changes in Alzheimer’s disease utilizing a new metric: the fractal
Clin Neurophysiol. 1994;91:163–170. dimension. Biol Psychiatry. 1994;35:381–387.
91. Park YM, Che HJ, Im CH, et al. Decreased EEG synchronization 111. Yagyu T, Wackermann J, Shigeta M, et al. Global dimensional
and its correlation with symptom severity in Alzheimer’s disease. complexity of multichannel EEG in mild Alzheimer’s disease
Neurosci Res. 2008;62:112–117. and age-matched controls. Dement Geriatr Cogn Disord.
92. Knott V, Mohr E, Mahoney C, et al. Electroencephalographic coher- 1997;8:343–347.
ence in Alzheimer’s disease: comparisons with a control group and 112. Jelles B, Jonkman EJ, Birgelen JH van, et al. Decrease of non-lin-
population norms. J Geriatr Psychiatry Neurol. 2000;13:1–8. ear structure in the EEG of Alzheimer patients compared to
93. Babiloni C, Ferri R, Binetti G, et al. Directionality of EEG syn- controls. Clin Neurophysiol. 1999;110:1159–1167.
chronization in Alzheimer’s disease subjects. Neurobiol Aging. 113. Jelles B, Scheltens P, van der Flier WM, et al. Global dynamical
2009;30:93–102. analysis of the EEG in Alzheimer’s disease: frequency-specific
94. Babiloni C, Ferri R, Moretti DV, et al. Abnormal fronto-parietal changes of functional interactions. Clin Neurophysiol.
coupling of brain rhythms in mild Alzheimer’s disease: a multi- 2008;119:837–841.
centric EEG study. Eur J Neurosci. 2004;19:2583–2590. 114. Van Cappellen van Walsum A-M, Pijnenburg YAL, Berendse
95. Jeong J, Gore JC, Peterson BS. Mutual information analysis of the HW, et al. A neural complexity measure applied to MEG data in
EEG in patients with Alzheimer’s disease. Clin Neurophysiol. Alzheimer’s disease. Clin Neurophysiol. 2003;114:1034–1040.
2001;112:827–835. 115. Reijneveld JC, Ponten SC, Berendse HW, et al. The application
96. Anderer P, Saletu B, Kloppel B, et al. Discrimination between of graph theoretical analysis to complex networks in the brain.
demented patients and normals based upon topographic EEG Clin Neurophysiol. 2007;118:2317–2331.
slow wave activity: comparison between z statistics, discriminant 116. Stam CJ, Reijneveld JC. Graph theoretical analysis of complex
analysis and artificial neural network classifiers. Electroenceph networks in the brain. Nonlinear Biomed Phys. 2007;1:3.
Clin Neurophysiol. 1994;91;108–117. 117. He Y, Chen ZJ, Evans AC. Small-world anatomical networks in
97. Pritchard WS, Duke DW, Coburn KL, et al. EEG-based, neural- the human brain revealed by cortical thickness from MRI. Cereb
net predictive classification of Alzheimer’s disease versus control Cortex. 2007;17:2407–2419.
subjects is augmented by non-linear EEG measures. Electroenceph 118. Stam CJ, Jones BF, Nolte G, et al. Small-world networks and
Clin Neurophysiol. 1994;91:118–130. functional connectivity in Alzheimer’s disease. Cereb Cortex.
98. Rossini PM, Buscema M, Capriotti M, et al. Is it possible to auto- 2007;17:92–99.
matically distinguish resting EEG data of normal elderly vs. mild 119. Stam CJ, de Haan W, Daffertshofer A, et al. Graph theoretical analy-
cognitive impairment subjects with high degree of accuracy? Clin sis of magnetoencephalographic functional connectivity in
Neurophysiol. 2008;119:1534–1545. Alzheimer’s disease. Brain. 2008, October 24. [Epub ahead of print]
99. Stam CJ. Nonlinear dynamical analysis of EEG and MEG: review 120. Supekar K, Menon V, Rubin D, et al. Network analysis of intrin-
of an emerging field. Clin Neurophysiol. 2005;116:2266–2301. sic functional brain connectivity in Alzheimer’s disease. PLoS
100. Stam CJ. Nonlinear Brain Dynamics. New York: Nova Science Comput Biol. 2008;4:e1000100.
Publishers; 2006. 121. Petit D, Montplaisir J, Lorrain D, et al. Spectral analysis of the
101. Jeong J. Nonlinear dynamics of EEG in Alzheimer’s disease. Drug rapid eye movement sleep EEG in right and left temporal reigns;
Dev Res. 2002;56:57–66. a biological marker for diagnosis of probable Alzheimer’s dis-
102. Besthorn C, Sattel H, Geiger-Kabisch C, et al. Parameters of EEG ease. Neurology. 1992;42:765–770.
dimensional complexity in Alzheimer’s disease. Electroenceph 122. Prinz PN, Larsen LH, Moe KE, et al. EEG markers of early
Clin Neurophysiol. 1995;95:84–89. Alzheimer’s disease in computer selected tonic REM sleep.
103. Hornero R, Abásolo D, Escudero J, et al. Nonlinear analysis of Electroenceph Clin Neurophysiol. 1992;83:36–43.
electroencephalogram and magnetoencephalogram recordings in 123. Moe KE, Larsen LH, Prinz PN, et al. Major unipolar depression
patients with Alzheimer’s disease. Philos Transact A Math Phys and mild Alzheimer’s disease: differentiation by quantitative
Eng Sci. 2009;367:317–336. tonic REM EEG. Electroenceph Clin Neurophysiol. 1993;86:
104. Jelles B, Strijers RLM, Hooijer Ch, et al. Nonlinear EEG analysis 238–246.
in early Alzheimer’s disease. Acta Neurologica Scandinavica. 124. Robinson DJ, Merskey H, Blume WT, et al.
1999;100:360–368. Electroencephalography as an aid in the exclusion of
105. Jeong J, Kim SY, Han SH. Non-linear dynamical analysis of the Alzheimer’s disease. Arch Neurol. 1994;51:280–284.
EEG in Alzheimer’s disease with optimal embedding dimension. 125. Strijers RLM, Scheltens Ph, Jonkman EJ, et al. Diagnosing
Electroenceph Clin Neurophysiol. 1998;106:220–228. Alzheimer’s disease in community-dwelling elderly: a compari-
106. Pritchard WS, Duke DW, Coburn KL. Altered EEG dynamical son of EEG and MRI. Dement Geratr Cogn Disord. 1997;8:
responsivity associated with normal aging and probable 198–202.
Alzheimer’s disease. Dementia. 1991;2:102–105. 126. Scheltens P, Leys D, Barkhof F, et al. Atrophy of medial temporal
107. Stam CJ, Tavy DLJ, Jelles B, et al. Non-linear dynamical analysis lobes on MRI in “probable” Alzheimer’s disease and normal age-
of multi channel EEG data: clinical applications in dementia and ing: diagnostic value and neuropsychological correlates. J Neurol
Parkinson's disease. Brain Topogr. 1994;7:141–150. Neurosurg Psychiatry. 1992;55:967–972.
Chapter 20 ■ Dementia and EEG 391

127. Claus JJ, Strijers RLM, Jonkman EJ, et al. The diagnostic value of 145. Ponomareva NV, Korovaitseva GI, Rogaev EI. EEG alterations in
electroencephalography in mild senile Alzheimer’s disease. Clin non-demented individuals related to apolipoprotein E genotype
Neurophysiol. 1999;110:825–832. and to risk of Alzheimer disease. Neurobiol Aging. 2008;29:
128. Brenner R, Ulrich RF, Spiker DG, et al. Computerized EEG spec- 819–827.
tral analysis in elderly normal, demented and depressed sub- 146. Coyle JT, Price DL, DeLong MR. Alzheimer’s disease: a disorder
jects. Electroenceph Clin Neurophysiol. 1986;64:483–492. of cortical cholinergic innervation. Science. 1983;219:
129. Brenner RP, Reynolds III ChF, Ulrich RF. Diagnostic efficacy of 1181–1190.
computerized spectral versus visual EEG analysis in elderly nor- 147. Collerton D. Cholinergic function and intellectual decline in
mal, demented and depressed subjects. Electroenceph Clin Alzheimer’s disease. Neuroscience. 1986;19:1–28.
Neurophysiol. 1988;69:110–117. 148. Perry EK, Perry RH, Blessed G, et al. Necropsy evidence of cen-
130. Hooijer Ch, Jonker C, Posthuma J, et al. Reliability, validity and tral cholinergic deficits in senile dementia. Lancet. 1977;1:189.
follow-up of the EEG in senile dementia: sequelae of sequential 149. Buzsaki G, Bickford RG, Ponomareff G, et al. Nucleus basalis
measurement. Electroenceph Clin Neurophysiol. 1990;76: and thalamic control of neocortical activity in the freely moving
400–412. rat. J Neurosci. 1988;8:4007–4026.
131. Babiloni C, Frisoni GB, Pievani M, et al. Hippocampal volume 150. Villa AEP, Tetko IV, Dutoit P, et al. Non-linear cortico-cortical
and cortical sources of EEG alpha rhythms in mild cognitive interactions modulated by cholinergic afferences from the rat
impairment and Alzheimer disease. Neuroimage. 2009;44: basal forebrain. BioSystems. 2000;58:219–228.
123–135. 151. Kikuchi M, Wada Y, Koshino Y, et al. Effects of scopolamine on
132. van der Hiele K, Vein AA, van der Welle A, et al. EEG and MRI interhemispheric EEG coherence in healthy subjects: analysis
correlates of mild cognitive impairment and Alzheimer’s dis- during rest and photic stimulation. Clin Electroencephalogr.
ease. Neurobiol Aging. 2007;28:1322–1329. 2000;31:109–115.
133. Dunkin JJ, Osato S, Leuchter A. Relationships between EEG 152. Ebert U, Grossmann M, Oertel R, et al. Pharmacokinetic-phar-
coherence and neuropsychological tests in dementia. Clin macodynamic modeling of the electroencephalogram effects of
Electroenceph. 1995;26:47–59. scopolamine in healthy volunteers. J Clin Pharmacol. 2001;41:
134. Lopez OL, Brenner RP, Becker JT, et al. EEG spectral abnormal- 51–60.
ities and psychosis as predictors of cognitive and functional 153. Weingartner H. Models of memory dysfunctions. Ann N Y Acad
decline in probable Alzheimer’s disease. Neurology. 1997;48: Sci. 1985;444:359–369.
1521–1525. 154. Riekkinen P, Buzsaki G, Riekkinen P Jr, et al. The cholinergic
135. Helkala EL, Laulumaa V, Soininen H, et al. Different patterns of system and EEG slow waves. Electroenceph Clin Neurophysiol.
cognitive decline related to normal or deteriorating EEG in a 3- 1991;78:89–96.
year follow-up study of patients with Alzheimer’s disease. 155. Agnoli A, Martucci N, Manna V, et al. Effect of cholinergic and
Neurology. 1991;41:528–532. anticholinergic drugs on short-term memory in Alzheimer’s
136. Claus JJ, Gool WA van, Teunisse S, et al. Predicting survival in dementia: a neuropsychological and computerized electroen-
patients with early Alzheimer’s disease. Dement Geriatr Cogn cephalographic study. Clin Neuropharmacol. 1983;6:311–323.
Disord. 1998;9:283–293. 156. Babiloni C, Cassetta E, Dal Forno G, et al. Donepezil effects on
137. Claus JJ, Ongerboer de Visser BW, Walstra GJM, et al. sources of cortical rhythms in mild Alzheimer’s disease:
Quantitative spectral electroencephalography in predicting sur- Responders vs. Non-Responders. Neuroimage. 2006;31:
vival in patients with early Alzheimer disease. Arch Neurol. 1650–1665.
1998;55:1105–1111. 157. Gianotti LR, Künig G, Faber PL, et al. Rivastigmine effects on
138. Nobili F, Copello F, Vitali P, et al. Timing of disease progression EEG spectra and three-dimensional LORETA functional imag-
by quantitative EEG in Alzheimer’s patients. J Clin Neurophysiol. ing in Alzheimer’s disease. Psychopharmacology (Berl). 2008;
1999;16:566–573. 198:323–332.
139. Strittmatter WJ, Roses AD. Apolipoprotein E and Alzheimer’s 158. Rodriguez G, Vitali P, De Leo C, et al. Quantitative EEG changes
disease. Proc Natl Acad Sci USA. 1995;92:4725–4727. in Alzheimer patients during long-term Donepezil therapy.
140. Lehtovirta M, Soininen H, Helisalmi S, et al. Clinical and neu- Neuropsychobiology. 2002;46:49–56.
ropsychological characteristics in familial sporadic Alzheimer’s 159. Adler G, Brassen S, Chwalek K, et al. Prediction of treatment
disease: relation to apolipoprotein E polymorphism. Neurology. response to rivastigmine in Alzheimer’s dementia. J Neurol
1996;46:413–419. Neurosurg Psychiat. 2004;75:292–294.
141. Lehtovirta M, Partanen J, Kononen M, et al. Spectral analysis of 160. Almkvist O, Jelic V, Hellstrom-Lindahl E, et al. Responder char-
EEG in Alzheimer’s disease: relation to Apolipoprotein E poly- acteristics to a single oral dose of cholinesterase inhibitor: a dou-
morphism. Neurobiol Ageing. 1996;4:523–526. ble-blind placebo-controlled study with Tacrine in Alzheimer
142. Lehtovirta M, Partanen J, Kononen M, et al. A longitudinal patients. Dement Geriatr Cogn Disord. 2001;12:22–32.
quantitative EEG study of Alzheimer’s disease: relation to 161. Knott V, Mohr E, Mahoney C, et al. Pharmaco-EEG test dose
apolipoprotein E polymorphism. Dem Geriatr Cogn Disord. response predicts cholinesterase inhibitor outcome in Alzheimer’s
2000;11:29–35. disease. Methods Find Exp Clin Pharmacol. 2000;22:115–122.
143. Jelic V, Julin P, Shigeta M, et al. Apolipoprotein E H4 allele 162. Dringenberg HC. Alzheimer’s disease: more than a ‘cholinergic
decreases functional connectivity in Alzheimer’s disease as disorder’—evidence that cholinergic—monoaminergic interac-
measured by EEG coherence. J Neurol Neurosurg Psychiatry. tions contribute to EEG slowing and dementia. Behav Brain Res.
1997;63:59–65. 2000;115:235–249.
144. Kramer G, van der Flier WM, de Langen C, et al. EEG functional 163. Di Lazzaro V, Oliviero A, Pilato F, et al. Motor cortex hyperex-
connectivity and ApoE genotype in Alzheimer’s disease and citability to transcranial magnetic stimulation in Alzheimer’s
controls. Clin Neurophysiol. 2008;119:2727–2732. disease. J Neurol Neurosurg Psychiatry. 2004;75:555–559.
392 Part III ■ Clinical EEG: General

164. Davis KL, Mohs RC, Marin D, et al. Cholinergic markers in eld- 184. Calzetti S, Bortone E, Negrotti A, et al. Frontal intermittent
erly patients with early signs of Alzheimer’s disease. J Am Med rhythmic delta activity (FIRDA) in patients with dementia with
Assoc. 1999;45:178–179. Lewy bodies: a diagnostic tool? Neurol Sci. 2002;23(suppl.
165. Visser FE, Kuilman M, Oosting J, et al. Use of electroencephalog- 2):S65–S66.
raphy to detect Alzheimer’s disease in Down’s syndrome. Acta 185. Barber PA, Varma AR, Lloyd JJ, et al. The electroencephalogram
Neurol Scand. 1996;94:97–103. in dementia with Lewy bodies. Acta Neurol Scand. 2000;
166. Jelic V, Blomberg M, Dierks T, et al. EEG slowing and cere- 101:53–56.
brospinal fluid tau levels in patients with cognitive decline. 186. Londos E, Passant U, Brun A, et al. Regional cerebral blood flow
Neuroreport. 1988;9:157–160. and EEG in clinically diagnosed dementia with Lewy bodies and
167. Babiloni C, Bosco P, Ghidoni R, et al. Homocysteine and elec- Alzheimer’s disease. Arch Gerontol Geriatr. 2003;36:231–245.
troencephalographic rhythms in Alzheimer disease: a multicen- 187. Bonanni L, Thomas A, Tiraboschi P, et al. EEG comparisons in
tric study. Neuroscience. 2007;145:942–954. early Alzheimer’s disease, dementia with Lewy bodies and
168. Schreiter-Gasser U, Rousson V, Hentschel F, et al. Alzheimer dis- Parkinson’s disease with dementia patients with a 2-year follow-
ease versus mixed dementias: an EEG perspective. Clin up. Brain. 2008;131:690–705.
Neurophysiol. 2008;119:2255–2259. 188. Briel RCG, McKeith IG, Barker WA, et al. EEG findings in
169. Johannesson G, Brun A, Gustafson I, et al. EEG in presenile dementia with Lewy bodies and Alzheimer’s disease. J Neurol
dementia related to cerebral blood flow and autopsy findings. Neurosurg Psychiatry. 1999;66:401–403.
Acta Neurol Scand. 1977;56:89–103. 189. Kai T, Asai Y, Sakuma K, et al. Quantitative electroencephalo-
170. Johannesson G, Hagberg B, Gustafson I, et al. EEG and cognitive gram analysis in dementia with Lewy bodies and Alzheimer’s
impairment in presenile dementia. Acta Neurol Scand. disease. J Neurol Sci. 2005;237:89–95.
1979;59:225–240. 190. Andersson M, Hansson O, Minthon L, et al. Electroencephalogram
171. Rosén I, Gustafson L, Risberg J. Multichannel EEG frequency variability in dementia with Lewy bodies, Alzheimer’s disease and
analysis and somatosensory-evoked potentials in patients with controls. Dement Geriatr Cogn Disord. 2008;26:284–290.
different types of organic dementia. Dementia. 1993;4:43–49. 191. Scott DF, Heathfield KW, Toone B, et al. The EEG in
172. Stigsby B, Johannesson G, Ingvar DH. Regional EEG analysis Huntington’s chorea: a clinical and neuropathological study.
and regional cerebral blood flow in Alzheimer’s disease and J Neurol Neurosurg Psychiatry. 1972;35:97–102.
Pick’s disease. Electroenceph Clin Neurophysiol. 1981;51: 192. Streletz LJ, Reyes PF, Zalewska M, et al. Computer analysis of
537–547. EEG activity in dementia of the Alzheimer’s type and
173. Forstl H, Besthorn C, Hentschel F, et al. Frontal lobe degenera- Huntington’s disease. Neurobiol Aging. 1990;11:15–20.
tion and Alzheimer’s disease: a controlled study on clinical 193. van der Hiele K, Jurgens CK, Vein AA, et al. Memory activation
findings, volumetric brain changes and quantitative electroen- reveals abnormal EEG in preclinical Huntington’s disease. Mov
cephalography data. Dementia. 1996;7:27–34. Disord. 2007;22:690–695.
174. Pijnenburg YA, Strijers RL, Made YV, et al. Investigation of rest- 194. Fowler CJ, Harrison MJ. EEG changes in subcortical dementia: a
ing-state EEG functional connectivity in frontotemporal lobar study of 22 patients with steele-richardson-olszewski (SRO)
degeneration. Clin Neurophysiol. 2008;119:1732–1738. syndrome. Electroencephalogr Clin Neurophysiol. 1986;64:
175. Yener GG, Leuchter AF, Jenden D, et al. Quantitative EEG in 301–303.
frontotemporal dementia. Clin Electroencephalogr. 1996;27: 195. Tashiro K, Ogata K, Goto Y, et al. EEG findings in early-stage
61–68. corticobasal degeneration and progressive supranuclear palsy: a
176. Chan D, Walters RJ, Sampson EL, et al. EEG abnormalities in retrospective study and literature review. Clin Neurophysiol.
frontotemporal lobar degeneration. Neurology. 2004;62: 2006;117:2236–2242.
1628–1630. 196. Román GC, Tatemichi TK, Erkinjuntti T, et al. Vascular demen-
177. Passant U, Rosén I, Gustafson L, et al. The heterogeneity of fron- tia: diagnostic criteria for research studies. Report of the
totemporal dementia with regard to initial symptoms, qEEG NINDS-AIREN International Workshop. Neurology. 1993;43:
and neuropathology. Int J Geriatr Psychiatry. 2005;20:983–988. 250–260.
178. Neufeld MY, Inzelberg R, Korczyn AD. EEG in demented and 197. Holmes C, Cairns N, Lantos P, et al. Validity of current clinical
non-demented parkinsonian patients. Acta Neurol Scand. criteria for Alzheimer’s disease, vascular dementia and dementia
1988;78:1–5. with Lewy bodies. Br J Psychiatry. 1999;174:45–50.
179. Caviness JN, Hentz JG, Evidente VG, et al. Both early and 198. Erkinjuntti T. Differential diagnosis between Alzheimer’s disease
late cognitive dysfunction affects the electroencephalogram and vascular dementia: evaluation of common clinical methods.
in Parkinson’s disease. Parkinsonism Relat Disord. 2007;13: Acta Neurol Scand. 1987;76:433–442.
348–354. 199. Erkinjuntti T, Larsen T, Sulkava R, et al. EEG in the differential
180. Domitrz I, Friedman A. Electroencephalography of demented diagnosis between Alzheimer’s disease and vascular dementia.
and non-demented Parkinson’s disease patients. Parkinsonism Acta Neurol Scand. 1988;77:36–43.
Relat Disord. 1999;5:37–41. 200. Jeong J, Chae J-H, Kim SY, et al. Nonlinear dynamic analysis of
181. Neufeld MY, Blumen S, Aitkin I, et al. EEG frequency analysis in the EEG in patients with Alzheimer’s disease and vascular
demented and nondemented parkinsonian patients. Dementia. dementia. J Clin Neurophysiol. 2001;18:58–67.
1994;5:23–28. 201. Moretti DV, Babiloni C, Binetti G, et al. Individual analysis of
182. Fogelson N, Kogan E, Korczyn AD, et al. Effects of rivastigmine EEG frequency and band power in mild Alzheimer’s disease.
on the quantitative EEG in demented Parkinsonian patients. Clin Neurophysiol. 2004;115:299–308.
Acta Neurol Scand. 2003;107:252–255. 202. Gawel M, Zalewska E, Szmidt-Sałkowska E, et al. Does EEG
183. Crystal HA, Dickson DW, Lizardi JE, et al. Antemortem diagno- (visual and quantitative) reflect mental impairment in subcorti-
sis of diffuse Lewy body disease. Neurology. 1990;40:1523–1528. cal vascular dementia? J Neurol Sci. 2007;257:11–16.
Chapter 20 ■ Dementia and EEG 393

203. Romano J, Engel GL. Delirium: I. Electroencephalographic data. 219. Steinhoff BJ, Zerr I, Glatting M, et al. Diagnostic value of peri-
Arch Neurol Psychiatry. 1944;51:356–377. odic complexes in Creutzfeldt-Jakob disease. Ann Neurol.
204. Koponen H, Partanen J, Pääkkönen A, et al. EEG spectral analy- 2004;56:702–708.
sis in delirium. J Neurol Neurosurg Psychiatry. 1989;52:980–985. 220. Zerr I, Pocchiari M, Collins S, et al. Analysis of EEG and CSF
205. Thomas C, Hestermann U, Walther S, et al. Prolonged activation 14-3-3 proteins as aids to the diagnosis of Creutzfeldt-Jakob
EEG differentiates dementia with and without delirium in frail disease. Neurology. 2000;55:811–815.
elderly patients. J Neurol Neurosurg Psychiatry. 2008;79:119–125. 221. Zerr I, Bodemer M, Otto M, et al. Diagnosis of Creutzfeldt-
206. Thomas C, Hestermann U, Kopitz J, et al. Serum anticholinergic Jakob disease by two-dimensional gel electrophoresis of cere-
activity and cerebral cholinergic dysfunction: an EEG study in brospinal fluid. Lancet. 1996;348:846–849.
frail elderly with and without delirium. BMC Neurosci. 222. Van Everbroeck B, Dobbeleir I, De Waele M, et al. Differential
2008;9:86. diagnosis of 201 possible Creutzfeldt-Jakob disease patients.
207. Boutros NN. A review of indications for routine EEG in clinical J Neurol. 2004;251;298–304.
psychiatry. Hosp Community Psych. 1992;43:716–719. 223. Kovacs GG, Trabbattoni G, Hainfellner JA, et al. Mutations of
208. Jacobson S, Jerrier H. EEG in delirium. Semin Clin Neuropsychiatry. the prion protein gene phenotypic spectrum. J Neurol.
2000;5:86–92. 2002;249:1567–1582.
209. Newman SE. The EEG manifestations of chronic ethanol abuse: 224. Tschampa HJ, Herms JW, Schulz-Schaeffer WJ, et al. Clinical
relation to cerebral cortical atrophy. Ann Neurol. 1978;3:299–304. findings in sporadic Creutzfeldt-Jakob disease correlate with
210. World Health Organization. Consensus on criteria for diagnosis thalamic pathology. Brain. 2002;125:2558–2566.
of sporadic CJD. Wkly Epidemiol Rec. 1998;73:361–365. 225. Stam CJ, van Woerkom TCAM, Keunen, RWM. Non-linear
211. Wieser HG, Schindler K, Zumsteg D. EEG in Creutzfeldt-Jakob analysis of the electroencephalogram in Creutzfeldt-Jakob dis-
disease. Clin Neurophysiol. 2006;117:935–951. ease. Biol Cybern. 1997,;77:247–256.
212. Will RG, Zeidler M, Stewart GE, et al. Diagnosis of new variant 226. Stam CJ, Vliegen JHR, Nicolai J. Investigation of the dynamics
Creutzfeldt-Jakob disease. Ann Neurol. 2000;47:575–582. underlying periodic complexes in the EEG. Biol Cybern. 1999;
213. Binelli S, Agazzi P, Giaccone G, et al. Periodic electroencephalo- 80:57–69.
gram complexes in a patient with variant Creutzfeldt-Jakob dis- 227. Radermecker J. Aspects electroencephalographiques dans trois
ease. Ann Neurol. 2006;59:423–427. cas d’encephalite subaigue. Acta Neurol Psychiatr Belg. 1949;49:
214. Cambier DM, Kantarci K, Worrell GA, et al. Lateralized and 222–232.
focal clinical, EEG, and FLAIR MRI abnormalities in Ceutzfelt- 228. Fariello RJ, Black JA. Pseudoperiodic bilateral EEG paroxysms in
Jakob disease. Clin Neurophysiol. 2003;114:1724–1728. a case of phencyclidine intoxication. J Clin Psychiatry.
215. Hirsch LJ, Brenner RP, Drislane FW, et al. The ACNS subcom- 1978;39:579–581.
mittee on research terminology for continuous EEG monitor- 229. Stockard JJ, Werner SS, Aalbers JA, et al. Electroencephalographic
ing: proposed standardized terminology for rhythmic and findings in phencyclidine intoxication. Arch Neurol. 1976;33:
periodic EEG patterns encountered in critically ill patients. J 200–203.
Clin Neurophysiol. 2005;22:128–135. 230. Gabuzda DH, Levy SR, Chiappa KH. Electroencephalography in
216. Hung CI, Wang PS, Soong BW, et al. Blind source separation of AIDS and AIDS-related complex. Clin Electroencephalogr.
concurrent disease-related patterns from EEG in Creutzfeldt- 1988;19:1–6.
Jakob disease for assisting early diagnosis. Ann Biomed Eng. 231. Nuwer MR, Miller EN, Visscher BR, et al. Asymptomatic HIV
2007;35:2168–2179. infection does not cause EEG abnormalities: results from the
217. Wang PS, Wu YT, Hung CI, et al. Early detection of periodic Multicenter AIDS Cohort Study (MACS). Neurology. 1992;42:
sharp wave complexes on EEG by independent component 1214–1219.
analysis in patients with Creutzfeldt-Jakob disease. J Clin 232. Harrison MJ, Newman SP, Hall-Craggs MA, et al. Evidence of
Neurophysiol. 2008;25:25–31. CNS impairment in HIV infection: clinical, neuropsychological,
218. Steinhoff BJ, Racker S, Herrendorf G, et al. Accuracy and relia- EEG, and MRI/MRS study. J Neurol Neurosurg Psychiatry. 1998;
bility of periodic sharp wave complexes in Creutzfeldt-Jakob 65:301–307.
disease. Arch Neurol. 1996;53:162–166.
CHAPTER

Metabolic Disorders and EEG


TRUDY PANG, MEGAN SELVITELLI, DONALD L. SCHOMER AND ERNST NIEDERMEYER 21
INTRODUCTION AND HISTORICAL the cortex disappears earlier than that of deep structures. Level
REMARKS of awareness, blood sugar level, and EEG changes do not neces-
sarily parallel each other (12).
Neuronal electrogenesis is dependent on metabolic homeosta- Early clinical EEG observations were reported by Berger,
sis. Nutritive and energy-providing metabolic systems represent Davis, Hoagland, Gibbs, Engel, Weinland and Weinland, Lafon,
the fuel for neuronal and glial structures and abnormalities in Gibbs and Murray, Hetzel and Niedermeyer, Shagass and
their composition may lead to pronounced clinical and elec- Roswell, and Regan and Browne-Mayers (1,13–22). It was
troencephalographic changes. These changes are usually found that very low blood sugar levels were compatible with a
reversible, but sometimes irreversible progression ensues. normal EEG and also with wakefulness (23). Clinical observa-
Cerebral metabolic disturbances and/or electroencephalo- tions had already shown that impairment of consciousness and
gram (EEG) changes due to blood alkalosis from hyperventila- disturbance of central nervous system (CNS) functions were
tion and anoxia are presented elsewhere. This chapter deals dependent mainly on the rapid fall of the blood sugar level,
mainly with alterations of blood sugar levels, failure of the rather than on absolute values (24). Patients with insulin-
hepatic and renal organ systems, states of electrolyte imbal- dependent diabetes mellitus may be unaware of hypoglycemia.
ances, changes due to endocrine disorders, and certain toxic- Their nearly normal glycosylated hemoglobin levels result in
metabolic conditions. Some chronic metabolic disturbances, normal cerebral glucose uptake and preserved cerebral metab-
such as inborn errors of metabolism, are discussed under the olism (25). Most of the aforementioned work was based on
heading of degenerative disorders. administration of insulin for therapeutic purposes in schizo-
Berger’s observation of slow EEG activity induced by hypo- phrenic patients (Sakel cure).
glycemia in schizophrenic patients treated with insulin marked All of this work shows an extremely impressive degree of
the beginning of EEG studies in human metabolic disorders slowing in the EEG. In particular, the EEG demonstrates a
(1). Over the ensuing years, EEG studies in metabolic disorders decrease in alpha and an increase in theta and delta frequencies
remained somewhat in the background when compared with with increasing hypoglycemia. Most often this is most marked
the activity in the fields of epileptic seizure disorder and struc- in the frontal region (26,27). In some patients, hypoglycemia
tural cerebral lesions. Most of the metabolic disorders belong to may also promote epileptiform discharges (28). Sperling found
the domain of internal medicine, a field of understandably lim- that induced hypoglycemia in medically refractory complex
ited interest in EEG investigations. partial epilepsy patients may activate or accentuate temporal
Major progress was made when the work of Foley, and lobe slowing and epileptiform discharges (29). In a more con-
Bickford and Butt on hepatic coma showed very pronounced trolled manner, induced hypoglycemia in adult cats results in
slowing and formation of triphasic waves (2,3). The contribu- an initial diffuse slowing of the EEG, followed by occasional
tions of Condon, Krump, Thiebaut, Cadilhac, and Glaser have spike waves and then near-isoelectric activity (30). Along this
provided further important information on the EEG correlates continuum and commonly seen in diabetic patients, Kirchner
of metabolic encephalopathies (4–8). and colleagues demonstrated that induced hypoglycemia
There has been persistent interest in the EEG of metabolic results in a faster onset of seizures in rats exposed to flurothyl,
disorders. This is probably due to the discovery of renal which corresponded with implanted hippocampal depth elec-
encephalopathies after dialysis in chronic renal insufficiency (9). trode recorded discharges (31).
Furthermore, EEG monitoring techniques are now being used to Spontaneous states of hypoglycemia may mimic complex
study the evolution of hepatic encephalopathy and the require- partial seizures, due to the concomitant impaired neuropsychi-
ment for and efficacy of hepatic and renal transplantation. atric state. However, the prolonged nature of these attacks,
atypical semiology, unresponsiveness to anticonvulsant thera-
HYPOGLYCEMIA pies, and provocation by fasting helps to differentiate between
these possibilities. Wang and fellow researchers recently
Cerebral functioning depends on an adequate supply of glu- demonstrated an initial theta and delta slowing on the EEG, fol-
cose. Hypoglycemia-induced EEG changes have been shown lowed by sporadic spike and sharp waves during a patient’s typ-
experimentally by Moruzzi, Himwich, Gellhorn and Kessler, ical attacks. Intravenous glucose administration terminated the
and Davis (10–13). It was found that the electrical activity of event (32). Prolonged fasting may be required to confirm the

395
396 Part III ■ Clinical EEG: General

diagnosis of an insulinoma, which requires surgical removal to hepatic coma becomes indistinguishable, regardless of the
promote cessation of events (33). Thus, the hypoglycemic mechanism (9).
attacks induced by an insulinoma may result initially in Patients presenting with hepatic encephalopathy demon-
impaired cognition, while increasing hypoglycemia may pro- strate a notable decline in mental functions, including con-
mote spike-wave discharges and seizures. structional apraxias, increased reaction times, and impaired
memory and concentration (46,47). In addition, altered per-
HYPERGLYCEMIA sonality traits may occur, such as apathy, euphoria, childishness,
and irritability. Motorically, patients demonstrate bradykinesia,
Hyperglycemic states are much less eloquent and predictable in asterixis, parkinsonian traits, and pyramidal signs, which may
their EEG manifestations than are states of hypoglycemia. be present before hypo- and hyperreflexia emerge in profound
Gibbs and colleagues found mixed slow and fast frequencies coma (46). The level of consciousness decreases from confusion
and some intermingled spiking in patients with blood sugar to lethargy and may progress to deep coma with unresponsive-
levels exceeding 400 mg/100 mL (15). In advanced diabetic ness. The concept of minimal hepatic encephalopathy has also
coma, however, very pronounced slowing may occur, and the been raised, in which patients appear clinically normal, with
record may become indistinguishable from a hypoglycemic subtle deficits on neuropsychiatric testing, including changes in
state (6). According to Cadilhac and colleagues, the EEG abnor- attention and psychomotor speed (47). The demonstration of
malities may persist for several days despite effective treatment minimal hepatic encephalopathy may correlate with decreased
(4). Occasionally, epileptogenic lesions may result from coma- ability to drive and decreased quality of life.
tose states, but such sequelae are more likely to be caused by the The biochemical and neuropathologic mechanisms
hypoglycemic state rather than hyperglycemic state (34). of hepatic encephalopathy are still being elucidated.
A distinction has to be made between ketotic and nonketotic Hyperammonemia and glutamate alterations are strongly sus-
hyperglycemia. The latter entity was described by DiBenedetto pected in both acute and chronic hepatic encephalopathy, as is
and colleagues and appears to be rich in neurologic complica- the role of increased GABA tone. Shawcross and Jalan summa-
tions (35). Epileptic seizures of focal character are common in rized the ammonia hypothesis, detailing the role of hyperam-
this condition (36–38), as are, above all, focal motor seizures monemia in contributing to increased astrocytic pH that results
with rolandic EEG spike foci and contralateral clonic motions in calcium-dependent release of glutamate, which contributes
(39,40). The focal motor seizures may be movement induced to NMDA activation and excitotoxicity (47). In contrast, with
(41,42). Other focal seizures including visual hallucinations chronic liver disease, hyperammonemia contributes to
with forced head and eye deviation and aphasia may also rarely decreased chronic astrocytic glutamate stores, resulting in inac-
occur (43–45). The hyperglycemic states do not have to be non- tivation of the glutamate transporter and decreased postsynap-
ketotic in order to cause focal seizures, but in such cases, a tic glutamate receptors on neurons and astrocytes, thereby
ketotic nonacidotic situation is found (36). Correction of the contributing to increased cerebral inhibition. However, these
hyperglycemic state with insulin, rather than treatment with authors also note that there is increased GABAergic cerebral
anticonvulsant medications, controls the epileptic seizures. tone that also contributes to chronic cerebral inhibition in liver
cirrhosis. Ahboucha and Butterworth reviewed PET studies
LIVER DISEASE (HEPATIC that suggest a role for increased GABAA receptor activation, also
ENCEPHALOPATHY) resulting in increased postsynaptic neuron inhibition (48).
Neuropathologic correlates of acute and chronic liver disease
Liver disease may produce variable degrees of EEG abnormali- include astrocytic and cytotoxic edema with the former and
ties, from mild slowing to isoelectric activity. The presence or Alzheimer II astrocytic change in the latter (47).
absence of notable EEG changes is dependent on the patho- The altered pathophysiology of hepatic encephalopathy may
physiologic mechanisms involved. For this reason, some discus- be demonstrated by both evoked potentials and brain MRI, as
sion of the pathophysiologic mechanisms and their clinical well as EEG. The varying dynamics of cognitive brain dysfunc-
expression is needed. tions are mirrored by altered event-related P300 potentials,
A distinction must be made between the concept of hepatic even with a minor degree of dysfunction and with less sensitiv-
encephalopathy and acute massive hepatic cellular failure. Portal- ity to medication use than the EEG (49). Even in the absence of
systemic hepatic encephalopathies are based on a portocaval overt hepatic encephalopathy, brain MRI may be abnormal and
anastomosis, whereby protein from gastrointestinal venous demonstrate low-grade cerebral edema (50).
absorption bypasses hepatic metabolism and is carried to the The electroencephalographic changes of hepatic encephalopa-
systemic venous system. This condition is found in chronic cir- thy have been well known since the studies of Foley et al. and
rhosis of the liver with portal-systemic venous shunts. In con- Bickford and Butt (2,3). The degree of slowing often parallels the
trast, acute fulminant hepatic failure results in sudden level of blood ammonia (51). Similarly, the degree of EEG slow-
hepatocellular breakdown and dysfunction. In both situations, ing correlates with severity of liver disease (52). Amodio and
toxic (mainly nitrogenous) substances reach the brain and con- Gatta summarized the common changes associated with hepatic
tribute to the cerebral pathology and clinical symptoms encephalopathy, including low-frequency alpha background
described below. In advanced stages, the distinction between rhythms with sudden intermittent random theta waves superim-
both the pathophysiologic and EEG changes of a full-fledged posed in either a temporal, frontal, or generalized distribution
Chapter 21 ■ Metabolic Disorders and EEG 397

Figure 21.2 Hepatic encephalopathy in a 42-year-old patient.


Predominant activity around 2.5 Hz, most notable over the frontocen-
tral regions. Several triphasic waves are seen in channels 4 and 15 of
the right portion of the tracing.
Figure 21.1 Hepatic encephalopathy in a 59-year-old patient. The EEG
demonstrates moderate slowing of the background rhythm (5 to 6 Hz),
with some intermixed 8 to 10 Hz alpha waves of lower voltage.
More recently, several authors have questioned the diagnos-
tic specificity of triphasic waves for hepatic encephalopathy.
Fisch and Klass demonstrated triphasic waves in numerous
(53). Subsequently, slow theta background activity may predom- conditions, including stroke, hypertensive encephalopathy,
inate (3,9,54–57). As the clinical presentation worsens, the EEG hyper/hyponatremia, hypercalcemia, hypoglycemia, encephali-
may progressively show more disorganization with diffuse and tis, cerebral abscess, septic shock, dementia, lithium toxicity,
asynchronous theta and delta slowing over both hemispheres. and the postictal state. However, no specific characteristic of the
With severe coma, the background activity may decrease in triphasic wave morphology could reliably differentiate the eti-
amplitude and frequency to the point of becoming isoelectric ology of the triphasic wave from the possibilities noted above
(53,58). Girier and colleagues investigated the EEG findings in (63). Aguglia and colleagues and Blatt and Brenner similarly
fulminant hepatic failure in children and described the stages of noted the occurrence of triphasic waves with etiologies ranging
progressive EEG deterioration (59). Inferences with regard to the from stroke, brain tumors, Binswanger’s subcortical
need for liver transplant can be made on the basis of the EEG encephalopathy, and lithium-treated aged psychiatric patients
findings. Figure 21.1 demonstrates the mild-to-moderate degree (64,65). More recently, Kenangil and colleagues and Eriksson
of slowing that may be seen with hepatic encephalopathy, while
Figure 21.2 demonstrates more marked slowing.
Bickford and Butt introduced the term triphasic waves, clas-
sically described as moderate to high-amplitude slow waves
occurring with a frequency of 1.5 to 3 Hz with an initial sharp
positive transient, followed by a smaller negative component
(Figs. 21.3 and 21.4) (3). The discharges are often noted to
have a frontal to occipital delay in expression, occur singly or
in runs lasting 0.5 to 2 seconds, and arise from a slowed and
disorganized background (60). Triphasic waves may be seen in
fully awake patients or may commonly occur with worsening
cognitive status. However, these waveforms resolve with sleep. Figure 21.3 Examples of triphasic waves taken from two patients with
In general, the incidence of triphasic waves in hepatic hepatic encephalopathy. Note wave morphology. The numbers 1, 2, and
encephalopathy lies around 25% (57,61,62). 3 indicate the phase of the triphasic wave.
398 Part III ■ Clinical EEG: General

schemes with neuropsychiatric test results, severity of liver dis-


ease, biochemical indices of liver function, and risk of death
(76). While all classification schemes, including visual assess-
ment and spectral analysis, were appropriately correlated with
these variables, the greatest reliability was noted with automatic
quantification systems. In addition, spectral EEG analysis has
also been used to identify minimal hepatic encephalopathy fea-
tures, including an increase in theta power in the posterior divi-
sions and less commonly a decrease in mean dominant
frequency and increase in relative delta power (53).
EEG spectral analysis may help identify the initial changes
associated with minimal hepatic encephalopathy, and the EEG
may assist with the prognosis and treatment response to liver
transplantation. Interestingly, while spectral EEG analysis may
improve after orthotopic liver transplantation, neuropsychi-
atric impairments may remain. As cerebral disturbances after
liver transplant are not uncommon, caused by factors such as
general anesthetics, impaired liver function, sepsis, or hypoxia
(77), the EEG may be useful in the assessment of cerebral dys-
function and possible electrographic seizures. In addition, the
EEGs of post-transplant patients who do not survive demon-
strate a greater incidence of epileptiform abnormalities than
survivors (78). Thus, the EEG may assist with the differential
Figure 21.4 Triphasic waves of unusually paroxysmal configuration in diagnosis of post-transplant cerebral disturbances and assist
a 59-year-old patient with hepatic encephalopathy and no history of with post-transplant prognosis.
seizures. Also note the distribution of the potentials and a posterior Despite the paroxysmal appearance of triphasic waves, clini-
maximum of the triphasic waves; this is not altogether uncommon. cally overt epileptic manifestations are not very common with
hepatic encephalopathy and occur much less frequently than in
and Wictor also described the occurrence of triphasic waves renal encephalopathies (79–81). Surprisingly, however, Adams
with bilateral paramedian thalamic stroke and CNS Lyme dis- and Foley found an approximately equal incidence of seizures
ease, respectively (66,67). Karnaze and Bickford also reviewed in renal and hepatic failure (82). Similarities between triphasic
the medical records of patients whose EEGs demonstrated waves and spike-wave complexes cannot be denied. The bound-
triphasic waves (68). Of the 50 patients identified, more than aries between these abnormalities were discussed by Hughes,
half had hepatic encephalopathy as the likely etiology of the Klass, and Ghigo (83–85). Ficker et al. demonstrated various
triphasic waves. However, other associated conditions included forms of spikes, focal sharp waves, and generalized spike waves
renal failure (10), anoxia (9), hyperosmolarity (2), and hypo- in patients with hepatic encephalopathy (86). These patients
glycemia (1). Of note, only 43% of hepatic encephalopathy also had epileptic seizures and poor outcomes. In addition,
patients demonstrated all the diagnostic features of triphasic Eleftheriadis and colleagues also discussed the case of a gentle-
waves noted above. man presenting with continuous generalized tonic–clonic
The presence of triphasic waves appears to be a poor prog- seizures in the setting of decompensated hepatitis C liver cir-
nostic indicator. Karnaze and Bickford found that patients with rhosis (87). Therefore, a continuum of epileptiform discharges
triphasic waves experienced mortalities ranging from 30% with and electrographic seizures may be seen with hepatic
renal encephalopathy, 60% with hepatic encephalopathy to encephalopathy.
100% with anoxia (68). Bahamon-Dussan and colleagues simi- Chronic hepatocerebral degeneration may lead to a variety of
larly found that patients with triphasic waves, regardless of eti- EEG changes. The acquired form is neurologically characterized
ology, had a 50% 1-month mortality rate and 77% mortality by dysarthria, ataxia, intention tremor, and choreoathetosis
rate at 22 months of follow-up (69). However, these studies affecting chiefly the cranial musculature (88,89). This clinical
were performed over 20 years ago and advances in medicine entity is considered to occur much more frequently than the
may have lowered the mortality rates associated with these con- familial form (Wilson disease) (88). The acquired form may
ditions. No recent evaluation of the prognostic significance of develop in various chronic liver diseases and often leads to the
triphasic waves has been published. above-described picture of hepatic encephalopathy. Such a
Quantification of EEG data has been done by some authors development is less common in Wilson disease. Nevsimalova
(9,55,70–75). Amodio and colleagues recently compared differ- and colleagues found only mild alterations in EEG recordings
ent EEG hepatic encephalopathy classification systems (modi- of patients diagnosed with Wilson disease, with predominantly
fied Parsons-Smith, Conn’s, spectral analysis according to Van generalized intermittent theta or delta slowing, occasionally
der Rijt and ANNES [artificial neural network expert system]) with associated sharp waves (90). Only 21% of patients
to assess for correlations between the individual classification demonstrated focal findings, of which 15% demonstrated focal
Chapter 21 ■ Metabolic Disorders and EEG 399

epileptiform discharges. Giagheddu and colleagues noted In chronic uremia, patients may experience decreased
abnormal EEGs in only 3 of 23 patients studied. These record- energy, poor memory, irritability, and agitation (100). The
ings demonstrated temporal theta slowing, left paramedian EEG most typically demonstrates a slow and disorganized
epileptiform activity (in a patient with a prior thalamotomy), background, with more pronounced slowing associated with
and diffuse slow activity with triphasic waves (in a patient expe- worsening symptoms of uremia and a prior history of malig-
riencing symptoms of hepatic encephalopathy) (91). nant hypertension (101–106) (Fig. 21.5). While Hughes noted
the closest parallel between increased urea concentrations and
RENAL DISEASE (RENAL more marked background slowing, this association was mini-
ENCEPHALOPATHY, UREMIA) mal and of limited clinical significance (100). Rarely, theta dis-
charges, sharp waves, and polyspike and wave complexes are
Various gradations of EEG abnormalities have been reported in noted (103,104). Photic stimulation may result in photomy-
both acute as well as chronic renal failure and may potentially oclonic or photoparoxysmal responses (103). Notably, children
be reversed with renal transplantation. with chronic uremia similarly demonstrate an increase in theta
In acute uremia, the neurologic picture is characterized by and delta and a decrease in beta activity with spectral EEG
agitation, confusion, tremor, fasciculations, myoclonus, and analysis (107), while only 20% of children demonstrate low-
coma (92). The EEG may demonstrate irregular low-voltage amplitude spikes or bilateral frontocentral spike and wave dis-
activity with slowing of the posterior basic rhythm and occa- charges (108).
sional theta bursts. Other cases show prolonged bursts of bilat- Patients with chronic renal failure also experience alterations
eral synchronous mixed slow and sharp activity or frank spikes. in their sleep, with a decrease in deep, rapid eye movement
Bilateral spike discharges may or may not be associated with (REM), and total sleep times, resulting in increased difficulty
widespread myoclonic jerking. Grand mal and partial seizures classifying sleep stages (109,110). Long bursts of high-voltage
as well as infrequent nonconvulsive status epilepticus may 12 to 13/sec waves with enhanced vertex sharp activity in
develop (93,94). About 10% to 30% of patients with renal drowsiness, lack of spindles (14/sec) in stage 2 sleep, and pro-
insufficiency develop epileptic seizures (95–97). Seizures are longed high-voltage slow bursts with awakening have been
usually due to water–electrolyte imbalance (98,99), although reported (111). In REM sleep, ocular movements are enhanced;
they may also occur in the setting of medication side effects, they may even be continuous and associated with blinking and
infection, malignant hypertension, or dialysis disequilibrium brief myoclonic motions. However, with hemodialysis, deeper
(95,96). stages of sleep are attained (110).

Figure 21.5 Left: Encephalopathy due to chronic renal failure (age 50 years). Note intermittent runs of delta slowing.
Right: The same patient in sleep. Trains of delta waves with superimposed spindles, not exceeding the normal limits
of variability. The tendency toward normalization in sleep is found in most forms of metabolic encephalopathy.
400 Part III ■ Clinical EEG: General

Dialysis helps to maintain patients with chronic renal failure, There is little EEG documentation of the ictus (126). In the
although it may result in transient worsening of EEG patterns, acute stage of eclampsia, the posterior alpha rhythm is seldom
with an increase in background slowing, bursts of frontal slow recorded, likely due to the preferential localization of the main
waves, and rarely, spike and wave complexes (100,101,112). process in the occipital lobes (127). This is congruent with clin-
However, with successive dialysis treatments, a slow, delayed ical findings of cortical blindness (128,129) as well as the MRI
increase in background frequency is noted (105). Occasionally, findings described above. In a group of 84 patients, Thomas et
dialysis disequilibrium syndrome may occur, typically at the al. found the interictal EEG to be abnormal in about 80% of
onset of dialysis therapy or with rapid and highly effective treat- patients with eclampsia (130) and the abnormalities included
ment (95). The symptoms may include irritability, nausea, diffuse slowing (56.4%), focal slowing (25.6%), spike discharges
headache, blurred vision, asterixis, confusion, and generalized (14.1%), alpha coma (1.3%), and electrical silence (2.6%). A
convulsions, which may occur within 24 hours of the dialysis study to compare cerebral magnetic resonance and interictal
session. The entity is felt to result from a rapid systemic clear- EEG findings in preeclamptic versus eclamptic women by
ance of urea, causing cerebral edema due to slower cerebral Osmanagaoglu et al. found that EEG abnormalities occurred at
clearance. The risk for dialysis disequilibrium is diminished with statistically higher rates in the eclamptic group than the
the use of aluminum-free dialysate. With chronic dialysis treat- preeclamptic group (131). In the group with mild preeclampsia,
ment, dialysis encephalopathy may also occur, characterized by 20% of patients demonstrated EEG abnormalities, including
speech and memory difficulties, psychiatric changes, involun- diffuse slowing, focal slowing, or hemispheric slowing, while in
tary movements, seizures, and death (100,113). The EEG fre- the group with severe preeclampsia, 36% of patients showed
quently reveals a slow and disorganized background rhythm, these abnormalities as well as focal discharges. In the eclamptic
frontal intermittent rhythmic delta activity, or paroxysmal fron- group, the percentage of patients showing EEG abnormalities
tocentral spike and wave discharges (100,103,113,114). was significantly higher at 83%, and the abnormalities included
Fortunately, renal transplantation may reverse the EEG diffuse slowing, focal slowing, focal spike discharges, and multi-
background slowing in as little as 2 weeks postsurgery (106). focal spike discharges. Although focal MRI abnormalities were
While there have been various reports of post-transplant noted in many cases, the EEG abnormalities tended to be diffuse.
seizures, more recent studies suggest an incidence of 2.3% in The authors found no significant correlation between EEG
adults (115) and none in children (116), presumably due to abnormalities and mean arterial blood pressure (130,132). Of
improved post-transplant care. Seizures were more commonly the 14 patients with abnormal EEGs, 94% of patients demon-
attributed to immunosuppressive agents, but also due to infec- strated resolution of these abnormalities at 1-month follow-up.
tion, hypertension, and biochemical alterations (116). Magnesium sulfate is frequently used in the treatment of
women with eclampsia and is typically administered during
ECLAMPSIA GRAVIDARUM labor and the 12 to 24-hour postpartum period (133).
Although a clinical benefit has been shown with treatment in
Eclampsia gravidarum refers to the development of one or this setting, magnesium infusion does not alter the EEG abnor-
more generalized convulsions and/or coma in pregnant or post- malities before, during, or after the infusion (132). A report by
partum patients with preeclampsia but in the absence of other Brophy described normalization of the EEG in eclampsia with
neurologic conditions (117). Preeclampsia is a syndrome char- pyridoxine infusion (134); however, there is currently no clear
acterized by hypertension and proteinuria that typically occurs preventive therapy for preeclampsia (117).
after 20 weeks gestation. A number of target organs are affected, Seizures occur acutely during eclampsia and chronically lin-
including the brain, liver, kidney, and placenta (118). The gering epileptic seizures known as “posteclamptic epilepsy” as a
underlying factors involved in the pathogenesis of preeclampsia consequence of eclampsia are rare (135).
are complex, involving a combination of maternal, fetal, and
placental factors. More specifically, abnormalities in the devel-
opment of placental vasculature early in pregnancy are thought ELECTROLYTE DISTURBANCES
to lead to release of angiogenic factors into the systemic circu-
lation causing maternal endothelial dysfunction and ultimately Hypocalcemia
systemic vasoconstriction (119,120). Similarly, vasoconstriction Hypocalcemia is frequently the consequence of hypoparathy-
or vasospasm is seen in the cerebral blood vessels, which has roidism and manifests with tetany of peripheral origin with
been demonstrated on cerebral angiography (121,122), and is carpopedal spasms and seizures due to CNS hyperexcitability. It
believed to be the cause of the observed pathologies, namely is important to emphasize that tetanic spasms are of peripheral
focal necrosis, hemorrhage, and vasogenic edema (123,124). origin rather than cerebral origin as the EEG is normal in
Neuroimaging with brain MRI also shows areas of cerebral tetany. In the CNS, hypocalcemia is highly epileptogenic, pro-
ischemia, infarction, hemorrhage, or edema, in the watershed ducing marked EEG changes with slowing and generalized
regions, with a particular predilection for the parieto-occipital bursts of spikes (Fig. 21.6). Corriol et al. found epileptic mani-
lobes (125). Given these findings, loss of cerebral autoregulation festations develop at calcium levels around 5 to 6 mg/dL in
leading to hypertensive encephalopathy is thought to play a key humans and animal models (136). Seizures tend to be general-
role in the pathogenesis of eclamptic convulsions (117). ized, although partial seizures may also occur and typically
Chapter 21 ■ Metabolic Disorders and EEG 401

triphasic waves (143,146–148), with or without diffuse slowing of


the basic rhythm (147). In general, epileptiform discharges are
not noted except for a case described by Huott et al. in which
massive spike activity was seen independently in the right and left
occipital regions in the setting of severe hypercalcemia (16.6
mg/dL), as well as two generalized seizures, and a right homony-
mous hemianopsia (149). The presence of occipital spike-slow-
wave complexes raises the suspicion for a vasospastic effect in the
posterior circulation associated with high calcium levels (150).

Hyponatremia and Water


Intoxication
Hyponatremia and acute water intoxication lead to severe EEG
Figure 21.6 Hypocalcemia (ca. 5.2 mg/ 100 mL). Note marked diffuse abnormalities with pronounced generalized slowing. Typically,
slowing along with scattered sharp activity. the rate or sodium change is more important than the absolute
degree of hyponatremia as the determining factor in the devel-
respond poorly to anticonvulsants; however, both the seizures opment of mental status changes, EEG abnormalities, and
and EEG abnormalities improve once normal serum calcium is seizures. Zwang and Cohn reported a case of acute compulsive
restored (137). The typical interictal EEG in the setting of water intake resulting in a serum sodium level of 116.4 mg/dL,
hypocalcemia shows high-amplitude delta activity and may be causing convulsive status epilepticus, papilledema, and an EEG
accompanied by focal spike and spike-wave discharges, all of with diffuse 0.5- to 2-Hz activity (151). Despite rapid normal-
which may be enhanced by hyperventilation (137). However, ization of the electrolytes, EEG recovery was slow. Okura et al.
no clear correlation was found among calcium serum level, also reported a case of convulsive status epilepticus due to poly-
onset of seizures, and EEG changes in other studies (138). dipsia, where the EEG demonstrated delta waves with spike-
Neonatal hypocalcemia produces acute epileptic seizures that wave complexes, but the EEG improved quickly as his
tend to be focal, with shifting focality and severe transient EEG hyponatremia was corrected (152). Other abnormalities have
changes, such as focal rhythmic, high-voltage, frontocentral dis- been described such as bursts of high-voltage rhythmic delta
charges that generalize rapidly and often lasting several seconds activity, central high-voltage 6- to 7-Hz activity with stimula-
(139). A case reported by Kossoff showed seizures consisting of tion-induced paroxysms of delta waves (153), periodic delta
3- to 4-Hz spike and wave discharges that resolved after intra- waves (154), triphasic waves and periodic lateralized epilepti-
venous calcium administration (140). Rachitic tetany (common form discharges (155,156), and absence status epilepticus (157).
infantile tetany) is associated with hypocalcemia, carpopedal
spasms, laryngospasm, convulsions, and the clinical signs of rick- Hypernatremia
ets; the EEG changes are usually mild with no spike activity (141). While hyponatremia is likely to be the underlying etiology of
In pseudohypoparathyroidism with short stature, osteo- seizures, hypernatremia may result as a consequence of seizures
chondral dysplasia, short fourth finger, hypocalcemia, and (158). Hypernatremia is associated with reduction of cerebral
tetany, the EEG is most often abnormal as in cases of hypocal- volume as water shifts from the intracellular space to the extra-
cemia (142,143). No significant EEG changes have been found cellular space, and again the rate of electrolyte change is more
in pseudopseudohypoparathyroidism. important than the absolute degree of hypernatremia as the
The EEG was found to be normal in five of six patients with determining factor in the development of clinical symptoms. In
Fahr disease; the remaining patient showed mild slowing (144). general, seizures are not seen except in cases of rapid correction
of hypernatremia. The EEG usually demonstrates mild nonspe-
Hypercalcemia cific slowing (159).
Hypercalcemia may be due to hyperplasia or neoplasm of the
parathyroid glands, excessive parathyroid activity caused by
Hypomagnesemia
renal failure or skeletal decalcification, vitamin D intoxication, Hypomagnesemia causes cardiac arrhythmias, hyperexcitability
paraneoplastic syndrome, or malignancy with bony metastases. of the peripheral nervous system (tetany), and hyperexcitability
Hypocalcemia is associated with CNS hyperexcitability, of the CNS (seizures, mental status changes, focal deficits). It is
whereas hypercalcemia is associated with depression of the CNS often associated with hypocalcemia since magnesium defi-
and epileptic activity rarely occurs. Clinically, irritability, ciency inhibits calcium mobilization via inhibition of secretion
lethargy, and malaise are seen with mild elevations of serum cal- and action of the pararathyroid hormone, causing secondary
cium, but as levels continue to rise, patients progress to coma. hypocalcemia (160). The effects of hypomagnesemia are
When calcium levels reach about 13 mg/dL, EEG changes start to thought to be potentiated by hypokalemia as well (160). Low
appear (145). Neurologic deficits and mental impairment are serum magnesium levels can result from excessive diuretic use,
accompanied by EEG abnormalities consisting mainly of bursts renal disease, inadequate intake, or impaired gastrointestinal
of frontal-dominant delta frequency activity and less frequently absorption, as in cases of malabsorption syndromes and
402 Part III ■ Clinical EEG: General

postgastrointestinal surgeries. Magnesium serves as a neuronal slowing with theta and delta activity that is reversible with
membrane stabilizer, and it is indicated in the treatment of niacin supplementation (170).
eclampsia (see the section “Eclampsia Gravidarum”). Unlike the
previously mentioned electrolyte disturbances, detailed studies Vitamin B6
of EEG changes with hypomagnesemia are lacking. Clinical Vitamin B6 (pyridoxine) deficiency on a nutritional basis has been
symptoms, including epileptic seizures, typically do not develop recognized as a rare cause of severe and even fatal convulsions in
until the magnesium level falls below 1.2 mg/dL (161). neonates and infants (171,172). Pyridoxine dependency develops
Although hypomagnesemia is a systemic disturbance, patients during fetal life as a genetic disorder and causes both intrauterine
may present with focal neurologic deficits, as was reported in and postnatal seizures (173–175). EEGs of affected patients show
the case of a young child who presented with hemiparesis and generalized spike activity. Both the EEG abnormalities and clini-
aphasia in the setting of severe hypomagnesemia and hypocal- cal seizures respond quickly to treatment with pyridoxine.
cemia. His EEG demonstrated background slowing in the EEG abnormalities have also been reported in controlled
affected hemisphere (162), which resolved along with his clini- studies of B6 depletion by Kretsch et al. in which healthy young
cal symptoms when his magnesium and calcium levels were women were placed on a diet containing less than 0.05 mg of B6
restored to the normal range. (176). Within 12 days, 2 of 11 study subjects exhibited abnor-
mal EEG patterns, including focal slowing and focal sharp wave
VITAMIN DEFICIENCIES activity, which resolved with vitamin B6 repletion, but no clear
seizures were seen.
Vitamin B1
Vitamin B12
Thiamine (vitamin B1) deficiency may give rise to manifesta-
tions in the cardiovascular and nervous systems. There are two Vitamin B12 (cobalamin) deficiency gives rise to subacute com-
forms: (i) dry beriberi, which consists of sensorimotor neu- bined degeneration characterized by changes in the peripheral
ropathy and Wernicke–Korsakoff syndrome, and (ii) wet and CNS. Patients present with large fiber peripheral neuropa-
beriberi, which consists of edema and congestive heart failure, thy and may exhibit hyperreflexia and mental status changes
but little CNS manifestations. The clinical picture of Wernicke’s such as delirium or dementia. Slow activity in the EEG may be
encephalopathy, with or without Korsakoff syndrome, is fre- quite pronounced (177,178) and temporal spike and sharp wave
quently encountered in alcoholics, with predominant oculomo- activity has been reported (178); but these findings are unrelated
tor and cerebellar symptomatology (163), although it can also to the presence of myelopathy or neuropathy (179). Pronounced
be seen in other conditions, such as hyperemesis, dialysis, and epileptiform EEG abnormalities have been reported in the early-
postgastrointestinal surgery. onset cobalamin C/D deficiency: an inborn error of intracellular
EEG changes range from mild to severe and, in general, paral- cobalamin metabolism with high plasma levels of methyl-
lel the severity of the neurologic deficits (164). Initial alpha slow- malonic acid, homocystine, and homocysteine (180). The EEG
ing is gradually followed by the appearance of theta and delta demonstrates massive focal or generalized spikes and the cere-
frequencies (165) and by diffuse sharp and slow wave complexes bral MRI shows diffuse loss of white matter.
in severely ill patients (166). In a group of 50 adult patients, 16
patients with neurologic manifestations demonstrated epilepti- ENDOCRINE DISORDERS
form activity on their EEGs (167). Thiamine deficiency in infants
is rare in developed countries and occurs mainly in breast-fed
Adrenal Cortex
infants of mothers who have inadequate thiamine intake or in Adrenocortical insufficiency (Addison disease) can be associ-
infants fed with thiamine-deficient formula (168). In a group of ated with pronounced background slowing with delta and theta
20 Israeli children who developed infantile thiamine deficiency, activity, but such changes are found in severe cases only
seven patients subsequently developed epilepsy (169). Upon ini- (181,182). In milder forms, the EEG shows little or no abnor-
tial presentation, all patients were noted to have episodes of mality. In severe cases, loss of reactivity to eye opening, increase
apnea, increased tone, and clinical seizures that were tonic, sensitivity to hyperventilation, and decreased beta activity also
myoclonic, or focal in nature. Initial EEG recording was available can be seen (183). Cortisone can dramatically improve the clin-
in six of the seven patients and demonstrated diffuse background ical and electroencephalographic picture, and it has been sug-
slowing, with or without lateralized epileptiform activity such as gested that the EEG abnormalities are caused mainly by altered
sharp waves or spikes, and in one case, a focal electrographic glucose metabolism.
seizure in the frontal region was recorded. The seizures resolved In adrenal cortical hyperfunction (Cushing disease), EEG
with thiamine repletion; however, after a seizure-free period of 1 abnormalities are less prominent than in Addisonian states. Slow
to 9 months, all children had recurrent focal motor seizures, as well as fast frequencies may be excessively developed (184).
complex partial seizures, or myoclonic seizures. No specific metabolic disturbance can be attributed to the EEG
changes. There are also reports that therapeutic dosages of
Vitamin B3 adrenocorticotropic hormone (ACTH) or cortisone may pro-
Vitamin B3 (niacin) deficiency results in pellagra, which is char- duce mild-to-moderate EEG changes along with frequent men-
acterized clinically by dermatitis, diarrhea, depression, and tal changes and enhanced seizure susceptibility (185). In adult
dementia. Electrographically it is associated with diffuse EEG epileptics with abnormal EEG tracings, the EEG changes are
Chapter 21 ■ Metabolic Disorders and EEG 403

enhanced by intravenous cortisone (186), in contrast to the ben- tion may respond poorly to anticonvulsants unless the underly-
eficial effect of ACTH and cortisone in hypsarrhythmic infants. ing endocrine disturbance is properly treated. In thyrotoxic cri-
Pheochromocytomas may cause extremely high blood pres- sis, prominent EEG abnormalities have been noted with
sure values, but in these critical states, the EEG may remain encephalopathy prompted by severe infection and surgical
normal (187) unless hypertensive encephalopathy develops. intervention. Cranial nerve signs, acute myeloencephalopathy,
and choreic movements may also occur. In these cases, the EEG
Pituitary Gland may be dominated by marked slowing with superimposed fast
Postpartum necrosis of the anterior lobe of the pituitary gland activity (199), triphasic waves (183), or bursts of anterior delta
(Sheehan syndrome) is a common cause of severe hypopitu- frequency activity (200).
itarism. The EEG shows massive diffuse intermittent or contin-
uous theta and delta activity, especially in association with Hashimoto Encephalopathy
impaired consciousness, and these findings most likely reflect Hashimoto encephalopathy is a clinical syndrome of
the secondary depression of adrenocortical function (188). encephalopathy, elevated serum levels of antithyroid antibodies,
and may present in two subtypes—a vasculitic subtype with
Thyroid Gland stroke-like episodes and a diffuse progressive type with dementia
Hyperthyroidism and psychiatric symptoms (201,202). Both forms may be accom-
Hyperthyroidism is commonly associated with acceleration of panied by depressed mental status, myoclonus, and tremor (202).
alpha rhythm frequency (Fig. 21.7). Such observations can be Epileptic seizures (partial and generalized), complex partial sta-
traced back to Rubin et al. (189), Lindsley and Rubinstein (190), tus epilepticus, and epilepsia partialis continua have also been
and Ross and Schwab (191). Enhancement of fast activity was reported (203,204). In Hashimoto’s thyroiditis, diffuse EEG
reported by Gibbs and Gibbs (192), Vague et al. (193,194), and abnormalities can be seen such as background slowing (205,206).
Condon et al. (5). Wilson et al. found the highest incidence of
Hypothyroidism
abnormalities in premenopausal females (195). According to
Vague et al., rolandic mu rhythm may be augmented by hyper- Hypothyroidism may present with a variety of clinical condi-
thyroidism (194). tionals such as congenital myxedema, cretinism, and myxedema
Paroxysmal bursts and clinical seizures, such as complex par- in adulthood with complications such as myxedema coma. In
tial seizures, focal motor seizures, and generalized seizures, have myxedematous infants, a delay of the EEG development and
been reported by Skanse and Nyman (196), Jabbari and Huott especially of the sleep spindle development has been reported
(197), and Miyazaki (198). Administration of thyroxin pro- by Schultz et al. (207), and low voltage and excessive slowing
duces epileptic activity. Epileptic patients with thyroid dysfunc- have been noted by Niemanan (208). In adults, an excess of
low-voltage slow activity has been reported (191) and reactivity
may be poor or absent (209). Epileptic seizures may precede
myxedema coma (210), and various EEG patterns have been
noted: triphasic waves and generalized periodic sharp waves
resembling Creutzfeldt–Jakob disease (211,212).

Other Hormonal Disturbances


The physiologic influence of estrogens on the EEG has been
demonstrated by some investigators. Mild-to-moderate slowing
of the alpha rhythm was found at the end of pregnancy (213).
However, subsequent studies could not replicate these findings,
and the authors hypothesize that the clinically observed changes
may be reflective of a pregnancy-related disorder (214). Although
an initial report by Dusser de Barenne and Biggs in 1942 found
slowing of alpha 1 to 3 days prior to menstruation (215), subse-
quent studies have not yielded consistent results (216).

ACUTE INTERMITTENT PORPHYRIA


In patients with abdominal pain and polyneuropathy due to
acute porphyria, the EEG is normal. However, when delirium
and mental impairment develop, the EEG shows theta and delta
frequencies (217), and occasionally asymmetrical slowing and
spike or spike-wave activity can be seen (218). Pronounced
Figure 21.7 Hyperthyroidism in a 52-year-old woman. Note fast basic slowing is found in central forms of porphyria (Fig. 21.8).
rhythm of 14 to 16 Hz with good blocking effect. Faster activity over Seizures can be seen in approximately 3% to 5% of the
central regions does not show the blocking response. patients with acute intermittent porphyria (219) and their
404 Part III ■ Clinical EEG: General

indisputable diffuse EEG slowing. A variety of medical condi-


tions (such as pneumonia or influenza in aged patients) and
postsurgical conditions (excluding neurosurgical cases) play a
major role in the genesis of mixed-type encephalopathy. The
length of time of anesthesia augments the problem in postsur-
gical cases; thus, introjecting a pharmacologic element. This
mixed type of encephalopathy usually carries a good progno-
sis. A special form of mixed-type encephalopathies occurs in
30% of severe burns; in these cases, however, the prognosis is
guarded (222).

CHRONIC RESPIRATORY (PULMONARY


INSUFFICIENCY)
Chronic bronchial disease and pulmonary emphysema, along
with right cardiac failure (cor pulmonale), may lead to a
chronic encephalopathy clinically characterized by headache,
papilledema, elevated cerebral spinal fluid pressure, and twitch-
ing of extremities (8,223). The EEG shows considerable diffuse
delta and theta activity; occasionally, anterior delta waves may
show the configuration of triphasic waves. Appropriate treat-
ment may quickly reverse these changes, but oxygen adminis-
tration may be dangerous.
Chronic alveolar hypoventilation in conjunction with obe-
sity gives rise to the Pickwickian syndrome with excessive som-
nolence (224,225) (Fig. 21.9). Most of these patients also suffer
from sleep apnea, especially from the obstructive type (see
Chapter 41). The routine EEG shows abnormal amounts of
diffuse slow activity (Fig. 21.10).

Figure 21.8 A: Cerebral form of acute intermittent porphyria in a


24-year-old woman with slow mentation, personality change, and
grand mal seizures. B: Same patient, improving although still lethargic.
Note runs of rhythmical slowing over left frontotemporal region.
This is an example of occasional focal characteristics in metabolic
encephalopathies.

treatment can be problematic because of the well-known


induction of porphyria with the use of antiepileptic medica-
tions (220). However, gabapentin is considered an exception
and thus is the treatment of choice (221).

MIXED-TYPE ENCEPHALOPATHY
This polyetiologic condition (222) stands apart from all of the
well-defined metabolic encephalopathies. It is a common con-
dition, mostly acute, characterized simply by change of menta- Figure 21.9 Abnormally slow and disorganized waking record in a
tion (delirium) with little or no neurologic deficits but with patient with pickwickian syndrome.
Chapter 21 ■ Metabolic Disorders and EEG 405

Figure 21.10 Burst of diffuse delta activity during wakefulness in a patient with severe chronic obstructive pul-
monary disease.

REFERENCES 9. MacGillivray BB. The EEG in liver disease. In: Remond A, ed.
Handbook of Electroencephalography and Clinical Neurophysiology.
1. Berger H. Das Elektrenkephalogramm des Menschen. Arch Amsterdam: Elsevier; 1976:26–50.
Psychiatry Nervenkr. 1937;106:165–187. 10. Moruzzi G. Action de l’hypoglycémie insulinique sur l’activité
2. Foley JM, Watson CW, Adams RD. Significance of the electroen- spontanée et provoquée de l’écorce cérébrale. C R Soc Biol.
cephalographic changes in hepatic coma. Trans Am Neurol Assoc. 1938;128:1181–1184.
1950;51:161–165. 11. Himwich HE, Frostig JP, Hoagland H, et al. Clinical electroen-
3. Bickford RG, Butt HR. Hepatic coma: the electroencephalographic cephalographic and biochemical changes during insulin hypo-
pattern. J Clin Invest. 1955;34(6):790–799. glycemia. Proc Soc Exp Biol. 1939;40:401–402.
4. Cadilhac J, Ribstein M, Jean R. EEG et troubles métaboliques. Rev 12. Gellhorn E, Kessler M. The effect of hypoglycemia on the elec-
Neurol (Paris). 1959;100:270–296. troencephalogram at varying degrees of oxygenation of the blood.
5. Condon JV, Becka DR, Gibbs FA. Electroencephalographic Am J Physiol. 1942;136:1–6.
abnormalities in endocrine disease. N Engl J Med. 1954;251(16): 13. Davis PA. Effect on the electroencephalogram of changing blood
638–641. sugar level. Arch Neurol Psychiatry. 1943;49:186–194.
6. Krump JE, ed. Die Klinische Bedeutung des Elecktroenzephalogrammes 14. Hoagland H, Cameron DE, Rubin MA. The electroencephalogram
bei Vergiftungen, Endotoxikosen und Endokrinopathien. Proceed of schizophrenics during insulin treatments. Am J Psychiatry.
Deutsche Internisten-Tagung; Berlin: VEB Verlag Volk und 1937;94:183–208.
Gesundheit; 1955. 15. Gibbs FA, Williams D, Gibbs EL. Modification of the cortical
7. Thiebaut F, Rohmer F, Wackenheim A. Electroencephalographical frequency spectrum by changes in CO2, blood sugar and oxygen.
study of endocrine diseases. Electroencephalogr Clin Neurophysiol. J Neurophysiol. 1940;3:49–58.
1958;10(1):1–30. 16. Engel GL, Romano J, Ferris EG, et al. A simple method of deter-
8. Glaser GH. Metabolic encephalopathy in hepatic, renal and mining the frequency spectrum of the electroencephalograph.
pulmonary disorders. Postgrad Med. 1960;27:611–619. Observations on physiological variations in glucose, oxygen,
406 Part III ■ Clinical EEG: General

posture and acid base balance on the normal electroencephalo- 38. Vastola EF, Maccario M, Homan R. Activation of epileptogenic foci
gram. Arch Neurol Psychiatry. 1944;51:134–146. by hyperosmolality. Neurology. 1967;17(5):520–526.
17. Weinland WL, Weinland W. Űber elektroencephalographische 39. Aquino A, Gabor AJ. Movement-induced seizures in nonketotic
Beobachtungen beim Insulinschock. Arch Psychiatr Nervenkr. hyperglycemia. Neurology. 1980;30(6):600–604.
1949;18:24–44. 40. Singh BM, Strobos RJ. Epilepsia partialis continua associated
18. Lafon R, Passouant P, Billet M, et al. Étude électroencéphalo- with nonketotic hyperglycemia: clinical and biochemical profile of
graphique du coma insulinique. Ann Méd Psychol. 1950;2: 21 patients. Ann Neurol. 1980;8(2):155–160.
251–254. 41. Gabor AJ. Focal seizures induced by movement without sensory
19. Gibbs FA, Murray EL. Hypoglycemic convulsions. Electro- feedback mechanisms. Electroencephalogr Clin Neurophysiol.
encephalgr Clin Neurophysiol. 1954;6:674. 1974;36(4):403–408.
20. Hetzel H, Niedermeyer E. Der Gehirnhydrolysat-Weckeffekt im 42. Duncan MB, Jabbari B, Rosenberg ML. Gaze-evoked visual
hypoglykaemischen Koma und sein hirnelektrisches. Bild Arch seizures in nonketotic hyperglycemia. Epilepsia. 1991;32(2):
Psychiatr Nervenkr. 1955;76:369–382. 221–224.
21. Shagass C, Rowsell PW. Serial electroencephalographic and clini- 43. Harden CL, Rosenbaum DH, Daras M. Hyperglycemia presenting
cal studies in a case of prolonged insulin coma. AMA Arch Neurol with occipital seizures. Epilepsia. 1991;32(2):215–220.
Psychiatry. 1954;72(6):705–711. 44. Huang CW, Hsieh YJ, Pai MC, et al. Nonketotic hyperglycemia-
22. Regan PF III, Browne-Mayers AN. Electroencephalography, fre- related epilepsia partialis continua with ictal unilateral parietal
quency analysis and consciousness; a correlation during insulin- hyperperfusion. Epilepsia. 2005;46(11):1843–1844.
induced hypoglycemia. J Nerv Ment Dis. 1956;124(2):142–147. 45. Manford M, Fuller GN, Wade JP. “Silent diabetes”: non-ketotic
23. Zeigler DK, Presthus J. Normal electroencephalogram at deep lev- hyperglycaemia presenting as aphasic status epilepticus. J Neurol
els of hypoglycemia. Electroencephalogr Clin Neurophysiol. Neurosurg Psychiatry. 1995;59(1):99–100.
1957;9:523–526. 46. Knell AJ, Davidson AR, Williams R, et al. Dopamine and serotonin
24. Von Braunmühl A. Insulinschock und Heilkrampf in der Psychiatrie. metabolism in hepatic encephalopathy. Br Med J. 1974;1(5907):
Stuttgart: Wissenschaftliche Verlagsgesellschaft; 1947. 549–551.
25. Boyle PJ, Kempers SF, O’Connor AM, et al. Brain glucose uptake 47. Shawcross D, Jalan R. The pathophysiologic basis of hepatic
and unawareness of hypoglycemia in patients with insulin- encephalopathy: central role for ammonia and inflammation. Cell
dependent diabetes mellitus. N Engl J Med. 1995;333(26): Mol Life Sci. 2005;62(19–20):2295–2304.
1726–1731. 48. Ahboucha S, Butterworth RF. Pathophysiology of hepatic
26. Tribl G, Howorka K, Heger G, et al. EEG topography during encephalopathy: a new look at GABA from the molecular stand-
insulin-induced hypoglycemia in patients with insulin-dependent point. Metab Brain Dis. 2004;19(3–4):331–343.
diabetes mellitus. Eur Neurol. 1996;36(5):303–309. 49. Kugler CF, Petter J, Taghavy A, et al. Dynamics of cognitive brain
27. Tamburrano G, Lala A, Locuratolo N, et al. Electro - dysfunction in patients with cirrhotic liver disease: an event-
encephalography and visually evoked potentials during related P300 potential perspective. Electroencephalogr Clin
moderate hypoglycemia. J Clin Endocrinol Metab. 1988;66(6): Neurophysiol. 1994;91(1):33–41.
1301–1306. 50. Haussinger D, Schliess F. Pathogenetic mechanisms of hepatic
28. Bjorgaas M, Sand T, Vik T, et al. Quantitative EEG during con- encephalopathy. Gut. 2008;57(8):1156–1165.
trolled hypoglycaemia in diabetic and non-diabetic children. 51. Senzolo M, Amodio P, D’Aloiso MC, et al. Neuropsychological and
Diabet Med. 1998;15(1):30–37. neurophysiological evaluation in cirrhotic patients with minimal
29. Sperling MR. Hypoglycemic activation of focal abnormalities in hepatic encephalopathy undergoing liver transplantation.
the EEG of patients considered for temporal lobectomy. Transplant Proc. 2005;37(2):1104–1107.
Electroencephalogr Clin Neurophysiol. 1984;58(6):506–512. 52. Amodio P, Del Piccolo F, Petteno E, et al. Prevalence and prognos-
30. Deutsch E, Sohmer H, Weidenfeld J, et al. Auditory nerve-brain tic value of quantified electroencephalogram (EEG) alterations in
stem evoked potentials and EEG during severe hypoglycemia. cirrhotic patients. J Hepatol. 2001;35(1):37–45.
Electroencephalogr Clin Neurophysiol. 1983;55(6):714–716. 53. Amodio P, Gatta A. Neurophysiological investigation of hepatic
31. Kirchner A, Veliskova J, Velisek L. Differential effects of low glucose encephalopathy. Metab Brain Dis. 2005;20(4):369–379.
concentrations on seizures and epileptiform activity in vivo and in 54. Friedlander WJ. Electroencephalographic changes in hyperam-
vitro. Eur J Neurosci. 2006;23(6):1512–1522. monemia. Electroencephalogr Clin Neurophysiol. 1956;8(3):
32. Wang S, Hu HT, Wen SQ, et al. An insulinoma with clinical and 513–516.
electroencephalographic features resembling complex partial 55. Laidlaw J. The application in general medical conditions of a visual
seizures. J Zhejiang Univ Sci B. 2008;9(6):496–499. method of assessing and representing generalized electroen-
33. Dion MH, Cossette P, St-Hilaire JM, et al. Insulinoma misdiag- cephalographic abnormalities. J Neurol Neurosurg Psychiatry.
nosed as intractable epilepsy. Neurology. 2004;62(8):1443–1445. 1959;22(1):69–76.
34. Engel R, Halberg F, Tichy FY, et al. Electrocerebral activity and 56. Laidlaw J, Read AE, Sherlock S. Morphine tolerance in hepatic cir-
epileptic attacks at various blood sugar levels; with a case report. rhosis. Gastroenterology. 1961;40:389–396.
Acta Neuroveg (Wien). 1954;9(1–4):147–167. 57. Silverman D. Some observations on the EEG in hepatic coma.
35. Dibenedetto RJ, Crocco JA, Soscia JL. Hyperglycemia nonketotic Electroencephalogr Clin Neurophysiol. 1962;14:53–59.
coma. Arch Intern Med. 1965;116:74–82. 58. Trewby PN, Casemore C, Williams R. Continuous bipolar record-
36. Daniels JC, Chokroverty S, Barron KD. Anacidotic hyperglycemia ing of the EEG in patients with fulminant hepatic failure.
and focal seizures. Arch Intern Med. 1969;124(6):701–706. Electroencephalogr Clin Neurophysiol. 1978;45(1):107–110.
37. Maccario M, Messis CP, Vastola EF. Focal seizures as a manifesta- 59. Girier B, Clouzeau J, Navelet Y, et al. Fulminant hepatic failure
tion of hyperglycemia without ketoacidosis. A report of seven (FHF) in children. EEG prognosis. Electroencephalogr Clin
cases with review of the literature. Neurology. 1965;15:195–206. Neurophysiol. 1989;73:48P–49P.
Chapter 21 ■ Metabolic Disorders and EEG 407

60. Sheridan PH, Sato S. Triphasic waves of metabolic encephalopathy 81. Plum F, Posner JB. Diagnosis of Stupor and Coma. Philadelphia:
versus spike-wave stupor. J Neurol Neurosurg Psychiatry. 1986; F. A. Davis; 1966.
49(1):108–109. 82. Adams RD, Foley JM. Neurological complications of liver disease.
61. Rumpl E, Hackl JM, Gerstenbrand F, et al. Zum EEG im Publ Assoc Res Neuro Ment Dis. 1953;32:198–237.
Leberkoma. Z EEG-EMG. 1979;10:88–94. 83. Hughes JR. Principles in the differentiation of atypical spike-
62. Simsarian JP, Harner RN. Diagnosis of metabolic encephalopathy: waves and triphasic waves. Am J EEG Technol. 1990;30:309–312.
significance of triphasic waves in the electroencephalogram. 84. Klass DW. Principles in the differentiation of atypical spike-waves
Neurology (Minneapolis). 1972;22:456. and triphasic waves. Am J EEG Technol. 1990;30:313–314.
63. Fisch BJ, Klass DW. The diagnostic specificity of triphasic wave 85. Ghigo J. Principles in the differentiation of atypical spike-waves
patterns. Electroencephalogr Clin Neurophysiol. 1988;70(1):1–8. and triphasic waves: a technologist’s view. Am J EEG Technol.
64. Aguglia U, Gambardella A, Oliveri RL, et al. Nonmetabolic causes 1990;30:315–316.
of triphasic waves: a reappraisal. Clin Electroencephalogr. 86. Ficker DM, Westmoreland BF, Sharbrough FW. Epileptiform
1990;21(3):120–125. abnormalities in hepatic encephalopathy. J Clin Neurophysiol.
65. Blatt I, Brenner RP. Triphasic waves in a psychiatric population: a 1997;14(3):230–234.
retrospective study. J Clin Neurophysiol. 1996;13(4):324–329. 87. Eleftheriadis N, Fourla E, Eleftheriadis D, et al. Status epilepticus
66. Kenangil G, Orken DN, Yalcin D, et al. Triphasic EEG pattern in as a manifestation of hepatic encephalopathy. Acta Neurol Scand.
bilateral paramedian thalamic infarction. Clin EEG Neurosci. 2003;107(2):142–144.
2008;39(4):185–190. 88. Victor M. Neurologic changes in liver disease. In: Plum F, ed.
67. Eriksson B, Wictor L. EEG with triphasic waves in Borrelia Brain Dysfunction in Metabolic Disorders. New York: Raven Press;
burgdorferi meningoencephalitis. Acta Neurol Scand. 2007;116(2): 1974:1–12.
133–136. 89. Victor M, Adams RD, Cole M. The acquired (non-Wilsonian)
68. Karnaze DS, Bickford RG. Triphasic waves: a reassessment of their type of chronic hepatocerebral degeneration. Medicine. 1965;
significance. Electroencephalogr Clin Neurophysiol. 1984;57(3): 44(5):345–396.
193–198. 90. Nevsimalova S, Marecek Z, Roth B. An EEG study of Wilson’s dis-
69. Bahamon-Dussan JE, Celesia GG, Grigg-Damberger MM. ease. Findings in patients and heterozygous relatives.
Prognostic significance of EEG triphasic waves in patients with Electroencephalogr Clin Neurophysiol. 1986;64(3):191–198.
altered state of consciousness. J Clin Neurophysiol. 1989;6(4): 91. Giagheddu M, Tamburini G, Piga M, et al. Comparison of MRI,
313–319. EEG, EPs and ECD-SPECT in Wilson’s disease. Acta Neurol
70. Cohn R, Castell DO. The effect of acute hyperammonemia on the Scand. 2001;103(2):71–81.
electroencephalogram. J Lab Clin Med. 1966;68(2):195–205. 92. Cadilhac J. The EEG in renal insufficiency. In: Remond A,
71. Kennedy J, Parbhoo SP, MacGillivray B, et al. Effect of extracorpo- ed. Handbook of Electroencephalography and Clinical
real liver perfusion on the electroencephalogram of patients in Neurophysiology. Amsterdam: Elsevier; 1976:351–369.
coma due to acute liver failure. Q J Med. 1973;42(167):549–561. 93. Grill MF, Maganti R. Cephalosporin-induced neurotoxicity: clin-
72. Kurtz D, Zenglein JP, Imler M, et al. Night sleep in porto-caval ical manifestations, potential pathogenic mechanisms, and the
encephalopathy. Electroencephalogr Clin Neurophysiol. 1972;33(2): role of electroencephalographic monitoring. Ann Pharmacother.
167–178. 2008;42(12):1843–1850.
73. MacGillivray BB. An EEG monitor incorporating simple pattern 94. Chuang CL, Chen KP, Kwan SY, et al. Creutzfeldt-Jakob-like EEG
recognition. J Physiol. 1969;201:65–67. in a patient with end-stage renal failure. Nephrol Dial Transplant.
74. Van der Rijt CC, Schalm SW, De Groot GH, et al. Objective meas- 2004;19(1):252–254.
urement of hepatic encephalopathy by means of automated EEG 95. Lacerda G, Krummel T, Sabourdy C, et al. Optimizing therapy of
analysis. Electroencephalogr Clin Neurophysiol. 1984;57(5): seizures in patients with renal or hepatic dysfunction. Neurology.
423–426. 2006;67(12 suppl 4):S28–S33.
75. Yaar I, Shapiro MB, Pottala EW. Spectral analysis of the EEG in 96. Boggs JG. Seizures in medically complex patients. Epilepsia.
hepatic encephalopathy treated with levodopa. Electroencephalogr 1997;38(suppl 4):S55–S59.
Clin Neurophysiol. 1981;52(6):617–625. 97. McEnery PT, Nathan J, Bates SR, et al. Convulsions in children
76. Amodio P, Pellegrini A, Ubiali E, et al. The EEG assessment of low- undergoing renal transplantation. J Pediatr. 1989;115(4):
grade hepatic encephalopathy: comparison of an artificial neural 532–536.
network-expert system (ANNES) based evaluation with visual 98. Glaser GH. Brain dysfunction in uremia. In: Plum F, ed. Brain
EEG readings and EEG spectral analysis. Clin Neurophysiol. Dysfunction in Metabolic Disorders. New York: Raven Press;
2006;117(10):2243–2251. 1974:3173–3197.
77. Lehmkuhl P, Kaukemuller J, Pohl S, et al. EEG monitoring 99. Zuckermann EG, Glaser GH. Urea-induced myoclonic seizures.
of intensive care patients after liver transplantation. An experimental study of site of action and mechanism. Arch
Electroencephalagr Clin Neurophysiol. 1988;69:17P. Neurol. 1972;27(1):14–28.
78. Wszolek ZK, Aksamit AJ, Ellingson RJ, et al. Epileptiform elec- 100. Hughes JR. Correlations between EEG and chemical changes in
troencephalographic abnormalities in liver transplant recipients. uremia. Electroencephalogr Clin Neurophysiol. 1980;48(5):583–594.
Ann Neurol. 1991;30(1):37–41. 101. Lavy S, Aviram A. EEG changes in uraemic patients undergoing
79. Bauer G, Niedermeyer E. Acute convulsions. Clin Electroencephalogr. regular haemodialysis. Electroencephalogr Clin Neurophysiol.
1979;10(3):127–144. 1969;27(2):217.
80. Gastaut H, Rohmer F, Cossette A, et al. Introduction to the study 102. Spehr W, Sartorius H, Berglund K, et al. EEG and haemodialysis. A
of functional generalized epilepsies. In: Gastaut H, Jasper HH, structural survey of EEG spectral analysis, Hjorth’s EEG descrip-
Bancaud J, et al., eds. The Physiopathogenesis of the Epilepsies. tors, blood variables and psychological data. Electroencephalogr
Springfield, IL: Charles C Thomas; 1969:15–25. Clin Neurophysiol. 1977;43(6):787–797.
408 Part III ■ Clinical EEG: General

103. Markand ON. Electroencephalography in diffuse 126. Guieu JD, Milbled G, Pruvot P. Unusual EEG events during a case
encephalopathies. J Clin Neurophysiol. 1984;1(4):357–407. of puerperal eclampsia. Electroencephalogr Clin Neurophysiol.
104. Zysno E, Reichenmiller HE, Durr F. EEG studies in uraemic 1976;40:549(abst).
patients during extracorporeal haemodialysis or intermittent 127. Calloni MV, Porazzi D, Rovetta P. Cerebral dysfunction in
peritoneal dialysis. Electroencephalogr Clin Neurophysiol. eclampsia: a longitudinal EEG study. Electroencephalogr Clin
1967;23(4):384. Neurophysiol. 1981;51:79P(abst).
105. Bourne JR, Ward JW, Teschan PE, et al. Quantitative assessment 128. Grimes DA, Ekbladh LE, McCartney WH. Cortical blindness in
of the electroencephalogram in renal disease. Electroencephalogr preeclampsia. Int J Gynaecol Obstet. 1980;17(6):601–603.
Clin Neurophysiol. 1975;39(4):377–388. 129. Gurjinder PS, Dhand UK, Chopra JS. Balint’s syndrome follow-
106. Kiley JE, Woodruff MW, Pratt KI. Evaluation of encephalopathy ing eclampsia. Clin Neurol Neurosurg. 1989;91(2):161–165.
by EEG frequency analysis in chronic dialysis patients. Clin 130. Thomas SV, Somanathan N, Radhakumari R. Interictal EEG
Nephrol. 1976;5(6):245–250. changes in eclampsia. Electroencephalogr Clin Neurophysiol.
107. Balzar E, Saletu B, Khoss A, et al. Quantitative EEG: investigation 1995;94(4):271–275.
in children with end stage renal disease before and after 131. Osmanagaoglu MA, Dinc G, Osmanagaoglu S, et al. Comparison
haemodialysis. Clin Electroencephalogr. 1986;17(4):195–202. of cerebral magnetic resonance and electroencephalogram find-
108. Fusco L, Picca S, Rizzoni G, et al. Long-term EEG monitoring in ings in pre-eclamptic and eclamptic women. Aust N Z J Obstet
uremic children on chronic dialysis treatment. Eur Neurol. Gynaecol. 2005;45(5):384–390.
1991;31(4):193–198. 132. Sibai BM, Spinnato JA, Watson DL, et al. Effect of magnesium
109. Zysno EA. The EEG in uraemia. Electroencephalogr Clin sulfate on electroencephalographic findings in preeclampsia-
Neurophysiol. 1970;29(2):212–213. eclampsia. Obstet Gynecol. 1984;64(2):261–266.
110. Reichenmiller HE, Reinhard U, Durr F. Sleep EEG and uraemia. 133. Sibai BM. Diagnosis and management of gestational hyperten-
Electroencephalogr Clin Neurophysiol. 1971;30(3):263–264. sion and preeclampsia. Obstet Gynecol. 2003;102(1):181–192.
111. Jacob JC, Gloor P, Elwan OH, et al. Electroencephalographic 134. Brophy E, Brophy MH. Pyridoxal phosphate normalization of the
changes in chronic renal failure. Neurology. 1965;15:419–429. EEG in eclampsia. Electroencephalogr Clin Neurophysiol. 1991;
112. Kerr DN, Osselton JW. The EEG as a monitor of cerebral distur- 79:36P(abst).
bance during fast and slow haemodialysis of patients in chronic 135. Ledermair O, Niedermeyer E. Posteklamptische Epilepsie.
renal failure. Electroencephalogr Clin Neurophysiol. 1967; Geburtshilfe Frauenheilkd. 1956;16(8):679–685.
23(5):488. 136. Corriol J, Papy J, Rohner JJ, et al. Electroclinical correlations
113. Noriega-Sanchez A, Martinez-Maldonado M, Haiffe RM. Clinical established during tetanic manifestations induced by parathyroid
and electroencephalographic changes in progressive uremic removal in the dog. In: Gastaut H, Jasper HH, Bancaud J, et al.
encephalopathy. Neurology. 1978;28(7):667–669. eds. The Physiopathogenesis of the Epilepsies. Springfield, IL:
114. Chokroverty S, Gandhi V. Electroencephalograms in patients Charles C Thomas; 1969:3128–3140.
with progressive dialytic encephalopathy. Clin Electroencephalogr. 137. Glaser GH, Levy L. Seizures and idiopathic hypoparathyroidism.
1982;13(2):122–127. A clinical-electroencephalographic study. Epilepsia. 1960;1:
115. Yardimci N, Colak T, Sevmis S, et al. Neurologic complications 454–465.
after renal transplant. Exp Clin Transplant. 2008;6(3):224–228. 138. Goldberg HH. The electroencephalogram in hypocalcemic syn-
116. Hamiwka LD, Midgley JP, Hamiwka LA. Seizures in children after dromes. Electroencephalogr Clin Neurophysiol. 1959;11:398.
kidney transplantation: has the risk changed and can we predict 139. Lynch BJ, Rust RS. Natural history and outcome of neonatal
who is at greatest risk? Pediatr Transplant. 2008;12(5):527–530. hypocalcemic and hypomagnesemic seizures. Pediatr Neurol.
117. Sibai BM. Diagnosis, prevention, and management of eclampsia. 1994;11(1):23–27.
Obstet Gynecol. 2005;105(2):402–410. 140. Kossoff EH, Silvia MT, Maret A, et al. Neonatal hypocalcemic
118. Lain KY, Roberts JM. Contemporary concepts of the pathogene- seizures: case report and literature review. J Child Neurol.
sis and management of preeclampsia. JAMA. 2002;287(24): 2002;17(3):236–239.
3183–3186. 141. Kurtz D. The EEG in parathyroid dysfunction. In: Remond A, ed.
119. Roberts JM, Taylor RN, Goldfien A. Clinical and biochemical evi- Handbook of Electroencephalography and Clinical Neurophysiology.
dence of endothelial cell dysfunction in the pregnancy syndrome Amsterdam: Elsevier; 1976:77–87.
preeclampsia. Am J Hypertens. 1991;4(8):700–708. 142. Guberman A, Jaworski ZF. Pseudohypoparathyroidism and
120. Ferris TF. Pregnancy, preeclampsia, and the endothelial cell. epilepsy: diagnostic value of computerized cranial tomography.
N Engl J Med. 1991;325(20):1439–1440. Epilepsia. 1979;20(5):541–553.
121. Will AD, Lewis KL, Hinshaw DB Jr, et al. Cerebral vasoconstric- 143. Swash M, Rowan AJ. Electroencephalographic criteria of
tion in toxemia. Neurology. 1987;37(9):1555–1557. hypocalcemia and hypercalcemia. Arch Neurol. 1972;26(3):
122. Trommer BL, Homer D, Mikhael MA. Cerebral vasospasm and 218–228.
eclampsia. Stroke. 1988;19(3):326–329. 144. Devlesc Howard AB, Manyam BV. Electroencephalography in
123. Sheehan HL, Lynch JB. Pathology of Toxemia of Pregnancy. Fahr’s disease. Electroencephalogr Clin Neurophysiol. 1989;73:
Edinburgh: Churchill Livingstone; 1973. 68P(abst).
124. Dahmus MA, Barton JR, Sibai BM. Cerebral imaging in eclamp- 145. Spatz R, Kugler J, Angstwurm H. The EEG in hypercalcemia
sia: magnetic resonance imaging versus computed tomography. (author’s transl). EEG EMG Z Elektroenzephalogr Elektromyogr
Am J Obstet Gynecol. 1992;167(4 Pt 1):935–941. Verwandte Geb. 1977;8(2):70–76.
125. Schwartz RB, Feske SK, Polak JF, et al. Preeclampsia-eclampsia: 146. Allen EM, Singer FR, Melamed D. Electroencephalographic
clinical and neuroradiographic correlates and insights into the abnormalities in hypercalcemia. Neurology. 1970;20(1):15–22.
pathogenesis of hypertensive encephalopathy. Radiology. 147. Moure JM. The electroencephalogram in hypercalcemia. Arch
2000;217(2):371–376. Neurol. 1967;17(1):34–51.
Chapter 21 ■ Metabolic Disorders and EEG 409

148. Strickland NJ, Bold AM, Medd WE. Bronchial carcinoma with 171. Coursin DB. Convulsive seizures in infants with pyridoxine-defi-
hypercalcaemia simulating cerebral metastases. Br Med J. cient diet. J Am Med Assoc. 1954;154(5):406–408.
1967;3(5565):590–592. 172. Reilly RH, Killam KF, Jenney EH, et al. Convulsant effects of iso-
149. Huott AD, Madison DS, Niedermeyer E. Occipital lobe epilepsy. niazid. J Am Med Assoc. 1953;152(14):1317–1321.
A clinical and electroencephalographic study. Eur Neurol. 173. Bejsovec M, Kulenda Z, Ponca E. Familial intrauterine convul-
1974;11(6):325–339. sions in pyridoxine dependency. Arch Dis Child. 1967;
150. Kaplan PW. Reversible hypercalcemic cerebral vasoconstriction 42(222):201–207.
with seizures and blindness: a paradigm for eclampsia? Clin 174. Hunt AD Jr, Stokes J Jr, Mc CW, et al. Pyridoxine dependency:
Electroencephalogr. 1998;29(3):120–123. report of a case of intractable convulsions in an infant controlled
151. Zwang HJ, Cohn D. Electroencephalographic changes in acute by pyridoxine. Pediatrics. 1954;13(2):140–145.
water intoxication. Clin Electroencephalogr. 1981;12(1):35–40. 175. Waldinger C, Berg RB. Signs of pyridoxine dependency manifest
152. Okura M, Okada K, Nagamine I, et al. Electroencephalographic at birth in siblings. Pediatrics. 1963;32:161–168.
changes during and after water intoxication. Jpn J Psychiatry 176. Kretsch MJ, Sauberlich HE, Newbrun E. Electroencephalographic
Neurol. 1990;44(4):729–734. changes and periodontal status during short-term vitamin B-6
153. Crawford JD, Dodge P. Complications of fluid therapy in neuro- depletion of young, nonpregnant women. Am J Clin Nutr.
logic disease; water intoxication and hypertonic dehydration. 1991;53(5):1266–1274.
Pediatr Clin North Am. 1964;11:1029–1052. 177. Diefenbach WC, Beyers MR, Meyer LM. The encephalographic
154. Nakayama Y, Tanaka A, Naritomi K, et al. Hyponatremia-induced and psychologic changes in pernicious anemia. Acta Haematol.
metabolic encephalopathy caused by Rathke’s cleft cyst: a case 1953;9(4):201–208.
report. Clin Neurol Neurosurg. 1999;101(2):114–117. 178. Walton JN, Kiloh LG, Osselton JW, et al. The electroencephalo-
155. Itoh N, Matsui N, Matsui S. Periodic lateralized epileptiform dis- gram in pernicious anaemia and subacute combined degenera-
charges in EEG during recovery from hyponatremia: a case tion of the cord. Electroencephalogr Clin Neurophysiol. 1954
report. Clin Electroencephalogr. 1994;25(4):164–169. Feb;6(1):45-64.
156. Maruyama T, Tabata K, Nakagawa S, et al. A case of acute water 179. Andersen K, Stigsby B. Quantitative EEG analysis in patients with
intoxication showing triphasic waves on EEG. Rinsho pernicious anemia. Electroencephalogr Clin Neurophysiol.
Shinkeigaku. 1991;31(5):523–527. 1985;61:33P(abst).
157. Azuma H, Akechi T, Furukawa TA. Absence status associated 180. Biancheri R, Cerone R, Rossi A, et al. Early-onset cobalamin C/D
with focal activity and polydipsia-induced hyponatremia. deficiency: epilepsy and electroencephalographic features.
Neuropsychiatr Dis Treat. 2008;4(2):495–498. Epilepsia. 2002;43(6):616–622.
158. Castilla-Guerra L, del Carmen Fernandez-Moreno M, Lopez- 181. Engel GL, Margolin SG. Neuropsychiatric disturbance in internal
Chozas JM, et al. Electrolytes disturbances and seizures. Epilepsia. disease: metabolic factors and electroencephalographic correla-
2006;47(12):1990–1998. tionss. Arch Intern Med. 1942;70:236–259.
159. Van der Helm-van Mil AH, Van Vugt JP, Lammers GJ, et al. 182. Hoffman WC, Lewis R, Thorn GW. The electroencephalogram in
Hypernatremia from a hunger strike as a cause of osmotic myeli- Addison’s disease. Bull Johns Hopkins Hosp. 1942;70:335–361.
nolysis. Neurology. 2005;64(3):574–575. 183. Vas GA, Cracco JB. Diffuse encephalopathies. In: Daly DD, Pedley
160. Shils ME. Magnesium, calcium, and parathyroid hormone inter- TA, eds. Current Practice of Clinical Electroencephalography. 2nd
actions. Ann N Y Acad Sci. 1980;355:165–180. ed. New York: Raven Press; 1990:371–399.
161. Riggs JE. Neurologic manifestations of electrolyte disturbances. 184. Garcia-Austt E, Torrents E, Mussio-Fournie F. The electroen-
Neurol Clin. 2002;20(1):227–239, vii. cephalogram in Cushings disease. Electroencephalogr Clin
162. Leicher CR, Mezoff AG, Hyams JS. Focal cerebral deficits in Neurophysiol. 1950;2:103.
severe hypomagnesemia. Pediatr Neurol. 1991;7(5):380–381. 185. Pine I, Engel FL, Schwartz TB. The electroencephalogram in
163. Victor M, Adams RD, Collins GH. The Wernicke-Korsakoff ACTH and cortisone treated patients. Electroencephalogr Clin
Syndrome. Philadelphia, PA: F.A. Davis; 1971. Neurophysiol. 1951;3(3):301–310.
164. Dreyfus PM, Victor M. Effects of thiamine deficiency on the cere- 186. Glaser GH, Komfeld DS, Knight RP. Intravenous hydrocortisone,
bral nervous system. Am J Clin Nutr. 1961;9:414–425. corticotropin and the electroencephalograms. Arch Neurol
165. Frantzen E. Wernicke’s encephalopathy. 3 Cases occurring in con- Psychiatry. 1955;73:338–344.
nection with severe malnutrition. Acta Neurol Scand. 1966;42(4): 187. Raab W, Smithwick RH. Pheochromocytoma with hypothalamic
426–441. manifestations and excessive hypermetabolism; a case report. J
166. Fournet A, Lanternier J. Constatations electroencephalo- Clin Endocrinol Metab. 1949;9(8):782–790.
graphiques dans 17 cas d’encephalopathie de Gayet-Wernicke. 188. Kennedy JM, Thomson AP, Whitfield IC. Coma and electro-
Rev Neurol (Paris). 1956;94(5):644–645. encephalographic changes in hypopituitarism. Lancet. 1955;
167. Keyser A, De Bruijn SF. Epileptic manifestations and vitamin B1 269(6896):907–908.
deficiency. Eur Neurol. 1991;31(3):121–125. 189. Lindsley DB, Rubenstein BB. Relationships between brain poten-
168. Fattal-Valevski A, Kesler A, Sela BA, et al. Outbreak of life-threat- tials and some other physiological variables. Proc Soc Exp Biol
ening thiamine deficiency in infants in Israel caused by a defec- (New York). 1937;35:558–563.
tive soy-based formula. Pediatrics. 2005;115(2):e233–e238. 190. Rubin MA, Cohen HC, Hoagland H. The effect of artificially
169. Fattal-Valevski A, Bloch-Mimouni A, Kivity S, et al. Epilepsy in raised metabolic rate and the electroencephalogram of schizo-
children with infantile thiamine deficiency. Neurology. 2009; phrenic patients. Endocrinology. 1937;25:536–540.
73(11):828–833. 191. Ross DA, Schwab RJ. The cortical alpha rhythm in thyroid disor-
170. Srikantia SG, Reddy MV, Krishnaswamy K. Electro - ders. Endocrinology. 1939;25:75–79.
encephalographic patterns in pellagra. Electroencephalogr Clin 192. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. 1st ed.
Neurophysiol. 1968;25(4):386–388. Cambridge, MA: Addison-Wesley; 1941.
410 Part III ■ Clinical EEG: General

193. Vague J, Gastaut H, Favier G, Rey J. Electroencephalography in 210. Forester CF. Coma in myxedema. Report of a case and review of
endocrine pathology. Mars Med. 1952;89(1):1–17. the world literature. Arch Intern Med. 1963;111:734–743.
194. Vague J, Gastaut H, Codaccioni JL, et al. EEG in thyroid diseases. 211. River Y, Zelig O. Triphasic waves in myxedema coma. Clin
Ann Endocrinol (Paris). 1957;18(6):996–1008. Electroencephalogr. 1993;24(3):146–150.
195. Wilson WP, Johnson JE, Feist FW. Thyroid hormone and brain 212. Wynn D, Lagerlund T, Mokri B, et al. Periodic complexes in
function. II. Changes in photically elicited EEG responses follow- hypothyroidism masquerading as Jakob-Creutzfeldt disease: a
ing the administration of triiodothyronine to normal subjects. case report. Electroencephalogr Clin Neurophysiol. 1989;72:
Electroencephalogr Clin Neurophysiol. 1964;16:329–331. 31P(abst).
196. Skanse B, Nyman GE. Thyrotoxicosis as a cause of cerebral dys- 213. Gibbs FA, Reid DE. The EEG in pregnancy. Am J Obstet Gynecol.
rhythmia and convulsive seizures. Acta Endocrinol (Copenh). 1942;44:672–675.
1956;22(3):246–263. 214. Keunen RW, Vliegen JH, van der Pol DA, et al. The electroen-
197. Jabbari B, Huott AD. Seizures in thyrotoxicosis. Epilepsia. cephalogram during normal third trimester pregnancy and six
1980;21(1):91–96. months postpartum. Br J Obstet Gynaecol. 1997;104(2):
198. Miyazaki Y, Fukuoka K, Murase T, et al. Abnormal MRI and EEG 256–258.
findings in thyroid storm resulting from Graves’disease. Thyroid. 215. Dusser de Barenne D, Gibbs FA. Variations in the electroen-
2008;18(10):1131–1132. cephalogram during the menstrual cycle. Am J Obstet Gynecol.
199. Cadilhac J. The EEG in thyroid dysfunction. In: Remond A, ed. 1942;44:687–690.
Handbook of Electroencephalography and Clinical Neurophysiology, 216. Holmes GL, Donaldson JO. Effect of sexual hormones on the
Vol 15C: Metabolic, Endocrine and Toxic Diseases. Amsterdam: electroencephalogram and seizures. J Clin Neurophysiol. 1987;
Elsevier; 1976:70–76. 4(1):1–22.
200. Berlit P, Rakicky J. The EEG in metabolic and endocrine 217. Goldberg A. Acute intermittent porphyria: a study of 50 cases. Q
encephalopathies. Clin Neurophysiol. 1991;78:36P(abst). J Med. 1959;28(110):183–209.
201. Chong JY, Rowland LP, Utiger RD. Hashimoto encephalopathy: 218. Dow RS. The electroencephalographic changes in acute intermit-
syndrome or myth? Arch Neurol. 2003;60(2):164–171. tent porphyria. Electroencephalogr Clin Neurophysiol. 1961;13:
202. Kothbauer-Margreiter I, Sturzenegger M, Komor J, et al. 425–437.
Encephalopathy associated with Hashimoto thyroiditis: diagnosis 219. Bylesjo I, Forsgren L, Lithner F, Boman K. Epidemiology and
and treatment. J Neurol. 1996;243(8):585–593. clinical characteristics of seizures in patients with acute intermit-
203. Tsai MH, Lee LH, Chen SD, et al. Complex partial status epilepti- tent porphyria. Epilepsia. 1996;37(3):230–235.
cus as a manifestation of Hashimoto’s encephalopathy. Seizure. 220. Birchfield RI, Cowger ML. Acute intermittent porphyria with
2007;16(8):713–716. seizures. Anticonvulsant medication-induced metabolic changes.
204. Aydin-Ozemir Z, Tuzun E, Baykan B, et al. Autoimmune thyroid Am J Dis Child. 1966;112(6):561–565.
encephalopathy presenting with epilepsia partialis continua. Clin 221. Krauss GL, Simmons-O’Brien E, Campbell M. Successful treat-
EEG Neurosci. 2006;37(3):204–209. ment of seizures and porphyria with gabapentin. Neurology.
205. Shaw PJ, Walls TJ, Newman PK, et al. Hashimoto’s encephalopa- 1995;45(3 Pt 1):594–595.
thy: a steroid-responsive disorder associated with high anti-thy- 222. Niedermeyer E, Ribeiro M, Hertz S. Mixed-type encephalopathies:
roid antibody titers—report of 5 cases. Neurology. 1991;41(2 Pt 1): preliminary considerations. Clin Electroencephalogr. 1999;30(1):
228–233. 12–15.
206. Henchey R, Cibula J, Helveston W, et al. Electroencephalographic 223. Austen FK, Carmichael MW, Adams RD. Neurologic manifesta-
findings in Hashimoto’s encephalopathy. Neurology. 1995;45(5): tions of chronic pulmonary insufficiency. N Engl J Med.
977–981. 1957;257(13):579–590.
207. Schultz MA Schulte FJ, Akiyama Y, et al. Development of EEG 224. Gastaut H, Tassinari CA, Duron B. Polygraphic study of diurnal
sleep phenomena in hypothyroid infants. Electroencephalogr Clin and nocturnal (hypnic and respiratory) episodal manifestations
Neurophysiol. 1968;25:351–358. of Pickwick syndrome. Rev Neurol (Paris). 1965;112(6):
208. Nieman EA. The electroencephalogram in congenital hypothy- 568–579.
roidism: a study of 10 cases. J Neurol Neurosurg Psychiatry. 225. Jung R, Kuhlo W. Neurophysiological studies of abnormal
1961;24:50–57. night sleep in the Pickwickian syndrome. In: Akert K, Bally C,
209. Scarpalezos S, Lygidakis C, Papageorgiou C, et al. Neural and Schade JP, eds. Sleep Mechanisms. Amsterdam: Elsevier; 1965:
muscular manifestations of hypothyroidism. Arch Neurol. 1973; 140–159.
29(3):140–144.
CHAPTER

EEG and Craniocerebral Trauma


ERIK RUMPL 22
A
craniocerebral trauma raises the important question of studies in brain injuries were done by Dow et al. (28), who
the degree of cerebral disturbance. This question may included patients with more severe injuries and repeated studies
be answered easily in cases of commotio cerebri or during the recovery of the patients. Dawson et al. (29) continued
slight concussion of the brain; this is a reversible impairment of to observe patients and studied the evolution of abnormalities
consciousness of brief duration with no clinical evidence of any with repeated records. Extensive work in this field was done by
gross structural change in the brain substance. The question of Meyer-Mickeleit (19), Schneider and Hubach (30), Courjon and
brain function becomes more important in cases of cerebral Scherzer (21), and Koufen and Dichgans (23).
contusion after more severe injury with prolonged unconscious- The impairment of consciousness to any degree was usually
ness and signs of brainstem dysfunction. Brainstem damage may found to be accompanied by an abnormal EEG (19,28–32).
result from primary brainstem injury (1,2) or from secondary Most of these authors directed their attention to the frequency
brainstem lesion due to downward transtentorial herniation (3). of the basic activity and to focal, general, or epileptic abnormal-
It has been shown that brainstem injury does not exist in isola- ities, but no further distinction of the EEG pattern was made.
tion; it is merely one aspect of diffuse brain damage (4). High-voltage delta activity was regarded as the electroen-
The clinical parameter of the grade of disintegration of brain cephalographic correlate of coma, which in turn was considered
function and impairment of the brainstem is demonstrated by the an equivalent of deep sleep (33–35). Further studies in coma
development of an acute secondary midbrain and bulbar brain (36–38) made it clear that states of coma were associated with a
syndrome (BBS). The symptoms of the well-known rostrocaudal variety of EEG patterns, often with the criteria of sleep
deterioration, first described by Mc-Nealy and Plum (5) and Plum (12,14,39). The manifestation of sleep patterns appeared to be
and Posner (6) and modified by Gerstenbrand and Lücking (7), of prognostic significance, supported by the results of overnight
allow a clear clinical statement about the depth of posttraumatic or prolonged EEG recordings (40–42). The role of reactivity in
coma. In addition to the neurologic examination, cranial com- the assessment of the depth of coma was first pointed out by
puted tomography (CT) and magnetic resonance imaging (MRI) Fischgold et al. (43) and is an important prognostic indicator
easily identify a space-occupying lesion and sometimes demon- for good outcome (44). In addition to these findings, asymme-
strates the displacement of the brainstem (8,9). These methods, try in reactivity was considered a reliable lateralizing sign (45).
however, fail to give any information about cerebral activity. Important reviews concerning posttraumatic comatose
Therefore, the electroencephalogram (EEG) is still important in states have given detailed insight into the EEG in altered states
the diagnosis of traumatic cerebral lesions, especially in the assess- of consciousness (10,46,47). Early and late changes of the EEG
ment of the degree of cortical activity, which shows reasonably after craniocerebral trauma were discussed extensively by
good correlation with the depth of posttraumatic coma (10–16). Courjon and Scherzer (21) and Stockard et al. (48).
Chronic stages of craniocerebral trauma demonstrate a
diminished electroencephalographic–neurologic correlation EXPERIMENTAL WORK
(17–20). However, there is an approximate correlation between
the EEG and clinical improvement, especially in patients who Williams and Denny-Brown (49) found an immediate reduc-
have had systematic follow-up studies of EEG and clinical tion in amplitude (sometimes approaching isoelectricity) in
examination (21–24). Occasionally, the EEG may return to nor- the EEG in cats subjected to experimental head concussion.
mal when neurologic or psychiatric abnormalities persist, a dis- After a short period of delta activity, the EEG returned to
parity that indicates a bad prognosis (18,25,26). By contrast, in normal within a period of seconds to a few minutes. Dow et al.
some patients with normal clinical findings, an abnormal EEG (28) studied the physical factors responsible for the injury in
may be the forerunner of an intracranial complication such as humans. These factors appeared to be of the same order as
posttraumatic epilepsy (21). those necessary to produce experimental concussion in ani-
mals (50). Interestingly, the threshold velocity that provoked
EARLIER EEG WORK IN concussion at the moment of impact in animals also marked
CRANIOCEREBRAL TRAUMA the limit between the average velocity in patients with border-
line and abnormal EEG (28). Other than this finding, patho-
Williams (27) studied a few patients after mild craniocerebral logic observations made in humans could not be convincingly
trauma and observed that normal records can be obtained duplicated in experimental studies. This might be due to the
within a few hours of injury. Further electroencephalographic smaller mass of brain in animals and the restriction in impact
411
412 Part III ■ Clinical EEG: General

techniques (51,52). Even in standardized animal experiments, Other signs apart from these grades can also be noted in the
the analysis of the EEG did not provide adequate indices of EEG: (i) superimposed fast activity (6 to 18 c/sec localized over
injury severity, and there was no correlation found with the the frontal region or diffusely spread); (ii) normal-looking
duration of coma (53). sleep records; (iii) diffuse slowing accompanied by “typical
Following the convulsive theory of slight concussion (54), sleep potentials” (spindles, vertex sharp waves, K complexes);
the energy imported to the brain may generate turbulent rota- (iv) diffuse slowing accompanied by altered “sleep potentials,”
tory and other movements of the cerebral hemispheres provok- listed under “atypical sleep potentials”; (v) spontaneously
ing a convulsive disorder, which might present a mild form of alternating EEG patterns with a rapid succession of low-
cerebral concussion with a sudden short loss of consciousness, voltage delta activity and high-voltage slow waves (this activity
paralysis of reflex activity, and loss of memory (commotio cere- can be classified as delta bursts [0.5 to 2 seconds] and short [2
bri). This theory may explain the observation that the EEG and 5 seconds] and long [more than 5 seconds in duration]
returns to normal within 24 hours in these cases. runs of delta with frequencies of 0.5 to 4 c/sec); (vi) “lateral-
Some closer correlations to EEG changes in comatose states ized signs” such as local slow activity (“local slowing”), unilat-
in humans were found in other animal experiments producing eral predominant slow activity, unilateral low-voltage output,
localized destructive brain lesions or using local electrical stim- and unilateral depression of superimposed fast activity listed
ulation. Local lesions in the white matter and in thalamic struc- under the heading of “unilateral predominant slowing,” asym-
tures were immediately followed by the appearance of delta metries of “typical or atypical sleep potentials,” and asymme-
waves, which were accompanied by spindles in the case of thal- tries of response to external stimuli; (vii) four types of reaction
amic lesions. Slow activity developed gradually in mesen- to external stimuli: (1) appearance of widespread 1 to 7 c/sec
cephalic reticular formation lesions; it was absent in cortical activity, (2) blocking of slow activity, (3) appearance of alter-
lesions (55). EEG changes similar to those seen in thalamic nating EEG patterns, and (4) appearance or disappearance of
destructive lesions were found after stimulation of thalamic “typical or atypical sleep potentials”; and (viii) epileptiform
nuclei (56,57). Stimulation at the level of the mesencephalic patterns: (1) focal or generalized runs of spikes and sharp spike
reticulum induced behavior and EEG arousal in sleeping in oth- waves, (2) periodic lateralized epileptiform discharges
erwise intact animals, whereas destructive lesions at mesen- (PLEDs), and (3) triphasic waves.
cephalic levels caused coma and EEG activity reminiscent of Reactivity may be immediate or delayed. Delayed reactiv-
different types of sleep (58). All these mechanisms may be ity is a slow EEG response inducing a new pattern under the
involved in posttraumatic comatose states as long as delta waves influence of repetitive stimulation (10). Fischgold and Mathis
and spindles are generated by otherwise relatively normal corti- (36) emphasize the usefulness of auditory and painful stimuli
cal neurons. in contrast to the insignificant effect of visual stimulation.
In lighter stages of coma, stimulation may also provoke
EEG CHANGES IN THE ACUTE STAGES extracerebral changes such as muscle activity and respiratory
OF POSTTRAUMATIC COMA artifacts.

Comatose posttraumatic states may be associated with a multi- CLASSIFICATION OF COMA


tude of abnormal waveforms other than delta (12,14,36,38).
These abnormal waveforms, especially the different patterns of Physiologically, the loss of consciousness implies that the
sleep, depend above all on the patient’s level of consciousness patient has suffered widespread dysfunction of cerebral hemi-
(46). Therefore, a detailed neurologic description of comatose spheres or the brainstem or both. When there is a diffuse
patients is necessary to define coma (59) in order to establish a swelling of the brain or bilateral hemispheric lesions, the cere-
sufficient correlation between different EEG changes and differ- bral hemispheres, the diencephalon, and the adjoining mid-
ent stages of coma. brain are displaced downward through the tentorial notch. In
this case, the herniation through the tentorium is symmetrical
CLASSIFICATION OF EEG (central tentorial herniation). If downward displacement con-
tinues, the contents of the posterior fossa will finally be pressed
Different parameters have been used in the breakdown of EEG through the foramen magnum. This brain shift evokes a num-
signs in comatose states (10–12,14,21,36,60–62). To increase the ber of characteristic clinical signs that have been described by
value of the EEG in posttraumatic coma, all these parameters various authors (5–7). The most prominent clinical signs of
were integrated and correlated with the stages of rostrocaudal central herniation are listed in Table 22.1. According to
deterioration (13). Gerstenbrand and Lücking (7) posttraumatic comatose states
The EEG abnormalities may be classified into five grades (13): can be broken down into six stages, including four stages of
grade 1, predominant alpha and little theta activity; grade 2, midbrain syndrome (MBS) and two stages of BBS. With regard
predominant theta and little delta activity; grade 3, predominant to the terminology of Plum and Posner (6), MBS 1 and MBS 2
high-voltage rhythmic and arrhythmic delta and subdelta acti- correspond to the early diencephalic stage, MBS 3 to the late
vity; grade 4, diffuse, mostly low-voltage delta and subdelta diencephalic stage, MBS 4 to the midbrain-upper pons stage,
activity and low-voltage activity only recognizable with increased BBS 1 to the lower pontine-upper medullary stage, and BBS 2
amplification (3.5 V/mm); and grade 5, isoelectric record. to the medullary stage.
Chapter 22 ■ EEG and Craniocerebral Trauma 413

Tabl e 2 2 . 1

Clinical Signs in Posttraumatic Patients at the Different Stages of the Midbrain Syndrome (MBS)
and Bulbar Brain Syndrome (BBS)

MBS 1 MBS 2 MBS 3 MBS 4 BBS 1 BBS2


Spontaneous Nonstereotyped Nonstereotyped Decorticate Decerebrate Flaccidity Flaccidity
limb postures movements movements posturing posturing
in the arms in the arms
and legs
Extension of
the legs
Motor response Nonstereotyped Nonstereotyped Decorticate Decerebrate Decerebrate
to pain withdrawal of withdrawal of response response response
the limbs the arms No response
Extensor
response of
the legs
Eye position Roving Roving, more Immobile, Immobile, Immobile, Immobile,
movements irregular straight divergent divergent divergent
ahead
Pupil size and Normal Normal small Small Enlarged Large Large
reaction
Reacting Reacting Small range Small range Unreacting Unreacting
of contraction of contraction
Respiration Normal Cheyne-Stoke Cheyne-Stokes Regular Ataxic No
pattern hyperven- respiration
tilation
Rapid regular
hyperven-
tilation

From Rumpl E, Prugger M, Bauer G, et al. Incidence and prognostic value of spindles in post-traumatic coma. Electroencephalogr Clin Neurophysiol. 1983;56:420–429.

In cases with expanding lesions in the lateral middle fossa or and respiratory abnormalities). The CT scan is diagnostic and
temporal lobe, the medial edges of the uncus and hippocampal eliminates a supratentorial lesion causing secondary brainstem
gyrus are commonly pushed toward the midline and over the free dysfunction. The MRI scan shows the extent of brainstem dam-
lateral edge of the tentorium, compressing the third cranial nerve age in the further course.
(early uncal herniation). During uncal herniation, a unilateral
dilated pupil is the most consistent finding (6). Other neurologic RELATION OF EEG ABNORMALITIES
signs may be present at this early stage and may provide hints TO THE STAGE OF COMA
concerning the original hemispheric lesion. Once the pupil
dilates fully (late uncal herniation), the patients become coma- The EEG patterns in the different stages of MBS and BBS are
tose, and abnormal postures and motor responses evolve (6,63). presented in Figure 22.1. The significance of this observation is
Eventually, with increasing intracranial pressure, lateralizing somewhat hampered by the fact that the figure presents EEG
signs disappear and the patients show the clinical signs of MBS 4 data obtained within 1 week after brain injury, thereby neglect-
or BBS 1. ing the special behavior of spindle activity (see the section
Neurologic signs different from this expected rostrocaudal “Relation of EEG Abnormalities to Outcome”). Nevertheless,
deterioration may be found in cases of primary brainstem the figure demonstrates well the principal relation of EEG to
injury. Maciver et al. (2) state that a primary brainstem lesion the stages of coma. There is a decrease in the number of differ-
can be suspected if the neurologic signs do not fit the classical ent EEG patterns related to the stages of MBS and BBS, indicat-
stages of rostrocaudal deterioration (i.e., the relatively intact ing the increase of intracranial pressure. Unfavorable prognosis
oculomotor functions are in contrast to decerebrate posturing is shown by the disappearance of “typical or atypical sleep
414 Part III ■ Clinical EEG: General

Figure 22.1. The EEG patterns at the different stages of acute traumatic secondary midbrain and bulbar brain syndrome.
see A–E above. Note the reduction in the variety of EEG patterns in the course of increasing rostrocaudal deterioration. At
bulbar brain syndrome stage 2, the EEG is isoelectric. The records were listed under one or more of the following cate-
gories: A/ T, predominant alpha and little theta activity; T/ D, predominant theta and little delta activity; D/ SD, predominant
diffuse rhythmic and arrhythmic delta and subdelta activity; DB, delta bursts; DS, short runs of delta; DL, long runs of
delta; TSP, typical sleep potentials; ASP, atypical sleep potentials; S, spindles; LS, local slowing; US, unilateral predominant
slowing; SFA, superimposed fast activity; MBS, midbrain syndrome; BBS, bulbar brain syndrome; N, number of records.
(From Rumpl, E. Elektro-neurologische Korrelationen in den frühen Phasen des posttraumatischen Komas. I. Das EEG in
den verschiedenen Phasen des akuten traumatischen sekundären. Mittelhirn und Bulbärhirnsyndroms. Z. EEG-EMG.
1979;10:148–157; and Rumpl E, Lorenzi E, Hackl JM, et al. The EEG at different stages of acute secondary traumatic mid-
brain and bulbar brain syndromes. Electroencephalogr Clin Neurophysiol. 1979;46:487–497, somewhat modified.)

potentials,” alternating patterns, and loss of reactivity (10,12– tials (14) and the shift of superimposed fast activity, diffusely
14,21,41). In accordance with the mildly diffuse cortical distur- spread in lighter stages of MBS, to the frontal regions (67). The
bance, the EEG is slightly or moderately abnormal in MBS 1. EEG patterns are still more simplified in MBS 4 and BBS 1. The
Stimuli to diencephalic or rostromesencephalic structures absence, reduction, and deterioration of sleep potentials or
(64) may induce delta bursts or short runs of delta waves. Local alternating patterns are suggestive of marked damage at the
slow activity can be seen only in an otherwise mildly abnormal diencephalic level (39,41). The telencephalon (10) must also be
EEG, while increasing diffuse slowing may overwhelm local involved, considering the diffuse brain edema in advanced
abnormalities (65). Borderline EEG patterns occur at MBS 1 but stages of MBS. In BBS 1, low-voltage delta and subdelta activity
not in deeper stages of MBS. In MBS 2, the amount of slow activ- appears, followed by low-voltage cerebral activity, often recog-
ity increases. This increasing abnormality may be due to a direct nizable only with increased amplification. No electrical cerebral
cortical disturbance (64) or due to a more remote effect from activity can be observed in patients with BBS 2 (13). Patients
deeper structures (64,66). However, “sleep or sleep-like poten- with an isoelectric EEG or an EEG with repeated isoelectric
tials” (see Figs. 22.6A and 22.7A) indicate a relatively intact cortex periods will die (68). The high prognostic value of an EEG clas-
(14). In MBS 2, the EEG shows the widest range of different EEG sification with a great number of different EEG patterns was
patterns. In MBS 3, a clear reduction in the number of EEG pat- supported by the development of an EEG score, which included
terns is noted. A decrease of spontaneous alternating patterns many of these phenomena weighted according to their per-
and “sleep potentials” has been thought to indicate an increasing ceived prognostic value and scored as dichotomous variables—
disturbance of the diencephalic and mesencephalic systems (12). present or absent (69).
Increasing rostrocaudal deterioration is further character- Unreactive alpha activity or “alpha-like” rhythms (70) are
ized by the increased change of typical or atypical sleep poten- not seen in patients in BBS 1 after supratentorial lesions causing
Chapter 22 ■ EEG and Craniocerebral Trauma 415

secondary brainstem involvement. This pattern usually appears


in polytraumatic patients at this stage and thereby strongly sug-
gests an additional hypoxic or anoxic cerebral lesion. The same
suggestion can be made with theta-pattern coma, which was
found to be a variant of alpha coma (71). Usually, both patterns
are predictors of fatal outcome. Rhythmic coma in children
especially in the alpha frequency range has generally a better
prognosis than in adults (72).
Primary brainstem injuries may produce clinical symp-
toms that are hardly separable from symptoms of secondary
brainstem injury caused by downward herniation. Although
primary brainstem damage rarely exists in pure form (4), a
primary brainstem lesion may be the principal cause of
coma. In these cases, the EEG can be a helpful diagnostic tool.
Patients in coma from caudally placed brainstem lesions may
have nonreactive alpha activity even in an unresponsive
decerebrate state (73). Sleep spindles, vertex sharp waves, and
K complexes suggest that the lesion is below the still intact
thalamocortical system (74). This suggestion has been con-
firmed by Steudel et al. (75), who described brainstem lesions
with evidence based on cranial CT and autopsy findings in all
their decerebrate patients with spindles and/or alpha activity
in the EEG. During the first week, these activities were
replaced by delta waves, indicating that EEG patterns similar
to the one generally seen in prolonged comatose states may
appear (46,76).
The reaction to external stimuli further characterizes the
depth of coma (10,21). In MBS 1, sensory stimulation may
briefly block the slow activity; sleep, easily recognizable at this
stage, may have a similar effect (48). In further stages of MBS,
reactivity consists of widespread delta activity, alternating pat-
terns, and typical or atypical sleep potentials (see Fig. 22.13A).
In BBS 1 and BBS 2, no reactivity can be observed.
Generalized burst suppression occurs in posttraumatic coma
only when cerebral anoxia was sustained with the trauma
(48,67). Burst suppression activity due to an overdose of sopo-
rific drugs tends to produce uniform intersuppression activity,
in contrast to the intersuppression activity in anoxic coma, in Figure 22.2. A: EEG from a 32-year-old woman in midbrain syndrome
which delta waves and intermittent spikes are observed fre- stage 2 and with clinical signs of a left hemisphere lesion. No clinical
quently (48). This type of activity carries a poor prognosis evidence of epileptic fits. Unilateral predominant slowing in the left
because it indicates a diffuse anoxic encephalopathy that inter- hemisphere. Sharp waves in left upper frontal region. B: EEG from the
feres with the original traumatic brain damage. same patient in midbrain syndrome stage 1, 1 day after first recording.
Metabolic derangements certainly have an important impact Right-sided clinical focal motor attacks. Continuous focal spikes in left
on coma and accompanying EEG abnormalities upper frontal-central region. (From Rumpl E. Elektro-neurologische
(10,13,14,39,77–79). The number of metabolic disturbances is Korrelationen in den frühen Phasen des posttraumatischen Komas. I.
obviously higher in patients in deep stages of posttraumatic Das EEG in den verschiedenen Phasen des akuten traumatischen
coma (13). Most of these patients suffer from multiple trauma sekundären. Mittelhirn und Bulbärhirnsyndroms. Z. EEG-EMG. 1979;
including the liver, kidneys, and lungs. The combination of 10:148–157.)
metabolic and traumatic encephalopathy carries the worst
prognosis. The EEG changes due to metabolic disturbance
interfere with the EEG changes because of the herniation itself, may have clinical seizures shortly before or shortly after the
thus blurring the rules concerning the decrease of different EEG discharges are found in the EEG (Fig. 22.2). Acute epileptic
patterns with advancing deterioration. Further influences on and paroxysmal EEG discharges may also accompany
the EEG are caused by the effects of sedative or anesthetic drugs nonconvulsive (subclinical) seizures or status epilepticus
frequently used in intensive care units. in severe brain injuries. In cases of nonconvulsive seizures
Early electroencephalographic seizure activity is only seen or status, a more severe clinical picture is simulated.
in cases of severe craniocerebral trauma, in which patients Seizures either are generalized from the start or more
416 Part III ■ Clinical EEG: General

often are focal with subsequent generalization (21). Focal


seizure activity consists of runs of spikes and sharp waves
and is more likely to occur in patients with underlying
intracranial hematoma. The EEG is of considerable localizing
value in these cases, and there has been only one reported
case of focal seizure activity occurring contralateral to the
posttraumatic hematoma (21).
Usually, the appearance of the electroencephalographic
seizure activity at these stages of coma carries a worse progno-
sis (21) and is associated with a mortality that is about three
times higher than in the absence of seizure activity (48).
Intracranial pressure may increase even in the presence of
nonconvulsive subclinical status epilepticus or repetitive elec-
troencephalographic seizures (80). Status epilepticus associ-
ated with generalized motor seizures may be the result of
severe cerebral trauma. Death occurs in more than 50% of Figure 22.3. Correlation of lateralizing signs in the EEG with the neuro-
these cases within the first few days (21). In children, post- logic or postmortem examinations. Dark columns demonstrate corre-
traumatic status epilepticus is associated with a far less omi- sponding neurologic and EEG signs of lateralization in midbrain
nous prognosis than in adults (81). Isolated spikes and slow syndrome (MBS) stages 1, 2, and 3. In midbrain syndrome stage 4 and
waves are rare after a recent injury (82), but they may occur in bulbar brain syndrome (BBS) stage 1 (listed in one column) results of
severe cases. Although prognosis in cases with early seizures is the autopsy have to be used in some cases to determine lateralization.
generally bad, acute convulsions are a more serious complica- (From Rumpl E, Lorenzi E, Hackl JM, et al. The EEG at different stages
tion in patients with brain tumor and metabolic of acute secondary traumatic midbrain and bulbar brain syndromes.
encephalopathies (83). Unfortunately, quantitative EEG has Electroencephalogr Clin Neurophysiol. 1979;46:487–497.)
difficulties detecting spikes, brief seizures, burst suppression,
PLEDs, triphasic waves, and intermittent theta and delta
activity. Therefore, the use of serial standard EEG recordings etative) state. Because there has been much doubt concerning
at least three times a week is still required in intensive care the prognostic significance of spindle activity in posttraumatic
patients (84). coma, special attention was paid to spindle activity in a study
PLEDs are rare events in posttraumatic coma; they are usu- based on 70 EEGs taken in the acute stage of coma (within the
ally seen in cases of subdural hematoma (85). More frequent first 2 days after brain injury) and 63 EEGs recorded in a pro-
PLEDs are seen in patients with acute unilateral lesions of vas- longed comatose state (days 3 through 12 after brain injury).
cular origin (86,87). Triphasic waves may also be produced by The incidence of spindles, form of spindles, and asymmetries of
subdural hematoma, but they are more characteristic of meta- spindle activity in the different outcome categories are listed in
bolic disturbances (88,89), especially when no asymmetry can Table 22.2 (91).
be detected.
The lateralizing signs in the EEG (other than epileptic dis-
charges) demonstrate a close correlation to the lateralizing
signs of the neurologic examination (Fig. 22.3). In MBS 4 and
BBS 1, the neurologic examination reveals no lateralizing
signs at all. At these stages, the EEG gives a hint of local cere-
bral lesion. The localized signs in the EEG may accompany all
local cerebral lesions including the three major types of
hematoma—subdural, epidural, and intracerebral (Fig. 22.4).
Asymmetry in reactivity (45) is helpful to ascertain uncertain
lateralizing signs in the acute stages of coma (see Chapter 23;
Fig. 23.9); in prolonged comatose states, asymmetry often
does not manifest itself unless the patient is stimulated (48).

RELATION OF EEG ABNORMALITIES


TO OUTCOME
According to Jennet and Bond (90), patient outcome may be Figure 22.4. EEG from a 42-year-old man in midbrain syndrome stage
classified into four categories: death, severe disability, moderate 3, 1 day after surgical evacuation of a right epidural hematoma.
disability, and good recovery. In the results presented here, the Unilateral slowing in right hemisphere and asymmetrical (and mildly
category of severe disability includes patients in an apallic (veg- atypical) spindles point to the original lesion.
Chapter 22 ■ EEG and Craniocerebral Trauma 417

Tabl e 2 2 . 2

Incidence of Spindles, Form of Spindles, and Asymmetries of Spindle Activity


in Different Outcome Categories a

Acute Coma (Total %) Prolonged Coma (Total %)


Outcome Asymmetry Asymmetry
Categories Nb Spindles Atypical Spindles N Spindles Atypical Spindles
Good 26 26 100 2 8 3 12 18 8 44 2 11 2 25
recovery
Moderate 10 9 90 3 33 3 33 7 4 57 2 50 1 25
disability
Severe 18 11 61 5 45 7 63 21 5 24 5 100 2 40
disability
Brain 16 9 56 4 44 8 88 17 2 12 1 50 2 100
death
Total 70 64 91 14 22 24 33 63 19 30 10 53 7 37
coma

a
The percentage of atypical spindles and symmetry of spindles remarkably increase with worsening of outcome. Spindles are seen in most cases in acute coma within
the first 2 days after injury but show clear reduction in prolonged coma (days 3 to 12 after injury). (From Rumpl E, Prugger M, Bauer G, et al. Incidence and prog-
nostic value of spindles in post-traumatic coma. Electroencephalogr Clin Neurophysiol. 1983;56:420–429.)
b
N, number of patients and EEGs.

In patients with good recovery, spindles are seen in all A spontaneous decrease of spindles in prolonged coma was
EEGs recorded during the acute stage of coma (100%). observed independent of the outcome (45,96). This observation
Atypical (8%) and asymmetrical spindles (12%) were rarely was further corroborated by Bricolo et al. (97), using com-
observed in these cases. In prolonged coma, spindles occurred pressed spectral array in long-term EEG monitoring. These
in only 44% of patients with good outcome; there were atypi- authors found the disappearance of frequencies of the 12 to
cal spindles in 11% and asymmetry in 25% of spindle EEGs. 14 c/sec range between days 3 and 5 after trauma to be one of
The EEGs of patients with moderate disability outcome their most striking results. On the third day after trauma, a
showed spindles in 90% of all records during the acute stage change of sleep-like activity with spindles to a slow monoto-
of coma, but atypical spindles (33%) and asymmetry (33%) nous pattern was also observed in children. There was no influ-
significantly increased. A remarkable increase of atypical spin- ence of this change on the final outcome (98). On the other
dles (50%) was seen in prolonged coma. The EEGs of patients hand a sleep organization close to normal based on 24-hour
who developed severe disability showed spindles in 61% of all polysomnographic recordings in prolonged stages of posttrau-
cases. Atypical spindles were found in 45%; asymmetry was matic coma was a reliable prognostic marker, both for survival
found in 63% of spindle EEGs. Patients with prolonged coma and for functional recovery (42).
and subsequent severe disability showed only atypical spin-
dles, and the percentage of spindles decreased to 24%.
Asymmetries were seen in 40%. Patients who suffered brain GOOD OUTCOME CATEGORY
death within 2 weeks after trauma showed spindles in 56% in
the acute stage of coma. Asymmetry of spindles was seen in Typical spindles and reactivity are seen in the majority of
88% and atypical spindles were seen in 44% of spindle coma. EEGs in acute coma, but spindles are clearly reduced in pro-
In prolonged coma, spindles were seen in only two cases longed coma (Fig. 22.5). Reactivity of the alerting type is the
(12%), and both were asymmetrical. most prominent response, but, in rare cases, reactivity of the
From these findings, one can conclude that the observation blocking type or disappearance of spindles was the result of
of spindles largely depends on the time of the EEG recording. auditory or painful stimulation. The variety of the different
Variations of the timing of EEG recordings account for the EEG patterns is high and only slightly reduced in prolonged
incidence of spindles varying between 14% and 67% in differ- coma. Hence, these EEG patterns strongly correspond to the
ent reports (75,92) and may also explain why the prognostic patterns found in MBS 2 (Fig. 22.1). Interestingly, eight
value of spindles was doubted (93–95). Within 2 days after patients showed decerebrate posture, leading to the classifica-
brain injury, the vast majority of EEGs showed spindles tion of MBS 4. In contrast to these alarming neurologic find-
(12,45,75,93). ings, the CT scan was normal or slightly abnormal in these
418 Part III ■ Clinical EEG: General

Figure 22.6. A: EEG from a 23-year-old man in acute coma due to sec-
ondary brainstem dysfunction (classical midbrain syndrome stage 2
without lateralization). Easily recognizable typical spindles, symmet-
rical over both hemispheres, accompanied by K complexes and diffuse
slowing. Computed tomography (CT) scan: moderate brain edema
Figure 22.5. The EEG patterns in acute (top) and prolonged (bottom) with scattered contusional areas in left frontoparietal and right tempo-
coma in patients with good recovery. Typical spindles (arrow) are seen ral regions. No signs of compression at tentorial level. Blood within
in the majority of EEGs in acute coma but are clearly reduced in pro- the interhemispheric fissure. B: EEG from the same patient in pro-
longed coma. REAC, reactivity; other abbreviations as in Figure 21.1. longed coma. No spindles, superimposed fast activity, and change to
(From Rumpl E, Prugger M, Bauer G, et al. Incidence and prognostic high-voltage short- and long-delta runs (alternating pattern). No later-
value of spindles in post-traumatic coma. Electroencephalogr Clin alizing signs in the EEG. Outcome: good recovery. (From Rumpl E,
Neurophysiol. 1983;56:420–429.) Prugger M, Bauer G, et al. Incidence and prognostic value of spindles
in post-traumatic coma. Electroencephalogr Clin Neurophysiol.
1983;56:420–429.)

cases. Therefore, a primary brainstem lesion was thought to be


the principal cause of coma. Typical spindles were noted in
patients with either primary or secondary brainstem lesions
SEVERE DISABILITY OUTCOME CATEGORY
(Figs. 22.6 and 22.7).
There is a decrease of spindles and reactivity and rarefaction of
MODERATE DISABILITY OUTCOME the number of EEG patterns in acute and prolonged coma
CATEGORY (Fig. 22.9). No typical spindles are found in prolonged coma.
These alterations in EEG patterns are similar not only to those
The EEG patterns in acute and prolonged coma show a wide found in MBS 3 and MBS 4 (Fig. 22.1) but also to those seen in
range of variability, similar to that seen in the good outcome cat- the transitional stage to the apallic syndrome (see the section
egory (Fig. 22.8). Typical spindles are reduced in prolonged coma, “EEG Changes in Prolonged Comatose States and the Apallic
but otherwise no significant change in other EEG patterns is seen. Syndrome”).
Chapter 22 ■ EEG and Craniocerebral Trauma 419

Figure 22.7. A: EEG from a 20-year-old man in acute coma due to pri- Figure 22.8. The EEG patterns in acute and prolonged coma in patients
mary brainstem injury. Neurologic signs of atypical midbrain syn- with moderate disability outcome. Typical spindles (arrow) are fre-
drome stage 4 (decerebrate posture). Typical spindles, delta bursts, quently seen in acute coma and show reduction in prolonged coma. No
and superimposed fast activity accompanied by diffuse slowing. CT dramatic change in other EEG patterns between acute and prolonged
scan: normal. B: EEG from the same patient in prolonged coma. coma. Reactivity is closely related to the capacity for spindling.
Predominant alpha/ theta activity, several delta waves, reactivity in Observations hampered by small number of patients. REAC, reactivity;
form of blocking slow waves, and no lateralizing EEG signs. other abbreviations as in Figure 21.1. (From Rumpl E, Prugger M, Bauer
Characteristic EEG pattern for pontine lesions. Outcome: good recov- G, et al. Incidence and prognostic value of spindles in post-traumatic
ery. (From Rumpl E, Prugger M, Bauer G, et al. Incidence and prog- coma. Electroencephalogr Clin Neurophysiol. 1983;56:420–429.)
nostic value of spindles in post-traumatic coma. Electroencephalogr Clin
Neurophysiol. 1983;56:420–429.)

trauma). With worsening of outcome, there is a steady decrease


of spindle activity accompanied by an increase of atypical spin-
BRAIN DEATH CATEGORY dles and asymmetry. In prolonged coma, spindles are less fre-
quent; atypical forms and asymmetries correspond with the
All EEG patterns already reduced in acute coma showed further outcome, as in cases of acute coma. The overall incidence of
reduction in prolonged coma (Fig. 22.10). Low-voltage output spindles in the first 2 days of coma is high (91%), but it shows
EEGs were seen in three cases in acute coma. In prolonged coma, a remarkable decrease of 30% in the following days. The brain’s
the number of low-voltage output EEGs increased to seven, and capacity for spindling is closely related to its capacity for EEG
isoelectric EEG was found in four cases. Reactivity was poor in arousal; both are partially lost in late stages of MBS and BBS, as
acute and prolonged coma. Typical spindles were rarely seen; well as in prolonged coma. The disappearance of spindles in
they were symmetrical only in one case. In rare cases, remnants prolonged coma suggests that spindle activity cannot predict
of spindles were noted (Fig. 22.11). On the whole, these patterns the outcome in prolonged coma. On the other hand, if spindle
correlate strongly with the findings in BBS 1 and BBS 2, activity is present, asymmetry and distortion are of prognostic
respectively. value. Atypical spindles and asymmetries significantly increase
There is good reason to assume that spindle activity is of in patients with bad outcome. Generally, the decrease of spin-
great prognostic value in acute coma (i.e., within 2 days after dles not only indicates the deepening of coma, but more
420 Part III ■ Clinical EEG: General

Figure 22.10. The EEG patterns in patients who suffered brain death
within 14 days after brain injury. In acute coma, typical spindles are
Figure 22.9. The EEG patterns of patients with severe disability out- rarely seen (symmetrical only in one case). In prolonged coma, typical
come in acute and prolonged coma. Note the decrease of typical spin- but asymmetrical spindles are seen in one case only. There is similar
dles (arrow) in acute coma and the total loss of typical spindles in decrease in reactivity and limited variety of EEG patterns. Columns of
prolonged coma. Also reactivity is less frequently seen in these cases. delta/ subdelta activity include three low-voltage EEGs in acute coma
Note the general decrease in the variety of EEG patterns. REAC, reactiv- and seven low-voltage EEGs and four isoelectric EEGs in prolonged
ity. A/ T, predominant alpha and little theta activity; T/ D, predominant coma. REAC, reactivity; other abbreviations as in Figure 21.1. (From
theta and little delta activity; D/ SD, predominant diffuse rhythmic and Rumpl E, Prugger M, Bauer G, et al. Incidence and prognostic value of
arrhythmic delta and subdelta activity; DB, delta bursts; DS, short runs spindles in post-traumatic coma. Electroencephalogr Clin Neurophysiol.
of delta; DL, long runs of delta; TSP, typical sleep potentials; ASP, atyp- 1983;56:420–429.)
ical sleep potentials; S, spindles; LS, local slowing; US, unilateral pre-
dominant slowing; SFA, superimposed fast activity; MBS, midbrain
syndrome; BBS, bulbar brain syndrome; N, number of records. (From correlation with symmetry and form of spindles as well as with
Rumpl E, Prugger M, Bauer G, et al. Incidence and prognostic value of the results of CT scan. Exceptions are seen in cases of primary
spindles in post-traumatic coma. Electroencephalogr Clin Neurophysiol. brainstem injury. Despite decerebrate posturing, many patients
1983;56:420–429.) recover well (12,91); others die or survive in a state resembling
a posttraumatic locked-in syndrome (99). In these cases, one
might assume either reversible or irreversible impairment of
frequently heralds the transition to a prolonged comatose state. the midbrain or brainstem, respectively, but undisturbed thala-
In such a case, the loss of spindles is of less prognostic signifi- mocortical connections subserve well-formed spindle activity.
cance when other EEG signs of favorable outcome, such as reac- On the other hand, there are no significant differences in spin-
tivity, alternating patterns, high voltage, and symmetry of dle activity in the different outcome categories when cases of
activities, remain unchanged. primary and secondary brainstem lesions are compared. One
There is no significant influence of small contusional areas must consider, however, that cases of isolated primary brain-
in the CT scans on spindle activity (91). Larger hemispheric stem injury are rare (4) and that the hemispheres are also fre-
lesions are accompanied by increasing asymmetry of spindle quently involved. This may be especially true in patients with
activity. Furthermore, the neurologic findings show close bad outcome.
Chapter 22 ■ EEG and Craniocerebral Trauma 421

doses most of these drugs evoke activation of beta activity


(superimposed fast activity), accompanied by a steady increase
of theta and delta waves with increasing anesthesia depth (see
Chapter 56). Since the neurologic examination is also severely
hampered by the influence of high-dose sedative or anesthetic
drugs, many intensive care units prefer to reduce sedation sys-
tematically, to allow useful neurologic examination. This point
of time would be also the best moment for doing an EEG con-
trol examination besides continuous EEG monitoring.

EEG CHANGES IN PROLONGED COMATOSE


STATES AND THE APALLIC SYNDROME
In prolonged coma, neurologic findings are similar to those
seen at the acute stage; only small differences are observed
(103). The EEG changes in this period have been described in
detail see EEG CHANGES IN THE ACUTE STAGES OF POST-
TRAUMATIC COMA. There are, however, more characteristic
EEG patterns during the transitional stage from MBS to the
traumatic apallic syndrome (104). The clinical symptomatology
of the transitional stage is not limited to patients with evolution
into the full stage of an apallic (vegetative) syndrome but also
occurs in patients who recover with a marked organic brain
syndrome characterized by various severe neurologic signs of
multilocal brain damage.

CLASSIFICATION OF PROLONGED COMA


AND THE APALLIC SYNDROME
Figure 22.11. A: EEG from a 17-year-old girl in acute coma due to sec-
ondary brainstem involvement (classical midbrain syndrome stage 4). Transitional Stage to the Apallic Syndrome
Typical (mildly abnormal) and asymmetrical spindles. Spindles are bet- During the prolonged evolution of a comatose state, the
ter seen on the left. Slight reduction in amplitudes of slow waves on the patients are still in deep coma with closed eyes. At the end of
right. CT scan: areas of local brain contusions at basal ganglia level on this stage, within 2 or 3 weeks, the survivors begin to open their
both sides. Small hemorrhagic lesion in the posterior part of the left eyes, initially for very short periods and after arousing stimuli.
internal capsule. Signs of tentorial compression. Blood in the third ven- A steady increase of chewing, a decrease of decorticate or decer-
tricle. B: EEG from the same patient in prolonged coma. Atypical ebrate rigidity, an increase of extrapyramidal symptoms, and
severely distorted spindles in the left frontal region. Spindles are hardly normalization of the reflex eye movements can be observed.
recognizable and can only be detected from knowledge of the previous Patients react to painful or acoustic stimuli with varying
EEG (A). Outcome: brain death. (From Rumpl E, Prugger M, Bauer G, degrees of extensor and flexor response in the limbs accompa-
et al. Incidence and prognostic value of spindles in post-traumatic nied by an altered respiratory pattern. The most important and
coma. Electroencephalogr Clin Neurophysiol. 1983;56:420–429.) characteristic feature of this stage is the onset of overactivity of
the sympathetic nervous system, leading to extensive tachycar-
Although there is increasing doubt about the effect of barbi- dia. This overactivity has been confirmed by the demonstration
turate anesthesia in the treatment of intracranial hypertension, of high catecholamine plasma levels (mainly of norepineph-
barbiturate anesthesia may be advisable in certain cases (100). rine) (105). This sequence of events may begin in stages 2 to 4
This therapy profoundly alters the EEG activity. The EEG by of MBS but is most frequently seen in MBS 3 and 4 (106).
itself is not sufficient to predict the level of barbiturate anesthe-
sia under clinical conditions in which adjuvant medications are Full Stage of the Apallic Syndrome
used (101). However, it is generally accepted that the appear- The term apallic is used for a clinical syndrome (107) without
ance of burst-suppression activity points to a therapeutic any implication of a particular cerebral lesion. The patients lie
plasma level of barbiturates (102). There is no doubt that the with open eyes for longer periods, paying no attention to events
EEG patterns mentioned above are widely suppressed by this around them. They seem to be awake, but not aware. They do
therapy; consequently, any prognostic value of the EEG is lost. not notice events around them. They may show no blink
Therefore, obtaining early EEG recordings before treating with response to menace and react with abnormal movements only
barbiturates is advocated. This also pertains to cases treated to painful stimulation. There are no emotional reactions. Chewing
with large doses of other sedative or anesthetic drugs. In low or teeth grinding occurs spontaneously or may be evoked by
422 Part III ■ Clinical EEG: General

tactile perioral stimuli. The muscle tone is increased, and only uniform, frequently high-voltage activity creates the impres-
fragments of coordinated movements can be observed. The sion of increasing rostrocaudal deterioration. This impression
sleep-waking rhythm seems to be controlled by exhaustion. In may be supported by the decrease of reactivity; there may be
the waking state, the overactivity of the sympathetic nervous no reactivity to any mode of sensory stimulation (48,76). A
system continues; during sleep, a shift to a parasympathetic set similar evolution of the EEG is seen in patients developing the
of responses may be observed. transition stage from MBS 2. In contrast to the unfavorable
outcome of patients with increasing intracranial pressure, the
State of Stupor and Confusion loss of the activities mentioned above does not carry a poorer
In less severe cases, the patients show prolonged symptoms of prognosis when seen in patients at the transition stage
early MBS (usually MBS 1) and pass into a state of stupor and (46,76). Patients in MBS 4 show an already-simplified EEG
confusion. The patients are usually drowsy, lying in a flexed atti- pattern with only slight further changes at the transition stage.
tude. During the waking state, patients may be resistant to nurs- Loss of atypical sleep potentials may be observed, or reactivity
ing care and examinations, disoriented, and sometimes noisy may be abolished (76).
and violent. Neurologic focal symptoms become more evident. The variety of EEG patterns is also reduced in the full stage of
Most of these patients need sedative drugs. This condition may the apallic syndrome, but this reduction is less marked than in
last for days or even weeks and gradually passes. Symptoms of the transitional stage. The variety of EEG patterns increases as
local brain damage may persist and may cause neurologic and soon as cyclic sleep and waking activity can be identified. This is
mental deficits of varying severity. associated with the spindles and long runs of delta (Fig. 22.14).
On the other hand, the EEG of patients developing the full stage
RELATION OF EEG ABNORMALITIES TO THE of the apallic syndrome from MBS 4 remains rather uniform
and shows only a little difference between sleep and waking
STAGES OF PROLONGED COMA AND THE activities. Periods of fast activity of low voltage, along with eye
APALLIC SYNDROME closure, usually characterize sleep, whereas high-voltage slow
The EEG patterns of patients in the transitional stage of the waves indicate the waking state (76,109). Similar alternating
apallic syndrome are compared with an EEG recorded during periods of faster low-voltage activity and high-voltage slow
the acute MBS 3 in Figure 22.12. There is a clear decrease in waves during were found in sleep in long-lasting nocturnal
the variety of EEG patterns, especially of sleep or sleep-like records (110–112). The appearance of EEG sleep patterns and
potentials and alternating patterns (76,108). A complete loss circadian EEG variations is a favorable prognostic sign (112).
of spindles is noted in these patients (Fig. 22.13). This evolu- Completely organized sleep patterns or increasing organiza-
tion of the EEG patterns from waveforms of sleep into rather tion of nocturnal sleep in successive recordings indicates a more
favorable outcome than the absence of such patterns. The
occurrence of REM sleep is especially significant in this regard
(108). This finding may be supported by reports of the reap-
pearance of slow-wave sleep with continued absence of the
REM phase in patients who never regained consciousness after
brainstem vascular accidents (113,114). In rare cases, positive
sharp waves in the occipital regions are seen during sleep (21).
In contrast to reports that suggest that sleep or sleep-like
activities point to a favorable outcome, there is no evidence
that sleep or sleep-like activity found at the full stage of the
apallic syndrome months after the brain injury gives any
information about further outcome (115). Patients with rela-
tively well-developed sleep stages persisted clinically at the
stage of an apallic syndrome or remained severely disabled
(Fig. 22.15), whereas patients with severely altered sleep
showed further recovery. However, the value of long-term
EEG recordings in these patients was hampered by the fact
that given stages of sleep or wakefulness lasted no longer than
a few seconds or minutes. This phenomenon appears to be
Figure 22.12. The EEG patterns of 13 patients in the transitional stage rather characteristic for apallic patients (109,115). REM sleep
to the traumatic apallic syndrome evolving from midbrain syndrome was hardly separated from sleep stage 1; rapid eye movements
stage 3. Dark columns demonstrate the decrease of EEG patterns at the appeared at any stage, even during synchronized sleep.
transition stage compared with the EEG patterns at the acute stage of Atypical and asymmetric spindle activity continued to be of
midbrain syndrome (white columns). Note the complete loss of spindles prognostic significance by indicating a persistent structural
in these cases. Abbreviations as in Figure 22.1. (From Rumpl E. Elektro- hemispheric lesion. Generally, the prognostic value of long-
neurologische Korrelationen in den frühen Phasen des posttraumatis- lasting nocturnal EEG recordings is very limited in apallic or
chen Komas. II. Das EEG im Übergang zum, und im Vollbild des severely disabled patients if done months after the craniocere-
traumatischen apallischen Syndroms. Z. EEG-EMG. 1980;11:43–50.) bral trauma.
Chapter 22 ■ EEG and Craniocerebral Trauma 423

Figure 22.13. A: EEG from a 24-year-old woman in acute


midbrain syndrome stage 3 without clinical signs of lateral-
ization. After acoustic stimulus (arrow), appearance of spin-
dles followed by delta and subdelta activity of higher
amplitude. Note the symmetry of reaction. B: EEG from the
same patient in the transition stage to the apallic syndrome
4 days after first recording. Uniform delta and subdelta
activity; loss of spindles. After acoustic stimulus (arrow),
long run of delta. Note the symmetry of reaction.

Figure 22.14. The EEG patterns of the same 13 patients as in Figure 22.12 Figure 22.15. Nocturnal EEG from a 29-year-old woman at the full stage
at the full stage of the apallic syndrome (dark columns) compared with the of an apallic (vegetative) syndrome. Later she has learned to move the
EEG patterns at the acute stage of midbrain syndrome (white columns). left lower extremity on command but remains severely disabled. Typical
Note the general acceleration and the reappearance of spindles and spindles, K complexes during sleep, stage 2. Continuous myogenic spik-
altered sleep activities. Abbreviations as in Figure 22.1. (From Rumpl E. ing in left temporal region due to palatal myoclonus. (From Rumpl E,
Elektro-neurologische Korrelationen in den frühen Phasen des posttrau- Prugger M, Bauer G. Zum prognostischen Wert elektroenzephalo-
matischen Komas. II. Das EEG im Übergang zum, und im Vollbild des graphischer Schlafbeobachtungen im traumatisch bedingten apallischen
traumatischen apallischen Syndroms. Z. EEG-EMG. 1980;11:43–50.) Syndrom. Neuropsychiatr Clin. 1984;3:219–232.)
424 Part III ■ Clinical EEG: General

The EEG patterns of patients in the state of stupor and EEG CHANGES AT EARLY STAGES AFTER
confusion show a decrease in the variety of EEG patterns sim- CRANIOCEREBRAL TRAUMA WITHOUT
ilar to the one seen in prolonged coma. However, sleep or
EVIDENCE OF BRAINSTEM DYSFUNCTION
sleep-like potentials are more frequently seen in these
patients in diurnal examinations. Generalized theta with less In mild or moderate head or brain injuries, there may be no or
delta activity is the predominating pattern at this stage; it only mild disturbances of consciousness with no clinical signs
is usually seen in waking periods and without further change of primary or secondary brainstem dysfunction. In general, the
after stimulation. The value of most records at this stage EEG recovery is fast in these patients, who fortunately form the
is hampered by the use of the sedative drugs needed to obtain vast majority of cases after craniocerebral trauma.
technically satisfactory recordings. Drug-induced sleep
activities might also indicate a favorable outcome in most of
these patients, and asymmetries in sleep potentials might be RELATION OF EEG ABNORMALITIES TO
of diagnostic significance in cases of chronic subdural MILD OR MODERATE STAGES OF CEREBRAL
hematoma. CONCUSSION
The reaction to external stimulation is remarkably reduced
in the transitional stage of the apallic syndrome. In most cases, In cases of commotio cerebri (cerebral concussion with loss of
no response can be observed (48,76). Less frequently, a delayed consciousness) but with no impairment of consciousness at the
reactivity characterized by long runs of delta is noted. No reac- time the records were taken, normal or borderline EEGs were
tivity is seen in patients in the full stage of the apallic syndrome found in the vast majority of cases within the first 24 hours
during the waking state, whereas stimulation during sleep fre- (27,28,32). No slowing of the alpha rhythm could be detected in
quently induces long-lasting waking activity (36,109). these recordings (19). The slowing of alpha rhythm is usually
Metabolic disturbances decrease in patients in prolonged considered to be the slightest degree of generalized disturbance
comatose states (10). Therefore, the EEG abnormalities are less after contusio cerebri and may only be found with repeated
influenced by metabolic factors than in the stages of MBS. EEG, because the significant slowing occurs in the alpha fre-
Nevertheless, the influence of metabolic disturbances and drugs quency range (19,23).
must be taken into consideration at these stages. The mildest disturbance of consciousness seen after cranio-
Epileptic discharges are not seen in patients in the transi- cerebral trauma is drowsiness and hypersomnia, which are
tional stage or full stage of the apallic syndrome (18,76) but accompanied by the EEG findings of normal sleep (48). There
may occur in the state of stupor and confusion and during the are usually generalized slowing of all frequencies and altered
remission stage from the apallic syndrome (18). sleep patterns in patients with barely existent periods of wake-
Lateralized signs in the EEG are slightly reduced in pro- fulness. Sensory stimulation will block the slowing and sleep.
longed comatose states, mainly due to the loss of asymmetrical There may be gliding clinical and electroencephalographic
typical or atypical sleep potentials. Asymmetry of reactivity is transitions to the first stage of MBS. However, the change from
diagnostically helpful (48). abnormal EEG activity to a more normal pattern is fast in these
Prognostic criteria can be derived from EEG findings only cases (Fig. 22.16).
with great care and caution. A gradual increase in frequency In contrast to the aforementioned fairly good correlation of
from delta to faster activity points to a favorable index only EEG and clinical findings, there are many patients with head
for survival, not for recovery (25). Improvement of EEG must injury who show marked discrepancies between EEG patterns
be accompanied by clinical improvement to have any prog- and clinical findings. A normal neurologic status in a patient
nostic significance (18,25). An isoelectric EEG indicates loss with no or little complaint may be accompanied by impressively
of bioelectrical activity, to be discussed in Chapter 23, on abnormal EEGs (Figs. 22.17 and 22.18). Similar EEG findings
coma and brain death. Patients who survive with an isoelec- may also be seen in patients with cerebral concussion without
tric EEG are classified as with complete apallic syndromes by loss of consciousness but with a short or longer lasting alter-
some authors (116,117). In cases of very low-voltage EEG in ation in mental status (119). The EEG would most likely detect
repeated recordings, one can expect survival without hope of cortical disease in acute cases with mild injury, even if the CT
recovery as a social human being (117). In patients who scan or MRI is normal. Very little is said in the literature about
develop the symptomatology of the apallic syndrome from these cases, and only a few authors regard them as mild forms
MBS 4 accompanied by rather rarified EEG patterns, the of concussion (120–122). In contrast, a normal EEG can be seen
prognosis for social recovery is also very poor (76,106). Most in patients with significant impairment in neuropsychological
of these patients remain at the full stage of the apallic syn- performance after mild traumatic brain injury within 24 hours
drome or at a minimally conscious state (118). The favorable and up to 6 weeks after injury (123).
clinical outcome of patients with organized sleep has been Particularly pronounced discrepancies between electrical
mentioned above. The persistence of sleep patterns or sleep- cortical activity and the clinical state may be found in children
like activities at the transitional stage of the apallic syndrome (127–129). Children may show high-amplitude generalized delta
might also indicate a favorable outcome although the value of activity for weeks or months after a mild craniocerebral trauma
this observation is hampered by the very small number of and in the presence of normal consciousness (48). Immaturity
cases studied (76,92). might enhance functional traumatic (postconcussion)
Chapter 22 ■ EEG and Craniocerebral Trauma 425

Figure 22.16. A: EEG from a 20-year-old woman with cerebral contu-


sion, drowsiness, and clinical signs of left frontoparietal lesion. Delta
and subdelta activity, unilateral slowing, and suppression of altered
spindles in the left hemisphere. B: EEG from the same patient, 1 day
after first recording. Reappearance of reactive alpha activity, local slow-
ing in left fronto-temporo-parietal region. C: EEG from the same patient,
3 days after first recording. The EEG is nearly normal; intermittent slow
waves in left temporal region. Full clinical recovery with the exception
of headache, lack of concentration, and dizziness.

disturbances and produce focal or generalized EEG abnormali- in elderly patients with early epileptic seizures (131). The loss of
ties (21). On the other hand, focal changes have been thought reactivity may appear in light stages of coma, indicating a bad
to be a significant sign of contusional brain damage (130). prognosis (21).
Elderly persons may have pretraumatic EEG abnormalities,
especially in the temporal regions, due to cerebrovascular insuf- Epileptic Discharges
ficiency. The combination of traumatic and vascular factors The term early epilepsy should be confined to fits in the first
makes it difficult to distinguish the EEG abnormalities caused week after injury (132). Localized focal motor epilepsy is most
by the cerebral contusion. There is particularly high mortality common in the first week, whereas acute temporal lobe
426 Part III ■ Clinical EEG: General

Figure 22.17. EEG from a 35-year-old man complaining of


vertigo after mild cerebral contusion. At the time of EEG
recording, normal neurologic status and CT scan. Intermittent
slow activity (theta and delta) in left frontotemporal region.

Figure 22.18. EEG from a 45-year-old woman after mild cerebral


contusion without signs of brainstem impairment. Some com-
plaints about vertigo. At the time of EEG recording, normal neu-
rologic and CT scan findings. Intermittent left temporal theta
focus. The recovery of EEG abnormalities follows the same pat-
tern as in adults (23). In children with transient cortical blind-
ness following mild head trauma, posterior slowing with
subsequent fast normalization can be observed (124).
Lateralized signs consist mainly of focal slow waves in the theta
and delta range (17,19,23,32), whereas the other cerebral
regions show only minor changes (125). A close correlation
with neurologic deficits is seen within the first months
(17,19,20,126).

epilepsy with psychomotor or complex partial seizures is occipital pathology on CT or MRI scanning is generally tran-
never found during this period. Generalized seizures were sient (Fig. 22.19), occipital status epilepticus accompanied by
found in 50% of the cases, while the other 50% had focal occipital edema may lead to persistent visual deficit if not
attacks (21). A higher percentage of focal seizures is assumed, detected for early treatment.
because of secondary generalization and doubtful clinical Long runs of continuous activity of focal spikes or unilateral
observation (21). After 2 weeks, adults with posttraumatic spike-wave discharges are the most common EEG pattern in
epilepsy may have any type of epileptic manifestation, with early epilepsy (21). In general, focal epileptiform patterns asso-
the exception of petit mal attacks with generalized rhythmic ciated with diffuse EEG abnormalities suggest a greater proba-
synchronous 3/sec spike-and-wave activity in the EEG (133). bility of subsequent chronic epilepsy than does focal
Although epilepsy during the first week recurred or persisted epileptiform activity in an otherwise normal EEG (48). Using
in less than one third of patients (the recurrence rate in the standard 24-hour recordings, seizure activity can be seen after
second week is over 70%), the risk for late epilepsy is signifi- trauma in up to 35% of patients with mild or severe head-brain
cantly greater for patients with early fits than for those with- injury in the first weeks after trauma, but accompanying epilep-
out (132). A threefold increase in the incidence of both early tic fits are rare (135). In cases of severe brain injury early elec-
and late seizures is seen in patients with depressed skull frac- troencephalographic seizure activity might be seen in patients
tures when compared with injuries accompanied by nonde- who will have clinical seizures shortly before or after the dis-
pressed fractures (48). Occipital status epilepticus is a rare charges are found in the EEG (Fig. 22.2).
event in posttraumatic blindness (134). While cortical blind- Epileptiform activities are more frequently seen in children
ness in traumatic brain-injured patients with no or minimal than in adults. These are isolated spikes or spike waves, sharp
Chapter 22 ■ EEG and Craniocerebral Trauma 427

Figure 22.19 A: EEG from a 6-year-old girl who had a fall on the back of her head after an unexpected push while playing.
She had a period of blurred vision, delayed answers to questions, disorientation, drowsiness, and stereotyped finger move-
ments, which lasted about 1 hour. The CT and magnetic resonance imaging scans were normal. At the time of the EEG
recording 2 days after the accident, the patient was alert with no neurologic or mental deficit. The EEG showed high-voltage
occipital delta activity intermingled with sharp transients. B: EEG from the same patient 3 days later within the range of
normal.
428 Part III ■ Clinical EEG: General

waves, and spike-wave complexes, localized or generalized. A However, it should be noted that most of the posttraumatic
single epileptic focus in the EEG after severe head trauma has complaints do not appreciably differ from nontraumatic neu-
the same favorable outcome as in benign focal epilepsy (136). rotic syndromes.
Generalized irregular spike-wave activity is occasionally seen
(48). The 14 and 6/sec positive spikes can be an abnormal find-
ing after mild or severe head injury in children. This pattern RELATION OF EEG ABNORMALITIES
may appear as a delayed reaction to mild injury, but may also be TO THE LATE STAGES OF
seen during recovery from severe injury (137). However, the CRANIOCEREBRAL TRAUMA
significance of 14 and 6/sec positive spikes is of less value,
because it may also be seen in healthy children (138). All of EEG recovery from the full stage of the apallic syndrome is gen-
these discharges can be observed within hours of the trauma erally associated with an acceleration of EEG activity. Theta and
(more commonly within days or weeks), and all are associated (fewer) delta waves characterize the apallic syndrome months
with a good prognosis. after the brain injury (18,142). Recovery from this stage is char-
acterized by the decline of theta and delta waves and reappear-
ance of reactive alpha activity. Favorable outcome is associated
EEG CHANGES AT LATE STAGES AFTER with faster regression of theta waves and more progressive rein-
CRANIOCEREBRAL TRAUMA stitution of alpha activity (142,143). In a summary of their
findings, Lücking et al. (18) have been able to draw the follow-
In late stages after craniocerebral trauma, clinical and EEG ing conclusions on the prognostic value of the EEG in the apal-
examination demonstrate diminished correlation, which fur- lic syndrome: (i) Predominant theta and delta activity
ther increases after a 3-month interval (17,19,21,130). This lack unchanged for several months points to a significant lesion of
of correlation may also occur in patients during recovery from the hemispheres. (ii) An early and progressive appearance of
an apallic syndrome (18). alpha activity in the absence of clinical improvement suggests
slight cortical, but extensive subcortical, lesions. (iii) Alpha fre-
quency may show a gradual acceleration within years without
CLINICAL CLASSIFICATION further clinical improvement. (iv) Focal symptoms become
Recovery from the Apallic Syndrome more marked after the decrease of general changes and may
shift from side to side in cases of temporal foci. (v) Epileptic
Although the symptomatology of the apallic syndrome is
discharges may occur in patients with no evidence of epileptic
remarkably uniform, there are different degrees of telencephalic
fits, although patients with focal epileptic attacks may have no
lesions, so that recovery is possible (107). During the remission
epileptic discharges in repeated EEGs.
stage (139), there is evidence of increasing reintegration of higher
EEG recovery of generalized abnormalities is characterized
brain function. The onset is indicated by primitive emotional
by a steady increase in frequency. After comatose states, gener-
reactions in response to painful stimuli followed by optical fixa-
alized theta activity is one of the most constant features of the
tion and displeasure reactions. The first voluntary movements
EEG recovery (48) and may show further improvement with
are usually seen in the fingers. The patient starts to respond to
gradually evolving reactive alpha activity. After mild head
simple orders. In patients who make a reasonable recovery, fur-
injuries with cerebral contusion and slowing of the basic
ther stages of remission appear, marked by the symptomatology
rhythm, follow-up EEGs are often characterized by a gradual
of the Klüver–Bucy syndrome, the Korsakoff syndrome, and,
increase of alpha frequency returning to normal within several
finally, the organic brain syndrome of varying severity.
weeks or months (17,19,23,32). Six months after trauma, alpha
Symptoms of local brain lesions can now be clearly determined.
slowing persists in only 4% of the cases (23).
These lesions may be permanent, but in some cases the apallic
EEG recovery of lateralizing signs shows a characteristic
syndrome is transient with complete recovery of the patient.
evolution in uncomplicated cases. Unilateral slowing or a delta
In mild and moderate brain injuries, the onset of the late
focus will gradually change into a theta focus or a focus of irreg-
stage can be clinically defined by the disappearance of distur-
ular waves of changing frequency. This focus may turn into the
bances of consciousness and confusion and by the stabilization
focal slowing of alpha rhythm in the parietal, posterior, and
of neurologic deficits and psychic syndromes (21). Usually, a
occipital regions with the slightest degree of local disturbance
gradual recovery is seen, but in some cases neurologic and
(17,19). This focal slowing usually disappears quickly and may
mental deficits may persist.
sometimes be followed by focal reduction in amplitude (21).
The diagnostic value of such alpha asymmetries is doubtful,
Posttraumatic Syndrome however, when one considers the frequent occurrence of such
This syndrome comprises headache, vertigo, dizziness, nervous- asymmetries in healthy persons (20). Another rare and tran-
ness, irritability, impaired memory, disturbances of concentra- sient local sign is focal activation of alpha rhythm (31) charac-
tion, excessive fatigue, insomnia, and intolerance to alcohol terized by a focal slowing and increase of alpha amplitude by
(140). Practically, in 50% of the patients, the symptoms disap- eye closure; eye opening fails to block the activity.
pear within 2 weeks after mild injury, while in 25% the symp- In cases with traumatic psychosis (delirium, Korsakoff
toms continue for months and sometimes years (141). syndrome, and changes of affect), EEG foci are demonstrated in
Chapter 22 ■ EEG and Craniocerebral Trauma 429

95% of cases, bilaterally in 70%. Normalization occurs within showed marked fluctuations in the degree of abnormality sug-
3 months in 48%, with foci persisting more than 2 years in gesting a vascular mechanism for these changes (147).
22% mostly in patients with traumatic epilepsy. Focal signs The EEG changes might indeed be due to fluctuations in the
initially consist of delta waves (85%) and finally of focal blood supply from the vertebral arteries, which can be impaired
dysrhythmia (73%) with temporal localization increasing from by hyperextension of the neck (148,149). Vertebral fractures with
58% to 82% (24). displacement of fragments may directly compress the vertebral
Even in the late posttraumatic stage, focal EEG abnormali- arteries. Similar patterns in the EEG may appear in this case.
ties may show a close correlation with the clinical focal deficit There are also alternative explanations for the genesis of EEG
(23,30). After closed head injuries, focal EEG abnormalities abnormalities in the wake of neck injuries. The acceleration–
tend to disappear within 6 months (23) to 2 years (19) in the deceleration as such may directly affect the cerebral parenchyma
vast majority of cases. In contrast, focal changes could be (50), or the rapid sequence of backward–forward movements of
found in 50% of cases 20 years after an open craniocerebral head and neck may cause traumatic lesions as the cerebrum
trauma (144), and focal changes in 40% of patients with pro- strikes the inner table of the skull (147). Mostly normal EEG (and
longed confusional states were seen (30). Focal abnormalities also 24-hour cassette recording) were found in 68 cases with
decrease more slowly than generalized ones and may even be whiplash injuries without accompanying head injury (150).
found some years after injury (21). The area of the focal
abnormality usually shrinks in size and may narrow down to
a temporal or parieto-occipital focus (21). In penetrating head COMPRESSED SPECTRAL ARRAY
injury, a close correlation between focal slowing and focal AND BRAIN MAPPING
neurologic deficit may be found even 12 to 16 years after
injury (22). In open craniocerebral trauma, foci often persist Continuous EEG monitoring of patients in the first days fol-
over the area of penetration, and migration to posterior lowing injury requires the use of computer-assisted (Fourier
regions is unusual (127). In rare cases, the location of the focal transform) analysis using compressed spectral analysis (CSA)
abnormality may shift from one area to another (23,29). More or similar techniques (see Chapters 54 and 56). With this
frequently, the foci shift to the frontal rather than to the occip- method the power spectra or specific EEG frequency bands may
ital region, but they may return to the original locus in both be monitored for the long term. The dominant frequencies,
cases (29). their distribution, and their amplitude may be better assessed
with CSA than with conventional EEG (15). Two or four chan-
Epileptic Discharges nels appear to be adequate to regain significant data regarding
Apart from early attacks, late epilepsy may develop within a the outcome of patients (75,97,151,152). Unfavorable outcome
month or two of the injury or may be delayed for many years. was seen in patients with slow and monotonous CSA and in
Early attacks will predispose to late ones (132). A detailed dis- patients with significant interhemispheric amplitude asymme-
cussion of posttraumatic epilepsy is presented in Chapter 27. try. Favorable outcome was frequently found in patients with
The EEG in the posttraumatic syndrome can neither prove changeable CSA, especially with sleep-like CSA (97,152).
nor rule out the existence of traumatic symptoms (145,146). Persistence or return of activity within the alpha or theta range
Even pronounced EEG abnormalities are not necessarily associ- pointed to good outcome in most cases (151). Repeated studies
ated with posttraumatic syndromes (21). In contrast, a normal of CSA showed an increase in the absolute and relative ampli-
EEG cannot exclude severe cerebral damage. tudes of the alpha and theta ranges in patients who survived
(153). Difficulties in interpretation may appear in patients in
EEG Changes in Whiplash Injuries, Cervical alpha or theta pattern coma, as well as in patients undergoing
Fractures, and Dislocations barbiturate therapy. Further difficulties are that CSA may fail to
Whiplash injuries may be accomplished by EEG abnormali- show paroxysmal patterns, such as spikes, brief seizures, burst
ties; similarly, patients with cervical fractures and dislocations suppression, PLEDs, triphasic waves, and intermittent theta and
may demonstrate EEG changes even in the absence of loss of delta activity (84).
consciousness at the moment of the accident (21). So the Calculation of the theta/beta power ratios in patients with
incidence of generalized and focal EEG changes (focal spikes deep coma revealed long-lasting EEG reactions after standard-
and focal slow waves) found in a selected group of whiplash ized repetitive stimulation (154). With this method EEG reac-
injury cases was the same as in patients with closed head injury tion may be elicited even in cases where other methods fail to
(147). An additional injury to the head was carefully excluded evoke an EEG response—acute or delayed—at all. The
in these patients. On the average, the EEGs were done 6 months theta/beta ratio decreased with recovery of cerebral functions. A
after the accident. Temporal lobe foci were more frequently seen bolus injection of 100 to 500 mg of thiopental evokes short-
in whiplash injury than in closed head trauma, but this did not lasting beta stimulation in CSA. This response was of high
prove to be statistically significant. Slow waves appeared to be prognostic significance since 84% of patients with this reaction
accentuated over the posterior regions of the brain. In contrast survived (155).
to the EEG changes after closed head injuries, the EEG became In mild head trauma discriminating EEG power spectral
increasingly abnormal in many of the whiplash injury cases analysis indicated three classes of variables that are attributable
when follow-up records were studied. Moreover, many cases to mechanical head injury: (i) increased coherence and
430 Part III ■ Clinical EEG: General

decreased phase in frontal and frontotemporal regions; (ii) signs of serious brain damage were found among amateur ath-
decreased power differences between anterior and posterior cor- letes (165). On the other hand, a significantly increased
tical regions; (iii) reduced alpha power in posterior cortical incidence of focal theta activity was found in soccer players at
regions (156). These authors speculate that these electrophysio- the end of their careers, probably as the result of a cumulative
logic features are consistent with the mechanics of cerebral effect due to repeated heading (166). Slightly abnormal to
trauma and may explain persistent symptoms after mild trauma abnormal EEGs were found in 35% of active and in 32% of for-
such as headache, dizziness, memory loss, short attention span, mer soccer players. Interestingly enough, there were fewer
and reduced ability to process complex information. abnormal EEG changes among the typical “headers” than
Subsequently, EEG measures of cerebral asymmetry such as EEG among the “nonheaders” (167).
coherence and phase and amplitude symmetry were found to be There is a chronic progressive traumatic encephalopathy in
best predictors of outcome also in patients with severe brain boxers, especially in those with a history of repeated knockout
injury. EEG coherence and phase have been shown to reflect to defeats. A significant increase of EEG abnormalities in boxers
some extent the magnitude of diffuse axonal injury (157). who suffered from this encephalopathy, which is known as the
Topographic brain mapping (TBM) can demonstrate sub- punch drunk state (168) or dementia pugilistica (169), in com-
tle asymmetries and lateralization and localization effects parison to the EEG of more successful boxers was found (170).
more efficiently than standard EEG, when the recording is However, no correlation between the degree of boxer’s
done properly from a technical standpoint (158,159). In 135 encephalopathy and the EEG was eventually seen (170,171).
patients after mild or moderate head injury, abnormal TBM Therefore, the EEG may be useful in determining the severity of
was found in 56% of cases years after injury. The tempo- trauma after a fight, but it is a less sensitive indicator of boxer’s
rofrontal regions were involved in 65%, the temporo-occipital encephalopathy during and beyond the active career. No corre-
regions in 25%, and the parieto-occipital regions in 9% of lation with clinical findings was established even with the use of
patients. The most common type of abnormality was absolute psychometric tests (172). As in EEG recovery after craniocere-
voltage asymmetry. In contrast the standard EEG showed bral trauma of other origins, a normal EEG cannot exclude the
abnormalities in 30% of cases consisting mainly of mild, non- existence or the development of encephalopathy.
specific generalized slowing (158). TBM appears to provide Other sports, such as horseback riding, American football,
better detection of low- amplitude slow activity not easily and rugby, may carry a similar risk of repeated concussion. The
discernible by routine EEG (159). number of injuries might be quite different from that of boxers,
but some of them may be more severe than those usually occur-
HEAD INJURY IN SPORTS ring in the boxing ring. Temporal lobe epilepsy and persisting
mental impairment was found in professional jockeys after
A generalized reduction in amplitude and slow irregular theta repeated riding accidents (173).
activity has been observed in boxers within 15 to 30 minutes
after fighting, with a significant increase of these findings in CONCLUSION
boxers who had sustained a knockout blow (160–162).
Although the EEG might detect even slight degrees of cere- There is a close correlation between clinical and electroen-
brovascular autoregulation disturbance, the method is not rou- cephalographic findings in acute posttraumatic comatose
tinely used to avoid second impact syndrome in contact-sports states. A declining number of observable EEG patterns may
head trauma (119,163). However, the method should be more suggest increasing rostrocaudal deterioration and poor progno-
sensitive than CT or MRI scanning. sis, as well as the development of a prolonged comatose state.
In long-term observations a correlation between the fre- An assessment of the EEG abnormalities without knowledge of
quency of abnormal EEGs and the number of fights and, fur- the clinical symptoms is difficult in these stages. Metabolic dis-
thermore, a correlation between EEG abnormalities and short turbances and drugs may also influence the EEG patterns. Early
time intervals between fights was found (162). Repeated fights epileptic discharges in the EEG are of prognostic significance if
within a short period have particularly ill effects on the brain, they appear prior to clinical seizures. In cases of primary brain-
and the extent and degree of EEG abnormalities increased after stem damage, the EEG can help to differentiate these lesions
each fight (21,162). Usually the EEG of young boxers shows from secondary brainstem involvement. In comatose states, fol-
more serious changes than that of older ones. No severe abnor- low-up EEG is necessary to determine the state of cortical activ-
malities were found in amateur boxers either with many or few ity and its evolution. Early EEG recordings are stressed in all
matches. There was a somewhat higher incidence of slight or cases of craniocerebral trauma because of increasing lack of
moderate EEG deviations among boxers than among soccer correlation between clinical and EEG data in later stages. No
players and track and field athletes. No neurophysiologic vari- conclusion can be drawn from a single EEG at a late stage as to
able was correlated with the number of bouts, number of lost the severity of the original trauma. A single record may only be
fights, or length of boxing career. Also an abnormal EEG did useful in the case of epileptic manifestations. Serial tracings
not predict the degree of clinical impairment. EEG abnormali- during acute and late stages accompanied by close clinical
ties showed no correlation with neurologic findings or the examinations will increase the value of the EEG in any case.
number of bouts the fighter has waged (164). In general, no Secondary deterioration of the EEG after a period of recovery
Chapter 22 ■ EEG and Craniocerebral Trauma 431

suggests late intracranial complications, such as epilepsy, 22. Jabbari B, Vengrow MI, Salazar AM, et al. Clinical and radiological
hematoma, hydrocephalus, abscess, and meningitis, or the correlates of EEG in the late phase of head injury: a study of 515
development of a concomitant cerebral disease. Vietnam veterans. Electroencephalogr Clin Neurophysiol. 1986;64:
285–293.
23. Koufen H, Dichgans J. Häufigkeit und Ablauf von traumatischen
REFERENCES EEG-Veränderungen und ihre klinischen Korrelationen. Fortschr
Neurol Psychiatr. 1978;46:165–177.
1. Jellinger K. Häufigkeit und Pathogenese zentraler Hirnläsionen 24. Koufen H, Hagel KH. Systematic EEG follow-up study of trau-
nach stumpfer Gewalteinwirkung auf den Schädel. Wien Z matic psychosis. Eur Arch Psychiatr Neurol Sci. 1987;237:2–7.
Nervenheilkd. 1967;25:223–249. 25. Strnad P, Strnadová V. Long-term follow-up EEG studies in patients
2. Maciver JN, Lassman LP, Thomas CW, et al. Treatment of severe with traumatic apallic syndrome. Eur Neurol. 1987;26:84–89.
head injuries. Lancet. 1958;2:544–550. 26. Walter W, Hill D, Williams D. Discussion of the electroencephalogram
3. Jefferson M. Altered consciousness associated with brain stem in organic cerebral disease. Proc Soc Med Lond. 1948;41:237–250.
lesions. Brain. 1952;75:55–67. 27. Williams D. The electro-encephalogram in acute head injuries.
4. Mitchell DE, Adams JH. Primary focal impact damage to the brain J Neurol Psychiatry 1941;4:107–130.
stem in blunt injuries. Does it exist? Lancet. 1973;2:215–218. 28. Dow R, Ulett G, Raaf J. Electroencephalographic studies immedi-
5. McNealy DE, Plum F. Brain stem dysfunction with supratentorial ately following head injury. Am J Psychiatr. 1944;101:174–183.
mass lesions. Arch Neurol. 1962;7:26–48. 29. Dawson EE, Webster JE, Gurdjian ES. Serial electroencephalogra-
6. Plum F, Posner JB. Diagnosis of Stupor and Coma. Philadelphia: phy in acute head injuries. J Neurosurg. 1951;8:613–630.
F.A. Davis; 1966. 30. Schneider E, Hubach H. Das EEG der traumatischen Psychosen.
7. Gerstenbrand F, Lücking CH. Die akuten traumatischen Dtsch Z Nervenheilkd. 1962;183:600–627.
Hirnstammschäden. Arch Psychiatr Nervenkr. 1970;213:264–281. 31. Duensing F. Die Alphawellenaktivierung als Herdsymptom in
8. Osborn AG. Diagnosis of descending herniation by cranial com- Elektroencephalogramm. Nervenarzt. 1948;19:544–552.
puted tomography. Radiology 1977;123:93–96. 32. Scherzer E. Wert der Elektroenzephalographie beim Schädel-
9. Reich JB, Sierra J, Camp W, et al. Magnetic resonance imaging meas- trauma. Wien Klin Wschr. 1965;77:543–547.
urements and clinical changes accompanying transtentorial and 33. Davis PA, Davis H. The electrical activity of the brain. Its relation
foramen magnum brain herniation. Ann Neurol. 1993;34:748–749. to physiological states and to states of impaired consciousness. Res
10. Arfel G. Introduction to clinical and EEG studies in coma. In: Publ Assoc Res Nerv Ment Dis. 1939;19:50–80.
Remond A, Harner R, Naquet R, eds. Handbook of 34. Hill D. Psychiatry. In: Hill D, Parr G, eds. Electroencephalography.
Electroencephalography and Neurophysiology, vol. 12. 1975;5–23. London: MacDonald; 1950:319–363.
11. Bricolo A, Turella G. Electroencephalographic patterns of acute 35. Schwab RS. Electroencephalography in Clinical Practice.
traumatic coma: diagnostic and prognostic value. J Neurosurg Sci. Philadelphia, PA: Saunders; 1951:195.
1973;17:278–285. 36. Fischgold H, Mathis P. Obnubilations, comas et stupeurs, etudes
12. Chatrian GE, White JR, Daly P. Electroencephalographic patterns electro-encephalographiques. Electroencephalogr Clin Neurophysiol.
resembling those of sleep in certain comatose states after injuries to 1959;11(suppl):27–68.
the head. Electroencephalogr Clin Neurophysiol. 1963;15:272–280. 37. Loeb C. Electroencephalographic changes during the state of
13. Rumpl E, Lorenzi E, Hackl JM, et al. The EEG at different stages of coma. Electroencephalogr Clin Neurophysiol. 1958;10:589–606.
acute secondary traumatic midbrain and bulbar brain syndromes. 38. Mathis P, Turrubia H, Fischgold H. Réactivité, périodicité et cor-
Electroencephalogr Clin Neurophysiol. 1979;46:487–497. rélation cortico-cardio-respiratoire dans le coma. Electro-
14. Silverman D. Retrospective study of EEG in coma. encephalogr Clin Neurophysiol. 1957;6(suppl):27–68.
Electroencephalogr Clin Neurophysiol. 1963;15:486–503. 39. Naquet R, Vigouroux RP, Choux Bourand C, et al. Étude élec-
15. Stone JL, Ghaly RF, Hughes JR. Electroencephalography in acute troencéphalographique des traumatismes craniens récents dans
head injury. J Clin Neurophysiol. 1988;5:125–133. un service de ré-animation. Rev Neurol. 1967;117:512–513(abst).
16. Synek VM. Value of a revised EEG coma scale for prognosis after 40. Bergamasco B, Bergamini L, Doriguzzi T, et al. EEG sleep patterns
cerebral anoxia and diffuse head injury. Clin Electroencephalogr. as a prognostic criterion in post-traumatic coma. Electro-
1990;21:25–30. encephalogr Clin Neurophysiol. 1968;24:374–377.
17. Jung R. Neurophysiologische untersuchungsmethoden. In: 41. Bricolo A, Gentilomo A, Rosadini G, et al. Akinetic mutism follow-
Bergmann G, Frey W, Schwiegk H. eds. Handbuch der Inneren ing cranio-cerebral trauma. Physiopathological considerations
Medizin V, vol. 1. Berlin: Springer; 1953:1286–1293. based on sleep studies. Acta Neurochir (Wien). 1968;18:68–77.
18. Lücking CH, Müllner E, Pateisky K, et al. Electroencephalographic 42. Valente M, Placidi F, Oliveira AJ, et al. Sleep organization pattern
findings in the apallic syndrome. In: Dalle Ore G, Gerstenbrand F, as a prognostic marker at the subacute stage of post-traumatic
Lücking CH, et al. eds. The Apallic Syndrome. Berlin/Heidelberg/ coma. 2002. Clin Neurophysiol. 2002;113:1798–1805.
New York: Springer; 1977:144–154. 43. Fischgold H, Torrubia H, Mathis P, et al. Réaction EEG d’éveil
19. Meyer-Mickeleit RW. Das Elektroenzephalogramm nach gedeck- (arousal) dans le coma. Corrélations cortico-cárdio-respiratoires.
ten Kopfver-letzungen. Dtsch Med Wschr. 1953;1:480–484. Presse Méd. 1955;61:1231–1233.
20. Radermecker J. Das EEG bei gedeckten Hirnschäden und seine 44. Fischer C, Mutschler V. Traumatic brain injuries in adults: from
Beziehung zu den subjektiven Beschwerden. Med Wschr. 1964; coma to wakefulness. Neurophysiological data. Ann Readapt Med
106:1315–1322. Phys. 2002;45:448–455.
21. Courjon J, Scherzer E. Traumatic disorders. In: Remond A, Magnus 45. Courjon J, Naquet R, Baurand C, et al. Valeur diagnostique de
O, Courjon J. eds. Handbook of Electroencephalography and Clinical l’E.E.G. dans les suites immédiates des traumatismes crâniens. Rev.
Neurophysiology, vol. 14. Amsterdam: Elsevier; 1972:8–95. EEG Neurophysiol. 1971;1:133–150.
432 Part III ■ Clinical EEG: General

46. Chatrian GE. Electrographic and behavioral signs of sleep in com- 68. Hutchinson DO, Frith RW, Shaw NA, et al. A comparison between
atose states. In: Remond A, Harner R, Naquet R, eds. Handbook of electroencephalography and somatosensory evoked potentials for
Electroencephalography and Clinical Neurophysiology, vol. 12. outcome prediction following severe head injury. Electroence-
Amsterdam: Elsevier; 1975:63–77. phalograph Clin Neurophysiol. 1991;78:228–233.
47. Vigouroux RP. Between life and death. In: Remond A, Harner R, 69. Rae-Grant AD, Barbour PJ, Reed J. Development of a novel EEG
Naquet R, eds. Handbook of Electroencephalography and Clinical rating scale for head injury using dichotomous variables.
Neurophysiology, vol. 12. Amsterdam: Elsevier; 1975:95–98. Electroencephalogr Clin Neurophysiol. 1991;79:349–357.
48. Stockard JJ, Bickford RG, Aung MH. The electroencephalogram in 70. Chokroverty S. “Alpha-like” rhythms in electroencephalograms in
traumatic brain injury. In: Vinken, PJ, Bruyn, GW, eds. Handbook coma after cardiac arrest. Neurology (Minneapolis) 1975;25:655–663.
of Clinical Neurology, vol. 23. Amsterdam/Oxford: North-Holland; 71. Synek VM, Synek BJL. Theta pattern coma, a variant of alpha pat-
1975;217–367. tern coma. Clin Electroencephalogr. 1984;15:116–121.
49. Williams D, Denny-Brown D. Cerebral electrical changes in exper- 72. Horton EJ, Goldie WD, Baram TZ. Rhythmic coma in children.
imental concussion. Brain. 1941;64:223–238. J Child Neurol. 1990;5:242–247.
50. Denny-Brown D, Russel WR. Experimental cerebral concussion. 73. Chatrian GE, Lowell MD, White E, Jr., et al. EEG pattern resem-
Brain. 1941;64:93–164. bling wakefulness in unresponsive decerebrate state following
51. Ommaya AK, Gennarelli A. Cerebral concussion and traumatic traumatic brain stem infarct. Electroencephalogr Clin Neurophysiol.
unconsciousness. Brain. 1974;97:633–654. 1964;16:285–289.
52. Ommaya AK, Grupp RL, Jr, Naumann RA. Coup and contre-coup: 74. Jasper HH, Van Buren J. Interrelationships between cortex and
observations on the mechanics of visible brain injuries in the rhe- subcortical structures. Clinical electroencephalographic studies.
sus monkey. J Neurosurg. 1971;35:503–517. Electroencephalogr Clin Neurophysiol. 1953;4(suppl):168–202.
53. Ommaya AK, Corrao PG, Letcher F. Head injury in the chim- 75. Steudel WI, Krüger J, Grau H. Zur Alpha- und Spindel-Aktivitt bei
panzee: part I. Biodynamics of traumatic unconsciousness. J komatösen Patienten nach einer Schädel-Hirn-Verletzung unter
Neurosurg. 1973;39:152–166. besonderer Berücksichtigung der Computertomographie. Z. EEG-
54. Shaw NA. The neurophysiology of concussion. Prog Neurobiol. EMG. 1979;10:143–147.
2002;67:281–344. 76. Rumpl E. Elektro-neurologische Korrelationen in den frühen
55. Gloor P, Ball G, Schaul N. Brain lesions that produce delta waves in Phasen desposttraumatischen Komas. II. Das EEG im Übergang
the EEG. Neurology. 1977;27:326–333. zum, und im Vollbild des traumatischen apallischen Syndroms. Z.
56. Akert K, Koella WP, Hess R, Jr. Sleep produced by electrical stimu- EEG-EMG. 1980;11:43–50.
lation of the thalamus. Am J Physiol. 1952;168:260–267. 77. Goulon M, Pocidalo JJ, Christophe M, et al. Clinical, electroen-
57. Hunter J, Jasper HH. Effects of thalamic stimulation in unanaes- cephalographic and biological correlations in 34 cases of bron-
thetized animals. Electroencephalogr Clin Neurophysiol. 1949;1: chopneumonia with asphyxia. In: Gastaut H, Meyer JS. eds.
305–324. Cerebral Anoxia and the Electroencephalogram. Springfield, IL:
58. Jouvet M. Telencephalic and rhombencephalic sleep in the cat. In: Charles C Thomas; 1959:565–577.
Wolstenholme GEW, O’Connor M. eds. The Nature of Sleep. 78. Merill JP, Hampers CL. Uremia (part 1). Engl J Med. 1970;289:
Boston, MA: Little, Brown; 1961:188–208. 953–961.
59. Fischer CM. The neurological examination of the comatose 79. Wilson WP, Sieker HL. A study of the factors responsible for
patient. Acta Neurol Scand. 1969;36(suppl):45–56. changes in the electroencephalogram in chronic pulmonary insuf-
60. Hockaday JM, Potts F, Epstein E, et al. EEG changes in acute cere- ficiency. Electroencephalogr Clin Neurophysiol. 1958;10:89–96.
bral anoxia from cardiac or respiratory arrest. Electroencephalogr 80. Vespa PM, Miller C, McArthur D, et al. Nonconvulsive electro-
Clin Neurophysiol. 1965;18:575–586. graphic seizures after traumatic brain injury result in a delayed,
61. Kubicki S, Rieger H, Busse G. EEG in fatal and near fatal poisoning prolonged increase in intracranial pressure and metabolic crisis.
with soporific drugs. I. Typical EEG patterns. Clin Crit Care Med. 2007;35:2830–2836.
Electroencephalogr. 1970;1:5–13. 81. Grand W. The significance of post-traumatic status epilepticus in
62. Prior PF. The EEG in Acute Cerebral Anoxia. Amsterdam: Excerpta childhood. J Neurol Neurosurg Psychiatry. 1974;37:178–180.
Medica; 1973:43–46. 82. Kiloh LG, Osselton JW. Clinical Electroencephalography. London:
63. Gerstenbrand F, Rumpl E. Das prolongierte Mittelhirnsyndrom Butterworths; 1966;125.
traumatischer genese. In: Neumärker KJ. ed. Hirnstammläsionen. 83. Bauer G, Niedermeyer E. Acute convulsions. Clin Electroen
Leipzig: Hirzel; 1983:236–248. Cephalogr. 1979;10:127–144.
64. Bricolo, A. Neurosurgical exploration and neurological pathology 84. Jordan KG. Continuous EEG and evoked potential monitoring in
as means for investigating human sleep semiology and mechanism. the neuroscience intensive care unit. J Clin Neurophysiol. 1993;
In: Lairy GC, Salzarulo P. eds. Experimental Study of Human Sleep: 10:445–475.
Methodological Problems. Amsterdam: Elsevier; 1975:55–82. 85. Toyonaga K, Schlagenhauff RE, Smith BH. Periodic lateralized
65. Hess R. Sleep and sleep disturbances in the electroencephalogram. epileptiform discharges in subdural hematoma. Case-reports and
Prog Brain Res. 1965;18:127–139. review in literature. Clin Electroencephalogr. 1974;5:113–118.
66. Lücking GH. Sleep-like patterns and abnormal arousal reactions in 86. Chatrian GE, Shaw CM, Leffman H. The significance of periodic
brain stem lesions. Electroencephalogr Clin Neurophysiol. lateralized epileptiform discharges in the EEG: an electroen-
1970;28:214 (abst). cephalographic, clinical and pathologic study. Electroencephalogr.
67. Rumpl E. Elektro-neurologische Korrelationen in den frühen Phasen Clin Neurophysiol. 1964;17:177–193.
des posttraumatischen Komas. I. Das EEG in den verschiedenen 87. Markand ON, Daly DD. Pseudoperiodic lateralized paroxysmal
Phasen des akuten traumatischen sekundären. Mittelhirn und discharges in electroencephalogram. Neurology (Minneapolis).
Bulbärhirnsyndroms. Z. EEG-EMG. 1979;10:148–157. 1971;21:975–981.
Chapter 22 ■ EEG and Craniocerebral Trauma 433

88. Bickford RG, Butt HR. Hepatic coma: the electroencephalic pat- 109. Butenuth J, Kubicki S. Klinisch-elektroenzephalographische
tern. J Clin Invest. 1955;34:790–799. Schlafbeobachtungen im apallischen Syndrome. Z EEG-EMG.
89. Hall RC, Joffe JR. Hypomagnesemia: physical and psychiatric 1975;6:185–188.
symptoms. JAMA. 1973;244:1749–1751. 110. Passouant P, Cadilhac J, Delange M, et al. Différentes stades élec-
90. Jennet B, Bond M. Assessment of outcome after severe brain triques et organization en cycle des comas posttraumatiques.
damage. Lancet. 1975;1:480–484. Enrégistrement polygraphique de long durée. Rev Neurol.
91. Rumpl E, Prugger M, Bauer G, et al. Incidence and prognostic 1964;111:391(abst).
value of spindles in post-traumatic coma. Electroencephalogr Clin 111. Cadilhac J, El Kassabgui M, Passouant P. La réorganisation du
Neurophysiol. 1983;56:420–429. sommeil nocturne après les comas posttraumatiques. Rev Neurol.
92. Bergamasco B, Bergamini L, Bricolo A, et al. Studies on sleep dur- 1966;115:529(abst).
ing the apallic syndrome. In: Dalle Ore G, Gerstenbrand F, 112. Bergamasco B, Bergamini L, Doriguzzi T, et al. EEG sleep pat-
Lücking CH, et al., eds., The Apallic Syndrome. Berlin/Heidel- terns as a prognostic criterion in post-traumatic coma. Electro-
berg/New York: Springer; 1977;155–159. encephalogr Clin Neurophysiol. 1968:24:374–377.
93. Hansotia P, Gottschalk P, Green P, et al. Spindle coma: incidence, 113. Chase TN, Moretti L, Prensky AL. Clinical and electroencephalo-
clinicopathologic correlates, and prognostic value. Neurology graphic manifestations of vascular lesions of the pons. Neurology
(Minneapolis) 1981;31:83–87. (Minneapolis) 1968;18:357–368.
94. Hughes JR, Boshes B, Leestma J. Electro-clinical and pathologic 114. Ferguson JM, Benett DR. Sleep in a patient with pontine infarc-
correlations in comatose patients. Clin Electroencephalogr. tion. Electroencephalogr Clin Neurophysiol. 1974;36:210–211(abst).
1976;7:13–30. 115. Rumpl E, Prugger M, Bauer G. Zum prognostischen Wert elek-
95. Lorenzoni E. Das EEG im posttraumatischen Koma. Fortschr troenzephalographischer Schlafbeobachtungen im traumatisch
Neurol Psychiatr. 1975;43:155–191. bedingten apallischen syndrom. Neuropsychiatr Clin. 1984;3:
96. Vigouroux R, Naquet R, Baurand C, et al. Évolution électroradio- 219–232.
clinique de comas graves prolongés post-traumatiques. Rev 116. Biniek R, Ferbert A, Rimpel J, et al. The complete apallic syn-
Neurol. 1964;110:72–81. drome—a case report. Intensive Care Med. 1989;15:212–215.
97. Bricolo A, Turazzi S, Faccioli F, et al. Clinical application of com- 117. Ingvar DH, Brun A. Das komplette apallische syndrom. Arch
pressed spectral array in long-term EEG monitoring of comatose Psychiatr Nervenheilkd. 1972;215:219–239.
patients. Electroencephalogr Clin Neurophysiol. 1978;45:211–225. 118. Giacino JT, Ashwal S, Childs N, et al. The minimally conscious state:
98. Dusser A, Navelet Y, Devictor D, et al. Short- and long-term prog- definition and diagnostic criteria. Neurology. 2002;58:349–353.
nostic value of the electroencephalogram in children with severe 119. Kelly JP, Rosenberg JH. Diagnosis and management of concus-
head injury. Electroencephalogr Clin Neurophysiol. 1989;73:85–93. sion in sports. Neurology (Minneapolis) 1997;48:575–580.
99. Britt RH, Herrick MK, Hamilton RD. Traumatic locked-in syn- 120. Denny-Brown D. Brain trauma and concussion. Arch Neurol.
drome. Ann Neurol. 1977;1:590–592. 1961;5:1–3.
100. Miller JD. Barbiturates and raised intracranial pressure (editor- 121. Tönnis A. Beobachtungen an frischen gedeckten Hirnschädi-
ial). Ann Neurol. 1979;6:189–193. gungen. In: Rehwald E., ed. Das Hirntrauma. Stuttgart: Thieme;
101. Myers RR, Stockard JJ, Saidman LJ. Monitoring of cerebral per- 1956:772.
fusion during anesthesia by time-compressed Fourier analysis of 122. Verjaal A. Trauma capitis. In: van den Bergh R, Folkerts JF, eds.
the electroencephalogram. Stroke. 1977;8:331–337. Neurologie vor de Algemene Praktijk. Brussels: Agon-Elsevier;
102. Newlon PG, Greenberg RP, Enas GG, et al. Effects of therapeutic 1972:170–187.
pentobarbital coma on multimodality evoked potentials 123. Voller B, Benke T, Benedetto K, et al. Neuropsychological, MRT
recorded from severely head-injured patients. Neurosurgery. and EEG findings after very mild traumatic brain injury. Brain
1983;12:613–619. Inj. 1999;13:821–827.
103. Overgaard J, Hvid-Hansen O, Land AM, et al. Prognosis after 124. Yamamoto LG, Bart RT. Transient blindness following mild head
head injury based on early clinical examination. Lancet. trauma. Clin Pediatr. 1989;27:479–483.
1973;2:631–635. 125. Hess R. Significance of EEG signs for localization of cerebral
104. Avenarius HJ, Gerstenbrand F. The transition stage from mid- tumours. Electroencephalogr. Clin Neurophysiol. 1961;19(suppl):
brain syndrome to the traumatic apallic syndrome. In: Dalle Ore 75–110.
G, Gerstenbrand F, Lücking CH, et al., eds. The Apallic Syndrome. 126. Mifka P, Scherzer E. Über die Wertigkeit des EEG im Spätstadium
Berlin/Heidelberg/New York: Springer; 1977;22–25. der Gehirnverletzung. Wien Klin Wschr. 1962;74:573–576.
105. Hörtnagl H, Hammerle AF, Hackl JM, et al. The activity of the 127. Dumermuth G. Elektroencephalographie im Kindesalter. Stutt -
sympathetic nervous system in the course of severe head injury. gart: Thieme; 1965:262–266.
Intensive Care Med. 1980;6:169–177. 128. Lenard HG. EEG-Veränderungen bei frischen Schädeltraumen
106. Gerstenbrand F, Rumpl E, Prugger M. Die apallische im Kindesalter. Med Wschr. 1965;107:1820–1827.
Symptomatik. In: Wiek HH., ed. Neurotraumatologie, Derzeitige 129. Melin KA. Electroencephalography following head injuries in
Schwerpunkte, 8. Internationales Symposium Erlangen 1979. children. Acta Pediatr (Uppsala). 1949;75:152–174.
Stuttgart: Thieme; 1980:235–239. 130. Christian W. Klinische Elektroenzephalographie. Lehrbuch und
107. Gerstenbrand F. Das Traumatische Apallische Syndrome. Atlas. Stuttgart: Thieme; 1968:264–274.
Wien/New York: Springer; 1967. 131. Vigouroux RP, Naquet R, Gastaut H. Aspects particuliers de cer-
108. Dolce G, Kaemmerer E. Contribute anatomico-clinico ed elet- taines manifestations épileptiques précoces lors de traumatismes
troencephalografico all conoscenza della sindromi apallica. crânio-cérébraux survénuschez des sujects âgés. Rev Neurol.
Studio dell evoluzione dell EEG da sonne in 5 casi. Sist Nerv 1966;115:976–980.
1967;1:12–23. 132. Jennett WB. Early traumatic epilepsy. Lancet. 1969;1:1023–1025.
434 Part III ■ Clinical EEG: General

133. Courjon J, Revol M. Traumatismes crâniens et épilepsie. Cah Méd 153. Steudel WI, Krüger J. Using the spectral analysis of the EEG for
Lyon. 1966;42:1343–1350. prognosis of severe brain injuries in the first post-traumatic
134. Black K, Obayan A, Zafonte RD, et al. Occipital status epilepticus: week. Acta Neurochir. 1979;28(suppl):40–42.
an unusual case of posttraumatic blindness. Neuro Rehabili- 154. Pfurtscheller G, Schwartz G, List W. Long-lasting EEG reactions
tation. 1996;7:219–221. in comatose patients after repetitive stimulation. Electro-
135. Kazibutowska Z, Stelmach-Wawrzyczek M, Majchrzak R. Results encephalogr Clin Neurophysiol. 1986;64:402–410.
of prospective 24-hour EEG studies of patients after craniocere- 155. Klein HJ, Rath SA, Göppel F. The use of EEG spectral analysis
bral injuries. Neurol Neurochir Pol. 1992;26:304–310. after thiopental bolus in the prognostic evaluation of comatose
136. Wohlrab G, Schmitt B, Botshauser E. Benign focal epileptiform patients with brain injuries. Acta Neurochirurg. 1988;
discharges in children after severe head trauma: prognostic value 42(suppl):31–34.
and clinical course. Epilepsia. 1997;38:275–278. 156. Thatcher RW, Walker RA, Gerson I, et al. EEG discriminant
137. Gibbs FA, Gibbs, EL. Electroencephalographic study of head analysis of mild head trauma. Electroencephalogr Clin
injury in childhood. Clin Electroencephalogr. 1987;18:10–11. Neurophysiol. 1989;73:94–106.
138. Niedermeyer E, Knott J. Über die Bedeutung der 14 and 6/sec 157. Thatcher RW, Cantor DS, McAlaster R, et al. Comprehensive pre-
positiven Spitzen in EEG. Arch Psychiatr Nervenkr. 1961;202: dictions of outcome in closed head-injured patients. Ann NY
266–280. Acad Sci. 1991;620:82–101.
139. Gerstenbrand F. The symptomatology of the apallic syndrome. 158. Hooshmand H, Beckner E, Radfor F. Technical and clinical
In: Dalle Ore G, Gerstenbrand F, Lücking CH, et al., eds. The aspects of topographic brain mapping. Clin Electroencephalogr.
Apallic Syndrome. Berlin/Heidelberg/New York: Springer; 1989;20:235–247.
1977:14–21. 159. Jerrett SA, Corsak J. Clinical utility of topographic EEG brain
140. Caveness WF. Posttraumatic sequelae. In: Caveness WF, Walker mapping. Clin Electroencephalogr. 1988;19:134–143.
AE, eds. Head Injury, Conference Proceedings. Philadelphia/ 160. Busse EW, Silverman AJ. Electroencephalographic changes in
Toronto: Lippincott; 1966:209–219. professional boxers. JAMA. 1952;149:1522–1525.
141. Verjaal A, Van’t Hooft F. Commotio and contusio cerebri (cere- 161. Larsson LE, Melin KA, Nordström-Öhrberg BO, et al. Acute head
bral concussion). In: Vinken PJ, Bruyn GW, eds. Handbook of injuries in boxers. Acta Psychiatr Scand. 1954;95(suppl):1–42.
Clinical Neurology, vol. 23. Amsterdam/Oxford: North-Holland; 162. Pampus E, Grote W. Elektroencephalographische und klinische
1975:417–444. Befunde bei Boxern und ihre Bedeutung fr die Pathophysiologie
142. Jellinger K, Gerstenbrand F, Pateisky K. Die protrahierte form der der traumatischen Hirnschädigung. Arch Psychiatr Nervenkr.
posttraumatischen Enzephalopathie. Nervenarzt. 1963;34: 1956;194:152–178.
145–163. 163. Saunders RL, Harbaugh RE. The second impact in catastrophic
143. Pateisky K, Gerstenbrand F, Jellinger K. The EEG in the posttrau- contact sports head trauma. JAMA. 1984;252:538–539.
matic apallic syndrome. Electroencephalogr Clin Neurophysiol. 164. Roberts AH. Brain Damage in Boxers. London: Pitman Medical
1963;15:713(abst). Scientific Publishing; 1969.
144. Goetze W. Das Hirnstrombild bei offenen Hirnverletzungen. 165. Haglund Y, Persson HE. Does Swedish amateur boxing lead to
Mschr Unfallheilk. 1953;56:297–305. chronic brain damage? A retrospective clinical neurophysiologi-
145. Courjon J. La place de l’électroencéphalographie en traumatolo- cal study. Acta Neurol Scand. 1990;82:353–360.
gie crânienne. Cah Méd Lyon. 1962;38:315–317. 166. Tysvaer AT, Storli OV, Bachen NI. Soccer injuries to the brain. A
146. Kugler J. Elektroenzephalographie in Klinik und Praxis 2. Aufl., neurologic and electroencephalographic study of former players.
Stuttgart: Thieme; 1966:203. Acta Neurol Scand. 1989;80:151–156.
147. Torres F, Shapiro SK. Electroencephalograms in whiplash injury. 167. Tysvaer AT. Head and neck injuries in soccer. Impact of minor
Arch Neurol. 1961;5:28–35. trauma. Sports Med. 1992;14:200–213.
148. Kuhlendahl K. Die neurologischen syndrome bei der 168. Martland HS. Punch drunk. JAMA. 1928;91:1103–1107.
Überstreckungsverletzung der Halswirbelsäule und dem soge- 169. Millspaugh JA. Dementia puglistica (punch drunk). US Nav Med
nannten Schleudertrauma. M Med Wschr. 1964;106:1025–1030. Bull. 1937;35:297–303.
149. Mifka P, Scherzer E. Zur Pathogenese zerebraler Symptomatik bei 170. Critchley M. Medical aspects of boxing, particularly from a neu-
Verletzungen der Halswirbelsäule. Münch Med Wschr. 1962;10: rological standpoint. Br Med J. 1957;1:357–362.
1686–1690. 171. Mawdsley C, Ferguson FR. Neurological disease in boxers. Lancet.
150. Jacome DE. EEG in whiplash: a reappraisal. Clin Electroence- 1963;2:795–801.
phalogr. 1987;18:41–45. 172. Johnson J. The EEG in the traumatic encephalopathy of boxers.
151. Cant BR, Shaw NA. Monitoring by compressed spectral array in Psychiatr Clin. 1969;2:204–211.
prolonged coma. Neurology. 1984;34:35–39. 173. Forster JB, Leiguarda R, Tilley PJB. Brain damage in national
152. Sironi VA, Ravagnati L, Signorini G, et al. Diagnostic and prog- hunt jockeys. Lancet. 1976;1:981–987.
nostic value of EEG compressed spectral analysis in posttraumatic
coma. In: Vilani R, Papo J, Giovanelli M, et al. eds. Advances in
Neurotraumatology. Amsterdam: Excerpta Medica; 1983:328–330.
CHAPTER

Anoxia, Coma, and Brain Death


PETER W. KAPLAN AND GERHARD BAUER 23
CLINICAL DEFINITIONS OF IMPAIRED is often diagnosed after about 3 months in nontraumatic brain
RESPONSIVENESS: FROM LOCKED-IN injury, or a year after brain trauma. Different reports of this
syndrome have used names such as apallic syndrome (4), com-
STATES TO COMA
plete and incomplete apallic syndrome, prolonged coma (5),
Consciousness and persistent vegetative state (PVS) (6). Absence of any clini-
Consciousness can be described as an awareness of self and the cal sign of cortical function combined with alternating periods
environment. The two primary components of consciousness of sleep and wakefulness is the essential of this condition. The
are awareness and arousal with the determination of conscious- eyes open periodically without evidence of appreciation of the
ness being made by the examination of the patient’s response to environment. More recently, multimodal imaging has shed
stimuli. The examination of brainstem reflexes and responses to light on brain activity in the VS (1,3,7). Pathologically, no com-
command is helpful in localizing the causes of coma to higher mon pattern of lesion can be defined. Prognosis, EEG signs, and
or lower CNS structures. appropriate management rest on the strict or broadened defini-
A further component of awareness depends on cortical tion of this condition (8–11). Terminological and prognostic
integrity and subcortical connections. When these are dis- controversies have been cleared by the Multi-Society Task Force
rupted, a patient may have a decreased consciousness to the on PVS (12). Questions remain about the boundaries to coma
point of being in a minimally conscious state or a vegetative state, and VS (12).
but less frequently is in actual coma (1).
Locked-in Syndrome (LIS)
Coma An LIS consists of almost total paralysis of limb and head
Coma is a state of unconsciousness without wakefulness, char- movements so that the patient cannot move or speak, but
acterized by the lack of arousal from internal or external stim- remains fully conscious (13,14). It is due to the interruption of
uli. The eyes are usually closed, and there is no awareness of self corticospinal and corticobulbar pathways, typically in the basis
or surroundings. Brainstem reflexes may be retained. Coma is pontis or midbrain (15–20) or from bilateral lesions in the
usually a transient condition that evolves to either wakefulness internal capsules (21), or less frequently from total peripheral
or death over days to weeks. paralysis (ALS, polyradiculopathy). This pathology disconnects
the brain from the ability to converse (interruption of efferent
Minimally Conscious States (MCS) pathways). The only retained movements are of the eyelids and
Relatively recently, a minimally conscious state (MCS) has of vertical eye movements. The patient can communicate with
been declared a separate clinical condition (2). A MCS is that a pattern of eye movements that the patient and observer have
of a severely altered consciousness. Behavioral manifestations established to allow discourse. In this way, the patient demon-
of awareness are minimal, but present. There may be fluctuat- strates awareness and hence consciousness. EEG has shown dif-
ing attention, as well as purposeful visual tracking and com- ferent patterns, but the most typical is a posterior, often reactive
munication, but all these elements should be present. fMRI has alpha rhythm (22,23).
demonstrated some patterns of brain activity in minimally
conscious patients that are also seen in normal people (3). Akinetic Mutism
These patterns are thought to represent conscious cognitive The term akinetic mutism was coined by Cairns and colleagues
processing. (1941) to describe a state of mutism, or minimal speech, and
hence communication (24). There are a number of anatomical
Vegetative States (VS) lesions that can cause this syndrome, and it is a term less used
In vegetative states (VS), the patient is awake. However, there at present.
is no evidence of awareness of self or of the environment,
although sleep–wake cycling occurs. There is also no evidence Coma Vigile
(during wakefulness) of cognitive processing. A VS is persistent A term also in declining use, it was used to refer to several of the
if it has lasted at least a month, but “persistent” does not neces- syndromes mentioned above including LIS, akinetic mutism
sarily imply irreversibility. A permanent VS is irreversible, and (6), or the apallic syndrome (25).

435
436 Part III ■ Clinical EEG: General

Sleep edema (41–43). The progressive impairment of consciousness


Although sleep represents a transient absence of consciousness, mirrors, to some extent, that of anesthesia induction.
a patient can be aroused to conscious wakefulness. In light A number of mineral and electrolyte disturbances can all
coma, it can be difficult to differentiate pathologic hypersom- contribute to encephalopathy, and in the extreme, to coma.
nolence (e.g., from drugs, alcohol, or sleep deprivation) from These include hyper- and hyponatremia, hyper- and hypocal-
physiologic sleepiness. cemia, hyper- and hypoglycemia, and uremia. Contributing to
these electrolyte disturbances, or constituting disorders in
themselves, are metabolic and endocrine disorders, and organ
THE ANATOMICAL AND PHYSIOLOGIC failures such as renal failure and dialysis effects, hypothy-
SUBSTRATES OF COMA roidism, hypocortisolism, and porphyria. There are rarely if any
pathognomonic EEG patterns with these encephalopathies and
Consciousness is maintained by the integration of ascending coma (44,45). The patterns reflect largely the degree of suppres-
arousal inputs via the ascending reticular activating system sion (and level of coma), the presence of associated epilepti-
through the thalami to both cerebral hemispheres. In animals form disturbances, and occasionally focal effects. One major
and humans, these ascending pathways pass through the pons exception is hepatic failure that may frequently be reflected on
and midbrain tegmentum (26). Structural or functional EEG by triphasic waves (TWs) and progressive disorganization
impairment or interruption of these pathways in the brainstem of the EEG as coma deepens even though such waves can be
or in both cerebral hemispheres leads to decreased conscious- seen in other conditions (see later on in this chapter).
ness and coma (27,28). Drug intoxications in coma can produce patterns that in part
reflect their cortical effects. Barbiturates progressively suppress
Structural Causes of Coma the EEG; benzodiazepines suppress voltage, increase the power of
The signal process needed to induce coma is compromise of frequencies in the beta range, and eventually slow the back-
brainstem function. This typically happens from compression ground. Lithium may produce epileptiform discharges, slowing,
or ischemia. Compression can arise from unilateral cerebral and TWs (46). Some stimulant drugs increase, voltage and fre-
mass lesions exerting traction and distortion of the brainstem quencies, but often do not produce coma. For more detailed
structures or compromise of circulation. Although bilateral pathophysiology and EEG findings on toxic/metabolic and organ
cerebral hemisphere dysfunction without brainstem involve- failure impairment of CNS function, please turn to Chapter 42.
ment can suppress cortical activity, coma per se rarely occurs.
Further, a markedly raised ICP in itself does not induce coma, Infection and Coma
except by brainstem compromise. Pathologic examination and Infection of the CNS, of individual body organs, as well as sep-
radiologic studies have demonstrated the effects of uncal and ticemia can all induce confusion and encephalopathy, but only
central herniation on diencephalic structures that mediate direct CNS infection readily causes coma (47–49). A large num-
arousal (29), but more current imaging techniques (CT and ber of microbes may directly affect the brain and/or the
MRI) give greatest credence to the transverse or horizontal type meninges, along with the increasing emergence of patients with
of herniation compromise (30,31), while evidence of down- the acquired immunodeficiency syndrome (AIDS). In addition,
ward, transtentorial herniation can be obtained with MRI there are conditions that may simulate infections. Conditions
(32–34). Brainstem compromise in the latter injury arises more such as neoplasms, autoimmune conditions, and slow viruses
from anoxia than direct pressure. are dealt elsewhere in this text. The pathophysiology of infection
More direct destruction by compression, trauma, or and coma has not been clearly elucidated. Mechanisms impli-
ischemia of the paramedian pontomesencephalic reticular for- cated include a direct release of toxins and their effect on CNS
mation can occur at the level of the pons and midbrain (35,36), structures, the involvement of microvasculature and larger ves-
although a direct pontine effect by trauma has been questioned sels, cerebral edema, a focal or generalized increase in intracra-
(37). Other authors note rare cases of traumatic brainstem nial pressure, and changes in cerebral blood flow (CBF), which
coma supported by MRI (38). Diffuse axonal injuries from tor- may all play a part. Once again, EEG changes in these various
sional or shearing stresses can markedly adversely affect prog- conditions reflect the focality, epileptogenicity, level of con-
nosis and be more difficult to evidence in imaging studies (39). sciousness, and the mechanism of CNS compromise produced
by the respective infectious process as it induces coma.
Toxic/ Metabolic Causes of Coma
Despite the prevalence and longstanding recognition of these Seizures, Epileptiform Activity, Status
conditions as causes of coma, there is a relative paucity of infor- Epilepticus, and Coma
mation on their mechanism. It is believed that synaptic transmis- Epileptic activity when generalized usually causes coma.
sion is variably impaired, with neuronal energy failure occurring Seizures of themselves and in the form of ongoing activity
late in the process. There is early impairment of complex interac- (status epilepticus) generally may cause either transient or a
tions among the cortical regions and polysynaptic pathways pro- more lasting loss of consciousness hours. Some conditions with
ducing an initial impairment of higher cortical functions. In this partial brain seizure activity may impair vigilance, but do not in
way, judgment, memory, attention, and concentration are early all cases cause coma. Coma from many causes may additionally
heralds of impending encephalopathy and coma (40). Coma is reveal epileptiform activity and seizure activity as an “epiphe-
believed to be produced by the induction of brainstem or brain nomenon.” In this sense, the underlying coma etiology is not
Chapter 23 ■ Anoxia, Coma, and Brain Death 437

that of seizure activity, but rather the reflection of cortical irri- difficult in adults to differentiate light coma from drowsiness.
tability or insult. Please see also Chapters 25, 26, and 28 to 30. Occasionally, higher frequencies can be seen, for example, in the
beta range, particularly with benzodiazepines and barbiturates.
Anoxia and Coma High doses of CNS-suppressant drugs can decrease EEG voltage.
Because anoxia is such a common cause of coma, as well as being Drug effects aside, low-voltage or flatline tracings after anoxia
one of its most morbid etiologies, more attention will be paid to predict an abysmal prognosis for return to consciousness.
this entity in this chapter. The brain at rest consumes 20% of the
total body oxygen consumption, even though it represents only Intermittent Delta Rhythms
2% by weight (50). Consciousness and normal background EEG Frontal intermittent rhythmic delta activity (FIRDA) (77) can be
patterns are impaired if the brain oxygen tension falls below 15 to seen in mild obtundation, but rarely occurs in coma. It is believed
20 mm Hg (51). An interruption in oxygen supply results in cell to arise most frequently from diffuse cerebral dysfunction (78),
dysfunction, and ultimately death, and can be induced by a but was initially described in deep midline dysfunctions in sub-
decrease in CBF, hypoxia or anoxia, a low oxygen concentration cortical regions (79), deep frontal (80), and with early herniation.
in respired air (hypoxic hypoxia). When ischemia reduces CBF When FIRDA exists with slow background activity, a more dif-
below 15 mL/min/100 g of tissue for several minutes, there may fuse cerebral dysfunction, including toxic, metabolic, or infec-
be subsequent cell death and degeneration over the ensuing days, tious etiologies, may be suspected (81,82). (see Fig. 23.1).
worsened by injury produced by resumed blood flow or reperfu-
sion (52–54). The postischemic period before cell death may rep- Prolonged Bursts of Delta Waves and the
resent a therapeutic window of opportunity. Causes of global Reactivity of EEG
ischemia include cardiorespiratory arrest (CRA), circulatory col- Some patients with head injury have paroxysms of potentiated
lapse, strangulation, drowning, suffocation, muscle paralysis, and slow activity, lasting up to minutes (83–86). These bursts may
toxins such as carbon monoxide. The less frequent hypoxia in occur spontaneously or after stimulation, particularly as the
pure form is rare as cardiac dysrrythmias usually also occur, as patient tries to communicate.
may systemic hypotension—both producing superadded In several of the patterns described above, spontaneous EEG
ischemic damage (55). Clinical signs of anoxic–ischemic brain variability or change in EEG patterns after stimulation indicates
injury largely follow the rate and severity of the deficiency. a more favorable prognosis. Conversely, the loss of variability or
Unconsciousness with cardiac arrest occurs in about 8 seconds in reactivity to stimuli can forebode deepening coma.
the upright position, and about 15 seconds supine, followed by
tonic posturing (56). When anoxic coma is prolonged, there may Epileptiform Morphologies
be persisting deficits, with permanent cognitive decline, rigidity, Epileptiform morphologies can be seen in many causes of
ataxia and spasticity, amnesia, visual dysfunction, movement dis- coma, even without apparent clinical or electrographic seizure
orders, and in some cases persistent coma or a VS (57). Epileptic activity. Continuous, unilateral epileptiform discharges with
phenomena include myoclonus, myoclonic status, or stimulus- little variability suggest pseudoperiodic lateralized epilepti-
sensitive myoclonus (15,58–63). It is important to distinguish form discharges (PLEDs), an indication of cortical irritability,
between the morbid postanoxic myoclonus in coma from the believed by most authors to fall short of actual seizures.
entity in which the patient returns to consciousness, but with Frequent causes are structural abnormalities (e.g., strokes,
stimulus-induced myoclonus following briefer anoxia/hypoxia trauma) with a recent seizure. Such patterns are frequently seen
(64,65). When the hypoxia is less marked, survivors are able to after seizure resolution in these settings, (87–90). When the dis-
speak, but may have some degree of cognitive deficit. CT of the charges are more generalized and synchronous, usually with a
head may be normal in the early hours of the day, but will even- relative paucity of intervening background activity, the term
tually reveal edema, sulcal effacement, basal ganglia lucency, and generalized periodic epileptiform discharges (GPEDs) has been
watershed changes (66,67). With MRI, there is better resolution used (91). This pattern usually reflects a more severe, diffuse
of abnormalities in the globus pallidus, putamen, caudate nuclei, cerebral insult such as is seen after anoxia (92), CNS infections,
hippocampus, cerebellum, brainstem, and subcortical white mat- or less frequently in the end-stages of status, and correspond-
ter (68–73). Magnetic resonance spectroscopy (MRS), diffusion- ingly indicates a worse prognosis. Generalized discharges can be
weighted MRI, and positron emission tomography (PET) are all accompanied by myoclonic movements, or conversely with lit-
more sensitive for hypoxic insults, and at an earlier point. With tle evident motor manifestations (15,92,93). Bilateral inde-
PET, De Volder et al. (1990) showed hypometabolism that corre- pendent discharges (BIPLEDs) probably lie in between with
lated with both location and severity of hypoxia. PET studies of regards to both severity of etiology and prognostic significance
patients in a VS have uncovered evidence of irreversible damage (88). There is ongoing controversy regarding where on the
to supratentorial cortical zones (74,75). ictal–interictal continuum these periodic discharge patterns lie,
and hence the need (or not) of more intensive seizure suppres-
EEG IN COMA sion with anesthetic agents or anesthetic coma. Evidence that
PLEDs represent a form of nonconvulsive status epilepticus
EEG patterns in coma may correlate with the depth of coma (94) arises from PET studies revealing increased metabolism or
(76). As consciousness decreases, there is a slowing of the preva- blood flow (95). In most cases, parenteral AEDs fail to lastingly
lent EEG frequencies from alpha toward delta. When there are suppress PLEDs or improve the patient’s level of consciousness.
excess theta frequencies in the posterior alpha rhythm, it may be See Figures 23.2 and 23.3.
438 Part III ■ Clinical EEG: General

Figure 23.1 This EEG shows


medium- to high-voltage theta and
a predominance of arrhythmic delta
activity in an unresponsive patient
with multiple organ failure includ-
ing renal and respiratory failure.

Triphasic Waves (TWs) TWs are seen with hepatic coma, after hypoxia (99–101), and fol-
Although this pattern was initially attributed to patients with lowing toxic exposure (see Fig. 23.6) (2,6,96,102,103). A number
hepatic failure and subsequently with raised serum ammonia, of other metabolic problems have been reported with TWs
later case series found this pattern with several different toxic or (78,104). Structural lesions have occasionally been reported with
metabolic problems, including renal failure (96). Some authors TWs, including brainstem strokes and subdural hematomas
have sought to distinguish a “typical” from an “atypical” form (105). Infection (106) and malignancy (cerebral carcinomatosis)
(Figs. 23.4 and 23.5) (97,98). Typical TW pattern consist of may also induce this pattern (107). Degenerative and spongi-
medium- to high-voltage three-phased bilateral, symmetric form encephalopathies may also produce TWs in the conscious
waveforms in runs, with a discharge frequency of 1.5 to 2.5 Hz. patient (108). An important differential diagnostic consideration
Typical TWs possess an anterior–posterior phase lag, which often is the continuously occurring sharp- and slow-wave runs seen
is not seen when using transverse or referential electrode arrays. with atypical absence status in Lennox–Gastaut syndrome (109).

Figure 23.2. This EEG sample


shows pseudoperiodic lateralized
epileptiform discharges (PLEDS) at
about 1 Hz over the right hemi-
sphere in an unresponsive patient.
Chapter 23 ■ Anoxia, Coma, and Brain Death 439

Figure 23.3 This EEG shows gene -


ralized pseudoperiodic epileptiform
discharges (GPEDs) in an anoxic
patient.

Burst-Suppression Patterns from less than a second to upwards of 10 seconds, but are char-
This pattern is largely associated with deeper levels of coma, acterized as burst-suppression patterns by the appearance of
and prominently occurs with anoxic insults after cardiac arrest. marked diffuse bilateral suppression of brain EEG activity
In this setting it carries an abysmal prognosis. Other more between these bursts. The periods of suppression again may
benign causes are in the setting of anesthetic use. In the thera- vary from about 1 second to periods exceeding 10 seconds.
peutic suppression of status epilepticus, propofol and other There may be a variable amount of background activity during
agents typically produce a burst-suppression pattern on the way these suppression periods (110,111). The bursts typically repeat
to producing more extensive, global cerebral suppression. quasi-periodically, appear diffusely and bilaterally, and exist
Overdoses or therapeutic use of barbiturates can also induce generally throughout the recording (112). They may be associ-
reversible patterns of burst-suppression. The pattern typically ated with single or salvos of myoclonic face, arm, chest, or leg
consists of high-voltage bursts of sharply contoured morpholo- jerks (15,93,113,114). In many patients, the jerks can be
gies, and polyspikes and spikes superimposed or associated induced by stimulating the patient by touch, suctioning, or loud
with underlying slow-wave activity. These paroxysms may last sounds. See Figure 23.7.

Figure 23.4 This EEG shows a pattern along the continuum of interictal to ictal, generalized periodic discharges (GPEDs)
in a patient who has had a prolonged cardiorespiratory arrest, but before therapeutic hypothermia.
440 Part III ■ Clinical EEG: General

Figure 23.5 This EEG shows the effects of lithium toxicity. There are runs of triphasic waves and some sharper, epilepti-
form morphologies seen diffusely, bilaterally. The background is slow. The patient was obtunded during the recording, but
recovered over several weeks.

Alpha and Theta Patterns in Coma Sleep-Like Patterns in Coma


Alpha frequency patterns may be recorded in some deep comas. Some coma EEG patterns contain spindles, slow activity, and
Originally described with brainstem dysfunction (strokes, vertex waves, resembling stage II sleep (81,131). They are usu-
tumors, trauma) (115–119), they were also recorded after ally seen after head trauma or structural lesions of the brain-
marked anoxia (119–124). Differentiating alpha coma from nor- stem (126,132,133), but may be seen with viral encephalitis
mal, physiologic waking alpha are its diffuse and indeed frontal (134), and benzodiazepines (135). Such patterns may evolve
predilection, general lack of reactivity to noxious stimulation (or to waking patterns, or diffuse slowing, and are thought to
at least a failure to induce a return to discernible clinical con- represent functional impairment of arousal mechanisms on
sciousness or wakefulness), and its steady presence. Alpha coma thalamocortical circuits responsible for sleep patterns. With
must be distinguished from the spindle-like patterns that can deepening coma, there is less variability and cycling, and
also appear in unresponsive states (125,126). Drug toxicity may the appearance of more monomorphic slow activity (see Fig.
cause faster rhythms in the 10- to 18-Hz range (123). In the 23.9) (136).
unresponsiveness of the LIS due to de-efferentation, the awake,
but paralyzed patient will evince waking alpha frequencies, typ- Low -Voltage or Diffusely Suppressed Patterns
ically reactive to eye-opening and closure. Alpha coma may fol- Marked voltage suppression can precede electrocerebral inac-
low burst-suppression pattern (111,125), heralding death. Some tivity (typically after anoxia), or occur (reversibly) with anes-
studies including unresponsive patients with alpha frequencies thetic suppression of cortical activity. It is usually defined as the
have found that prognosis may vary according to the etiology of totality of EEG-recorded brain activity that remains below
coma, and the presence of a degree of reactivity. Unreactive pat- 20 V. Low-voltage records can occur in conscious patients
terns and anoxia, or brainstem infarction carried the worst prog- after head trauma, alcoholism, and hydrocephalus, and are not
nosis (see Fig. 23.8) (115–130). to be confused with the morbid pattern seen in coma.
Chapter 23 ■ Anoxia, Coma, and Brain Death 441

Figure 23.6 The EEG shows disorganized activity, with no dominant alpha background activity. There are high-voltage
triphasic waves, some exhibiting an anteroposterior lag. This patient had baclofen toxicity.

EEG AND THE ETIOLOGY OF COMA are suggestive of a metabolic disorder (see Fig. 23.11), especially
of hepatic origin (46,96,102,103,142). The diagnostic signifi-
The EEG is not specific for etiology of coma, but can reflect cance of TWs may have been overstressed, but it is still impor-
some types of underlying disease. tant when this pattern is encountered in an undiagnosed,
insidiously developing coma. With hepatic coma, spikes may
Infectious and Inflammatory Processes also be encountered. Uremia is frequently complicated by gener-
With massive cerebral infection, there may be prominent, diffuse, alized tonic–clonic seizures and generalized polyspikes associ-
often rhythmic slowing, with few faster frequencies in the alpha ated with diffuse slow activity. Photomyoclonic responses are
and theta ranges. With the late, immune disorder of measles- not infrequent. Diffuse slowing may become exaggerated during
associated subacute sclerosing panencephalitis (SSPE), there may or after dialysis with or without clinical signs of a disequilibrium
be extremely high-voltage complexes reaching 300 V, lasting 1 syndrome. Paroxysmal bursts of slow waves and slowing of the
to 2 seconds, with a periodicity exceeding 5 to 10 seconds (137) basic rhythm were seen in all cases of the dialysis dementia syn-
or Jakob–Creutzfeldt disease (138,139). These patterns can also drome (143,144). Modern advances in dialysis techniques made
exist in other diffuse encephalitides. Periodic complexes are typ- this complication an exceptional event.
ically lateralized in the encephalitis of herpes simplex (140,141).
Epileptic Conditions
EEG in Metabolic Coma Epileptic conditions lead to prolonged coma in convulsive
The enormous complexity of pathophysiologic mechanisms in status epilepticus, postictal states with lingering subclinical
metabolic coma stands in contrast to the lack of specificity of paroxysmal activities, typical and atypical absence status
accompanying EEG signs. Diffuse slowing is always seen in (145,147,148), and other types of nonconclusive status epilep-
metabolic coma (see Fig. 23.10) (44,45). In cases of dispropor- ticus (145–148). In most instances, the EEG shows prominent
tionate degrees of coma and slowing, a metabolic cause is seizure activity and is instrumental in the establishment of
unlikely (78). Focal slowing does not exclude the diagnosis. TWs the correct diagnosis, although an EEG without spikes does not
442 Part III ■ Clinical EEG: General

Figure 23.7 This EEG shows a burst-suppression pattern in a patient who had anoxia and cardiac arrest. There are bilate -
ral, generalized polyspike–slow-wave complexes. The patient died without regaining consciousness.

categorically exclude the epileptic nature of the altered state of EEG in Relation to the Depth of Coma
consciousness. On the other hand, many comatose states are The EEG as a functional test should be closely related to the
complicated by interspersed epileptic seizures. Coma with acute degree of impaired cerebral function. There are several reasons
convulsions is accompanied by a markedly abnormal EEG, why this relation is far from being perfect. The main reason is
which in most instances is characterized by seizure discharges found in local brainstem coma, which leaves the cortex and the
(see Fig. 23.12) (15). Occasionally, seizures on EEG take the EEG comparatively unaffected.
form of rhythmic theta and delta activity (see Fig. 23.13).

Figure 23.8 This composite of four 10-second segments of EEG shows a diffuse alpha frequency pattern, present also
anteriorly. After two arousal efforts, the EEG continues to show alpha frequencies. This pattern constitutes alpha coma.
Chapter 23 ■ Anoxia, Coma, and Brain Death 443

Figure 23.9 This EEG shows sleep


spindle patterns in an unresponsive
patient, consistent with a spindle
coma pattern. This pattern carries a
good prognosis for return to con-
sciousness, if not due to anoxia.

Attempts to classify coma into clinical stages are numerous. rostral–caudal deterioration can be supported by EEG evi-
The most logical coma grading is based on an orderly sequence dence in the following manner:
of neurologic symptoms corresponding to the levels of the
1. The degree and generalization of slowing is related to the
neuraxis in cases of transtentorial herniations. Regardless of
level of unresponsiveness (78,149,150). The most important
the etiology, the following information about the grade of

Figure 23.10 This EEG shows sinusoidal diffuse theta activity in a comatose patient, from hypoglycemia.
444 Part III ■ Clinical EEG: General

Figure 23.11 There are runs of


triphasic waves seen synchronously,
bilaterally in this patient with hyper-
ammonemia.

exception to this rule is prolonged bursts of delta waves They might be mediated by the lower brainstem or even by
secondary to exogenous stimuli. Furthermore, in children the spinal cord and can be present with cortical death and
the degree of slowing is frequently disproportionate to the electrocerebral silence (ECS).
clinical state.
EEG activity seen during sleep becomes progressively scarcer
2. The effect of stimulation yields good information about the
and finally disappears with deepening coma (132,152–154).
depth of coma. As one proceeds down the scale of coma, the
Patterns highly suggestive of a late midbrain or initial bulbar
blocking type of response is replaced by the alerting type.
brain syndrome are (i) progressive voltage depression
Finally, the EEG becomes unreactive even to repeated stim-
(83,84,151); (ii) extreme slowing with extinction of superim-
ulation (83,84,151). Motor responses without changes of
posed fast activities (84); (iii) intermittent isoelectric periods;
EEG should not be regarded as a sign of cortical functions.

Figure 23.12 The high-voltage


bursts of generalized polyspike–slow-
wave activity seen synchronously,
bilaterally in this lethargic patient
with an idiopathic generalized
epilepsy indicate generalized non-
convulsive status epilepticus. Note
that in contrast to some burst-
suppression patterns after anoxia,
there is background activity in the
form of spindles, alpha, and theta
activity.
Chapter 23 ■ Anoxia, Coma, and Brain Death 445

Figure 23.13 This EEG shows rhythmic theta and delta waves, diffusely, but with slight frequency shift over time from
side-to-side. The patient was in coma with limbic encephalitis.

(iv) periodic spiking or burst-suppression activity (15,84,101); EEG in Anoxic Coma


and (v) monorhythmic unreactive alpha and theta frequencies Patients in postanoxic coma may exhibit a number of EEG pat-
(119,155) and spindle patterns (132). terns including TWs, continuous epileptiform discharges in
“electrographic status in coma,” burst-suppression patterns,
and monorhythmic alpha, theta, or mixed alpha–theta activity.
EEG ABNORMALITIES IN ANOXIA The patient typically is deeply comatose, and may have
AND ANOXIC COMA myoclonic jerks and an EEG reflective of bursts of epileptiform
activity (status myoclonicus) (59,61,157,158).
Basic Mechanisms of EEG Changes
In prolonged anoxia, the EEG patterns are similar to those
The EEG reflects decreases in cerebral metabolism due to seen in other causes of coma. There may be diffuse slowing (see
anoxia. As pO2 falls, there is a fall in membrane potential pro- Fig. 23.14) (100,101). FIRDA, a pattern of frontal delta waves,
gressing to a cessation of electrical activity. EEG voltage declines can also be seen, but is nonspecific, occurring in deep midline
to flatline (51). An increase in glutamate is associated with cell dysfunction as well as in other diffuse cerebral dysfunctions,
death, initially affecting the forebrain, followed by brainstem particularly in the elderly (82) (Fig. 23.15). Another pattern in
structures (156). postcardiac arrest coma is the appearance of sharp activity
with some morphologies assuming a triphasic appearance
EEG Changes during Arrest of Cerebral (99,101), PLEDs (Fig. 23.2), or bilaterally independent peri-
Circulation and w ith Syncope odic discharges (BIPLEDs) (88). With prolonged, severe
A progression of abnormalities follows the cerebral ischemia hypoxia, there may be intermittent suppression with bursts of
that arises from hypotension and cardiac arrest. In the first sev- spikes. A frank burst-suppression EEG pattern (Figs. 23.16
eral seconds, little change is seen, but after about 10 seconds, and 23.17) indicates severe prolonged diffuse cortical and
there is higher voltage slow activity, followed by flattening as the subcortical insults (15). Similarly, there is variability and evo-
period of ischemia increases. Recovery may occur in the reverse lution among patterns of burst-suppression, and alpha–theta
order. coma patterns overtime. As some have noted, there is often an
446 Part III ■ Clinical EEG: General

Figure 23.14 This EEG is from a patient several days after cardiorespiratory arrest. It shows generalized, sinusoidal 4- to
5-Hz activity, and carries a moderate prognosis for good outcome. The patient awoke with significant cognitive deficits.

inconstant association among clinical signs and paroxysmal anoxic comas induce alpha and theta frequency patterns
EEG activity. With spike or burst activity, there may be sponta- (100,119,127,155,109–161). Both can occur in the same record
neous or stimulus-induced eye-opening, and facial and limb (130) or appear in sequence. Reactivity signals a better progno-
myoclonus, but similar activities can occur without an EEG sis, although overall the mortality of these patterns is 80% to
correlate (15,159). As coma progresses, posturing can regress, 90% (127). An irreversible depolarization of membrane poten-
suggesting further involvement of brainstem structures. Some tial is reflected as a flatline EEG pattern.

Figure 23.15 This EEG panel shows a


burst of frontal intermittent, rhythmic
delta activity.
Chapter 23 ■ Anoxia, Coma, and Brain Death 447

Figure 23.16 This EEG panel


shows polyspike bursts on a sup-
pressed background in a patient
after cardiac arrest and anoxia. The
patient was comatose with
myoclonic face and limb move-
ments. The patient died without
regaining consciousness.

PROGNOSIS IN COMA USING EEG states. In these patients, neurologic and EEG examinations may be
misleading, and valuable monitoring seems to be confined to
Electrophysiology is optimally used in guiding early diagnostic evoked potentials (163–165). The barbiturate treatment of coma-
investigation, and in excluding other differential considera- tose states, however, has never been generally accepted (166,167),
tions including severe encephalopathy, NCSE, or psychogenic and its use has declined progressively (168,169).
issues. In general, the prognostic significance of EEG patterns par-
The EEG is certainly of great prognostic value in comatose allels their correlations to the grade of rostral–caudal deteriora-
states (162). Its significance is limited by the fact that the outcome tion. Prognostically favorable signs are reactions to exogenous
is often not determined by the brain disturbance itself but by the stimuli (83,170–172) and normal-looking sleep potentials or sleep
underlying metabolic or cardiovascular problems. Furthermore, cycles (83,131,152,173–175). Spindle coma indicates relative
prognostic criteria do not apply if anesthetics or other CNS- integrity of the cerebral hemispheres (126). Its prognostic signif-
depressant drugs have been used. This fact has become especially icance depends on the underlying etiology (176). Best outcomes
important with the widespread use of sedative drugs in comatose occurred when spindle coma was due to drugs, encephalopathy,

Figure 23.17 This EEG shows pro-


longed suppression with variable
polymorphic bursts occurring syn-
chronously, bilaterally in a patient
who had anoxia and cardiac arrest.
This is also a burst-suppression pat-
tern. Care was withdrawn for this
patient.
448 Part III ■ Clinical EEG: General

Figure 23.18 This patient was


obtunded with an excess of benzodi-
azepines. The EEG shows diffuse 20-
to 25-Hz beta activity, reflecting the
medication effect, in a pattern
resembling sleep spindles, but of
higher frequency.

or seizures (Fig. 23.18). With posttraumatic coma, normal- the EP testing resources are not available in many hospitals,
looking sleep spindles carry a good prognosis (Fig. 23.9). On the excepting some academic centers. Imaging techniques are still
other hand, several posttraumatic cases exhibiting spindle coma largely experimental, but may be useful in individual cases.
have a poor outcome probably due to prominent primary or sec- When patients reach the chronic stages with LIS, VS, PVS, or
ondary brainstem lesions (174,176). The absence of spindles in MCS, electrophysiology does not accrue further prognostic value.
anoxic coma was associated with poor outcome, whereas its pres-
ence did not indicate a favorable course (177). Improvement of Vegetative States
EEG signs at serial recordings may represent the first indication Regarding VS, there is a wide variety of possible EEG patterns,
of a favorable course. including normal reactive alpha, diffuse slowing and ranging to
On the other hand, absence of reactivity has been judged an continuous arrhythmic delta slowing, seen in most patients.
ominous sign (83). With rostrocaudal deterioration, the number Other types of pattern include an unreactive alpha, theta, or
of patterns decreases (153). A very poor prognosis is indicated by spindle coma pattern, reactive theta background, or diffuse
monotonous, high-voltage 0.5- to 3-Hz activity without changes voltage suppression consistent with electrocerebral inactivity.
occurring spontaneously or secondary to exogenous stimuli Hence, EEG is of limited value in VS. There may be EEG evi-
(178), the burst-suppression pattern (15,100,101,159,179–181), dence of wakefulness or sleep activity. Such testing does not
generalized periodic phenomena (100,120,180,181), intermittent evaluate consciousness, and EEG and SSEP may not reliably
flattening, by monorhythmic alpha frequencies (83,100,119,120), separate VS from LIS in some patients.
and a low-voltage-output EEG (15,100,101,120,159,179). With
intoxications, the aforementioned patterns are of less serious sig-
nificance. TWs indicate a poor prognosis in liver diseases (182; see PROGNOSIS IN ANOXIC COMA;
Fig. 23.11). Paroxysmal abnormalities in coma represent an unfa- CLINICAL AND ELECTROPHYSIOLOGIC
vorable sign (120), except in posttraumatic coma (15). CONSIDERATIONS
Patients typically evolve over hours to days from comatose
states. A typical progression would be from coma to alertness, Methodologic approaches can vary widely in studies of coma.
coma to death, or, in a minority of patients, coma to VS. In The duration of anoxia is a major determinant, as is the duration
some reports, progression from coma to VS is not accompanied of CPR. The duration of hypoxia may be difficult to quantify, but
by a change in EEG. In some cases, EEG change will not be data suggest that if the coma lasts 2 to 3 days, there is rarely a
reflected in a clinical change. complete recovery (183). Most comas lasting a few hours recover
(184). About 80% of CA survivors regained consciousness in 1
PROGNOSTIC CRITERIA FROM EEG hour after CPR (185), while others have found that about 20% of
patients who are unconscious for 10 minutes after CPR are nor-
For prognosis, EEG can be obtained shortly after an irreversible mal or nearly normal at 1 year (186)—the remainder had persist-
insult such as ischemia/anoxia from CRA. It can then be used to ent severe deficits. Edgren et al. (1994) and Levy et al. (1981)
help predict outcome, and the probabilities of making a good, found that absent motor responses on day 3 with unreactive
poor, or no recovery. After anoxic coma, EPs also provide reliable pupils predicted PVS in all patients. Unreactive pupils at initial
prognostication of a poor or negative outcome. Combined with examination foretold that 0/52 patients had a moderate or good
EPs, an Index of Global Cortical Function (IGCF) can also be outcome (187,188). Wiilloughby and Leach (1974) also found
generated to provide a graded prognostication, but frequently that no response or stereotyped reflexes at 1 hour after CA bode
Chapter 23 ■ Anoxia, Coma, and Brain Death 449

a poor prognosis (189). Poor outcome occurred if at 3 days, no later, and there may be a progression through several EEG pat-
motor response or flexor and extensor posturing was present. A terns of coma, seizure, or prognostic significance in the early
minority of 5% to 7% of patients with no pupillary light reaction period (200,205). A number of investigators have promulgated
and motor responses had favorable outcomes (190). The pres- different EEG scales of pattern severity over the years, with sub-
ence of myoclonic status epilepticus after CA is also indicative of division varying from 5 to 13 grades, and with greater or lesser
a poor outcome (15,59–61). Briefer anoxia or hypoxia may result homogeneity of the study populations. Early work by Fischgold
in stimulus-sensitive myoclonus in an awake patient and Mathis, and by Prior (1974) provided excellent qualitative
(Lance–Adams syndrome), with good prognosis (64). guidance to the types of abnormalities, and some insight into
Overall 50% of out-of-hospital CA patients survive after the evolution over time (84). Scales by Hockaday as early as
resuscitation (191–193), and 20% at 1 year (168); about one 1965, later modified by Synek (1988, 1990), Scollo-Lavizzari and
sixth to one half will recover to normal wakefulness and cogni- Bassetti (1987), and Young and colleagues (2004, 2005, 2006),
tion. Other workers report a 20% good recovery; 80% died or have led to increasing use of grading scales to predict outcome
vegetative (194). after CRA (159,206). For the high and low grades, an accuracy of
After in-hospital CPR, more than 90% of patients had a above 80% has been touted by a number of investigators. Data
favorable recovery (195). The use of EEG in predicting outcome analysis of the early hours of EEG after CRA may yield greater
has been the subject of much study over the years opportunities for therapeutic intervention. The availability of
(110,125,127,137,196–203). Quite aside from the challenges electrophysiology has a proven role in the decision-making of
faced in determining the value of EEG in accurately predicting ICU specialists in the withdrawal of care.
prognosis for the range of possibilities from normal to death or All scales agree that the EEG patterns of electrographic status
PVS are the problems of interference in producing accurate EEG in coma, status myoclonicus with a burst-suppression pattern, a
data because of administered medications that modify the EEG low-voltage EEG, or ECS are reliable indicators of death or a VS
or the clinical examination. Furthermore, more recent use of (Figs. 23.19 to 23.23). More recent studies have in part removed
hypothermia also modifies the interpretation of EEG and clini- patterns such as spindle or alpha coma from the extremely poor
cal findings. Electrophysiology (EEG and evoked potentials) has prognosis, particularly if there is some reactivity (160,176).
been most successful in accurately prognosticating patients at Others have more systematically excluded EEG responsiveness to
the extremes of outcome—that is to say those who will do well external stimuli in the diagnosis of these coma patterns (com-
or very poorly. Patients who lie in between these extremes have plete or incomplete alpha/theta coma), and hence found the
been far more problematic with regards to accurate prognostica- worse prognosis (119,125). A similar distinction can be found
tion (100,101,159,200,204). Furthermore, the time of electro- with types of burst-suppression patterns. A summary of our
physiologic assessment has also been found to be important, present understanding of the reliability of electrophysiology in
particularly for EEG. Early EEG ( 24 hours) after coma onset is CRA outcome has been provided by the excellent meta-analysis
less reliable for prognostic purposes as a record obtained days of 33 studies, totaling more than 4500 patients. Reliable predictors

Figure 23.19 This EEG shows a pattern that is intermediate between that of generalized periodic discharges (GPEDs) and
generalized electrographic status epilepticus in coma. This patient has had prolonged cardiorespiratory arrest.
450 Part III ■ Clinical EEG: General

Figure 23.20 In this obtunded patient after cardiac arrest,


there is diffuse theta activity with some arrhythmic delta
activity, representing an intermediate grade 3 on the Synek
scale of recovery after anoxic insult.

of poor outcome (death or PVS) were found to be the absence of (vii) cerebral death syndrome; and (viii) irreversible breakdown
early cortical somatosensory evoked responses (0% to 2% for of cerebral functions.
95% confidence interval [CI] of pooled false-positive tests), elec- From a neurologic point of view, there are two problems with
trographic seizures, grade 4 to 5 EEG ( 5 V or flatline tracing), non-heart-beating donors. No data exist about how long the heart
and burst-suppression (196). From this analysis and for subse- beat has to be arrested until the brain is irreversibly out of any
quent prognostication, the authors suggest a stepwise approach function. Category 3 patients, according to Koostra et al. (1995),
beginning 3 days after coma onset. Those patients without pupil- are ICU patients dying from severe brain trauma with no chance
lary reflexes or motor response to pain (better than flexion) for recovery but who do not meet the criterion of brain death. To
should be tested with SSEPs. Patients lacking cortical responses harvest organs, medical support is withdrawn until cardiac arrest
can then be considered as having no prognosis for meaningful occurs. Ethically, this procedure can hardly be accepted. With the
recovery and guided toward the option of palliative care. The pre- other categories of non-heart-beating donors, neurologists and
dictive value of neuroimaging, and of particular laboratory electroencephalographers might not be involved in the decision-
parameters in the serum and cerebrospinal fluid fall outside the making process.
scope of this chapter and will not be addressed here.
Neurologic Signs
BRAIN DEATH In the diagnosis of cerebral death, the first step is the demon-
stration of a brain death syndrome based on neurologic exam-
Definition ination. Neurologic signs of cortical functions and brainstem
The entire problem of cerebral death is a by-product of improved activity must be absent. Pupils must be fixed even with a strong
medical care; it has become urgent because of the need for organs light stimulus. Peripheral third nerve injuries should be
for transplantation surgery. Numerous terms are used for the excluded. Oculovestibular responses must be absent (185,186).
same clinical entity: (i) aperceptive, areactive, apathic, and atonic No motor activity must be observed in the muscles innervated
syndrome (183); (ii) brain death; (iii) stage IV coma (84); (iv) by cranial nerves. Spontaneous respiration must be absent; no
coma depasse; (v) irreversible coma (5); (vi) cerebral death; respiration movements should be detectable after removal from

Figure 23.21 This EEG sample is from a patient after


cardiac arrest, with a Glasgow Coma Scale of 3. A flatline
tracing like this is classified as a 5 on the 5-point Synek
scale, and represents electrocerebral inactivity. If after car-
diac arrest and in the absence of hypothermia, or CNS-
suppressant drugs, it carries a 100% bad outcome.
Chapter 23 ■ Anoxia, Coma, and Brain Death 451

Figure 23.22 After anoxia this patient showed prolonged


bursts and brief suppressions (burst-suppression pattern),
and died.

the respirator. Testing for apnea is necessary to confirm the EEG activity with isolated brainstem death proves this state-
neurologic brain death syndrome. ment. In cases of primary brainstem lesions, the EEG is not
The practical procedure differs markedly within different only confirmatory, but also essential for correct diagnosis. The
centers (186). A standardized procedure has been suggested for same is true for fulminant demyelinating polyradiculopathy
apnea testing. The varying criteria used across the United States resembling brainstem death.
have been reviewed, and a more standardized approach was The relationship between brain death diagnosed on the basis
suggested. of neurologic and EEG examinations and respirator brain at
autopsy has been reassessed. Overly rigid criteria for the differ-
Electrocerebral Silence (ECS) entiation of ECS from the very low-voltage-output EEG and
An isoelectric EEG is confirmatory for the establishment of excess noise have been criticized. In spite of certain shortcom-
cerebral death. It must be emphasized, however, that ECS per ings, EEG has proved to be of paramount importance in the
se does not mean cerebral death. Isoelectric EEG recordings evaluation of brain death. Shortcomings are of a technical
have been found with intoxication and full recovery (187), nature, as are conceptual misunderstandings, such as with pri-
hypothermia (100), and transient decorticate states followed mary brainstem death and the overinterpretation of artifacts.
by varying degrees of recovery (84). Therefore, ECS can be However, real cerebral EEG waves exclude brain death by defi-
taken as a sign of brain death only if neurologic signs of corti- nition. To bypass these difficulties, several attempts have been
cal and brainstem functions are lacking, and intoxication and made to redefine brain death and to confine it to brainstem
marked hypothermia can be excluded. A brain death syndrome death. The concept of whole-brain death delivers high diagnos-
at the neurologic examination is based on brainstem death and tic practicability and certainty and has achieved a high level of
does not exclude ongoing cortical activities reflected by EEG acceptance in the Western society. There is no urgent need to
activity (188,189). To consider EEG activity with brainstem change it to brainstem death or neocortical death.
death as a pitfall of EEG in determining brain death, because The definition and reliability of ECS is based on the technical
brainstem death equals brain death, turns the logic upside standards applied for its registration. Many records referred to as
down. Brainstem death does not mean brain death, defined as showing ECS show low-voltage-output EEG or use insufficient
irreversible loss of functions of all parts of the brain. Ongoing technical standards. Several national EEG societies have published

Figure 23.23 This EEG tracing was obtained after CRA,


but during hypothermia. Although SE after CRA usually is
fatal, hypothermia may improve prognosis in some, but
complicates EEG interpretation and prognostication. Is this
GPEDs or status epilepticus?
452 Part III ■ Clinical EEG: General

recommendations for EEG recordings in suspected brain death. 14. Laureys S, Owen AM, Schiff ND. Brain function in coma, vegeta-
These suggestions do not differ essentially among the national tive state, and related disorders. Lancet Neurol. 2004;3(9):537–546.
recommendations. They include the following: a minimum of 15. Bauer G, Niedermeyer E. Acute convulsions. Clin Electro-
eight scalp electrodes and reference electrodes to cover the major encephalogr. 1979;10:127–144.
16. Chia LG. Locked-in syndrome with bilateral ventral midbrain
brain areas; interelectrode impedances under 10,000 but over
infarcts. Neurology. 1991;41:445–446.
100 ; testing the integrity of the entire recording system;
17. Dehaene J, Dom R. A mesencephalic locked-in syndrome. J Neurol.
interelectrode distances of at least 10 cm to magnify the ampli- 1982;227:255–259.
tudes and to pick up electrical fields originating in deep struc- 18. Hochmann MS, Sowers JJ, Bruce-Gregorius J. Syndrome of the
tures; sensitivity increased up to 2 V/mm during most of the mesencephalic artery: report of a case with CT and necropsy find-
recording to distinguish ECS from low-voltage-output EEG; ings. J Neurol Neurosurg Psychiatry. 1985;48:1179–1181.
the use of time constants of 0.3 to 0.4 second; the use of mon- 19. Karp JS, Hurtig HI. “Locked-in” state with bilateral midbrain
itoring techniques, with simultaneous ECG recording to be infarcts. Arch Neurol. 1974;30:176–178.
mandatory; testing of EEG reactivity to exogenous stimuli; a 20. Uematsu D, Suematsu M, Fukunchi Y, et al. Midbrain locked-in
recording time of at least 30 minutes; and a recording to be state with oculomotor subnucleus lesion. J Neurol Neurosurg
made only by a qualified technologist. The most important Psychiatry. 1985;48:952–956.
21. Chia LG. Locked-in state with bilateral internal capsule infarcts.
recommendation is the use of high instrument sensitivity
Neurology. 1984;34:1365–1367.
along with the need for continuous recognition and elimina-
22. Gütling E, Isenmann S, Wichmann W. Electrophysiology in the
tion of artifacts. This is indeed a painstaking task in a busy locked-in-syndrome. Neurology. 1996;46(4):1092–1101.
ICU. If the EEG is chosen as a confirmatory laboratory test, the 23. Jacome DE, Morilla-Pastor D. Unreactive EEG: pattern in locked-
length of observation time until a second and final EEG should in syndrome. Clin Electroencephalogr. 1990;21(1):31–36.
be recorded has been under discussion for a long time, with 24. Wijdicks EF, Cranford RE. Clinical diagnosis of prolonged states of
intervals between recordings and clinical observation varying impaired consciousness in adults. Mayo Clin Proc. 2005;80(8):
from 6 to 24 hours. In cases with diagnostic or technical uncer- 1037–1046.
tainties, repeated recordings are advised. The usefulness of 25. Matsuo F. EEG features of the apallic syndrome resulting from
EEG in the evaluation of brain death in children has been dis- cerebral anoxia. Electroencephalogr Clin Neurophysiol. 1985;61(2):
cussed and remains under contention. With optimal technical 113–122.
26. French JD, Magoun HW. Effects of chronic lesions in central cephalic
standards, similar criteria to those used in adults can be applied,
brain stem of monkeys. Arch Neurol Psychiatry. 1952;68:591–604.
particularly for older children.
27. Plum F. Organic disturbances of consciousness. In: Critchley M,
O’Leary JLO, Jennet B, eds. Scientific Foundations of Neurology.
REFERENCES Philadelphia, PA: F.A. Davis; 1972:193–201.
28. Parvizi J, Damasio R. Neuroanatomical correlates of brainstem
1. Schiff ND, Fins JJ. Disorders of consciousness. Mayo Clin Proc. coma. Brain. 2003;126:1524–1536.
2007;82(2):250–251. 29. McNealy DE, Plum F. Brainstem dysfunction with supratentorial
2. Giacino JT, Ashwal S, Childs N, et al. The minimally conscious mass lesions. Arch Neurol. 1962;7:10–32.
state. Neurology. 2002;58:349–353. 30. Ropper AH. Lateral displacement of the brain and level of con-
3. Schiff ND. Bringing neuroimaging tools closer to diagnostic use in sciousness in patients with an acute hemispheral mass. N Engl
the severely injured brain. Brain. 2007;30(pt 10):2482–2483. J Med. 1986;314:953–958.
4. Gerstenbrand F. Das Traumatische Apallische Syndrome. Wien- 31. Ropper AH. Syndrome of transtentorial herniation: is vertical dis-
New York: Springer-Verlag; 1967. placement necessary? J Neurol Neurosurg Psychiatry. 1993;56(8):
5. Vigouroux RP. Between life and death. In: Harner R, Naquet R, 932–935.
eds. Altered States of Consciousness, Coma, Cerebral Death. 32. Feldmann E, Gandy SE, Becker R, et al. MRI demonstrates
Handbook of Electroencephalography and Clinical Neurophysiology. descending transtentorial herniation. Neurology. 1988;38:697–701.
Vol 11. Amsterdam: Elsevier; 1975:95–99. 33. Reich JB, Sierra J, Camp W, et al. Magnetic resonance imaging and
6. Jennet B, Plum F. Persistent vegetative state after brain damage. A clinical changes accompanying transtentorial and foramen mag-
syndrome in search of a name. Lancet. 1972;1:734–737. num brain herniation. Ann Neurol. 1993;33:159–170.
7. Schiff ND, Rodriguez-Moreno D, Kamal A, et al. fMRI reveals 34. Wijdicks EFM, Miller GM. MR imaging of progressive downward
large-scale network activation in minimally conscious patients. herniation of the diencephalon. Neurology. 1997;48:1456–1459.
Neurology. 2005;64(3):514–523. 35. Nyberg-Hansen R, Loken AC, Tenstad O. Brainstem lesion with
8. Levin HS, Saydjari Ch, Eisenburg HM, et al. Vegetative state after coma for five years following manipulation of the cervical spine.
closed-head injury. Arch Neurol. 1991;48:580–585. J Neurol. 1978;218:97–105.
9. Molinari GF. Persistent vegetative state, do not resuscitate … and 36. Dinsdale HB. Spontaneous hemorrhage in the posterior fossa.
still more words doctors use. J Neurol Sci. 1991;102:125–127. Arch Neurol. 1964;10:200–217.
10. Munsat ThL, Stuart WH, Cranford RE. Guidelines on the vegeta- 37. Mitchell DE, Adams JM. Primary focal impact damage to the
tive state. Neurology. 1989;39:123–124. brainstem in blunt head injuries. Does it exist? Lancet. 1973;
11. Spudis EV. The persistent vegetative state—1990. J Neurol Sci. II:215–218.
1991;102:128–136. 38. Gentry LR, Godersky JC, Thompson BH. Traumatic brain stem
12. Fins JJ, Schiff ND. Shades of gray: new insights into the vegetative injury: MR imaging. Radiology. 1989;171:177–187.
state. Hastings Cent Rep. 2006;36(6):8. 39. Adams JH, Graham DI, Genarelli TA, et al. Diffuse axonal injury in
13. Plum F, Posner JB. The Diagnosis of Stupor and Coma. non-missile head injury. J Neurol Neurosurg Psychiatry. 1991;54:
Philadelphia, PA: F.A. Davis; 1966. 481–483.
Chapter 23 ■ Anoxia, Coma, and Brain Death 453

40. Gastaut H, Fischer-Williams M. The physiopathology of epileptic 63. Lance JW, Adams RD. The syndrome of intention or action
seizures. In: Field Y, et al., eds. Handbook of Physiology. Vol 1. myoclonus as a sequel to hypoxic encephalopathy. Brain. 1963;86:
Washington: American Physiological Society; 1959:329–363. 111–133.
41. New PFJ, Scott WR. Computerized Tomography of the Brain and 64. Morris HR, Howard RS, Brown P. Early myoclonic status and out-
Orbit. Baltimore, MD: Williams & Wilkins; 1975. come after cardiorespiratory arrest. J Neurol Neurosurg Psychiatry.
42. Ruggiero G, Sabattini L, Nuzzo G. Computerized tomography and 1998;64:267–268.
encephalography. Neuroradiology. 1977;13:45–48. 65. Werhan KJ, Brown P, Thompson PD, et al. The clinical features and
43. Torack RM, Alcala H, Gado M, et al. Correlative assay of comput- prognosis of chronic post hypoxic myoclonus. Mov Disord.
erized cranial tomography (CCT), water content and specific 1997;12:216–220.
gravity in normal and pathological post mortem brain. 66. Kjos BO, Brant-Zawadzki M, Young RG. Early CT findings of
J Neuropathol Exp Neurol. 1976;34:385–392. global central nervous system hypoperfusion. AJNR Am
44. Kaplan PW. Stupor and coma: metabolic encephalopathies. Suppl J Neuroradiol. 1983;4:1043–1048.
Clin Neurophysiol. 2004;57:667–680. 67. Morimoto Y, Kemmotsu O, Kitami K, et al. Acute brain swelling
45. Kaplan PW. The EEG in metabolic encephalopathy and coma. after out-of-hospital cardiac arrest: pathogenesis and outcome.
J Clin Neurophysiol. 2004;21(5):307–318. Crit Care Med. 1993;21:104–110.
46. Kaplan PW, Birbeck G. Lithium-induced confusional states: non- 68. Birbamer G, Aichner F, Felber S, et al. MRI of cerebral hypoxia.
convulsive status epilepticus or triphasic encephalopathy? Neuroradiology. 1991;33:53–55.
Epilepsia. 2006;47(12):2071–2074. 69. Christophe C, Fonteyne C, Ziereisen F, et al. Value of MR imaging
47. Kalita J, Misra UK. EEG in dengue virus infection with neurologi- of the brain in children with hypoxic coma. AJNR Am
cal manifestations: a clinical and CT/MRI correlation. Clin J Neuroradiol. 2002;23:716–723.
Neurophysiol. 2006;117(10):2252–2256. 70. Sawada H, Udaka F, Seriu N, et al. MRI demonstration of cortical
48. Clarke M, Newton RW, Klapper PE, et al. Childhood encephalopa- laminar necrosis and delayed white matter injury in anoxic
thy: viruses, immune response, and outcome. Dev Med Child encephalopathy. Neuroradiology. 1990;32:319–321.
Neurol. 2006;48(4):294–300. 71. Singhal AB, Topcuoglu MA, Koroshetz WJ. Diffusion MRI in three
49. Sejvar JJ, Haddad MB, Tierney BC, et al. Neurologic manifestations types of anoxic encephalopathy. J Neurol Sci. 2002;196:37–40.
and outcome of West Nile virus infection. JAMA. 2003;290(4): 72. Takahashi S, Higano S, Ishii K, et al. Hypoxic brain damage: corti-
511–515. cal laminar necrosis and delayed changes in white matter at
50. Clarke DD, Sokoloff L. Circulation and energy metabolism of the sequential MR imaging. Radiology. 1993;189:449–456.
brain. In: Siegel GJ, et al., eds. Basic Neurochemistry: Molecular, 73. Chalela JA, Wolf RL, Maldjian JA, et al. MRI identification of early
Cellular, and Medical Aspects. 5th ed. New York, NY: Raven Press; white matter injury in anoxic–ischemic encephalopathy.
1994:645–680. Neurology. 2001;56:481–485.
51. Martin RL, Lloyd HGE, Cowan AL. The early events of oxygen and 74. Rudolf J. Positron emission tomography in diagnosis and progno-
glucose deprivation: setting the scene for neuronal death. Trends sis of postanoxic cerebral dysfunctions. Fortschr Neurol Psychiatr.
Neurosci. 1994;17:251–257. 2000;68:344–351.
52. Hori A, Hirose G, Kataoka S, et al. Delayed postanoxic 75. Schaafsma A, de Jong BM, Bams JL, et al. Cerebral perfusion and
encephalopathy after strangulation. Arch Neurol. 1991;48:871–874. metabolism in resuscitated patients with severe post-hypoxic
53. Petito CH, Feldmann E, Pulsinelli WA, et al. Delayed hippocampal encephalopathy. J Neurol Sci. 2003;210:23–30.
damage in humans following cardiorespiratory arrest. Neurology. 76. Gibbs FA, Gibbs EL, Lennox WG. Effect on the electroencephalo-
1987;37:1281–1286. gram of certain drugs which influence nervous activity. Arch Intern
54. Hallenbeck JM, Dutka AJ. Background review and current con- Med. 1937;60:154–166.
cepts of reperfusion injury. Arch Neurol. 1990;47:1245–1254. 77. Daly DD. Use of EEG for diagnosis and evaluation of epileptic
55. Miyamoto O, Auer RN. Hypoxia, hyperoxia, ischemia, and brain seizures and nonepileptic episodic disorders. In: Klass DD, Daly
necrosis. Neurology. 2000;54:362–371. DD, eds. Current Practice of Clinical Electroencephalography. New
56. Heiss WD, Rosner G. Functional recovery of cortical necrosis as York, NY: Raven Press; 1979:221–268.
related to degree and duration of ischemia. Ann Neurol. 1983;14: 78. Harner RN, Katz RI. Electroencephalography in metabolic coma.
294–301. In: Harner R, Naquet R, eds. Altered States of Consciousness,
57. Snyder BD, Loewenson RB, Gumnit RJ, et al. Neurological progno- Coma, Cerebral Death. Handbook of Electroencephalography and
sis after cardiac arrest: III. Seizure activity. Neurology. 1980;30: Clinical Neurophysiology. Vol 12. Amsterdam: Elsevier; 1975:
1292–1297. 47–62.
58. Jumao-as A, Brenner RP. Myoclonic status epilepticus: a clinical 79. Gloor P. Generalized and widespread bilateral paroxysmal activi-
and electroencephalographic study. Neurology. 1990;40:1199–1202. ties. In: Cobb W, ed. EEG Interpretation in Clinical Medicine.
59. Krumholz A, Stern BJ, Weiss HD. Outcome from coma after car- Handbook of Electroencephalography and Clinical Neurology. Vol
diopulmonary resuscitation: relation to seizures and myoclonus. 11B. Amsterdam: Elsevier; 1976:52–87.
Neurology. 1988;38:401–405. 80. Arfel G, Fischgold H. EEG signs in tumors of the brain. In:
60. Wijdicks EFM, Parisi JE, Sharbrough FW. Prognostic value of Electroencephalography and Cerebral Tumors. Electroencephalogr
myoclonic status in comatose survivors of cardiac arrest. Ann Neurol. Clin Neurophysiol Suppl. 1961;19:36–50.
1994;35:239–243. 81. Hess R. Die bioelektrischen Zeichen der cerebralen Massenver -
61. Young GB, Gilbert JJ, Zochodne DW. The significance of schiebung bei Hirntumoren. Schweiz Med Wochenschr. 1962;92:
myoclonic status epilepticus in postanoxic coma. Neurology. 1990; 1537–1542.
40:1843–1848. 82. Schaul N, Gloor P, Gotman J. The EEG in deep midline lesions.
62. Gatzonis SD, Zournas Ch, Michalopoulos A, et al. Area-selective Neurology (New York). 1981;31:157–167.
stimulus-provoked seizures in post-anoxic coma. Seizure. 2001;10: 83. Arfel G. Introduction to clinical and EEG studies in coma. In:
294–297. Harner R, Naquet R, eds. Altered States of Consciousness, Coma,
454 Part III ■ Clinical EEG: General

Cerebral Death. Handbook of Electroencephalography and Clinical 107. Miller JW, Klass DW, Mokri B, et al. Triphasic waves in cerebral
Neurophysiology. Vol 12. Amsterdam: Elsevier; 1975:5–23. carcinomatosis. Arch Neurol. 1986;43:1191–1193.
84. Fischgold H, Mathis P. Obnubilations, comas et stupeurs: ƒtudes 108. Blatt I, Brenner RP. Triphasic waves in a psychiatric population: a
Electroencephalographique. Electroencephalogr Clin Neurophysiol. retrospective study. J Clin Neurophysiol. 1996;13:324–329.
(suppl 11). Paris: Masson; 1959. 109. Bauer G, Aichner F, Mayr U. Status atypischer Absencen in
85. Kubicki S, Rieger H, Busse G, et al. Elektroenzephalographische Jugend- und Erwachsenenalter. Nervenarzt. 1983;54:100–105.
Befunde bei schweren Schlafmittelvergiftungen. Z EEG-EMG. 110. Bauer G, Prugger M, Bohr K, et al. Relationship between the sup-
1970;1:80–93. pression burst pattern and rhythmical activities in the alpha and
86. Schwartz MS, Scott DF. Pathological stimulus-related slow theta bands of EEG of comatose patients. Electroencephalogr Clin
wave arousal response in the EEG. Acta Neurol Scand. 1978;57: Neurophysiol. 1989;76:72P.
300–304. 111. Zaret BS. Prognostic and neurophysiological implications of
87. Chatrian GE, Shaw CM, Leffman H. The significance of periodic concurrent burst suppression and alpha patterns in the EEG of
lateralized epileptiform discharges in EEG: an electrographic, postanoxic coma. Electroencephalogr Clin Neurophysiol. 1985;61:
clinical and pathological study. Electroencephalogr Clin 199–209.
Neurophysiol. 1964;17:177–193. 112. Niedermeyer E, Sherman DL, Geocadin RJ, et al. The burst-
88. de la Paz D, Brenner RP. Bilateral independent periodic lateral- suppression electroencephalogram. Clin Electroencephalogr.
ized epileptiform discharges. Clinical significance. Arch Neurol. 1999;30(3):99–105.
1981;38(11):713–715. 113. Butenuth J, Kubicki S. Über die prognostische Bedeutung bes-
89. Kuroiwa Y, Celesia GG. Clinical significance of periodic EEG pat- timmter Formen der Myoklonien und korrespondierender EEG-
terns. Arch Neurol. 1980;37:15–20. Muster nach Hypoxien. Z EEG-EMG. 1971;2:78–83.
90. Schwartz MS, Prior PF, Scott DF. The occurrence and evolution 114. Pampiglione G, Harden A. Resuscitation after cardio-vascular
in the EEG of a lateralized periodic phenomenon. Brain. 1973; arrest. Prognostic evaluation and early electroencephalographic
96:613–622. findings. Lancet. 1968;1:1261–1264.
91. Husain AM, Mebust KA, Radtke RA. Generalized periodic 115. Chatrian GE, White LE, Shaw C-M. EEG pattern resembling
epileptiform discharges: etiologies, relationship to status epilep- wakefulness in unresponsive decerebrate state following trau-
ticus, and prognosis. J Neurophysiol Clin. 1999;16(1):51–58. matic brain stem infarct. Electroencephalogr Clin Neurophysiol.
92. Lowenstein DH, Aminoff MJ. Clinical and EEG features of status 1964;16:285–289.
epilepticus in comatose patients. Neurology. 1992;42:100–104. 116. Loeb C, Poggio G. Electroencephalograms in a case with ponto-
93. Suter C. Clinical advances in the evaluation of deep coma. MCV mesencephalic haemorrhage. Electroencephalogr Clin Neuro-
Q. 1974;10:152–162. physiol. 1953;5:295–296.
94. Terzano MG, Parrino L, Mazzuchi A, et al. Confusional states with 117. Lundervold A, Hange T, Lšken AC. Unusual EEG in unconscious
periodic lateralized epileptiform discharges (PLEDs): a peculiar patient with brain stem atrophy. Electroencephalogr Clin
epileptic syndrome in the elderly. Epilepsia. 1986;27:446–457. Neurophysiol. 1956;8:665–670.
95. Handforth A, Cheng JT, Mandelkern MA, et al. Markedly 118. Rohmer R, Kurtz D, Kiffer A. étude critique de l’activité E.E.G.
increased mesiotemporal lobe metabolism in a case with PLEDs: dan les syndromes vascularies du tronc cérébral. Rev Neurol. 1965;
further evidence that PLEDs are a manifestation of partial status 113:278–284.
epilepticus. Epilepsia. 1994;35:876–881. 119. Westmoreland BF, Klass DW, Sharbrough FM, et al. Alpha-coma.
96. Fisch BJ, Klass DW. The diagnostic specificity of triphasic wave Electroencephalographic, pathologic and etiologic correlations.
patterns. Electroencephalogr Clin Neurophysiol. 1988;70(1):1–8. Arch Neurol. 1975;32:713–718.
97. Bickford RG, Butt HR. Hepatic coma: the electroencephalo- 120. Binnie CD, Prior PF, Lloyd DSL, et al. Electroencephalographic
graphic pattern. J Clin Invest. 1955;34:790–799. prediction of fatal anoxic brain damage after resuscitation from
98. Reiher J. The electroencephalogram in the investigation of meta- cardiac arrest. Br Med J. 1970;4:265–268.
bolic comas. Electroencephalogr Clin Neurophysiol. 1970;28:104. 121. Chokroverty S. “Alpha-like” rhythms in electroencephalograms
99. Calham CL, Ettinger MG. Unusual EEG in coma after cardiac in coma after cardiac arrest. Neurology (Minneapolis). 1975;25:
arrest. Electroencephalogr Clin Neurophysiol. 1966;21:385–388. 655–663.
100. Prior PF. The EEG in Acute Cerebral Anoxia. Amsterdam: 122. Grindal AB, Suter C. “Alpha-pattern coma” in high voltage electri-
Excerpta Medica; 1973. cal injury. Electroencephalogr Clin Neurophysiol. 1975;38:521–526.
101. Silverman D. The electroencephalogram in anoxic coma. In: 123. Sharbrough FW, Westmoreland BF, Reagan TJ, et al. The signifi-
Harner R, Naquet R, eds. Altered States of Consciousness. Coma, cance of a transitional monorhythmic EEG pattern in patients
Cerebral Death/Handbook of Electroencephalography and Clinical after cardiopulmonary arrest. Neurology (Minneapolis). 1975;27:
Neurophysiology. Vol 12. Amsterdam: Elsevier; 1975: 81–94. 384–385.
102. Vulliemoz S, Iwanowski P, Landis T, et al. Levetiracetam accumu- 124. Vignaendra VMB, Wilkus RJ, Copass MK, et al. Electro -
lation in renal failure causing myoclonic encephalopathy with encephalographic rhythms of alpha frequency in comatose
triphasic waves. Seizure. 2009;18:376–378. patients after cardiopulmonary arrest. Neurology (Minneapolis).
103. Neufeld MY. Periodic triphasic waves in levodopa-induced 1974;24:582–588.
encephalopathy. Neurology. 1992;42(2):444–446. 125. Berkhoff M, Donati F, Bassetti C. Postanoxic alpha (theta) coma:
104. Madison D, Niedermeyer E. Epileptic seizures resulting from acute a reappraisal of its prognostic significance. Clin Neurophysiol.
cerebral anoxia. J Neurol Neurosurg Psychiatry. 1970;33:381–386. 2000;111:297–304.
105. Towsend JB, Drury J. Triphasic waves in coma from brainstem 126. Nogueira de Melo A, Krauss GL, Niedermeyer E. Spindle coma:
infarction. Eur Neurol. 1991;31:47–49. observations and thoughts. Clin Electroencephalogr. 1990;21:
106. Young GB, Bolton CF, Archibald YM, et al. The electroencephalo- 151–161.
gram in sepsis-associated encephalopathy. J Clin Neurophysiol. 127. Iragui VJ, McCutchen ChB. Physiologic and prognostic significance
1992;9:145–152. of “alpha coma”. J Neurol Neurosurg Psychiatry. 1983;46:632–638.
Chapter 23 ■ Anoxia, Coma, and Brain Death 455

128. Kaplan PW, Genoud D, Ho TW, et al. Etiology, neurologic corre- 154. Silverman D. Retrospective study of the EEG in coma.
lations, and prognosis in alpha coma. Clin Neurophysiol. 1999; Electroencephalogr Clin Neurophysiol. 1963;15:486–503.
110(2):205–213. 155. Synek VM, Synek BJL. Theta pattern coma, a variant of alpha
129. Tomassen W, Kamphuisen HAC. Alpha coma. J Neurol Sci. pattern coma. Clin Electroencephalogr. 1984;15:116–121.
1986;76:1–11. 156. Naquet R, Fernandez-Guardiola A. Effects of various types of
130. Young GB, Blume WT, Campbell VM, et al. Alpha, theta and anoxia on spontaneous and evoked cerebral activity in the cat. In:
alpha–theta coma: a clinical outcome study utilizing serial Gastaut H, Meyer JS, eds. Cerebral Anoxia and the Electroencephalo-
records. Electroencephalogr Clin Neurophysiol. 1994;91:93–99. gram. Springfield, IL: Charles C. Thomas; 1959:72–88.
131. Chatrian GE, White LE, Daly D. Electroencephalographic pattern 157. Kuroiwa Y, Celesia GG, Chung HD. Periodic EEG discharges of
resembling those of sleep in certain comatose states after injuries to the cerebral cortex in anoxic encephalopathy: a necropsy case
the head. Electroencephalogr Clin Neurophysiol. 1963;15:272–280. report. J Neurol Neurosurg Psychiatry. 1982;45:740–742.
132. Seet RC, Lim EC, Wilder-Smith EP. Spindle coma from acute 158. Simon RP, Aminoff MJ. Electroencephalographic status epilepti-
midbrain infarction. Neurology. 2005;64(12):2159–2160. cus in fatal anoxic coma. Ann Neurol. 1986;20:351–355.
133. Ganji SS, Henry R, Furlow J. Diffuse theta activity and spindle-like 159. Van Cott AC, Blatt I, Brenner RP. Stimulus-sensitive seizures in
bursts during coma after cardiac arrest. Clin Electroencephalogr. postanoxic coma. Epilepsia. 1996;37:868–874.
1996;27(2):89–94. 160. Hockaday JM, Potts F, Epstein E, et al. EEG changes in acute cere-
134. Dadmehr N, Pakalnis A, Drake ME Jr. Spindle coma in viral bral anoxia from cardiac or respiratory arrest. Electroencephalogr
encephalitis. Clin Electroencephalogr. 1987;18(1):34–37. Clin Neurophysiol. 1965;18:575.
135. Mouradian MD, Penovich PE. Spindle coma in benzodiazepine 161. McKeown MJ, Young GB. Comparison between the alpha pattern
toxicity: case report. Clin Electroencephalogr. 1985;16(4): 213–218. in normal subjects and in alpha pattern coma. J Clin Neurophysiol.
136. Hess R. Sleep and sleep disturbances in the electroencephalo- 1997;14:414–418.
gram. Prog Brain Res. 1965;18:127–139. 162. Celesia GG. EEG and coma: is there a prognostic role for EEG?
137. Radermecker J, Macken J. Aspects électroencéphalographiques et Editorial. Clin Neurophysiol. 1999;110:203–204.
cliniques de la leucoencéphalite sclérosante subaigue. Rev Neurol. 163. Hume AI, Cant BR. Central somatosensory conduction after
1951;83:341. head injury. Ann Neurol. 1981;10:411–419.
138. Gloor P, Kalabay O, Giard N. The electroencephalogram in dif- 164. Hutchinson DO, Frith RW, Shaw NA, et al. A comparison
fuse encephalopathies: electroencephalographic correlates of between electroencephalography and somatosensory evoked
grey and white matter lesions. Brain. 1968;91:779–802. potentials for outcome predictions following severe head injury.
139. Lee RG, Blair RDG. Evolution of EEG and visual evoked response Electroencephalogr Clin Neurophysiol. 1991;78:228–233.
changes in Jakob–Creutzfeldt disease. Electroencephalogr Clin 165. Rumpl E, Prugger M, Gerstenbrand F, et al. Central somatosen-
Neurophysiol. 1975;35:133–142. sory evoked potentials in posttraumatic coma. Electroencephalogr
140. Illis LS, Taylor FM. The electroencephalogram in herpes-simplex Clin Neurophysiol. 1983;56:583–596.
encephalitis. Lancet. 1972;1:718–721. 166. Cremer OL, Moons KGM, Bouman EAC, et al. Long-term propo-
141. Kugler J, Martin JJ, Radermecker FJ, et al. Periodische Komplexe fol infusion and cardiac failure in adult head-injured patients.
im EEG bei nekrotisierender Herpes-Enzephalitis. Z EEG-EMG. Lancet. 2001;357:117–118.
1976;7:63–71. 167. Schwab S, Spranger M, Schwarz S, et al. Barbiturate coma in
142. Kaplan PW. Electrophysiological prognostication and brain severe hemispheric stroke: useful or obsolete? Neurology. 1997;48:
injury from cardiac arrest. Semin Neurol. 2006;26:403–412. 1608–1613.
143. Chang Chui H, Damasio AR. Progressive dialysis encephalopathy 168. Brain Resuscitation Clinical Trial I Study Group. Randomized
(“dialysis dementia”). J Neurol. 1980;222:145–157. clinical study of thiopental loading in comatose survivors of car-
144. Garret PJ, Mulcahy D, Carmody M, et al. Aluminium encephalopa- diac arrest. N Engl J Med. 1986;314:397–403.
thy: clinical and immunological features. Q J Med. 1988;69:775–783. 169. Dearden NM. Ischaemic brain. Lancet. 1985;2:255–259.
145. Bauer G. Der Wert von EEG-Kontrollen mšglichst bald nach 170. Evans BM, Bartlett JR. Prediction of outcome in severe head
einem epileptischen Anfall. Z EEG-EMG. 1975;6:125–130. injury based on recognition of sleep related activity in the poly-
146. Bauer G. Psychische Veränderungen bei kontinuierlichen epilep- graphic electroencephalogram. J Neurol Neurosurg Psychiatry. 1995;
tischen Entladungen. Schweiz Arch Neurol Neurochir Psychiatr. 59:17–25.
1975;116:241–255. 171. Gutling E, Gonser A, Imhof H-G, et al. EEG reactivity in the
147. Brett EM. Minor epileptic status. J Neurol Sci. 1966;3:53–75. prognosis of severe head injury. Neurology. 1995;45:915–918.
148. Niedermeyer E, Khalifeh R. Petit mal status (“spike wave 172. Luecking CH. Sleep-like patterns and abnormal arousal reactions
stupor”). Epilepsia. 1965;6:250–262. in brain stem lesions. Electroencephalogr Clin Neurophysiol. 1970;
149. Loeb C. Correlative EEG and clinico-pathological studies of patients 28:214 [abstract].
in coma. In: Harner R, Naquet R, eds. Altered States of Consciousness, 173. Bergamasco B, Bergamini L, Doriguzzi T, et al. EEG sleep pat-
Coma, Cerebral Death. Vol 11. Amsterdam: Elsevier; 1975:24–36. terns as a prognostic criterion in post-traumatic coma. Electroen-
150. Romano J, Engel GL. Delirium. I: electroencephalographic data. cephalogr Clin Neurophysiol. 1968;24:374–377.
Arch Neurol Psychiatry. 1944;52:290–295. 174. Rumpl E, Prugger M, Bauer G, et al. Incidence and prognostic
151. Vigouroux R, Naquet R, Baurand C, et al. Evolution électroradio- value of spindles in posttraumatic coma. Electroencephalogr Clin
clinique de comas graves prolongés post traumatiques. Rev Neurophysiol. 1983;56:420–429.
Neurol. 1964;110:72–80. 175. Valente M, Placidi F, Oliveira AJ, et al. Sleep organization pattern
152. Bergamini L, Bergamasco B, Mombelli A, et al. Autocorrelazione as a prognostic marker at the subacute stage of post-traumatic
in sogetti in coma. Rev Neurol. 1966;36:233–236. coma. Clin Neurophysiol. 2002;113:1798–1805.
153. Rumpl E, Lorenzi E, Hackl JM, et al. The EEG at different stages 176. Kaplan PW, Genoud D, Ho TW, et al. Clinical correlates and
of acute secondary traumatic midbrain and bulbar brain syn- prognosis in early spindle coma. Clin Neurophysiol. 2000;111:
dromes. Electroencephalogr Clin Neurophysiol. 1979;46:487–497. 584–590.
456 Part III ■ Clinical EEG: General

177. Hulihan JF, Syna DR. Electroencephalographic sleep patterns in 193. Snyder BD, Loewenson RB, Gumnit RJ, et al. Neurologic progno-
post-anoxic stupor and coma. Neurology. 1994;44:758–760. sis after cardiac arrest: II. Level of consciousness. Neurology. 1980;
178. Dusser A, Nevelet Y, Devictor D, et al. Short- and long-term 30:52–58.
prognostic value of electroencephalogram in children with 194. Bassetti C, Bomio F, Mathis H, et al. Early prognosis after cardiac
severe head injury. Electroencephalogr Clin Neurophysiol. 1989; arrest: a prospective clinical, electrophysiological, and biochemical
73:85–93. study of 60 patients. J Neurol Neurosurg Psychiatry. 1996;61:
179. Bickford RG, Dawson B, Takeshita H. EEG evidence of neuro- 610–615.
logic death. Electroencephalogr Clin Neurophysiol. 1965;18:513. 195. Murphy DJ, Murray AM. Outcome of cardiopulmonary resusci-
180. Pampiglione G. EEG studies after cardio-pulmonary resuscita- tation in the elderly. Ann Intern Med. 1989;111:199–203.
tion. Proc R Soc Med. 1962;55:653. 196. Zandbergen EGJ, de Haan RJ, Stoutenbeek CP, et al. Systematic
181. Pampiglione G. Resuscitation after cardiac arrest. Electroen- review of early prediction of poor outcome in anoxic-ischaemic
cephalogr Clin Neurophysiol. 1962;14:294. coma. Lancet. 1998;352:1808–1812.
182. MacGillivray BB. The EEG in liver disease. In: Daly DD, ed. 197. Guérit JM, Amantini A, Amodio P, et. al. Consensus on the use of
Metabolic, Endocrine and Toxic Diseases. Handbook of Electroen- neurophysiological tests in the intensive care unit (ICU): elec-
cephalography and Clinical Neurophysiology. Vol 15. Amsterdam: troencephalogram (EEG), evoked potentials (EP), and elec-
Elsevier; 1976:26–50. troneuromyography (ENMG). Neurophysiol Clin. 2009;39:71–83.
183. Bell JA, Hodgson HJF. Coma after cardiac arrest. Brain. 1974; 198. Guérit JM. Electroencephalography: the worst traditionally rec-
97:361–372. ommended tool for brain death confirmation. Intensive Care
184. Thomassen A, Wernberg M. Prevalence and prognostic signifi- Med. 2007;33:9–10.
cance of coma after cardiac arrest outside intensive care and 199. Young GB. Clinical practice. Neurologic prognosis after cardiac
coronary units. Acta Anaesth. 1979;23:143–148. arrest. N Engl J Med. 2009;361:605–611.
185. Bates D, Caronna JJ, Cartlidge NEF, et al. A prospective study of 200. Chen R, Bolton CF, Young B. Prediction of outcome in patients
non-traumatic coma: methods and results in 310 patients. Ann with anoxic coma: a clinical and electrophysiological study. Crit
Neurol. 1977;2:211–220. Care Med. 1996;24:672–678.
186. Abramson NS, Meisel A, Safar P, Deferred consent. A new 201. Zandbergen EG, Hijdra A, Koelman JH, et. al. PROPAC Study
approach for resuscitation research on comatose patients. JAMA. Group. Prediction of poor outcome within the first 3 days of
1986;255:2466–2471. postanoxic coma. Neurology. 2006;66:62–68.
187. Edgren E, Hedstrand U, Kelsey S, et al., and BRCT 1 Study Group. 202. Jia X, Koenig MA, Shin HC, et. al. Quantitative EEG and neuro-
Assessment of neurological prognosis in comatose survivors of logical recovery with therapeutic hypothermia after asphyxial
cardiac arrest. Lancet. 1994;343:1055–1059. cardiac arrest in rats. Brain Res. 2006;1111:166–175.
188. Levy DE, Bates D, Caronna JJ, et al. Prognosis in nontraumatic 203. Rothstein TL, Thomas EM, Sumi SM. Predicting outcome in
coma. Ann Intern Med. 1981;94:293–301. hypoxic-ischemic coma. A prospective clinical and electrophysio-
189. Willoughby JO, Leach BG. Relation of neurological findings after logic study. Electroencephalogr Clin Neurophysiol. 1991;79:101–107.
cardiac arrest to outcome. Br Med J. 1974;3:437–439. 204. Pressler RM, Boylan GB, Morton M, et al. Early serial EEG in
190. Levy DE, Caronna JJ, Singer BH, et al. Predicting outcome from hypoxic ischaemic encephalopathy. Clin Neurophysiol. 2001;112:
hypoxic ischemic coma. JAMA. 1985;253:1420–1426. 31–37.
191. Earnest MP, Breckinridge JC, Yarnell PR, et al. Quality of survival 205. Synek VM. EEG abnormality grades and subdivisions of prog-
after out-of-hospital cardiac arrest; predictive value of early neu- nostic importance in traumatic and anoxic coma in adults. Clin
rologic evaluation. Neurology. 1979;29:56–60. Electroencephalogr. 1988;19:160–166.
192. Longstreth WT Jr, Diehr P, Inui TS. Prediction of awakening 206. Synek VM. Value of a revised EEG coma scale for prognosis after
after out-of-hospital cardiac arrest. N Engl J Med. 1983;308: cerebral anoxia and diffuse head injury. Clin Electroencephalogr.
1378–1382. 1990;21:25–30.
CHAPTER
The EEG in Patients with Migraine and
Other Forms of Headache
ERNST NIEDERMEYER AND DONALD L. SCHOMER
24
P
atients with headaches are usually referred to the elec- field, migraine has remained a poorly understood disorder. A
troencephalography (EEG) laboratory to rule out under- plethora of clinical data is found in the work of Sacks (4).
lying cerebral pathology rather than for a clarification of The clinical symptomatology of the migrainous attack is
the type of headache. This type of referral has become less fre- well known. In the classical form, visual symptoms herald the
quent with the greater availability of modern neuroimaging. attack; there are scintillating scotoma with teichoscopy and
Headache is one of the most common complaints. As a other forms of visual field cuts. Within a short time (about 5 to
symptom, it may herald a wide variety of infectious, neoplastic, 20 minutes), this stage is supplanted by headache, which is
and vascular intracranial lesions, but it also may be a sign of var- mostly unilateral, with shifting lateralization from attack to
ious dysfunctions impinging on neural, vascular, and muscular attack in most cases. This is accompanied by severe nausea,
structures. It may arise from the vicinity of the cranial cavity or vomiting, irritability, and photophobia. This stage lasts for
even from distant structures. Metabolic, toxic, and hormonal hours or a full day. Wolff (5) has ascribed the initiating visual
disturbances are further causes. In other words, headache is a symptoms to intracerebral local vasoconstriction and the ensu-
challenge for the diagnostic acumen of neurologists and other ing phase of headache and nausea to an abnormal degree of
specialists. It has been stated that about 20% of the U.S. popula- extracranial vasodilation, which can easily be palpated along
tion complains of headache, and about half of them receive the temporal artery on the painful side.
some form of symptomatic medical treatment (1). Table 24.1 A genetic predisposition is present or even pronounced, but
shows a classification of the types of headache. New criteria of the mode of genetic transmission is not fully understood. The
classification have been proposed by Silberstein et al. (3). attacks tend to start in adolescence; in childhood, attacks of
abdominal pain may be precursors of migraine. It has been
thought that a fall of the plasma serotonin level (6) plays a cru-
MIGRAINE (CLASSICAL AND cial role in the physiopathogenesis of the migrainous attack, but
COMPLICATED FORMS) this concept has not been generally accepted. Allergic (anti-
gen–antibody) reactions, free fatty acids, and prostaglandin E
General Considerations and Clinical Features are also believed to be involved in migraine (7). Activation of 5-
Migraine has been known to humanity for ages; a Sumerian hydroxytryptamine receptors has been stressed by Fozard (8).
poem written 5000 years ago gives an account of this disorder. According to Moskowitz (9), migraine is caused by a distur-
In spite of a remarkable upsurge of research interest in this bance of the “trigeminovascular system” (connections between

Tabl e 2 4 . 1

Classification of Headache

Vascular Headache Muscle Contraction Traction and Inflammatory


(Psychogenic) Headache Headache
Migraine Cervical osteoarthritis Mass lesions (tumors, edema, hematomas, cerebral hemorrhage)
1. Classic
2. Common
3. Hemiplegic Complicated migraine
4. Ophthalmoplegic
Cluster (histamine) Chronic myositis Diseases of the eye, ear, nose, throat, teeth
Toxic vascular Depressive equivalents and Infection
conversion reactions
Hypertensive Arteritis, phlebitis; cranial neuralgias; occlusive vascular disease

Modified from Dalessio, DJ. Classification and mechanism of migraine. Headache. 1979;19:114–120.

457
458 Part III ■ Clinical EEG: General

trigeminal ganglia and cerebral blood vessels) involving the migraine and EEG were frequently flawed by sampling prob-
neurotransmitter peptide, substance P. Experimentally, “neu- lems and heterogeneous populations of migrainous persons.
roinflammation” was produced in animals by electrical stimu- The contrast between various reports on the EEG in migraine
lation of the trigeminal ganglion causing the release of sensory in the interval between attacks is due to (i) composition of mate-
neuropeptides from nerve terminals (the model of “neurogenic rial (adults vs. children, inclusion or exclusion of hemiplegic
inflammation” (10)). Biggs and Johnson (11) have placed spe- cases), (ii) different criteria for normality and abnormality in
cial emphasis on the adrenergic system and its role in migraine the investigators’ EEG interpretation, and (iii) difficulties in the
pathogenesis. A unified neurogenic concept of migraine has delineation of migraine as a nosologic entity (inclusion or strict
been proposed by Diamond and Dalessio (12). exclusion of cluster headaches or symptomatic forms of
Not all cases of migraine are due to an inherited dysfunction; migraine with cerebral pathology). Keeping all this in mind, it is
neuropathologic processes such as arteriovenous malformations still difficult to understand the disparity of the reports.
of neurosyphilis are known as cases of “symptomatic migraine.” The predominance of normal-interval EEG records was
This differential diagnosis may occasionally become difficult, stressed by Ulett et al. (27), Jung (28), Becher (29), Krischek
because migraine attacks are capable of proceeding to a state of (30), Wissfeld and Neu (31), Bille (32), and Gibbs and Gibbs
ischemic infarction (13,14) and of producing regional computed (33). The work of other authors places the emphasis on a variety
tomography (CT) scan changes (15). In regional cerebral blood of abnormalities. Heyck (34,35) found mainly “hypersynchro-
flow studies using xenon-133 intra-arterially, reduced blood flow nous bursts” and occasional focal slowing. Weil (36,37) noted
was demonstrable during migraine attacks starting posteriorly pronounced delta responses to hyperventilation. Various types
and very slowly spreading to the rolandic region (16). According of abnormalities were noted by Dow and Whitty (38) and Selby
to Hansen et al. (17) and Olesen (18), spreading depression is and Lance (39). A high incidence of abnormal EEG records was
considered a useful model of migraine aura and presumably also also emphasized by Barolin (40), Gschwend (41), and Pithova
for the subsequent headache. This concept was reemphasized by (42). Almost equal numbers of normal and abnormal records
Olesen (19), especially on the basis of the positron emission (with about 45% abnormal tracings) were reported in the exten-
tomography (PET) scan observation (oxygen-15-labeled water) sive work of Smyth and Winter (43). The reported abnormali-
of Woods et al. (20). There is good evidence of hypoperfusion ties, however, were predominantly mild to moderate, with some
within the occipital lobe (20), which can just as well (if not bet- bursts, slowing, or sharp transients. With the use of computer
ter) be used as supportive of the vascular concept. frequency analysis, Jonkman and Lelieveld (44) demonstrated
Special involvement of the central visual system is a very abnormal interval EEG findings in 55% of migrainous patients.
common feature of classical migraine with visual initiation. According to Drake et al. (45), the EEG of patients with migraine
Huang et al. (21) have shown with the use of functional mag- does not differ significantly from the EEG of normal individu-
netic resonance imaging (fMRI) the excessive responses to als. This is essentially congruent with my personal views.
visual stimuli and a particular sensitivity to a pattern of regu- Intermittent photic stimulation often shows an occipital
larly spaced parallel lines of stripes (see also Lashley (22)). driving response extending into the range above 20 flashes/sec
Whoever reads the original experimental techniques used in (“H response” after Golla and Winter (46)); according to Smyth
the production of spreading depression (23,24) will have nag- and Winter (43), this is almost specific for migraine. This has
ging doubts concerning the appropriateness of the spreading- been substantiated by Slater (47). Personal observations essen-
depression model for a neurogenic migraine concept. The mode tially support this view. Further substantiation of these findings
of elicitation implies all sorts of mechanical and chemical has been provided by Simon et al. (48) with the use of spectral
trauma to the brain tissue; electrocorticographic recording (24) analysis during photic stimulation (Fig. 24.1).
shows flattening of the record followed by several recurrent EEG findings in the migraine attack range from normal to
prominent spikes (against a flat background) and another phase mildly abnormal (alpha depression) in the initiating ophthalmic
of flattening before the baseline character of the record is phase; even severely abnormal findings have been reported in
restored. It is difficult to imagine that similar electrical processes special cases (49–51). Based on his large material, Heyck (34)
would occur in migrainous human beings. The primordial found normal tracings in the ophthalmic vasoconstrictive as
nature of vasomotor changes according to H. G. Wolff’s (5) orig- well as in the headache-nausea phase. Schoenen et al. (52) found
inal theory seems to be a lot more plausible. reduced alpha activity over one occipital region in 19 out of 22
On the basis of data derived from animal experiments patients recorded during an attack of common migraine. In the
(Wistar rats), Ebersberger et al. (25) doubt that spreading light of these observations, the statement that “EEGs have
depression initiates migraine. almost always been normal in migraineurs during attacks” (53)
might be slightly exaggerated, but EEG abnormalities should be
Electroencephalographic Findings viewed as exceptions.
The literature in this field is very confusing because almost A neuronal dysfunction as the cause of migraine was
equal numbers of reports stress the predominance of normal assumed by Soysal et al. (54) on the basis of significantly pro-
and abnormal tracings. Relatively few records have been longed P100 latencies of visual evoked potentials in the interval
obtained during the attacks; these data are discussed later. Daly between attacks. On the other hand, EEG abnormalities were
and Markand (26) have pointed out that previous studies of observed in only 4 of the 13 patients.
Chapter 24 ■ The EEG in Patients with Migraine and Other Forms of Headache 459

has been pointed out that in familial hemiplegia the lateralization


of the affected hemisphere remains unchanged in every attack
and is the same in all involved family members. This is not con-
gruent with a personal observation. EEG studies in familial hemi-
plegia show a varying degree of slowing over the brain’s affected
side.
The contingent negative variation (CNV) has been used for
the differential diagnosis of headaches (62). Ahmed (63) clearly
demonstrated an enhanced CNV in patients with classic and
common migraine; their CNV was moderately larger than in
patients with tension headaches and much larger than in nor-
mal controls. These statements pertain to the pain-free interval
between migraine attacks. The CNV changes are believed to
reflect catecholamine hyperactivity. According to CNV
research, this slow potential measures expectancy, attention,
preparation, and motivation. When one considers that, accord-
ing to Ahmed (63), the enlargement of the CNV would persist
after successful antimigraine treatment, then one wonders if
enhanced motivation and a powerful desire to perform tasks
perfectly are personality traits of migrainous persons.

ATYPICAL FORMS OF MIGRAINE


Abdominal manifestations of migraine are relatively common in
children. Headache is usually absent in these attacks, whereas
abdominal pain is in the foreground. Such attacks may last for
hours. Temporary agitation and obnubilation during the attack
have been noted by Lérique-Koechlin and Mises (64).
In childhood migraine, normal EEG records are the rule
(89%), but “benign” focal spikes, mostly rolandic, were found in
9% (65). Kellaway et al. (66) stressed the high incidence of 14
and 6/sec positive spikes in the sleep records of these children.
During the attack of abdominal pain, the EEG is unremarkable
(personal observations). Gibbs and Gibbs (33) separate abdom-
inal pain attack (with frequent 14 and 6/sec positive spike dis-
charges in the interval) from migraine with normal-interval
tracings. Lérique-Koechlin and Mises (64) reported a very high
Figure 24.1 A: A 31-year-old woman with a history of classical migraine incidence of paroxysmal EEG changes in children with a history
(experiences flashing lights and also some left-sided numbness). EEG of abdominal pain attacks. Moore (67) introduced the term
obtained in interval. Note good occipital photic driving response to a flash abdominal epilepsy for such cases of acute abdominal pain, but
rate of 22/ sec (“H response”). The right frontal spiky discharge is artifac- this has not been widely accepted. This term is rather mislead-
tual. B: Same patient. Very good occipital lambda activation presentation ing, and its use should be discouraged. In migrainous children,
of pattern vision test tables. temporal lobe-type seizures have been described (68). These
children experienced strange sensations or misperceptions; the
EEG in most cases showed sharp activity over the temporal
When the migraine attack is complicated by mild hemiparetic region. “Benign paroxysmal vertigo of childhood” has been
or dysphasic deficits (migraine accompagnŽe), the EEG may found to be a migrainous equivalent (69).
remain normal (55). In cases with pronounced hemiplegia and Basilar migraine represents a syndrome described by
aphasia, there is good evidence of delta and theta activity over the Bickerstaff (70), consisting of a sudden transient blurring of
affected hemisphere (34,56–59). The delta activity over the vision or blindness, vertigo, gait ataxia, dysarthria, acroparesthe-
affected hemisphere may be very impressive (see cases of Isler sias, and pulsatile occipital headache with vomiting. Even syn -
(60)). The neurologic deficit subsides within days (sometimes copal states and loss of consciousness may occur (71). Basilar
within weeks), and the focal or lateralized slowing my also linger migraine manifestations are more common in older children,
on for some period of time (Fig. 24.2). Cases of familial hemi- adolescents, and young adults; females are more often affected. In
plegic migraine have been reported by Whitty (14), Rosenbaum adulthood, these attacks may be supplanted by attacks of classical
(59), Bradshaw and Parsons (56), and Müller and Müller (61). It or common migraine. According to Caplan (72), basilar migraine
460 Part III ■ Clinical EEG: General

Figure 24.2 A: A 13-year-old boy with migraine attacks. EEG recorded during a migraine attack with moderate right hemi-
paresis and global aphasia. Note marked delta activity over left occipital-posterotemporal region. B: EEG taken 5 days later.
Clinically normal, EEG normalized.

is not always benign. It affects both sexes and a wider age range; According to Ramelli et al. (79), the EEG of children (11
it also may be associated with strokes. to 13 years) with basilar artery migraine showed diffuse
The EEG literature is meager in this domain; a case recorded subdelta–delta activity during the attack and occipital
by Slater (47) during a presumed attack showed diffuse 1.5 to delta–theta activity hours afterward. The authors warn against
4/sec activity with subsequent normalization. This patient, how- concepts of structural lesions (infarction, inflammation) and
ever, was 46 years old; this age could cast some doubt on the diag- presume a temporary disfunction.
nosis. Lapkin et al. (73) reported two cases (ages 12 and 10) with Cernibori and Bouquet (80) observed children ages 2 to 14
diffuse and chiefly posterior slowing in the 1.5 to 2/sec and 3 to years with episodes of loss of consciousness ranging from 1 hour
4/sec ranges, respectively, during the attack. This activity van- to several days; these episodes were attributed to basilar artery
ished with serial recordings. Camfield et al. (74) observed four migraine of childhood. Diffuse or lateralized slow activity was
adolescents (two girls and two boys) with presumed basilar noted in the EEG, which improved over the ensuing days.
migraine. These cases were complicated by epileptic seizures Slightly abnormal interval EEG tracings were found in 4 of 12
(mostly grand mal, but also focal motor). The EEG showed very children. Episodic comatose states have also been reported by
prominent spike activity and slowing accentuated over the poste- Ganji et al. (81).
rior regions. All of these patients had a strongly positive history
of classical or common migraine. Similar paroxysmal findings Status Migrainosus
over posterior regions were reported by Panayatopoulos (75). On the basis of personal observations, there is good reason to
Another form with predominant beta activity during the attack presume that prolonged or constantly recurrent migraine
was delineated by Parain and Samson-Dollfus (76). Gastaut (77) attacks are the result of severe mental–emotional stress. In this
has cast much doubt on the observations of Camfield et al. (74); condition, EEG findings are bland and noncontributory.
he feels that these patients were suffering from benign occipital Whatever neurologic–organic basis may be demonstrated
lobe epilepsy (see Chapter 27). Simple partial status epilepticus in in migraine, the involvement of psychogenic factors must not
the occipital lobe can be misdiagnosed as migraine (78). be counted out. Evidence of a link between migraine and
Tabl e 2 4 . 2

Differentiation of Migrainous and Certain Epileptic Conditions

Scintillating Visual Hallucination, Nausea


Condition Occurrence Sex Age Scotoma (Elementary, Figurative) Headache (Vomiting)
Common Very common Probably females Mostly after age 0 0
migraine preponderant 15 years
Classic Common Mostly males if without Mostly after age ? 0– 0–
migraine headache-nausea 15 years
Basilar artery Moderately Probably females only 15–35 years 0– 0– 0– 0–
migraine common
Migraine- Very rare ? (? mainly females) ? 15–40 years (immediately ? – (after 0– (after
triggered followed by grand mal) grand mal) grand mal)
epilepsy
Benign Rare No sex 5–40 years 0 – – (after
occipital preponderance (after seizure) seizure)
lobe epilepsy
Benign Very rare ? (? mainly females) 5–20 years for 0 (for epileptic), 0 0– 0–
rolandic epileptic, past 15 (for migrainous attacks) (after seizure), (after seizure),
epilepsy- for migrainous
migraine manifestations (in migraine) (in migraine)
Epileptic EEG (During
Syncope Seizures EEG (Interval) Episodes) Therapy Prognosis
0 reduction 0 Mostly normal Mostly normal Antimigrainous, Mostly very good (albeit
stress reduction no real cure)
0 (? ) 0 Mostly normal Mostly normal Antimigrainous, Mostly very good (albeit
stress reduction no real cure)
0– 0– Normal to slightly Normal to slightly Antimigrainous, Mostly very good
abnormal (nonspecific) abnormal (with stress reduction, (? spontaneous cure
changes due to may require in middle adulthood)
syncope or antiepileptics
convulsion)
0 Normal Typical EEG of a Probably preventive Unclear
(grand mal) tonic–clonic antimigrainous therapy,
convulsion antiepileptics ineffective
0 Abnormal with Abnormal with Antiepileptics Mostly good
(mostly visual, recurrent posterior bilateral posterior
also others) spike-waves or spikes spike-waves
0 (focal Normal or abnormal Probably abnormal Antiepileptics, in later Probably very good
motor) with rolandic spikes, during focal motor phase antimigrainous, (albeit no real cure
in adulthood normal attacks, normal stress reduction for migraine)

461
during migraine
462 Part III ■ Clinical EEG: General

neuroticism has been reported by Breslau et al. (82). Let us by Heyck and Hess (99). A physiopathogenetic basis of such
assume that, while many migrainous persons are in full control dysfunctional states with headache is still obscure.
of such subthreshold mechanisms, others may lose their control A rather nonspecific type of headache (“fullness of head,
temporarily (under stress) or perhaps even permanently. Olesen’s pressure, heat, pounding”) was found to be an ictal epileptic
(19) concept of a “clean neurologic” migraine—uncontaminated manifestation in the limbic portion of the right temporal lobe
with psychogenic features—cannot be upheld in view of the recorded with depth electrodes (100). These patients benefited
clinical facts. from surgical resection.
It was found that EEG studies done with spectral analysis in
RELATIONSHIPS BETWEEN MIGRAINE patients with tension headaches did not differ from normal
AND EPILEPTIC SEIZURE DISORDERS persons (and were also not significantly different from the EEG
of migrainous patients) (45).
A combination of migraine and epileptic seizure disorder may
occur, but it is uncommon (40,83,84). A true link between both ACKNOWLEDGMENT
disorders is highly debatable (85–87). There is some reason to
support the view of Camfield et al. (74) that, under certain cir- The assistance of Dr. Fowzia Siddiqui is gratefully acknow-
cumstances, migraine can trigger an epileptic attack. This subject ledged.
has been reviewed by Hess (88), who presumes that headache
may be a secondary symptom in patients with epileptic seizure
REFERENCES
disorder. There is no cogent need to establish a special form
called “dysrhythmic migraine” (36,37). 1. Diamond S. Headache: its diagnosis and management (introduc-
The entire subject of migraine and epilepsy was reinvesti- tion to symposium). Headache. 1979;19:113.
gated in extensive work edited by Andermann and Lugaresi (89) 2. Dalessio DJ. Classification and mechanism of migraine. Headache.
and in the overview of Andermann and Andermann (90). 1979;19:114–120.
An entity named “migraine-triggered epilepsy” has been 3. Silberstein SD, Lipton RB, Sliwinski M. Classification of daily and
described by Niedermeyer (91–93). Typical grand mal seizures near-daily headaches: field trial of revised IHS criteria. Neurology.
1996;47:871–875.
occur after a very short visual initiation; migrainous headache
4. Sacks O. Migraine. Stuttgart: Kohlhammer. (Original English ver-
and nausea follow the seizure. The EEG is mostly normal in sion: Migraine. The Evolution of a Common Disorder. London:
the interval and the response to antiepileptic medication Faber & Faber; 1985.)
leaves much to be desired. Postepileptic headaches are quite 5. Wolff HG. Headache and Other Head Pain. New York: Oxford
common and there is also a possibility of “epilepsy-triggered University Press; 1948.
migraine” (94). 6. Anthony M, Hinterberger H, Lance JW. Plasma serotonin in
On the basis of very extensive data collected (1957 adult migraine and stress. Arch Neurol (Chicago). 1967;16:544–552.
probands with epilepsy), Ottman and Lipton (95) were unable 7. Anthony M. Plasma fatty acids and prostaglandin E in migraine
to support the theory of a shared genetic susceptibility for and stress. Headache. 1970;16:58.
migraine and epileptic disorders. Table 24.2 demonstrates the 8. Fozard JR. Proceedings of the International Headache Congress,
differentiation of migrainous and certain epileptic conditions. Florence, 1980. Clin Psychiatry News. 1980;8(9).
9. Moskowitz MA. The neurobiology of vascular head pain. Ann
Neurol. 1984;16:157–168.
THE EEG IN OTHER TYPES OF HEADACHE 10. Moskowitz MA, Nozaki K, Draig RP. Neocortical spreading depres-
sion provokes the expression of c-fos-protein-like immunoreactivity
The EEG in nonmigrainous forms of headaches has been thus within trigeminal nucleus caudalis via trigeminovascular mecha-
far a barren field; one is unable to correlate any type of nisms. J Neurosci. 1993;13:1167–1177.
headache with some type of EEG change during the attacks or 11. Biggs MJ, Johnson ES. The autonomous nervous system and
in the interval. Cluster headache is a well-defined entity, but has migraine pathogenesis. In: Amery WK, Van Nueten JM, Wauquier
no special EEG correlate. Hyperventilation-related headache A, eds. The Pharmacological Basis of Migraine Therapy. London:
associated with EEG slowing has been reported by Sbrascini Pitman; 1984:99–107.
and Bassi (96). Hypnic headache has been described by Raskin 12. Diamond S, Dalessio DJ. The Practicing Physician’s Approach to
(97) and related to REM sleep; the EEG is normal (98). Headache. Baltimore: Williams & Wilkins; 1982.
In general, patients with habitual headaches and no organic 13. Schumacher GA, Wolff HG. Experimental studies on headache.
disease may show EEG patterns that are believed to reveal some Arch Neurol Psychiatry (Chicago). 1941;45:199–214.
14. Whitty CWM. Familial hemiplegic migraine. J Neurol Neurosurg
degree of “neuronal hyperexcitability”; this is a vague term
Psychiatry. 1953;16:172–177.
without any precise scientific foundation, but with some merit 15. Skinhoj E, Hoedt-Rasmussen K, Paulson OB, et al. Regional cere-
in the domain of medical practice. Rolandic mu rhythm is quite bral blood flow and its autoregulation in patients with transient
often found in patients referred for headaches without demon- focal ischemic attacks. Neurology (Minneapolis). 1970;20:485–493.
strable organic cause. Other patients show mildly paroxysmal 16. Lauritzen M, Skyhoj Olsen T, Lassen NA, et al. Changes in regional
flicker responses; still others show 14 and 6/sec positive spikes cerebral blood flow during the course of classic migraine attacks.
or even categorical EEG abnormalities such as psychomotor Ann Neurol. 1983;13:633–641.
variant pattern and 6/sec spike waves. The possibility of vasomo- 17. Hansen AJ, Lauritzen M, Tfeldt-Hansen P. Spreading cortical depres-
tor headaches as a symptom of larval epilepsy has been discussed sion and antimigrainous days. In: Amery WK, Van Neuten JM,
Chapter 24 ■ The EEG in Patients with Migraine and Other Forms of Headache 463

Wauquier A, eds. The Pharmacological Basis of Migraine Therapy. 43. Smyth VOG, Winter AL. The EEG in migraine. Electroencephalogr
London: Pitman; 1984:161–170. Clin Neurophysiol. 1964;16:194–202.
18. Olesen J. Conclusions and prospects for the future. In: Olesen J, ed. 44. Jonkman EJ, Lelieveld MHJ. EEG computer analysis in patients
Migraine and Other Headaches. New York: Raven Press; 1991: with migraine. Electroencephalogr Clin Neurophysiol. 1981;52:
347–349. 652–655.
19. Olesen J. Understanding the biologic basis of migraine. N Engl J 45. Drake ME, Huber SJ, Pakalnis A, et al. Computerized EEG spectral
Med. 1994;331:1713–1714. analysis in migraine and tension headaches. J Clin Neurophysiol.
20. Woods RP, Iacoboni M, Mazziota JC. Brief report: bilateral spread- 1987;4:301.
ing cerebral hypoperfusion during spontaneous migraine 46. Golla FL, Winter AL. Analysis of cerebral responses to flicker in
headache. N Engl J Med. 1994;331:1689–1692. patients complaining of episodic headache. Electroencephalogr Clin
21. Huang J, Cooper TG, Santana B, et al. Visual distortion provoked Neurophysiol. 1959;11:539–549.
by a stimulus in migraine associated with hyperneuronal activity. 47. Slater KH. Some clinical and EEG findings in migraine. Brain.
Headache. 2003;43:664–671. 1968;91:85–98.
22. Lashley KS. Patterns of cerebral integration indicated by scotomas 48. Simon RH, Zimmerman A, Tasman A, et al. Spectral analysis of
of migraine. Arch Neurol Psychiatry (Chicago). 1941;46:331–339. photic stimulation in migraine. Electroencephalogr Clin Neurophysiol.
23. Leão AAP. Spreading depression of activity in the cerebral cortex. 1982;53:270–276.
J Neurophysiol. 1944;7:359–390. 49. Kugler J. Elektroenzephalographie und Beziehungen zur Epilepsie.
24. Leão AAP. Spreading depression. In: Purpura DP, Penry JK, Tower In: Soyka D, ed. Die MigrŠne. Dusseldorf: Labaz; 1979:51–62.
D, et al., eds. Experimental Models of Epilepsy. New York: Raven 50. Scollo-Lavizzari G. Das Elektroenzephalogramm bei der Migräne.
Press; 1972:173–196. Schweiz Rdsch Med (Praxis). 1975;64:234–237.
25. Ebersberger A, Schaible HG, Averbeck B, et al. Is there a correlation 51. Westmoreland B. EEG in the evaluation of headaches. In: Klass DW,
between spreading depression, neurogenic inflammation, and noci- Daly DD, eds. Current Practice of Clinical Electroencephalography.
ception that might cause migraine headache? Ann Neurol. 2001;49: New York: Raven Press; 1978;381–394.
7–13. 52. Schoenen J, Jamart B, De Pasqua V, et al. Mapping of EEG and
26. Daly DD, Markand ON. Focal brain lesions. In: Daly DD, Pedley auditory event-potentials in migraine. Electroencephalogr Clin
TA, eds. Current Practice of Clinical Electroencephalography. 2nd Neurophysiol. 1990;75:S134(abst.).
ed. New York: Raven Press; 1990:35–370. 53. Gorman MJ, Welch KMA. Cerebral blood flow and migraine. In:
27. Ulett GA, Evans D, O’Leary JL. Survey of EEG findings in 1,000 Phillips JW, ed. The Regulation of Cerebral Blood Flow. Boca Raton:
patients with chief complaint of headache. Electroencephalogr Clin CRC Press; 1993:399–410.
Neurophysiol. 1952;4:463–470. 54. Soysal A, Atay T, Ozturk M, et al. Pattern reversal visual evoked
28. Jung R. Neurophysiologische Untersuchungsmethoden. In: potentials and EEG in migraine with and without visual aura.
Handbuch der Inneren Medizin. 4th ed., Vol. 5/1. Berlin: Springer; J Neurol Sci (Turkish). 2001;18:1–8.
1953:1206–1314. 55. Farkas V, Szeg L, Kohlhéb O. Hemiplegic migraine in childhood.
29. Becher F. Hirnelektrische und elektromyographische. Unter - Differential diagnosis and EEG aspects. Electroencephalogr Clin
suchungen beim Migränesyndrom. Dtsch Z Nervenheilk. 1955; Neurophysiol. 1985;61:4P–5P(abst.).
172:556. 56. Bradshaw P, Parsons M. Hemiplegic migraine: a clinical study.
30. Krischek J. Elektroenzephalographische Befunde bei Migräne. J Med. 1965;34:65–85.
Dtsch Z Nervenheilk. 1956;175:43. 57. Degen R, Degen HE, Palm D, et al. Die Migraine hémiplégique im
31. Wissfeld E, Neu O. Über die EEG-Verändeungen bei Migräne und Kindesalter. Dtsch Med Wochenschr. 1980;105:640–645.
die Bedeutung occipitaler Delta-Wellen im EEG. Nervenarzt. 1960; 58. Heron JR. Migraine and cerebrovascular disease. Neurology
31:418. (Minneapolis). 1966;16:1097–1104.
32. Bille B. Migraine in school children. Acta Paediatr Suppl (Uppsala). 59. Rosenbaum HE. Familial hemiplegic migraine. Neurology
1962;51:136. (Minneapolis). 1960;10:164–170.
33. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol. 3. 60. Isler W. Akute Hemiplegien und Hemisyndrome im Kindesalter.
Reading: Addison-Wesley; 1964. Stuttgart: Thieme; 1969.
34. Heyck H. Neue BeitrŠge zur Klinik und Pathogenese der MigrŠne. 61. Müller D, Müller J. Die familiäre hemiplegische Migräne. Z Artzl
Stuttgart: Thieme; 1956. Fortibild. 1977;71:763–767.
35. Heyck H. Der Kopfschmerz. Stuttgart: Thieme; 1958. 62. Martens de Nordhout A, Timsit-Bertheir M, Timsit M, et al.
36. Weil AA. EEG findings in a certain type of psychosomatic headache: Contingent negative variation in headache. Ann Neurol. 1986;19:
dysrhythmic migraine. Electroencephalogr Clin Neurophysiol. 1952; 78–80.
4:181–186. 63. Ahmed I. Contingent negative variation in migraine: effect of beta
37. Weil AA. Observation on dysrhythmic migraine. J Neurol Ment blocker therapy. Clin Electroencephaologr. 1999;30:21–23.
Dis. 1962;134:277–281. 64. Lérique-Koechlin A, Mises J. L’EEG dans une manifestation parox-
38. Dow DJ, Whitty CW. Electroencephalographic changes in migraine. ystique non-épileptique de l’enfant: la migraine. Electroencephalogr
Review of 51 cases. Lancet. 1947;2:52–54. Clin Neurophysiol. 1964;16:203–204.
39. Selby G, Lance JW. Observations on 500 cases of migraine and allied 65. Kinast M, Lueders H, Rothner AD, et al. Benign focal epileptiform
vascular headache. J Neurol Neurosurg Psychiatry. 1960;23:23–32. discharges in childhood migraine (BFEDC). Neurology (New York).
40. Barolin GS. Migraines and epilepsies—a relationship? Epilepsia 1982;32:1309–1311.
(Amsterdam). 1966;7:53–66. 66. Kellaway P, Crawley JW, Kagawa N. Paroxysmal pain and auto-
41. Gschwend J. EEG-Befunde und ihre Interpretation bei einfacher nomic disturbances of cerebral origin. A specific electroclinical
Migräne. J Neurol (Berlin). 1972;201:279–292. syndrome. Epilepsia (Amsterdam). 1960;1:466–483.
42. Pithova B. Clinico-EEG correlation in migraine. Electroencephalogr 67. Moore MT. Paroxysmal abdominal pain. A form of symptomatic
Clin Neurophysiol. 1983;55:31P(abst.). epilepsy. JAMA. 1945;129:1233–1240.
464 Part III ■ Clinical EEG: General

68. Seshia SS, Reggin JD, Stanwich RS. Migraine and complex seizures 84. Giardina M, Sideri S. Migraine and epilepsy. Electroencephalogr
in children. Epilepsia (New York). 1985;26:232–236. Clin Neurophysiol. 1985;60:71P(abst.).
69. Koehler B. Benign paroxysmal vertigo of childhood: a migraine 85. Basser LS. The relation of migraine and epilepsy. Brain. 1969;92:
equivalent. Eur J Pediatr. 1980;134:149–151. 285–300.
70. Bickerstaff ER. Basilar artery migraine. Lancet. 1961a;1:15–17. 86. Lance JW, Anthony M. Some clinical aspects of migraine. A
71. Bickerstaff ER. Impairment of consciousness in migraine. Lancet. prospective survey of 500 patients. Arch Neurol (Chicago). 1966;
1961b;2:1057–1059. 15:356–361.
72. Caplan LR. Migraine and vertebrobasilar ischemia. Neurology. 87. Lees R, Watkins SM. Loss of consciousness in migraine. Lancet.
1991;41:55–61. 1963;2:647–650.
73. Lapkin ML, French JH, Golden GS, et al. The electroencephalogram 88. Hess R. Epilepsie und Kopfschmerzen. A. EEG-EMG. 1977;8:
in childhood basilar artery migraine. Neurology (Minneapolis). 1977; 125–136.
27:580–583. 89. Andermann F, Lugaresi E, eds. Migraine and Epilepsy. Boston:
74. Camfield PR, Metrakos K, Andermann F. Basilar migraine, Butterworth; 1987.
seizures and severe epileptiform EEG abnormalities. Neurology 90. Andermann F, Andermann E. Migraine and epilepsy with special
(Minneapolis). 1978;28:584–588. reference to the benign epilepsies of childhood. In: Degen R,
75. Panayatopoulos CP. Basilar migraine, seizures and severe epilep- Dreifuss FE, eds. Benign Localized and Generalized Epilepsies of
tic EEG abnormalities. Neurology (Minneapolis). 1980;30: Early Childhood. Amsterdam: Elsevier; 1992:207–214.
1122–1125. 91. Niedermeyer, E. Epilepsy Guide. Baltimore: Urban & Schwarzenberg;
76. Parain D, Samson-Dollfus D. Electroencephalograms in basilar 1983.
artery migraine. Electroencephalogr Clin Neurophysiol. 1984;58: 92. Niedermeyer, E. The Epilepsies. Baltimore: Urban & Schwarzenberg;
392–399. 1990.
77. Gastaut H. A new type of epilepsy: benign partial epilepsy of child- 93. Niedermeyer, E. Migraine-triggered epilepsy. Clin Electroencephalogr.
hood with occipital spike-waves. Clin Electroencephalogr. 1982;13: 1993;24:37–43.
13–22. 94. Jacobs J, Goadsby PJ, Duncan JS. Use of sumatriptan in post-ictal
78. Walker MC, Smith SJM, Sisodiya SM, et al. Case of simple partial migraine headache. Neurology. 1996;47:1104.
status epilepticus in electrophysiological, and magnetic resonance 95. Ottman R, Lipton RB. Is the comorbidity of epilepsy and migraine
imaging characteristics. Epilepsia. 1995;36:1233–1236. due to shared genetic susceptibility? Neurology. 1996;47:918–924.
79. Ramelli GP, Sturzenegger M, Donati F, et al. EEG findings during 96. Sbrascini S, Bassi P. Headache and slow hypersynchronization of
basilar migraine attacks in children. Electroencephalogr Clin the EEG during hyperventilation. Electroencephalogr Clin
Neurophysiol. 1998;107:374–378. Neurophysiol. 1983;55:3P(abst.).
80. Cernibori A, Bouquet F. Loss of consciousness during basilar 97. Raskin NH. The hypnic headache syndrome. Headache. 1988;28:
artery migraine attack in childhood: EEG and clinical studies. 534–536.
Electroencephalogr Clin Neurophysiol. 1984;58:72P(abst.). 98. Evers S, Goadsby PJ. Hypnic headache. Neurology. 2003;60:
81. Ganji S, Hellman S, Stagg S, et al. Episodic coma due to acute basi- 905–909.
lar artery migraine: correlation of EEG and brain stem auditory 99. Heyck H, Hess R. Vasomotorische Kopfschmerzen als Symptom
evoked potentials. Clin Electroencephalogr. 1993;24:44–48. larvierter Epilepsien. Schweiz Med Wochenschr. 1955;85:573–575.
82. Breslau N, Chilcoat HD, Andreski P. Further evidence on the 100. Laplante P, Saint-Hilaire JM, Bouvier G. Headache as an epilep-
link between migraine and neuroticism. Neurology. 1996;47: tic manifestation—two cases studied with depth electrodes.
663–667. Electroencephalogr Clin Neurophysiol. 1982;54:36P–37P(abst.).
83. Alvarez WC. Migraine plus epilepsy. Neurology (Minneapolis). 1959; 101. Olesen J, Tfelt-Hansen P, Welch DMA, eds. The Headaches. New
9:487–491. York: Raven Press; 1993.
Part IV Clinical EEG in Epilepsy and Related Disorders
CHAPTER
Seizures and Epilepsies in the
Preterm and Term Neonate
EMMA LAURETA, ELI M. MIZRAHI, AND SOLOMON L. MOSHÉ
25
NEONATAL SEIZURES Terminology
In the term infant, the neonatal period is defined as the first 28
Historical Aspects
days of life. In the preterm infant, this period extends to the 44
Initial investigations and attempts to characterize and classify completed weeks gestational age. It is a period of increased vul-
neonatal seizures began in the 1950s and 1960s, as neonatal nerability to anoxic and metabolic stress, and most seizures dur-
electroencephalography (EEG) began to emerge and well before ing this period are acute reactive seizures, as opposed to
video-EEG recordings became established as diagnostic tools manifestations of neonatal epilepsy––although the latter may
(1–4). Early investigators relied on clinical observation or occur with early neonatal seizures being the first of a chronic
observation supplemented by interictal EEG recordings (5,6). condition (19). However, the term “epileptic” is used in current
Later, bedside EEG recordings of infants who were experiencing classification systems to denote presumed underlying pathophys-
seizures combined with cinematography, improved correlation iology––seizures generated by hypersynchronous cortical neu-
of observed clinical phenomenon with electrographic activity ronal discharges as demonstrated by temporally related EEG
(7–10). changes. This is in contrast to “nonepileptic” seizures that occur
These pioneering French investigators observed that seizures without any EEG correlate and are of unclear pathophysiology,
in the neonate had unique clinical features that distinguished most likely based in reflex physiology. Focal clonic, focal tonic
them from seizures generated by more mature brains. For events, some myoclonic events and spasms are most often epilep-
example, generalized tonic–clonic seizures were found not to tic. Generalized tonic posturing and motor automatisms are
occur (5,10–12). The more common focal clonic seizures were often purely clinical. Seizures recorded in the absence of clinical
often asynchronous if they occurred bilaterally and did not activity are termed “electrical only or electrographic seizures.”
spread in typical Jacksonian sequence (2,8–10). These may occur in infants not treated with antiepileptic drugs
Also, in the 1950s and 1960s, paroxysmal phenomenon with (AEDs) with encephalopathy or those with electroclinical
minimal motor manifestations began to be recognized as clini- seizures who have been treated with AEDs with subsequent
cal seizures. These included paroxysmal ocular and oro-buccal- “decoupling” of electrical and clinical manifestations (17).
lingual movements, repetitive limb movements resembling
pedaling, rowing or swimming movements, changes in skin Epidemiology
color, respiration, and other autonomic changes (5,7). They Reported incidence of neonatal seizures range from 1.8 to 5.5
were initially characterized as “anarchic or atypical” and later per 1000 newborns (20–23). The National Collaborative
various other terms have been used to describe these seizures Perinatal Project, which studied infants of 54,000 pregnant
including “slight,” “minimal,” “subtle,” or “motor automatisms” women between 1959 and 1966, reported an incidence of
(9,10,13–16). Many of the so-called subtle seizures were subse- 5.1/1000 live births (24). More recent population-based studies
quently found not to be associated with simultaneous electro- in Fayette County, Kentucky; Newfoundland, Canada; and
graphic discharges. This was also observed to be true for other Harris County, Texas have reported lower incidences rates,
seizures, such as those characterized by generalized tonic pos- between 1.8/1000 and 2.5/1000 live births (20–23). Differences
turing. Over the years, this has led to controversy over their in target population, periods of surveillance, methods used in
pathophysiology. Observing that these events were similar to case identification, and use of clinical versus clinical and EEG
certain reflex behaviors, Mizrahi and Kellaway postulated that criteria in the identification of cases likely account for the vari-
these were primitive brainstem reflexes “released” by forebrain ability in reported rates (25). The incidence of neonatal seizures
depression, rather than true epileptic events (17,18). is higher in premature and low birth weight infants, as well as
These observations and controversies have led to the evolu- infants cared for in intensive care units (23). Neonatal seizures
tion of current classification systems of neonatal seizures, are most common in the first week of life (20,26).
which are primarily based on clinical semiology, electroen-
cephalographic features, the temporal relationship of clinical Pathophysiology
and EEG features, and presumed pathophysiology. They have Seizures occur more frequently in the neonatal period than at any
also led to several issues regarding diagnosis and management other time in life. Basic science research has shown various fac-
of neonatal seizures especially those unaccompanied by promi- tors underlie this increased susceptibility to seizures, based pri-
nent clinical manifestations. marily on relatively enhanced excitation in the immature brain

465
466 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

(27–32). For example, in the immature brain, potassium tends to ation of clinical behaviors with presumed pathophysiology based
accumulate in the extracellular space, secondary to decreased on the presence or absence of simultaneous EEG changes (16,17).
Na , K-ATPase activity, and immature enzyme systems. This In their classification scheme, neonatal seizures are broadly cate-
leads to the development of a hyperexcitable state and decreased gorized as (i) epileptic events—when associated with consistent
seizure threshold (27). Excitation may also be enhanced by a rel- electrocortical signature, (ii) nonepileptic events— when unac-
ative increase in the density of glutamate receptors and NMDA- companied by time-synchronized EEG changes, and (iii) electri-
gated channels. On the other hand, synapses and receptors for cal seizures—when EEG seizure discharges are not associated
inhibitory neurotransmitters may be less abundant in the devel- with obvious clinical manifestations (17).
oping brain. Also, GABA, the main inhibitory neurotransmitter The types of seizures found to be most consistently associ-
in the CNS, may have excitatory functions early on (33–35). In ated with EEG changes include focal clonic, focal tonic, some
animal models, the substantia nigra pars reticularis, which in myoclonic seizures, and spasms. Seizures not consistently asso-
adults is involved in the control of seizures, has been shown to ciated with an electrographic signature include generalized
amplify seizures in immature animals. These and other factors, tonic, some myoclonic, and motor automatisms. Mizrahi and
not only lower the overall seizure threshold, but likely, also allow Kellaway used the term “motor automatisms” to describe
seizures to spread more readily (25). seizures consisting of a variety of paroxysmal behaviors such as
Long-term consequences of neonatal seizures on brain devel- oral-buccal- lingual movements, ocular signs, progression
opment are unclear and likely affected by multiple factors, such movements, and complex purposeful movements. Each clinical
as seizure burden, age of the infant when seizures occur, and seizure type and their associated EEG abnormalities are
underlying etiology. Despite the increased susceptibility to described in detail below.
seizures, studies using animal models/rodents provide evidence Focal Clonic: These seizures consist of rhythmic, usually slow
to suggest that the immature brain is more resistant to seizure- (approximately one to three jerks per second) repetitive move-
induced hippocampal injury (28). However, in rats with a preex- ments of the face, proximal or distal arm or leg muscles, or axial
isting lesion, the risk of status epilpeticus (SE)–induced injury is structures. The movements may be unifocal (confined to one
significantly elevated (36). Also severe recurrent seizures can part of the body); hemiconvulsive (involving one side of the
lead to impaired synaptogenesis, myelination, and brain growth. body); or multifocal (involving multiple body parts on both
Galanopoulou has shown that three SE in P4-6 rat pups may sides). Clonic seizures that involve both sides of the body simul-
upregulate the expression of the chloride cotransporter KCC2 in taneously are usually asynchronous unlike true generalized
immature neurons with still depolarizing GABAA receptor clonic seizures in older individuals. Seizures may alternate
responses, which may prevent the maintenance of the GABA- between sites within a particular seizure, or may migrate from
mediated effects on cell maturation (37,38). This effect differs one site to another, not necessarily spreading in traditional
from those described in adults and may therefore underlie and Jacksonian sequence. Focal clonic seizures are usually associ-
partially explain the age-specific consequences of SE. ated with focal brain lesions such as cerebral infarctions but can
There are also studies that suggest that exposure to currently also occur with more diffuse neuropathologic processes. By far,
available AEDs used to control seizures during this period, may they are the type of seizures most consistently associated with
have similar detrimental effects causing increased apoptosis or electrographic correlate.
even on occasion promote seizures. In each case, the cost bene- Focal Tonic: Sustained posturing of a limb, or unilateral flex-
fit ratio of the effects of seizures or AEDs must be determined, ion of the trunk characterize these seizures that can also be
although ideally age-specific and even gender-specific treat- accompanied by sustained conjugate deviation to one side. Like
ments should be developed (see the following section). focal clonic seizure, tonic seizures are usually associated with
synchronized EEG discharges. They occur less frequently than
focal clonic seizures.
Clinical and EEG Features of Neonatal Seizures
Generalized Tonic: These seizures are characterized by sus-
Characterization and Classification of Neonatal Seizures tained bilateral extension or flexion of the limbs or trunk,
Traditional classification schemes for seizures have been diffi- resembling decerebrate or decorticate posturing. Unlike their
cult to apply to neonatal seizures that are generally fleeting more focal counterpart, these seizures are generally not associ-
events occurring in infants with limited repertoire of normal ated with EEG correlate and are classified as nonepileptic.
behavior to begin with. More than one seizure type can occur Because they tend to occur in association with other automatic
in a particular infant. behavior in obtunded infants with diffuse central nervous
The two most widely accepted classification schemes for pathologic processes, Mizrahi and Kellaway proposed that such
neonatal seizures are by Volpe and by Mizrahi and Kellaway. Each seizures are “brainstem release phenomenon” from disinhibi-
characterizes seizures based on semiology of the most prominent tion of forebrain structures in clinically depressed infants.
movement. In Volpe’s classification scheme, these neonatal Myoclonic: Irregular, erratic twitching, or contractions of
seizures are classified as (i) clonic, (ii) tonic, (iii) myoclonic, and muscle groups involving the face, limbs, or trunk are termed
(iv) subtle (15,39). Mizrahi and Kellaway’s classification scheme, myoclonus. These can be either epileptic or nonepileptic.
developed from findings on their EEG-polygraphic monitoring Myoclonic movements associated with EEG changes are also
studies, not only considers semiology, but emphasizes the associ- called “cortical myoclonus,” while myoclonus with no EEG
Chapter 25 ■ Seizures and Epilepsies in the Preterm and Term Neonate 467

correlate is termed “sub-cortical myoclonus.” The latter term Autonomic Signs: A variety of paroxysmal changes in auto-
implies that more caudal structures in the brainstem or spinal nomic activity such as alterations in breathing, heart rate, blood
cord responsible for generating the movements. The seizures pressure, salivation, sweating, color changes, have also been
can be focal, usually involving muscles of one upper extremity; described as manifestations of subtle seizures (42–44). However,
or they may be generalized or multifocal. Generalized in isolation, they rarely occur as epileptic phenomena. When
myoclonus consists of bilateral symmetric flexion jerks of the they are accompanied by ictal EEG correlates, they occur in asso-
limbs or trunk. Multifocal or “fragmentary” myoclonus is char- ciation with other clinical behavioral or motor phenomenon. In
acterized by brief asynchronous twitching of different muscle a study by Watanabe et al., 14 of the 21 infants with apneic
groups. Of the three types, fragmentary myoclonus is the type seizures with electrical EEG correlate exhibited other subtle phe-
least associated with EEG changes, while two thirds of general- nomenon such as eye-opening, “staring,” deviation of the eyes
ized myoclonus have EEG correlate. and mouth movements (45). Bradycardia was also found to be
Spasms: Spasms characterized by bilateral flexor, extensor, or more common in “convulsive apnea” versus “nonconvulsive”
mixed flexion and extension contractions of limb and truncal apnea in another study (46).
muscles occur rarely in the neonate. Like spasms in older
infants, they can occur in clusters and can be more frequent Ictal EEG Correlates
upon awakening. On EEG, they are associated with a general- Electrical seizures are more common in term than in preterm
ized attenuation of the background or a high-voltage slow wave. infants. When they occur in infants less than 34 to 35 weeks,
Spasms are not included in Volpe’s classification scheme. they are shorter in duration (23). Onset is usually focal, usually
Motor Automatisms: Also called “subtle seizures,” paroxysmal in the central and temporal regions (Fig. 25.1). Less common
changes in behavior or autonomic functions with minimal sites of origin are the frontal and occipital regions. Seizures can
motor manifestations occur frequently in the newborn. These arise from more than one area, propagating asynchronously
consist of various irregular and disconjugate ocular move- from different foci (Fig. 25.2). Frequency, morphology, and
ments, eye-opening, chewing, oral-buccal movements, and voltage of the discharge may vary within the same seizure, or
peculiar extremity movements such as pedaling, stepping, box- from one seizure to the next in a given infant. The seizure dis-
ing, swimming movements. Mizrahi and Kellaway found these charge can consist of repetitive spikes, sharps or slow waves, or
behaviors to be inconsistently associated with EEG seizure and a combination of different waveforms. Seizures may remain
classify them as nonepileptic. However, similar behaviors, in localized to a specific area, slowly spread to involve contiguous
preterm, and some full term infants have been demonstrated by regions, abruptly involve one of hemisphere, or migrate to the
other investigators to be ictal phenomena associated with other hemisphere (25).
simultaneous abnormal EEG discharges (40). Similarly, in older Electrographic seizures in the absence of clinical activity in
individuals, paroxysmal automatic behaviors are known to severely encephalopathic neonates are characterized by back-
occur without surface EEG correlate as manifestations of psy- ground EEG activity that is usually depressed and undifferenti-
chomotor seizures or postictal behavior (41). Studies in neona- ated. Some are lower in voltage, longer in duration, and show
tal animals have also shown that epileptiform discharges can little tendency to spread or change. These have been referred to
occur in deep limbic structures with minimal or no propaga- as “seizure discharges of the depressed brain” (26) (Fig. 25.3).
tion to the cortical surface (30). These observations raise the Alpha seizure discharges are characterized as paroxysmal 8 to
possibility that subtle seizures in neonates may be true epileptic 12 Hz alpha activity in the central or temporal regions in
events with generators in deeper limbic structures if concomi- encephalopathic newborns and suggest a poor prognosis
tant electrographic seizure activity is the only criteria. These (45,47,48) (Fig. 25.4).
observations, however, do not exclude the possibility that some When neonates are treated for seizures with antiepileptic
of these events behavior may be reflex behaviors. medications, persistent electrical seizure activity can be found

Figure 25.1 Central onset of electrical seizure


activity. Rhythmic sharp waves arise in the left
central region and remain confined to that region,
associated with a clinic focal clonic seizure involv-
ing the right hand. The EEG was recorded from a
40-week C.A. infant with a left frontal lobe infarc-
tion. (Reprinted with permission from Mizrahi
EM, Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia:
Lippincott Williams & Wilkins; 2004:250.)
468 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 25.2 Multifocal electrical seizure activ-


ity. Seizure discharges occur simultaneously but
asynchronously from the two hemispheres in this
39-week C.A. infant with hypoxic-ischemic
encephalopathy. There is initial rhythmic,
moderate-voltage, sharp-wave activity in the
right centrotemporal region and then an inde-
pendent discharge arising from the left temporal
region characterized by sharp- and slow-wave
activity with complex morphology. No clinical
seizures accompanied these discharges.
(Reprinted with permission from Mizrahi EM,
Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia:
Lippincott Williams & Wilkins; 2004:250.)

long after clinical seizures have stopped. This “decoupling” of However, in the setting of neonatal seizures, assessment of
electrical from clinical activity, first described by Mizrahi and the interictal EEG background is still clinically useful. It can
Kellaway, has been demonstrated by other investigators (17). In suggest possible etiologies for the seizures, help in determining
a study by Clancy and coworkers, 79% of 393 electrographic an infant’s degree of cerebral dysfunction, risk for persistent
seizures recorded were not accompanied by clinical seizure seizures, and prognosis for long-term outcome.
activity. Seizures were the result of diverse etiologies and most EEG findings correlating with diffuse cerebral dysfunction
infants had received one or more AED prior to the EEG record- include a depressed undifferentiated background, suppression-
ings (49). Similar seizures without clinical correlates were also burst pattern, multifocal sharp transients, and dysmaturity.
noted by Scher, Bye and Flanagan, and Murray to occur fre- These abnormalities, although not typically etiologic specific,
quently (23,40,50). suggest etiologies which result in generalized or multifocal brain
injury, such as hypoxia, acute metabolic disturbances, inborn
Interictal EEG Correlates errors of metabolism, infection, and bilateral brain hemorrhage.
Although the documentation of simultaneous EEG discharge On the other hand, persistent asymmetries of the background
during clinical events is useful in the diagnosis of seizures in the and persistently lateralized sharp transients may indicate focal
neonate, the finding of sharp-wave transients on interictal structural brain abnormalities such as focal cerebral infarction
records does not aid in the diagnosis of seizures, or predict or areas of dysgenesis as underlying etiologies. The absence of
epileptogenicity. Unlike in adults, transient sharp waves, fre- focal features does not rule a focal structural pathology (25).
quently found in the central and temporal regions are a nonspe- The EEG obtained within the first 24 hours can help in
cific finding that can be seen in pathologically or as normal determining prognosis and risk for further seizures. Prospective
finding in term and preterm infants (26,51,52). Early studies studies in infants presenting with seizures in the neonatal
suggested that positive rolandic sharp waves were most specific period have found that the background EEG can be used for
for intraventricular hemorrhage (53–55), but subsequently have prognostic purposes to predict survival and long-term outcome
been more closely associated with periventricular leukomalacia. (25,56–58). A normal EEG suggests a good prognosis while the

Figure 25.3 Seizure discharge of the depressed


brain. Low-voltage, rhythmic, monomorphic,
slow sharp waves on the left persist virtually
unchanged throughout the recorded seizure. No
clinical seizures were present. The background
activity is depressed and undifferentiated. The
infant is 39-week C.A. with hypoxic-ischemic
encephalopathy. (Reprinted with permission
from Mizrahi EM, Hrachovy RA, Kellaway P.
Atlas of Neonatal Electroencephalography. 3rd
ed. Philadelphia: Lippincott Williams & Wilkins;
2004:250.)
Chapter 25 ■ Seizures and Epilepsies in the Preterm and Term Neonate 469

Figure 25.4 Alpha seizure discharge. A seizure


discharge in the left temporal region is characteri-
zed by sinusoidal rhythmic 10 to 11 Hz activity
that evolved from rhythmic sharp wave activity
in that region. There is also an independent
seizure discharge in the left occipital region
appearing as semiperiodic sharp waves. The
background activity is depressed and undifferen-
tiated in this 38 week C.A. infant with meningi-
tis. (Reprinted with permission from Mizrahi EM,
Hrachovy RA, Kellaway P. Atlas of Neonatal
Electroencephalography. 3rd ed. Philadelphia:
Lippincott Williams & Wilkins; 2004:250.)

presence and persistence of diffuse abnormalities on serial Common acquired structural brain lesions include intracra-
EEGs suggest poor outcome. nial hemorrhage and cerebral infarctions. Focal areas of cerebral
EEG background may also predict persistent seizures in the dysgenesis and more diffuse anomalies from major congenital
acute period as well as in the postneonatal period. Laroia and brain malformations, such as in lissencephaly, can produce
coworkers, studying 51 infants at risk for neonatal seizures, found seizures as well in the neonatal period. The presence of a struc-
that a normal or immature background strongly predicted the tural brain lesion can be suggested by focal seizures, electro-
absence of electrographic seizures in subsequent 18- to 24-hour graphically arising from the same region, or from asymmetries
period of video-EEG monitoring, while background abnormali- in voltage or frequency of background activities. Neuroimaging
ties predicted the occurrence of electrographic seizures within is essential when such abnormalities are found on EEG.
that same time frame (59). Among the 27 survivors of neonatal Drug withdrawal or intoxication is another cause of tran-
seizures followed for a mean of 31 months by Clancy and sient seizures in the newborn. History of maternal drug use or
coworkers, postneonatal epilepsy occurred in 68% (13 of 19) of of administration of medications during delivery is suggestive
patients with moderately or markedly abnormal EEG back- of the diagnosis. The affected neonate can appear depressed and
grounds but in only 25% (2 of 8) without (60). Ortibus and col- exhibit symptoms such as jitteriness that can be confused with
leagues however, did not find background abnormalities to be a seizures.
strong predictor of seizures past the neonatal period (56). When infectious, metabolic, and structural causes of seizures
are excluded, neonatal encephalopathy secondary to hypoxic
Diagnosis and Treatment of Acute Seizures injury is often implicated, especially if accompanied by other sys-
Etiology temic signs of anoxia such as persistently low Apgar scores, sys-
Major etiologies for acute seizures in the neonate include (i) temic acidosis, and multiorgan involvement. Grading schemes
and criteria used to diagnose neonatal encephalopathy vary and
neonatal encephalopathy and hypoxic-ischemic encephalopa-
it is difficult to assess the true proportion due to asphyxia (62,63).
thy; (ii) intracranial hemorrhage and infarctions; (iii) infec-
Perlman and colleagues looked at various clinical and laboratory
tions; (iv) metabolic disturbances; (v) congenital structural
markers within the first hour of life that identify newborn infants
brain lesions; and (vi) drug withdrawal (25,61). The term
at high risk for seizures secondary to hypoxia ischemia. In their
neonatal encephalopathy has gained favor in the literature over
study of 96 infants, significant relationships with seizures were
recent years over the more specific term, hypoxic-ischemic
found with the following variables: 5-minute Apgar score of 5 or
encephalopathy, as oftentimes, a direct causal relationship
less, the need for intubation in the DR, umbilical cord arterial pH
between hypoxia and encephalopathy is hard to establish.
of 7.00 or less, and base deficit of –14 mEq/L (64). In 49 infants
Metabolic disturbances causing seizures are important to
monitored with continuous EEG, Murray and colleagues found
identify as many acute disturbances are potentially treatable with
similar positive predictive value for seizures for low Apgar scores,
etiology-specific therapy. These include electrolyte abnormalities
systemic acidosis, and the need for intubation.
such as hypocalcemia, hypomagnesemia, and hypoglycemia.
Surgery for congenital heart disease in the neonatal period
Rare inborn errors of metabolism such as urea cycle defects,
is a procedure that carries increased risk for seizures.
aminoacidurias, and organic acidurias can cause neonatal
Electrographic seizures were found in 11% of 183 post-op
seizures refractory to AED treatment. These usually require
infants monitored by video-EEG in a study by Clancy and col-
dietary manipulation to control. Pyridoxine-dependent
leagues (65) (Table 25.1).
seizures can produce severe seizures, including spasms that may
respond to pyridoxine.
Systemic and CNS infections can be secondary to postna- Differential Diagnosis
tally acquired bacterial and viral agents, or congenital, in-utero The differential diagnosis of neonatal seizures includes physio-
infections. logic and nonphysiologic events that may be abnormal or a
470 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 2 5 . 1 neonatal behavioral repertoire. Apnea or changes in autonomic


functions are rarely epileptic in origin when they occur in the
Most Frequently Identified Etiologies of Neonatal absence of other autonomic or motor manifestations (45).
Seizures a
Treatment
Hypoxia-ischemia Seizures are often the first sign of neurologic dysfunction in the
Intracranial hemorrhage newborn. Prolonged seizures can adversely affect an infant’s
homeostasis and protracted or frequent seizures have been
Intraventricular associated with increased morbidity (74). Thus, seizures in the
Intracerebral neonate are considered to be urgent medical conditions that
Subdural require prompt evaluation and treatment.
Management begins with stabilization of the infant with par-
Subarachnoid ticular attention given to maintenance of airway, breathing, and
Infection––CNS circulation. Establishing intravenous access for medications is of
Meningitis prime importance since this is the most reliable route for achiev-
ing immediate therapeutic blood levels of anticonvulsants.
Encephalitis Although determining etiology should not delay the treat-
Intrauterine ment of seizures, in certain circumstances, etiology-specific ther-
Infarction apy alone is required to control seizures. This is particularly true
with seizures secondary to metabolic disturbances such as hypo-
Metabolic glycemia, hypocalcemia, hypomagnesemia, hypo- or hyperna-
Hypoglycemia tremia, hyperammonemia, or pyridoxine-dependent seizures.
Hypocalcemia Similarly, antibiotics and antiviral agents are used empirically
when infectious causes are suspected, until ruled out by CSF and
Hypomagnesemia blood cultures (61).
Chromosomal anomalies The current practice is to treat highly suspicious clinical
Congenital abnormalities of the brain behavior as seizures with anticonvulsants even without initial
EEG confirmation. Focal clonic and tonic movements correlate
Neurodegenerative disorders well with electrographic seizures in the neonate. On the other
Inborn errors of metabolism hand, motor automatisms, myoclonus, bilateral tonic stiffening,
Benign neonatal convulsions as well as movements that are highly stimulus sensitive, corre-
late poorly with electrographic seizures. Therefore, it is ideal to
Benign familial neonatal convulsions confirm their epileptic pathophysiology with EEG before con-
Drug withdrawal or intoxication tinuing treatment with anticonvulsants.
When initiating antiepileptic therapy, physiologic factors
a
Listed in relative order of frequency. Not listed is “unknown” etiology, which is unique to the neonatal period have to be taken into considera-
encountered in approximately 10% of cases. (From Mizrahi EM, Kellaway P. tion. These may influence the choice, route of administration,
Diagnosis and Management of Neonatal Seizures. 1st ed. Philadelphia: Lippincott
and dosing of medications. The oral route of administration for
Williams & Wilkins; 1998.)
initiation of AED therapy is not appropriate. Absorption of
medication is decreased due to immaturity of the GI system and
variation of normal neonatal behavior. Infants withdrawing from from early variations in gastric pH (75). Intramuscular absorp-
in-utero drug exposure or who have suffered anoxic or acute tion of medications is also erratic and inconsistent due to
metabolic insults can have tremulous or jittery movements, as increased water content and smaller muscle mass in the new-
well as exaggerated clonus (66,67). Unlike seizures, these move- born. The same factors also lead to higher risk of local injury
ments are very stimulus sensitive and can be suppressed by repo- and muscle necrosis with this route of administration (76).
sitioning or restraint. Physiologic myoclonus is a common sleep Therefore, the most dependable route for administering medica-
phenomenon mistaken for seizures. Some infants have a particu- tions remains intravascular, and establishing access is critical
larly exaggerated form of it that persists for the first few months early on.
but is benign. Because of its occurrence in sleep it is called benign The half-lives and free drug concentration of many medica-
neonatal sleep myoclonus (68). Infants with hyperekplexia, a tions are increased due to a combination of factors. These
condition caused by a mutation in genes encoding for inhibitory include a greater proportion of total body water in the neonate,
glycine receptor, also tend to have prominent myoclonus in sleep decreased albumin concentration and plasma binding of drugs,
or upon arousal. In addition, they have excessive startle response decreased drug metabolism, and excretion from immature
triggered by minimal tactile or auditory stimulus, as well as hepatic and renal systems (19). This scenario makes anticonvul-
episodes of generalized tonic stiffening (68–73). As mentioned sant drug levels highly variable, especially when the infant is
earlier, subtle sudden or repetitive mouth or limb movements systemically ill and receiving other medications. Close monitor-
can raise the suspicion for seizures, but can be part of normal ing of drug levels is necessary especially early on in the course
Chapter 25 ■ Seizures and Epilepsies in the Preterm and Term Neonate 471

of treatment that needs to be individualized. Medication contradictory results. In a nonrandomized retrospective study
dosages often need adjustment over time, as the infant’s physi- of midazolam in the setting of refractory neonatal seizures,
ologic systems mature. electrographically confirmed seizures were rapidly controlled in
13 of 13 nonresponders to phenobarbital/phenytoin treated
Antiepileptic Therapy AED therapy is begun with an initial with midazolam (88). However, a prospective trial looking at its
intravenous bolus given to achieve targeted therapeutic levels of effectiveness as a first-line agent found it to be ineffective (87).
the drug. Subsequent smaller boluses of the same medication Valproic acid is more widely used in Europe, but because of
can be given to control persistent clinical seizures. If seizures its potential to cause fatal hepatotoxicity in this age group, it is
continue despite titration to maximal levels or clinical toxicity, rarely used in the United States. In a small study, lidocaine was
a second agent is added. The ideal is to control both clinical and found be effective in controlling seizures in 50% of infants with
electrographic seizures; however, it is controversial to aggres- refractory seizures (89).
sively pursue purely electrographic seizures that often require More recently there has been interest in the use of lidocaine
high dosages of potentially toxic AED levels for control. as an AED for otherwise refractory neonatal seizures. There is a
Two medications most frequently used as first-line agents narrow therapeutic range, with reports of dose-related and
are phenobarbital and phenytoin/fosphenytoin (77), although reversible cardiac arrhythmias, with infusions (95).
phenobarbital is the most favored. Studies have shown them to There is no universally accepted protocol for duration of
be equally effective in controlling seizures, although overall treatment of the acute period. Duration of treatment ranges
effectiveness only reaches 50% (78). from 2 weeks to 1 year (100–103). Regardless of duration, most
Phenobarbital is more widely used as a first-line agent. The clinicians obtain an EEG prior to gradually weaning off AEDs,
usual loading dose is 20 mg/kg IV, with initial maintenance doses looking for persistent subclinical seizures (77).
of 3 to 4 mg/kg/day in two divided doses. Titration of the dose to
achieve levels of up to 40 mcg/mL may be necessary in refractory Prognosis
cases; however, further control of seizures is hardly achieved Neonatal seizures are associated with significant incidence of
above this maximal therapeutic level (79). Common toxic effects mortality and morbidity. Reported mortality rates range from
at this level include lethargy, central respiratory depression with 24% to 30%, with decreasing incidence over the years from
apnea, bradycardia, and hypotension. Apparent half-life after 5 to improved neonatal and obstetric care. The risk of neurodevel-
7 days is 100 hours. It is greater in premature infants and in those opmental sequelae is also high but varies depending on etiology
suffering from impaired hepatic metabolism and renal clearance and degree of brain injury. Abnormalities in the neurologic
from hypoxic-ischemic encephalopathy. examination and developmental delay are reported in about
Phenytoin is used after phenobarbital, although the prodrug half of survivors. Cerebral palsy and mental retardation have
form, fosphenytoin is the preferred agent due to its better side been reported in about a third (56,104,105). Prognosis was bet-
effect profile. A loading dose of 15 to 20 mg PE/kg IV is recom- ter for term than for preterm infants (104). Epilepsy occurs in
mended with subsequent maintenance doses of 3 to 8 mg 20% to 30%, usually with onset within the first 6 to 9 months
PE/kg/day in two to three divided doses. Average therapeutic of life (23,56,57,60,100,104).
range is 10 to 20 mcg/mL. Because of highly variable metabo- Etiology is the most important determinant of overall out-
lism and oral absorption, half-life ranges from 40 to 100 hours. come in neonates who experience seizures (4,13,25,67,106–108).
Due to nonlinear kinetics, small dose changes near maximal Infants whose seizures are secondary to severe or diffuse brain
therapeutic levels can lead to acute toxicity, therefore close insults such as hypoxic-ischemic encephalopathy, intraventricu-
monitoring and an individualized approach to therapy is neces- lar hemorrhage, or congenital cerebral malformations have
sary (80–82). Common side effects include arrhythmia and poorer prognosis compared to those whose seizures are second-
local irritation with phenytoin. ary to transient causes, such as hypoglycemia, hypocalcemia,
There is little consensus in the choice of AEDs to treat and drug withdrawal. This observation holds true for the risk of
seizures that are refractory to phenobarbital and phenytoin. developing both neurodevelopmental sequela and postneonatal
Medications reported to have been used in such scenarios epilepsy. The highest risk of epilepsy is reported in the setting of
include benzodiazepines (79,83–88), valproic acid (89), primi- cerebral dysgenesis and infections and severe intraventricular
done (90), carbamazepine (91,92), anesthetic agents (87,93–96), hemorrhage (13,56,58,104).
and some of the newer antiepileptic agents such as lamotrigine Certain clinical characteristics of the seizures themselves
(97), vigabatrin (98), topiramate, and levetiracetam (99) admin- have been found to correlate with outcome. Mizrahi and
istered nasogastrically. Kellaway found that focal tonic and focal clonic seizures, typi-
Lorazepam at a dose of 0.05 mg/kg and repeated up to a total cally associated with focal brain lesion, predicted better prog-
dose of 0.15 mg/kg (85) and diazepam at 0.25 mg can be nosis. Myoclonic seizures, motor automatisms, and generalized
administered as IV boluses, with subsequent additional boluses tonic posturing on the other hand were more likely to be asso-
given for persistent seizures (19). Many intensivists prefer the ciated with diffuse brain involvement and a worse prognosis
use of midazolam given as an infusion of 0.05 to 0.4 mg/kg/hr (109). These observations have been supported by later investi-
after an initial bolus of 0.1 to 0.3 mg/kg. Its short half-life lends gators (104,105).
to ease of titration, and discontinuation of the drug once The duration of seizures and their responsiveness to medica-
seizures are controlled. Published data on its effectiveness show tions also determine prognosis. Seizures that are prolonged
472 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

(56,74), frequent (110), or require multiple drugs to control maternal and delivery history and laboratory and neuroimag-
(104) have been associated with poorer prognosis. ing studies (121).
EEG can have prognostic significance as well. As mentioned Clinical seizures are described to have clonic or tonic com-
earlier, the presence EEG background abnormalities, is associ- ponents and can be partial or generalized with asymmetric
ated with poorer outcome. A normal EEG or mildly abnormal motor activity. Myoclonic seizures or spasms have not been
EEG, on the other hand, predicted normal neurologic develop- described. Apnea is a frequent component (122). Seizures
ment in more than 80% in a recent study (58). The presence of described in kindred of 69 affected individuals consisted of
interictal epileptiform activity is not as clearly related to out- mixed seizure types, starting with tonic posture, ocular symp-
come or postneonatal seizures. Rowe and coworkers found toms, apnea, and other autonomic features, and progressing to
epileptiform abnormalities on interictal EEG to correlate with clonic movements and motor automatisms. The ictal EEG pat-
adverse outcomes, but not to be as significant a predictor of out- tern was characterized by generalized suppression of amplitude
come as background rhythms (111). Ortibus found the number on onset (123). Hirsch also reported generalized onset with
of negative sharp waves in the temporal region to correlate with bilateral, symmetrical flattening of the EEG in three infants.
both outcome and postneonatal seizures (56). In Clancy’s study, This was associated with simultaneous tonic motor activity and
PNE occurred in only 27% (3 of 11) of patients without spikes apnea, and followed by bilateral discharge with associated
or sharp waves on postnatal EEGs performed at the age of 3 vocalizations, chewing, and focal or generalized clonic activity
months but in 100% (3 of 3) of patients with spikes or sharp (124). However, Bye recorded focal onset seizures with and
waves (60). Electroencephalographic seizures, whether associ- without secondary generalization in one neonate (125), and
ated with clinical manifestations have been correlated with Plouin and Anderson suggested that the electroclinical presen-
higher mortality (up to 50% in a study by Scher and coworkers) tation is closer to partial epilepsies (121).
and increased risk of developmental abnormalities (23,111). The interictal EEG is nonspecific and may be normal or
abnormal (119). Some abnormalities reported in the literature
NEONATAL EPILEPSY SYNDROMES include interictal sharp transients, the theta pointu alternant
pattern (126–129). Centrotemporal spikes have also been
Four neonatal onset seizure disorders are recognized by the reported in one patient who later on developed one clinical
International League Against Epilepsy (ILAE). Two of these are rolandic seizure at the age of 4 years (130).
relatively benign syndromes characterized by transient seizures Prognosis is generally good with resolution of seizures by 1
and good prognosis neurodevelopmental outcome. The two to 6 months and normal neurologic outcome in the majority of
others do not have similar favorable prognosis and present with cases. Mental retardation and learning disabilities do not occur
seizures that persist beyond the neonatal period and progressive at an increased rate compared to the general population (120).
neurologic deterioration. However, epilepsy develops in 11% to 14% of cases with 5%
risk of febrile seizures (121,122).
Benign Syndromes Seizures are generally treated with antiepileptics because of
Benign Familial Neonatal Seizures (BFNS) their frequency even though they are generally self-limited.
As its name implies, the syndrome of benign familial neonatal Phenobarbital is most commonly used although phenytoin and
seizures (BFNS) is a familial epileptic disorder that occurs in the valproate have been used in refractory cases (121). Novel
neonatal period, and runs a benign course with good prognosis. approaches to treatment with antiepileptic and anti-inflamma-
The name was coined by Bjerre and Corelius in 1968 (112); how- tory medications that target KCNQ2/KCNQ3 channels are
ever, the first familial case was described by Rett and Teubel 4 being investigated in animal models (131–133).
years earlier (113). It was first included in the ILAE classification
of epilepsy as a syndrome in 1989. In recent years, it has been Benign Neonatal Convulsions (BNCs)
identified as a CNS channelopathy caused by mutations in the Navelet and Dehan were the first to describe nonfamilial cases of
genes encoding potassium channels (114–118). BFNS is a rare self-limited idiopathic seizures in the newborn that ran a benign
disorder that may be under recognized. Ronen and colleagues course (134). It was later recognized as an epileptic syndrome by
found 5 familial cases out of 34,615 live births in their popula- the ILAE and given the name “benign neonatal convulsions.”
tion-based study of neonatal seizures in Newfoundland (104). Emphasizing its unclear etiology, the term “benign idiopathic
Seizures in affected infants begin by 2 to 8 days of life, with neonatal convulsions” has also been used in the literature. Because
80% appearing on the second or third day (19). Onset of seizures of the tendency of the seizures to occur on the fourth or fifth day
can be delayed up to 3.5 months especially in premature infants of life, the term “fifth day fits” has also been commonly used.
(104,119). The seizures that are brief, typically 1 to 2 minutes, can Although the exact incidence is unknown, BNCs are sug-
occur multiple times a day during the vulnerable period, with gested to represent 2% to 7% of seizures in the neonate (121).
most seizures remitting by 16 months of life (104,119,120). Affected infants are usually term or near term and are products
The diagnosis is suspected when typical seizures occur of uneventful pregnancies and deliveries. Males are more
without obvious precipitants in an otherwise normal newborn affected than females (121).
with a family history of similar seizures in the neonatal period. Like the familial form, its diagnosis requires the exclusion of
It requires exclusion of other causes of seizures by normal other transient causes for seizures that present with the typical
Chapter 25 ■ Seizures and Epilepsies in the Preterm and Term Neonate 473

EEG abnormalities, such as hypocalcemia, subarachnoid hemor- Catastrophic Epilepsy Syndromes


rhage, and neonatal meningitis (121). Plouin proposed the fol- Two so-called catastrophic epilepsy syndromes occur in the
lowing diagnostic clinical criteria: (i) Apgar score greater than neonatal period and have been classified as generalized sympto-
seven at 1 minute, (ii) typical interval between birth and matic epilepsies by the ILAE. Unlike the previously discussed
seizures onset (4 to 6 days), (iii) normal neurologic examination benign disorders, these epilepsy syndromes are invariably associ-
before seizures and interictally, (iv) normal laboratory findings ated with abnormal mentation, psychomotor retardation, and
(metabolic studies, neuroimaging, and CSF analysis), (v) no persistent, difficult to control seizures, often consisting of various
family history of neonatal seizures or postneonatal epilepsy types. It is thought that in these syndromes, the epileptiform
(135). The final criterion distinguishes this syndrome from the abnormalities themselves, and not just the underlying disorder,
familial form. contribute to the eventual neurologic deterioration. Thus, they
The etiology and pathogenesis of the seizures are unknown. join the syndromes of infantile spasms, Lennox–Gastaut, Dravet,
One intriguing study found rotavirus in the stool of 95% of 19 and electrical status epilepticus during sleep under the category
“fifth day fitters,” compared to 40% in the control group (136). epileptic encephalopathies.
Another study found low levels of zinc in the CSF of affected
infants (43). In four cases, a mutation in the gene encoding for Early Myoclonic Encephalopathy
KCNQ2 (a mutation frequently found in the familial form of The syndrome was first described by Aicardi and Goutieres
BNCs) was identified (117). However, no consistent links have in 1978, who used the term neonatal myoclonic encephalopa-
been found to metabolic, infectious, or genetic causes. thy (140). Other subsequent nomenclatures have included
Typical seizures in affected infants consist of brief focal, “myoclonic encephalopathy with neonatal onset” (141), “neona-
clonic activity typically lasting 1 to 3 minutes. The seizures tend tal epileptic encephalopathy” (142), and “early myoclonic epilep-
to occur in clusters beginning on the fourth or seventh day of tic encephalopathy” (143).
life. In 80% of cases, the seizures occur on the fourth or fifth In this syndrome, erratic and fragmentary myoclonic seizures
day. Multiple clusters of seizure can occur in a day and can lead occur early on, often within the first few days or months of life.
to status epilepticus. Average duration of seizures is 20 hours. Myoclonic movements can affect the face, proximal, or distal
Although the syndrome is classified as a generalized epilepsy limb muscles. The jerks can be restricted to a few muscles or can
syndrome, focal clonic seizures are more common (121,137). affect multiple muscles asynchronously, shifting erratically from
These may or may not be associated with apnea. Focal tonic one area to another. Generalized massive myoclonus is less com-
seizures are rare although Guerra reported one patient with mon. Frequency of seizures is variable but can be almost contin-
fifth day fits presenting with generalized tonic–clonic seizures uous. Partial seizures follow the appearance of the erratic
and generalized electrical discharges on video-EEG (138). Ictal myoclonus. These seizures can be characterized by focal clonic
EEG patterns are nonspecific and can be focal or more general- movements of a limb or subtle eye-deviation with respiratory
ized, characterized by rhythmic spikes or rhythmic slow waves. changes. Tonic spasms are the last type of seizures to appear,
The seizure discharge can arise from any area but is most com- usually between 3 to 4 months, although earlier onset has been
monly seen in the rolandic region (121). described.
The interictal EEG is more characteristic, although nonspe- The interictal EEG is highly abnormal and is characterized
cific. Plouin reviewed EEG patterns described in 101 of 278 by brief, 1 to 3 seconds, bursts of high amplitude slow waves
cases (121). In 60% of cases, a theta pointu alternant pattern with intermixed spikes and sharp waves. Burst alternate with
was identified consisting of discontinuous, nonreactive, 4 to periods of suppression that can last up to 10 seconds. This burst
7 Hz theta activity that frequently alternates between hemi- suppression pattern is more pronounced in sleep and can later
spheres and is intermixed with sharp activities. The pattern can on evolve into atypical hypsarrhythmia. The EEG can be nor-
be present in wakefulness as well as quiet sleep. This is clearly an mal at the onset of the seizures, and repetitive EEGs may be
interictal pattern as it can persist days after cessation of clinical necessary for the diagnosis (144). Partial seizures are often
seizures. Other abnormal patterns that have been described accompanied by electrographic seizure activity but the erratic
include centrotemporal sharps, discontinuous patterns, and myoclonus usually has no EEG correlate.
focal or multifocal nonspecific abnormalities. The EEG is nor- The neurologic examination is abnormal from the onset and
mal in 10% of patients (121). affected infants are often lethargic, hypotonic, and have psy-
The course of BNC is considered generally benign, although chomotor delays. Developmental regression has also been
favorable outcome needs confirmation by more extensive stud- described (143). The mortality rate is high (up to 50%) and
ies (121). Seizures usually remit by 48 hours of onset (139). many die within the first 2 years of life (145).
Some argue against treating these self-limited seizures, but EME is a rare disorder. Of 2220 cases of epilepsy in children
most clinicians treat acute seizures with antiepileptics––usually under 10 years of age, 2 were classified as having EME (preva-
phenobarbital and phenytoin. Although the exact incidence of lence rate of 0.1%) by Oka and colleagues in their population-
postneonatal epilepsy is unknown, seizures rarely occur later in based survey in Okayama, Japan (146). Various etiologies have
life. Mild psychomotor retardation has been described in some been associated with this syndrome, suggesting that EME may
survivors; however, prognosis for neurodevelopmental out- represent a broad spectrum of clinical entities (147). Inborn
come is considered to be generally favorable (121). errors of metabolism are commonly reported. Cases associated
474 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

with nonketotic hyperglycinemia (145,147–149), methyl- childhood after a transient evolution into hypsarrhythmia in
malonic acidemia (148), propionic acidemia (148,150), molyb- middle to late infancy (154,157).
denum cofactor deficiency (151), and pyridoxine-dependency EEG during tonic spasms principally shows desynchroniza-
(147) have been reported. Structural brain abnormalities have tion with or without evident rapid activity (156). Using video-
also been described but are less common than in the other cat- EEG with polymyographic recordings from deltoid muscles,
astrophic neonatal epilepsy syndrome––early infantile epileptic Fusco and coworkers found each burst to be associated with
encephalopathy (152–154). tonic contraction of variable duration in all but one of the eight
infants in their study (159). They also recorded partial seizures
Early Infantile Epileptic Encephalopathy clinically consisting of tonic eye-deviation, unilateral clonic
Also known as Ohtahara syndrome, after Ohtahara who first contractions, or subtle or subclinical phenomena. There was no
described the disorder in 1976 (155), early infantile epileptic particular localization of ictal discharges on the EEG. Yamatogi
encephalopathy is another catastrophic epilepsy syndrome with described partial seizures to arise from a fixed focus and to be
neonatal onset. In fact, intrauterine onset has been suggested followed by tonic spasms in series (156).
(153). Certainly, it occurs very soon after birth, usually within Both congenital and acquired structural brain lesions have
the first month of life (156). The diagnosis is suspected when been described in affected infants. In Ohtahara’s original series
the typical newborn infant presents with tonic spasms and has of 16 cases, 11 were found to have structural abnormalities. Four
a suppression-burst pattern on EEG. Often, a structural brain had cerebral dysgenesis, two had Aicardi syndrome, and there
abnormality is found on neuroimaging studies, and the infant were one case each of hemimegalencephaly, lissencephaly, poren-
does poorly with severe psychomotor retardation and refrac- cephaly, hydrocephalus, and subacute diffuse encephalopathy
tory seizures. (157). Schlumberger identified six cases of hemimegalen-
The most characteristic seizures in this syndrome are tonic cephaly in his series of eight patients. One had Aicardi syn-
spasms, which can occur up to 300 times a day (157). They can drome and another had dentato-olivary dysplasia (158). Other
be lateralized, or generalized but asymmetric. Other seizure associated brain malformations reported in the literature
types such as partial motor seizures and hemiconvulsions include diffuse cerebral migrational disorders (163) and
occur in about a third of patients (156). Although generalized polymicrogyria (164).
tonic–clonic seizures and rare myoclonic seizures can develop Metabolic causes are rare but single cases of Leigh
later, the erratic myoclonus, which characterizes early myoclonic encephalopathy (165), nonketotic hyperglycinemia (160),
encephalopathy, does not occur in this syndrome (158). cytochrome c oxidase deficiency (166), pyridoxine-dependency,
The characteristic suppression-burst pattern found on EEG and carnitine palmitoyltransferase deficiency (159) have been
is an interictal pattern, although some have observed clinical reported.
seizures to correlate with the bursts (159). The pattern consists Seizures are typically refractory to conventional anticonvul-
of bursts of high-voltage (150 to 350 V) slow waves intermin- sant medications, and affected infants generally do poorly.
gled with spikes, alternating with periods of suppression of Ohtahara reported ACTH to be effective in two cases in the
electrical activity. The bursts of spikes and polyspikes can last 1 acute period and in four cases during the transition to West syn-
to 6 seconds, while the periods of suppression are usually 2 to 5 drome (157). However, ACTH used in four of Clarke’s cases was
seconds long (145,156), although they can last up to 18 seconds not effective (160). He reported only transient improvement of
(160). The interval measured from beginning to beginning of seizures in two out of three infants given the ketogenic diet, and
each burst ranges from 5 to 10 seconds. The pattern can involve in three out of four given chloral hydrate. Cases responding to
only one hemisphere, or be more widespread, involving both vitamin B6 (159), clonazepam and acetazolamide (157), zon-
hemispheres symmetrically or asymmetrically. Asynchrony is isamide (156,167), and vigabatrin (98) have been described.
not common, although has been described in Aicardi syn- Surgical treatment with hemispherectomy or focal resection of
drome (156). Focal epileptiform discharges have also been lesions has been reported to improve seizures in patients with
described during the periods of suppression (161). Ohtahara megalencephaly and cortical dysplasias (168–170).
stressed that the distinguishing features of suppression-burst Treatment of the seizures does not appear to prevent relent-
pattern in Ohtahara syndrome were its consistent appearance less neurologic deterioration. Subsequent brain atrophy is fre-
in both waking and sleeping states and regular appearance of quently found on imaging studies. In 16 cases followed by
periodicity (157). This is in contrast to the suppression-burst Ohtahara in 1992, all 8 survivors were profoundly mentally
pattern in EME, which can be present only during sleep, or be retarded and physically handicapped. Two had persistent
enhanced by it. seizures and six had persistent epileptiform EEG abnormalities
The suppression-burst pattern can evolve into hypsarrhyth- at time of follow-up (156).
mia between 3 and 6 months of age, coincident with the devel-
opment of more typical infantile spasms. This occurred in 8 of
16 cases followed by Ohtahara (156,157,162). Later transition REFERENCES
into slow-spike and wave morphology (characteristic of 1. Burke JB. The prognostic significance of neonatal convulsions.
Lennox–Gastaut syndrome) occurred in two survivors in the Arch Dis Child. 1954;29(146):342–345.
group. This early transition into hypsarrhythmia is less seen in 2. Craig WS. Convulsive movements occurring in the first 10 days of
EME wherein the suppression-burst pattern can persist into life. Arch Dis Child. 1960;35:336–344.
Chapter 25 ■ Seizures and Epilepsies in the Preterm and Term Neonate 475

3. Keen JH. Significance of hypocalcaemia in neonatal convulsions. 24. Holden KR, Mellits ED, Freeman JM. Neonatal seizures. I.
Arch Dis Child. 1969;44(235):356–361. Correlation of prenatal and perinatal events with outcomes.
4. McInerny TK, Schubert WK. Prognosis of neonatal seizures. Am J Pediatrics. 1982;70(2):165–176.
Dis Child. 1969;117(3):261–264. 25. Mizrahi EM, Kellaway P. Diagnosis and Management of Neonatal
5. Schulte FJ. Neonatal convulsions and their relation to epilepsy in Seizures. Philadelphia: Lippincott-Raven; 1998.
early childhood. Dev Med Child Neurol. 1966;8(4):381–392. 26. Kellaway P, Hrachovy RA. Status epilepticus in newborns: a per-
6. Massa T, Niedermeyer E. Convulsive disorders during the first spective on neonatal seizures. In: Delgado-Escueta AV, Wasterlain
three months of life. Epilepsia. 1968;9(1):1–9. CG, Treiman DM, et al., eds. Status Epilepticus. New York: Raven
7. Cadilhac J, Passouant P, Ribstein M. Convulsions in the newborn: Press; 1983:93.
EEG and clinical aspects. Electroencephalogr Clin Neurophysiol. 1959; 27. Swann JW, Moshe SL. Developmental issues in animal models. In:
11:604. Engel J, Pedley TA, eds. 2nd ed. Epilepsy: A Comprehensive Textbook.
8. Harris R, Tizard JP. The electroencephalogram in neonatal convul- New York: Raven Press; 1997:467–480.
sions. J Pediatr. 1960;57:501–520. 28. Sperber EF, Moshe SL. The effects of seizures on the hippocampus
9. Passouant P, Cadilhac J. EEG and clinical study of epilepsy during of the immature brain. In: Engel J, Morrell F, eds. Brain Plasticity
maturation in man. Epilepsia. 1962;3:14–43. and Epilepsy: The Legacy of Frank Morell. San Diego: Academic
10. Dreyfus-Brisac C, Monod N. Electroclinical studies of status Press; 2001.
epilepticus and convulsions in the newborn. In: Kellaway P, 29. Moshe SL, Sperber EF, Velisek L. Critical issues of developmental
Petersen I, eds. Neurological and Electroencephalographic seizure disorders. Physiol Res. 1993;42(3):145–154.
Correlative Studies in Infancy. New York: Grune and Stratton; 30. Moshe SL. Epileptogenesis and the immature brain. Epilepsia.
1964:250–272. 1987;28(suppl 1):S3–S15.
11. The EEG society: At the annual general meeting held on October 4, 31. Moshe SL, Albala BJ. Maturational changes in postictal refractori-
1958, the following officers were elected: Chairman: Dr. J. R. ness and seizure susceptibility in developing rats. Ann Neurol. 1983;
Davidson; Hon. Treasurer: Dr. W. A. Cobb; Hon. Secretary: Dr. S. L. 13(5):552–557.
Sherwood; Hon. Foreign Secretary: Dr. W. Grey Walter. The follow- 32. Moshe SL, Albala BJ, Ackermann RF, et al. Increased seizure
ing were elected to the council: Dr. J. A. V. Bates, Dr. M. Jefferson, susceptibility of the immature brain. Brain Res. 1983;283(1):
Dr. P. Bradley, Dr. G. Pampliglione, Dr. G. D. Dawson, Dr. J. A. 81–85.
Simpson. Summaries of papers read at the centres in London and 33. Mueller AL, Chesnut RM, Schwartzkroin PA. Actions of GABA in
Edinburgh during 1958 Proceedings Secretary: Dr. G. Parr 68, developing rabbit hippocampus: an in vitro study. Neurosci Lett.
Compton road, Winchmore hill, London, N. 21. Electroencephalogr 1983;39(2):193–198.
Clin Neurophysiol. 1959;11(3):601. 34. Ben-Ari Y, Khazipov R, Leinekugel X, et al. GABAA, NMDA and
12. Ribstein M, Walter M. Convulsion in the first month of life. Rev AMPA receptors: a developmentally regulated ‘menage a trois’.
Neurol (Paris). 1958;99(1):91–99. Trends Neurosci. 1997;20(11):523–529.
13. Rose AL, Lombroso CT. A study of clinical, pathological, and elec- 35. Michelson HB, Wong RK. Excitatory synaptic responses mediated
troencephalographic features in 137 full-term babies with a long- by GABAA receptors in the hippocampus. Science. 1991;253(5026):
term follow-up. Pediatrics. 1970;45(3):404–425. 1420–1423.
14. Lombroso CT. Seizures in the newborn. In: Vinken PJ, Bruyn GW, 36. Scantlebury MH, Ouellet PL, Psarropoulou C, et al. Freeze lesion-
eds. Handbook of Clinical Neurology. New York: Elsevier; 1974: induced focal cortical dysplasia predisposes to atypical hyperther-
189–218. mic seizures in the immature rat. Epilepsia. 2004;45(6):592–600.
15. Volpe JJ. Neonatal seizures. N Engl J Med. 1973;289(8):413–416. 37. Galanopoulou AS. Dissociated gender-specific effects of recurrent
16. Mizrahi EM. Electroencephalographic/polygraphic/video moni- seizures on GABA signaling in CA1 pyramidal neurons: role of
toring in childhood epilepsy. J Pediatr. 1984;105(1):1–9. GABA(A) receptors. J Neurosci. 2008;28(7):1557–1567.
17. Mizrahi EM, Kellaway P. Characterization and classification of 38. Galanopoulou AS. GABA receptors as broadcasters of sexually differ-
neonatal seizures. Neurology. 1987;37(12):1837–1844. entiating signals in the brain. Epilepsia. 2005;46(suppl 5):107–112.
18. Kellaway P, Hrachovy RA. Electroencephalography. In: Swainman 39. Volpe JJ. Neonatal seizures: current concepts and revised classifica-
KF, Wright FS, eds. The Practice of Pediatric Neurology. 2nd ed. St. tion. Pediatrics. 1989;84(3):422–428.
Louis: Mosby; 1982:96. 40. Murray DM, Boylan GB, Ali I, et al. Defining the gap between elec-
19. LaJoie J, Moshe SL. Neonatal seizures and neonatal epilepsy trographic seizure burden, clinical expression and staff recogni-
syndromes. In: Devinsky O, Westbrook LE, eds. Epilepsy and tion of neonatal seizures. Arch Dis Child Fetal Neonatal Ed. 2008;
Developmental Disabilities. Boston: Butterworth-Heinemann; 2002. 93(3):F187–F191.
20. Lanska MJ, Lanska DJ, Baumann RJ, et al. A population-based 41. Devinsky O, Kelley K, Porter RJ, et al. Clinical and electroen-
study of neonatal seizures in Fayette County, Kentucky. Neurology. cephalographic features of simple partial seizures. Neurology. 1988;
1995;45(4):724–732. 38(9):1347–1352.
21. Ronen GM, Penney S, Andrews W. The epidemiology of clinical 42. Watanabe K, Hara K, Miyazaki S, et al. Electroclinical studies of
neonatal seizures in Newfoundland: a population-based study. seizures in the newborn. Folia Psychiatr Neurol Jpn. 1977;31(3):
J Pediatr. 1999;134(1):71–75. 383–392.
22. Saliba RM, Annegers JF, Waller DK, et al. Incidence of neonatal 43. Goldberg HJ, Sheehy EM. Fifth day fits: an acute zinc deficiency
seizures in Harris County, Texas, 1992–1994. Am J Epidemiol. 1999; syndrome? Arch Dis Child. 1982;57(8):633–635.
150(7):763–769. 44. Lou HC, Friis-Hansen B. Arterial blood pressure elevations during
23. Scher MS, Aso K, Beggarly ME, et al. Electrographic seizures in motor activity and epileptic seizures in the newborn. Acta Paediatr
preterm and full-term neonates: clinical correlates, associated brain Scand. 1979;68(6):803–806.
lesions, and risk for neurologic sequelae. Pediatrics. 1993;91(1): 45. Watanabe K, Hara K, Miyazaki S, et al. Apneic seizures in the new-
128–134. born. Am J Dis Child. 1982;136(11):980–984.
476 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

46. Fenichel GM, Olson BJ, Fitzpatrick JE. Heart rate changes in con- 69. Giacoia GP, Ryan SG. Hyperekplexia associated with apnea and
vulsive and nonconvulsive neonatal apnea. Ann Neurol. 1980; sudden infant death syndrome. Arch Pediatr Adolesc Med. 1994;
7(6):577–582. 148(5):540–543.
47. Knauss TA, Carlson CB. Neonatal paroxysmal monorhythmic 70. Zhou L, Chillag KL, Nigro MA. Hyperekplexia: a treatable neuro-
alpha activity. Arch Neurol. 1978;35(2):104–107. genetic disease. Brain Dev. 2002;24(7):669–674.
48. Willis J, Gould JB. Periodic alpha seizures with apnea in a new- 71. Tijssen MA, Vergouwe MN, van Dijk JG, et al. Major and minor
born. Dev Med Child Neurol. 1980;22(2):214–222. form of hereditary hyperekplexia. Mov Disord. 2002;17(4):
49. Clancy RR, Legido A, Lewis D. Occult neonatal seizures. Epilepsia. 826–830.
1988;29(3):256–261. 72. Bakker MJ, van Dijk JG, van den Maagdenberg AM, et al. Startle
50. Bye A, Flanagan D. Electroencephalograms, clinical observations syndromes. Lancet Neurol. 2006;5(6):513–524.
and the monitoring of neonatal seizures. J Paediatr Child Health. 73. Andrew M, Owen MJ. Hyperekplexia: abnormal startle response
1995;31(6):503–507. due to glycine receptor mutations. Br J Psychiatry. 1997;170:
51. Mizrahi EM, Hrachovy RA, Kellaway P. Atlas of Neonatal 106–108.
Electroencephalography. 3rd ed. Philadelphia: Lippincott Williams 74. Pisani F, Cerminara C, Fusco C, et al. Neonatal status epilepticus
& Wilkins; 2003. vs. recurrent neonatal seizures: clinical findings and outcome.
52. Scher MS, Bova JM, Dokianakis SG, et al. Positive temporal sharp Neurology. 2007;69(23):2177–2185.
waves on EEG recordings of healthy neonates: a benign pattern of 75. Heimann G. Enteral absorption and bioavailability in children in
dysmaturity in pre-term infants at post-conceptional term ages. relation to age. Eur J Clin Pharmacol. 1980;18(1):43–50.
Electroencephalogr Clin Neurophysiol. 1994;90(3):173–178. 76. Guillet R, Morselli PL. Pharmacokinetics of anticonvulsants in the
53. Cukier F, Andre M, Monod N, et al. Contribution of EEG to the neonate. In: Wasterlain CG, Vert P, eds. Neonatal Seizures. New
diagnosis of intraventricular hemorrhages in the premature infant. York: Raven Press, Ltd.; 1990:257–267.
Rev Electroencephalogr Neurophysiol Clin. 1972;2(3):318–322. 77. Boer HR, Gal P. Neonatal seizures: a survey of current practice.
54. Clancy RR, Tharp BR. Positive rolandic sharp waves in the elec- Clin Pediatr (Phila). 1982;21(8):453–457.
troencephalograms of premature neonates with intraventricular 78. Painter MJ, Scher MS, Stein AD, et al. Phenobarbital compared
hemorrhage. Electroencephalogr Clin Neurophysiol. 1984;57(5): with phenytoin for the treatment of neonatal seizures. N Engl J
395–404. Med. 1999;341(7):485–489.
55. Novotny EJ Jr, Tharp BR, Coen RW, et al. Positive rolandic sharp 79. Gal P, Toback J, Boer HR, et al. Efficacy of phenobarbital monother-
waves in the EEG of the premature infant. Neurology. 1987;37(9): apy in treatment of neonatal seizures––relationship to blood levels.
1481–1486. Neurology. 1982;32(12):1401–1404.
56. Ortibus EL, Sum JM, Hahn JS. Predictive value of EEG for out- 80. Dodson WE. Special pharmacokinetic considerations in children.
come and epilepsy following neonatal seizures. Electroencephalogr Epilepsia. 1987;28(suppl 1):S56–S70.
Clin Neurophysiol. 1996;98(3):175–185. 81. Dodson WE. Antiepileptic drug utilization in pediatric patients.
57. Bye AM, Cunningham CA, Chee KY, et al. Outcome of neonates Epilepsia. 1984;25(suppl 2):S132–S139.
with electrographically identified seizures, or at risk of seizures. 82. Bourgeois BF, Dodson WE. Phenytoin elimination in newborns.
Pediatr Neurol. 1997;16(3):225–231. Neurology. 1983;33(2):173–178.
58. Tekgul H, Gauvreau K, Soul J, et al. The current etiologic profile 83. Andre M, Boutroy MJ, Bianchetti G, et al. Clonazepam in neona-
and neurodevelopmental outcome of seizures in term newborn tal seizures: dose regimens and therapeutic efficacy. Eur J Clin
infants. Pediatrics. 2006;117(4):1270–1280. Pharmacol. 1991;40(2):193–195.
59. Laroia N, Guillet R, Burchfiel J, et al. EEG background as predictor 84. Sheth RD, Buckley DJ, Gutierrez AR, et al. Midazolam in the treat-
of electrographic seizures in high-risk neonates. Epilepsia. 1998; ment of refractory neonatal seizures. Clin Neuropharmacol. 1996;
39(5):545–551. 19(2):165–170.
60. Clancy RR, Legido A. Postnatal epilepsy after EEG-confirmed 85. Maytal J, Novak GP, King KC. Lorazepam in the treatment of
neonatal seizures. Epilepsia. 1991;32(1):69–76. refractory neonatal seizures. J Child Neurol. 1991;6(4):319–323.
61. Co JP, Elia M, Engel J Jr, et al. Proposal of an algorithm for diag- 86. Claassen J, Hirsch LJ, Emerson RG, et al. Continuous EEG moni-
nosis and treatment of neonatal seizures in developing countries. toring and midazolam infusion for refractory nonconvulsive sta-
Epilepsia. 2007;48(6):1158–1164. tus epilepticus. Neurology. 2001;57(6):1036–1042.
62. Nelson KB, Broman SH. Perinatal risk factors in children with seri- 87. Boylan GB, Rennie JM, Chorley G, et al. Second-line anticonvul-
ous motor and mental handicaps. Ann Neurol. 1977;2(5):371–377. sant treatment of neonatal seizures: a video-EEG monitoring
63. Leviton A, Nelson KB. Problems with definitions and classifications study. Neurology. 2004;62(3):486–488.
of newborn encephalopathy. Pediatr Neurol. 1992;8(2):85–90. 88. Castro Conde JR, Hernandez Borges AA, Domenech Martinez E,
64. Perlman JM. Summary proceedings from the neurology group on et al. Midazolam in neonatal seizures with no response to pheno-
hypoxic-ischemic encephalopathy. Pediatrics. 2006;117(3 pt 2): barbital. Neurology. 2005;64(5):876–879.
S28–S33. 89. Gal P, Oles KS, Gilman JT, et al. Valproic acid efficacy, toxicity, and
65. Clancy RR, Sharif U, Ichord R, et al. Electrographic neonatal pharmacokinetics in neonates with intractable seizures. Neurology.
seizures after infant heart surgery. Epilepsia. 2005;46(1):84–90. 1988;38(3):467–471.
66. Parker S, Zuckerman B, Bauchner H, et al. Jitteriness in full-term 90. Sapin JI, Riviello JJ Jr, Grover WD. Efficacy of primidone for
neonates: prevalence and correlates. Pediatrics. 1990;85(1):17–23. seizure control in neonates and young infants. Pediatr Neurol.
67. Volpe JJ. Neurology of the Newborn. Philadelphia: W.B. Saunders; 1988;4(5):292–295.
2001. 91. MacKintosh DA, Baird-Lampert J, Buchanan N. Is carbamazepine
68. Coulter DL, Allen RJ. Benign neonatal sleep myoclonus. Arch an alternative maintenance therapy for neonatal seizures? Dev
Neurol. 1982;39(3):191–192. Pharmacol Ther. 1987;10(2):100–106.
Chapter 25 ■ Seizures and Epilepsies in the Preterm and Term Neonate 477

92. Hoppen T, Elger CE, Bartmann P. Carbamazepine in phenobar- 113. Rett A, Teubel R. Neugeborenen krampfe im rahmen einer
bital-nonresponders: experience with ten preterm infants. Eur J epileptisch belasten familie. Wiener Klinische Wochenschr.
Pediatr. 2001;160(7):444–447. 1964;76:609–613.
93. Koren G, Butt W, Rajchgot P, et al. Intravenous paraldehyde for 114. Leppert M, Singh N. Benign familial neonatal epilepsy with
seizure control in newborn infants. Neurology. 1986;36(1):108–111. mutations in two potassium channel genes. Curr Opin Neurol.
94. Hellstrom-Westas L, Svenningsen NW, Westgren U, et al. 1999;12(2):143–147.
Lidocaine for treatment of severe seizures in newborn infants. II. 115. Leppert M, Anderson VE, Quattlebaum T, et al. Benign familial
Blood concentrations of lidocaine and metabolites during intra- neonatal convulsions linked to genetic markers on chromosome
venous infusion. Acta Paediatr. 1992;81(1):35–39. 20. Nature. 1989;337(6208):647–648.
95. Hellstrom-Westas L, Westgren U, Rosen I, et al. Lidocaine for 116. Singh NA, Charlier C, Stauffer D, et al. A novel potassium chan-
treatment of severe seizures in newborn infants. I. Clinical effects nel gene, KCNQ2, is mutated in an inherited epilepsy of new-
and cerebral electrical activity monitoring. Acta Paediatr Scand. borns. Nat Genet. 1998;18(1):25–29.
1988;77(1):79–84. 117. Claes LR, Ceulemans B, Audenaert D, et al. De novo KCNQ2
96. Malingre MM, Van Rooij LG, Rademaker CM, et al. Development mutations in patients with benign neonatal seizures. Neurology.
of an optimal lidocaine infusion strategy for neonatal seizures. 2004;63(11):2155–2158.
Eur J Pediatr. 2006;165(9):598–604. 118. Ryan SG, Wiznitzer M, Hollman C, et al. Benign familial neona-
97. Barr PA, Buettiker VE, Antony JH. Efficacy of lamotrigine in tal convulsions: evidence for clinical and genetic heterogeneity.
refractory neonatal seizures. Pediatr Neurol. 1999;20(2):161–163. Ann Neurol. 1991;29(5):469–473.
98. Baxter PS, Gardner-Medwin D, Barwick DD, et al. Vigabatrin 119. Miles DK, Holmes GL. Benign neonatal seizures. J Clin
monotherapy in resistant neonatal seizures. Seizure. 1995;4(1): Neurophysiol. 1990;7(3):369–379.
57–59. 120. Zonana J, Silvey K, Strimling B. Familial neonatal and infantile
99. Silverstein FS, Ferriero DM. Off-label use of antiepileptic drugs seizures: an autosomal-dominant disorder. Am J Med Genet.
for the treatment of neonatal seizures. Pediatr Neurol. 2008; 1984;18(3):455–459.
39(2):77–79. 121. Plouin P, Anderson VE. Benign familial and nonfamilial neonatal
100. Hodson A. A case for long-term treatment with anticonvulsants. seizures. In: Roger J, Bureau M, Dravet C, et al., eds. Epileptic
Pharmacotherapy. 1985;5(5):274–277. Syndromes in Infancy, Childhood and Adolescence. 4th ed.
101. Gal P. Anticonvulsant therapy after neonatal seizures––how long Montrouge, France: John Libbey Eurotext Ltd.; 2005:3–15.
should it be continued? I. A case for early discontinuation of 122. Proposal for classification of epilepsies and epileptic syndromes.
anticonvulsants. Pharmacotherapy. 1985;5(5):268–273. commission on classification and terminology of the interna-
102. Hellstrom-Westas L, Blennow G, Lindroth M, et al. Low risk of tional league against epilepsy. Epilepsia. 1985;26(3):268–278.
seizure recurrence after early withdrawal of antiepileptic treat- 123. Ronen GM, Rosales TO, Connolly M, et al. Seizure characteristics
ment in the neonatal period. Arch Dis Child Fetal Neonatal Ed. in chromosome 20 benign familial neonatal convulsions.
1995;72(2):F97–F101. Neurology. 1993;43(7):1355–1360.
103. Guillet R, Kwon JM. Prophylactic phenobarbital administration 124. Hirsch E, Velez A, Sellal F, et al. Electroclinical signs of
after resolution of neonatal seizures: survey of current practice. benign neonatal familial convulsions. Ann Neurol. 1993;34(6):
Pediatrics. 2008;122(4):731–735. 835–841.
104. Ronen GM, Buckley D, Penney S, et al. Long-term prognosis in 125. Bye AM. Neonate with benign familial neonatal convulsions:
children with neonatal seizures: a population-based study. recorded generalized and focal seizures. Pediatr Neurol. 1994;
Neurology. 2007;69(19):1816–1822. 10(2):164–165.
105. Brunquell PJ, Glennon CM, DiMario FJ Jr, et al. Prediction of 126. Dobrescu O, Larbrisseau A. Benign familial neonatal convul-
outcome based on clinical seizure type in newborn infants. sions. Can J Neurol Sci. 1982;9(3):345–347.
J Pediatr. 2002;140(6):707–712. 127. Camfield PR, Dooley J, Gordon K, et al. Benign familial neonatal
106. Hill A, Volpe JJ. Seizures, hypoxic-ischemic brain injury, and convulsions are epileptic. J Child Neurol. 1991;6(4):340–342.
intraventricular hemorrhage in the newborn. Ann Neurol. 1981; 128. Pettit RE, Fenichel GM. Benign familial neonatal seizures. Arch
10(2):109–121. Neurol. 1980;37(1):47–48.
107. Bergman I, Painter MJ, Hirsch RP, et al. Outcome in neonates 129. Cronin C, Cosgrove J. Benign familial neonatal convulsions. Ir
with convulsions treated in an intensive care unit. Ann Neurol. Med J. 1986;79(11):325–326.
1983;14(6):642–647. 130. Maihara T, Tsuji M, Higuchi Y, et al. Benign familial neonatal
108. Lombroso CT. Prognosis in neonatal seizures. Adv Neurol. 1983; convulsions followed by benign epilepsy with centrotemporal
34:101–113. spikes in two siblings. Epilepsia. 1999;40(1):110–113.
109. Mizrahi EM. Neonatal seizures: the need for precise clinical char- 131. Peretz A, Degani N, Nachman R, et al. Meclofenamic acid and
acterization. Ann Neurol. 1987;21(3):312–313. diclofenac, novel templates of KCNQ2/Q3 potassium channel
110. Legido A, Clancy RR, Berman PH. Neurologic outcome after openers, depress cortical neuron activity and exhibit anticonvul-
electroencephalographically proven neonatal seizures. Pediatrics. sant properties. Mol Pharmacol. 2005;67(4):1053–1066.
1991;88(3):583–596. 132. Rundfeldt C, Netzer R. The novel anticonvulsant retigabine
111. Rowe JC, Holmes GL, Hafford J, et al. Prognostic value of the activates M-currents in Chinese hamster ovary-cells transfected
electroencephalogram in term and preterm infants following with human KCNQ2/3 subunits. Neurosci Lett. 2000;282(1–2):
neonatal seizures. Electroencephalogr Clin Neurophysiol. 1985; 73–76.
60(3):183–196. 133. Wickenden AD, Yu W, Zou A, et al. Retigabine, a novel anticon-
112. Bjerre I, Corelius E. Benign familial neonatal convulsions. Acta vulsant, enhances activation of KCNQ2/Q3 potassium channels.
Paediatr Scand. 1968;57(6):557–561. Mol Pharmacol. 2000;58(3):591–600.
478 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

134. Navelet Y, D’Allest AM, Dehan M, et al. Are convulsions on the 153. du Plessis AJ, Kaufmann WE, Kupsky WJ. Intrauterine-onset
fifth day of life a distinct clinical and electrophysiological entity? myoclonic encephalopathy associated with cerebral cortical dys-
(author’s transl.). Rev Electroencephalogr Neurophysiol Clin. 1977; genesis. J Child Neurol. 1993;8(2):164–170.
7(3):366–370. 154. Djukic A, Lado FA, Shinnar S, et al. Are early myoclonic
135. Plouin P. Benign familial neonatal seizures and benign idiopathic encephalopathy (EME) and the Ohtahara syndrome (EIEE)
neonatal seizures. In: Engel J, Pedley TA, eds. Epilepsy: A independent of each other? Epilepsy Res. 2006;70(suppl 1):68–76.
Comprehensive Textbook. Philadelphia: Lippincott-Raven 155. Ohtahara S, Ishida T, Oka E, et al. On the specific age dependent
Publishers; 1997:2247–2255. epileptic syndrome: the early-infantile epileptic encephalopathy
136. Herrmann B, Lawrenz-Wolf B, Seewald C, et al. 5th day convul- with suppression-bursts [in Japanese]. No To Hattatsu. 1976;8:
sions of the newborn infant in rotavirus infections. Monatsschr 270–280.
Kinderheilkd. 1993;141(2):120–123. 156. Yamatogi Y, Ohtahara S. Early-infantile epileptic encephalopathy
137. Watanabe K, Miura K, Natsume J, et al. Epilepsies of neonatal with suppression-bursts, Ohtahara syndrome; its overview refer-
onset: seizure type and evolution. Dev Med Child Neurol. 1999; ring to our 16 cases. Brain Dev. 2002;24(1):13–23.
41(5):318–322. 157. Ohtahara S, Yamatogi Y. Ohtahara syndrome: with special refer-
138. Guerra MP, Wilson GA, Boylan GB, et al. An unusual presenta- ence to its developmental aspects for differentiating from early
tion of fifth-day fits in the newborn. Pediatr Neurol. 2002;26(5): myoclonic encephalopathy. Epilepsy Res. 2006;70(suppl 1):
398–401. S58–S67.
139. Pryor DS, Don N, Macourt DC. Fifth day fits: a syndrome of 158. Schlumberger E, Dulac O, Plouin P. Early-infantile epileptic syn-
neonatal convulsions. Arch Dis Child. 1981;56(10):753–758. drome(s) with suppression-burst: nosological considerations. In:
140. Aicardi J, Goutieres F. Neonatal myoclonic encephalopathy Roger J, Bureau M, Dravet C, et al., eds. Epileptic Syndromes in
(author’s transl.). Rev Electroencephalogr Neurophysiol Clin. Infancy, Childhood and Adolescence. Montrouge, France: John
1978;8(1):99–101. Libbey & Company Ltd.; 1992:35–42.
141. Cavazzuti GB, Nalin A, Ferrari F, et al. Encefalopatia epilettica ad 159. Fusco L, Pachatz C, Di Capua M, et al. Video/EEG aspects of
insorgenza neonatale. Clin Pediatr. 1978;60:239–246. early-infantile epileptic encephalopathy with suppression-bursts
142. Martin HJ, Deroubaix-Tella P, Thelliez PH. Encéphalopathie (Ohtahara syndrome). Brain Dev. 2001;23(7):708–714.
épileptique néonatale à bouffées périodiques. Rev EEG 160. Clarke M, Gill J, Noronha M, et al. Early infantile epileptic
Neurophysiol. 1981;11:397–403. encephalopathy with suppression burst: Ohtahara syndrome.
143. Dalla Bernardina B, Dulac O, Fejerman N, et al. Early myoclonic Dev Med Child Neurol. 1987;29(4):520–528.
epileptic encephalopathy (E.M.E.E.). Eur J Pediatr. 1983;140(3): 161. Al-Futaisi A, Banwell B, Ochi A, et al. Hidden focal EEG seizures
248–252. during prolonged suppressions and high-amplitude bursts in
144. Ozyurek H, Turanli G, Aliefendioglu D, et al. Repetitive EEG early infantile epileptic encephalopathy. Clin Neurophysiol. 2005;
recordings are necessary for the diagnosis of early myoclonic 116(5):1113–1117.
encephalopathy. Neurol India. 2005;53(2):235–237. 162. Ohtahara S, Ohtsuka Y, Yamatogi Y, et al. The early-infantile
145. Aicardi J, Ohtahara S. Severe neonatal epilepsies with suppres- epileptic encephalopathy with suppression-burst: developmental
sion-burst pattern. In: Roger J, Bureau M, Dravet C, et al., eds. aspects. Brain Dev. 1987;9(4):371–376.
Epileptic Syndromes in Infancy, Childhood and Adolescence. 4th 163. Miller SP, Dilenge ME, Meagher-Villemure K, et al. Infantile
ed. Montrouge, France: John Libbey Eurotext Ltd.; 2005:39–50. epileptic encephalopathy (Ohtahara syndrome) and migrational
146. Oka E, Ishida S, Ohtsuka Y, et al. Neuroepidemiological study of disorder. Pediatr Neurol. 1998;19(1):50–54.
childhood epilepsy by application of international classification 164. Tominaga I, Kaihou M, Kimura T, et al. Early infantile epileptic
of epilepsies and epileptic syndromes (ILAE, 1989). Epilepsia. encephalopathy (Ohtahara syndrome) with poly-microgyria. Rev
1995;36(7):658–661. Neurol (Paris). 1993;149(10):532–535.
147. Wang PJ, Lee WT, Hwu WL, et al. The controversy regarding 165. Tatsuno M, Hayashi M, Iwamoto H, et al. Autopsy case of Leigh’s
diagnostic criteria for early myoclonic encephalopathy. Brain encephalopathy with wide lesions in central nervous system and
Dev. 1998;20(7):530–535. early infantile epileptic encephalopathy with burst suppression.
148. Lombroso CT. Early myoclonic encephalopathy, early infantile No To Hattatsu. 1984;16(1):68–75.
epileptic encephalopathy, and benign and severe infantile 166. Williams AN, Gray RG, Poulton K, et al. A case of Ohtahara syn-
myoclonic epilepsies: a critical review and personal contribu- drome with cytochrome oxidase deficiency. Dev Med Child Neurol.
tions. J Clin Neurophysiol. 1990;7(3):380–408. 1998;40(8):568–570.
149. Dalla Bernardina B, Aicardi J, Goutieres F, et al. Glycine 167. Ohno M, Shimotsuji Y, Abe J, et al. Zonisamide treatment of early
encephalopathy. Neuropadiatrie. 1979;10(3):209–225. infantile epileptic encephalopathy. Pediatr Neurol. 2000;23(4):
150. Vigevano F, Maccagnani F, Bertini E, et al. Encefalopatia mio- 341–344.
clonica precoce associata ad alti livelli di acido propionico nel 168. Pedespan JM, Loiseau H, Vital A, et al. Surgical treatment of an
siero. Boll Lega Ital Epil. 1982;39:181–182. early epileptic encephalopathy with suppression-bursts and focal
151. Aukett A, Bennett MJ, Hosking GP. Molybdenum co-factor defi- cortical dysplasia. Epilepsia. 1995;36(1):37–40.
ciency: an easily missed inborn error of metabolism. Dev Med 169. Komaki H, Sugai K, Maehara T, et al. Surgical treatment of early-
Child Neurol. 1988;30(4):531–535. infantile epileptic encephalopathy with suppression-bursts associ-
152. Spreafico R, Angelini L, Binelli S, et al. Burst suppression and ated with focal cortical dysplasia. Brain Dev. 2001;23(7):727–731.
impairment of neocortical ontogenesis: electroclinical and neu- 170. Hmaimess G, Raftopoulos C, Kadhim H, et al. Impact of early
ropathologic findings in two infants with early myoclonic hemispherotomy in a case of Ohtahara syndrome with left
encephalopathy. Epilepsia. 1993;34(5):800–808. parieto-occipital megalencephaly. Seizure. 2005;14(6):439–442.
CHAPTER
Seizures and Epilepsy in
Infants to Adolescents
DOUGLAS R. NORDLI, JR., JAMES J. RIVIELLO, JR., AND ERNST NIEDERMEYER
26
BASIC CONSIDERATIONS Principal Differences between Interictal
and Ictal Discharges
Definition and Prevalence
A distinction is made between ictal and interictal paroxysmal
There is no disease named “epilepsy.” Rather, epileptic seizures activity. This logical differentiation, however, is beset with con-
are abnormal reactions of the brain caused by a large number fusion. It is sometimes difficult or even impossible to distin-
of diseases. The entire brain or parts of it may be involved; the guish between ictal-clinical, ictal-subclinical, and interictal
extent of involvement largely determines the seizure type. paroxysmal EEG activity, even using neuropsychological tests.
Epilepsy originates in the brain, but systemic disorders and Computerized testing demonstrates the existence of a gray zone
resulting metabolic or toxic effects may cause secondary between interictal and ictal paroxysmal activities (e.g., transient
encephalopathies, and thus, secondary epileptic seizures. cognitive impairment).
Genetic predisposition also plays a very important role.
The prevalence of epileptic seizures has been estimated to Problems of Terminology
be between 0.5% (core group) and 5% (fringe group with at Definitions of EEG events are indispensable, but they are also
least one seizure during life). More precise figures of Juul- quite difficult to arrive at and are often unsatisfactory. Attempts
Jensen and Foldspang (1) lie between 1.27% and 2.44%. to reassess EEG terminology have been made periodically; they
Gumnit (2) estimates that core and fringe group may add up often result in neologistic construction of terms that fail to
to 9% of the population. receive general acceptance. EEG terminology is filled with
An overview of various epilepsy prevalence studies (3) shows widely used, popular, and, often quite sloppy and inaccurate
fairly consistent figures: Mumbai, India 3.7; Rochester, Minnesota, terms. Nevertheless, most electroencephalographers know what
6.66; Denmark, 6.9; Poland, 7.8; Nigeria, 5.3; Mississippi, 6.78; and is meant by such expressions in the professional jargon.
China, 4.57 (prevalence per 1000). Similar figures were presented One could argue that the busy electroencephalographer will
by Greulich and Gerber (4) and Hauser (5). always be satisfied with the nomenclature learned during the
training period, whereas the academicians among electroen-
Introduction and Historical Remarks cephalographers constantly strive for an unobjectionable and
Electroencephalography (EEG) has revolutionized the entire field crystal clear terminology free from linguistic flaws and capable
of epileptology. No one can deny that ingenious and even great of defining an EEG event from a variety of aspects. This view,
concepts arose in the pre-EEG era; clinical acumen and keen however, is oversimplified. Academia sometimes has a vested
analysis of the observed phenomena resulted in remarkable interest in certain terms; it may bitterly resent new terms rec-
descriptions that have stood the test of time. Indeed, focal motor ommended by international committees.
seizures, infantile spasms, rolandic seizures, and absence seizures, A preliminary proposal by the International Federation
to list but a few, were all recognized prior to the development of Societies for Electroencephalography and Clinical Neuro -
of EEG. Conventional EEG has opened new vistas and modern physiology (6) was followed by a definitive proposal (7) and
EEG techniques, especially the prolonged EEG monitoring of a glossary of terms used by clinical electroencephalogra-
people with epilepsy, have made further important epileptologic phers (8). This glossary was updated in 1999 (9). The hand-
contributions. These noninvasive techniques are complemented book catalogue of Dutertre (10) deserves special admiration.
by invasive recordings from cortex and deep structures that con- Unfortunately, the definition of electrophysiologic events can
tinue to provide insights into fundamentals of epilepsy. never do justice to all facets of the phenomenon. The ques-
We review the role of the EEG in the epilepsies, including the tion then arises as to whether we must rigidly follow the new
correlation of the semiology with the electrographic findings nomenclature. A negativistic attitude must be castigated, but
(electroclinical correlation) that are contributory to the EEG electroencephalographers should not be pressured to strictly
diagnosis of epileptic phenomena in ictal states or the interictal adhere to newly proposed terms as time may be needed for
period in various epileptic conditions. these terms to achieve consensus.
The interictal paroxysmal phenomena are discussed first. As to epileptic discharges, there has been considerable dis-
These discharges represent the basic elements of more complex unity in the ranks of electroencephalographers. The term
ictal patterns. epileptic discharge has been criticized because such discharges

479
480 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

may occur in the absence of clinical seizure manifestations or in lead (11). Spikes appear to be hypersynchronous events due to
individuals who have never had seizures. The same is true for excessive simultaneous neuronal discharge. This view, however,
the widely used term seizure discharge; even paroxysmal dis- is not congruent with the desynchronized (low voltage) EEG
charge, certainly a more cautious and unassuming term, has not aspect of “electrodecremental seizures” or the electroincre-
found general acceptance. The term epileptiform discharge is an mental initiation of focal or generalized attacks. Definitions
elegant solution since it addresses the critical issue: an epilepti- such as “epilepsy is paroxysmal hypersynchrony” (12) may be
form discharge demonstrates an underlying epileptic patho- correct on the neuronal level but partly incorrect from the
physiology but its presence alone does not make the diagnosis viewpoint of the “macro-EEG.”
of epilepsy. The diagnosis of epilepsy remains a clinical one, Basic neurophysiologists have been cautioning against the
which must be based on actual clinical events. casual use of terms such as synchronization and desynchronization.
Low-voltage fast rhythms are not necessarily desynchronized and
INTERICTAL EPILEPTIFORM DISCHARGES may be associated with enhancement and synchronization within
intracortical networks.
Spike (Single or Random Spike) On the basis of long-term monitoring and computer analy-
According to IFSECN (8), a spike is a transient, clearly distin- sis, Gotman (13) has raised the question as to whether interic-
guished from the background activity, with pointed peak at con- tal spikes are in reality postictal. Gotman (13) and Gotman and
ventional paper speed and a duration from 20 to under 70 msec; Marciani (14) observed marked and prolonged increase of
the main component is generally negative and the amplitude is interictal spikes in a patient after a seizure (awake as well as
variable. asleep). Drug levels had little influence on the rate of spike
The distinction from the background activity is based on activity, and there was no increase of spiking prior to a seizure.
wave morphology and amplitude. In many cases, spikes stand The multiphasic character of a single spike warrants partic-
out against the background because of their high voltage. If the ular emphasis. A sequence of a minor positive (the “positive
voltage of spikes and background activity is approximately prespike”), a major negative, and a second minor positive com-
equal, the faster character (i.e., the shorter duration) of the spike ponent (Fig. 26.1) is typical in most instances. A slow negative
is its distinctive feature. Is it possible that a spike of 50 msec component may trail the spike discharge and often attain about
duration and moderate amplitude may be embedded in 20 Hz the same amplitude as the negative main component of the
activity (50 msec wave duration), for instance, in an epileptic spike. This trailing slow component of a single spike should not
with considerable drug-induced fast activity? Under these cir- be regarded as evidence of a spike-wave complex. Dutertre (10)
cumstances, the spike activity may be undetectable. In such a in his handbook catalogue shows examples of single spike-wave
case, a fast paper speed could demonstrate the morphologic complexes that one could just as well describe as compounded
features of the spike (multiphasic and more pointed character or multiphasic single spikes. From the vantage point of basic
with a dominant negative phase) in contrast with the more neurophysiology, there is some reason to presume that the trail-
monotonous appearance of the fast waves. In the digital era, ing slow wave is caused by the same type of hyperpolarization
paper speed is reflected in the sweep time. demonstrated by Pollen et al. (15) in the experimental analysis
Spikes have many characteristics in common and yet there are of the spike-wave discharge.
also remarkable inter- and intraindividual variations (Fig. 26.1) The electroencephalographer very seldom finds single spikes
and inconsistencies between one spike and the next in the same on the scalp with predominant positive component (16,17).
Positive single spikes are more common in electroencephalo-
graphic and depth recordings; on the scalp, predominant posi-
tivity of single spikes raises the question of defective superficial
cortical laminae. Following surgery of cortical arteriovenous
(AV) malformations or after traumatic laceration (18), such
positive spikes may be present occasionally. With dipole forma-
tion (especially in benign rolandic epilepsy), a positive spike
discharge may be found in a moderate distance from the nega-
tive spike focus (19,20).
Spikes represent the basic element of epileptiform activity in
the EEG. A unitarian view that all spikes mean a hidden or overt
epileptic event would be erroneous. The fine semiology of
spikes is extremely important and the EEG interpreter must
consider the following questions:
Figure 26.1 Various wave morphologies of spikes recorded from the What is the precise wave morphology?
same patient (age 6 years). Upper lead, T3–T5; lower lead, T5–O1. (First Where do the spikes occur?
and second tracing from the top, continued in tracings 3 and 4, respec- What is the patient’s age?
tively 5 and 6.) Note multiphasic character of spikes with predomi- What is the state of awareness?
nance of the negative phase and occasional formation of double spikes. Is there any possibility of an artifact of similar appearance?
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 481

Is there any possibility of a physiologic potential of similar


appearance?
Discrepancies between spikes and behavioral epileptic events
are not uncommon at all. This highly complex situation has
been reinvestigated by Niedermeyer (21).

Wave Morphology
The largest and most pronounced spikes are not necessarily
associated with more serious epileptic seizure disorders. On the
contrary, rolandic spikes in a child age 4 to 10 years are very
prominent; however, the seizure disorder itself is usually quite
benign or there may be no clinical seizures at all. Alternatively,
“small sharp spikes” are indeed marginal spike discharges and
will be discussed separately.

Spatial Distribution
In childhood, occipital spikes are, in general, the most benign
spike discharges, with less than 50% having clinical seizures;
rolandic central–midtemporal–parietal spikes are also quite
benign, whereas frontal spikes or multifocal spikes are more
epileptogenic.

Age
The significance of the age factor is enormous. From the spikes
of an epileptic newborn to a seizure focus of old age, age-deter- Figure 26.2 Coexistence of rolandic spikes and physiologic vertex
mined varieties of spikes can be distinguished. waves in light sleep (8-year-old boy, attacks of abdominal pain, no
proven epileptic seizures). Right centroparietal spikes with occasional
Level of Awareness spread to the left are marked X; typical examples of a vertex wave are
Random spikes may occur at any state of waking–sleeping cycle marked O.
and occur even in rapid eye movement (REM) sleep when bilat-
eral synchronous bursts of spikes or spike waves are usually
suppressed.
in the more uncommon case of epileptiform spike discharges
Repercussions on Perceptual Function over the vertex, the differentiation from physiologic vertex
Shewmon and Erwin (22,23) investigated the impact of spikes sharp waves is not difficult.
located over the occipital region and its vicinity on visual percep- The physiologic nature of the occipital lambda waves and
tion and reaction time in three subjects with posterior spikes. “lambdoid” activity (positive occipital transients of sleep)
Their work shows that the visuomotor reaction time was pro- must also be considered. The main component of this pattern
longed (or the response was absent) when the task was time is positive.
locked to the spike. Furthermore, “spike-induced dysfunction
was most pronounced when either response hand or visual field Distinction from Artifacts of Similar Appearance
of stimulus was contralateral to the spike” (23). In a further study, This distinction depends on the electroencephalographer’s
Shewmon and Erwin (24) emphasized the significance of the experience and is usually an easy one.
trailing slow wave that follows a single spike discharge. The per- The interpretation of the clinical significance of spikes can
ceptive disturbance was particularly pronounced when the task be extremely difficult and depends on the electroencephalogra-
was presented during the slow wave. These fascinating findings pher’s experience in the art of reading the EEG recording and
indicate that even single spikes of rapid duration may be capable also on the clinical understanding of epileptologic problems.
of momentary impairment of the cortical visual function. Extensive personal laboratory experience is just as essential as
scientific knowledge in interpreting the EEG.
Distinction from Similar Physiologic Patterns
This differentiation is particularly important in the case of ver- Are Interictal Spikes Nothing but EEG Signals?
tex sharp waves during deep drowsiness and stage 2 of light Our earlier view (found in this chapter in previous editions)—
non-REM sleep. In childhood (after age 4), these waves may namely that single spikes are an EEG signal and nothing else—
have a particularly spiky appearance and may be misinterpreted is in need of some correction. This seems to be particularly true
as paroxysmal spikes. for “negative myoclonus” (ictally suppressed motor activity),
Physiologic sharp or spiky vertex waves are usually quite eas- examples of which have been presented by Gambardella et al.
ily distinguished from rolandic spikes in sleep (Fig. 26.2). Even (25) and Werhahn and Noachtar (26) (also see Ref. 27). A single
482 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

spike may also cause changes in cerebral blood flow and increase
of metabolic demands (28), demonstrated with the simultane-
ous use of EEG and functional magnetic resonance imaging
(fMRI).
Even behavioral changes with impaired test performance
may be associated with single interictal spikes (29,30). Further
information is found in the work of Fisch (27).

Sharp Waves
According to IFSECN (8), a sharp wave is a transient, clearly
distinguished from background activity, with pointed peak at
conventional paper speeds and duration of 70 to 200 msec, that
is, more than approximately 1/14 to 1/5 sec. The main compo-
nent is generally negative relative to the other components. The
term is restricted to epileptiform discharges and does not apply
to distinctive physiologic waveforms, such as vertex sharp tran-
sients, lambda waves, and POSTS.
Jasper (31) pointed out that the rising phase of the sharp wave
is of the same order of magnitude as in spikes, but the descending
phase is prolonged. The configuration with a steeper ascending
phase, however, is not always present. Examples of sharp waves
are shown in Figure 26.3.
A very unusual type of repetitive slow sharp wave activity
Figure 26.4 An example of a very slow sharp wave (blunted sharp
has been reported in prematurely born infants with intraven-
wave), maximal over T6, obtained from a 6-year-old boy with cranio-
tricular hemorrhage; these discharges show predominantly
cerebral trauma at age 4 followed by seizures.
positive polarity and are recorded mainly over the rolandic
region (32,33). The maximum of this activity is sometimes
found over the vertex. even a small biphasic spike discharge may precede the much
Spikes and sharp waves are neurophysiologically closely larger sharp wave. Some sharp waves even exceed the maximum
related phenomena; both of them are typical epileptiform dis- length of 200 msec of the IFSECN definition (8) (Fig. 26.4), also
charges and highly suggestive of an epileptic seizure disorder, named “slow sharp waves” (35) or in cases of particularly long
although both phenomena may occur in patients without a his- duration, “blunted sharp waves.” Others become very complex.
tory of seizure disorder. They consist of a constantly varying number of components;
Sharp waves are usually found as random focal discharges; such compounded sharp waves may occur as the periodic dis-
most anterior temporal spikes are, in a strict sense, sharp waves. charges (periodic lateralized epileptiform discharges) described
This is also true for most benign rolandic spikes of childhood. by Chatrian et al. (36).
Sharp waves are more seldom found in generalized synchro- It is certainly not incorrect to use the terms spikes and sharp
nous bursts where single spikes, spike waves, and polyspikes waves synonymously when discussing the clinical significance
predominate. of a focal transient.
It has been contemplated that sharp waves on the scalp cor-
respond with spikes in the depth or on the cortex. Combined Multiple Spike Complex or Polyspike Complex
depth and scalp recording clearly refutes this view (34). One can This discharge type represents a complex of spikes and may also
detect spikes as well as sharp waves in depth recordings; a deep be called a polyspike complex. In modern terminology (8), the
sharp wave usually corresponds with a sharp wave on the scalp term multiple spike complex is preferred because “polyspikes” is an
if there is any corresponding scalp activity at all. etymologic hybrid. It has been defined as a complex paroxysmal
The long duration of a sharp wave permits better insight EEG pattern with close association of two or more diphasic spikes
into the multiphasic character of this potential. A small preced- occurring more or less rhythmically in bursts of variable duration,
ing positive component may be very fast and qualify as a spike; generally with large amplitudes (10) (Figs. 26.5 and 26.6).
Polyspike bursts are readily elicited by electrical stimulation
of single depth leads, especially in limbic regions. On the scalp,
however, polyspikes occur mostly as bilateral or generalized
synchronous discharges. Exceptional focal polyspikes are occa-
sionally encountered; these usually have a frontal maximum,
except for occipital accentuation in hypsarrhythmia. Polyspikes
Figure 26.3 Recurrent sharp wave recorded from a 61-year-old patient. and also polyspike-wave complexes are sometimes associated
Upper lead, F3–C3; lower lead, C3–P3. A prominent negative phase is fol- with concomitant myoclonus, especially in primary generalized
lowed by a large and relatively slow trailing positive wave. epilepsy and in photosensitive individuals with this type of
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 483

Figure 26.6 The differentiation between polyspikes and drug-induced


fast activity may be difficult. The run of beta waves (center) certainly
falls into the category of fast activity, but the more outstanding and
burst-like run (right) is more likely to represent polyspike activity. A
Figure 26.5 Polyspikes in light non-rapid eye movement (non-REM) definitive statement depends on the impressions derived from the entire
sleep, especially over the central region, associated with K complexes. record.
(With permission of AMA.)

seizure disorder. Children with Lennox–Gastaut syndrome may bursts of more than 5-second duration are usually associated
also show association of polyspikes and myoclonus. with tonic seizures and thus represent an ictal pattern.
The obvious misnomer “grand mal discharges” is based on
Runs of Rapid Spikes certain similarities with the ictal EEG of a generalized tonic–
This pattern has been described as “grand mal discharge” clonic seizure. In a patient population with generalized
(37,38), “fast paroxysmal rhythms” (39), “rhythmic spikes” (40), tonic–clonic seizures, runs of rapid spikes as an interictal pat-
and “generalized paroxysmal fast activity” (41). It is seen only in tern are a very rare finding. This has been confirmed by the data
sleep and occurs in older children, adolescents, and younger of Chayasirisobhon et al. (42). The inappropriateness of the
adults (Fig. 26.7). It consists of bursts of spike discharges at a rate term grand mal discharge was stressed by Rodin et al. (43), who
from 10 to 25 Hz, usually generalized but with a well-defined thought that this pattern occurred in patients with primary
maximum over frontal regions; it may even be confined to the generalized epilepsy who suffered from more than one type of
frontal leads. The voltage is in the medium to high range, often seizure and especially from akinetic seizures. As already noted
exceeding 100 or even 200 V. The discharge rate is in most cases by Gastaut et al. (40), this discharge is very typical in patients
somewhat irregular. The bursts last for about 2 to 10 seconds; with Lennox–Gastaut syndrome; it is hardly ever found outside

Figure 26.7 A run of rapid spikes in a 19-year-old epilep-


tic patient (Lennox–Gastaut syndrome). Note anterior
maximum of the discharge. A few slow spike wave com-
plexes are also seen (right temporo-occipital region).
484 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

this clinical entity (44–46). It may be found in very rare simple visual analysis at 30 mm/sec paper speed; special record-
instances of posttraumatic epilepsy with EEG traits of the ing methods will better reveal these extra components. Gastaut
Lennox–Gastaut syndrome (47). Brenner and Atkinson (41), and Hunter (55) and Cohn (56) and Weir (57) have elucidated
however, insist that runs of rapid spikes (“generalized paroxys- the components of this pattern. Most important is a spike of
mal fast activity”) may be found outside the Lennox–Gastaut positive polarity during the descending portion of the slow
syndrome. In our experience, runs of rapid spikes may occur in wave (57), becoming evident as notching under usual recording
all “imitators” of the Lennox–Gastaut syndrome (48,49). conditions.
Little is known about the underlying neurophysiologic mech- On the basis of experimental studies of cortical faradiza-
anisms of this pattern. It is clearly a frontal lobe discharge, and tion, Jung and Toennies (58) stressed the inhibitory character
the associated tonic seizures are probably arising from the pre- of the slow wave following a spike discharge (“Bremswelle”).
motor and supplementary motor portions of the frontal lobes. Experimentally induced spike-wave-like activity in cats, elicited
There could be certain similarities to the electrodecremental by electrical stimulation of thalamic intralaminar structures, was
seizure patterns, which also originate most commonly from the studied with intracellular cortical microelectrodes (15,59,60).
frontal lobe (50). This electrodecremental pattern may actually According to Pollen (59), all components of the spike-wave com-
be associated with high-frequency discharges (high-frequency plex were the “consequence of postsynaptic activity.” This author
oscillations) that are not detectable at the scalp, giving the false points out that “the initial positivity which may precede a nega-
impression of a decrement in cortical activity (51). tive spike (depending on the intensity of the stimulation) results
Unfortunately, those electroencephalographers who never from depolarization of middle and deep lying neurons, whereas
use sleep portions in routine records will never see this fascinat- the surface negative spike is the result of EPSP (excitatory postsy-
ing pattern, which is definitely in need of further basic research. naptic potential) generation primarily upon apical dendrites of
vertically oriented pyramidal type neurons.” According to Pollen
Spike-and-Slow -Wave Complex (59), the long surface negative wave is generated by inhibitory
(Classical 3 Hz Spike Wave) postsynaptic potentials (IPSPs) predominantly on or near the
Classical 3 Hz spike-wave complexes are widely known even out- soma of similar pyramidal cells. Thus, the slow wave indeed
side the community of electroencephalographers. It is officially appears to be related to inhibitory impulses.
termed spike-and-slow-wave complex (8); this term comprises all In this view, the spike-wave discharge apparently represents an
types of spike-wave complexes, which are listed separately alternating succession of excitation and inhibition. The clinical
because of markedly differing associated clinical–epileptologic ictal activities are thus constantly curbed by intervening inhibitory
conditions. The official definition (8) is quite simple: “A pattern impulses that prevent the discharge from progressing into massive
consisting of a spike followed by a slow wave.” Older terms such downward discharges with motor effects (polyspikes with massive
as spike-and-dome complex, wave-and-spike complex, and dart- myoclonus) or to a full-blown generalized tonic–clonic seizure.
and-dome complex are seldom used these days and should be For this reason, motor manifestations of ictal episodes character-
discouraged. The abridged term 3 Hz spike wave is certainly ized by spike waves are almost always inconspicuous or modest.
acceptable, because the omitted word complex is automatically Furthermore, spike-wave bursts rarely proceed into generalized
implied. tonic–clonic seizures; such rare exceptions have been demon-
Spike-wave complexes of the classical variety (3 Hz) have strated by Halász (61) and Niedermeyer (62).
been described as the EEG pattern of absence seizures (38). The distinction between classical (3 Hz), slow (1 to 2.5 Hz),
Since that time, the 3 Hz spike-wave discharge has been equated and fast (4 to 5 Hz) spike-wave complexes and the smaller
with absence epilepsy. Regrettable mistakes have occurred due to 6 Hz spike-wave discharge is justified on the basis of different
lack of communication between electroencephalographers and clinical–epileptologic correlates of each spike-wave type; it is
clinicians who initiated treatment with anticonvulsants specific not an example of electroencephalographic hair splitting. The
for an absence seizure due to the use of the term 3 Hz spike-wave classical 3 Hz spike-wave discharge is most typical and most
complexes alone. Unfortunately, the term petit mal discharge (37) pronounced in children with absence seizures. Clinical absences
has reinforced this type of reflex thinking. Fortunately, elec- are usually present when the burst duration is greater than
troencephalographers and epileptologists have always cautioned 5 sec. Thus, shorter bursts are usually subclinical, but numerous
against the misinterpretation of the term petit mal discharges; computerized neuropsychological studies have been employed
this should be interpreted as “discharges that occur in absences to demonstrate a clinical component (fluctuations of level of
but also in other conditions.” The work of Silverman (52), Clark awareness) during an apparent subclinical spike-wave bursts.
and Knott (53), and Lundervold et al. (54) has clearly shown that The classical spike-wave complex is, in most cases, easily acti-
the spike-wave discharge as an interictal event correlates with vated by hyperventilation, whereas the slow and the fast forms
the occurrence of petit mal absences only in moderate percent- of this discharge show little or no enhancement.
age of patients (16% according to Clark and Knott). A classical 3 Hz spike-wave burst does not run exactly at a rate
The 3 Hz spike-wave discharge is a worthwhile subject for of 3 Hz. The complexes are faster at the onset of the burst (mostly
more detailed discussion. As to wave morphology, there is good around 4 Hz), then slow down to 3.5 and 3 Hz for the main por-
evidence that the spike-wave complex is not simply an associa- tion, and eventually slow down to 2.5 Hz at the end of the burst.
tion of a spike and a slow wave, although the above-mentioned During a burst, the spike discharges become gradually smaller,
definition is justifiable as a simplification. The spike-wave com- often shrinking to insignificance (Fig. 26.8) as in drowsiness and
plex contains hidden components that can be seen even with sleep.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 485

Figure 26.8 Petit mal absence with generalized synchro-


nous spike-wave complexes. The enormous amplitude of
the discharges necessitates considerable lowering of the
gain. The frontal voltage maximum is evident. Note grad-
ual decline of the spike component at the end of attack,
along with slight slow down of the frequency to about
2.5 Hz.

The spatial distribution of the bursts is very typical; the max- be blocked for a moment by a few (even by a single) spike waves
imum almost always lies over the frontal midline region, and interfere with the working memory. In a clinical 3 Hz
whereas a minimum is found over temporal and occipital areas. spike-wave absence, blocking of the working memory might be
The different conclusion by Dondey (63) was based on EEG the most likely explanation for the mild disturbance of con-
recordings in which no midline electrodes were used. Rodin sciousness and its immediate full recovery with the termination
and Ancheta (64) have confirmed the concept of a frontal mid- of the attack (71). No “true disturbance of consciousness”—
line maximum using brain mapping. Almost all spike-wave epileptic or nonepileptic—can recover immediately. In other
bursts are bilateral synchronous or generalized synchronous; words, the function of working memory is simply suspended
the synchrony, however, is not perfect (56,65). The age of distri- for the duration of the spike waves.
bution lies mainly in the range from 4 to 16 years. Due to the uniqueness of primary generalized epilepsy with
Computerized analysis by Lemieux and Blume (66) has also 3 Hz spike waves in the human (72), animal models for this
shed light on the spatial distribution of the spike-wave com- response have great limitations. The arrest reaction of the monkey
plex: the field distribution of spikes differed from that of the (73) produced by alumina cream injection into the fronto-orbital
ensuing slow waves. Spikes “arose in one frontal region and cortex has some resemblance to the human absence epilepsy; this
propagated to the homologous part of the other frontal region cannot be said about the sudden freeze of motility noted in the cat
with a peak-to-peak interval less than 15 msec. Less commonly, stimulation of the cat’s mesial anterior thalamus (74); there is no
spikes first moved anteriorly within the initiating frontal region spike-wave activity associated with this type of arrest reaction.
before contralateral propagation occurred.” By contrast, nega- According to Sperling and Skolnick (75), there is a general
tive slow waves were more diffuse, more symmetrical in evolu- decrease of cerebral blood flow (133 xenon method) during
tion, and more posteriorly centered than the spikes. human generalized spike-wave activity, which, however, is less
The onset of the spike component in one frontal lobe is con- pronounced in frontal regions than in the parietal lobes.
gruent with the earlier findings of Cohn (56) and Lüders et al. Transcranial magnetic stimulation of the motor cortex in
(67), who noted asynchronies ranging from 5 to 20 msec. The patients with primary generalized epilepsy and 3 Hz spike
frontal lobe is a large structure and subdivisions are necessary. waves produced motor evoked potentials of significantly
Onset of spike-wave activity (and, in particular, of the spike reduced size when the cortical stimulus was time locked to the
component) over the frontal midline means in reality that a slow wave component of the spike-wave complex (but slightly
frontal portion in the immediate vicinity of the interhemi- reduced or unchanged when the stimulus was time locked with
spheric fissure is the origin of the discharge; this area belongs to the spike) (76). This underscores the special inhibitory char-
the supplementary motor zone. acter of the slow wave component, which has been known since
What are the neuropsychological correlates of subclinical the work of Jung and Toennies (58).
generalized spike-wave bursts and clinical absences with spike
waves? In this context, it should be pointed out that the impact Spike-Wave Complex (Slow 1 to 2.5 Hz)
of apparently subclinical 3 Hz spike-wave bursts might be After the demonstration of the 3 Hz spike-wave complex and its
stronger than one would expect from routine clinical observa- relationship to the absence seizure (38), the Gibbses and William
tion. With its maximum over the frontal midline (frontal lobe G. Lennox were struck by the occurrence of slow spike-wave
close to interhemispheric fissure), there might be some impact complexes in a much different type of patients with seizures
on prefrontal lobe functions and, in particular, on the working other than the petit mal absence. The classical 3 Hz spike-wave
memory. This concept of working memory, introduced by complex was termed petit mal discharge. Consequently, the slow
Baddeley (68) and enormously refined by Fuster (69,70), spike-wave complex was termed petit mal variant discharge. This
implies a system of constant perceptory afferences to the pre- term was used first by Gibbs et al. (77). In that study, hypo-
frontal dorsolateral region whence motor impulses emanate, glycemia was induced in order to demonstrate an increase of
resulting in readiness (set) for motor action. This system could classical 3 Hz spike-wave activity in the hypoglycemic state; an
486 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

keep in mind, however, that many children or adolescents with


slow spike waves also show series of 3 Hz or even 4 Hz spike-
wave complexes; the presence of these spike-wave types is obvi-
ously of no significance in such cases and what counts is the slow
type. On the other hand, patients with classical 3 Hz spike-wave
complexes and a usually benign type of epileptic seizure disor-
der almost never show slow spike waves except when a 3 Hz or
4 Hz spike-wave burst may slow down to 2.5 Hz at the end.

Spike-Wave Complex (Fast 4 Hz, 4 to 5 Hz)


This pattern is closely related to the classical 3 Hz spike-wave com-
plex; both discharge types are most commonly lumped together.
A differentiation, however, is justified on clinical grounds.
Gastaut (79) deserves credit for an outline of the distinctive
features between the 3 Hz and the 4 Hz (or 4 to 5 Hz) spike-
wave discharge. According to his investigations, the fast spike-
wave burst is usually of shorter duration (1 to 3 sec), it occurs
in patients older than 15 years, the bursts are always subclinical,
Figure 26.9 Generalized slow spike-wave complexes (mostly around and the associated seizure disorder is usually characterized by
2 Hz) in a child with severe epileptic seizure disorder (Lennox–Gastaut myoclonic jerking, grand mal attacks, or a combination of both
syndrome). seizure types, whereas absence seizures are quite uncommon.
Paroxysmal flicker responses are common in such patients. A
increase of slow spike-wave activity, however, was not elicited positive family history of epileptic seizures disorder is fre-
with this activation method. quently found in this group of patients.
The wave morphology of this pattern varies considerably. In The 4 Hz spike-wave discharge is spatially distributed in the
the majority, the complex consists of a rather slow spike (accord- same manner as the 3 Hz spike-wave discharge; the frontal mid-
ing to the definition, a sharp wave, lasting 70 msec or longer) and line maximum is quite prominent.
a slow wave. In a sizable number of cases, true spikes (60 msec or
less duration) are followed by a slow wave (Fig. 26.9). The 6 Hz Spike-and-Slow -Wave Complex
The spatial distribution is, in most instances, quite similar to (Phantom Spike and Wave, Wave and Spike
that of the 3 Hz spike-wave complex. In the vast majority of the Phantom)
cases, the bursts are bilateral or generalized synchronous with This is now considered a pattern of uncertain clinical signifi-
imperfect synchrony and a frontal midline maximum is the rule cance, or a normal variant (marginal paroxysmal pattern),
in these discharges. Lateralization or occasional focal occur- described by Gibbs and Gibbs (37): “The spike in this pattern
rence is sometimes observed, usually in children with severe has a strong positive component but the entire wave complex
residual brain damage in certain areas where parenchymatous looks in general like a miniature reproduction of the 3 per sec-
destruction abolishes the spike-wave discharge. ond spike and wave of petit mal.” Because of the positive spike
Generalized slow spike-wave discharges are often quite pro- component, Silverman (80) stressed the similarity to the 6 Hz
longed. In some children or adolescents, the entire sleep por- component of the 14 and 6 Hz positive spike burst and reported
tion (light and moderately deep non-REM sleep) consists of a transition from the former to the latter with deepening
unabated generalized slow spike-wave activity. The diagnosis of drowsiness. Marshall (81) introduced the term wave and spike
an “electrical status epilepticus” may or may not be justified in phantom, which has found but limited acceptance.
such cases; there are usually no behavioral or polygraphic char- The clinical correlates of 6 Hz spike-wave complex have been
acteristics to suggest an ongoing ictal event. In some cases, the elucidated by Thomas (82), Gibbs and Gibbs (83), Hughes et al.
slow spike-wave activity is found only in non-REM sleep. (84), and Thomas and Klass (85). It is usually a pattern of adult-
Although the classical 3 Hz spike-wave complex is seldom hood but may also occur in adolescents and children. About
seen before the age of 4 and almost never before 3.5 years, its slow 50% to 60% of the patients suffer from indubitable epileptic
counterpart appears much earlier, sometimes before the age of 6 seizures, mostly generalized tonic–clonic seizures; the remain-
months. At this early age, the frontal maximum may not be read- der show a history of syncopal attacks, posttraumatic states, or
ily demonstrable. (as emphasized by Small in Ref. 86) psychiatric problems.
The slow spike-wave complex is almost always associated The 6 Hz spike-wave complex is an uncommon but not rare
with a severe and often uncontrollable type of childhood pattern (about 0.5% to 1% in a central EEG laboratory).
epilepsy (seldom with onset between ages 11 and 20 years) called The discharge may be recorded in waking state, drowsiness,
Lennox–Gastaut syndrome (40,44,46,77,78). Most of these chil- and light non-REM sleep; light drowsiness appears to be the
dren have a mixed seizure disorder and mental retardation. optimal recording condition (Fig. 26.10).
Thus, the distinction between slow (1 to 2.5 Hz) and classical Investigations regarding the spatial distribution of this
(3 Hz) spike waves is of great clinical significance. One has to pattern have led to an important dichotomy. Hecker et al. (87)
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 487

Kocher et al. (90) were struck by the occurrence of 6 Hz


spike-wave complexes in the abstinence or withdrawal phase of
drug-dependent individuals. Tharp (91) produced the dis-
charge by intravenous diphenhydramine (50 mg) in normal
subjects. Hecker et al. (87) found that the occipital form of the
6 Hz spike-wave complex is often related to drug dependence
(hypoanalgesics, barbiturates) and withdrawal. All this under-
scores the need for a distinction of the two forms. Thus, the
frontal form will have to be regarded as a truly epilepsy-related
paroxysmal pattern, whereas the occipital form finds itself on
the fringe of paroxysmal discharge types.

Patterns of Uncertain Clinical Significance,


or Benign EEG Variants, Benign Epileptiform
Variants, and Marginal Patterns
The just-mentioned 6 Hz spike-wave discharge is considered a
“benign EEG variant and pattern,” which include patterns with
an “epileptiform morphology.” According to Westmoreland
(92), the 6 Hz spike-wave discharge is not a reliable indicator of
seizures (also see Refs. 85,93). Other patterns listed under the
heading of benign patterns are small sharp spikes, 14 and 6 Hz
positive bursts, wicket spikes, and the rhythmic temporal theta
bursts of drowsiness (psychomotor variant pattern).
According to Westmoreland, these patterns “have an epilep-
tiform appearance but are considered normal or benign vari-
Figure 26.10 A short run of 6 Hz spike waves, posterior type,
ants of EEG activity or activity of uncertain significance.” These
recorded in a 52-year-old woman with a history of head injury 2 years
patterns occur in the healthy individual and there is no doubt
earlier and subsequent headache, dizziness, and memory loss. There
that various EEG abnormalities may occur in someone who
was computed tomography (CT) scan evidence of cortical atrophy.
functions normally (48,49,94). Although these may be associ-
ated with potential epileptogenicity, they should be interpreted
pointed out a distinction between a frontal versus an occipital with caution, especially if the only finding on the EEG.
accentuation. The frontal type is most commonly associated Westmoreland (92) suggests that “critical evaluation of the var-
with epileptic seizure disorders and sometimes found in combi- ious types of EEG activity is important.” From a single labora-
nation with other epileptiform discharges. The occipital types tory in London, Ontario, benign epileptiform variants occurred
are found predominantly in patients with no evidence of epilep- in 3.4% of 1183/35,249 recordings (95).
tic seizure disorder.
In a study of 839 patients with 6 Hz spike-wave complexes, Rudimentary Spike-Wave Complex
Hughes (88) placed particular emphasis on the distinction (“Pseudo Petit Mal Discharge”)
between frontal and occipital maximum. He summarized the Gibbs and Gibbs (83) described a paroxysmal discharge that
principal features of both forms by the acronyms WHAM and consists of generalized or nearly generalized high-voltage 3 to
FOLD: 4 Hz waves “with a poorly developed spike in the positive
trough between the slow waves, occurring in drowsiness only.”
W: waking record F: females This pattern is most prominent over parietal areas and was
found only in infancy and early childhood with hypnagogic
H: high in amplitude O: occipital hypersynchrony. This hypogognic hypersynchrony can occur in
generalized bursts rather than in prolonged stretches. These
A: anterior L: low in amplitude bursts, sometimes with a frequency as slow as 3 to 4 Hz, may
contain small spike elements that justify a term like rudimen-
M: males D: drowsy record
tary spike waves. The Gibbsian term pseudo petit mal discharge
suggests a relationship to spike-wave discharges and absence
An interesting sidelight of this observation is the uneven epilepsy. However, this pattern should not be equated to a spe-
gender distribution, which is rather unique; no other EEG cific epileptiform discharge and a transition into a classical or
abnormalities show any gender preference. other spike-wave pattern does not occur (83). A history of
According to Hughes (89), patients with anterior 6 Hz spike febrile illness is very often found in these children with rudi-
waves are more likely to have epileptic seizures, and those with mentary spike waves and a history of febrile convulsions is also
posterior discharges tend to have neuroautonomic disturbances. quite common (83,96).
488 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Small Sharp Spikes It is interesting to note that Gibbs and Gibbs (83) found a
Small sharp spikes, also known as benign epileptiform tran- rather high prevalence of small sharp spikes in presumed nor-
sients of sleep (BETS), were delineated by Gibbs and Gibbs mal adult control subjects, reaching 7.9% in the range from 40
(37,83). It is the most inconspicuous paroxysmal discharge and to 49 years. Reiher and Klass (100), Lebel et al. (101), White et
hence is easily overlooked. The main negative and positive com- al. (102), Klass and Westmoreland (93), and Gutrecht (103)
ponents are of about equally spiky character (Fig. 26.11), it is have stressed the nonspecific nature of this pattern and do not
fairly widespread but seen chiefly over temporal and frontal regard its occurrence as an abnormality. By contrast, Low et al.
areas, either shifting from side to side or synchronously firing. (104) as well as Hughes and Gruener (105) have pointed out
The pattern is almost exclusively found in drowsiness and/or that small sharp spikes are not a normal or meaningless pattern.
light non-REM sleep. It is essentially a pattern of adulthood The data of these authors clearly show that this pattern indi-
with a peak between ages 30 to 60 years; occurrence in adoles- cates “a moderate degree of epileptogenicity” (105). This has
cence and old age is somewhat less common. It is virtually been strongly confirmed by Saito et al. (106) and also by Molaie
absent in the first 10 years of life. et al. (107). This is another indication of the inherent epilepto-
Koshino and Niedermeyer (97) found a prevalence of 1.36% genic potentialities in so-called benign patterns.
with a peak of 2.9% between ages 30 and 40. In this study, two
thirds (67.4%) of the patients had a history of epileptic seizures; Needlelike Occipital Spikes of the Blind
this underscores the paroxysmal character of the discharge. Spike discharges of a particularly fast and needlelike character
From Ontario, small sharp spikes occurred in 1.85% of EEG develop over the occipital region in most congenitally blind
recordings (95). children (83,108,109). These spikes are completely innocuous,
Small sharp spikes sometimes are precursors of typical ante- unrelated to epileptic seizure disorder, and probably due to a
rior temporal spikes or sharp waves, which, in such patients, state of functional deafferentation (110). The discharges disap-
simply occur somewhat later in drowsiness or sleep. This asso- pear during childhood or adolescence.
ciation was also noted by Gibbs and Gibbs (83). In a depth EEG
study by Westmoreland et al. (98), an extensive generator area The 14 and 6 Hz Positive Bursts
of small sharp spikes was found. Among people without (Fourteen and Six Positive Bursts)
epilepsy, the discharges may occur in patients with cerebrovas- In the IFSECN (8), “Fourteen and Six Hz Positive Bursts,” are
cular disorder, syncopal attacks, and psychiatric problems. defined as: “Bursts of arch shaped waves at 13 to 17 Hz and/or 5
Small (86) and Small et al. (99) have placed special emphasis on to 7 Hz, most commonly at 14 and/or 6 Hz, seen generally over
the occurrence of this spike discharge in a psychiatric popula- the posterior temporal and adjacent areas of one or both sides of
tion and especially in patients with manic-depressive illness. the head during sleep. The sharp peaks of its component are

Figure 26.11 Small sharp spikes (51-year-old patient).


Note the subtle character and moderate voltage of the
discharge; also note its predominance in the left
nasopharyngeal lead. There is evidence of spread into
T3, as well as into the right nasopharyngeal lead. The
left section was recorded in the waking state (transition
to earliest drowsiness); the middle and right sections
were recorded in sleep. (Reproduced with permission
from Koshino Y, Niedermeyer E. The clinical signifi-
cance of small sharp spikes in the electroencephalo-
gram. Clin Electroencephalogr. 1975;6:131–140.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 489

Little is known about origin and neurophysiologic discharge


demonstrated in depth recordings at the thalamic as well as the
basal ganglia level (132); the discharge was also repeatedly
recorded in the amygdaloid complex.
Much of the controversy about this pattern has arisen from
differences of recording techniques and the use of routine sleep
tracings and, in particular, the length of the sleep portion. The
prevalence used to range from 10% to 30% at the age of 10 to 25
years, but there has been an unexplained marked decrease of the
prevalence of “14 and 6” Hz discharge according to Gibbs (133).
We interpret the 14 and 6 Hz positive burst itself as “within
normal limits,” unless it is extremely frequent and pronounced;
then it is regarded as minimally abnormal. It appears to be a
marginal pattern of no or very little epileptologic significance
Figure 26.12 Various examples of 14 Hz and 6 Hz positive spikes and proven cases of epileptic seizures very seldom have a 14 and
(underlined). Note the posterior predominance for this pattern and 6 Hz positive burst as the only significant finding.
shifting asymmetrics. Also note the sometimes-blurred distinction However, the 14 and 6 Hz positive burst may occur in the
between the 14 and 6 components, due to notch formation. The record- advanced states of metabolic encephalopathies, especially in
ing was obtained from a 12-year-old patient; montages to ipsilateral ear. hepatic coma (134,135). Drury (136) reported periodic 14 and
6 Hz positive bursts in 7/121 children with hepatic coma, mostly
in association with Reye’s syndrome. All cases had a severe back-
positive with respect to other regions.” For these bursts, the ampli- ground EEG abnormality; these bursts were activated by stimu-
tudes are generally below 75 V, the pattern is best demonstrated lation in four children, and disappeared on follow-up recordings
by referential recording using contralateral ear lobes or other in six. The “14 and 6” pattern may also occur as a transient toxic
remote reference leads, and the clinical significance is controver- effect of diphenhydramine hydrochloride (Benadryl).
sial. The synonymous term ctenoids was introduced by Lombroso
et al. (111) with regard to the comblike shape. Contrary to Rhythmic Temporal Theta Burst of Drowsiness,
Dutertre’s (10) statement, “mitten” is not a synonymous term. Rhythmic Midtemporal Discharges, or
The 14 and the 6 component may be observed independ- Psychomotor Variant
ently. According to Gibbs and Gibbs (83), the 6 Hz component This pattern was widely known as “psychomotor variant dis-
prevails in early childhood and adulthood, while the 14 Hz charge” (37,83) because of certain similarities to rhythmical
component is more prominent in older children and adoles- ictal activity occurring in psychomotor seizures (temporal lobe
cents. Hughes (112,113) emphasized the harmonic character seizures, complex partial seizures). This term has been discour-
of the major frequencies and preferred the term 14 and 7 Hz aged by IFSECN (8), which recommended the term rhythmical
positive spikes (Fig. 26.12). From Ontario, 14 and 6 positive theta bursts of drowsiness. The term rhythmic midtemporal dis-
bursts occurred in 0.52% of EEG recordings (95). charge of Lipman and Hughes (137) is also widely used.
This pattern was described first by Gibbs and Gibbs (114) and The pattern consists of long runs of rhythmical activity in the
regarded as “evidence of thalamic and hypothalamic epilepsy.” range of 5 to 6.5 Hz with a maximum over the midtemporal
The following 15 years produced exciting correlations to auto- region, often with considerable spread into posterior temporal,
nomic nervous system dysfunctions and behavioral disorders anterior temporal, and occipital areas. The theta activity shows a
(115–126). Enthusiasm flagged when it became clear that this well-defined negative sharp component (Fig. 26.13) that infers
pattern is too often found in healthy individuals (111,127). Little the epileptiform character of the discharge. These trains of sharp
(128) even presumed that this pattern “is more likely to be a sign theta waves may occur in a unilateral, bilateral shifting, or syn-
of health than of disease.” chronous distribution type. The duration of a single run usually
The “14 and 6 pattern” may be confined to the regions lying exceeds 10 seconds and may reach 1 minute or more. Very often,
beneath a skull defect (129) and hence could denote a “hidden the first run is noted in early drowsiness; in deep drowsiness and
pattern” that is not detectable from the scalp unless it reaches light sleep, the pattern tends to disappear. According to Egli et al.
major proportions. (138), patients with rhythmical theta bursts show shortened
The pattern occurs most commonly in children after age 4 periods of REM sleep and also, to a lesser degree, of slow sleep
and adolescents and declines in adulthood; Gibbs and Gibbs in nocturnal sleep studies at the expense of log drowsy periods
(83,130) recorded the 14 and 6 Hz activity even in infants. Its with the rhythmic theta pattern. Hughes (139) has emphasized
occurrence in the waking state is exceptional; it typically occurs that the rhythmic midtemporal discharge is not limited by the
in deep drowsiness and very light non-REM sleep, although drowsy state and may be very active in wakefulness.
deep sleep might be more conducive in very young children This pattern is rare. Egli et al. (138) found a prevalence of
(83). Okada et al. (131) noted 14 and 6 Hz positive bursts in 0.1% in a large laboratory, and Santoshkumar et al. (95) in 0.12%
association with simultaneous negative spikes over the frontal of EEG recordings. Although occurring mainly in younger or
area (suggestive of dipole formation). middle-aged adults, it is also seen in adolescents and children.
490 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

maximum of this discharge is usually found over the centropari-


etal region and especially over the vertex. Hyperventilation may
induce this pattern.

Periodic or Quasiperiodic Discharges


These rhythmically (periodic), or nearly rhythmically (“quasi-
periodic”) recurring discharges are of an epileptiform charac-
ter, but are not usually associated with chronic epilepsy. Periodic
discharges occur with a specific interval duration and quaiperi-
odic discharges occur with a varying interval duration. These are
an important EEG feature of a severe ongoing central nervous
system (CNS) disorder. They are hence disease-suggestive and
sometimes virtually disease-specific, rather than suggestive of
epilepsy in general. A review of periodic activities was presented
by Gaches (148) as well as by Bauer and Pieber (149). The peri-
Figure 26.13 Psychomotor variant discharge of the right midtemporal
odic character of these patterns remains enigmatic. This rhyth-
region (T4). Tracing was obtained from a 21-year-old woman. The
mical firing is markedly different from the predominantly
record had low-voltage character; this necessitated the use of high gain.
random character of interictal seizure discharges (spikes, sharp
wave, etc.). There is no satisfactory model to explain the perio-
The clinical significance is not clear. Despite its strongly epilepti- dicity of the discharges.
form appearance, its epileptogenic properties seem to be very Periodic discharges are always of large amplitude, mostly in the
low; most patients have no history of clinical seizures, although range of 100 to 300 V. Morphologically, these may be simple
Lipman and Hughes (137) found seizures in 36% of their sharp waves, usually with a duration exceeding 150 msec, but may
patients. Personality disorders and some autonomic nervous be a compounded waveform. Periodic discharges may be focal,
system dysfunctions are common in patients with this pattern widely scattered, or generalized synchronous. A fine tabulated
(83,137,140). This view is shared by Eeg-Olofsson and Petersén review of periodic discharges has been presented by Spehlmann
(1982; cited in Boutros et al. (141)), who also studied the (150). Periodic discharges are differentiated by the interval: either
genetic transmission of this pattern and wondered about possi- long-interval or short-interval periodic discharges.
ble autosomal-dominant inheritance.
Periodic Complexes in Subacute Sclerosing
Subclinical Rhythmic EEG Discharge Panencephalitis (SSPE)
of Adults (SREDA) SSPE is now exceptionally rare in the developed world following
This pattern was originally described by Naquet et al. (142) as a measles immunization. Periodic complexes dominate the second
“paroxysmal discharge of the temporo–parietooccipital junction,” state of SSPE, a prolonged phase in which the clinical diagnosis is
facilitated or triggered by temporary hypoxic conditions. In spite usually made. This type of complex discharge almost never
of certain epileptiform features and its rhythmical sharp charac- occurs in other clinical conditions and is hence almost disease-
ter, the possibility of an epileptic (ictal) event has been strongly specific. It is a very reliable and highly contributory diagnostic
de-emphasized. Herranz and Lopez (143) have described this pat- finding in this disease. The discharge duration varies from 0.5 to
tern as “subclinical paroxysmal activity” in 31 patients and 3 seconds and are typically formed by two or more waves with
stressed its rarity; it has an incidence of 0.02% to 0.045%. From mean amplitude of 500 V (100 to 1000 V) (150, reaching as
Ontario, SREDA occurred in 0.07% of EEG recordings (95). The high as 1400 V (11)). The enormous discharge amplitude might
authors have seen this pattern once in a child. be confused with movement artifact, which is made more con-
Westmoreland and Klass (144,145) and Miller et al. (146) fusing by the associated myoclonus or sudden loss of tone.
deserve particular credit for the elucidation of this discharge The periodicity develops with disease progression. The earlier
type. These authors disagree with the view of Naquet et al. stages are usually dominated by diffuse 1 to 3 Hz activity of fairly
(142) concerning the major role of cerebrovascular insuffi- rhythmical character, but the compounded discharge may be
ciency and hypoxia, although the average age of their patients aperiodic. Sometimes, the periodic discharge is present in the
was 61 years. The term “SREDA” has been proposed by Miller et earliest stage of the disease and may persist to the fatal outcome;
al. (146). O’Brien et al. (147) analyzed this pattern in seven more often, this pattern disappears in the terminal or third stage.
recent patients and only one of them showed evidence of cere- The discharge is more impressive in the waking state (Fig.
brovascular disorder. 26.14). The elements of the discharge are variable; a giant slow
This pattern occurs mainly in the waking state or in light wave is usually mixed with several sharp waves. The discharge is
drowsiness. The onset may be fairly abrupt with widespread generalized, with a maximum over the frontocentral areas and
sharp rhythmical theta (4 to 7 Hz) and occasionally with delta vertex. The frequency of the complexes ranges from 4 to 16 Hz
activity. In other cases, the onset is gradual with a few single sharp (151). This is an example of a long-interval periodic discharge.
discharges that increasingly develop rhythmical character, first at Accompanying myoclonus is fairly synchronous with the
delta, then at theta frequency. As to the spatial distribution, a discharge; the motor activity may precede or trail the discharge
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 491

Figure 26.14 Periodic discharges in subacute sclerosing panencephalitis (SSPE). A: At an earlier state with some preser-
vation of other activities. B: At a more advanced state (recorded from two different patients, ages 8 and 9 years).

in a range of 200 to 800 msec. There may be minor interhemi- The origin of the periodic SSPE discharge is controversial.
spheric asynchronies (15 msec) and an earlier appearance in Radermecker and Poser (157) have proposed a deep thalamic or
parietooccipital areas than in frontocentral areas, where the dis- mesencephalic-reticular origin; this view has been supported by
charge shows its most impressive voltage (152). Cobb (158,159), who performed depth EEG recordings, and by
The background activity between the complexes is quite Lombroso (160), who studied direct current (DC) potential
variable. The background may be normal with posterior alpha changes during the periodic discharge. Other authors favor the
rhythm; however, this is exceptional; severely disordered back- idea of a cortical origin (161–163).
ground activity is most commonly found. Slow activity prevails In an autopsy-confirmed case of SSPE, Martinovic (164)
and numerous spikes are sometimes recorded, especially over demonstrated periodically occurring generalized high-voltage
anterior areas. Spike-wave complexes may also be present; even bursts firing at a rate of 11 to 13 Hz with a spiky contour. There
clinical absence seizures with classical 3 Hz spike-wave com- was a repetition interval of 27 to 28 seconds. This most unusual
plexes have been observed (153). A case of acute fulminant pattern was gradually replaced by the conventional type of peri-
SSPE with only 2 months’ duration was reported by Gökcil et odic discharges.
al. (154); the EEG showed very pronounced discharges.
Periodic discharges mimicking SSPE have been reported by Periodic Complexes in Herpes Simplex
Giovanardi Rossi et al. (155) in children with myoclonic juve- Encephalitis
nile epilepsy of nonprogressive character. These discharges, In this extremely severe but not invariably fatal necrotizing
however, were confined to ictal episodes. Most unusual is the encephalitis, slow repetitive spikes have been noted by
observation of Prier et al. (156): convincing periodic complexes Radermecker (165). These observations were subsequently con-
of the aforementioned type in an indubitable case of multiple firmed by Millar and Coey (166), Perier et al. (167), Carmon et
sclerosis (age 15 years). al. (168), Rawls et al. (169), Adams and Jennett (170), Upton and
492 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Gumpert (171), Gaches and Arfel (172), Elian (173), Kugler et al.
(174), and Cobb (175). The discharge was also found in experi-
mental disease inoculated rabbits by Gupta and Seth (176).
In an earlier stage, local mostly unilateral temporal polymor-
phic delta waves are the most striking feature, but soon large sharp
waves emerge over the most affected region. These discharges usu-
ally fire at intervals from 2 to 4 seconds. This is a short-interval
periodic discharge. The discharge may be quite slow and exceed
1000 msec; consider that a sharp wave is defined as having a dura-
tion from 70 to 150 msec. The amplitudes are in the range from
100 to about 500 V. Similar periodic sharp discharges have been
demonstrated in neonatal herpes simplex encephalitis (177). In
the course of the disease, the originally regional discharge tends to
become generalized synchronous; asynchronous bilateral dis-
charges may also occur. Predominantly occipital localization of
the periodic discharges has been reported by Bergey et al. (178).
In contrast with the long-interval periodic complexes found
in SSPE, the periodic discharge of herpes simplex encephalitis
has a short interval, with the longest intervals of 4 seconds or
less; according to Gaches (148), this criterion is always met in
the herpes simplex discharge. In some cases, the periodic dis-
charge can be detected only when almost daily repeat records
are carried out. This explains the fact that periodic discharges
are not reported in all cases of herpes simplex encephalitis.

Periodic Discharges in Creutzfeldt–Jakob Disease Figure 26.15 Periodic discharges in Creutzfeldt–Jakob disease. Note
The clinical and neuropathologic features of this subacute dis- the mild lateralization of the recurrent sharp waves to the left. The 68-
ease were established by the work of Creutzfeldt (179), Jakob year-old patient was demented, with psychotic features; there were long
(180), and Kirschbaum (181,182). A very similar syndrome tract signs, fasciculation, and frequent myoclonus. Myoclonic jerking
known as spongiform encephalopathy (183) and a special vari- did not alter the tracing.
ant described by Heidenhain (184) were eventually found to be
the same disease. Although originally presumed to be a degen-
erative CNS disorder, a slow virus was eventually demonstrated Myoclonus is a typical clinical feature of this disease. The peri-
as the causative agent (185,108). These investigators transmit- odic patterns may or may not be associated with myoclonus (192).
ted this virus to the chimpanzee with intracerebral inoculation In addition to periodic discharges, cyclic changes may also
and produced a similar invariably fatal disease. It is interesting occur, and the periodic activity may get temporarily replaced by
to note that chimpanzees with the full clinical picture of diffuse slow activity in the delta and theta range (193).
Creutzfeldt–Jakob disease showed prominent delta activity in The periodic EEG discharge of Creutzfeldt–Jakob disease may
their EEG but neither spikes nor periodic discharges (186). be caused by other conditions in rare instances. Hypothyroidism
The periodic discharge consists of a sharp wave or a sharp is mentioned as a treatable cause of this pattern.
triphasic complex of 100 to 300 msec duration with a repetition It has to be emphasized that the periodic discharges of
rate of 0.5 to 2 Hz or intervals of 500 to 2000 msec. This is also a Creutzfeldt–Jakob disease (i) do not occur in every case of
short-interval periodic discharge. The discharges occur against a this disease (even with numerous repeat-recordings) (194) and
severely disordered background of activity, mostly in generalized (ii) may occasionally be found in other disorders.
synchrony. Some cases show unilateral onset with discharges over
one hemisphere or lateralized to one hemisphere for several days Periodic Lateralized Epileptiform
or weeks (187). According to Tariska et al. (188), the periodic dis- Discharges (PLEDs)
charges may even remain unilateral in the course of the disease. Chatrian et al. (36) gave an extensive account of periodic later-
The periodic activity usually shows a maximum over the ante- alized or focal discharges that may occur in a variety of acute
rior region except for the Heidenhain form, which has a posterior neurologic conditions. This pattern is most often associated
maximum (Fig. 26.15); in this special form, cortical blindness is a with acute cerebral infarctions but may also occur in neoplastic,
common feature. In sleep, the periodic discharges tend to disap- inflammatory, and epileptic disorders, especially status epilepti-
pear. The absence of periodic discharges after 10 weeks of illness cus. Periodic discharges occurring in herpes simplex encephali-
militates against Creutzfeldt–Jakob disease (189). EEG studies of tis are discussed earlier in this chapter. PLED activity, however,
Chiofalo et al. (190) are based on the observation of 27 cases. The may also occur in other types of encephalitis—among others,
differential diagnosis of the combined clinical and electroen- in infectious mononucleosis encephalitis, according to Aminoff
cephalographic picture of this disease is quite complex (191). et al. (195). As to acute vascular pathology, watershed-type
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 493

infarctions are the most common structural substratum in ing right frontal MRI lesion and PLEDs in the EEG. Another
patients with PLED. Markand and Daly (196), Dauben and report on PLEDs in MS (right temporal PLEDs and MRI
Adams (197), and Bauer et al. (198) presented further impor- lesion) has been presented by Gandelman-Marton et al. (210).
tant work on this pattern, which also plays a major role in We have seen PLEDs in a child with hemiplegic migraine fol-
experimental cerebral embolism (199). lowing a seizure.
PLEDs may be simple and large sharp waves or complex A very thoughtful study of PLEDs deserves special attention:
(compounded) discharges with mixed spiky and slower ele- a critical review by Pohlmann-Eden et al. (211). These authors
ments. The amplitudes are usually around 100 to 300 V, but consider PLEDs as “an EEG signature of a dynamic pathophys-
may occasionally be much higher. The firing rate may be as fast iologic state in which unstable neurobiologic processes create
as 3 Hz or as slow as 12/min (36) (Fig. 26.16). an ictal–interictal continuum, with the nature of the underlying
The discharge is found over the maximally involved area, and neuronal injury, the patient’s preexisting propensity to have
the local background activity is almost always severely disor- seizures, and the coexistence of any acute metabolic derange-
dered. The focal maximum is customarily located over the ments all contributing to whether seizures occur or not.”
boundary zone between middle and posterior cerebral arteries Pohlmann-Eden et al. have strongly reemphasized the domi-
and thus over the posterior temporal region and its immediate nant etiologic role of strokes in the generation of PLEDs. From
vicinity. Watershed infarctions are seldom found between the the evidence of functional neuroimaging studies, these authors
territories of middle and anterior cerebral arteries, with a maxi- feel that PLEDs “might reflect a key pattern for focal hyperex-
mum of PLED activity over the superofrontal region. citability in the penumbra zone of ischemic stroke.”
The term PLED implies lateralization. It is possible, however, A nonlateralized variant of PLEDs are periodic epileptiform
that PLED activity is generated independently over both hemi- discharges occurring in the midline (PEDIM), as cited by
spheres. De la Paz and Brenner (200) have investigated the bilat- Westmoreland et al. (212). These midline discharges are also
eral type of PLED referred to as “BI-PLED” (also “bi-PLED”). presumed to arise from watershed areas: anterior and middle,
Although strokes were found to be the leading cause in the middle, and posterior cerebral arteries.
PLED group, CNS infection and epileptic seizure disorders,
especially status epilepticus, predominated in the BIPLEDs Periodic Discharges in Acute Cerebral Anoxia
group. Bilateral synchronous (generalized synchronous) peri- Repetitive simple or compounded sharp waves in generalized
odic activity should not be listed as BIPLED in view of the orig- synchrony may occur against a flat (or at least seemingly flat)
inal definition of Chatrian et al. (36). This distinction, however, background of activity in patients with acute cerebral anoxia.
is not generally observed (201). Filley et al. (202) have broken Myoclonus is often found in association with this type of activ-
down their large material of PLEDs into typical and “indeter- ity (213–215). In most cases, these discharges are probably
minate” discharges. The term cerebral bigeminy has been used aborted bursts in a suppression-burst pattern. True periodic
by Aldrich and Pugh (203) for bilaterally independent PLED discharges, however, may also occur (Fig. 26.17).
activity. Even TRI-PLEDs, arising from three different areas,
have been reported (left frontal, left posterotemporal, and right Periodic Discharges of Other Etiologies
posterotemporal) in an 85-year-old patient with renal Periodic discharges may occasionally occur in other conditions,
encephalopathy (204). but such observations are rare. The presence of such discharges,
PLEDs are often associated with simultaneous focal motor whether regional or generalized, merely underscores the severity
twitching in the contralateral face or fingers, hand, arm, leg, of the basic conditions. Figure 26.18 demonstrates periodic dis-
foot, etc. This underscores the paroxysmal character of the pat- charges over the right posterior quadrant in a 15-year-old girl
tern. It is usually a temporary pattern that changes into other with a history of Dandy–Walker syndrome, numerous shunt
abnormalities within 1 to 2 days. Patients with PLEDs are in operations, shunt infections, and a severe epileptic seizure disor-
most cases acutely ill and often show a history of a variety of der. These rhythmical discharges also show the tempo of propa-
mixed problems, such as cerebral arteriosclerosis plus renal gation over the right posterior quadrant of the cerebrum.
insufficiency or chronic alcoholism or diabetes mellitus. The
possibility of an underlying neoplasm must also be considered. SPIKES AND OTHER PAROXYSMAL
Patients with PLEDs are commonly older adults; this pat- DISCHARGES IN HEALTHY CHILDREN
tern occasionally occurs in young adults and children (205).
Andriola (206) noted PLEDs as well as BIPLEDs in 12 children Focal or generalized paroxysmal discharges may occur in appar-
with various types of acute CNS disease; those with bilateral ently healthy individuals in all age ranges. These findings may be
activity (BIPLED) expired in the acute state. Ritaccio and termed as “false positives,” but the EEG abnormalities are real
March (207) reported the association of BIPLEDs with com- and such spikes are evidence of an underlying epileptic patho-
plex partial status epilepticus. Silbert et al. (208) described physiology that may or may not subsequently become manifest.
another variant of PLEDs with independent ipsilateral peri- Let us contemplate the indubitable fact that a complete medical
odic lateralized discharges. A clinical curiosity appeared to be evaluation will yield certain abnormalities in almost every
the observation of Chabolla et al. (209): a young man suffering healthy individual. What the electroencephalographer needs in
from multiple sclerosis (MS) with exacerbations ushered in by such cases is a commonsense philosophy as a basis for a wise
complex partial status epilepticus associated with an enhanc- interpretation. There are many examples of seemingly irrelevant
494 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 26.16 A: Periodic lateralized epilep-


tiform discharge (PLED) in a 70-year-old
patient, widespread over left hemisphere
with left midtemporal maximum. Note the
poorly developed background of activity.
Cerebrovascular accident associated with
twitching of right arm. Patient lethargic and
obtunded. B: PLED over right central–
midtemporal region, sometimes also involv-
ing the right parietal area. Note the well-
preserved physiologic activities of waking
and light drowsiness. Also note the complex
and multiphasic character of the discharge.
Patient is 68 years old with cerebrovascular
disease with right centroparietal low-density
area in CT scan (infarction).
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 495

Figure 26.17 Comatose patient (acute cerebral anoxia), age 57.


Constantly repetitive compounded spike discharges against a virtually
flat background of activity. These discharges were associated with occa-
sional generalized myoclonus.
Figure 26.18 Waking record of a 15-year-old girl with Dandy–Walker
syndrome in combination with a brainstem lipoma. The patient suffers
and yet unmistakably present abnormalities that the prudent, from a severe epileptic seizure disorder; she had been treated with
seasoned physician will integrate into a holistic view of the indi- numerous shunt operations. Against a diffuse background of disorgan-
vidual. Similarly, EEG does not differ from the rest of medicine. ized slow activity, a periodic discharge of “blunted” sharp character is
The EEG evaluation of comparatively large healthy popula- very prominent over the right posterior quadrant. Both bipolar and
tions usually shows a certain percentage of abnormalities. unipolar derivations show a focal origin in the right posterotemporal lead
Thorner (216) showed paroxysmal discharges in 0.3% of 1100 (T6) with prominent spread into right occipital (O2) and right parietal (P4)
flying cadets; Gibbs et al. (217) found epileptic abnormalities in regions. This spread is so slow that the traveling character of the periodic
0.9% of normal adult controls; Harty et al. (218) observed such discharges can be gleaned from bipolar and referential derivations.
abnormalities in 3% of candidates for medical service; and
Williams (219) noted “larval epileptic disturbances” in 9% of
healthy controls. A study of 682 Air Force applicants showed (3.52%). Generalized, mainly polyspike, wave discharges were
paroxysmal changes in 2.6%; about one fourth were focal and found in 41 cases, midtemporal spikes in 50, centroparietal
the rest were of nonfocal character (220). spikes in 27, occipital spikes in 2, and bilateral (midtemporal or
Zivin and Ajmone Marsan (221) and Chatrian (222) have parietal) spikes in 11 children. Changes in spike location and
contemplated the clinical significance of spikes in healthy per- shifts from focal to generalized discharges were rare. The follow-
sons. Above all, the interpretation must take into consideration up period was up to 9 years. The abnormalities disappeared in
age. In childhood, the occurrence of central–midtemporal (also most, usually during elementary school age or in early adoles-
parietal) spikes is associated with overt seizures in only 50% to cence. Only seven children developed clinical epileptic seizures;
70% of the cases; this pertains mainly to the age from 3 to 12 five had generalized discharges, one had midtemporal, and one
years. In occipital spikes (mainly age 3 to 5 years), the epilepto- had centroparietal spikes. In two subjects there was evidence of
genicity is even lower. Major studies on this subject based on epileptic seizures in the family, while six children had siblings
large healthy populations were performed by Nekhorocheff with spikes, either generalized or rolandic.
(223), Kellaway and Fox (224), Brandt and Brandt (225), Corbin Both generalized synchronous (spike wave, polyspike wave)
and Bickford (226), Trojaborg (227), and Eeg-Olofsson et al. and rolandic (centroparieto–midtemporal) spikes in nonepilep-
(228). In general, “benign” focal spikes (such as seen in benign tic children suggest a genetic predisposition if no neurologic
rolandic epilepsy) outnumber generalized synchronous bursts deficit and no history of insult to the CNS are present. In chil-
of spikes or spike waves by a slight to considerable margin. dren with a history of cerebral palsy and with no seizures but
The study in children by Cavazzuti et al. (229) demands spe- prominent spiking, the spike activity may herald future
cial attention. EEG recordings were performed on 3726 children epileptic seizures (83). Even in perfectly healthy children with
from 6 to 13 years of age without evidence of seizure or neuro- spikes, the possibility of future seizures cannot be completely
logic deficits. Paroxysmal discharges were found in 131 cases ruled out. In addition, epileptiform abnormalities may be an
496 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

epiphenomenon associated with an underlying neurologic tially reiterated by Dreifuss (233)), this classification did not
disorder, such as attention deficit disorder, headaches, or find universal acceptance. This may have been due to an excess
behavioral problems, and psychiatric disorders, to name a few. of terms and the use of cumbersome formulations. Also, in
This section must be capped by a strong plea to clinicians to pediatric practice the classification required several modifica-
refrain from rash and ill-advised statements that a person tions for practical application. It was not always possible to
without seizures has epilepsy and must be treated because of differentiate simple from complex focal seizures because
spikes in the EEG. There may be need for further medical immaturity, limited communication skills, and autistic fea-
attention, and in some circumstances a repeat EEG may be tures could prevent the accurate assessment of alteration of
prudent but anticonvulsive treatment is seldom needed and consciousness in some children (234). Infantile spasms were
indeed is ill-advised in most cases. missing from the classification, as were myoclonic–astatic or
myoclonic–atonic seizures. A breakdown into small ictal
Types of Epileptic Seizures behavioral or ictal-EEG detail may render a classification more
The character of an epileptic seizure is more or less strongly nebulous. There are, of course, limitations for every attempt at
determined by the chief cerebral area involved. Another impor- seizure-type classifications. The classification of 1981 could
tant factor is the underlying basic epileptic condition. As a not accommodate a considerable number of seizures, which
matter of fact, the underlying basic condition may be of para- then were listed as unclassifiable because they did not fit the
mount significance, especially in an infant with infantile scheme. A recent recommendation from the ILAE taskforce on
spasms—hypsarrhythmia and jackknife seizures, which are not classification suggested a marked simplification of the seizure
found outside this basic epileptic condition, or in a child with classification and tried to address some of these issues (235)
Lennox–Gastaut syndrome and atonic drop attacks, which are (Table 26.1).
germane to this condition and alien to others (also see Ref. 230).
These basic epileptic conditions are age-determined or age- Generalized Tonic–Clonic Seizure (Grand Mal)
dependant; therefore, age is an important factor that is capable of Clinical Manifestations
modifying the character of epileptic seizures. Furthermore, the The sequence of clinical manifestations has been masterfully
nature and extent of an underlying cerebral lesion may be a neg- described by Gowers (236, see also republication in 1964) and is
ligible factor in the determination of the seizure type in the group given in detail below. Further detailed descriptions were given by
of age-determined epileptic conditions. Janz (237), Gastaut and Broughton (238), and Karbowski (239).
Certain types of seizures, such as focal motor and psy- The generalized tonic–clonic (GTC) seizure lasts about 40 to
chomotor seizures, are highly indicative of special cerebral 70 seconds, or sometimes up to 90 seconds. Initial massive
areas, whereas others are of much less localizing value. The
localization depends heavily on such confirming EEG evidence
as local spikes and recorded ictal episodes and, in some special
cases, on the demonstration of regional changes with depth Tabl e 2 6 . 1
electrodes. Structural neurodiagnostic tests are needed for the Classification of Seizure Types
demonstration of significant morphologic changes.
Focal seizures may be a prelude to an ensuing grand mal Generalized seizures
convulsion. The term aura has been used for such initiating
focal seizures, especially for attacks impinging on afferent sys- Tonic–clonic (in any combination)
tems (visual, acoustic, olfactory, somatosensory, gustatory, and Absence
so forth). Typical
Simultaneous EEG recording and fMRI is beset with prob- Atypical
lems since the EEG is being obtained in a high-field magnet and Absence with special features
during scanning. Baudewig et al. (231) have reported a method Myoclonic absence
demonstrating simultaneous blood-oxygenation-dependent Eyelid myoclonia
(“BOLD”) MRI sequences and EEG activity with very little arti- Myoclonic
factual distortion (in epileptic patients). Myoclonic
Classification of Epileptic Seizures Myoclonic–atonic
Myoclonic–tonic
The classification of epileptic seizures is a thankless job. The
preferences of “splitters” and “lumpers” must be reconciled in Clonic
the tedious work of an international committee entrusted with Tonic
this task by the International League Against Epilepsy, pro- Atonic
gressing from the older classifications of reputed epileptolo-
gists that have sometimes been marred by personal biases. The Focal seizures
most commonly used classification had been proposed by Unknown
the Commission on Classification and Terminology (232). Epileptic spasms
Although widely used and officially recommended (and essen-
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 497

generalized tonic spasm is sometimes initiated with a cry (“a Karbowski (239). These attacks are characterized by shorter
wild, harsh, screaming sound,” 236). It is immediately associated duration and rudimentary tonic or clonic phases, possibly due
with loss of consciousness (profound coma); the arms are usually to the effect of anticonvulsive medication.
in semiflexion and the legs in extension. After about 10 to 20 sec-
onds, it is supplanted by the clonic phase in which “the vibratory EEG Manifestations
phenomenon (noted with palpation of the extremely tense mus- The tonic–clonic seizure is initiated by an abrupt loss of voltage
cles) becomes sufficiently prolonged to interrupt completely the (desynchronization, electrodecremental period) of a few seconds
tonic contraction” (238). duration; there is evidence of very fast (20 to 40 Hz) activity in all
This leads to a succession of brief and violent flexor spasms of leads. In patients with primary generalized epilepsy, several gen-
the entire body. Accompanying apnea leads to a grayish livid eralized bursts of polyspike-wave complexes with massive bilat-
complexion, while the rhythmical clonic spasms slow down until eral myoclonus may precede the phase of desynchronization.
a final massive myoclonus marks the end of the seizure. Muscle activity rapidly obscures the recording; the vertex
According to Gastaut and Broughton (238), tongue biting usually derivation, however, may remain artifact-free due to the lack of
materializes during the clonic phase. Mydriasis, arterial hyper- underlying muscles. Informative recordings can be secured only
tension, and tachycardia accompany the attack. Enuresis usually from patients with muscle relaxation from curarization and
occurs at the termination of the seizure (238); there may occa- artificial respiration. Removal of muscle artifact by means of
sionally be fecal incontinence. The patient is completely flaccid digital filtering has been achieved by Gotman et al. (241).
after the last clonic jerk, after which respiration returns. Only a After the phase of desynchronization, which may be as short
few seconds after the beginning of the flaccid, immediate postic- as 1 to 3 seconds, rhythmical activity at about 10 Hz with rap-
tal phase, a tonic muscle spasm returns that is most intense and idly increasing amplitude dominates the EEG. Gastaut and
prolonged in the masseter (238). This postictal trismus tem- Broughton (238) have laid much stress on this frequency
porarily blocks respiratory effect. The respiration becomes regu- (“epileptic recruiting rhythm,” 242), which is better discernible
lar in the ensuing recuperative phase, during which the patient with the use of automatic frequency analysis.
returns to consciousness unless he slips directly into a period of About 10 seconds after the onset of a seizure, slower fre-
postictal sleep. A fall caused by a GTC can be traumatizing; even quencies are noted, gradually slowing into the theta and delta
epidural hematoma has been reported (240). range. Once the frequency of 4 Hz is reached, “each slow wave
Tonic–clonic seizures may occasionally show unilateral interrupts the recruiting rhythm, giving rise to polyspikes and
predominance of the clinical manifestations. Gastaut and wave complexes, themselves decreasing in frequency” (238).
Broughton (238) feel strongly that these attacks (which are more The clonic activity corresponds with generalized polyspike
common in children and infants) are in fact generalized seizures bursts at each myoclonic jerk (Fig. 26.19).
with unilateral expression. Do attenuated or mitigated grand The last clonic contraction is followed by postictal flatness
mal seizures exist? The answer is affirmative according to for several seconds (243). Very slow irregular delta activity

Figure 26.19 The EEG correlate of a grand mal


(generalized tonic–clonic) seizure, recorded in a
curarized patient and almost free of superim-
posed muscle activity. The bottom channel of
this continuous recording shows the deflections
of a frequency analyzer (after W. Grey Walter).
The seizure activity is characterized by fast spik-
ing of increasing amplitude during the tonic
phase. There are repetitive bursts of spikes mixed
with persisting weak muscle potentials during
the clonic phase (lower portion). There is unre-
markable EEG activity prior to the seizure; the
termination of the convulsion is characterized
by general voltage depression. (From Gastaut H,
Broughton R. Epileptic Seizures. Springfield, IL:
Charles C Thomas; 1972, with permission.)
498 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

(“postseizure stupor,” 37) then dominates the EEG, with grad- term absence was introduced by Calmei (251). The modern
ual frequency increase into the theta and alpha band; the nomenclature (Gastaut et al., 1970) recommends the term
appearance of an organized posterior alpha rhythm signals the absence, but the term petit mal is deeply rooted. Petit mal
return to the waking state. absence seems to be an acceptable compromise.
Prevalence and Age Factor Clinical Manifestations
Tonic–clonic seizures are common at any age, except for the The petit mal absence occurs mainly in children older than 3
first year of life, during which they are largely absent (244). years, with a declining incidence throughout adolescence and
Insufficient myelination of the brain probably precludes them early adulthood; persistence beyond middle adult life is very rare.
at this early age. In the research of Gibbs and Gibbs (37), 5598 The attack consists of a sudden lapse of consciousness with
patients of a population of 11,612 people with epilepsy (48.2%) impairment of mental functions. Its usual duration ranges from
had grand mal only; another 3290 patients (28.3%) had grand 5 to 20 seconds; longer absences may occur over 1 to 2 minutes,
mal in combination with other types of seizures. but such an unusual length is a sign of a somewhat complicated
epileptic seizure disorder with some degree of automatism-like
Neurophysiologic Aspects ictal behavior. The absence is associated with interruption of
One is tempted to regard the tonic–clonic seizure as a standard- ongoing activity and, due to moderate impairment of con-
ized maximal and global epileptic response of the brain. sciousness, the patient is unable to see or hear. There is usually
Electroconvulsion studies in the cat, however, have shown that a blank facial expression that contradicts the frequently used
the cerebellum and the lower brainstem do not fully participate term staring spells; a true stare is more likely to occur in tempo-
in the ictal activity (245). The view of a standardized all-out ral lobe epileptics with psychomotor seizures. The eyes drift
response of the brain requires some correction. Schmidt and upward (“star gazing,” 237,252); rhythmical beating of the eye-
Wilder (1968) feel that GTCs “vary in their severity and in the lids at 3 Hz is very common and may be the only apparent
degree to which they incorporate the various tonic and clonic motor manifestation of the attack. Marked orofacial move-
phases.” Variations of the degree of cerebral cortical participa- ments during the absence are suggestive of a more complex
tion in tonic–clonic seizures have been demonstrated in the cat type with poorer response to therapy.
as well as in the human by Rodin et al. (246). This view has been Petit mal absences show a wide variety of mild-to-moderate
supported by the experimental findings of Petsche (247). motor accompaniment; rhythmical eye blinking in synchrony
This implies that tonic–clonic seizures are graded rather than with the spike waves is the most common motor component.
maximal cerebral epileptic responses. These variations could Retropulsion of the head is quite common (“retropulsive petit
explain differences in the response to electroconvulsive therapy mal,” 237); adversive movements and some rhythmically repet-
in psychiatric illness; following the electrically induced GTC, the itive oral motions may also occur. Automatisms occurred in
EEG may show little or marked slowing, probably depending on 163/405 (40%) of children and were more likely with hyperven-
the degree of neuronal participation in the seizure. tilation and with longer seizures, occurring in only 23% of
Quite different is the view of Gastaut and his coworkers. The spontaneous awake seizures (253,254). The most common clin-
emphasis placed on rhythmical 10 Hz activity during a large ical manifestations include swallowing, mouthing, chewing,
portion of the grand mal attack has been discussed; this “epilep- lip smacking, and lip licking and occurred across all absence
tic recruiting rhythm” will follow stimulation only in nonspe- syndromes.
cific reticular thalamic structures projecting diffusely to both There is good reason to assume that the unique nature of
hemispheres over still uncertain connecting pathways. It was felt loss of consciousness during the classical absence is based on a
that the motor, autonomic, and EEG phenomena of the tonic temporary suspension of the “working memory” due to the
phase could be explained by massive discharge of the thalamic powerful accentuation of spike-wave activity in the frontal cor-
and subthalamic brainstem reticular structures; this would also tex (255–257). This explains the unmatched immediate restora-
account for the loss of consciousness. In the clonic phase, the tion of the working memory along with consciousness at the
appearance of slow waves has been attributed to cortical end of the absence (no other form of brief loss of conscious-
inhibitory systems via thalamic and lower brainstem structures ness—syncope, for instance—reverses so quickly). As to the
or, more specifically, via a thalamocaudate circuit branched crucial function of working memory, see Ref. 69.
from the thalamocorticospinal system. These views have been The postural control is grossly maintained, but swaying and
laid down by Gastaut et al. (248), Gastaut and Fischer-Williams stumbling movements may be noted in the standing patient
(242), and, in condensed form, by Gastaut and Broughton (238). (Schmidt and Wilder, 1968). The posture may be altered by sus-
tained or saccadic retroflexion of the head (“retropulsive petit
Absences (Petit Mal)
mal,” 258). Autonomic and especially vasomotor changes are
Terminology common during the petit mal absence (259,260).
The term petit mal arose from the jargon of physicians and Impairment of the level of awareness has been the object of
attendants in the hospitals of Paris early in the 19th century. some studies (260–265). A variety of tests, such as rhythmically
According to Temkin (249), Esquirol (250) distinguished more pressing a button, have been used to demonstrate the lapse of con-
or less severe epileptic attacks as “grand mal” and “petit mal,” sciousness (266). Mirsky and Tecce (267) demonstrated the per-
but his definition of these two terms “is obviously vague.” The sistence of visual evoked potentials during spike-wave discharges.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 499

Absence seizures are divided into typical absence and atypi- (usually slower), may be asymmetric, and usually has a slow
cal absence seizures. Typical absence seizures are classified as background (see above).
simple or complex; a simple absence has a sudden onset without
motor activity; complex absences have associated motor activity EEG Manifestations
or autonomic activity. In a typical absence seizure, it is usually The ictal EEG of the absence seizure is characterized by the gen-
not difficult for an observer to identify the electrographic seizure eralized synchronous 3 Hz spike-wave discharge. No typical
onset and the seizure cessation by clinical observation. absence seizure can occur without this classical pattern, but the
The atypical absence seizure has a less clearly defined onset 3 Hz spike-wave pattern may occur, usually in bursts of less
or cessation, so that it may be difficult to identify the beginning than 5-second duration, without an accompanying clinical
or the end of the seizure without EEG. The atypical absence absence seizure. This should serve as a stern warning not to
seizure may have more pronounced tone changes, a longer equate generalized 3 Hz spike waves with absence seizures; the
duration, and are typically associated with other seizure types latter cannot occur without the former, but the former fre-
and mental retardation. In contrast to the typical absence quently occur without clinical absences (52–54) (Fig. 26.20).
seizure, it is usually very difficult for an observer to identify the The spike-wave discharge is maximal over the frontal mid-
electrographic seizure onset by clinical observation alone. line and may start at a rate of around 4 Hz, quickly slow down
Penry et al. (268) studied 374 absence seizures with video- to 3 to 3.5 Hz, and, during the final phase of the attack, slow
EEG in 48 patients; a simple absence occurred in only 9% of down to about 2.5 Hz. Onset and termination are abrupt; the
patients; automatisms occurred in at least one episode in 88%, attacks may be preceded and immediately followed by normal
clonic components occurred in 41%, and seizures lasted less EEG activity, especially when recorded in the waking–resting
than 20 seconds in duration in 85%. Automatisms may be per- state rather than during hyperventilation or sleep.
severative or occurring de novo, and autonomic abnormalities There may also be a regularity and organization to the
may consist of pupillary dilatation, pallor, flushing, salivation, generalized spike-wave discharges; another term has been frag-
or incontinence. In a recent study, absence seizures ranged mentation, describing spike-wave discharges that occur in frag-
from 1 to 44 seconds in duration, the average duration was 9.4 ments, rather than in a continuous burst.
seconds, and only 65% lasted between 4 and 20 seconds and In the rare cases of the absence seizures of middle or old age,
66% had major impairment in awareness; no myoclonia, the spike-wave complexes are somewhat less impressive, with the
except in the face, occurred in 74%, no photic induction spike component being slower and less prominent (Fig. 26.20).
occurred in 83% (269).
The EEG in a typical absence seizure shows a symmetric 3 Hz Precipitating Factors
generalized spike and wave discharge, which has a faster fre- Hyperventilation is an extremely potent activator of the 3 Hz
quency at the beginning and a slower frequency at the end. The spike-wave discharge with or without clinical petit mal absences.
atypical absence seizure has a frequency between 1.5 to 2.5 Hz Further powerful facilitating mechanisms are non-REM sleep

Figure 26.20 Petit


mal absence, in an
8-year-old patient.
The spike-wave burst
is preceded by an
aborted run of rhyth-
mical posterior slow
3 Hz waves; after
two slow waves, the
attack begins.
500 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

and hypoglycemia. In some cases, intermittent photic stimula- Myoclonic Seizures


tion is very effective. Myoclonus is characterized by a rapid involuntary muscle con-
traction, subtle or massive, usually with locomotor effect, gen-
Age Factor and Prevalence
eralized or limited to certain muscular segments, mostly
Petit mal absences frequently start at the age of 3 years, are predominant in flexor muscles, and more pronounced in upper
almost never found prior to the age of 2.5 years, and if so, carry extremities. The resulting jerk may be synchronous or moder-
a poor prognosis (270). Early onset absence seizures have been ately asynchronous.
associated with the glucose transporter defect (De Vivo syn- Myoclonus may be epileptic or it may represent a dyskinetic
drome) (271). In 34 cases of absence seizures in an age less than disturbance caused by the breakdown of the motor control sys-
4 years, 4/34 had a Glut-1 deficiency (272). In a 24-hour EEG, tems of the cerebellum. A combination of epileptic and appar-
2.5 to 4 Hz spike-wave discharges occurred in 41%, and gener- ently nonepileptic myoclonus may occur. The term myoclonic
alized slowing or attenuation, or no background abnormalities stresses the singular nature (one distinct jerk), whereas clonic
each occurred in 34% (271). Focal discharges were more com- refers to the repetitive type of muscular twitching.
mon in younger children, less than age 2 years, whereas gener- The complexities of the underlying neurophysiologic mech-
alized discharges were more common in children 2 to 8 years anisms can be gleaned from the work of Watson and Denny-
(271). The EEG may improve when the child is postprandial, so Brown (280), Halliday (281–284), Shibasaki and Kuroiwa (285),
an EEG before and after feeding may demonstrate a decrease in Chadwick et al. (286), Hallett et al. (287), and Kelly et al. (288).
epileptiform features (273). Progress in myoclonus research was sparked by the introduc-
Some of the children with absence epilepsy may have a his- tion of myoclonus-triggered back averaging of the concomitant
tory of febrile convulsion in infancy. There is decline of petit EEG (285, also independently developed by Chadwick et al.
mal epilepsy during the second decade, aside from treatment (286)). Hallett (289) has summarized the most recent insights
effects of drastic reduction of attacks. In some cases, the into the physiologic nature of myoclonus. Three major mecha-
absences start around age 9 to 10 years; German schools (see nisms have been pointed out: (i) cortical reflex myoclonus,
Ref. 237) have studied the differences in the course of children often occurring in focal motor epilepsy and associated with
with onset earlier (around 4 years) and later (9 to 15 years). giant somatosensory evoked potentials (based on hyperex-
This and the occasional persistence of petit mal into adult life citability of the sensorimotor cortex; (ii) reticular reflex
are discussed below (274,275). myoclonus, thought to be due to hyperexcitability of the caudal
Janz et al. (276) have de-emphasized the significance of the brainstem reticular formation; EEG spikes are not time locked
dichotomy of pyknolepsy and juvenile absences. In 163 patients with myoclonus and are maximal over vertex; (iii) primary gen-
with absence epilepsy, Trinka et al. (277) reported that 22% eralized epileptic myoclonus, as found in primary generalized
were in an “overlap” group. epilepsy with bilateral predominantly frontocentral EEG event
A clinical absence with generalized 3 Hz spike waves was preceding the myoclonus. Hallett’s (289) subdivision of
triggered by tapping the head of a 2½-year-old child. This well- myoclonic phenomena is strictly physiologic and not etiologi-
documented case of DeMarco (278) makes one wonder if such cally oriented. The authors make it clear that either cortical
an unusually early manifestation (and precipitation) belongs in reflex myoclonus or reticular reflex myoclonus occur in clinical
the category of primary generalized epilepsy. disorders associated with myoclonus (Table 26.2).
The prevalence can be gleaned from the figures of Gibbs and The clinical–semiologic details of the myoclonic movements
Gibbs (37). Among a total of 11,612 patients with epilepsy, only have been extensively described by Gastaut (290) and cine-
335 (2.9%) had absence seizures without other seizure types. matographically demonstrated by Oller-Daurella and Oller-
There were 896 patients (7.7%) with absence seizures in combi- Ferrer-Vidal (266,291).
nation with other seizure types; 706 of them (6.1%) had a com- Myoclonic phenomena are found in various epileptic con-
bination of absence and generalized tonic–clonic seizures. ditions such as primary generalized epilepsy, infantile spasms
These figures increase moderately when one deals with a popu- with hypsarrhythmia, and Lennox–Gastaut syndrome, and in
lation of children and adolescents. degenerative CNS disease, such as Tay–Sachs disease, forms of
Very prolonged absence-like stages, attacks of petit mal–like CNS lipidosis, essential hereditary myoclonus epilepsy
stupor, or petit mal automatisms have been termed petit mal (Lafora–Unverricht–Lundborg), encephalitis, acute or chronic
status (279), whereas the modern terminology recommends the renal failure, acute cerebral anoxia, or postanoxic states. More
term absence status. These states are discussed in the section on extensive studies on the clinical significance of myoclonus
status epilepticus. Whereas all of the ictal manifestations of pri- have been presented by Weingarten (292), Aigner and Mulder
mary generalized epilepsy tend to occur in children and adoles- (293), Gastaut (290), Bauer (294), Kinsbourne and Rosenfield
cents, the absence status not only occurs in elderly patients but (295), Sulibhavi and Schneck (296), Farrell and Swanson (297),
may even have its onset in old age. Charlton (298), Niedermeyer et al. (299), and Aicardi (291).
It has been pointed out that, in neonates, nonketotic
Neurophysiologic Aspects and Clinical Significance hyperglycinemia represents an important cause of early
These subjects are discussed in the section on primary general- myoclonic epilepsy. In infants and young children, progressive
ized epilepsy. myoclonic encephalopathies are usually due to metabolic or
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 501

Tabl e 2 6 . 2

Characteristics of Major Neurophysiologic Types of Myoclonus a

Cortical Reflex Reticular Reflex Primary Generalized


Myoclonus Myoclonus Epilepsy Myoclonus
Clinical setting of Fragment of focal (partial) Fragment of bilateral or Fragment of primary generalized
occurrence epileptic seizures generalized epileptic epilepsy
manifestations (aside
from primary generalized
epilepsy)
Localization of Involves only a few adjacent Myoclonus tends to affect Small bilateral myoclonic, often
myoclonus muscles the whole body fingers only, minimyoclonus
(mimicking tremulousness); also
major whole-body myclonic jerks
Precipitation Spontaneous or induced Spontaneous or induced No extrinsic precipitation except for
(accentuated) by voluntary (accentuate) by voluntary photosensitivity
movement (motor action) movement (motor action)
Bilateral synchrony Asynchronous; usually Usually synchronous, but Synchronous (fairly precise bilateral
of myoclonus unilateral synchrony may be quite synchrony)
imperfect
Somatosensory Enhanced (“giant SSEP”) Unremarkable Unremarkable
evoked potentials
(SSEP)
Focal time-locked Well demonstrable, with EEG event (spike) often Negative event precedes myoclonus,
EEG event preceding spontaneous and associated myoclonus but with bilateral frontocentral
reflex-induced jerks located not time locked; spike maximum, lasting 30–100 msec for
over appropriated motor generalized maximal over major jerks (well time locked) and
cortex region vertex 100–250 msec for minimyoclonus
(less well time locked)
CNS origin of Focal: motor cortex (local ? Caudal brainstem reticular Presumably cortical (frontal,
neurophysiologic hyperexcitability) formation supplementary motor cortex
event as starting point)
Clinical Focal motor (elementary Postanoxic action Primary generalized epilepsy and
significance partial) epileptic seizure, (intention) myoclonus, related epileptic seizure disorders
probably also Creutzfeldt– renal encephalopathy
Jakob disease and others (uremia), others

aModified from Hallett M. Myoclonus: relation to epilepsy. Epilepsia (New York). 1985;11:567–577.

genetic disorders: mitochondrial diseases, gangliosidoses and rhythmia (West syndrome), the EEG correlate of the myoclonus
ceroid-lipofuscinoses, and in older children, Lafora’s disease is variable and reaches from sudden flattening or desynchroniza-
and its variants are in the foreground as the cause of tion to massive spiking and unaltered ictal recordings. Myoclonus
myoclonus (300). This investigator has also emphasized that a may occur with or without spiking in degenerative CNS disease.
majority of myoclonic epilepsies do not belong in the group of The term negative myoclonus was introduced by Tassinari (301)
neurodegenerative disorders; consequently, their prognosis is and denotes brief repetitive lapses of postural tone time locked to
much better. spikes over the contralateral central region (302,303). In some
From the EEG viewpoint, myoclonus is associated with mas- patients, this phenomenon is associated with acute valproate
sive spike discharges and especially with bursts of bilateral or encephalopathy (valproate stupor). According to Rubboli et al.
generalized synchronous polyspikes in patients with primary (304), epileptic negative myoclonus is accompanied by an
generalized epilepsy, and thus with the related photoconvulsive inhibitory frontal spike component.
response to intermittent photic stimulation, and also in patients Table 26.3 shows the EEG correlates in a variety of clinical
with Lennox–Gastaut syndrome. In infantile spasms with hypsar- conditions with myoclonic seizures.
502 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 2 6 . 3

Clinical and EEG Characteristics of Chronic Conditions with Myoclonus

Epileptologic Neurologic or
Age Symptomatology Mental Deficits EEG
Primary generalized Mainly 4–16 Petit mal absences None Principally generalized
epilepsy (synonyms: years; in case Petit mal–grand mal- or bilateral anterior
common generalized, grand mal, 10–50 myoclonus synchronous spikes,
centrencephalic, years spike waves (3 Hz, 4 Hz),
corticoreticular Grand mal- polyspikes, activated by
epilepsy) myoclonus hyperventilation, flicker,
Absence (petit mal) sleep (non-REM)
status, often with
myoclonus
Infantile spasms Age 4 months– Myoclonic, mainly Varying from normal Hypsarrhythmia: high-
2.5 years head nodding findings to forms of voltage output, irregular
(hypsarrhythmia) Jackknifing with cerebral palsy, slowing with massive
tonic component hydrocephaly, spikes, polyspikes, mostly
microcephaly, mental occipital maximum
retardation Asleep: enhanced bursts
with stretches of
depressed voltage
Lennox–Gastaut Onset mostly age Numerous minor Varying from normal Slow spike-wave
syndrome 1–10 years, motor types of or more or less complex (“petit mal
becoming poorly seizures; tonic, severe neurologic variant” after Gibbs and
distinct after atonic–akinetic, deficits, types of coworkers), 1–2.5 Hz; in
age 30 myoclonic, also cerebral palsy, etc; sleep, runs of rapid
psychomotor, grand mental retardation spikes, 10–20 Hz; also
mal petit mal polyspikes; frequently
general slowing
Cherry-red spot- Childhood to Massive myoclonus, Fundi: cherry-red- Positive spikes over
myoclonus syndrome adolescence also facial; grand mal spot; relatively mild vertex in brief bursts
may occur neurologic deficit,
normal intelligence
Essential hereditary Onset in Myoclonus Cerebellar Several disorganized,
myoclonus epilepsy childhood or Grand mal symptomatology of predominantly slow with
(Lafora–Unverricht– adolescence (the varying degree: numerous single spikes,
Lundborg) earlier the onset, dysmetria, often with needlelike
the more serious intentional tremor, spikes of myogenic
the progression) speech and gait character; few or no
disturbance; polyspikes; sometimes
progressive dementia more rhythmical slow
activity with spikes
Benign myclonus Onset in Myoclonus None Mostly normal (a
(Hartung) probably childhood personal observation of
related to an abnormal record is an
paramyoclonus exception)
multiplex (Friedreich)
Postanoxic Any age Myoclonus, intention Cerebellar deficit, Disorganized, with
myoclonus or action type, EEG possibly other numerous spikes,
spikes maximal over posterior anoxic bilateral synchronous,
vertex deficits vertex or frontocentral
Dementia
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 503

Focal Seizures Adversive Seizures (Versive, Ipsiversive,


Focal seizures are discussed in greater detail in the chapter on or Contraversive Seizures)
epilepsy in adults. A few focal seizures that are particular to Adversive seizures may be limited to conjugated eye move-
childhood are mentioned here. ments; there is usually extreme lateral gaze to the side contralat-
eral to the lesion. The eye movement may be tonic (oculogyric)
Focal Motor Seizures (Rolandic Motor Seizures) or clonic (oculoclonic, also called epileptic nystagmus). These
Clonic twitching of the contralateral muscles of the body is the manifestations must be carefully distinguished from nonepilep-
typical seizure manifestation of a localized discharge within the tic nystagmus and oculogyric crises such as brainstem disease
precentral gyrus. Bravais (305) studied focal motor seizures in and basal ganglia disease.
his thesis. Jackson’s work, however, and the classical electrical More commonly, the adversion consists of deviation of head
stimulation studies of Fritsch and Hitzig (306), Ferrier (307), and eyes; even the entire body may rotate to one side (usually
and Luciani (308) have brought the excitability of the human contralateral, i.e., opposite side of the focus). Sometimes, the
cortex into the right perspective. contralateral arm is raised in tonic extension, with the subject
In accordance with the somatotopic arrangement within apparently looking at his raised hand (238). This has been
the precentral gyrus, the clonic movements are initially limited impressively demonstrated by the illustrations of Aird and
to the corresponding area of the body and tend to spread dur- Woodbury (314) and Matthes (315).
ing the attack. Such a spread (for instance, from the facial mus- These attacks may occur with a loss of consciousness (fron-
cles to the leg) is well known as the Jacksonian march. topolar region, middle, and superior frontal gyrus) or in a
Such a Jacksonian march does not materialize in many cases conscious state (posterior portion of frontal lobe, vicinity of
and the attacks remain limited to one body region, usually precentral gyrus) (316,317).
thumb, fingers, lips, eyelids, or great toe. The cortical represen- The ictal EEG usually shows more or less rhythmical slowing
tation of these functions, according to Gastaut and Broughton with spikes over the affected area and, in most cases, over a large
(238), represents a phylogenetic acquisition of humans, who neighboring region. For this reason, a precise focal EEG diagno-
have been endowed with a particularly large cortical motor sis is difficult or impossible.
zone. Any region may be involved; even trunk muscles may par- Adversive seizure phenomena are not limited to the frontal
ticipate. There is no impairment of consciousness during these region; oculoclonic seizures may be frontal as well as occipital.
attacks. Involuntary lingual movements were described as “lin- The occipital origin of combined oculoclonic and adversive
gual seizures” occurring in conjunction with contralateral cen- seizures has been clearly shown by Fossas et al. (318). The initi-
trofrontal ictal spiking (309). ation of grand mal seizures by a brief adversive movement is
A status of constant clonic activity in one muscular region is quite common and cannot be regarded as a reliable localizing
known as epilepsia partialis continua or Koshevnikov syndrome. sign. This has been further corroborated by Robillard et al.
Bilateral focal motor attacks are very rare. Focal motor seizures (319), as well as by Ochs et al. (320). Further considerations of
may be caused by an ipsilateral lesion (310), but such cases are this topic are found in the work of Quesney et al. (321).
exceptional (paradoxical lateralization).
The ictal EEG is expected to show impressive spiking con- Epileptic Spasm/Partial Seizures
tralaterally over the involved motor cortex, but such cases with A combination of flexor spasms with preceding or subsequent
precise focal EEG documentation are seldom observed. partial clonic seizures has been described by Pachatz et al.
Widespread EEG changes with desynchronization, spiking, (322), showing in the EEG a juxtaposition of focal and general-
and more or less rhythmical theta or delta activity may be ized ictal activity. These attacks occur in childhood/adolescence
noted, or there may be a complete lack of ictal EEG activity, with cryptogenic or residual-acquired etiologies; their progno-
also caused in part by movement artifact. The discharge may sis is relatively favorable.
be subcortical in a three-dimensional involvement of cortical
and deep structures (discussed under rolandic epilepsy), or Hypomotor Seizures
cortical spiking may be too small and desynchronized. The dis-
sociation between spiking over the motor cortex and corre- This term was introduced by Kallen et al. (323) and pertains
sponding motor effects has been investigated by Elger and to seizures of infancy and childhood consisting of diminished
Speckmann (311) in the rat; spiking in lamina V after local motor activity along with an undetermined level of con-
penicillin gives rise to contralateral twitching without con- sciousness. These have also been referred to as behavioral
comitant spiking on the cortical surface. With spike activity arrest seizures, and may often be accompanied by version of
in superficial and deep cortical layers, there are accompany- the eyes (244). The EEG shows typical ictal changes of focal
ing motor effects. These authors also introduced the term seizures, often arising from the temporal, parietal, or occipital
vertical inhibition for a mechanism responsible for the failure lobes.
of descending neuronal activity during epicortical epilepti-
form potentials (312). Quiet epileptogenic foci within the Seizures Occurring in Infantile Spasms
motor cortex can be activated by transcranial magnetic stimu- Only (Jackknifing, Salaam)
lation (313). These are discussed below (see Infantile Spasms).
504 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Primary Generalized Epilepsy Tabl e 2 6 . 4 A


(“Idiopathic Generalized Epilepsy”)
versus Secondarily Generalized Epilepsy Classification of Epilepsy Syndromes
The term primary generalized epilepsy has fallen out of vogue
Epilepsy syndromes
with the ILAE classification commission, but the concept is an
Neonatal
important one for many neurologists and a suitable substitute
Early myoclonic encephalopathy
term has yet to be officially suggested. A secondary generalized
Ohtahara syndrome
epilepsy, or secondary bilateral synchrony, implies that there is
a focal onset with secondary generalization. Infancy
The entire concept of a primary generalized epilepsy has its Epilepsy of infancy with migrating focal seizures
foundation in the EEG. It implies that the clinical and EEG phe- West syndrome
nomena of the seizures are generalized from the start and that Myoclonic epilepsy in infancy
there is no structural epileptogenic focus. The total lack of Benign infantile epilepsy
structural lesions has been challenged by Meencke and Janz Benign familial infantile epilepsy
(324,325), who found areas of minor dysgenetic changes in Dravet syndrome
autopsy studies of patients with presumed primary generalized Myoclonic encephalopathy in nonprogressive disorders
epilepsy. However, Lyon and Gastaut (326) have minimized the Childhood
significance of microdysgenesis, which may occur in other con- Febrile seizures plus
ditions or in neurologically normal controls. Panayiotopoulos syndrome
Currently, it is suggested that the terms focal and general- Epilepsy with myoclonic–atonic seizures
ized should apply to seizures and not to epilepsies (epileptic Benign epilepsy with centrotemporal spikes
syndromes) (327). For example, focal seizures have occurred Autosomal-dominant nocturnal frontal lobe epilepsy
in several of the generalized epilepsy syndromes, such as juve- Late onset childhood occipital epilepsy
nile myoclonic epilepsy and “benign rolandic spikes” may Epilepsy with myoclonic absences
occasionally develop in childhood absence epilepsy, especially Lennox–Gastaut syndrome
after suppression of the absences and the spike waves with Epileptic encephalopathy with continuous spike and wave
medication. Landau–Kleffner syndrome
Lombroso (328) reported his experience with a mean long- Childhood absence epilepsy
term follow-up of 16 years (up to 22 years) in 58 patients with Epilepsy with eyelid myoclonia
various primary generalized epilepsies. Thirty-two of these
Adolescence
patients had focal features present without structural lesion. He
Juvenile absence epilepsy
hypothesized either microdysgenesis or the development of
Juvenile myoclonic epilepsy
localized hyperexcitability in low threshold structures second-
Epilepsy with generalized tonic–clonic seizures alone
ary to long-term generalized spike-wave activity.
Progressive myoclonus epilepsies
Autosomal-dominant epilepsy with auditory features
EPILEPTIC SYNDROMES Familial temporal lobe epilepsies
The syndromatologic approach to the epilepsies is of fairly
recent origin. It has been recognized over the last few decades
that certain forms of epileptic seizure disorders have special
clinical and EEG characteristics regardless of their polyetiologic (332), Helbig et al. (333), Hirose et al. (334), Ottman et al. (335),
background. These forms represent epileptologic entities or Scheffer et al. (336) or are specified. Epigenetic mechanisms may
epileptic syndromes with important differences in course and also be important (337). These references are used for the genetic
prognosis. The correct diagnosis of these syndromes is there- information for the following epilepsy syndromes, unless other-
fore of considerable practical significance. Furthermore, these wise specified.
syndromes are age-dependent and chiefly occur in certain age The frequency of the epilepsy syndromes is remarkably sim-
ranges, especially infancy and childhood but frequently persist- ilar across series. Most epilepsies can be diagnosed early on,
ing through adolescence into adult life. Accordingly, Berg and but some require follow-up for proper identification (338)
others have presented an age-related approach to the listing of (Table 26.4B).
the various epilepsy syndromes (235) (Table 26.4A).
Teaming modern genomic technologies with improved classi- NEONATAL
fication of seizures and epileptic syndromes is the identification of
the underlying genes, receptors and channels that are associated Benign Neonatal Seizures (BNS)
with seizure types and specific syndromes. The genetic findings There are familial (benign familial neonatal seizures [BFNS])
for the various syndromes come from several sources: Chahine and nonfamilial forms. BFNS typically begin on day 2 or 3 of
and Mikati (329), Lagae (330), Lu and Wang (331), Lucarini et al. life, may be difficult to control initially, but typically resolve
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 505

Tabl e 2 6 . 4 B

Frequency of Epilepsy Syndromes

Kramer Dura-Trave Berg (N = 613) at Berg (N = 613)


Epilepsy Syndrome (N = 440) (N = 365) Initial Diagnosis After 2 Years
Benign rolandic epilepsy 8% 11.5% 9.6% 10.3%
CEOP 2 9 0.3 0.5
BMEI 0.8 0.2 0.3
CAE 12.3 12.1 12.4
JAE 2.2 2.4 2.8
JME 0.9 0.8 2.0 2.4
GTC on awakening 0.5 0.3 0.3
West syndrome 9 4.1 3.0 3.1
LGS 1.5 0.5 0.7 3.1
Myoclonic–astatic 0.2 6 1.6 1.3
Myoclonic absence 1.9 0.3 0.3
SMEI 1.4 0.2 0.2
ESES 0.2 1.1 0.2 0.2
Acquired Ep aphasia (LKS) 0.2 0.5
Ohtahara syndrome 0.2

within the first several months. In the nonfamilial form, which encodes the alpha-2 subunit of the voltage-gated
seizures typically begin during the first week of life, commonly sodium channel (2q24).
on Day 5 of life. The clinical manifestations consist of tonic
posturing, automatisms, apnea, or focal or generalized clonic Early Myoclonic Encephalopathy (EME)
activity. The seizures may rarely persist into adulthood. (Neonatal Myoclonic Encephalopathy)
Typically the interictal EEG is normal in both forms but The onset of EME is almost always in the newborn period. First
can have focal or multifocal epileptiform discharges, a discon- described by Aicardi and Goutires (342), EME is characterized
tinuous background, or the theta-pointu alternant pattern. by fragmentary or erratic partial myoclonus, massive body
This theta pattern consists of runs of 4 to 7 Hz activity, with myoclonus, focal motor seizures, and tonic spasms. Erratic par-
admixed sharp waves, alternating from side to side. It is not tial myoclonus (face, limbs, sometimes just a finger, or the orbic-
specific for this disorder. In the few recorded seizures in the ular area) may appear in the very first hours of life (343) and
familial variety, seizures start with an electrodecrement for 5 may persist during sleep (344,345). The myoclonus may shift
to 19 seconds, followed by 1 to 2 minute bilateral spikes or from one part of the body to another in an anarchic and asyn-
sharp waves, the prominence could vary from side to side, and chronous manner (343). Massive myoclonus may not always be
had no postictal EEG depression (339). Focal seizures (340) present. Tonic spasms usually appear later, mostly around the
and focal seizures with generalization have also been recorded age of 3 months, and are similar to those found in infantile
(341). In the nonfamilial variety, the ictal EEG shows rhyth- spasms (West syndrome). Focal (partial) motor seizures, includ-
mic spikes or slow waves that may predominate in the central ing only eye deviation, may occur (344). Autonomic phenomena
regions. such as apnea or flushing of the face may be present (343).
The familial form, BFNS, has been associated with muta- There are very typical EEG changes (Fig. 26.21) with con-
tions in the KCNQ2 (20q13) and KCNQ3 (8q24) voltage- stantly repetitive generalized bursts of high-voltage slow waves
gated potassium channels, responsible for the M-current. This with spikes, lasting a few seconds and separated from each other
slowly activating current regulates subthreshold neuronal by brief stretches of sudden voltage depression or near flatness.
excitability. Retigabine may be helpful since it blocks the Although the bursts are synchronous, the spikes themselves
M-current. The benign neonatal-infantile seizures have been show no bilateral synchrony. The bursts of this burst-suppression
associated with a missense mutation in the SCN2A gene, pattern are not associated with motor manifestations. The
506 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Murakami et al. (351) have emphasized the essentially differ-


ent nature of early myoclonic encephalopathy and Ohtahara
syndrome. Transition into West syndrome and Lennox–Gastaut
syndrome is common in the Ohtahara syndrome and rare in
early myoclonic encephalopathy (351).

INFANCY
Epilepsy of Infancy w ith Migrating Focal Seizures
Malignant migrating partial seizures in infancy is a severe
epileptic disorder described in 1995 (352). It is characterized by
nearly continuous electrographic seizures involving multiple
independent areas of onset, beginning in the first 6 months of
life in normal infants with developmental delay and intractable
seizures. No underlying cause has been found, but a functional
or metabolic dysfunction is suspected. Its prevalence is
unknown; series involving only small numbers of patients have
been published, but the spectrum of clinical manifestations
appears to be large. Most seizures are focal and accompanied by
autonomic manifestations or subtle version; many have motor
manifestations that migrate from one side of the body to the
Figure 26.21 Early infantile myoclonic encephalopathy in a 3-month- other. Two of the original 14 patients described by Coppola and
old patient. Note the burst-suppression-like alternation of mixed slow colleagues presented with status epilepticus. Sometimes very
activity (with some intermingled slow and spiky discharges) and mild clinical features were observed during habitual seizures.
stretches of flattening. The interictal EEG was normal in three patients at onset, but
was always abnormal during evolution, with multifocal spikes
and sleep abnormalities. The ictal EEG is characterized by
burst-suppression pattern varies, in that it may only be seen monomorphic rhythmic theta or alpha activity progressively
during sleep, but not in the waking EEG. involving multiple sites, moving from one area to another (thus
There is pronounced impairment of the neurologic and men- the term migrating), and progressively spreading to involve
tal status; marked hypotonia with hyperreflexia is common and larger area. Additional seizures starting in other areas in either
microcephaly may develop. Neuroimaging is usually unremark- hemisphere could start before the end of the first event, or
able. The prevalence of this syndrome is quite low. Familial immediately follow it.
occurrence has been reported and there is some evidence of an Neuroimaging has not revealed any consistent abnormality
autosomal-recessive inheritance. No single progressive meta- but most patients develop microcephaly during the first year of
bolic or neurodegenerative CNS disease has been identified as life. During short-term follow-up, 3/14 patients described by
causative, although nonketotic hyperglycinemia must be con- Coppola and colleagues died. Conventional treatments were
sidered. We have seen this syndrome with the ARX mutation. ineffective, except for the combination of stiripentol and clon-
The prognosis is very poor; most children die prior to the age azepam in two children, in which some neurologic improvement
of 2 years. was also noted. We (personal observations) and others (353) sus-
pect that there is a wider clinical spectrum of this disorder. In
Early Infantile Epileptic Encephalopathy particular we have observed patients with less-than-constant
(Ohtahara Syndrome) seizures and Marsh and colleagues report patients with slightly
Ohtahara et al. (346) and Ohtahara (347) described this better outcomes than those reported by Coppola et al.
syndrome, introducing the term early infantile epileptic
encephalopathy (EIEE). It is characterized by tonic spasms occur- INFANTILE SPASMS (WEST SYNDROME)
ring before the age of 20 days and EEG changes similar to those
in EME (342,348). In contrast to EME, the burst-suppression Infantile spasms (IS) are an age-dependent “malignant” epilep-
pattern occurs in both the awake and sleep states and the inter- tic syndrome characterized by the triad: myoclonic or tonic
burst interval tends to be relatively constant, often about 3 to seizures, hypsarrhythmia, and mental retardation, referred to as
5 seconds. The clinical picture and EEG closely resemble those of West syndrome. The peak onset is between 4 and 7 months and
nonketotic hyperglycinemia (343). The prognosis is extremely always before 12 months of age. Hypsarrhythmia (37) is an EEG
poor, with a mortality of 50% before the age of 1 month. Its term that denotes the EEG correlate of the condition. Infantile
prevalence is very low. Ohtahara syndrome has been associated spasms are considered an epileptic encephalopathy, defined as a
with a de novo mutation in the gene encoding STXBP1 (also disorder in which the epileptiform activity, in this specific case,
known as MUNC18-1), a protein essential for synaptic vesicle the hypsarrhythmia, causes neurologic dysfunction. This is
release and with the ARX mutation (349,350). referred to as a catastrophic epilepsy, with a very poor prognosis
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 507

unless the epilepsy is controlled. The hypsarrhythmia pattern


may be a variant form of nonconvulsive status epilepticus (354)
and it is possible that the lack of an organized background and
normal brain rhythms critically interferes with the ability of the
brain to process information in an orderly manner.
The current classification system (2001 glossary, 235) now
uses the term “epileptic spasms” because spasms may continue
or develop after infancy (355). We continue using the term
infantile spasms here.
Infantile spasms are divided into symptomatic, cryptogenic,
and idiopathic forms. Symptomatic infantile spasms are sec-
ondary to a known neurologic disorder, such as tuberous scle-
rosis; cryptogenic refers to a suspected, but not definitely
identified neurologic disorder; idiopathic refers to a case in
which no specific disorder has been identified. The prognosis is
related to the underlying etiology and is worse when the spasms Figure 26.23 Infantile spasms-hypsarrhythmia in an 8-month-old
are either symptomatic or cryptogenic. The spasms themselves patient. Note the high-voltage output (see parameter of sensitivity) and
consist of sudden tonic and myoclonic phenomena, including posterior maximum of spikes. Sleep record.
brief head nods, with quick extension and flexion movements
of the trunk, arms, and legs, occurring in clusters and especially
during transitions from sleeping to awaking. These are also single spikes and sharp waves, as well as very brief sequences of
described as flexor, extensor, or mixed. The actual spasms may polyspikes that are usually of smaller amplitude and the spike
resolve, but other seizure types occur later. activity almost always has a posterior predominance (Fig. 26.23).
The term hypsarrhythmia (37) is derived from the Greek word The ictal EEG, occurring at the time of the actual infantile
hypselos, which means “high,” thus indicating the high voltage spasms, is quite variable. The most common ictal finding is an
that generally predominates. No hypsarrhythmic recording can electrodecremental response, a sudden suppression of the EEG
be appropriately obtained at the standard sensitivity of the EEG activity that lasts for several seconds (118,356). However, fast
apparatus; lowering the sensitivity is required in order to prevent activity and high-voltage spikes (37) or polyspikes and slow
“blocking.” Bursts of very high voltage slow waves occur in an waves may be present (118). Fusco and Vigevano (357) reported
irregular fashion with a varying degree of bilateral synchrony, that the characteristic ictal pattern is a high-amplitude vertex
which usually increases in sleep. The stages of early non-REM positive wave. A sleep recording is a necessity because the hyp-
sleep are particularly conducive to a typical hypsarrhythmic sarrhythmia may be confined only to the sleep portion in some
recording. Long stretches of high-voltage slow and admixed spike children (356).
activity may suddenly be interrupted by a generalized elec- The great variability of EEG abnormalities in long-term
trodecrement, or less commonly in only a few EEG channels or recordings has been described by Hrachovy et al. (358):
over one hemisphere; these electrodecrements are practically increased interhemispheric synchronization, asymmetrical
limited to sleep tracings (Fig. 26.22). The spike activity shows hypsarrhythmia, hypsarrhythmia with a consistent focus of
abnormal discharge, hypsarrhythmia with episodes of attenua-
tion, and hypsarrhythmia comprising primarily high-voltage
slow activity with little sharp wave or spike activity. Kramer et
al. (359) studied these hypsarrhythmia variant patterns and
devised a scoring system. The variant patterns occurred in 69%
of 53 consecutive EEGs. Prognosis was not related to the pres-
ence of any one variant pattern, but was associated with faster
background activity ( 75% delta), a lower total hypsarrhyth-
mia score ( or = 10), and with absence of electrodecremental
discharges on the pre–ACTH EEG.
The evaluation for infantile spasms includes neuroimaging
to exclude anatomic lesions, but other disorders, especially
metabolic and infectious, and now, specific genetic disorders,
need to be excluded. X-linked infantile spasms have been asso-
ciated with a defect in the ARX gene (360) and CDKL5 (cyclin-
dependent kinase like 5) gene. Secondary focal spasms may
occur from focal lesions, such as tumor, stroke, or a focal corti-
cal dysplasia. ACTH has been considered the drug of choice by
Figure 26.22 Markedly depressed stretches in an 8-month-old patient many, although benzodiazepines, especially nitrazepam, val-
with infantile spasms-hypsarrhythmia. Record was obtained in sleep. proic acid, and vigabatrin have been used. A UK study showed
508 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

that ACTH and oral prednisone have a better efficacy than viga- Tabl e 2 6 . 5
batrin (361). ACTH and vigabatrin are the only AEDs for which
evidence exists for efficacy and for controlling the hypsarrhyth- Infantile Spasms: Differential Diagnosis
mia (362,363). Vigabatrin may be especially effective when
spasms are caused by tuberous sclerosis (364). However, vigaba- Neonatal sleep myoclonus
trin has been associated with retinal dysfunction and visual Benign myoclonus of infancy
field defects and has only just been released in the United States
Head banging (jactatio capitis nocturnis)
20 years after its introduction.
The EEG shows an almost immediate improvement under Spasmus nutans
effective therapy, but a clinical improvement may not necessar- Sandifer syndrome, abdominal pain (colic), esophageal
ily follow. Complete normalization may occur, but this may be spasm
only temporary, and epileptiform activity, mostly over posterior
Shuddering attacks
regions, and other seizure types may develop. In many cases with
a poor therapeutic response and especially in those with preex- Hyperekplexia
isting brain damage, a transition to the Lennox–Gastaut syn- Paroxysmal infantile dystonia, choreoathetosis, torticollis
drome is common. This occurs in 58.7% according to Ohtahara
Benign paroxysmal tonic upward gaze
et al. (365) and in 50% to 70% according to Hughes (366). In
addition, infantile spasms may persist as epileptic spasms (367), Infantile self-gratification
or infantile spasms in remission may recur and persist as epilep- Breath-holding episodes with myoclonus/myotonic
tic spasms (368). posturing
If a focal lesion, especially a tumor, is seen on neuroimaging,
Intoxications
then surgical resection may be curative. Even in the absence of
a lesion on neuroimaging, surgery may be done on refractory Increased moro reflex with opisthotonus
cases if an epileptogenic focus is identified, especially using the Myoclonic encephalopathy
PET scan (369). Most of these cases result from focal cortical
dysplasias. From Fejerman N. Differential diagnosis. In: Dulac O, Chugani HT, Dalla
A fine clinical–electrical analysis of infantile spasms was car- Bernardina B, eds. Infantile Spasms and West Syndrome. London: WB Saunders;
ried out by Fusco and Vigevano (357). In both cryptogenic and 1994:88–98.
symptomatic cases, the EEG pattern that corresponds to the
spasm consists of a slow wave at the vertex. Two other patterns weeks. Hypsarrhythmia may evolve later into the multiple inde-
include fast activity, called “spindle-like” and the electrodecre- pendent spike syndrome.
ment. The vertex positive slow wave was associated with the On the other hand, if the expected hypsarrhythmia is not
actual spasm, the fast activity with clinical staring, and the elec- found in a suspected case of infantile spasms, the presumptive
trodecrement was postictal. The persistence of the hypsarrhyth- diagnosis may be wrong. The clinical differential diagnosis of
mic pattern during a cluster of spasms is found in both infantile spasms or hypsarrhythmia has been beautifully
symptomatic and the idiopathic and cryptogenic groups and demonstrated by Roger et al. (371) and includes a variety of con-
asymmetric spasms suggest a symptomatic etiology. Persistence ditions. In spasmus nutans, the EEG is normal. Jactatio capitis
of EEG hypsarrhythmia during a cluster of spasms appears to nocturna also has a normal EEG. With salaam tic or “salutatory”
indicate a favorable prognosis in the idiopathic group (357). spasms (Moro), there are nonspecific EEG abnormalities, some-
According to Koo and Hwang (370), occipital lesions are asso- times with spikes in combination with epileptic seizures, but no
ciated with the earliest onset of spasms and frontal lesions with hypsarrhythmia. In myoclonic encephalopathy (372), the EEG is
late onset. Our clinical experience has been different and we normal (Table 26.5).
have not been able to confirm these observations. The reader is
cautioned to avoid placing too much emphasis on the prognos- Benign Myoclonic Epilepsy in Infancy
tic value of ictal hypsarrhythmia or to infer much about the Myoclonic seizures are the predominant manifestation in this
location of the lesion based upon the age of presentation. epilepsy syndrome. Infants with normal behavioral and cognition
Hypsarrhythmia is not always a reliable EEG correlate of develop myoclonic jerks mostly of the head and proximal arms,
infantile spasms. There are clinically convincing cases with no occurring in isolation or in brief runs of recurrent jerks. There is
hypsarrhythmia, but in these rare exceptions the voltage output often a vocalization associated with an expiratory noise (Lin,
is unusually high and there are almost invariably interictal 1998), considered characteristic of this disorder. Rarely, general-
epileptiform discharges. A completely normal EEG obtained ized clonic seizures may occur during sleep or upon awakening
awake and asleep virtually excludes the diagnosis of infantile that may be prolonged and associated with cyanosis (Vegevano,
spasms. It should be stressed here that it does not exclude later 1990). The ictal EEG shows a burst of generalized or diffuse spike-
forms of spasms like periodic spasms. In infantile spasms unless wave activity with the myoclonia, otherwise the interictal EEG is
there is a rapid response to treatment, hypsarrhythmia is likely normal (Auvin, 2005). However, focal discharges, especially in the
to appear in the further course of such infants, commonly for rolandic and vertex regions may mimic a focal epilepsy (373).
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 509

Benign Infantile Epilepsy and Benign closure. All of these children had a genetic cause for their
Familial Infantile Epilepsy epilepsy. The second subgroup included five patients with
Benign infantile epilepsy may be seen in sporadic or familial inhibitory phenomena associated with a dystonic component
forms (374,375; Watanabe, 1993; Fukuyama, 1963). In these and sudden irregular rapid lightning-like jerks. The EEG
disorders, infants manifest focal seizures that may present with showed subcontinuous multifocal slow spike waves, predomi-
subtle behavioral arrest or, on the other extreme, bilateral con- nating in frontocentral regions. These patients either had corti-
vulsive features. The interictal EEG is usually normal, though cal malformations or the etiology was unknown. The third
some benign infantile epilepsies may have peculiar low or subgroup included six children who initially suffered from
medium voltage vertex spikes or sharp waves with a dome-like myoclonic absences. The status was characterized by subcontin-
morphology (376). Ictal discharges often arise from the tempo- uous generalized spike-wave-type paroxysms correlating with
ral region or posterior quadrant, but this is relatively nonspe- the rhythmic myoclonia of face and limbs. After 1 to 3 weeks,
cific for focal seizures in the immature. the EEG showed sharp theta waves with very slow pseudorhyth-
mic continuous spikes in the central regions and vertex. The
cause in this subgroup was perinatal anoxic injury.
Severe Myoclonic Epilepsy in Infants
(Dravet Syndrome)
First reported by Dravet (377) and subsequently confirmed CHILDHOOD
by Dalla Bernardina et al. (378), Severe Myoclonic Epilepsy of
Infancy (SMEI) is an uncommon syndrome. There is typically a Febrile Convulsions
family history of epilepsy or febrile seizures (FS), and seizures Age and Definition
begin in the first year of life (typically around 6 months of age) The ICES considers FS a situation-related seizure: the situation
in a previously normal infant who develops prolonged general- being the illness with fever. A benign febrile seizure is defined as
ized or unilateral febrile clonic seizures, with then frequently a short ( 15 minutes) generalized tonic–clonic seizure without
occurring convulsive seizures. In addition, there may be attacks of significant postictal depression, in a child with a nonfocal neu-
pallor, cyanosis, atonic phenomena, and automatisms (379). rologic examination without a family history of epilepsy. A
Moderate fever may accompany the first attacks. Multiple seizure febrile seizure is otherwise considered complex if it is longer or
types, especially myoclonic seizures then develop between 1 and predominantly focal. This condition is probably the most com-
4 years of age, and focal seizures may also occur. mon epileptic seizure disorder; about 3% to 4% of all children
The interictal EEG may initially be normal although we have have at least one febrile seizure in infancy or early childhood
seen low amplitude occipital polyspikes in some infants in the (2.2% to 4% according to Knudsen (381)). The seizures tend to
first year of life. The theta-pointu pattern may be seen. More occur between the ages of 6 months and 5 years, especially
prominent paroxysmal abnormalities tend to develop during between 6 months and 3 years. The onset falls into the range of
the second year of life with generalized spikes, polyspikes, and 6 to 24 months. Beaumanoir (382) thinks that it is unwise to
spike-wave-like activity, which may be sleep-activated and pro- call fever-induced convulsions after the age of 4 years “simple
gressive background slowing develops. Photosensitivity is com- febrile convulsions.”
mon and develops in the first phase of the disorder, and focal The conventional wisdom regarding the EEG in benign FS
abnormalities may be present. Dravet syndrome has been asso- is that the interictal EEG is normal, whereas in a child with
ciated with mutations in the SCN1A gene (19q13.1) and in the epilepsy, the interictal EEG may be abnormal, with associated
GABA RA2 (5q34) receptor. These occur in about 70% to 80% epileptiform features. The recent practice parameter on benign
of those with Dravet syndrome (336). FS does not require an EEG in the evaluation of a benign febrile
Psychomotor retardation occurs, usually beginning in the seizure (383). However, a careful literature review shows that
second year of life, with ataxia, interictal myoclonus, and hyper- there may not be such a clear EEG distinction for a “benign
reflexia. The seizures are typically treatment resistant. febrile seizure.” Alvarez (96) reported hypnagogic paroxysmal
spike-wave activity (minimal epileptiform features, sharp waves
Myoclonic Encephalopathy in embedded into hyperventilation or hypnagogic hypersyn-
Nonprogressive Disorders chrony) occurring with a higher incidence in children with FS.
Myoclonic encephalopathy in nonprogressive disorders is char- This is similar to the epileptiform activity admixed into vertex
acterized by recurrent myoclonic status in infants and young waves and sleep spindles, called dyshormia, which refers to
children with nonprogressive encephalopathy. In a study abnormal epileptiform arousal features during sleep. These fea-
reviewing the findings in 29 patients, 3 main subgroups were tures may indicate a lower seizure threshold and explains why
described (380). The first subgroup of 18 patients presented they occur in both the normal population and in those with
with myoclonic absences and rhythmic myoclonias. These were epilepsy. Japanese child neurologists rely more on EEG and have
followed by a brief silent period during which there was sub- introduced the concept of epileptic versus nonepileptic febrile
continuous delta–theta activity in the central areas, and rhyth- convulsions (384). However, children with epileptic febrile con-
mic delta waves with superimposed spikes mainly involving the vulsions treated with AEDs had the same recurrence rate as
parietooccipital regions. The latter were often activated by eye those not treated with AEDs.
510 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

In one classic reference, Frantzen et al. concluded that EEG Findings


epileptiform discharges were not predictive of later develop- Ictal EEG tracings are hard to obtain in a truly simple febrile
ment of a FS (Frantzen, 1968). They did not, however, convulsion. Although generalized tonic–clonic seizure EEG
separate out the various types of interictal epileptiform changes might be inferred from the apparent widespread and
discharges in their analysis. Frantzen and colleagues did symmetric body movements described by many parents, this
note that focal slowing was seen in the posterior head sequence is rarely seen in infants (Korrf, 2005). Indeed, EEG
region in children with prolonged FS. They noted that foci recordings obtained in inpatients with acute FS show lateralized
of spikes could develop in these children approximately EEG changes (see samples by Lennox-Buchthal (387)), but an
1.5 years later, but their study did not have sufficient follow- argument could be made that these are most likely the more dra-
up to determine if spikes were predictive of later develop- matic and severe cases.
ment of epilepsy. In a pure benign febrile seizure, the interictal recording should
be normal. Gibbs and Gibbs (83) pointed out that short spike-
Genetics of Febrile Seizures wave-like bursts in drowsiness and sleep may occur (“pseudo petit
It has been thought that FS are inherited in an autosomal-dom- mal discharge”). Des Termes et al. (388) found 31 children with
inant manner with incomplete penetrance. In families with FS, spike foci among 500 patients with febrile convulsions. The most
six genetic loci have been mapped: FEB 1 (8q13-q21), FEB 2 common site was the occipital lobe and, in 88%, the spike focus
(19p13.3), FEB 3 (2q23-q24), FEB 4 (5q14-q15), FEB 5 (6q22- disappeared within 3 years. Yamatogi et al. (389) have stressed the
q24), and FEB 6 (18p11.2) (385). Defects in the interleukin excellent prognosis of children with febrile convulsions and per-
genes interleukin 1 beta and interleukin 1 receptor antagonist fectly normal EEG tracings. Yamatogi and Ohtahara (390) have
have also been associated with FS (386) (Table 26.6). placed emphasis on the clinical value of follow-up EEG studies.

Tabl e 2 6 . 6

Differences between Febrile Convulsions and Seizures due to Febrile Brain Disease

Febrile Convulsions Seizures in Febrile Brain Disease


Typical age range 6 months to 3 years (seldom 5 years) Mainly 0–3 years
Genetic predisposition May be strong Mostly minor or insignificant
to seizures
Type of seizure Tonic–clonic (modified or Tonic–clonic (grand mal-like) or hemiconvulsions
attenuated grand mal)
Duration of seizure Mostly 1–3 min seldom prolonged Often prolonged, 10 min to hours, status-like or in
rapid succession
Clinical setting in At the onset of a febrile disease, In a variety of CNS infections (encephalitis,
which seizures occur mostly upper respiratory illness, meningoencephalitis), intracranial venous
often coinciding with the first sharp thrombosis, cerebrovascular accidents of infancy;
rise in temperature also in exanthema subitum and after smallpox
vaccination, but usually less severe
Type of underlying None Various types of inflammatory and vascular
cerebral pathology changes, in milder cases limited to edema
Postictal neurologic Very uncommon Common and often mixed with pathology-
deficit (Todd’s paralysis) determined neurologic defect
EEG Rapidly normalizes after convulsion Abnormal throughout febrile episode, abnormal
normal interval tracings in 80–90% in interval (except for mild encephalitis)
Anticonvulsive Not necessary (neither for acute Acutely needed (preferably benzodiazepines,
medication convulsions nor for prevention of phenobarbital), long-term treatment required
further seizures) afterward (except for mild encephalitis)
Prognosis Excellent in the vast majority Guarded; neurologic defects and further seizures
(especially those with normal common
interval EEG)
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 511

Abnormal interictal EEG tracings are likely to indicate may be much higher if children with atypical and inconspicu-
underlying cerebral impairment with paroxysmal properties; ous presentation are included.
these infants might be candidates for a febrile epileptic manifes- The aforementioned “mainly autonomic” seizures consist of
tation (i.e., for a chronic epileptic seizure disorder) in the feeling sick, looking pale, nausea, retching, and vomiting. There
future. According to Des Termes et al. (388), the prognosis for may also be cyanosis, mydriasis (or miosis), cardiorespiratory
these children is mostly favorable. and thermoregulatory alterations, incontinence of urine and/or
feces. Apnea and cardiac asystole may be exceptionally severe.
Generalized Epilepsy w ith Febrile Two thirds of the seizures occur in sleep. Unilateral eye devia-
Seizures Plus (GEFS ) tion is common. The seizures commonly end with hemiconvul-
Generalized epilepsy with FS plus (GEFS ) was described in sions or with Jacksonian march or rarely with a generalized
1997 (Scheffer, 1997). It is characterized by the association of tonic–clonic seizure, which, quite rarely, evolve into status
generalized FS beyond the age of 6 years and afebrile generalized epilepticus. The duration varies from 1 to 30 minutes, and
convulsions; a positive family history of epilepsy with variable autonomic status epilepticus may occur. The final portion of
phenotypes; and a favorable evolution in the majority of cases. the seizure may consist of ictal syncope with flaccidity. After
The first description in a large family revealed that the clinical sufficient sleep, the child is perfectly normal.
spectrum of GEFS comprised cases of simple FS; FS beyond In a prospective study of 192 children with PS by Caraballo
the age of 6 years (FS ); FS and absences; FS and myoclonic et al. (393), the following inclusion criteria were used: (i) auto-
seizures; FS and atonic seizures; and myoclonic–astatic nomic manifestations, visual symptoms, and/or simple motor
epilepsy. This spectrum was further extended with other types of focal seizures followed or not by impairment of consciousness
epilepsy, such as FS and temporal lobe epilepsy (Singh, 1999), with or without secondary generalization; (ii) functional occip-
Dravet syndrome, and cryptogenic Lennox–Gastaut syndrome ital and extra-occipital spikes alone or combined on the inter-
(Singh, 2001). The pattern of transmission in the first family ictal EEG, or a normal EEG; (iii) normal neurologic and mental
suggested an autosomal-dominant inheritance. The authors status; and (iv) normal brain imaging. Of the 172 treated chil-
hypothesized that a single major gene defect, with possible dren, 152 became seizure free with treatment, 6% had infre-
additive gene effect, was responsible for the phenotypic vari- quent seizures, and 6% had frequent seizures (380).
ability on the entire spectrum of the generalized epilepsies The EEG is the most useful test, once neuroimaging is nor-
(Scheffer, Singh, 1999). Extensive genetic analyses in these mal. Ninety percent of EEGs show multifocal spikes and spike
families identified a mutation in the sodium-channel 1 waves with posterior accentuation. The spikes were occipital in
subunit gene (SCN 1) on chromosome region 19q13.1 75%, temporal in 26%, and frontal in 10%; with the occipital
(Wallace). Additional mutations were later discovered in other spikes, 60% were bilateral and synchronous, with voltage asym-
genes, such as the sodium-channel 1 subunit gene on chro- metries, in the other 40%, spikes were unilateral and greater on
mosome region 2q24 (Escayg), or the GABAA receptor 2- the right side (70%). The spikes were sleep-activated in 90% and
subunit gene on chromosome region 5q34 (Baulac). These three had generalized spikes. The ictal EEG demonstrates rhyth-
findings underline the genetic complexity of this disorder, mic delta activity intermixed with usually small spikes. Onset is
which remains to be completely understood. Some have unilateral, often posterior, but may also be anterior and not
argued that this is not a specific epilepsy syndrome, but rather strictly localized to one electrode. In four EEGs with seizures
a description of a genetic susceptibility. In either case, this recorded from the onset, the onset was occiptotemporal in two,
research has been one of the most exciting and productive and one each from the frontal or frontotemporal regions (394).
areas in pediatric epilepsy. The EEG in PS may show shifting or multiple epileptic foci,
some with an occipital predominance (Ferrie, 2006). In follow-
The Panayiotopoulos Syndrome up EEGs, 17% had a shift of epileptiform activity from the
In an excellent overview of benign childhood partial seizures, occipital to centrotemporal areas and 3% to frontal areas (394).
Panayiotopoulos (391) described a special type of early- In PS, frontal spikes may be activated by occipital foci (395) and
onset occipital seizure for which he coined the term the “synchronous occipital and frontopolar spike phenomenon”
Panayiotopoulos syndrome. This syndrome was subsequently (an occipital and frontopolar spike) has been reported (396), in
highlighted in a special study (392) with the following initial which negative occipital spikes precede the frontal spikes, sug-
characterization of this syndrome: “Panayiotopoulos syndrome gesting propagation; this did not occur in all children.
is a childhood-related idiopathic benign susceptibility to The family history is usually negative and there is no relation-
partial, mainly autonomic, seizures that may be genetically ship to migraine in the family. The alternative diagnoses made in
determined. The children have normal physical and neuropsy- these children include encephalitis, syncope, migraine, sleep dis-
chological development.” order, and gastroenteritis, with SE occurring in a large percent-
The age of onset shows a range from 1 to 14 years with a age of cases. A recent consensus statement defined autonomic SE
peak at 4 to 5; 76% start between ages 3 and 6 years. The preva- as epileptic activity manifesting as autonomic dysfunction at
lence is about 13% among children with a seizure onset seizure onset lasting greater than 30 minutes and emphasized
between ages 3 and 6 years and overall, about 0.2% to 0.3% of that it also occurs in childhood symptomatic epilepsies, rarely in
the general population of children may be affected. This figure adults, and is not limited to the Panayiotopoulos syndrome.
512 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Myoclonic–Astatic Epilepsy of Childhood Age and Prevalence


This syndrome has been described by Doose (397) (“akinetic Benign rolandic epilepsy occurs at age 3 to 12 years, with the
petit mal”). This condition is often confused with Lennox– majority 6 to 10 years old. Disappearance of the seizures during
Gastaut syndrome, but is clearly different. Doose (398) feels that adolescence or even prior to puberty is the rule. The seizures may
this epileptic condition is taxonomically closer to primary gen- occasionally recur much later in life, probably due to seizure-
eralized epilepsy rather than to the Lennox–Gastaut syndrome. facilitating factors such as severe illness or toxic-metabolic factors.
Deonna et al. (1986) reported a combination of myoclonic– The gender distribution shows that boys are more often
astatic epilepsy and “benign focal epilepsy of childhood.” affected. The prevalence is not quite clear and might be some-
There are now indications that the Lennox–Gastaut syndrome where between 5% and 10% in a population of epileptics below
and the syndrome of myoclonic–astatic epilepsy are indeed two age 15. Beaumanoir (382) goes even further; she thinks that the
separate entities with different neurophysiologic mechanisms prevalence of benign rolandic epilepsy exceeds that of petit mal
generating the myoclonus. In the Lennox–Gastaut syndrome, absences and reaches 14% to 15% of all childhood epilepsy
myoclonus originates from a stable generator in the frontal cor- cases (Fig. 26.24).
tex. Spreading to contra- and ipsilateral cortical areas, whereas in
myoclonic–astatic epilepsy, myoclonus appears to be a primary Ictal Manifestations
generalized epileptic phenomenon (399). The actual seizures are hardly ever seen by the physician, even by
Onset is typically between 7 months and 6 years of age. The the epileptologist who sees sizable numbers of these children
seizures consist primarily of generalized myoclonic, astatic, or (413,420,421). One therefore depends heavily on descriptions by
myoclonic–astatic seizures, short absences, and mostly GTC the patient or the patient’s family; nocturnal videotape or long-
seizures typically without tonic seizure or drop attacks. term monitoring recordings from the Epilepsy Monitoring Unit
However, tonic seizures may occur in cases with a poor outcome. are quite helpful. Whether the seizures are focal or generalized,
At the onset, the EEG background may be normal and char- they tend to occur during nocturnal sleep (413), mostly during
acterized by a 4 to 7 Hz rhythm with a parietal midline accen- the last hour of sleep or in the first 2 hours. About 80% of the
tuation. There are generalized 2 to 3 Hz spike and wave attacks occur in sleep; of the remaining 20%, about 10% take
discharges with admixed slow waves. There is a high incidence place shortly after awakening. Nocturnal seizures may awaken
of seizures or EEG abnormalities in relatives. Prior to the onset the child afterward. Preservation of consciousness and hence the
of the seizures, which may start as FS, the development is nor- ability to describe the experienced seizure, indicating focality,
mal. Valproic acid and ethosuximide have been considered the was found in 58% and generalized tonic–clonic seizures were
drugs of choice, and the ketogenic diet has also been effective noted in 26% (413).
(400). The prognosis is variable, but in our experience many Focal seizures often involve the face. The midtemporal spike
can do well with prompt initiation of effective treatment. localization has been thought to be related to paroxysmal activ-
The myoclonic–astatic seizure is now referred to as a ity in the very closely located lower portion of the motor strip
myoclonic–atonic seizure (235). (faciolaryngopharyngeal muscles); this has been stressed by
Lombroso (412), who uses the term sylvian seizures. Hemifacial
Benign Rolandic Epilepsy twitching is definitely more common than clonic motions in
In children with spikes over the central region and/or adjacent the contralateral arm; least common is clonic activity in the leg.
midtemporal and parietal areas, a benign and readily control- In some cases, the entire half of the body participates, but a typ-
lable type of epileptic seizure disorder with focal motor seizures ical Jacksonian march does not seem to occur. Speech arrest is
and/or generalized tonic–clonic seizures are the rule. This is a quite common (39% of the seizures, 413). This is apparently an
special form of childhood epilepsy. Earlier observations of ictal anarthria with preserved internal speech.
Gastaut (401), Hess (402), Nayrac and Beaussart (403), Isler Oropharyngeal involvement is very often reported (53%,
and Hess (404), and Gibbs and Gibbs (83) have set the stage for 413), with sounds described as “guttural,” “gargling,” “throaty,”
more recent work in this field (46,405–415). This work has out- “wheezing,” or “as if going to vomit.” The feeling of suffocation
lined the characteristics of benign rolandic epilepsy, as this con- is reported as coming from the mouth but not from the chest.
dition has been termed over the past few years. These patients also have focal seizures that are not rolandic,
Beaumanoir (382) has listed this form of childhood epilepsy with blindness, vertigo, and torsion of the body as ictal signs.
among the primary generalized epilepsies despite its prominent This underscores the complexity of the underlying neurophys-
focal features in the ictal and EEG semiology. Such a classifica- iologic mechanisms.
tion certainly appears to be provocative. There are indeed cer-
tain relationships between benign rolandic epilepsy and EEG
primary generalized epilepsies, and conversion from one form Spiking over the central–midtemporal area in children is of lim-
to the other may occur (416–419). There is certainly good rea- ited epileptogenicity. It is reasonable to presume that 50% to
son to separate benign rolandic epilepsies from the bulk of focal 70% of these children have seizures and the remaining 30% to
(partial) epileptic seizure disorders, which will be presented 50% are seizure free. These latter patients are referred to the EEG
somewhat later. In this section, we also discuss benign seizure laboratory because of a variety of symptoms such as behavior
disorders in children with spikes outside the rolandic region. disorder, headaches, and other complaints or deviations.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 513

Figure 26.24 Presumed physiopathogenetic mechanisms


involved in primary generalized epilepsy. The dyshormic
and the photosensitive subgroups show considerable over-
lap. Note the greater significance of the age factor in the
dyshormic group; also note differences of spike-wave fre-
quencies and types of seizures. CGE, common generalized
epilepsy.

The spatial distribution of the spike activity requires an considered pathognomonic for benign rolandic epilepsy show
appropriate number of electrodes; the International 10-20 dipole configurations with the negative pole over the central
Electrode System is particularly suitable. Otherwise, the rolandic (centrotemporal) region and a positive pole over the frontal
cortex may lie between a frontal and a parietal electrode, and region (303,425). Typical spikes indicating benign rolandic
strictly local spiking may escape detection, especially when the seizure disorder start with a spike of centroparietal negativity
midtemporal region is not explored ideally. In our experience, a and superior frontal negativity, changing within 12 msec to sole
central maximum of spike discharge is slightly more often noted central negativity and after further 16 msec to a dipole of central
than a midtemporal maximum. As above, the EEG has distinc- negativity and frontomedian positivity (424). Similar findings
tive, high amplitude, diphasic spikes or sharp waves with a were observed with visual analysis during ictal manifestations of
prominent slow wave, in the midtemporal (T3,T4) and central benign rolandic epilepsy (426). According to Legarda and
(C3,C4) regions. However, when additional scalp electrodes are Jayakar (427), the dipolar distribution of rolandic spikes was not
placed, maximum negativity was in the central region, either in found to be a reliable indicator of potential epileptogenicity.
a “high” (C3,C4) or “low” (C5,C6) location (Legarda, 1994). Yoshinaga et al. (428) have given strong support to the
The spikes themselves are large and may be either spikes in importance of dipole tracing in benign rolandic and other types
the strict sense or sharp waves (Fig. 26.25). of childhood epilepsies. Confusing elements in the assessment
Central spiking with dipole formation has been reported by of rolandic spikes and their dipoles may arise from the assump-
Gregory and Wong (19,422). In fact, Gregory and Wong (422) tion of a single dipole—a concept presently widely supplanted
have stressed the clinical relevance of presence or absence of by the concept of multiple sources and extended source geom-
dipole fields in children with epileptic seizure disorder and etry (303,422). It has to be added that every investigator of the
rolandic spikes. According to their data, the presence of dipole cerebral dipole problem has to take into account the stringent
discharges (usually located over frontal or temporal regions) is limitations of dipole theory (429).
associated with a benign functional rolandic focus with little or Spiking is usually enhanced in light non-REM sleep, during
no clinical problems, whereas absence of a dipole discharge is which the discharges may become extremely abundant. Their
more likely to be found in children with neurologic and behav- random character may give way to quasirhythmic or periodic
ioral abnormalities. In our opinion, however, the differential spiking at intervals of less than 1 second; previously unilateral
diagnosis of benign rolandic epilepsy should not hinge on cen- spikes become bilateral synchronous or asynchronous (382). In
tral spikes of dipole or nondipole type. many children, rolandic spikes are found in the sleep portion
In the course of the 1990s, presence and orientation of only. According to Beaumanoir (382), REM sleep restores the
dipoles of rolandic spikes have become a widely debated issue. unilateral character of the spiking. Beaumanoir (382) also
According to Wong (423), the dipole-dependent topography of described the occurrence of bilateral parietooccipital 4 Hz
rolandic spikes “contains information on the behavior or func- spike-wave-like discharges of moderate voltage seen in the wak-
tional state of brain tissue even distant to the brain region ing patient (Fig. 26.26).
responsible for the spike generation.” Van der Meij et al. (424) In some patients, interictal rolandic spikes can be blocked
feel that sequential mapping can differentiate between epileptic with contralateral finger or hand movements (Niedermeyer,
and nonepileptic rolandic spikes. Classically, rolandic spikes 1970), either actively or passively (in sleep) performed. The
514 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 26.25 A: Left, a few suggestions of right cen-


tral spikes in a 9-year-old boy. Center, some activity
over right central area during flicker (same patient).
Right, marked spikes and sharp waves over central
regions, especially on the left, during non-REM sleep
(same patient). (From Niedermeyer E, Koshino Y. My-
rhythmus: vorkommen und klinische bedeutung. EEG-
EMG. 1975;6:69–78.) B: Sleep tracing in a 7-year-old
boy with grand mal attacks since age 3. Postictally, the
patient was temporarily aphasic and hemiparetic on
the right side. Note the left midtemporal spikes and a
burst of generalized spikes mixed with slow activity.

Figure 26.26 A: A 5-year-old patient with recent onset of petit mal absences. The recorded generalized spike-wave burst
was subclinical. B: The same patient at age 8 years. There are numerous independent spikes over left central and right cen-
troparietal regions. This case is a typical example of crossover between primary generalized epilepsy and benign rolandic
epilepsy.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 515

Figure 26.27 A 14-year-old patient with severe personality disorder


but no history of epileptic seizures. The lower portion was recorded
with identical montage, sensitivity, and paper speed. Note abundance
of right central spikes and excellent blocking response to contralateral
movement (clenching left fist).

reactivity of rolandic spikes was studied by Fonseca et al. (430),


who noted a strong spike blocking response tongue movements
inside or outside the mouth. On the other hand, Nadkarni et al.
(431) reported a child with centro-midtemporal spikes that
could be triggered by blinking.
The rest of the tracing is usually normal in these patients,
and the frequency spectrum corresponds to age. A central mu
rhythm is sometimes present (382,415), and there is reason to
presume that central spikes of childhood may be gradually Figure 26.28 A: An 8-year-old boy with epilepsia partialis continua or
replaced by mu rhythm, at least in some of the patients (Fig. focal motor status (clonic motions, left arm). The EEG shows consider-
26.27). There is indubitable evidence that, in a limited number able slowing (note lowered gain). There is evidence of constant muscle
of patients, the central spike activity can be blocked by con- activity, but no authentic cerebral spikes are demonstrable. B: Same
tralateral fist clenching or, even better, by alternate clenching patient. Electrocorticography. In general anesthesia, suppression of
and opening of the fist (45,415). This provides further evidence focal motor attacks but prominent spiking, especially in leads 4 (pre-
for the functional character of the spikes (Fig. 26.28). central gyrus) and 7 (postcentral gyrus).
Mitsudome et al. (432) have pointed out that, in benign
rolandic epilepsy, central spikes may be associated with local
slowing. Both rolandic spikes and local slowing were found to that omission of midline leads would result in missing the abnor-
respond to clonazepam. mality (436). Some of these patients show focal motor or sensory
In a small number of cases, the spike activity shows a consis- ictal activity of leg predominance; more often, the ictal symptoms
tent maximum over the vertex (433–435) or over the centropari- do not correspond with the spike localization. Even the parietal
etal midline region, which may be the sole region of spiking, so midline may be strongly involved in the spike activity (435).
516 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Clinical Signs of Nonictal Character has had central spikes in the past, but such conclusions can be
One must agree with Beaumanoir (382) when she points out made only with reservations.
that neurologic deficits are not compatible with benign A younger age of onset is associated with a tendency for more
rolandic epilepsy. This condition is based on dysfunction rather frequent seizures (443). Massa et al. (444) identified the follow-
than structural pathology. It is worthwhile, however, to search ing EEG characteristics as predictive of a “complicated” course:
carefully for a true intracranial lesion. Arteriovenous malfor- intermittent slow wave focus; multiple, asynchronous spike-
mation, tumor, or cortical dysplasia occasionally may be the wave foci; spike-wave clusters; generalized 3 Hz spike-wave dis-
cause of discharges and associated seizure. charges; discharges with positive or negative myoclonia; or
Central, midtemporal, or parietal spikes or spikes over the abundance of interictal abnormalities during waking or sleep.
midline may also occur in children with evidence of cerebral
Etiologic Considerations
palsy, in mostly diplegic, quadriplegic, or choreoathetoid forms.
In these children, rolandic spikes have a different connotation This form of epilepsy is due to dysfunction rather than to pathol-
and do not herald a good prognosis for the seizure disorder. ogy. A genetic basis is the most logical thought. The work of Bray
The reader will find more extensive discussion in the section on and Wiser (445) on familial occurrence of midtemporal spikes
cerebral palsy. has pioneered in this area, and other studies will certainly follow
Behavior disorders are very common in children with true suit. The work of Doose et al. (446) has further substantiated the
rolandic spikes; they may range from hyperkinetic behavior and significance of genetic factors. A positive family history of
signs of minimal cerebral dysfunction to severe anxiety neuro- epilepsy was reported in 11.3% of 80 children with benign
sis. Various types of headaches may occur; see the symptom rolandic epilepsy (447). Gelisse et al. (448) have pointed out that
profile of Gibbs and Gibbs (83). Migraine headaches may be benign rolandic epilepsy can also affect children with static brain
associated with the centro-temporal spike (Anderman, 341). lesions, which, however, have no influences on the benign course.
The intelligence is normal in true benign rolandic epilepsy. These authors feel that neuroimaging procedures are usually
Much research has recently been devoted to the neuropsy- unnecessary in children with benign rolandic epilepsy.
chological deficits that may accompany benign rolandic The centro-temporal sharp waves seen in “benign rolandic
epilepsy. There have been learning disabilities, attention deficit epilepsy” have an autosomal-dominant inheritance (449) and
disorder, attention deficit disorder with hyperactivity, impaired are associated with a defect in the elongator protein complex 4
motor and visual coordination, and specific language disorders. (ELP4), involved in transcription and tRNA modification (450).
Weglage et al. (437) correlated IQ deficits with greater than 6
spikes per minute in the waking EEG; children with 5 spikes or Problems of Differential Diagnosis
less had no deficits. Staden et al. (438) found language dysfunc- Children with benign rolandic epilepsy must be differentiated
tion in 13/20 children and correlated this with greater than 10 from the following groups:
spikes per minute. Croona et al. (439) demonstrated improve- 1. Children with rolandic spikes and who have no seizures what-
ment in the follow-up studies, after the spikes had resolved. soever (these children should not be considered as epileptics;
Massa (2001) correlated the following EEG findings with about 30% to 50% of the children with rolandic spikes have
cognitive and behavioral problems: intermittent slow wave no overt clinical seizures (451);
focus, multiple asysnchronous spike-wave foci, long spike-wave 2. Children with rolandic spikes and a history of antecedent
clusters, generalized 3 Hz discharges, atonia, myoclonia or brief brain damage or cerebral palsy;
alteration of awareness with the discharges, and abundance of 3. Children who have typical psychomotor seizures and evidence
interictal epileptiform discharges. Nicolai et al. reported that of temporal lobe epilepsy that may gradually progress in
dysfunction was related to the spike-wave index. severity; these children may have atypical spike localization
Course (central, midtemporal), whereas the classical anterotemporal
sharp wave focus does not materialize before adolescence;
The seizures are easily controlled with routine anticonvulsive 4. Children with midtemporal spikes, marked tendency to gen-
treatment; oxcarbazepine and levetiracetam are preferable. eralization of spike-wave formation, clinically with aphasia
Treatment may even be withheld unless seizures repeat them- (Landau–Kleffner syndrome);
selves (382). A population study in Canada demonstrated that 5. Children with frequent focal motor seizures that become
treatment is not required (440). The seizures typically remit progressively worse: “malignant” rolandic epilepsy of child-
before 15 to 16 years of age. According to Lerman, the seizure hood; and
frequency is low, 13% may have just one seizure, 66% have 6. Children with centroparietal spikes elicited by tactile stimu-
infrequent seizures, and 21% have frequent seizures (441). lation of corresponding cutaneous areas of the body.
Freedom from seizures in adolescence is the rule. Lerman
and Kivity (442) reported EEG normalization by adulthood in The differentiation of these conditions rests on a careful com-
all patients. The return of a single major convulsion may occa- bined clinical–electroencephalographic assessment of each case.
sionally occur under the influence of infections, stress, or toxic
substances. These cases show no resurgence of the central spike Spike Foci Outside the Rolandic Region in Children
focus, which renders the EEG diagnosis very difficult. The pres- Occipital spike foci are usually found between the ages of 2 and
ence of central mu rhythm may serve as a hint that the patient 5 years. These children show no neurologic or ophthalmologic
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 517

deficit; about 40% of them have clinical seizures, mostly gener- syndrome) are quite rare but do constitute a special epileptologic
alized tonic–clonic, with good prognosis. entity (456; Niedermeyer et al., 1977c; 457). Motor deficits and
According to Gibbs and Gibbs (83), frontal spike foci in chil- mental decline are associated with the seizure disorder (Figs.
dren are associated with epileptogenicity, with about 80% hav- 26.29 and 26.30). The etiology is poorly defined; chronic local-
ing overt seizures, and a guarded prognosis. Multiple spike foci ized encephalitis may be one of the causes (458–460). Chronic
(two or more areas of independent spiking) are also highly encephalitis, or Rasmussen syndrome, is characterized by refrac-
epileptogenic; the prognosis is guarded and probably fairly tory focal seizures, frequently epilepsia partialis continua, and
good if rolandic spikes predominate. progressive hemiatrophy with progressive contralateral hemi-
A theory of spike migration of childhood from occipital to paresis (461,462). Although various autoimmune treatments,
midtemporal and then to conversion into 14 and 6 Hz positive such as IVIG, corticosteroids, or plasmapheresis have been used
bursts, with good prognosis, or anterior temporal spike activity, (462,463), hemispherectomy seems to be the only effective cure,
with poor prognosis, was advanced by Gibbs (452) but has found and limited cortical excisions or lobectomies are ineffective (464;
little acceptance (see Ref. 404). “Focus-migration” must be Muto, 2010) (Fig. 26.30).
understood strictly in terms of dysfunction-induced epileptoge- The EEG shows endless sequences of ictal spike discharges
nesis, which may undergo changes of spatial origin and distribu- during focal motor attacks but may not demonstrate any elec-
tion. A structurally determined epileptogenic focus is unlikely to trographic seizure activity during epilepsia partialis continua,
display any wanderlust. Andermann and Oguni (453) and Blume which probably originate from deep structures or possibly from
(454) have discussed the pros and cons of the focus-migration lamina V of the motor cortex without participation of the
theory; both authors have cautioned against the facile use of this superficial layers (Elger and Speckman 1979, 1980, 1983).
term. In a recent study, the migration of spike foci was correlated A recent European conference outlined the following two-
with a better outcome in pediatric epilepsy: seizure frequency tiered clinical, EEG, and MRI criteria (A and B) for diagnosis.
and the number of prescribed AEDs were significantly greater in Tier A includes: clinical: focal seizures, with or without epilep-
those with a “fixed focus” versus a “migrating focus” (455). sia partialis continua and unilateral cortical deficits; EEG: uni-
hemispheric slowing with or without epileptiform activity and
Considerations of Basic Mechanisms unilateral seizure onset; MRI: unihemispheric cortical atrophy
True benign rolandic epilepsy is likely to be based on temporary and at least one of either gray or white matter T2/FLAIR hyper-
paroxysmal hyperirritability of the motor cortex, which natu- intensity or hyperintense signal or atrophy of the caudate. If all
rally has a lower threshold of epileptic excitability. This is merely three of A are present, this confirms the diagnosis, or if not, 2
a working hypothesis in need of further substantiating evidence. out of 3 of B: epilepsia partialis continua or progressive unilat-
eral cortical deficits, on MRI, progressive unihemispheric corti-
Rasmussen Encephalitis and Other Causes of cal atrophy, or histopatholgy showing encephalitis with
“Malignant” Rolandic Epilepsy activated microglial cells and reactive astrocytosis (461).
Cases of progressively worsening focal motor seizures and pro- A malignant rolandic-sylvian seizure syndrome has been
longed episodes of epilepsia partialis continua (Kozhevnikov described in children consisting of refractory sensorimotor

Figure 26.29 Ictal spiking in


right frontal leads (depth and
scalp) associated with left facial
twitching in an 8-year-old boy with
chronic encephalitis, not relieved
by right frontal lobectomy. Much
improved after right hemispherec-
tomy at age 15.
518 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Genetic screening for the nicotinic acetylcholine receptor gene


mutation (CHRNA4 and CHRNB2) is commercially available.

Benign Occipital Lobe Epilepsy


This epileptologic entity has been individualized by Gastaut
(468) and updated by Taylor et al. (469,470). It has been divided
into early onset, or Panayiotopoulos syndrome, or later onset,
Gastaut syndrome. However, Panayiotopoulos syndrome is now
considered an autonomic, rather than an occipital epilepsy (471).
Benign occipital epilepsy occurs in children and adolescents;
there is no persistence into adult life. Seizures are mostly initiated
by visual symptoms (amaurosis, phosphenes, or figurative hallu-
cinations), which may be followed by other seizure manifesta-
tions (mostly hemiclonic attacks, but also psychomotor seizures
and grand mal). Migrainous or pseudomigrainous symptoms
with headache, nausea, and vomiting often follow the seizure.
The EEG shows very characteristic changes. In the interval,
there are frequently recurrent stretches of rhythmical and well-
formed spike-wave discharges of high voltage, at 1.5 to 3 Hz,
located over occipital regions and the immediate vicinity.
Moderate lateralization is common. Ictal EEG discharges consist
of continuous spike-wave activity over one occipital region even
though the interictal spike-wave pattern is bilateral (468). Figure
26.31 shows an example of the occipital spike-wave discharge.

Figure 26.30 The patient discussed in Figure 26.29 after right hemi-
spherectomy. Note the almost complete loss of activities on the right.

seizures, typically presenting as “tics,” frontotemporal EEG


spikes, rolandic-sylvian spike sources on dipole analysis or
MEG, and cognitive regression (465). However, even with very
frequent seizures, remission may occur (466).

Autosomal-Dominant Nocturnal
Frontal Lobe Epilepsy
Patients with autosomal-dominant nocturnal frontal lobe
epilepsy have clinical manifestations that may include sudden
awakenings with dystonic movements (tonic posturing) or vio-
lent movements and the hypermotor seizures or tonic postur-
ing may often be brief and nocturnal as the name suggests. The
EEG findings may be relatively bland or may show focal fea-
tures such as slowing and spikes. Ictal recordings are character-
ized by rhythmic frontal discharges with a diffuse frontal
predominance. The ictal discharges may lateralize or even local-
ize to a region suggesting a focal structural lesion. However,
interictal and ictal epileptiform activity may be lacking and the
seizures mistaken for sleep disorders, nocturnal paroxysmal
dystonia, or nonepileptic seizures. In a study of 40 patients,
interictal and ictal EEGs were normal in 26% (Oldani, 1998) Figure 26.31 A 13-year-old boy with benign occipital lobe epilepsy.
and in a study of children, the interictal EEG was normal in 3/8 There are widespread 1.5 to 2 Hz spike-wave complexes with occipital-
but ictal EEG was abnormal in all showing bifrontal beta activ- posterotemporal maximum, occurring in long bursts during wakeful-
ity with or without rhythmic slowing (467). ness. The spike waves are lateralized to the left (but the lateralization
A careful family history is warranted before considering focal was not consistent). At other times, the patient had a well-developed
respective surgery in people with nocturnal frontal lobe seizures. posterior alpha rhythm.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 519

When one considers the indubitably existing relationship to pies such as corticosteroid therapy, the ketogenic diet, or vagus
migraine, the question arises as to whether there is a continuum nerve stimulation (VNS) may be helpful.
of more or less epileptogenic conditions between benign occip- The EEG hallmark of LGS is the slow spike and wave dis-
ital lobe epilepsy and forms of basilar artery migraine with charge (1.5 to 2.5 Hz) superimposed on a slow background.
paroxysmal posterior EEG changes (472–474). Lugaresi et al. Bursts of fast rhythms (10 Hz) are also a prerequisite; these may
(475) reported cases of “scotosensitive seizures” elicited by dark- be frequent, sleep-activated, and typically occur during the
ness and eye closure; these authors feel that this seizure type is tonic seizure. Multifocal spikes may also be seen and in some
closely related to benign occipital lobe epilepsy. cases may be the predominant epileptiform activity.
Benign occipital lobe epilepsy is unassociated with photosen- A multitude of etiologies may cause this condition, and in
sitivity; there are no paroxysmal responses to flickering light. many cases the cause remains unknown. According to Gastaut et
al. (485), the prevalence of the Lennox–Gastaut syndrome in a
Epilepsy w ith Myoclonic Absences major epilepsy center (5.1% with 10.2% of patients below age 15
(Myoclonic Absence Epilepsy) and 0.6% of patients above this age) is greater than infantile
This was first described by Tassinari (476,477). Epilepsy with spasms. LGS usually starts between the ages of 1 and 10 years;
myoclonic absences consists of myoclonic absence seizures with onset in the second decade of life is much less common (16% after
variable alteration of awareness associated with bilateral rhyth- Komai (486)) and adult onset is rare (487). Onset at age 6 to 12
mic myoclonic jerks that may have a tonic component (478). The months has been observed and requires solid EEG documentation
myoclonias typically involve the shoulders, arms, and legs with a for differentiation from hypsarrhythmia-infantile spasms. We have
typical progressive elevation of the arms. Eyelid involvement is observed a form of epilepsy intermediate between the two that we
rare. The myoclonic absences have an abrupt onset and cessation, describe as late infantile epileptogenic encephalopathy (244).
similar to CHILDHOOD ABSENCE EPILEPSY (CAE). Simple About 10% to 20% of the children with LGS have passed through
absence seizures, generalized tonic–clonic seizures, and drop a period of infantile spasms-hypsarrhythmia before the LGS
attacks may occur. This is now classified as an absence seizure becomes evident (44–46). The transition from infantile spasms to
with special features, the myoclonic absence (235). LGS has been investigated by Olmos-Garcia de Alba et al. (488).
The interictal EEG has a normal background. Generalized Even in LGS, EEG focality may suggest a hidden focal process.
spike waves may occur, and rarely, these discharges may have
focality. The ictal discharge is a 3 Hz spike and wave discharge. Ictal Manifestations
In one study of 14 children (479), chromosomal abnormali- The types of seizure occurring in the Lennox–Gastaut syn-
ties were present in 7, including Angelman syndrome, 12p tri- drome are best divided into the following groups (Table 26.7).
somy, and an inversion/duplication of chromosome 15. Tonic seizures show a variety of manifestations; see Gastaut
and Broughton (238) and their subdivision into axial, axorhi-
Lennox–Gastaut Syndrome zomelic, and global tonic seizures. The attacks are short and last
This is a severe, intractable, mixed seizure disorder with tonic, from about 5 to 20 seconds. There is extension of the axial mus-
atonic, and atypical absence as the “core” seizures, although gen- culature with some opisthotonus; moderate flexion of the arms is
eralized tonic–clonic seizures, myoclonic seizures, atypical noted, and this may be followed by extension. Tracheobronchial
absence, and focal seizures do occur. Tonic and atonic seizures hypersecretion occurs with repeated attacks, and a fairly danger-
are referred to as “drop attacks.” Mental retardation is frequent, ous status may evolve (489). Tonic seizures are quite common in
and a slow spike and wave pattern is seen on the EEG. This is a non-REM sleep and often are observed in a routine sleep tracing.
typical example of an “epileptic encephalopathy.” Gibbs et al. Bilateral synchronous fast or moderately fast spike activity of
(77) first noticed the severity and poor prognosis of the seizure about 10 to 25 Hz of medium to high voltage and frontal accen-
disorder in patients with slow spike-wave complexes (called the tuation is the EEG concomitant of these attacks (“runs of rapid
“petit mal variant,” in contrast to the classical 3 to 4 Hz spike- spikes”). Electrodecrements may also occur. A diffuse slow ictal
wave complexes in patients with petit mal absences and the pattern has been mentioned by Gastaut and Broughton (238),
prognostically more favorable “primary generalized epilepsy”). who consider it extremely rare.
A detailed account of the clinical and ictal symptomatology of
these epileptics was given by Lennox (78), and further important EEG
work in this field was done by Gastaut et al. (40). The clinical In the atypical absence seizure, the clinical onset and termina-
features were codified by Dravet in her doctoral thesis. The term tion are less abrupt than in classical absences and may be asso-
Lennox–Gastaut syndrome was introduced by Niedermeyer ciated with marked changes in posture and tone. Atypical
(44,480). The work of Oller-Daurella (481–483) deserves special absence status epilepticus may occur.
mention in this context. EEG findings are crucial in the diagnosis of Lennox–Gastaut
Tonic seizures are a major seizure type in LGS and strict cri- syndrome. The outstanding feature is the slow spike-wave com-
teria for LGS require tonic seizures for the diagnosis (484). plex ranging from 1 to 2.5 Hz. It is more often an interictal rather
Tonic seizures are typically sleep activated and repetitive, than an ictal discharge, is most often seen in a generalized
although long-term monitoring may be needed to discover synchronous pattern, although lateralization is also fairly com-
these, as the tonic component may be subtle. LGS is typically mon. Focal slow spike-wave activity is quite rare. A maximum
resistant to therapy with standard AEDs and alternative thera- over the frontal midline is the rule (Fig. 26.32). This discharge is
520 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 2 6 . 7

Types of Seizure Occurring in Lennox–Gastaut


Syndrome

Incidence in
Lennox–Gastaut
Syndrome
Seizures occurring in other seizure
disorders
Grand mal (generalized tonic–clonic)
Psychomotor (complex partial)
Myoclonus
Focal motor and other focal (elementary
partial)
Adversive
Seizures occurring almost exclusively in
the Lennox–Gastaut syndrome
Tonic
Atonic drop (with tonic and myoclonic
elements)
Clonic (rapidly repetitive clonic
movements)
Hemiclonic

enhanced in non-REM sleep and may become almost continu-


ous. Such an abundance of generalized slow spike-wave activity
in non-REM sleep, however, must be carefully distinguished
from the condition known as electrical status epilepticus during
sleep in children (ESES) syndrome (490,491).
Another important pattern is runs of rapid spikes (Figs. 26.33
and 26.34), which are seen in non-REM sleep only. This pattern
is more common in older children, adolescents, or adults. The
EEG as such (“background EEG”) is often disorganized and
excessively slow, some patients show an unremarkable posterior
alpha rhythm, the basic rhythm. The degree of general slowing
and disorganization usually underscores the severity or
advanced stage of the case.
These children are often overmedicated with anticonvulsants, Figure 26.32 A: Tracing of an 8-year-old boy with Lennox–Gastaut syn-
resulting adverse effects including somnolence and incoordina- drome (mentally retarded, uncontrolled psychomotor automatisms, and
tion. Interestingly, high and toxic levels of phenobarbital may be occasion grand mal). Note the irregular slow (1.5 to 2.5 Hz) spike-wave
completely unassociated with fast EEG activity; this absence of complexes. B: Pronounced slow (about 1.5 Hz) spike-wave complexes in
barbiturate-induced fast frequencies is quite characteristic in generalized synchrony with superior frontal maximum in an 11-year-old
advanced cases with considerable cerebral impairment (492). patient with Lennox–Gastaut syndrome. The patient was awake at the
The adult patient may gradually lose the characteristics of the time of this recording; there were no clinical ictal manifestations.
Lennox–Gastaut syndrome, as the slow spike-wave complex may
disappear after age 20 or 15, replaced by only runs of rapid spikes
in sleep. This pattern, too, may gradually vanish and spikes or (495) secondary temporalization is a very common development
sharp waves of temporal and especially anterotemporal localiza- (78% of their patients).
tion may then become predominant. This change may be associ- Clinical entities need revisiting from time to time. Refreshing
ated with the development of complex partial seizures of a new thoughts have been presented by Ohtahara et al. (496) who
temporal origin. This process has been described as “secondary have reemphasized the wide borderland of this syndrome, the
temporalization” (45,493,494). According to Hughes and Patil role of focal cortical elements and secondary bilateral synchrony.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 521

Figure 26.34 A tonic seizure in light sleep in a 20-year-old patient


with Lennox–Gastaut syndrome. A generalized run of rapid spikes is
preceded by a few large slow waves mixed with spikes.

mild mental retardation. The seizures consist of generalized


tonic–clonic and absence seizures. The unique EEG manifesta-
tions during non-REM sleep are coupled with rather bland EEG
Figure 26.33 A run of rapid spikes in a 14-year-old patient. findings in the waking state and REM sleep; occasional focal or
bilaterally synchronous spikes may be present while awake. The
dramatic changes in non-REM sleep disappear during adoles-
These authors therefore advocate a more refined subclassifica- cence. From the viewpoint of the sleep researcher, it is interesting
tion, especially in view of the suitability of neurosurgical treat- to note that these children awaken from such a severely disturbed
ment (callosotomy or vagus nerve stimulator). nocturnal sleep pattern in a normally refreshed state. Although
The highly critical approach of Hirt (497) attempts to cast also referred to as continuous spike-waves of slow sleep, this is
doubt on practically all of the electroclinical diagnostic pillars not absolutely correct, in that the discharges are activated in
of the diagnosis of Lennox–Gastaut syndrome. Such negativis- non-REM sleep, and typically at the onset of drowsiness.
tic analyses could be done in most clinical diagnostic entities; This disorder is rare, occurring in 0.2% of pediatric epilepsy
with sufficient destructive nihilism, the “gestalt” of diseases and syndromes (499) and is divided into symptomatic and crypto-
syndromes could be turned into an empty and worthless con- genic groups, determined by whether normal neurologic or psy-
struct. Criticism is always welcome but has to be coupled with chomotor development was present before the onset of the
a constructive attitude. CSWS. These children commonly have seizures, but the hallmark
is regression. Tassinari reported 29 children with CSWS. All except
Electrical Status Epilepticus during Sleep one child had seizures; one had a single seizure and one had only
(ESES Syndrome) three seizures. Eighteen had normal, and 11 had abnormal psy-
This syndrome is characterized by continuous slow spike-wave chomotor development prior to onset. In the 18 with normal
activity during non-REM sleep. The spike-wave complexes are development, all had severe loss of IQ and behavioral distur-
not well formed, and the spike component is usually more pro- bances, defined as decreased attention span, hyperactivity, aggres-
nounced than the slow wave component. The maximum is vari- sion, difficulties with interaction and inhibition, and 2 patients
able; a maximum over the vertex but also over the occipital developed a psychotic state. In the 11 with abnormal psychomo-
region may be present, according to personal observations. tor development, mental deterioration occurred in all, 3 devel-
This electroclinical condition has been described by Patry et al. oped a marked hyperactivity, and 1 showed “massive regression”
(490) and Tassinari et al. (491,498). ESES was originally defined as including language and a loss of interest in all activities.
having spike and wave discharges present during 85% of slow The regression occurs in a more generalized manner, predom-
wave sleep. This disorder occurs in children (mostly around age inantly with cognitive and behavioral manifestations, rather than
8 years) with chiefly nocturnal but also diurnal seizures and predominantly the language regression seen in Landau–Kleffner
522 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

syndrome (LKS). Guilhoto and Morrell (500) reported that a may differentiate an expressive from a receptive aphasia, and
more focal EEG pattern is associated with language regression, localize the superior temporal gyrus (503), but is not widely
whereas if the EEG pattern is more generalized, the regression is available. All children should have intensive speech therapy
more global, and not predominantly in language skills. and in our experience, even if the EEG activity is controlled,
There has been a recent upsurge of interest in ESES and (LKS). there is a persistent language disorder.
New waves of research in this domain have resulted in a book
dealing with both conditions that was edited by Beaumanoir et al. CHILDHOOD ABSENCE EPILEPSY (CAE)
(1995). There have been advocates of a unified syndrome com-
prising both conditions (501) and also opponents of this idea. Absence seizures were discussed previously under seizure types.
Beaumanoir (1996) as well as Tassinari (1996) have expressed a This section shall refer more to the syndrome of CAE rather than
cautious view aiming at preservation of the status quo. absence seizures. For syndrome classification, historically there are
two different forms of absence seizures: (i) simple absence
LANDAU–KLEFFNER SYNDROME seizures, starting at age 4 or shortly thereafter, with a large num-
ber of absences per day (sometimes exceeding 100/day); and
(ACQUIRED EPILEPTIC APHASIA)
(ii) juvenile absence seizures with an onset at age 9 to 15 years,
Landau–Kleffner syndrome (LKS) (502) is also a rare epileptic more prolonged or mixed with more motor activity. The absences
syndrome, occurring in 0.2% of pediatric epilepsies (499) and of the first type have been called “pycnoleptic” (237,315,504),
is characterized by language regression and an abnormal EEG. using an old term introduced by Sauer (504), which characterizes
LKS usually develops in a previously normal child older than 4 the high frequency of the attacks. The absences with onset around
years and may first manifest as an apparent word deafness, a age 10 to 15 have been called “spanioleptic” (“spanios,” meaning
“verbal auditory agnosia.” Seizures and behavior disturbances, seldom, because of their smaller number of daily seizures) (397).
particularly hyperactivity, each occur in approximately 2/3 of A cyclic change of the frequency has also been noted (“cycloleptic
the children. The majority are classified as idiopathic, although attacks”) (237,315). This distinction (pycnoleptic vs. spanioleptic
LKS may be caused by any pathologic process affecting auditory absences) has been doubted by Loiseau et al. (506). In a study of
cortex. We have seen “symptomatic” cases, one with a left tem- various absence syndromes, there were no syndrome-specific fea-
poral lobe tumor and another with a left sided middle cranial tures to absence seizures (507). The absence seizures were shorter
fossa arachnoid cyst. Therefore, neuroimaging is needed. in Juvenile Myoclonic Epilepsy (JME) and there was greater
The term LKS variant refers to children without the classic awareness.
features. These include children with involvement of more ante- Children with childhood absences often start having gener-
rior language areas with dysfunction characterized by an expres- alized tonic–clonic seizures in early adolescence, with the inci-
sive disorder with oral-motor apraxia, sialorrhea, seizures, and dence ranging from 31% (508) to 54% (509), to Matthes (315),
an abnormal EEG (centro-temporal spikes similar to those seen who reports that about two thirds of the children with petit mal
with benign focal epilepsy), children with PDD (autism) with absences will have generalized tonic–clonic seizures. In most of
language regression and abnormal EEGs, and even children with these cases, the generalized tonic–clonic seizures do not pose a
congenital aphasias, also called developmental language disor- major problem and are readily brought under control. Janz
ders, with epileptiform EEGs. In our experience, children with (510,511) has stressed the role of the stage of awakening as a
PDD comprise the largest group of the “variants.” strongly facilitating factor in these major convulsions. This view
The EEG in LKS shows bilateral, multifocal spikes, and spike has shed much light on the pathophysiologic basis of these
and wave discharges, occurring usually in the posterior regions, attacks and deserves full support.
especially the temporal region, with a marked activation during In a recent study of childhood absence seizures, using strict
sleep. However, discharges occur in many locations, and may classification criteria, only 5/47 (11%) would be classified as
even be generalized. Some centers require ESES to diagnose CAE (269).
LKS, but the strict ESES definition would exclude many cases. Regarding the EEG findings, the organization and regularity
However, sleep-activated epileptiform discharges are required of spike-wave discharges are greater (less fragmented) in CAE,
for the diagnosis. The EEG may improve over time, either spon- than in other absence syndromes (253,254). There is a tendency
taneously or with treatment and the EEG abnormalities may be for the spike-wave frequency and morphology to vary with
an epiphenomenon. absence syndromes: 3 Hz with CAE, faster spikes with Juvenile
The evaluation of ESES or LKS includes a baseline history, Absence Epilepsy (JAE), and polyspikes with JME (512).
physical examination, and sleep-deprived EEG, a formal neu- CAE has been associated with GABARA1 and GABARG2
ropsychologic evaluation, and neuroimaging, with MRI pre- (5q34), chloride channel 2 (3q26), and the voltage-dependent
ferred. Although long-term video-EEG monitoring (LTM) has T-type alpha1H subunit of the calcium channel (16p13).
been required, the presence of strict ESES in an overnight EEG
may be predicted by the abundance of sleep-activated epilep- EYELID MYOCLONIA WITH ABSENCES
tiform features with drowsiness. In certain cases, spike map- (JEAVONS SYNDROME)
ping or functional neuroimaging with either SPECT or PET
scan, and magnetoencephalography (MEG) might be helpful, This seizure type is now classified as an absence seizure with
especially in the child with medically refractory LKS. The special features, eyelid myoclonia (235). The eyelid myoclonia
frequency-modulated auditory evoked response (FM-AER) and absence seizures may occur independently. Eyelid myoclo-
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 523

nia typically occurs immediately after eye closure, is associated Delgado-Escueta et al. (1984) reported that GTC seizures
with brief bilateral spike and wave activity, is more frequent in occurred in 41/43, and Asconape and Penry (518) reported GTC
bright light and does not occur in the dark. Eyelid myoclonia seizures in 83%. Myoclonic jerks usually precede the GTC and
looks like rapid blinking, with upward deviation. However, may evolve into a GTC seizure. GTC seizures usually occur shortly
eyelid myoclonia occurs in other idiopathic generalized epilep- after awakening. Absence seizures occurred in 40% in the series of
sies, especially with photosensitive generalized epilepsy, and Delgado-Escueta et al. (1984). These usually occur in association
have occurred in other family members without seizures, with GTC seizures, and most commonly occur shortly after awak-
referred to as paroxysmal eye movements (513). The frequency ening. Postmyoclonic inhibition may occur (520).
may help to differentiate these: the eyelid flutter with an absence The interictal EEG in JME is characterized by a fast spike
seizure has a frequency of 3 Hz, eyelid myoclonia has a fre- and wave discharge: 3.5 to 6 Hz spike and wave and polyspike
quency of 4 to 6 Hz, and paroxysmal eyelid movements have a and wave complexes. However, patients may have spike-wave
frequency of 10 Hz (513). discharges less than 3.5 Hz (521). Ten to sixteen hertz rapid
spikes with slow waves occur with myoclonic seizures, photo-
ADOLESCENCE sensitivity is common, and abnormalities may occur in sleep.
Although classified as “generalized epilepsy,” focal EEG abnor-
Juvenile Absence Epilepsy (JAE) malities have been reported (522,523); in one study, focal
The onset of JAE onset is usually around puberty, from 9 to 17 spikes, focal sharp waves or slow waves occurred in 37% of
years of age, but in contrast to the very frequent seizures in EEGs (524), and in another 12/85 had focal EEG findings,
CAE, the absence seizures in JAE may be less frequent but more 12/85 had focal clinical findings, and only 2/85 had both clini-
prolonged and have more associated motor activity. GTC and cal and EEG focality (Asconape, 1984). The interictal EEG has
myoclonic seizures may also occur. There are few outcome been normal in 6% (521) or 10% and sleep deprivation may
studies of JAE. In a larger study of 64 patients, only 24/64 (37%) activate epileptiform activity and the discharges may have a
were seizure free after a mean follow-up of 22 years; in a smaller longer duration (525).
study, only 8/17 (44%) were seizure free after a mean follow-up JME, similar to JAE, may require lifelong treatment. Twelve
of 6 years (range 2 to 12 years) (514). Both studies found that out of thirteen patients who discontinued valproate after seizure
those with GTC seizures had less chance of remission. freedom had recurrence (517). In 24 patients followed for 25
The EEG background is normal in JAE. The spike-wave dis- years, only 175 were seizure free and 13% had myoclonus after
charge is usually faster than 3 Hz (3 to 4 Hz), and similar to stopping AEDs (526). In addition, children with absence epilepsy
CAE, may be faster, 4 to 5 Hz, at the onset. The spike-wave dis- without remission (527) and patients with JAE (277) may evolve
charge is less organized (more fragmented) than in CAE, but into JME.
has greater organization (less fragmented) than JME (253,254). The calcium channel, voltage-dependent beta-4 subunit
Focal features have been reported (515). JAE has been associ- (2q24), chloride channel 2 (3q26), GABA1RA (5q34), GABAR
ated with a defect in the chloride channel 2 (3q26) and the EF- delta (1p36), and the EF-hand domain (C-terminal)-containing
hand domain (C-terminal)-containing 1 (6p12-11). 1 (6p12-11) have been associated with JME.

Juvenile Myoclonic Epilepsy (JME) Epilepsy w ith GTC Seizures upon Awakening
JME of Janz is characterized by mild myoclonic, generalized In this disorder, GTC seizures occur exclusively or predomi-
tonic–clonic, sometimes clonic–tonic–clonic seizures and nantly shortly after awakening. The onset is usually in the sec-
absence seizures and has been referred to as impulsive petit mal. ond decade. Absence or myoclonic seizures may occur. The
Onset is typically around puberty. The myoclonic seizures are EEG shows the fast spike and wave burst, similar to JME. These
usually mild-to-moderate in intensity, may involve the entire bursts are relatively short and dominated by 4 Hz or 4 to 5 Hz
extremity, rather than isolated muscles, and generally are bilat- spike-wave complexes that are, contrary to the classical 3 Hz or
eral. These myoclonic seizures typically occur after awakening 3 to 4 Hz spike waves, not readily activated by hyperventila-
and are aggravated by fatigue, sleep deprivation, or alcohol. The tion. Many of these have photosensitivity (photoconvulsive
patient may drop objects or fall. Myoclonus may also be associ- response), and positive family histories of epilepsy are often
ated with brief petit mal absences and myoclonic status epilep- obtained in this group. This syndrome may represent JME
ticus has occurred. GTC seizures are frequent. without myoclonus.
Lennox (279) conceived of this combination (“myoclonic petit
mal”) as a part of his “petit mal triad.” Janz (237) has extensively CHILDREN WITH MULTIFOCAL SPIKES
investigated this type of attack and uses the term impulsive petit (MULTIFOCAL INDEPENDENT SPIKE
mal; Matthes (315) prefers the term Janz syndrome. An extensive SYNDROME, MISS)
study on myoclonic petit mal in 399 patients with genetic evalua-
tion has been carried out by Tsuboi (516). Delgado-Escueta and Multiple independent spikes are defined as three or more inde-
Enrile-Bascal (517) first stressed the good response to sodium val- pendent spike foci, from noncontiguous electrodes, with at least
proate and hence the ultimately good prognosis, which contrasts one focus originating in each hemisphere and the spikes may
with the experience prior to valproate. This is similar to the find- vary from different locations over time. First described by
ings of Asconape and Penry (518). Further information is found Noriega-Sanchez and Markand (1977) in 108 patients, prior
in the work of Schmitz and Sander (519). EEGs could show a hypsarrhythmia or slow spike and wave
524 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

pattern, and the most common age range was 4 to 7 years (40%). seizure documentation, that is, ictal rather than interictal pat-
Blume reported multiple independent spikes, from noncontigu- terns. The interictal spike discharges, however, must not be
ous electrodes, in 63/1500 (4.2%) of EEGs (528). This disorder underrated as an important indicator of paroxysmal activity
was termed severe epilepsy with multiple independent spike foci (21,536). In the epilepsy surgery evaluation, interictal spikes
by Ohtahara (529), with seizures consisting predominantly of define the irritative zone.
tonic spasms. Kotagal (530) supported this as a distinct electro-
clinical syndrome. The MISS may occur following infantile USE OF ACTIVATION METHODS
spasms or LGS. Background slowing and sleep activated dis-
charges are common, and similar to infantile spasms, increased Various activation methods have been used to activate otherwise
synchrony may occur during sleep. undetectable epileptiform activity, such as hyperventilation and
Some children with rather dramatic EEG findings show sur- intermittent photic stimulation. In most major North American
prisingly normal neurologic and mental findings. In the latter EEG laboratories, sleep, either natural or with sedation, is a stan-
group, the widespread generation of spike activity is likely to be dard part of the recording. Sleep deprivation further facilitates
due to temporary paroxysmal hyperexcitability. Genetic predis- epileptiform features, is a true activation, and avoids concerns
position has been suggested by Hauser et al. (531). Diffuse EEG regarding conscious sedation. A whole list of procedures can be
background slowing differentiates these benign epilepsies from put together; all of them aim at the demonstrability of otherwise
the pure MISS. hidden epileptic discharges (Table 26.8). The timing of the
recording may be important. Focal as well as generalized spiking
PRAGMATIC USE OF THE EEG IN DIAGNOSIS may activate shortly after a seizure (537–539), but postictal sup-
pression or attenuation of a focus may also occur. A very useful
Clinical evaluation of a presumed epileptic individual without review of the practical value of EEG studies in epileptic patients
the use of the EEG is almost unthinkable. As a function- has been presented by Engel (534).
oriented test, the EEG is ideally suited for the demonstration of Eye opening and closing will activate certain forms of visu-
epileptiform abnormalities. ally induced epilepsies, especially eye closure. Hyperventilation
Epileptic seizures are necessarily accompanied by an electro-
graphic seizure, which is a transient hypersynchronous neu-
ronal discharge consisting of repetitive epileptiform discharges Tabl e 2 6 . 8
that have an evolution in frequency, amplitude, and morphol- EEG Activation Methods for Enhancement of
ogy in addition to a temporal and spatial evolution. Single Paroxysmal Discharges
spikes or sharp waves do not constitute an electrographic
seizure. However, these may be undetectable on the scalp A. First-line methods
because the generator is buried in a cortical sulcus or in deep
limbic or subcortical regions. A limitation of the ictal firing to Eye opening and closure
lamina V may be responsible for a lack of spikes on the cortical Hyperventilation
surface (532). Without concomitant activation of larger cortical Intermittent photic stimulation (flicker)
areas, deep discharges may not make it to the surface, where the Sleep (short diagnostic non-REM sleep)
recording electrodes are routinely placed. Invasive EEG record- Cautious reduction of anticonvulsive medication (timing
ings must be restricted to only those who are candidates for depending on type of medication and its metabolism)
seizure surgery, and even certain local epileptic events may not Sleep deprivation
be detected. Invasive monitoring is myopic: the recording is Timing of EEG recording to menstrual cycle
from a very limited area. (premenstrual hydration)
Alternatively, epileptiform discharges may also occur in a B. Second-line methods (mostly historical)* indicates historical
substantial number of patients without epilepsy (221,533). Convulsants*
Hence, epileptic discharges may be either too concealed or too Pentylenetetrazol (Metrazol) i.v.
readily available, enticing the electroencephalographer to Bemegride (Megimide) i.v.
imprudent interpretations. It must be kept in mind, however, Combined flicker and pentylenetetrazol i.v.*
that epileptiform discharges in nonepileptic patients are false- Methohexital (Brevital) i.v.
positive findings in a very limited sense only; they are not Use of special trigger mechanisms (cutaneous stimuli,
meaningless. Spikes in nonepileptics clearly indicate a certain viewing geometrical structures, visual exploration,
predisposition to an epileptic seizure or may foreshadow a startling stimuli)
future seizure disorder. The diagnosis of an epileptic seizure Induced hypoglycemia (insulin, fasting)*
disorder must never be made based solely on EEG spikes alone. Hypoxia (breathing nitrogen)*
Useful practical guides for routine studies in epilepsy have been Alpha-chloralose*
presented by Engel (534) and Dinner and Lüders (535).
In the course of the development of epilepsy monitoring C. Use of special technology for greater length of recording
units, the clinical value of interictal spikes has been belittled by Long-term monitoring
most investigators, who, in these special units, are hunting for
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 525

is the classical activation for childhood absences and the 3 Hz but the fatigue; it is important that the patient not be allowed
spike-wave discharge; it is much less effective in other epileptic to falling asleep in the laboratory. An impressive bulk of literature
seizure disorders but is capable of inducing practically every on sleep deprivation has accumulated during the past 15 years.
type of seizure or EEG discharge. Even in some cases of tempo- More information is found in the work of Binnie et al. (543),
ral lobe epilepsy, hyperventilation may be remarkably informa- Degen and Degen (544), and Ellingson et al. (545), to name just
tive. Children below the age of 3.5 years usually do not a few of the numerous investigators in this field.
cooperate with this test; some older children enjoy it. The yield Bilateral synchronous spikes, polyspikes, and spike-wave
of intermittent photic stimulation is enormous, although lim- complexes are usually enhanced in light non-REM sleep
ited to a rather small domain of epileptology, but it may induce (mainly stage 2) and may be found in conjunction with K com-
epileptiform responses in nonepileptics. plexes. The role of arousal has been pointed out in detail by
Sleep records made in periods of deep drowsiness, stage 1 and Niedermeyer (45,72,414,546–548), who introduced the term
non-REM sleep and stage 2, usually 10 to 30 minutes, are an dyshormia, which means faulty or deviant arousal. It is thought
important part of the EEG evaluation. In generalized as well as that arousing stimuli generate K complexes contaminated with
in focal (partial) epilepsies, the epileptiform discharges may be spikes. These K complexes are maximal over the frontal midline
limited to light sleep. The informative value of short sleep trac- (Fz electrode) and thus differ from the vast majority of K com-
ings with or without sedation has been emphasized by Gastaut plexes that have a vertex (C2) maximum (Fig. 26.35). A
et al. (540,541) and Niedermeyer and Hšller (542). Long-term schematic view is shown in Figure 26.36. These data and the
EEG monitoring has demonstrated the increase of spike activity resulting concept have been confirmed by the work of Halász
during nocturnal sleep (Martins da Silva et al., 1984). and his coworkers (61,549–553). Nocturnal (all night) sleep
Sleep deprivation works differently; the important element recordings also demonstrate an increase of generalized paroxys-
in facilitation of epileptiform discharges is not the sleep itself, mal discharges in light non-REM sleep (554,555). Massive

Figure 26.35 A: Spike-wave discharges with frequently intermixed polyspikes in a 30-year-old patient with unusually late
onset of petit mal absences at age 18. Stage 2, non-rapid eye movement (non-REM) sleep. Marked frontal maximum.
B: Same patient, stage 2, non-REM sleep. Transversal montages clearly demonstrate a voltage peak (based on phase rever-
sal) of spike waves and polyspikes over frontal and central midline, but when Fz and Cz are run against each other (chan-
nels 15 to 16), it becomes clear that the frontal midline is the maximally involved area.
526 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 26.35 (Continued) C: Spikes and polyspikes riding on K com-


plexes in stage 2, non-REM sleep in an 18-year-old patient. Well-defined
maximum of spiking over frontal midline (Fz ). (From Niedermeyer E.
Generalisierte epilepsien. EEG Labor. 1979;1:119–131.)

arousal leading to immediate awakening usually does not pro-


duce epileptic K complexes; more subtle arousing stimuli are Figure 26.36 Schematic view of the paroxysmal arousal response over
more effective. Age may be a factor: the sharp component of the frontal midline and the physiologic arousal responses in sleep over ver-
K complex is particularly prominent after the age of 4 years and tex (K complex, vertex waves). Individual predisposition for paroxys-
starts to decline during the second decade. Such naturally sharp mal responses might be based on a local biochemical defect. (From
K complexes apparently facilitate paroxysmal responses. Niedermeyer E. Generalisierte Epilepsien. EEG Labor. 1979;1:119–131.)
There may even be an association of spike bursts and spin-
dles in patients with generalized epilepsies (556). This may be
similar to the aforementioned dyshormia concept. The rela- dow in the patient’s daily EEG activity, but this limited record-
tionship of sleep spindles and spike-wave bursts is bolstered by ing period can be exploited with the crafty use of numerous
the experimental work of Gloor and his coworkers (557–559). channels for better localization and an arsenal of activating
The reduction of long-standing anticonvulsive medication methods. This window can be enlarged by prolonged recording
must be handled with great caution and we typically limit this to in the laboratory over, say, several hours.
inpatients in the Epilepsy Monitoring Unit, since status epilepti- The pragmatist may ask for a numerically precise answer to
cus may be precipitated. Interestingly, interictal focal spike activ- the following questions: “How useful is the EEG in epileptic
ity is not significantly enhanced during the withdrawal phase seizure disorder? How many false-positive or false-negative
despite the enormously heightened risk of grand mal seizures EEG findings occur in the EEG evaluation for presumed
(560). No effect on spike localization and seizure symptomatol- seizures?” Unfortunately, there are no simple answers and this is
ogy was noted with acute withdrawal of antiepileptics (561). controversial. A practice parameter for afebrile seizures in chil-
There are certain mysterious day-to-day changes in the dren recommends an EEG for a single seizure. This has been
degree of a patient’s epileptic activity. It may require a detec- criticized because we do not usually treat a single seizure but it
tive’s acumen to obtain some insight into certain cycles of certainly may give information about the seizure type or
epileptogenicity in a given patient. In epileptic patients with epilepsy syndrome that is helpful in patient management. Table
improvement of seizure frequency after initiation of antiepilep- 26.9 demonstrates the degree of usefulness of EEG findings in
tic medication, the interpretation of repeat tracings requires various forms of epileptic seizure disorder.
particular caution and restraint because the congruence of clin- The value of EEG is determined by intangibles such as the
ical improvement and the amount of paroxysmal EEG activity technical quality of the laboratory technicians and the caliber of
is quite limited (562). the electroencephalographer; on his or her experience and
Repetition of EEG recordings in a patient who stubbornly effort hinge the thoroughness and trustfulness of the EEG
refuses to show the expected paroxysmal discharges will gradu- interpretation. For such obvious reasons, the reader will look in
ally lead to success, but this philosophy is neither elegant nor vain for easy answers; there are none. There are no shortcuts on
economical. The EEG obtained in the laboratory is a small win- the thorny path to expertise in EEG.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 527

Tabl e 2 6 . 9

Usefulness of EEG in Various Epileptic Conditions

Overall
Usefulness False Negatives False Positives Comments
Neonatal convulsions Normal in benign Most valuable in differentiation
forms of severe and benign forms
West syndrome May occur (normal Hypsarrhythmic pattern practically
(infantile spasms) but very high voltage) diagnostic
Febrile convulsions Normal EEG the Abnormal EGG suggestive Value of EEG lies in predominantly
rule! of febrile seizures normal findings
Lennox–Gastaut Almost negligible (Difficult differential A diagnosis grossly based upon
diagnosis) EEG; sleep record helpful
Primary generalized Almost negligible (Seizure-free relatives, Often sleep record needed for
epilepsy (in general) other conditions) EEG evidence, photic stimulation
helpful
Pure petit mal No petit mal absence Extremely reliable correlation
absences without spike waves
Juvenile myoclonic ? Sleep record, photic simulation
epilepsy helpful
Benign rolandic Almost negligible (Seizure free despite Often sleep record needed for
epilepsy predisposition) EEG evidence
Benign occipital lobe
epilepsy
Temporal lobe (Repeat records Sleep record a necessity, EEG
epilepsy often needed) diagnosis may require patience
Frontal lobe epilepsy Difficult EEG diagnosis
Epilepsy of motor 0– May be ? A weak spot in the EEG diagnosis
cortex
Other focal epilepsies 0– May or may not be reliable
Epilepsies due to ? Value of EEG depends on
unusual triggering imaginative use of activation
factors
Alcohol-withdrawal Normal EEG the Abnormal record suggests Value of EEG lies in predominantly
epilepsy rule! (mostly of different genesis of normal findings
low voltage) epilepsy
Status epilepticus / Negligible May indicate severity of status—
convulsive helps differential grand
mal/tonic/myoclonic
Status epilepticus Negligible Excellent in detection of absence
nonconvulsive status, good in detection of
psychomotor status
Psychogenic seizures Normal EEG Abnormal records not EEG useful in spite of its
(pseudoseizures) expected to be the uncommon in patients limitations (Video EEG
rule but there are with psychogenic seizures monitoring may be needed)
many exceptions
528 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

REFERENCES 22. Shewmon DA, Erwin RJ. The effect of focal interictal spikes on
perception and reaction time. I. General considerations.
1. Juul-Jensen P, Foldspang A. Natural history of epileptic seizures. Electroencephalogr Clin Neurophysiol. 1988;69:319–337.
Epilepsy (New York). 1983;24:297–312. 23. Shewmon DA, Erwin RJ. The effect of focal interictal spikes on
2. Gumnit RJ. The Epilepsy Handbook. New York: Raven Press; 1984. perception and reaction time. II. Neuroanatomic specificity.
3. Annegers JF. The epidemiology of epilepsy. In: Wyllie E, ed. The Electroencephalogr Clin Neurophysiol. 1988;69:338–352.
Treatment of Epilepsy. Philadelphia, PA: Lea and Febiger; 1993: 24. Shewmon DA, Erwin RJ. Focal spike-induced cerebral dysfunction
157–164. is related to the after-coming slow wave. Ann Neurol. 1988;23:
4. Greulich W, Gerber U. Epidemiologie der Epilepsien. In: Froescher 131–137.
W, Vassella F, eds. Die Epilepsien. Berlin: De Gruyter; 1994:43–55. 25. Gambardella A, Aguglia U, Oliveri RI, et al. Negative myoclonic
5. Hauser WA. Incidence and prevalence. In: Engel J Jr, Pedley T, eds. status due to antiepileptic drug tapering: report of three cases.
Epilepsy. A Comprehensive Textbook (3 vol.). Vol 1. Philadelphia, Epilepsia. 1997;38:819–823.
PA: Lippincott-Raven; 1998:47–57. 26. Werhahn KJ, Noachtar S. Epileptic negative myoclonus. In: Lueders
6. IFSECN. Preliminary proposal for an EEG terminology by the termi- HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology and Clinical
nology committee of the International Federation for Electroen- Semiology. New York: Churchill Livingstone; 2000:473–483.
cephalography and Clinical Neurophysiology. Electroencephalogr 27. Fisch BJ. Interictal epileptiform activity: diagnostic and behavioral
Clin Neurophysiol. 1961;13:646–650. implications. J Clin Neurophysiol. 2003;20:155–162.
7. IFSECN. Proposal for an EEG terminology committee of the 28. Warach S, Ives JR, Schlaug G, et al. EEG-triggered echo-planar func-
International Federation for Electroencephalography and Clinical tional MRI in epilepsy. Neurology. 1996;47:89–93.
Neurophysiology. Electroencephalogr Clin Neurophysiol. 1966;30: 29. Aarts JHP, Binnie CD, Smit AM, et al. Selective cognitive impair-
306–310. ment during focal and generalized epileptiform EEG activity. Brain.
8. IFSECN. A glossary of terms commonly used by clinical electroen- 1984;107:293–308.
cephalographers. Electroencephalogr Clin Neurophysiol. 1974;37: 30. Hughes JR. The significance of interictal spike discharge: a review.
538–548. Clin Electroencephalogr. 1997;28:60. Abstract.
9. Noachtar S, Binnie C, Ebersole J, et al. A glossary of terms most 31. Jasper HH. Epilepsy. In: Penfield W, Erickson TC, eds. Epilepsy and
commonly used by clinical electroencephalographers and pro- Cerebral Localization. Springfield, IL: Charles C Thomas; 1941:
posal for the report form for the EEG findings. Recommendations 380–454.
for the Practice of Clinical Neurophysiology: guidelines of the 32. Cukier F, André N, Monod N, et al. EEG contribution to diagnosing
International Federation of Clinical Physiology. Electroencephalogr intraventricular haemorrhage in premature infants. Electroen-
Clin Neurophysiol Suppl. 1999;52:21–41. cephalogr Clin Neurophysiol. 1974;36:840. Abstract.
10. Dutertre F. Catalogue of the main EEG patterns. In: Remond A, 33. Werner SS, Stockard JE, Bickford RG. Atlas of Neonatal
ed-in-chief. Handbook of Electroencephalography and Clinical Electroencephalography. New York: Raven Press; 1977.
Neurophysiology. Vol 11A. Amsterdam: Elsevier; 1977:40–79. 34. Zobniw AM, Yaworth S, Niedermeyer E. Depth electroencephalog-
11. Kiloh LG, McComas AJ, Osselton JW. Clinical Electroencephalo- raphy. J Electrophysiol Technol. 1975;2:215–240.
graphy. 3rd ed. London: Butterworth; 1972. 35. Ribeiro M, Niedermyer E, Hertz S. Slow sharp waves. Clin
12. Roger H, Cornil L, Paillas JE. Les Épilepsies. Paris: Flammarion; Electroencephalogr. 1999;30:114–117.
1949. 36. Chatrian GE, Shaw CM, Leffman H. The significance of periodic
13. Gotman J. Could interictal epileptic spikes actually be postictal lateralized epileptiform discharges in EEG: an electrographic, clin-
epileptic spikes? Electroencephalogr Clin Neurophysiol. 1984;58:9P. ical and pathological study. Electroencephalogr Clin Neurophysiol.
Abstract. 1964;17:177–193.
14. Gotman J, Marciani MG. Electroencephalographic spiking activ- 37. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 2. 2nd ed.
ity, drug levels and seizure occurrence in epileptic patients. Ann Cambridge, MA: Addison-Wesley; 1952.
Neurol. 1985;17:597–603. 38. Gibbs FA, Davis H, Lennox WG. The electroencephalogram in
15. Pollen DA, Reid KH, Perol P. Micro-electrode studies of experimen- epilepsy and in conditions of impaired consciousness. Arch Neurol
tal wave and spike in the cat. Electroencephalogr Clin Neurophysiol. Psychiatry (Chicago). 1935;34:1133–1148.
1964;17:57–67. 39. Jasper HH, Kershman J. Classification of the EEG in epilepsy.
16. Matsuo F, Knott JR. Focal positive spikes in electroencephalogra- Electroencephalogr Clin Neurophysiol. 1949;2(suppl 2):123–131.
phy. Electroencephalogr Clin Neurophysiol. 1977;42:15–25. 40. Gastaut H, Roger J, Soulayrol R, et al. Childhood epileptic
17. Maulsby RL. Some guidelines for assessment of spikes and sharp encephalopathy with diffuse slow spike-waves (otherwise known as
waves in EEG tracings. Am J EEG Technol. 1971;11:3–16. “petit mal variant”) or Lennox syndrome. Epilepsia (Amsterdam).
18. Binnie CD, Polkey CE, Spencer S. Positive spikes over a cortical lacer- 1966;7:139–179.
ation. Electroencephalogr Clin Neurophysiol. 1989;72:106P. Abstract. 41. Brenner RP, Atkinson R. Generalized paroxysmal fast activity: elec-
19. Gregory DL, Wong PK. Topographical analysis of the centrotempo- troencephalographic and clinical features. Ann Neurol. 1982;11:
ral discharges in benign rolandic epilepsy of childhood. Epilepsia 386–390.
(New York). 1984;25:705–711. 42. Chayasirisobhon S, Cullis P, Sack R, et al. Grand mal discharge.
20. Wong PKH, Gregory D. Dipole fields in rolandic discharges. Am J Clin Electroencephalogr. 1984;15:155–158.
EEG Technol. 1988;28:243–250. 43. Rodin E, Smid N, Mason K. The grand mal pattern of Gibbs,
21. Niedermeyer E. Clinical relevance of EEG signals in epilepsies. In: Gibbs and Lennox. Electroencephalogr Clin Neurophysiol. 1976;40:
Wada JA, Ellingson RJ, eds. Handbook of Electroencephalography 401–406.
and Clinical Neurophysiology. Revised series, Vol 4. Amsterdam: 44. Niedermeyer E. The Lennox–Gastaut syndrome: a severe type of
Elsevier; 1990:237–261. childhood epilepsy. Dtsch Z Nervenheilk. 1969;195:263–282.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 529

45. Niedermeyer E. The Generalized Epilepsies. Springfield, IL: Charles 68. Baddeley AD. Working Memory. Oxford: Oxford University Press;
C Thomas; 1972. 1986.
46. Niedermeyer E. Compendium of the Epilepsies. Springfield, IL: 69. Fuster JM. Memory and planning. In: Jasper HH, Riggio S, Patricia
Charles C Thomas; 1974. Goldman-Rakic PS, eds. Epilepsy and the Functional Anatomy of
47. Niedermeyer E, Walker AE, Burton C. The slow spike-wave com- the Frontal Lobe. New York: Raven Press; 1995:9–19.
plex as a correlate of frontal and fronto-temporal posttraumatic 70. Fuster JM. Memory in the Cerebral Cortex. Cambridge, MA: MIT
epilepsy. Eur Neurol (Basel). 1970;3:330–346. Press; 1996.
48. Niedermeyer E. Widersprüchlich beurteilte EEG-Tätigkeiten und 71. Niedermeyer E. Frontal lobe functions and dysfunctions.
ihre Bedeutung. EEG-Labor. 1988a;10:3–7. Presented at Symposium; 1997; Lima (Peru).
49. Niedermeyer E. The electroencephalogram in the differential diag- 72. Niedermeyer E. Primary (idiopathic) generalized epilepsy. Clin
nosis of the Lennox–Gastaut syndrome. In: Niedermeyer E, Degen Electroencephalogr. 1996;27:1–21.
R, eds. The Lennox–Gastaut Syndrome. New York: Alan R. Liss; 1988: 73. Walker A, Morello G. Experimental petit mal. Trans Am Neurol
177–220. Assoc. 1967;97:57–61.
50. Arroyo S, Lesser RP, Fisher RS, et al. Clinical and electroencephalo- 74. Hunter J, Jasper HH. Effects of thalamic stimulation in unanes-
graphic evidence for sites of origin of seizures with diffuse elec- thetized animals. Electroencephalogr Clin Neurophysiol. 1949;1:
trodecremental pattern. Epilepsia. 1994;35:974–987. 303–324.
51. RamachandraNair R, Ochi A, Imai K, et al. Epileptic spasms in 75. Sperling MR, Skolnick BE. Cerebral blood flow during spikewave
older pediatric patients: MEG and ictal high frequency oscillations discharges. Epilepsia. 1995;36:156–163.
suggest focal onset seizures in a subset of epileptic spasms. Epilepsy 76. Gianelli M, Cantello R, Civardi C, et al. Idiopathic generalized
Research. 2008;78:216–224. epilepsy: magnetic stimulation of motor cortex time-locked and
52. Silverman D. Clinical correlates of the spike-wave complexes. unlocked to 3-Hz spike-and-wave discharges. Epilepsia. 1994;35:
Electroencephalogr Clin Neurophysiol. 1954;6:663–669. 53–60.
53. Clark EC, Knott JR. Paroxysmal wave and spike activity and diag- 77. Gibbs FA, Gibbs EL, Lennox WG. The influence of the blood sugar
nostic subclassification. Electroencephalogr Clin Neurophysiol. 1955; level on the wave and spike formation in petit mal epilepsy. Arch
7:161–164. Neurol Psychiatry (Chicago). 1939;47:1111–1116.
54. Lundervold A, Henriksen GF, Fegersten L. The spike wave com- 78. Lennox WG. Epilepsy and Related Disorders. Boston, MA: Little,
plex. A clinical correlation. Electroencephalogr Clin Neurophysiol. Brown; 1960.
1959;11:13–22. 79. Gastaut H. Clinical and electroencephalographic correlates of gen-
55. Gastaut H, Hunter J. An experimental study of the mechanism of eralized spike and wave bursts occurring spontaneously in man.
photic activation in idiopathic epilepsy. Electroencephalogr Clin Epilepsia (Amsterdam). 1968;9:179–184.
Neurophysiol. 1950;2:263–287. 80. Silverman D. Phantom spike-wave and the fourteen and six per
56. Cohn R. Spike-dome complex in the human electroencephalo- second positive spike pattern: a consideration of their relationship.
gram. Arch Neurol Psychiatry (Chicago). 1954;71:699–706. Electroencephalogr Clin Neurophysiol. 1967;23:203–217.
57. Weir B. The morphology of the spike-wave complex. Electroen- 81. Marshall C. Some clinical correlates of the wave and spike phan-
cephalogr Clin Neurophysiol. 1965;19:284–290. tom. Electroencephalogr Clin Neurophysiol. 1955;7:633–636.
58. Jung R, Toennies F. Über Entstehung and Erhaltung von 82. Thomas JE. A rare electroencephalographic pattern: the six-per
Krampfentladungen. Die Vorgänge im Reizort und die Krampf- second spike and wave discharge. Neurology (Minneapolis). 1957;7:
fähigkeit des Gehirns. Arch Psychiatr Nervenkr. 1950;1985:701–735. 438–442.
59. Pollen DA. Intracellular studies of cortical neurons during thala- 83. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 3.
mic induced wave and spike. Electroencephalogr Clin Neurophysiol. Reading, MA: Addison-Wesley; 1964.
1964;17:398–406. 84. Hughes JR, Schlagenhauff RE, Magoss M. Electro-clinical correla-
60. Weir B, Sie PG. Extracellular unit activity in cat during the spike- tions in the six per second spike and wave complex. Electroen-
wave complex. Epilepsia (Amsterdam). 1966;7:30–43. cephalogr Clin Neurophysiol. 1965;18:71–77.
61. Halász P. The generalized epileptic spike-wave mechanism and the 85. Thomas JE, Klass DW. Six-per-second spike and wave pattern in
sleep-wakefulness system. Acta Physiol Acad Scient Hung. 1972; the electroencephalogram: a reappraisal of clinical significance.
42:293–314. Neurology (Minneapolis). 1968;18:587–593.
62. Niedermeyer E. Immediate transition from a petit mal absence 86. Small JG. Small sharp spikes in a psychiatric population. Arch Gen
into a grand mal seizure. Eur Neurol (Basel). 1976;14:11–16. Psychiatry. 1970;22:277–284.
63. Dondey M. Transverse topographical analysis of petit mal dis- 87. Hecker A, Kocher R, Ladewig D, et al. Das Minatur-Spike-Wave-
charges: diagnostical and pathogenic implications. Electroen- Muster. Das EEG Labor. 1979;1:51–56.
cephalogr Clin Neurophysiol. 1983;55:361–371. 88. Hughes JR. Two forms of the 6/sec spike and wave complex.
64. Rodin E, Ancheta O. Cerebral electrical fields during petit mal Electroencephalogr Clin Neurophysiol. 1980;48:535–550.
absences. Electroencephalogr Clin Neurophysiol. 1987;66:457–466. 89. Hughes JR. Changes in the annual incidence of the 6/sec spike and
65. Gotman J. Interhemispheric relations during bilateral spike-and- wave pattern. Clin Electroencephalogr. 1991;22:71–74.
wave activity. Epilepsia (New York). 1981;22:453–466. 90. Kocher R, Scollo-Lavizzari G, Ladewig D. Miniature-spikewave-
66. Lemieux JF, Blume WT. Topographical evolution of the spike-wave Muster: Elektroencephalographisches Korrelat in der Abstinenzphase
complexes. Electroencephalogr Clin Neurophysiol. 1983;56:30P. bei Medikamentenabhängigkeit. Z EEG-EMG. 1975;6:78–82.
Abstract. 91. Tharp BR. The 6-per-second spike and wave complex. Arch Neurol
67. Lüders H, Daube J, Johnson R, et al. Computer analysis of gener- (Chicago). 1966;15:533–537.
alized spike-and-wave complexes. Epilepsia (New York). 1980;21: 92. Westmoreland BF. Benign EEG variants and patterns of uncertain
183. Abstract. clinical significance. In: Daly DD, Pedley TA, eds. Current Practice
530 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

of Clinical Electroencephalography. 2nd ed. New York: Raven 115. Gibbs FA, Gibbs EL. Fourteen and six per second positive spikes.
Press; 1990:243–252. Electroencephalogr Clin Neurophysiol. 1963;15:353–358.
93. Klass DW, Westmoreland BF. Nonepileptogenic epileptiform 116. Henry EE. Positive spike discharges in the EEG and behavior
electroencephalographic activity. Ann Neurol. 1985;18:627–635. abnormality. In: Glaser GE, ed. EEG and Behavior. New York:
94. Niedermeyer E. Controversial EEG patterns and their signifi- Basic Books; 1963:315–344.
cance. Editorial. Am J EEG Technol. 1987;27:129–132. 117. Hughes JR, Gianturco D, Stein W. Electro-clinical correlations in
95. Santoshkumar B, Chong JJR, Blume WT, et al. Prevalence of benign the positive spike phenomenon. Electroencephalogr Clin Neuro-
epileptiform variants. Clin Neurophysiol. 2009;120:856–861. physiol. 1961;13:599.
96. Alvarez N, Lombroso CT, Medina C, et al. Paroxysmal spike and 118. Kellaway P. Neurologic status of patients with hypsarrhythmia.
wave activity in drowsiness in young children: its relationship to In: Gibbs FA, ed. Molecules and Health. Philadelphia, PA: JB
febrile convulsions. Electroencephalogr Clin Neurophysiol. 1983; Lippincott; 1959:134–149.
56:406–413. 119. Millen FJ, White B. Fourteen and six per second positive spike
97. Koshino Y, Niedermeyer E. The clinical significance of small activity in children. Neurology (Minneapolis). 1954;4:541–549.
sharp spikes in the electroencephalogram. Clin Electroencephalogr. 120. Niedermeyer E, Knott JR. Über die Bedeutung der 14 and 6/sec-
1975;6:131–140. positiven Spitzen im EEG. Arch Psychiatr Nervenkr. 1961;202:
98. Westmoreland BF, Reiher J, Klass DW. Recording small sharp 266–280.
spikes with depth electroencephalography. Epilepsia (New York). 121. Poser CM, Ziegler DK. Clinical significance of 14 and 6 per sec-
1979;20:599–612. ond positive spike complexes. Neurology (Minneapolis). 1958;8:
99. Small JG, Small IF, Milstein V, et al. Familial associations with 903–912.
EEG variants in manic depressive disease. Arch Gen Psychiatry. 122. Refsum S, Presthus J, Skulstad AA, et al. Clinical correlates
1975;32:43–48. of the 14 and 6 per second positive spikes. An electroen-
100. Reiher J, Klass DW. Two common EEG patterns of doubtful clin- cephalographic and clinical study. Acta Psychiatr Scand. 1960;35:
ical significance. Med Clin North Am. 1968;52:933–940. 330–344.
101. Lebel M, Reiher J, Klass D. Small sharp spikes (SSS). Reassessment 123. Schwade ED, Geiger SG. Matricide with electroencephalographic
of electroencephalographic and clinical significance. Electroen- evidence of thalamic or hypothalamic disorder. Dis Nerv Syst.
cephalogr Clin Neurophysiol. 1977;43:463. Abstract. 1953;14:18–20.
102. White JC, Langston JW, Pedley TA. Benign epileptiform tran- 124. Shimoda Y. The clinical and electroencephalographic study of the
sients of sleep: clarification of the small sharp spike controversy. primary diencephalic epilepsy or epilepsy of brain stem. Acta
Neurology. 1977;27:1061–1068. Neurovegetat (Vienna). 1961;23:181–191.
103. Gutrecht JA. Clinical implications of benign epileptiform transients 125. Stephenson WA. Intracranial neoplasm associated with fourteen
of sleep. Electroencephalogr Clin Neurophysiol. 1989;72:486–490. and six per second positive spikes. Neurology (Minneapolis). 1951;
104. Low MD, Henrikson K, Lamont D, et al. The relationship of small 1:372–376.
sharp spikes to seizures. Electroencephalogr Clin Neurophysiol. 1984; 126. Walter RD, Colbert EG, Koegler RR, et al. A controlled study of the
58:11P. Abstract. fourteen-and-six-per-second EEG pattern. Arch Gen Psychiatry.
105. Hughes JR, Gruener GT. Small sharp spikes revisited: further 1960;2:559–566.
data on this controversial pattern. Electroencephalogr Clin Neuro- 127. Long MT, Johnson LC. Fourteen and six positive spikes in
physiol. 1984;15:208–213. nonpatient population. Neurology (Minneapolis). 1967;17:316.
106. Saito F, Fukushima Y, Kubota S. Small sharp spikes: possible rela- Abstract.
tionship to epilepsy. Clin Electroencephalogr. 1987;18:144–119. 128. Little SC. A general analysis of the fourteen and six per second
107. Molaie M, Santana HB, Cavanaugh WA. Benign epileptiform dysrhythmia. Proceedings of the Sixth International Congress of
transients of sleep (BETS) revisited. Electroencephalogr Clin Neuro- Electroencephalography and Clinical Neurophysiology. Vienna:
physiol. 1989;72:19P. Abstract. Wiener Med. Akad; 1965:313–315.
108. Gibbs CJ Jr, Gajdusek DC, Asher DM, et al. Creutzfeldt–Jakob 129. Beydoun A, Drury I. Unilateral 14 and 6 Hz positive bursts.
disease (spongiform encephalopathy): transmission to the chim- Electroencephalogr Clin Neurophysiol. 1992;82:310–312.
panzee. Science. 1968;161:388–389. 130. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 4. Menlo
109. Lairy GC, Harrison A, Leger EM. Foyers EEG biocciptaux asyn- Park, CA: Addison-Wesley; 1978.
chrones de pointes chez l’enfant mal voyant et aveugle d’âge sco- 131. Okada S, Kato T, Miyashita K, et al. 14 and 6 Hz positive spikes:
laire. Rev Neurol (Paris). 1964;111:351–353. relationship with a negative spiky phase in the frontal area.
110. Cannon WB, Rosenblueth A. The Supersensitivity of Denervated Electroencephalogr Clin Neurophysiol. 1990;75:S108. Abstract.
Structures: A Law of Denervation. New York: Macmillan; 1949. 132. Niedermeyer E, Ray CD, Walker AE. Depth studies in a patient
111. Lombroso CT, Schwarts IH, Clark DM, et al. Ctenoids in healthy with fourteen and six per second positive spikes. Electroen-
youth. Controlled study of 14 and 6-per-second positive spiking. cephalogr Clin Neurophysiol. 1967;22:86–89.
Neurology (Minneapolis). 1966;16:1152–1158. 133. Gibbs FA. An epidemiological note? Clin Electroencephalogr. 1990;
112. Hughes JR. The 14 and 7 per second positive spikes—a reappraisal 21:112–118.
following a frequency count. Electroencephalogr Clin Neurophysiol. 134. Silverman D. Fourteen and six per second positive spike pattern
1960;12:495–496. in a patient with hepatic coma. Electroencephalogr Clin Neuro-
113. Hughes JR. A review of the positive spike phenomenon. In: Wilson physiol. 1964;16:395–398.
WP, ed. Application of Electroencephalography in Psychiatry. 135. Ford RG, Freeman AM. Positive spike bursts in a comatose adult.
Durham: Duke University Press; 1965:54–101. Electroencephalogr Clin Neurophysiol. 1982;53:29P. Abstract.
114. Gibbs EL, Gibbs FA. Electroencephalographic evidence of thala- 136. Drury I. 14- and 6-Hz positive bursts in diverse encephalopathies
mic and hypothalamic epilepsy. Neurology (Minneapolis). 1951;1: of childhood. Electroencephalogr Clin Neurophysiol. 1989;72:12P.
136–144. Abstract.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 531

137. Lipman IL, Hughes JR. Rhythmic mid-temporal discharges. An Children. New York: Grune & Stratton (Stockholm: Almquist and
electroclinical study. Electroencephalogr Clin Neurophysiol. 1969; Wiksell); 1968:275–286.
27:43–47. 160. Lombroso CT. Remarks on the EEG and movement disorder in
138. Egli M, Hess R, Kuritzke G. Die Bedeutung der “rhythmic mid- SSPE. Neurology (Minneapolis). 1968;18:69–75.
temporal discharges.” Z EEG-EMG. 1978;9:74–85. 161. Bogacz J, Castells C, San Julian J, et al. Nonepidemic progressive
139. Hughes JR. The continuous rhythmic mid-temporal discharge. subacute encephalitis (type Van Bogaert). II. Serial EEG abnormal-
Clin Electroencephalogr. 2001;32:10–13. ities and deep electrography. Acta Neurol Lat Am. 1959;5:158–183.
140. Garvin JS. Psychomotor variant pattern. Dis Nerv Syst. 1968;29: 162. Lesse S, Hoefer PFA, Austin JH. The electroencephalogram in dif-
59–76. fuse encephalopathies. Arch Neurol Psychiatry (Chicago).
141. Boutros NN, Hughes JR, Weiler M. Psychiatric correlates of the 1958;79:359–375.
rhythmic mid-temporal discharges and the 6/sec spike and wave 163. Storm van Leeuwen W. Electroencephalographical and neuro-
complexes. Biol Psychiatry. 1986;21:94–99. physiological aspects of subacute sclerosing leuco-encephalitis.
142. Naquet R, Louard C, Rhodes J, et al. À propos de certaines Psychiatr Neurol Neurochir (Amsterdam). 1964;67:312–322.
décharges paroxystiques du carrefour temporo-pariéto-occipital: 164. Martinovic Z. Periodic generalized bursts of fast waves in suba-
leur activation par l’hypoxie. Rev Neurol (Paris). 1961;105:203–207. cute sclerosing panencephalitis. Electroencephalogr Clin Neuro-
143. Herranz F, Lopez S. Subclinical paroxysmal rhythmic activity. physiol. 1986;63:236–238.
Electroencephalogr Clin Neurophysiol. 1984;4:419–442. 165. Radermecker FJ. Systématique et Électroencéphalographie des
144. Westmoreland BF, Klass DW. A distinctive rhythmic EEG discharge Encéphalites et Encéphalopathies. Amsterdam: Elsevier; 1956
of adults. Electroencephalogr Clin Neurophysiol. 1981;51:186–191. (Electroencephalogr Clin Neurophysiol Suppl 5).
145. Westmoreland BF, Klass DW. Unusual variants of subclinical 166. Millar JHD, Coey A. The EEG in necrotizing encephalitis.
rhythmic electrographic discharge of adults (SREDA). Electroen- Electroencephalogr Clin Neurophysiol. 1959;11:582–585.
cephalogr Clin Neurophysiol. 1997;102:1–4. 167. Perier O, Parmentier B, Brihaye J, et al. A case of inclusion body
146. Miller CR, Westmoreland BF, Klass DW. Subclinical rhythmic necrotizing encephalitis. In: Van Bogaert L, et al., eds.
EEG discharge of adults (SREDA): further observations. Am J Encephalitides. Amsterdam: Elsevier; 1961:235–242.
EEG Technol. 1985;25:217–224. 168. Carmon A, Behar A, Beller A. Acute necrotizing encephalitis
147. O’Brien TJ, Sharbrough FW, Westmoreland BF, et al. Subclinical presenting clinically as a space-occupying lesion. A clinico-
rhythmic electrographic discharges of adults (SREDA) revisited: pathological study of six cases. J Neurol Sci. 1965;2:328–343.
a study using digital EEG analysis. J Clin Neurophysiol. 1998l;15: 169. Rawls WE, Dyck PJ, Klass DW, et al. Encephalitis associated with
493–501. herpes simplex virus. Ann Intern Med. 1966;64:104–115.
148. Gaches J. Les activitiés périodiques en EEG. Rev EEG Neuro- 170. Adams JH, Jennett WB. Acute necrotizing encephalitis: a problem
physiol. 1971;1:9–33. in diagnosis. J Neurol Neurosurg Psychiatry. 1967;30:248–260.
149. Bauer G, Pieber R. Über periodische Komplexe im EEG. Z EEG- 171. Upton A, Gumpert J. Electroencephalography in diagnosis of
EMG. 1974;5:75–86. herpes simplex encephalitis. Lancet. 1970;1:650–652.
150. Spehlmann R. EEG Primer. Amsterdam: Elsevier; 1981. 172. Gaches J, Arfel G. Certitude et suspicions d’encéphalites herpé-
151. Rabending G, Radermecker FJ. Subacute sclerosing panen- tiques. Aspects électroencéphalographiques. Encéphale. 1972;61:
cephalitis (SSPE). In: Remond A, ed-in-chief. Handbook of 510–549.
Electroencephalography and Clinical Neurophysiology. Vol. 15A. 173. Elian M. Herpes simplex virus encephalitis and EEG. J
Amsterdam: Elsevier; 1977:28–35. Electrophysiol Technol. 1975;1:161–170.
152. Celesia GG. Pathophysiology of periodic EEG complexes in sub- 174. Kugler J, Marltin JJ, Radermecker RJ, et al. Periodische Komplexe
acute sclerosing panencephalitis (SSPE). Electroencephalogr Clin im EEG bei nekrotisierender Herpes-Enzephalitis. Z EEG-EMG.
Neurophysiol. 1973;35:293–300. 1976;7:63–71.
153. Broughton R, Nelson R, Gloor P, et al. Petit mal epilepsy evolving 175. Cobb WA. Evidence on the periodic mechanism in herpes sim-
to subacute sclerosing panencephalitis. In: Lugaresi E, Pazzaglia P, plex encephalitis. Electroencephalogr Clin Neurophysiol. 1979;46:
Tassinari CA, eds. Evolution and Prognosis of Epilepsies. Bologna: 345–350.
Gaggi; 1973:63–72. 176. Gupta PC, Seth P. Periodic complexes in herpes simplex
154. Gökcil Z, Odabasi Z, Aksu A, et al. Acute fulminant SSPE: clini- encephalitis. A clinical and experimental study. Electroencephalogr
cal and EEG features. Clin Electroencephalogr. 1998;29:43–48. Clin Neurophysiol. 1973;35:67–74.
155. Giovanardi Rossi P, Gobbi G, Moschen R, et al. Subacute scleros- 177. Fineyre R. Periodic discharge of unusually slow character in
ing panencephalitis-like EEG pattern and periodic myoclonic neonatal status epilepticus caused by necrotizing encephalitis
seizures: two case reports. Electroencephalogr Clin Neurophysiol. (herpes simplex). Electroencephalogr Clin Neurophysiol. 1979;46:80.
1982;54:3P. Abstract. Abstract.
156. Prier S, Benoit C, Cambier J. Bilateral periodic stereo-typed EEG 178. Bergey GK, Coyle PK, Krumholz A, et al. Herpes simplex
complexes in multiple sclerosis. Clin Neurophysiol. 1992;22(suppl encephalitis with occipital localization. Arch Neurol (Chicago).
1–1992):107s. Abstract. 1982;39:312–313.
157. Radermecker FJ, Poser CM. The significance of repetitive 179. Creutzfeldt HG. Über eine eigenartige herdförmige Erkrankung
paroxysmal electroencephalographic patterns. Their specificity des Zentralnervensystems. Z Gesamte Neurol Psychiatr. 1920;57:1.
in subacute sclerosing leukoencephalitis. World Neurol. 1960;1: (Quoted after Kirschbaum, W.R., 1968.)
422–433. 180. Jakob A. Über eigenartige Erkrankungen des Zentralnervensys-
158. Cobb WA. The periodic events of subacute sclerosing leucoen- tems mit bemerkenswerten anatomischen Befunden (spastische
cephalitis. Electroencephalogr Clin Neurophysiol. 1966;21:278–294. Pseudosklerose, Encephalomyelopathie mit disseminierten
159. Cobb WA. Depth recording in subacute sclerosing leucoencephali- Degenerationsbeschwerden). Dtsch Z Nervenheilkd. 1921;70:132.
tis. In: Kellaway P, Petersén I, eds. Clinical Electroencephalography of (Quoted after Kirschbaum, W.R., 1968.)
532 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

181. Kirschbaum WR. Zwei eigenartige Erkrankungen des Zentralner - 202. Filley CM, Kelly MA, Hwang PALS, et al. An analysis of typical
vensystems nach Art der spastischen Pseudosklerose (Jakob). Z and “indeterminate” PLEDs. Electroencephalogr Clin Neurophysiol.
Gesamte Neurol Psychiatr. 1924;92:157. (Quoted after Kirschbaum, 1984;58:32P. Abstract.
W.R., 1968.) 203. Aldrich MS, Pugh JE. Cerebral bigeminy: alternating periodic
182. Kirschbaum WR. Jakob–Creutzfeldt’s Disease. New York: American epileptiform discharges. Electroencephalogr Clin Neurophysiol.
Elsevier; 1968. 1984;58:35P. Abstract.
183. Jones DP, Nevin S. Rapidly progressive cerebral degeneration 204. Hughes JR, Taber J, Uppal H. TRI-PLEDs: a case report. Clin
(subacute vascular encephalopathy) with mental disorder, focal Electroencephalogr. 1998;29:106–108.
disturbances and myoclonic epilepsy. J Neurol Neurosurg 205. PeBenito R, Cracco JB. Periodic lateralized epileptiform dis-
Psychiatry. 1954;17:148–159. charges in children. Ann Neurol. 1979;6:47–50.
184. Heidenhain A. Klinische und anatomische Untersuchungen einer 206. Andriola M. PLEDS and bi-PLEDS in children. Electroen -
eignartiger organische Erkrankung des Zentralnervensystems im cephalogr Clin Neurophysiol. 1982;53:88P. Abstract.
Praesenium. Z Gesamte Neurol Psychiatr. 1929;118:49–111. 207. Ritaccio AL, March G. The significance of BIPLEDs in complex
185. Gibbs CJ Jr, Gajdusek DC. Infection as the etiology of spongi- partial status epilepticus. Am J EEG Technol. 1993;33:27–34.
form encephalopathy (Creutzfeldt–Jakob disease). Science. 1969; 208. Silbert PL, Radhakrishnan K, Sharbrough FW, et al. Ipsilateral
165:1023–1025. independent periodic lateralized epileptiform discharges.
186. Niedermeyer E, Gibbs CB Jr, Marsh R, et al. EEG studies in sub- Electroencephalogr Clin Neurophysiol. 1996;98:223–226.
acute and degenerative neurological diseases experimentally pro- 209. Chabolla DR, Moore JL, Westmoreland BF. Periodic lateralized
duced in ferrets and chimpanzees. Electroencephalogr Clin epileptiform discharges in multiple sclerosis. Electroencephalogr
Neurophysiol. 1972;33:351–352. Abstract. Clin Neurophysiol. 1996;98:5–8.
187. Drury I, Beydoun A. Evolution of periodic complexes in 210. Gandelman-Marton R, Rabey JM, Flechter S. Periodic lateralized
Creutzfeldt–Jakob disease. Am J EEG Technol. 1996;36:230–234. epileptiform discharges in multiple sclerosis: a case report. J Clin
188. Tariska P, Gereby G, Majtenyi K. The clinical significance of peri- Neurophysiol. 2003;20:117–121.
odic lateralized epileptiform discharges (PLEDs). Electroen - 211. Pohlmann-Eden B, Hoch DB, Cochius JI, et al. Periodic lateralized
cephalogr Clin Neurophysiol. 1985;61:11P–12P. Abstract. epileptiform discharges—a critical review. J Clin Neurophysiol.
189. Chiappa K, Young R. The EEG as a definitive diagnostic tool early 1996;13:519–530.
in the course of Creutzfeldt–Jakob disease. Electroencephalogr 212. Westmoreland BF, Frere RC, Klass DW. Periodic epileptiform dis-
Clin Neurophysiol. 1978;45:26. Abstract. charges in the midline. J Clin Neurophysiol. 1997;14:495–498.
190. Chiofalo N, Fuentes A, Galvez S. Serial EEG findings in 27 cases of 213. Butenuth J, Kubicki St. Über die prognostische Bedeutung bes-
Creutzfeldt–Jakob disease. Arch Neurol (Chicago). 1980;37:143–145. timmter Formen der Myoklonien und korrespondierender EEG-
191. Poole EW. Jakob–Creutzfeldt disease. Patients who might have Muster nach Hypoxien. Z EEG-EMG. 1971;2:78–83.
had this. Electroencephalogr Clin Neurophysiol. 1980;49:89P. 214. Madison DS, Niedermeyer E. Epileptic seizures resulting from acute
192. Niedermeyer E, Fineyre R, Riley T, et al. Myoclonus and the elec- cerebral anoxia. J Neurol Neurosurg Psychiatry. 1970;33:381–386.
troencephalogram. A review. Clin Electroencephalogr. 1979;10: 215. Takahashi M, Kubota F, Nishi Y, et al. Persistent synchronous
75–95. periodic discharges caused by anoxic encephalopathy due to car-
193. Terzano MG, Mancia D, Calzetti S, et al. Diagnostic value of EEG diopulmonary arrest. Clin Electroencephalogr. 1993;24:166–172.
periodic discharges and cyclic changes in Creutzfeldt–Jakob dis- 216. Thorner MW. Procurement of electroencephalograph tracings in
ease. Electroencephalogr Clin Neurophysiol. 1981;52:52P. Abstract. 1000 flying cadets for evaluating the Gibbs technique in relation
194. Bortone E, Bettoni L, Giorgi C, et al. Reliability of EEG in the to flying ability. USAD School of Aviation Medical Research
diagnosis of Creutzfeldt–Jakob disease. Electroencephalogr Clin Report, No. 7-1; 1942.
Neurophysiol. 1994;90:323–330. 217. Gibbs FA, Gibbs EL, Lennox WG. Electroencephalographic clas-
195. Aminoff MJ, Greenberg DA, Wiekle DJ. Periodic EEG complexes sification of epileptic patients and control subjects. Arch Neurol
in infectious mononucleosis encephalitis. Electroencephalogr Clin Psychiatry. 1943;50:111–128.
Neurophysiol. 1982;53:28P. Abstract. 218. Harty JE, Gibbs EG, Gibbs FA. Electroencephalographic study of
196. Markand ON, Daly DD. Pseudoperiodic lateralized paroxysmal two hundred and seventy-five candidates for military service.
discharges in the electroencephalogram. Neurology (Minneapolis). War Med (Chicago). 1942;2:923–930.
1971;21:975–981. 219. Williams D. The nature of transient outbursts in the electroen-
197. Dauben RD, Adams AH. Periodic lateralized epileptiform dis- cephalogram of epileptics. Brain. 1944;67:10–37.
charges in EEG. A review with special attention to etiology and 220. Buchthal F, Lennox M. The EEG effect of Metrazol and photic
recurrence. Clin Electroencephalogr. 1974;8:116–124. stimulation in 682 normal subjects. Electroencephalogr Clin
198. Bauer G, Aichner F, Hengl W. Der diagnostische Wert periodisch Neurophysiol. 1953;5:545–558.
lateralisierter Komplexe im EEG. Z EEG-EMG. 1981;12:135–141. 221. Zivin L, Ajmone Marsan C. Incidence and prognostic signifi-
199. Naquet R, Vigouroux RA. The EEG in relation to pathology in cance of “epileptiform” activity in the EEG of nonepileptic sub-
simple cerebral ischaemia. In: Remond A, ed-in-chief. Handbook jects. Brain. 1968;91:751–777.
of Electroencephalography and Clinical Neurophysiology. Vol 14A. 222. Chatrian GE. Paroxysmal pattern in “normal” subjects. In: Remond
Amsterdam: Elsevier; 1972:38–44. A, ed-in-chief. Handbook of Electroencephalography and Clinical
200. De la Paz D, Brenner RP. Bilateral independent periodic lateralized Neurophysiology. Vol 6A. Amsterdam: Elsevier; 1976:114–123.
epileptiform discharges—clinical significance. Electroencephalogr 223. Nekhorocheff MI. La stimulation lumineuse intermittante chez
Clin Neurophysiol. 1982;53:27P. Abstract. l’enfant normal. Rev Neurol (Paris). 1950;83:601–602.
201. Schear HE. Periodic EEG activity. Clin Electroencephalogr. 1984; 224. Kellaway P, Fox BJ. Electroencephalographic diagnosis of cerebral
15:32–39. pathology in infants during sleep. I. Rationale, technique and the
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 533

characteristics of normal sleep in infants. J Pediatr. 1952;41: 246. Rodin EA, Gonzales S, Caldwell D, et al. Photic evoked responses
262–287. during induced epileptic seizures. Epilepsia (Amsterdam). 1966;
225. Brandt S, Brandt H. The electroencephalographic patterns in 7:202–214.
young healthy children from 0 to five years of life. Acta Psychiatr 247. Petsche H. Epileptischer Anfall und kortikale Neuronenpopula-
Scand. 1955;30:77–89. tion. Wien Z Nervenheilkd. 1968;26:45–55.
226. Corbin HPF, Bickford RG. Studies of the electroencephalogram of 248. Gastaut H, Naquet R, Fischer-Williams M. The pathophysiology
normal children: comparison of visual and automatic frequency of grand mal seizures generalized from the start. J Nerv Ment Dis.
analysis. Electroencephalogr Clin Neurophysiol. 1955;7:15–28. 1958;127:21–33.
227. Trojaborg W. Focal spike discharges in children. Acta Paediatr 249. Temkin O. The Falling Sickness. 2nd ed. Baltimore, MD: Johns
Scand Suppl. 1966;168:1–13. Hopkins Press; 1971.
228. Eeg-Olofsson O, Petersén I, Selldén U. The development of the 250. Esquirol. Quoted in Temkin, O. 1971; 1815.
electroencephalogram in normal children from the age of 1 251. Calmei L-F. De l’épilepsie, étudiée sous la rapport de son siège et de
through 15 years. Paroxysmal activity. Neuropaediatrie. 1971;2: son influence sur la production de l’aliénation mentale. Paris:
375–404. Thése de Paris; 1824.
229. Cavazzuti GB, Cappella L, Nalin A. Longitudinal study of epilep- 252. Bamberger P, Matthes A. Anfalle im Kindesalter. Basel: S. Karger;
tiform EEG patterns in normal children. Epilepsia (New York). 1959.
1980;21:43–55. 253. Sadleir LG, Scheffer IE, Smith S, et al. Automatisms in absence
230. Wolf P. The classification of seizures and the epilepsies. In: Porter seizures in children with idiopathic generalized epilepsy. Arch
RJ, Morselli PL, eds. The Epilepsies. London: Butterworth; 1985: Neurol. 2009;66:729–734.
107–124. 254. Sadleir LG, Scheffer IE, Smith S, et al. EEG features of absence
231. Baudewig J, Bittermann HJ, Paulus W, et al. Simultaneous EEG seizures in idiopathic generalized epilepsy: impact of syndrome,
and functional MRI of epileptic activity: a case report. Clin age, and state. Epilepsia. 2009;50:1572–1578.
Neurophysiol. 2001;112:1196–1200. 255. Niedermeyer E. Frontal lobe epilepsy: the next frontier. Clin
232. Commission on Classification and Terminology of the Electroencephalogr. 1998;29:163–169.
International League Against Epilepsy. Proposal for revised clinical 256. Niedermeyer E. Electrophysiology of the frontal lobe. Clin
and electroencephalographic classification of epileptic seizures. Electroencephalogr. 2003;34:5–12.
Epilepsia (New York). 1981;22:489–501. 257. Pavone A, Niedermeyer E. Absence seizures and the frontal lobe.
233. Dreifuss FE. The international classification of seizures and Clin Electroencephalogr. 2000;31:153–156.
epilepsies: advantages. In: Wolf P, ed. Epileptic Seizures and 258. Janz D. Die klinische Stellung der Pyknolepsie. Dtsch Med
Syndromes. London: Libbey; 1994:9–14. Wochenschr. 1955;80:1392–1400.
234. Nordli DR Jr, Bazil CW, Scheuer ML, et al. Recognition and clas- 259. Jung R. Uber vegetative Reaktionen und Hemmungswirkungen
sification of seizures in infants. Epilepsia. 1997;38:553–560. von Sinnesreizen im kleinen epileptischen Anfall. Nervenarzt.
235. Berg AT, Berkovic SF, Brodie MJ, et al. Revised terminology and 1939;12:169–185.
concepts for organization of seizures and epilepsies: report of the 260. Mirsky AF, Van Buren JM. On the nature of the “absence” in cen-
ILAE Commission on Classification and Terminology, 2005–2009. trencephalic epilepsy. A study of some behavioral, electroen-
Epilepsia. 2010;51:676–685. cephalographic and autonomic factors. Electroencephalogr Clin
236. Gowers WR. Epilepsy and Other Chronic Convulsive Diseases: Their Neurophysiol. 1965;18:334–348.
Causes, Symptoms and Treatment. New York: Dover; 1881 261. Cornil L, Gastaut H, Corriol J. Appréciation du degré de con-
(reprinted 1964) (American Academy of Neurology reprint series). science au cours des paroxysmes épileptiques “Petit Mal.” Rev
237. Janz D. Die Epilepsien. Stuttgart: Thieme; 1969. Neurol (Paris). 1951;84:149–151.
238. Gastaut H, Broughton R. Epileptic Seizures. Springfield, IL: 262. Fischgold H. La conscience et ses modifications. Systemes de
Charles C Thomas; 1972. references en E.E.G. clinique. In: Proceedings of the First
239. Karbowski K. Epileptische AnfŠlle. Berlin: Springer; 1985. International Congress of Neurological Sciences. Reports and
240. Tabbador K, Balagura S. Acute epidural hematoma following discussions. Vol 2. Brussels: Les editions Acta Med Belg; 1957:
epileptic seizures. Arch Neurol (Chicago). 1981;38:198–199. 181–213.
241. Gotman J, Ives JR, Gloor P. Frequency content of EEG and EMG 263. Lehmann HJ. Praparoxysmale Weckreaktionen bei pyknoleptis-
at seizure onset: possibility of removal of EMG artifact by digital chen Absenzen. Arch Psychiatr Nervenkr. 1963;204:417–426.
filtering. Electroencephalogr Clin Neurophysiol. 1981;52:626–639. 264. Schwab RS. Methods of measuring consciousness in attacks
242. Gastaut H, Fischer-Williams M. The physiopathology of epilep- of petit mal. Arch Neurol Psychiatry (Chicago). 1939;41:215–217.
tic seizures. In: Field J, Magoun HW, Hall VE, eds. Handbook of 265. Shimazono Y, Hirai T, Okuma T, et al. Disturbance of conscious-
Physiology. Vol 1. Baltimore, MD: Williams & Wilkins; 1959: ness in petit mal epilepsy. Epilepsia (Boston). 1953;2:49–55.
329–364. 266. Oller-Daurella L, Oller-Ferrer-Vidal L. Atlas de Crisis Epilepticas.
243. Zifkin B, Dravet C. Generalized convulsive seizures. In: Engel J Jr, Spain, Barcelona: Geigy Division Farmaceut; 1977.
Pedley T, eds. Epilepsy. A Comprehensive Textbook (3 vol.). Vol 1. 267. Mirsky AF, Tecce JJ. The analysis of visual potentials during spike
Philadelphia, PA: Lippincott-Raven; 1998:567–577. and wave activity. Epilepsia (Amsterdam). 1968;9:211–220.
244. Korff C, Nordli DR Jr. Do generalized tonic–clonic seizures in 268. Penry JK, Porter RJ, Dreifuss RE. Simultaneous recording of
infancy exist? Neurology. 2005;65:1750–1753. absence seizures with video tape and electroencephalography. A
245. Jung R. Hirnelekrische Untersuchungen über den Elektrokrampf. study of 374 seizures in 48 patients. Brain. 1975;98:427–440.
Die Erregungsabläufe in corticalen und subcorticalen Hirnregio- 269. Sadleir LG, Farrell K, Smith S, et al. Electroclinical features of
nen bei Katze und Hund. Arch Psychiatr Nervenkr. 1949;183: absence seizures in childhood absence epilepsy. Neurology. 2006;
206–244. 67:413–418.
534 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

270. Chaix Y, Daquin G, Monteiro F, et al. Absence epilepsy with onset 295. Kinsbourne M, Rosenfield DB. Nonprogressive myoclonus. In:
before age three years: a heterogeneous and often severe condi- Charlton MH, ed. Myoclonic Seizures. Amsterdam: Excerpta
tion. Epilepsia. 2003;44:944–949. Medica; 1975:30–59.
271. Leary LD, Wang D, Nordli DR Jr, et al. Seizure characterization 296. Sulibhavi DC, Schneck L. Myoclonus epilepsy in progressive dis-
and electroencephalographic features in Glut-1 deficiency. ease. In: Charlton MH, ed. Myoclonic Seizures. Amsterdam:
Epilepsia. 2003;44:701–707. Excerpta Medica; 1975:60–76.
272. Suls A, Mullen SA, Weber YG, et al. Early-onset absence epilepsy 297. Farrell DF, Swanson PD. Infectious diseases associated with
caused by mutations in the glucose transporter GLUT1. Ann myoclonus. In: Charlton MH, ed. Myoclonic Seizures. Amsterdam:
Neurol. 2009;66:415–419. Excerpta Medica; 1975:77–110.
273. Von Moers A, Brockmann K, Wang D, et al. EEG features of Glut- 298. Charlton MH. Infantile spasms. In: Charlton MH, ed. Myoclonic
1 deficiency syndrome. Epilepsia. 2002;43:941–945. Seizures. Amsterdam: Excerpta Medica; 1975:111–120.
274. Rütti W. Absenzen-Epilepsie im Erwachsenenalter. Schweiz Med 299. Niedermeyer E, Fineyre F, Riley T, et al. Myoclonus and the elec-
Wochenschr. 1982;112:434–441. troencephalogram. Clin Electroencephalogr. 1979;10:75–95.
275. Rütti W, Karbowski K. Absence epilepsy in adults. Electroen- 300. Aicardi J. Myoclonus as a manifestation of degenerative disorders
cephalogr Clin Neurophysiol. 1983;55:18P. Abstract. of the central nervous system in childhood. Electroencephalogr
276. Janz D, Beck-Mannagetta G, Spröder B, et al. Childhood absence Clin Neurophysiol. 1983;56:2P.
epilepsy (pyknolepsy) and juvenile absence epilepsy: one or two 301. Tassinari CA. New perspectives in epileptology. In: Japanese
syndromes? In: Wolf P, ed. Epileptic Seizures and Syndromes. Epilepsy Association, ed. Trends in Modern Epileptology. Tokyo:
London: Libbey; 1994:115–126. Proceedings of the International Public Seminar of Epileptology;
277. Trinka E, Baumgartner S, Unterberger I, et al. Long-term prog- 1981:42–59.
nosis for childhood and juvenile absence epilepsy. J Neurol. 2004; 302. Aguglia U, Gambardella A, Zappia M, et al. Negative myoclonus
251:1235–1241. during valproate-related stupor. Neurophysiological evidence of a
278. DeMarco P. Reflex petit mal absence? Clin Electroencephalogr. cortical non-epileptic origin. Electroencephalogr Clin Neurophysiol.
1990;21:74–76. 1995;94:103–108.
279. Lennox WG. The petit mal epilepsies; their treatment with trid- 303. Baumgartner C, Podreka I, Olbrich A, et al. Epileptic negative
ione. JAMA. 1945;129:1069–1074. myoclonus. An EEG-single-photon-emission CT study indicating
280. Watson CW, Denny-Brown D. Studies of the mechanism of involvement of premotor cortex. Neurology. 1996;46:753–758.
stimulus-sensitive myoclonus in man. Electroencephalogr Clin 304. Rubboli G, Parmeggiani L, Tassinari CA. Frontal inhibitory spike
Neurophysiol. 1955;7:341–356. component associated with epileptic negative myoclonus.
281. Halliday AM. The electrophysiological study of myoclonus. Electroencephalogr Clin Neurophysiol. 1995;95:201–205.
Brain. 1967;90:241–284. 305. Bravais LF. Recherches sur les Symptomes et le Traitement de
282. Halliday AM. Cerebral evoked potentials in familial progressive l’épilepsie Hémiplégique. Paris: Thèse de Paris No. 118; 1827.
myoclonic epilepsy. J R Coll Physicians Lond. 1967;1:123–134. 306. Fritsch G, Hitzig E. Uber die elekrische Erregbarkeit des
283. Halliday AM. The clinical incidence of myoclonus. In: Williams Grosshirns. Arch Anat Physiol. 1870;37:300–332.
D, ed. Modern Trends in Neurology. London: Butterworth; 1967: 307. Ferrier D. Experimental researches in cerebral physiology and
69–105. pathology. West Riding Lunatic Asylum Med Rep. 1873;3:30–96.
284. Halliday AM. The neurophysiology of myoclonus—a reappraisal. 308. Luciani L. Sulla patogenesi dell’epilessia. Riv Sper Freniatr Med
In: Charlton MH, ed. Myoclonic Seizures. Amsterdam: Excerpta Leg. 1878;4:617–646.
Medica; 1975:1–29. 309. Neufeld MY, Blumen S, Nisipeanu P. Lingual seizures.
285. Shibasaki H, Kuroiwa Y. Electroencephalographic correlates of Electroencephalogr Clin Neurophysiol. 1988;69:59P. Abstract.
myoclonus. Electroencephalogr Clin Neurophysiol. 1975;39:455–463. 310. Ahuja GK, Tharakan J. Ipsilateral seizures. Epilepsia (New York).
286. Chadwick D, Mallett M, Harris R, et al. Clinical biochemical and 1981;23:555–561.
physiological features distinguishing myoclonus response to 5- 311. Elger CE, Speckmann E-J. Focal interictal epileptiform discharges
hydroxytryptophan, tryptophan with a monoamino oxidase (FIED) in the epicortical EEG and their relations to spinal field
inhibitor and clonazepam. Brain. 1977;100:455–487. potentials in the rat. Electroencephalogr Clin Neurophysiol. 1980;
287. Hallett M, Chadwick D, Marsden CD. Ballistic movement overflow 48:447–460.
myoclonus: a form of essential myoclonus. Brain. 1977;100:299–312. 312. Elger CE, Speckmann E-J. Vertical inhibition in motor cortical
288. Kelly JJ Jr, Sharbrough FW, Daube JR. A clinical and electrophys- epileptic foci and its consequences for descending neuronal activity
iological evaluation of myoclonus. Neurology (New York). 1981; to the spinal cord. In: Speckmann E-J, Elger CE, eds. Epilepsy and
31:581–589. Motor System. Munich: Urban & Schwarzenberg; 1983:152–160.
289. Hallett M. Myoclonus: relation to epilepsy. Epilepsia (New York). 313. Classen J, Witte OW, Schlaug G, et al. Epileptic seizures triggered
1985;26(suppl 1):567–577. directly by focal transcranial stimulation. Electroencephalogr Clin
290. Gastaut H. Sémiologie des myoclonies et nosologie analytique Neurophysiol. 1995;94:19–25.
des syndromes myocloniques. Rev Neurol (Paris). 1968;119:1–30. 314. Aird RB, Woodbury DM. The Management of Epilepsy.
291. Oller-Daurella L, Oller-Ferrer-Fidal L. Altas de Crisis Epilepticas. Springfield, IL: Charles C Thomas; 1974.
2nd ed. Basel: Geigy; 1981. 315. Matthes A. Epilepsie. 3rd ed. Stuttgart: Thieme; 1977.
292. Weingarten K. Die Myoklonischen Syndrome. Vienna: Maudrich; 316. Penfield W, Jasper HH. Epilepsy and the Functional Anatomy of
1957. the Human Brain. Boston, MA: Little, Brown; 1954.
293. Aigner BR, Mulder DW. Myoclonus: clinical significance 317. Penfield W, Kristiansen K. Epileptic Seizure Patterns. Springfield,
and approach to classifications. Arch Neurol (Chicago). 1960;2: IL: Charles C Thomas; 1951.
600–615. 318. Fossas P, Sanchez ME, Oller L. Occipital origin of partial oculo-
294. Bauer G. Myoklonien: Erscheinungsbild, Pathophysiologie und clonic versive epileptic seizures in a case of post-traumatic
klinische Bedeutung. Wien Klin Wochenschr. 1974;124:577–581. epilepsy. Electroencephalogr Clin Neurophysiol. 1985;60:58P.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 535

319. Robillard A, Saint-Hilaire J-M, Mercier M, et al. The lateralizing 341. Bye AM. Neonate with benign familial neonatal convulsions:
and localizing value of adversion in seizures. Electroencephalogr recorded generalized and focal seizures. Pediatr Neurol. 1994;10:
Clin Neurophysiol. 1982;54:41P. Abstract. 164–165.
320. Ochs RF, Gloor P, Ives JR. The diagnostic value of head turning in 342. Aicardi J, Goutieres F. Encephalopathie myoclonique neonatale.
the localization of seizures. Electroencephalogr Clin Neurophysiol. Rev EEG Neurophysiol. 1978;8:99–101.
1981;51:21P–22P. Abstract. 343. Aicardi J. Early myoclonic encephalopathy. In: Roger J, Dravet C,
321. Quesney LF, Constain M, Rasmussen T. Seizures from the dorso- Bureau M, et al., eds. Epileptic Syndromes in Infancy, Childhood,
lateral frontal lobe. In: Chauvel P, Delgado-Escueta AV, Halgren E, and Adolescence. London: Libbey; 1985:12–21.
et al., eds. Frontal Lobe Seizures and Epilepsies. New York: Raven 344. Dalla Bernardina B, Capovilla G, Gattoni MB, et al. Épilepsie
Press; 1992:233–243. myoclonique grave de la prémière année. Rev. EEG Neurophysiol.
322. Pachatz C, Fusco L, Vigevano F. Epileptic spasms and partial 1982;12:21–25.
seizures as a single ictal event. Epilepsia. 2003;44:693–700. 345. Dalla Bernardina B, Dulac O, Fejerman N, et al. Early myoclonic
323. Kallen C, Wyllie E, Luders HO, et al. Hypomotor seizures in epileptic encephalopathy (E.M.E.E.). Eur J Pediatr. 1983;140:
infants and children. Epilepsia. 2002;43:882–888. 248–252.
324. Meencke H-J, Janz D. Neuropathological findings in primary 346. Ohtahara S, Ishida T, Oka E, et al. On the specific age-dependent
generalized epilepsy: a study of eight cases. Epilepsia (New York). epileptic syndrome. The early infantile epileptic encephalopathy
1984;25:8–21. with suppression-burst [in Japanese]. No-to-Hattatsu (Tokyo).
325. Meencke HJ, Janz D. The significance of microdysgenesia 1976;8:270–280.
in primary generalized epilepsy: an answer to the considera- 347. Ohtahara S. Clinico-electrical delineation of epileptic
tions to Lyon and Gastaut. Epilepsia (New York). 1985;26: encephalopathies in childhood. Asian Med J. 1978;21:499–509.
368–371. 348. Talada Serra M. Early epileptic encephalopathy with a pattern of
326. Lyon G, Gastaut H. Considerations on the significance attributed periodic bursts (Ohtahara syndrome). Electroencephalogr Clin
to unusual histological findings recently described in eight Neurophysiol. 1985;60:55P. Abstract.
patients with primary generalized epilepsy. Epilepsia (New York). 349. Saitsu H, Kato M, Mizuguchi T, et al. De novo mutations in the
1985;26:365–367. gene encoding STXBP1 (MUNC18-1) cause early infantile
327. Capovilla G, Berg AT, Cross JH, et al. Conceptual dichotomies in encephalopathy. Nat Genet. 2008;40:782–788.
classifying epilepsies: partial versus generalized and idiopathic 350. Fullston T, Brueton L, Willis T, et al. Ohtahara syndrome in a
versus symptomatic. Epilepsia. 2009;50:1645–1649. family with an ARX protein truncation. Eur J Hum Genet. 2010;18:
328. Lombroso CT. Consistent EEG focalities detected in subjects 157–162.
with primary generalized epilepsies monitored for two decades. 351. Murakami N, Ohtsuka Y, Ohtahara S. Early infantile epileptic
Epilepsia. 1997;38:797–812. syndromes with suppression-bursts. Jpn J Psychiatr Neurol. 1993;47:
329. Chahine LM, Mikati MA. Benign pediatric localization-related 197–200.
epilepsies. Part II. Syndromes in childhood. Epileptic Disord. 352. Coppola G, Plouin P, Chiron C, et al. Migrating partial seizures
2006;8:243–258. in infancy: a malignant disorder with developmental arrest.
330. Lagae L. What’s new in: “genetics in childhood epilepsy.” Eur J Epilepsia. 1995;36:1017–1024.
Pediatr. 2008;167:715–722. 353. Marsh E, Melamed SE, Barron T, et al. Migrating partial seizures
331. Lu Y, Wang X. Genes associated with idiopathic epilepsies: a in infancy: expanding the phenotype of a rare seizure syndrome.
current review. Neurol Res. 2009;31:135–143. Epilepsia. 2005;46:568–72.
332. Lucarini N, Verrotti A, Napolioni V, et al. Genetic polymorphisms 354. Cross JH. When is epileptic encephalopathy nonconvulsive status
and idiopathic generalized epilepsies. Pediatr Neurol. 2007;37: epilepticus? Epilepsia. 2007;48(suppl 8):42–43.
157–164. 355. Goldstein J, Slomski J. Epileptic spasms: a variety of etiologies
333. Helbig I, Scheffer IE, Mulley JC, et al. Navigating the channels and and associated syndromes. J Child Neurol. 2008;23:407–414.
beyond: unraveling the genetics of the epilepsies. Lancet Neurol. 356. Jeavons PM, Bower BD. Infantile Spasms. A Review of the
2008;7:231–245. Literature and a Study of 112 Cases. London: Heinemann; 1964.
334. Hirose S, Mitsudome A, Okada M, et al., on behalf of The 357. Fusco L, Vigevano F. Ictal clinical electroencephalographic
Epilepsy Study Group, Japan. Genetics of idiopathic epilepsies. findings of spasms in West syndrome. Epilepsia. 1993;34:
Epilepsia. 2005;46(suppl 1):38–43. 671–673.
335. Ottman R, Hirose S, Jain S, et al. Genetic testing in the epilepsies— 358. Hrachovy RA, Frost JD Jr, Kellaway P. Hypsarrhythmia: varia-
Report of the ILAE genetics commission. Epilepsia. 2010;51: tions on a theme. Epilepsia (New York). 1984;25:317–325.
655–670. 359. Kramer U, Sue WC, Mikati MA. Hypsarrhythmia: frequency of
336. Scheffer IE, Zhang UH, Gecz J, et al. Genetics of the epilepsies: variant patterns and correlation with etiology and outcome.
genetic twists in the channels and other tales. Epilepsia. 2010; Neurology. 1997;48:197–203.
51(suppl 1):33–26. 360. Kato M, Das S, Petras K, et al. Polyalanine expansion of ARX
337. Urdinguio RG, Sanchez-Mut JV, Esteller M. Epigenetic mecha- associated with cryptogenic West syndrome. Neurology. 2003;
nisms in neurological diseases: genes, syndromes, and therapies. 61:267–268.
Lancet Neurol. 2009;8:1056–1072. 361. Lux AL, Edwards SW, Hancock E, et al. The United Kingdom
338. Berg AT, Shinnar S, Levy SR, et al. How well can epilepsy syn- Infantile spasms study comparing vigabatrin with prednisolone
dromes be identified at diagnosis? A reassessment 2 years after or tetracosatide at 14 days: a multicenter randomized controlled
initial diagnosis. Epilepsia. 2000;41:1269–1275. trial. Lancet. 2004;364:1773–1778.
339. Hirsch E, Velez A, Sellal F, et al. Electroclinical signs of benign 362. Mackay MT, Weiss SK, Adams-Webber T, et al. Practice parameter:
familial convulsions. Ann Neurol. 1993;34:835–841. medical treatment of infantile spasms: report of the American
340. Aso K, Watanabe K. Benign familial neonatal convulsions: gener- Academy of Neurology and the Child Neurology Society. Neurology.
alized epilepsy? Pediatr Neurol. 1992;8:226–228. 2004;62:1668–1681.
536 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

363. Pellock JM. Infantile spasms: US consensus report. Epilepsia. 2010, 385. Nakayama J, Arinami T. Molecular genetics of febrile seizures.
DOI: 10.1111/j.1528–1167.2010.02657.x. Epilepsy Res. 2006;70S:S190–S198.
364. Elterman RD, Shields WD, Mansfield KA, et al. Randomized trial 386. Heida JG, Moshe SL, Pittman QJ. The role of interleukin 1-B in
of vigabatrin in patients with infantile spasms. Neurology. 2001; febrile seizures. Brain Dev. 2009;31:388–393.
57:1416–1421. 387. Lennox-Buchthal MA. Fieberkrémpfe. Stuttgart: Hippokrates; 1977.
365. Ohtahara S, Yamatogi Y, Ohtsuka Y, et al. Prognosis of the West 388. Des Termes H, Mises J, Plouin P, et al. The “spike focus” during the
syndrome with special reference to Lennox syndrome: a develop- evolution of febrile convulsions: an electrophysiological and clin-
mental study. In: Wada JA, Penry JK, eds. Advances in Epileptology. ical study of 35 patients. Electroencephalogr Clin Neurophysiol.
Xth Epilepsy International Symposium. New York: Raven Press; 1978;45:370. Abstract.
1980:149–154. 389. Yamatogi Y, Ishida S, Terasaki T, et al. An electroencephalo-
366. Hughes JR. Natural history of hypsarrhythmia. Clin Electroen- graphic study of febrile convulsions. Electroencephalogr Clin
cephalogr. 1985;16:128–130. Neurophysiol. 1982;54:27P–28P. Abstract.
367. Talwar D, Baldwin MA, Hutzler R, et al. Epileptic spasms in older 390. Yamatogi Y, Ohtahara S. EEG in febrile convulsions. Am J EEG
children: persistence beyond infancy. Epilepsia. 1995;36:151–155. Technol. 1990;30:267–280.
368. Camfield P, Camfield C, Lortie A, et al. Infantile spasms in remis- 391. Panayiotopoulos CP. Benign Childhood Partial Seizures and
sion may reemerge as intractable epileptic spasms. Epilepsia. 2003; Related Epileptic Syndromes. London: Libbey; 1999.
44:1592–1593. 392. Panayiotopoulos CP. The Panyiotopoulos Syndrome. London:
369. Chugani HT, Shields WD, Shewmon DA, et al. Infantile spasms: Libbey; 2002.
I. PET identifies focal cortical dysgenesis in cryptogenic cases for 393. Caraballo R, Cersosimo R, Fejerman N. Panayiotopoulos syn-
surgical treatment. Ann Neurol. 1990;27:406–413. drome: a prospective study of 192 patients. Epilepsia. 2007;48:
370. Koo B, Hwang P. Localization of focal cortical lesions influences 1054–1061.
age of onset of infantile spasms. Epilepsia. 1996;37:1068–1071. 394. Specchio N, Trivisano M, Di Ciommo V, et al. Panayiotopoulos
371. Roger J, Soulayrol R, Pinsard N. Diagnostic différential. In: syndrome: a clinical, EEG, and neuropsychological study of 93
Gastaut H, Soulayrol R, Roger J, et al., eds. L’Encéphalopathie consecutive patients. Epilepsia. 2010, DOI: 10.1111/j.1528–1167.
Myoclonique Infantile avec Hypsarrhythmie (Syndrome de West). 2010.02639.x.
Paris: Masson; 1964:143–149. 395. Leal AJR, Ferreira JC, Dias AI, et al. Origin of frontal lobe spikes
372. Kinsbourne M. Myoclonic encephalopathy of infants. J Neurol in the early onset benign occipital lobe epilepsy (Panayiotopoulos
Neurosurg Psychiatry. 1962;25:271–276. syndrome). Clin Neurophysiol. 2008;119:1985–1991.
373. Darra F, Fiorini E, Zoccante L, et al. Benign myoclonic epilepsy of 396. Yoshinaga H, Kobayashi K, Ohtsuka Y. Characteristics of the
infancy (BMEI): a longitudinal electroclinical study of 22 cases. synchronous occipital and frontal spike phenomenon in
Epilepsia. 2006;47(suppl 5):31–35. Panayiotopoulos syndrome. Brain Dev. 2010;32(8):603–608.
374. Vigevano F. Benign familial infantile seizures. Brain Dev. 2005; 397. Doose H. Das Akinetische Petit mal. Arch Psychiatr Nervenkr.
27:172–177. 1964;205:625–654.
375. Specchio N, Vigevano F. The spectrum of benign infantile 398. Doose H. Myoclonic astatic epilepsy of early childhood. In: Roger
seizures. Epilepsy Res. 2006;70(suppl 1):156–167. J, Dravet C, Bureau M, et al., eds. Epileptic Syndromes in Infancy,
376. Capovilla G, Beccaria F. Benign partial epilepsy in infancy and Childhood and Adolescence. London: Libbey; 1985:78–88.
early childhood with vertex spikes and waves during sleep: a new 399. Bonanni P, Parmeggiani L, Guerrini R. Different neurophysiologi-
epileptic form. Brain Dev. 2000;22:93–98. cal patterns of myoclonus characterize Lennox–Gastaut syndrome
377. Dravet C. Les Épilepsies graves de l’enfant. Vie Med. 1978;8: and myoclonic-astatic epilepsy. Epilepsia. 2002;43:609–615.
543–548. 400. Kilaru S, Bergqvist C. Current treatment of myoclonic astatic
378. Dalla Bernardina B, Dulac O, Bureau M, et al. Encephalopathie epilepsy: clinical experience at the Children’s Hospital of
myoclonique precoce avec epilepsie. Rev EEG Neurophysiol. 1982; Philadelphia, PA. Epilepsia. 2007;48:1703–1707.
12:8–14. 401. Gastaut Y. Un élément Džroutant De La Séméiologie élec-
379. Dravet C, Bureau M, Roger J. Severe myoclonic epilepsy in troencžphalographique: les pointes rolandiques sans signfication
infants. In: Roger J, Dravet C, Bureau M, et al., eds. Epileptic focale. Rev Neurol (Paris). 1952;87:448–450.
Syndromes in Infancy, Childhood, and Adolescence. London: Libbey; 402. Hess R. Verlaufsuntersuchungen über Anfälle und EEG bei
1985:58–67. kindlichen Epilepsien. Arch Psychiatr Nervenkr. 1958;197:568–593.
380. Caraballo RH, Cersosimo RO, Espeche A, et al. Myoclonic status 403. Nayrac P, Beaussart M. Les pointes-ondes prérolandiques.
in nonprogressive encephalopathies: study of 29 cases. Epilepsia. Expression EEG trés particulière: étude électroclinique de 21 cas.
2007;48:107–113. Rev Neurol (Paris). 1958;99:201–206.
381. Knudsen FU. Febrile convulsions. In: Dam M, Gram L, eds. 404. Isler W, Hess R. Verlaufsuntersuchungen Ÿber AnfŠlle und EEG
Comprehensive Epileptology. New York: Raven Press; 1991:133–143. bei fokalen Epilepsien im Kindesalter. Arch Psychiatr Nervenkr.
382. Beaumanoir A. Les Epilepsies infantiles. Problèmes de Diagnostic et 1960;200:257–266.
de Traitement. Basel: Editiones Roche; 1976. 405. Beaumanoir A, Ballis T, Varfis G, et al. Benign epilepsy of childhood
383. Provisional Committee on Quality Improvement, Subcommittee with rolandic spike. A clinical electroencephalographic and telen-
on Febrile Seizures. Practice Parameter: the neurodiagnostic eval- cephalographic study. Epilepsia (Amsterdam). 1974;15:301–315.
uation of the child with a first simple febrile seizure. Pediatrics. 406. Beaussart M. Benign epilepsy of children with rolandic (centro-
1996;97:769–775. temporal) paroxysmal foci. A clinical entity. Study of 221 cases.
384. Okumura A, Ishiguro Y, Sofue A, et al. Treatment and outcome in Epilepsia (Amsterdam). 1972;13:795–811.
patients with in patients with febrile convulsion associated with 407. Blom S, Brorson LO. Central spikes or sharp waves (rolandic
epileptiform discharges on electroencephalography. Brain Dev. spikes) in children’s EEG and their clinical significance. Acta
2004;26:241–244. Pediatr Scand. 1966;55:385–393.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 537

408. Blom S, Heijbel J. Benign epilepsy of children with centrotempo- 429. Niedermeyer E. Dipole theory and electroencephalography. Clin
ral EEG foci: a follow-up study in adulthood of patients initially Electroencephalogr. 1996;27:121–131.
studied as children. Epilepsia (New York). 1982;23:629–632. 430. Fonseca LC, Tedrus GM, Bastos A, et al. Reactivity to rolandic
409. Dalla Bernardina B, Beghini G. Rolandic spikes in children with spikes. Clin Electroencephalogr. 1996;27:116–120.
and without epilepsy (20 subjects photographically studied dur- 431. Nadkarni MA, Postolache V, Gold A, et al. Central mid-temporal
ing sleep). Epilepsia (New York). 1976;17:161–168. spikes triggered by blinking. Electroencephalogr Clin Neurophysiol.
410. Hejbel J, Blom S, Bergfors PG. Benign epilepsy of children with 1994;90:36–39.
centrotemporal EEG foci. A study of incidence rate in outpatient 432. Mitsudome A, Ohu M, Yasumoto S, et al. Rhythmic slow activity
care. Epilepsia (New York). 1975;16:657–664. in benign childhood epilepsy with centrotemporal spikes. Clin
411. Lairy GC, Harrison A. Functional aspects of EEG foci in children. Electroencephalogr. 1997;28:44–48.
In: Kellaway P, Petersen I, eds. Clinical Electroencephalography in 433. Nelson KR, Brenner RP, De La Paz D. Midline spikes—electroen-
Children. New York: Grune & Stratton (Stockholm: Almqvist & cephalographic and clinical features. Electroencephalogr Clin
Wiksell); 1968:197–212. Neurophysiol. 1984;57:42P–43P. Abstract.
412. Lombroso CT. Sylvian seizures and midtemporal spike foci in 434. Pedley TA, Tharp BR, Herman KR. Electroencephalographic and
children. Arch Neurol (Chicago). 1967;17:52–59. clinical correlates of vertex spike foci. Electroencephalogr Clin
413. Loiseau P, Beaussart M. The seizures of benign childhood epilepsy Neurophysiol. 1980;50:153P. Abstract.
with Rolandic paroxysmal discharges. Epilepsia (Amsterdam). 435. Pourmand RA, Markand ON, Thomas C. Midline spike dis-
1973;14:381–389. charges: clinical and EEG correlates. Clin Electroencephalogr. 1984;
414. Niedermeyer E. Spitzen über der Zentralregion und mu 15:232–236.
Rhythmus: gedanken zum Problem der “funktionellen” Spitzen. 436. Ehle AL. Midline spike foci in children. Electroencephalogr Clin
Z EEG-EMG. 1970;1:133–141. Neurophysiol. 1980;50:153P. Abstract.
415. Niedermeyer E. Focal and generalized seizure discharges in the 437. Weglage J, Demsky A, Pietsky M, et al. Neuropsycholgical, intel-
electroencephalogram and their responses to intravenous lectual, and behaviorla findings in patients with centrotemporal
diazepam. Int Med Dig. 1972;7:49–61. spikes with and without seizures. Dev Med Child Neurol. 1997;
416. Gastaut H. Individualization of so-called benign and functional 39:646–651.
epilepsy at different times of life. Electroencephalogr Clin Neuro- 438. Staden U, Isaacs E, Boyd SG, et al. Language dysfunction in chil-
physiol. 1982;53:79P. Abstract. dren with rolandic epilepsy. Neuropediatrics. 1998;29:242–248.
417. Loiseau P, Pestre M, Dartigues F, et al. Long-term prognosis in 439. Croona C, Kihlgren M, Lundberg S, et al. Neuropsychological
two forms of childhood epilepsy: typical absence seizures and findings in children with benign childhood epilepsy with cen-
epilepsy with rolandic (centrotemporal) EEG foci. Ann Neurol. trotemporal spikes. Dev Med Child Neurol. 1999;41:813–818.
1983;13:642–648. 440. Peters JM, Camfield CS, Camfield PR. Population study of benign
418. Niedermeyer E. Complexities of primary generalized epilepsy. Rolandic epilepsy: is treatment needed? Neurology. 2001;57:
Clin Electroencephalogr. 1981;12:177–191. 537–539.
419. Nogueira de Melo A, Niedermeyer E. Crossover phenomena in 441. Lerman P. Benign partial epilepsy with centro-temporal spikes.
epileptic syndromes in childhood epilepsies. Clin Electroen- In: Roger J, Bureau M, Dravet C, et al., eds. Epileptic Syndromes in
cephalogr. 1991;22:75–82. Infancy, Childhood and Adolescence. 2nd ed. London: John Libbey
420. Aicardi J, Chevrie JJ, Rousselie F. Le syndrome spasmes en and Company; 1992:189–200.
flexion, agénesie calleuse, anomalies choriorétiniennes. Archive 442. Lerman P, Kivity S. Benign focal epilepsy of childhood. A follow-
Francaise de Pediatric. 1969;26:1103–1120. up study of 100 recovered patients. Arch Neurol. 1975;23:
421. Blom S, Heijbel J, Bergfors PG. Benign epilepsy of children with 261–264.
centrotemporal EEG foci. Prevalence and follow-up study of 40 443. Kramer U, Zelnik N, Lerman-Sagie T, et al. Benign childhood
patients. Epilepsia (Amsterdam). 1972;13:609–613. epilepsy with centrotemporal spikes: clinical characteristics and
422. Gregory DL, Wong PKH. Clinical relevance of a dipole field in identification of patients at risk for multiple seizures. J Child
rolandic spikes. Epilepsia (New York). 1992;33:36–44. Neurol. 2002;17:17–19.
423. Wong PK. The importance of source behavior in distinguishing 444. Massa R, de Saint Martin A, Carcangiu R, et al. EEG criteria pre-
populations of epileptic foci. J Clin Neurophysiol. 1993;10:314–322. dictive of a complicated evolution in idiopathic rolandic epilepsy.
424. Van der Meij W, Van Huffelen AC, Wieneke GH, et al. Sequential Neurology. 2001;57:1071–1079.
EEG mapping may differentiate “epileptic” from “nonepileptic” 445. Bray PF, Wiser WC. The relation of focal to diffuse epileptiform
rolandic spikes. Electroencephalogr Clin Neurophysiol. 1992;82: EEG discharges in genetic epilepsy. Arch Neurol (Chicago). 1965;13:
408–414. 223–237.
425. Luders H, Lesser RP, Dinner DS, et al. Benign focal epilepsy of 446. Doose H, Gerken H, Kiefer R, et al. Genetic factors in childhood
childhood. In: Luders H, Lesser RP, eds. Epilepsy Electroclinical epilepsy with focal sharp waves. II. EEG findings in patients and
Syndromes. Berlin: Springer; 1987:303–346. siblings. Neuropadiatrie. 1977;8:10–20.
426. Gutierrez AR, Brick JF, Bodensteiner J. Dipole reversal: an ictal 447. Gereby G. Benign centro-temporal epilepsy of childhood. EEG
feature of benign partial epilepsy with centrotemporal spikes. and clinical follow-up of 80 cases. Electroencephalogr Clin
Epilepsia. 1990;31:544–548. Neurophysiol. 1985;61:7P. Abstract.
427. Legarda S, Jayakar P. Electroclinical significance of rolandic 448. Gelisse P, Corda D, Raybaud C, et al. Abnormal neuroimaging in
spikes and dipoles in neurodevelopmentally normal children. patients with benign epilepsy with centrotemporal spikes.
Electroencephalogr Clin Neurophysiol. 1995;95:257–259. Epilepsia. 2003;44:372–378.
428. Yoshinaga H, Amano R, Oka E, et al. Dipole tracing in childhood 449. Bali B, Kull LL, Strug LJ, et al. Autosomal dominant inheritance
epilepsy with special reference to rolandic. Brain Topogr. 1992; of centrotemporal sharp waves in Rolandic epilepsy families.
4:193–199. Epilepsia. 2007;48:2266–2272.
538 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

450. Strug LJ, Clarke T, Chiang T, et al. Centrotemporal sharp wave 473. Panayiotopoulos CP. Basilar migraine, seizures and severe epileptic
EEG trait in rolandic epilepsy maps to Elongator Protein EEG abnormalities. Neurology (Minneapolis). 1980;30:1122–1125.
Complex 4 (ELP4). Eur J Hum Genet. 2009;17:1171–1181. 474. Riggio S, Santiago M, Niedermeyer E. Benign occipital lobe
451. Lerman P, Kivity-Ephraim S. Focal epileptic EEG discharges in chil- epilepsy. Neurology. 1987;37(suppl I):106. Abstract.
dren not suffering from clinical epilepsy: etiology, clinical signifi- 475. Lugaresi E, Cirignotta F, Montagna P. Occipital lobe epilepsy with
cance and management. Epilepsia (New York). 1981;22:551–558. scotosensitive seizures: the role of central vision. Epilepsia (New
452. Gibbs FA. Differentiation of mid-temporal, anterior temporal York). 1984;25:115–120.
and diencephalic epilepsy. In: Baldwin M, Bailey P, eds. Temporal 476. Tassinari CA, Bureau M. Epilepsy with myoclonic absences. In:
Lobe Epilepsy. Springfield, IL: Charles C Thomas; 1958:109–117. Roger J, Dravet C, Bureau M, et al., eds. Epileptic Syndromes in
453. Andermann F, Oguni H. Do epileptic foci migrate? The pros. Infancy, Childhood and Adolescence. London: John Libbey and
Electroencephalogr Clin Neurophysiol. 1991;76:96–99. Co; 1985:121–129.
454. Blume WT. Do epileptic foci migrate? The cons. Electroen- 477. Bureau M, Tassinari CA. Epilepsy with myoclonic absences. Brain
cephalogr Clin Neurophysiol. 1991;76:100–105. Dev. 2005;27:178–184.
455. Lee IC, Chen YJ, Lee HS. Migration of epileptic spike foci in 478. Manonmani V, Wallace SJ. Epilepsy with myoclonic absences.
encephalograms may correlate with a better outcome in pediatric Arch Dis Child. 1994;70:288–290.
epilepsy. Brain Dev. 2010; Jan 8. 479. Elia M, Guerrini R, Musumeci SA, et al. Myoclonic absence-like
456. Niedermeyer E, Rocca U. Scalp, cortical and depth EEG contribu- seizures and chromosomal abnormalities. Epilepsia. 1998;39:
tion to focal motor epilepsy and epilepsia partialis continua. 660–663.
Electroencephalogr Clin Neurophysiol. 1980;50:160P. Abstract. 480. Niedermeyer E. The Lennox–Gastaut syndrome: a severe type of
457. Rocca U, Niedermeyer E. Severe forms of focal motor seizure dis- childhood epilepsy. Electroencephalogr Clin Neurophysiol.
orders in childhood. In: Proc. Congr. Latin. Amer. (Buenos Aires), 1968;24:283. Abstract.
1982:277–291. 481. Oller-Daurella L. Sindrome de Lennox. Barcelona: Editorial
458. Andrews PI, McNamara JO, Lewis DV. Clinical and electroen- Espaxs; 1967.
cephalographic correlates in Rasmussen’s encephalitis. Epilepsia. 482. Oller-Daurella L. Evolution et pronostic du syndrome de
1997;38:189–194. Lennox–Gastaut. In: Lugaresi E, ed. Evolution and Prognosis of the
459. Aguilar MJ, Rasmussen T. Role of encephalitis in pathogenesis of Epilepsies. Bologna: Gaggi; 1973:155–164.
epilepsy. Arch Neurol (Chicago). 1960;2:663–676. 483. Oller-Daurella L. Las fronteras entre et petit mal y el sindrome de
460. Rasmussen T, Olszewski J, Lloyd-Smith D. Focal seizures due to Lennox–Gastaut. Rev Esp Otoneurooftalmol Neurocir. 1976;34:
chronic localized encephalitis. Neurology (Minneapolis). 1958;8: 27–44.
435–445. 484. Arzimanoglou A, French J, Blume WT, et al. Lennox–Gastaut
461. Bien CG, Granata T, Antozzi C, et al. Pathogenesis, diagnosis, and syndrome: a consensus approach on diagnosis, assessment,
treatment of Rasmussen encephalitis. A European consensus management, and trial methodology. Lancet Neurol. 2009;8:
statement. Brain. 2005;128:454–471. 82–93.
462. Bien CG, Schramm J. Treatment of Rasmussen encephalitis half 485. Gastaut H, Gastaut JL, Gonçalves e Silva E, et al. Relative fre-
a century after its initial description: promising prospects and a quency of different types of epilepsy: a study employing the clas-
dilemma. Epilepsy Res. 2009;86:101–112. sification by the International League against Epilepsy. Epilepsia
463. Granata T, Fusco L, Gobbi G, et al. Experience with immunomod- (New York). 1975;16:457–461.
ulatory treatments in Rasmussen’s encephalitis. Neurology. 2003; 486. Komai S. Lennox–Gastaut’s syndrome. Prognosis of the secondary
61:1807–1810. generalized epilepsies. Epilepsia (New York). 1977;18:131. Abstract.
464. Daniel RT, Villemure J-G, Granata T, et al. Experience with 487. Bauer G, Benke T, Bohr K. The Lennox–Gastaut syndrome in
immunomodulatory treatments of Rasmussen encephalitis. adulthood. In: Niedermeyer E, Degen R, eds. The Lennox–Gastaut
Neurology. 2004;63:1761–1762. Syndrome. New York: Alan R. Liss; 1988:317–327.
465. Otsubo H, Chitoku S, Ochi A, et al. Malignant rolandic-sylvian 488. Olmos-Garcia de Alba G, Valdez JM, Crespo FV. West syndrome
epilepsy in children. Diagnosis, treatment and outcomes. Neuro- evolving into the Lennox–Gastaut syndrome. Clin Electroen-
logy. 2001;57:590–596. cephalogr. 1984;15:61–68.
466. Ong HT, Wyllie E. Benign childhood epilepsy with centrotempo- 489. Gastaut H, Roger J, Soulayrol R, et al., eds. L’Encéphalopathie
ral spikes: is it always benign? Neurology. 2000;54:1182–1185. Myoclonique Infantile avec Hypsarrhythmie (Syndrome de West).
467. Raju P, Sarco DP, Poduri A, et al. Oxcarbazepine in children with Paris: Masson; 1964.
nocturnal frontal lobe epilepsy. Pediatr Neurol. 2007;37:345–349. 490. Patry G, Lyagoubi S, Tassinari A. Subclinical “electrical status
468. Gastaut H. A new type of epilepsy: benign partial epilepsy of epilepticus” induced by sleep in children. A clinical and electroen-
childhood with occipital spike-waves. Clin Electroencephalogr. cephalographic study of six cases. Arch Neurol. 1971;24:242–252.
1982;13:13–22. 491. Tassinari CA, Daniele O, Dravet C, et al. Sleep polygraphic studies
469. Taylor I, Scheffer IE, Berkovic SF. Occipital epilepsies: identifica- in some epileptic encephalopathies from infancy to adolescence. In:
tion of specific and newly recognized syndromes. Brain. 2003; Degen R, Niedermeyer E, eds. Epilepsy, Sleep and Sleep Deprivation.
126:753–769. Amsterdam: Elsevier; 1984:175–189.
470. Taylor I, Berkovic SF, Kivity S, et al. Benign occipital epilepsies of 492. Niedermeyer E, Freeman JM, Long DM, et al. EEG studies in
childhood. Brain. 2008;131:2287–2294. recalcitrant and disabling focal motor seizures. Epilepsia (New
471. Panayiotopoulos CP, Michael M, Sanders S, et al. Benign child- York). 1977;18:289. Abstract.
hood focal epilepsies: assessment of established and newly recog- 493. Niedermeyer E. Considerations of the centrencephalic (general-
nized syndromes. Brain. 2008;131:2264–2286. ized) type of epilepsy. Del Med J. 1966;38:341–348.
472. Niedermeyer E, Riggio S, Santiago M. Benign occipital lobe 494. Nogueira de Melo A, Niedermeyer E. Considerations of second-
epilepsy. J Epilepsy. 1988;1:3–11. ary temporalization. Clin Electroencephalogr. 1991;22:161–171.
Chapter 26 ■ Seizures and Epilepsy in Infants to Adolescents 539

495. Hughes JR, Patil VK. Long-term electro-clinical changes in the 518. Asconape J, Penry JK. Some clinical and EEG aspects of benign
Lennox–Gastaut syndrome before, during, and after the flow juvenile myoclonic epilepsy. Epilepsia. 1984;25:108–114.
spikewave pattern. Clin Electroencephalogr. 2002;33:1–7. 519. Schmitz B, Sander T, eds. Juvenile Myoclonic Epilepsy. Wrightson:
496. Ohtahara S, Ohtsuka Y, Kobayashi K. Lennox–Gastaut syndrome: Petersfield; 2000.
a new vista. Psychiatry Clin Neurosci. 1995;49:S179–S183. 520. Oguni H, Mukahira K, Oguni M, et al. Video-polygraphic analy-
497. Hirt HR. Zur Nosologie des Lennox–Gastaut Syndromes. sis of myoclonic seizures in juvenile myoclonic epilepsy. Epilepsia.
Nervenarzt. 1996;67:109–122. 1994;35:307–316.
498. Tassinari CA, Bureau M, Dravet C, et al. Electrical status epilep- 521. Montalenti E, Imperiale D, Rovera A, et al. Clinical features, EEG
ticus during sleep in children (ESES). In: Sterman MB, Shouse findings, and diagnostic pitfalls in juvenile myoclonic epilepsy: a
MN, Passouant P, eds. Sleep and Epilepsy. New York: Academic series of 63 patients. J Neurol Sci. 2001;184:65–70.
Press; 1982:465–479. 522. Lancman ME, Asconape JJ, Penry JK. Clinical and EEG asymme-
499. Kramer U, Nevo Y, Neufeld MY, et al. Epidemiology of epilepsy in tries in juvenile myoclonic epilepsy. Epilepsia. 1994;35:302–306.
childhood: a cohort of 440 consecutive patients. Pediatr Neurol. 523. So GM, Thiele EA, Sanger T, et al. Electroencephalogram and
1998;18:46–50. clinical focalities in juvenile myoclonic epilepsy. J Child Neurol.
500. Guilhoto LMFF, Morrell F. Electrophysiological differences 1998;13:541–545.
between Landau–Kleffner syndrome and other conditions show- 524. Aliberti V, Grunewald RA, Panayiotopoulos CP, et al. Focal elec-
ing the CSWS electrical pattern. Epilepsia. 1994;35(suppl 8):126. troencephalographic abnormalities in juvenile myoclonic
501. Hirsch E, Maquet P, Metz-Lutz M-N, et al. The eponym epilepsy. Epilepsia. 1994;35:297–301.
“Landau–Kleffner syndrome” should not be restricted to child- 525. de Carvalho Sousa NA, da Silva Sousa P, Garzon E, et al. EEG
hood-acquired aphasia with epilepsy. In: Beaumanoir A, Bureaus recording after sleep deprivation in a series of patients with juvenile
M, Deonna T, et al., eds. Continuous Spikes and Waves During myoclonic epilepsy. Arq Neuropsiquiatr. 2005;63(2-B):383–388.
Slow Sleep. Electrical Status Epilepticus During Slow Sleep. 526. Camfield CS, Camfield PR. Juvenile myoclonic epilepsy 25 years
London: Libbey; 1995:57–62. after seizure onset: a population-based study. Neurology. 2009;73:
502. Landau W, Kleffner FR. Syndrome of acquired aphasia with con- 1041–1045.
vulsive disorder in children. Neurology. 1957;7:523–530. 527. Wirrell EC, Camfields CS, Camfield PR, et al. Long-term progno-
503. Stefanatos GA, Green GG, Ratcliff GG. Neurophysiological evi- sis of typical childhood absence epilepsy: remission or progres-
dence of auditory channel anomalies in developmental dyspha- sion to juvenile myoclonic epilepsy. Neurology. 1996;47:912–918.
sia. Arch Neurol. 1989;46:871–875. 528. Blume WT. Clinical and electroencephalographic correlates of
504. Kuhlo W. Petit-mal-Epilepsie. In: Niedermeyer E, ed. Epilepsy, the multiple independent spike foci pattern in children. Ann
Recent Views on Theory, Diagnosis and Therapy of Epilepsy. Basel: Neurol. 1978;4:541–547.
S. Karger; 1970:120–138. 529. Yamatogi Y, Ohtahara S. Multiple independent spike foci with
505. Sauer H. Über gehäufte kleine Anfälle bei Kindern (Pyknolepsie). epilepsy, with special reference to a new epileptic syndrome of
Monatsschr Psychiatr Neurol. 1916;40:267–300. “severe epilepsy with multiple independent spike foci.” Epilepsy
506. Loiseau P, Duché B, Pédespan J-M. Absence epilepsies. Epilepsia. Res. 2006;70(S1):S96–104.
1995;36:1182–1186. 530. Kotagal P. Multifocal independent Spike syndrome: relationship
507. Sadleir LG, Scheffer IE, Smith S, et al. Factors influencing clinical to hypsarrhythmia and the slow spike-wave (Lennox–Gastaut)
features of absence seizures. Epilepsia. 2008;49:2100–2107. syndrome. Clin Electroencephalogr. 1995;26:23–29.
508. Paal G. Katamnestische Untersuchungen und EEG bei Pyknolepsie. 531. Hauser WA, Rich S, Anderson VE. The multifocal spike pattern
Arch Psychiatr Nervenkr. 1957;196:48–62. and sibling risk for epilepsy. Electroencephalogr Clin Neurophysiol.
509. Livingston S, Torres I, Pauli L, et al. Petit mal epilepsy. Results 1984;57:44P–45P. Abstract.
of a prolonged follow-up of 117 patients. JAMA. 1965;194: 532. Elger CE, Speckmann E-J. Interiktale epileptiforme Potentiale im
227–232. corticalen OberflŠchen-EEG und ihre Beziehungen zu spinalen
510. Janz D. Aufwach-Epilepsien. Arch Psychiatr Nervenkr. 1953;191: Feldpotentialen bei der Ratte. In: Doose M, Gross-Selbeck G, eds.
73–98. Epilepsie 1978. Stuttgart: Thieme; 1979:245–249.
511. Janz D. The grand mal epilepsies and the sleeping-waking cycle. 533. Lugaresi E, Pazzaglia P. The EEG in the positive and differential
Epilepsia (Amsterdam). 1962;3:69–109. diagnosis of epilepsy. In: Remond A, ed-in-chief. Handbook of
512. Waltz S, Beck-Managetta G, Janz D. Are there syndrome-related Electroencephalography and Clinical Neurophysiology. Vol 13A.
genetically determined spike and wave patterns? A comparison Amsterdam: Elsevier; 1975:69–71.
between syndromes of generalized epilepsy. Epilepsia 1990;31: 534. Engel J Jr. A practical guide for routine EEG studies in epilepsy. J
819 (abstract). Clin Neurophysiol. 1984;1:109–142.
513. Camfield CS, Camfield PR, Sadler M, et al. Paroxysmal eyelid 535. Dinner, D.S., and Lueders, H. The electroencephalogram in the
movements: a confusing feature of generalized photosensitive routine evaluation of patients with epilepsy. In: Porter RJ, Morselli
epilepsy. Neurology. 2004;63:40–42. PL, eds. The Epilepsies. London: Butterworth; 1985:142–173.
514. Tovia E, Goldberg-Stern H, Shahar E, et al. Outcome of children 536. Hughes JR. The significance of the interictal spike discharges. A
with juvenile absence epilepsy. J Child Neurol. 2006;21:766–768. review. J Clin Neurophysiol. 1989;6:207–226.
515. Tezer FI, Sahin G, Ciger A, et al. Focal EEG findings in juvenile 537. Ajmone Marsan C, Zivin LS. Factors related to the occurrence of
absence syndrome and the effect of antiepileptic drugs. Clin EEG typical paroxysmal abnormalities in the EEG records of epileptic
Neurosci. 2008;39:33–38. patients. Epilepsia (Amsterdam). 1970;11:361–381.
516. Tsuboi T. Primary Generalized Epilepsy with Sporadic Myoclonias 538. Bauer G. Der Wert von EEG-Kontrollen mglichst bald nach
of Myoclonic Petit Mal Type. Stuttgart: Thieme; 1977. einem epileptischen Anfall. Z EEG-EMG. 1975;6:125–130.
517. Delgado-Escueta AV, Enrile-Bascal F. Juvenile myoclonic epilepsy 539. Kaibara M, Blume WT. The postictal electroencephalogram.
of Janz. Neurology. 1984;34:285–294. Electroencephalogr Clin Neurophysiol. 1988;70:99–104.
540 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

540. Gastaut H, Gomez-Almanzar M, Taury M. Der provozierte 555. Tomka I. Die funktionelle Beziehung zwischen Petit mal-
Mittagsschlaf: eine einfache erfolgreiche Methode zur Schlafak- Epilepsie und Schlaf. Z EEG-EMG. 1983;14:154–159.
tivierung bei Epileptikern. Z EEG-EMG. 1983;14:1–5. 556. Kellaway P, Frost JD Jr, Crawley JW. The relationship between
541. Gastaut H, Gomex-Almanzar M, Taury M. The enforced nap: a sleep spindles and spike-and-wave bursts in human epilepsy. In:
simple effective method of inducing sleep activation in epileptics. Avoli M, Gloor P, Kostopoulos G, et al., eds. Generalized Epilepsy.
In: Degen R, Niedermeyer E, eds. Epilepsy, Sleep and Sleep Boston, MA: Birkhauser; 1990:36–84.
Deprivation. Amsterdam: Elsevier; 1984:75–83. 557. Avoli M, Gloor P, Kostopoulos G, et al. An analysis of penicillin-
542. Niedermeyer E, Hšller L. Kurzschlaf im EEG-Eine Fundgrube sonst induced generalized spike and wave discharges using simultane-
tibersehener EEG-AbnormalitŠten. Z EEG-EMG. 1984;15: 57–66. ous recordings of cortical and thalamic single neurons. J
543. Binnie CD, Veldhuizen R, Beintema DJ. Evaluation of recording Neurophysiol. 1983;50:819–837.
after sleep deprivation in the diagnostic EEG assessment of epilepsy. 558. Gloor P, Avoli M, Kostopoulos G. Thalamocortical relation-
Electroencephalogr Clin Neurophysiol. 1982;54:21P–22P. Abstract. ships in generalized epilepsy with bilaterally synchronous
544. Degen R, Degen H-E. Sleep and sleep deprivation in epileptology. spike-and-wave discharge. In: Avoli M, Gloor P, Kostopoulos
In: Degen R, Niedermeyer E, eds. Epilepsy, Sleep and Sleep G, et al., eds. Generalized Epilepsy. Boston, MA: Birkhauser; 1990:
Deprivation. Amsterdam: Elsevier; 1984:273–286. 190–212.
545. Ellingson RJ, Wilken K, Bennett DR. Efficacy of sleep deprivation 559. Kostopoulos G, Gloor P, Pellegrini A, et al. A study of the tran-
as an activation and procedure in epilepsy patients. J Clin sition from spindles to spike and wave discharges in feline gen-
Neurophysiol. 1984;1:83–101. eralized penicillin epilepsy: EEG features. Exp Neurol. 1981;73:
546. Niedermeyer E. Generalized seizure discharges and possible pre- 43–54.
cipitating mechanisms. Epilepsia (Amsterdam). 1966;7:23–29. 560. Marciani MG, Gotman J, Andermann, F. Changes in interictal and
547. Niedermeyer E. Petit mal, primary generalized epilepsy and sleep. ictal activity during anticonvulsant withdrawal in epileptic patients.
In: Sterman MB, Shouse MN, Passouant P, eds. Sleep and Electroencephalogr Clin Neurophysiol. 1984;58:32P. Abstract.
Epilepsy. New York: Academic Press; 1982:191–207. 561. Marks DA, Katz A, Scheyer R, et al. Clinical and electrographic
548. Niedermeyer E. Awakening epilepsy (“Aufwach-Epilepsie”) revis- effects of acute anticonvulsant withdrawal in epileptic patients.
ited 30 years later. In: Degen SR, Niedermeyer E, eds. Epilepsy, Neurology. 1991;41:508–512.
Sleep and Sleep Deprivation. Amsterdam: Elsevier; 1984:85–94. 562. Van Parys JAP. The use of EEG as a method of control in epilepsy.
549. Halász P. Generalized epilepsy with spike-wave paroxysms as an Electroencephalogr Clin Neurophysiol. 1981;52:41P. Abstract.
epileptic disorder of the function of sleep promotion. Acta 563. Auvin S, Pandit F, De Bellecize J, et al. Benign myoclonic epilepsy
Physiol Acad Sci Hung. 1981;57:51–86. in infancy: electroclinical features and long-term follow-up of 34
550. Halász P. Sleep, arousal and electroclinical manifestations of gener- patients. Epilepsia. 2006;47:387–393.
alized epilepsy with spike wave pattern. In: Degen R, Niedermeyer 564. Blume WT, Luders HO, Mizrahi E, et al. Glossary of descriptive
E, eds. Epilepsy, Sleep and Sleep Deprivation. Amsterdam: Elsevier; terminology for ictal semiology: Report of the ILAE task force on
1984:97–107. classification and terminology. Epilepsia. 2001;42:1212–1218.
551. Halász P, Dévényi E. Petit mal absences in night sleep with special 565. Dura-Trave T, Yoldi-Petri ME, Gallinas-Victoriano F. Epilepsy in
reference to transitional sleep and REM periods. Acta Med Acad children in Navarre, Spain: epileptic seizure types and epileptic
Sci Hung. 1974;31:31–45. syndromes. J Child Neurol. 2007;22:823–828.
552. Nowack WJ. Dyshormia revisited: generalized seizures and 566. Ferrie CD, Caraballo R, Covanis A, et al. Autonomic status epilep-
arousal. Clin Electroencephalogr. 1996;27:22–25. ticus in Panayiotopoulos syndrome and other childhood and
553. Sato S, Dreifuss FE, Penry JK. The effect of sleep on spike wave adult epilepsies: a consensus view. Epilepsia. 2007;48:1165–1172.
discharges in absence seizures. Neurology (Minneapolis). 1973;23: 567. Legarda S, Jayakar P, Duchowny M, et al. High central and low
1335–1345. central subgroups. Epilepsia. 1994;35:1125–1129.
554. Passouant P, Cadilhac J. Décharges épileptiques. In: Niedermeyer 568. Legarda S, Jayakar P, Duchowny M, et al. Benign rolandic epilepsy:
E, ed. Epilepsy. Recent Views on Theory, Diagnosis and Therapy of high central and low central subgroups. Epilepsia. 1994;35:
Epilepsy. Basel: S. Karger; 1970:87–104. 1125–1129.
CHAPTER

Epilepsy in Adults and the Elderly


BERNARD S. CHANG, DONALD L. SCHOMER AND ERNST NIEDERMEYER 27
INTRODUCTION voltage activity may gradually increase in voltage with decreas-
ing frequencies. Ictal rhythmic activity may soon become pre-
It is difficult to imagine the practice of modern epileptology ponderant, similar to that found in the sudden change of
without the availability of electroencephalography (EEG), in all frequency. Arroyo et al. (1) have stressed the role of the frontal
of its various forms. The two fields have been closely inter- lobes in electrodecremental seizures. Electrodecremental
twined over the past decades, the latter being the objective seizure represents the most common EEG expression of infan-
accompaniment to the clinical art of the former. As some tile spasms occurring in the West syndrome (2,3).
epileptologists have remarked, “one feels that one knows a
patient a little better after seeing his EEG.” While electroen- Sudden Increase of Voltage
cephalography (EEG) clearly has many other important uses in
The classical example is the sudden steep rise of amplitude in a
neurologic practice (detailed throughout this volume), it has
classical petit mal absence with 3/sec spike waves. There is
revolutionized the entire field of epileptology and continues to
hence considerable variability in the ictal EEG events. This has
do so with each technical advance.
been quite discouraging for those concentrating on automatic
This chapter defines and describes the various EEG abnor-
devices for seizure detection. Even the eye of the experienced
malities seen in patients with epilepsy, including both those that
electroencephalographer may have difficulties in the determi-
occur during seizures and those that occur between seizures, as
nation of ictal episodes.
well as similar EEG findings that have less certain significance
and relationship to seizure disorders.
Ultraslow and Ultrafast Frequencies
These two aspects of paroxysmal activity are playing an increas-
PRINCIPAL DIFFERENCES BETWEEN
ing role in our understanding of epileptiform EEG activity, and
INTERICTAL AND ICTAL DISCHARGES are discussed separately within this volume.
Naturally, one would like a distinction to be made between ictal
and interictal paroxysmal activities. This logical differentiation, Problems of Terminology
however, is beset with great difficulties. It is sometimes difficult Definitions of EEG events are indispensable, but they are also
or even impossible to distinguish between ictal–clinical, quite difficult to arrive at and are often unsatisfactory. Attempts
ictal–subclinical, and interictal paroxysmal EEG activities, even to reassess the EEG terminology have been made from time to
with the use of neuropsychologic tests. We must acknowledge time; they often result in neologistic construction of terms that
the existence of a gray zone between interictal and ictal parox- fail to receive general acceptance. The EEG terminology is filled
ysmal activities. with widely used, popular, and, alas, often quite sloppy and
inaccurate terms.
Sudden Change of Frequency A preliminary proposal by the International Federation of
There are valid guidelines for the detection of true ictal events. Societies for Electroencephalography and Clinical Neuro -
The onset of a clinical seizure may be characterized by a sudden physiology (4) was followed by a definitive proposal (5) and a
change of frequency. A new type of rhythm appears, hesitantly, glossary of terms used by clinical electroencephalographers (6).
and then more distinctly; soon it boldly dominates the tracing. The handbook catalog of Dutertre (7) exudes Cartesian lucid-
This rhythm may be in alpha frequency or it may be faster or ity and deserves admiration. Unfortunately, the definition of
slower; it clearly demonstrates a new element of the tracing, electrophysiologic events can never do justice to all facets of the
indicative of a completely new electrophysiologic event. The phenomenon.
abnormal rhythm may or may not show spiky character. It As to epileptic discharges, there has been considerable dis-
tends to become slower with increasing amplitudes and more unity in the ranks of electroencephalographers. The term
distinct spiky phases of the rhythmic waves. epileptic discharge has been attacked and condemned on the
ground that such discharges may occur in the absence of clini-
Sudden Loss of Voltage cal seizure manifestations or in individuals who have never had
Sudden “desynchronization” of electrical activity is found in seizures. The same is true for the widely used term seizure dis-
electrodecremental seizures. The onset of these attacks may look charge; even paroxysmal discharge, certainly a more cautious
almost flat locally and/or diffusely, but extremely fast very low and unassuming term, has not found general acceptance.
541
542 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

INTERICTAL EPILEPTIFORM DISCHARGES nized and may be associated with enhancement and synchro-
nization within intracortical networks (10).
Spikes (Single or Random Spikes) On the basis of long-term monitoring and computer analy-
According to IFSECN (6), a spike is a transient, clearly distin- sis, Gotman (11) has raised the question of whether interictal
guished from the background activity, with pointed peak at spikes are in reality postictal. Gotman (11) and Gotman and
conventional paper speed or screen display and a duration from Marciani (12) observed marked and prolonged increase of
20 to under 70 msec; the main component is generally negative interictal spikes in a patient after a seizure (awake as well as
(Fig. 27.1). Amplitude is variable. asleep). Drug levels had little influence on the rate of spike
The distinction from the background activity is based on activity, and there was no increase of spiking prior to a seizure.
wave morphology and amplitude. In many cases, spikes stand The multiphasic character of a single spike warrants particu-
out against the background because of their high voltage. If the lar emphasis. A sequence of a minor positive, a major negative,
voltage of spikes and background activity is approximately and a second minor positive component (Fig. 27.1) is typical in
equal, the faster character (i.e., the shorter duration) of the most instances. A slow negative component may trail the spike
spike is its distinctive feature. It is possible that a spike of discharge and often attain about the same amplitude as the neg-
50-msec duration and moderate amplitude may be embedded ative main component of the spike. This trailing slow component
in 20/sec activity (50-msec wave duration), for instance, in a of a single spike should not be regarded as evidence of a spike-
person with epilepsy with considerable drug-induced fast activ- wave complex. Dutertre (7) in his handbook catalogue shows
ity. Under these circumstances, the spike activity may be unde- examples of single spike-wave complexes that one could just as
tectable. In such a case, a fast paper speed or widened screen well describe as compounded or multiphasic single spikes. From
display could demonstrate the morphologic features of the the vantage point of basic neurophysiology, there is some reason
spike (multiphasic and more pointed character with a domi- to presume that the trailing slow wave is caused by the same type
nant negative phase) in contrast with the more monotonous of hyperpolarization that was demonstrated by Pollen et al. (13)
appearance of the fast waves. in the experimental analysis of the spike-wave discharge.
Spikes have many characteristics in common and yet there The electroencephalographer very seldom finds single spikes
are also remarkable inter- and intraindividual variations and on the scalp with predominant positive component (14,15).
inconsistencies between one spike and the next in the same lead Positive single spikes are more common in depth recordings; on
(8). Spikes appear to be hypersynchronous events due to exces- the scalp, predominant positivity of single spikes raises the ques-
sive simultaneous neuronal discharge. This view, however, is tion of defective superficial cortical laminae. Following surgery
not congruent with the desynchronized (low-voltage) EEG of cortical arteriovenous malformations (AVMs) or after trau-
aspect of “electrodecremental seizures” or the electrode- matic laceration (16), such positive spikes may be present occa-
incremental initiation of focal or generalized attacks. sionally. A special type of positive spiking over the vertex has
Definitions such as “epilepsy is paroxysmal hypersynchrony” been described by Bergen (17) in a case that combined features
(9) may be correct on the neuronal level but partly incorrect of sphingolipidosis and mucopolysaccharidosis. (Also see the
from the viewpoint of the “macro-EEG.” work of Engel et al. (18) on the cherry-red spot-myoclonus syn-
Basic neurophysiologists have been cautioning against the drome.) With dipole formation (especially in benign rolandic
casual use of terms such as synchronization and desynchroniza- epilepsy), a positive spike discharge may be found in a moderate
tion. Low-voltage fast rhythms are not necessarily desynchro- distance from the negative spike focus (19,20).
Spikes represent the basic element of paroxysmal activity in
the EEG. A unitarian view that all spikes mean a hidden or overt
paroxysmal event would be erroneous. The fine semiology of
spikes is extremely important and the EEG interpreter ought to
consider the following questions:
1. What is the precise wave morphology?
2. Where do the spikes occur?
3. What is the patient’s age?
4. What is the state of awareness?
5. Is there any possibility of an artifact of similar appearance?
6. Is there any possibility of a physiologic potential of similar
appearance?
Discrepancies between spikes and behavioral epileptic events
Figure 27.1 Various wave morphologies of spikes recorded from the are not uncommon at all. This highly complex situation has
same patient (age 6 years). Upper lead, T3 T5; lower lead, T5 O1. been reinvestigated by Niedermeyer (21).
(First and second tracing from the top, continued in tracings 3 and 4,
respectively, 5 and 6.) Note multiphasic character of spikes with pre- Wave Morphology
dominance of the negative phase (see Chapter 8) and occasional forma- The largest and most pronounced spikes are not necessarily
tion of double spikes. associated with more serious epileptic seizure disorders. On the
Chapter 27 ■ Epilepsy in Adults and the Elderly 543

contrary, rolandic spikes in a child aged 4 to 10 are very promi-


nent; however, the seizure disorder is usually quite benign or
there may be no clinical seizures at all.
On the other hand, “small sharp spikes” are indeed marginal
spike discharges and will be discussed separately. A very
unusual type of rhythmically repetitive and yet interictal spike
activity in two epileptic brothers was described by Bauer and
Markoff (22).

Spatial Distribution
In childhood, occipital spikes are, in general, the most benign
spike discharges, with less than 50% having clinical seizures;
rolandic central–midtemporal–parietal spikes are also quite
benign, whereas frontal spikes or multifocal spikes are more
epileptogenic.

Age
The significance of the age factor is enormous. From the spikes
of an epileptic newborn to a seizure focus of old age, age-
determined varieties of spikes can be distinguished.

Level of Awareness
Random spikes may occur at any state of waking–sleeping cycle Figure 27.2 Coexistence of rolandic spikes and physiologic vertex
and occur even in rapid-eye-movement (REM) sleep when waves in light sleep (8-year-old boy, attacks of abdominal pain and no
bilateral-synchronous bursts of spikes or spike waves are usu- proven epileptic seizures). Right centroparietal spikes with occasional
ally suppressed. spread to the left are marked X; typical examples of a vertex wave are
Relationship to Perceptual Function marked O.

Shewmon and Erwin (23,24) investigated the impact of spikes


located over the occipital region and its vicinity on visual per- The physiologic nature of the occipital lambda waves and
ception and reaction time in three subjects with posterior “lambdoid” activity (positive occipital transients of sleep) must
spikes. Their work shows that the visuomotor reaction time was also be considered. The main component of this pattern is positive.
prolonged (or the response was absent) when the task was
spike-locked. Furthermore, “spike-induced dysfunction was Distinction from Artifacts of Similar Appearance
most pronounced when either response hand or visual field of This distinction depends on the electroencephalographer’s
stimulus was contralateral to the spike” (24). In a further study, experience and is usually an easy one.
Shewmon and Erwin (25) emphasized the significance of the
trailing slow wave that follows a single spike discharge. The per- Automatic Spike Detection
ceptive disturbance was particularly pronounced when the task Since the pioneering work of Gotman et al. (27) considerable
was presented during the slow wave. These fascinating findings controversy has arisen regarding computer-based automatic
indicate that even single spikes of rapid duration may be capa- detection of spikes in the EEG. The pivotal question is “What
ble of momentary impairment of the cortical visual function. should the computer imitate?” (28), and this question hinges on
This applies to visual tasks. During an ictal–subclinical episode the precise definitions of spikes (29): “A spike detected by any
of repetitive fast spiking over the occipital area and its neigh- reader is a spike, a spike detected by the majority of readers is a
borhood, correct mental arithmetic is possible (26). spike, a spike detected by the weighted average of readers is a
spike, etc.” According to these authors, average spike attributes
Distinction from Similar Physiologic Patterns are calculated, and the resulting database can serve as a “gold
This differentiation is particularly important in the case of ver- standard” for testing computer algorithms or other readers.
tex sharp waves during deep drowsiness and stage 2 of light With the use of an “off-line” artificial neural network, attempts
non-REM sleep. In childhood (after age 4), these waves may have been made at real-time detection of spikes (30).
have a particularly spiky appearance and may be misinterpreted
as paroxysmal spikes. Are Interictal Spikes Nothing but EEG Signals?
Physiologic sharp or spiky vertex waves are usually quite eas- Our earlier view (found in this chapter in previous editions)—
ily distinguished from rolandic spikes in sleep (Fig. 27.2). Even namely that single spikes are an EEG signal and nothing else—
in the more uncommon case of paroxysmal spike discharges appears to be in need of some correction. This seems to be
over the vertex, the differentiation from physiologic vertex particularly true for “negative myoclonus” (ictally suppressed
sharp waves is not difficult. motor activity). Beautiful examples have been presented by
544 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 27.3 Recurrent sharp wave recorded from a 61-year-old patient.


Upper lead, F3 C3; lower lead, C3 P3. A prominent negative phase is
followed by a large and relatively slow trailing positive wave.

Gambardella et al. (31) and Werhahn and Noachtar (32) (also


see Ref. 33). A single spike may also cause changes in cerebral
blood flow and increase of metabolic demands (34), demon-
strated with the simultaneous use of EEG and functional mag-
netic resonance imaging (fMRI). Even behavioral changes with
impaired test performance may be associated with single inter-
ictal spikes (35,36). Further information is found in the work of
Fisch (33).

Sharp Waves
According to IFSECN (6), a sharp wave is a transient, clearly
distinguished from background activity, with pointed peak at
conventional paper speed or screen display and duration of 70 Figure 27.4 An example of a very slow sharp wave (blunted sharp
to 200 msec, that is, more than approximately 1/14 to 1/5 sec- wave), maximal over T6, obtained from a 6-year-old boy with cranio-
onds. The main component is generally negative (Fig. 27.3). cerebral trauma at age 4 followed by seizures.
Jasper (37) pointed out that the rising phase of the sharp
wave is of the same order of magnitude as in spikes, but the
descending phase is prolonged. The configuration with a named “slow sharp waves” (41) or, in cases of particularly long
steeper ascending phase, however, is not always present. duration, “blunted sharp waves” (Fig. 27.4). Others become
A very unusual type of repetitive slow sharp wave activity very complex. They consist of a constantly varying number of
has been reported in prematurely born infants with intraven- components; such compounded sharp waves may occur as the
tricular hemorrhage; these discharges show predominantly periodic discharges (periodic lateralized epileptiform dis-
positive polarity and are recorded mainly over the rolandic charges [PLEDs]) of Chatrian et al. (42).
region (38,39). The maximum of this activity is sometimes It is certainly not incorrect to use the terms spikes and sharp
found over the vertex. waves synonymously when a local paroxysmal event is dis-
Spikes and sharp waves are neurophysiologically closely cussed, although purists of nomenclature would regard this as
related phenomena; both of them are typical paroxysmal dis- a breach of etiquette.
charges and highly suggestive of an epileptic seizure disorder,
although both phenomena may occur in patients without a his- Polyspikes or Multiple Spikes
tory of seizure disorder. This discharge type represents a complex of spikes and may also
Sharp waves are usually found as random focal discharges; be called polyspike complex. In modern terminology (6), the
most anterior temporal spikes are, in a strict sense, sharp term multiple spike complex is preferred on the grounds of lin-
waves. This is also true for most benign rolandic spikes of guistic considerations, because “polyspikes” is an etymological
childhood. Sharp waves are more seldom found in generalized- hybrid. It has been defined as a complex paroxysmal EEG pat-
synchronous bursts where single spikes, spike waves, and poly- tern with close association of two or more diphasic spikes
spikes predominate. occurring more or less rhythmically in bursts of variable dura-
It has been contemplated that sharp waves on the scalp cor- tion, generally with large amplitudes (Figs. 27.5 and 27.6) (7).
respond with spikes in the depth or on the cortex. Combined Polyspike bursts are readily elicited by electrical stimulation
depth and scalp recording clearly refutes this view (40). One can of single depth leads, especially in limbic regions. On the scalp,
detect spikes as well as sharp waves in depth recordings; a deep however, polyspikes occur mostly as bilateral- or generalized-
sharp wave usually corresponds with a sharp wave on the scalp synchronous discharges. Exceptional focal polyspikes are occa-
if there is any corresponding scalp activity at all. sionally encountered; these usually have a frontal maximum,
The long duration of a sharp wave permits better insight except for occipital accentuation in hypsarrhythmia. Polyspikes
into the multiphasic character of this potential. A small preced- and also polyspike-wave complexes are sometimes associated
ing positive component may be very fast and qualify as a spike; with concomitant myoclonus, especially in primary generalized
even a small biphasic spike discharge may precede the much epilepsy and in photosensitive individuals with this type of
larger sharp wave. Some sharp waves even exceed the maximum seizure disorder. Children with Lennox–Gastaut syndrome may
length of 200 msec of the IFSECN definition (6) (Fig. 27.4), also also show association of polyspikes and myoclonus.
Chapter 27 ■ Epilepsy in Adults and the Elderly 545

Figure 27.5 Polyspikes in light nonrapid-eye-movement (REM) sleep,


especially over central region, associated with K complexes. (Reproduced
with permission from AMA Archives of Neurology.)

Figure 27.7 A run of rapid spikes in a 19-year-old epileptic patient


Runs of Rapid Spikes (Lennox–Gastaut syndrome). Note anterior maximum of the discharge.
A few slow spike-wave complexes are also seen (right temporo-occipi-
This pattern has been described as “grand mal discharge”
tal region).
(43,44), “fast paroxysmal rhythms” (45), “rhythmic spikes” (46),
and “generalized paroxysmal fast activity” (47). It is seen only in
sleep and occurs in older children, adolescents, and younger discharge is very typical in patients with Lennox–Gastaut syn-
adults (Fig. 27.7). It consists of bursts of spike discharges at a drome; it is hardly ever found outside this clinical entity
rate from 10 to 25/sec, usually generalized but with a well- (50–52). It may be found in very rare instances of posttrau-
defined maximum over frontal regions; it may even be confined matic epilepsy with EEG traits of the Lennox–Gastaut syn-
to the frontal leads. The voltage is in the medium to high range, drome (53). Brenner and Atkinson (47), however, insist that
often exceeding 100 or even 200 V. The discharge rate is in runs of rapid spikes (“generalized paroxysmal fast activity”)
most cases somewhat irregular. The bursts last for about 2 to may be found outside the Lennox–Gastaut syndrome. There is
10 seconds; bursts of more than 5-second duration are usually evidence that runs of rapid spikes may occur in all “imitators”
associated with tonic seizures and thus represent an ictal pattern. of the Lennox–Gastaut syndrome (54).
The obvious misnomer “grand mal discharges” is based on We know little about the neurophysiologic underpinnings of
certain similarities with the ictal EEG of a generalized this pattern. It is clearly a frontal lobe discharge, and the associ-
tonic–clonic seizure. In a patient population with such seizures, ated tonic seizures are probably arising from the premotor and
runs of rapid spikes as an interictal pattern are a very rare find- supplementary motor portions of the frontal lobes. There could
ing. This has been confirmed by the data of Chayasirisobhon et be certain similarities to the decremental seizure patterns, which
al. (48). The inappropriateness of the term grand mal discharge also originate most commonly from the frontal lobe (1).
was stressed by Rodin et al. (49), who thought that this pattern Unfortunately, those electroencephalographers who never
occurred in patients with primary generalized epilepsy who suf- use sleep portions in routine records will never see this fascinat-
fered from more than one type of seizure and especially from ing pattern, which is definitely in need of further basic research.
akinetic seizures. As already noted by Gastaut et al. (46), this
Three-Per-Second Spike-Wave Complexes
Classical 3/sec spike-wave complexes are widely known even
outside the community of electroencephalographers. The dis-
charge is officially termed spike- and slow-wave complex (6); this
term comprises all types of spike-wave complexes, which are
listed separately because of markedly differing associated clini-
cal-epileptologic conditions. The official definition (6) is quite
simple: “A pattern consisting of a spike followed by a slow
wave.” Older terms such as spike-and-dome complex, wave-
and-spike complex, and dart-and-dome complex are seldom used
Figure 27.6 The differentiation between polyspikes and drug-induced these days and should be discouraged. The abridged term 3/sec
fast activity may be difficult. The run of beta waves (center) certainly falls spike wave is certainly acceptable, because the omitted word
into the category of fast activity, but the more outstanding and burst-like complex is automatically implied.
run (right) is more likely to represent polyspike activity. A definitive Spike-wave complexes of the classical variety (3/sec) have been
statement depends on the impressions derived from the entire record. described as the EEG pattern of petit mal absences (Fig. 27.8) (44).
546 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

studied with intracellular cortical microelectrodes (13,61,62).


According to Pollen (61), all components of the spike-wave com-
plex were the “consequence of postsynaptic activity.” This author
points out that “the initial positivity which may precede a nega-
tive spike (depending upon the intensity of the stimulation)
results from depolarization of middle and deep lying neurons,
whereas the surface negative spike is the result of EPSP [excita-
tory postsynaptic potential] generation primarily upon apical
dendrites of vertically oriented pyramidal type neurons.”
According to Pollen (61), the long surface negative wave is gener-
ated by inhibitory postsynaptic potentials (IPSPs) predominantly
on or near the soma of similar pyramidal cells. Thus, the slow
wave indeed appears to be related to inhibitory impulses.
From this vantage point, the spike-wave discharge apparently
represents an alternating succession of excitation and inhibition.
The clinical–ictal activities are thus constantly curbed by interven-
ing inhibitory impulses that prevent the attack from progressing
into massive downward discharges with motor effects (polyspikes
with massive myoclonus) or into a full-blown tonic–clonic attack.
Figure 27.8 Three-per-second generalized synchronous spike-wave
For this reason, motor manifestations of ictal episodes character-
complexes. There is a clear voltage maximum over the frontal regions
ized by spike waves are almost always inconspicuous or modest.
bilaterally. The spike component sometimes becomes less evident
Furthermore, spike-wave bursts rarely proceed into generalized
toward the end of a run of discharges, and there can be a slight slow-
convulsions; such rare exceptions have been demonstrated by
down of the usual frequency as well.
Halász (63) and Niedermeyer (64).
The distinction between classical (3/sec), slow (2 to 2.5/sec),
Since that time, the 3/sec spike-wave discharge has been equated and fast (4/sec) spike-wave complexes and the smaller 6/sec
with absence epilepsy. Regrettable mistakes have occurred due to spike-wave discharge is justified on the basis of different
lack of communication between electroencephalographers and clinical-epileptologic correlates of each spike-wave type; it is
clinicians who initiated treatment with anticonvulsants specific most definitely not an example of electroencephalographic hair
for absence seizures due to the use of the term 3/sec spike-wave splitting. The classical 3/sec spike-wave discharge is most typi-
complexes alone. The term petit mal discharge (43) has reinforced cal and most pronounced in children with absence seizures.
this type of reflex thinking. Fortunately, electroencephalographers Clinical absences are usually present when the burst lasts for
and epileptologists have always cautioned against the misinterpre- longer than 5 seconds. Thus, shorter bursts are usually subclin-
tation of the term petit mal discharges; this should be interpreted ical, but numerous psychophysiological attempts have been
as “discharges that occur in petit mal absences but also in other made to demonstrate certain fluctuations of level of awareness
conditions.” The works of Silverman (55), Clark and Knott (56), even in apparently subclinical spike-wave bursts. The classical
and Lundervold et al. (57) have clearly shown that the spike-wave spike-wave complex is, in most cases, easily activated by hyper-
discharge as an interictal event correlates with the occurrence of ventilation, whereas the slow and the fast forms of this dis-
absence seizures only in a moderate percentage of patients (16% charge show little or no enhancement.
according to Clark and Knott). A classical 3/sec spike-wave burst does not run exactly at a
The 3/sec spike-wave discharge is a worthwhile subject for rate of 3/sec. The complexes are faster at the onset of the burst
more detailed discussion. As to wave morphology, there is good (mostly around 4/sec), and then slow down to 3.5 and 3/sec for
evidence that the spike-wave complex is not simply an association the main portion, and eventually slow to 2.5/sec at the end of
of a spike and a slow wave, although the above-mentioned defini- the burst. During a burst, the spike discharges become gradu-
tion is justifiable as a simplification. The spike-wave complex con- ally smaller (Fig. 27.8), often shrinking to insignificance as in
tains hidden components that can be seen even with simple visual drowsiness and sleep.
analysis at 30 mm/sec paper speed or equivalent screen display; The spatial distribution of the bursts is very typical; the maxi-
special recording methods will reveal these extra components mum almost always lies over the frontal midline region, whereas
much better. Cohn (58) and Weir (59) have elucidated the com- the minimum is found over temporal and occipital areas.
ponents of this pattern. Most important is a spike of positive Because somewhat different findings obtained by Dondey (65)
polarity during the descending portion of the slow wave (59), were based on EEG recordings in which no midline electrodes
becoming evident as notching under usual recording conditions. were used, they do not invalidate the concept of a frontal mid-
On the basis of experimental studies of cortical faradization, line maximum, which has been confirmed by Rodin and
Jung and Toennies (60) stressed the inhibitory character of Ancheta (66) with the use of a brain mapping technique. The
the slow wave following a spike discharge (“Bremswelle”). understanding of this distribution type is highly conducive to a
Experimentally induced spike-wave-like activity in cats, elicited better comprehension of the underlying mechanisms. Almost
by electrical stimulation of thalamic intralaminar structures, was all spike-wave bursts are bilateral synchronous or generalized
Chapter 27 ■ Epilepsy in Adults and the Elderly 547

synchronous; the synchrony, however, is not perfect (58,67). The patients with primary generalized epilepsy and 3/sec spike-
age of distribution lies mainly in the range from 4 to 16 years. wave complexes produced motor evoked potentials of signifi-
The work of Lemieux and Blume (68) has shed more light cantly reduced size when the cortical stimulus was time-locked
on the spatial distribution of the spike-wave complex on the to the slow-wave component of the spike-wave complex (but
basis of computer analysis. According to these authors, field dis- slightly reduced or unchanged when the stimulus was time-
tribution of spikes differed from that of the ensuing slow waves. locked with the spike) (78). This underscores the special
Spikes “arose in one frontal region and propagated to the inhibitory character of the slow-wave component, which has
homologous part of the other frontal region with a peak-to- been known since the work of Jung and Toennies (60).
peak interval less than 15 msec. Less commonly, spikes first
moved anteriorly within the initiating frontal region before Slow Spike-Wave Complexes
contralateral propagation occurred.” By contrast, negative slow After the demonstration of the 3/sec spike-wave complex and
waves were more diffuse, more symmetrical in evolution, and its relationship to the petit mal absence (44), the Gibbses and
more posteriorly centered than the spikes. William G. Lennox were struck by the occurrence of slow spike-
The onset of the spike component in one frontal lobe is con- wave complexes in a much different type of patients with
gruent with the earlier findings of Cohn (58) and Lüders et al. seizures other than absence. The classical 3/sec spike-wave com-
(69), who noted asynchronies ranging from 5 to 20 msec. The plex was termed petit mal discharge. Consequently, the slow
frontal lobe is a large structure and subdivisions are necessary. spike-wave complex was termed petit mal variant discharge.
Onset of spike-wave activity (and, in particular, of the spike This term was used first by Gibbs et al. (79). In that study, hypo-
component) over the frontal midline means in reality that a glycemia was induced in order to demonstrate an increase of
frontal portion in the immediate vicinity of the interhemi- classical 3/sec spike-wave activity in the hypoglycemic state; an
spheric fissure is the origin of the discharge; this area belongs to increase of slow spike-wave activity, however, was not elicited
the supplementary motor zone. with this activation method when used in appropriate patients.
As to the neuropsychological correlates of subclinical general- The wave morphology of this pattern varies considerably. In
ized spike-wave bursts and clinical absences with spike waves, it the majority of the cases, the complex consists of a rather slow
should be pointed out that the impact of apparently subclinical spike (according to the definition, a sharp wave, lasting 70 msec
3/sec spike-wave bursts might be stronger than one would expect or longer) and a slow wave. In a sizable number of cases, true
from routine clinical observation. With its maximum over the spikes (60 msec or less duration) are followed by a slow wave
frontal midline (frontal lobe close to interhemispheric fissure), (Fig. 27.9).
there might be some impact on prefrontal lobe functions and, in The spatial distribution is, in most instances, quite similar to
particular, on working memory. The concept of working mem- that of the 3/sec spike-wave complex. In the vast majority of the
ory, introduced by Baddeley (70) and enormously refined by cases, the bursts are bilateral or generalized synchronous (with
Fuster (71,72), implies a system of constant perceptory afferences imperfect synchrony); a frontal midline maximum is the rule in
to the prefrontal dorsolateral region whence motor impulses these discharges. Lateralization or occasional focal occurrence
emanate, resulting in readiness (set) for motor action. This sys- is sometimes observed, usually in children with severe residual
tem could be blocked for a moment by a few spike-wave com- brain damage in certain areas where parenchymatous destruc-
plexes (or even by a single one) and interfere with working tion abolishes the spike-wave discharge.
memory. In a clinical 3/sec spike-wave absence, blocking of
working memory might be the most likely explanation for the
mild disturbance of consciousness and its immediate full recov-
ery with the termination of the attack (73). No “true disturbance
of consciousness”—epileptic or nonepileptic—can recover
immediately. In other words, the function of working memory is
simply suspended for the duration of the spike-wave complexes.
Due to the uniqueness of primary generalized epilepsy with
3/sec spike-wave complexes in the human (74), animal models
for this response have great limitations. The arrest reaction of
the monkey (75) produced by alumina cream injection into the
fronto-orbital cortex has some resemblance to the human
absence epilepsy. However, this cannot be said about the sudden
freeze of motility noted in the cat stimulation of the cat’s mesial
anterior thalamus (76), since there is no spike-wave activity
associated with this type of arrest reaction.
According to Sperling and Skolnick (77), there is a general
decrease of cerebral blood flow (by 133 xenon method) during
human generalized spike-wave activity, which, however, is less Figure 27.9 Generalized slow spike-wave complexes (mostly around
pronounced in frontal regions than in the parietal lobes. 2/ sec) in a child with severe epileptic seizure disorder (Lennox–Gastaut
Transcranial magnetic stimulation of the motor cortex in syndrome).
548 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Generalized slow spike-wave discharges are often quite pro- observed transition from the former to the latter with deepening
longed. In some children or adolescents, the entire sleep por- drowsiness. Marshall (83) introduced the term wave and spike
tion (light and moderately deep non-REM sleep) consists of phantom, which has found only limited acceptance.
unabated generalized slow spike-wave activity. The diagnosis of The clinical correlates of 6/sec spike-wave complex have been
an “electrical status epilepticus” may or may not be justified in elucidated by Thomas (84), Gibbs and Gibbs (85), Hughes et al.
such cases; there are usually no behavioral or polygraphic char- (86), and Thomas and Klass (87). It is usually a pattern of adult-
acteristics to suggest an ongoing ictal event. In some cases, the hood but may also occur in adolescents and children. About
slow spike-wave activity may be found only in non-REM sleep. 50% to 60% of the patients suffer from clear epileptic seizures
Although the classical 3/sec spike-wave complex is seldom (mostly generalized tonic–clonic); the remainder show a history
seen before the age of 4 and almost never before 3.5 years, its of syncopal attacks, posttraumatic states, or (as emphasized by
slow counterpart appears much earlier, sometimes before the Small (87)) psychiatric problems.
age of 6 months. At this early age, the frontal maximum may The 6/sec spike-wave complex is an uncommon but not rare
not be readily demonstrable. pattern (about 0.5% to 1% in a central EEG laboratory). The
The slow spike-wave complex is almost always associated discharge may be recorded in waking state, drowsiness, and
with a severe and often uncontrollable type of childhood light non-REM sleep; light drowsiness appears to be the opti-
epilepsy (seldom with onset between ages 11 and 20 years) mal recording condition (Fig. 27.10).
called Lennox–Gastaut syndrome (46,50,52,79,80). Most of Investigations regarding the spatial distribution of this pat-
these children show a variety of minor attacks and evidence of tern have led to an important dichotomy. Hecker et al. (88)
mental retardation. pointed out that a distinction must be made between frontal
Thus, the distinction between slow (1 to 2.5/sec) and classical accentuation and occipital accentuation. We are evidently deal-
(3/sec) spike waves is of great clinical significance. One has to ing with two different types of discharge; the frontal type is
keep in mind, however, that many children or adolescents with most commonly associated with epileptic seizure disorders and
slow spike waves also show series of 3/sec or even 4/sec spike- sometimes found in combination with other paroxysmal dis-
wave complexes; the presence of these spike-wave types is obvi- charge types. The occipital types are found predominantly in
ously of no significance in such cases and what counts is the slow patients with no evidence of epileptic seizure disorder.
type. On the other hand, patients with classical 3/sec spike-wave In a study of 839 patients with 6/sec spike-wave complexes,
complexes and a usually benign type of epileptic seizure disor- Hughes (89) placed particular emphasis on the distinction
der almost never show slow spike waves except when a 3 or 4/sec between frontal and occipital maximum. He summarized the
spike-wave burst may slow down to 2.5/sec at the end.

Fast Spike-Wave Complexes


This pattern is closely related to the classical 3/sec spike-wave
complex; both discharge types are most commonly lumped
together. A differentiation, however, is justified on clinical
grounds.
Gastaut (81) deserves credit for an outline of the distinctive
features between the 3 and the 4/sec (or 4 to 5/sec) spike-wave
discharge. According to his investigations, the fast spike-wave
burst is usually of shorter duration (1 to 3 seconds), it occurs
in patients older than 15 years, the bursts are always subclinical,
and the associated seizure disorder is usually characterized by
myoclonic jerking, grand mal attacks, or a combination of both
seizure types, whereas petit mal absences are quite uncommon.
Paroxysmal flicker responses are common in such patients. A
positive family history of epileptic seizures disorder is frequently
found in this group of patients. The 4/sec spike-wave discharge is
spatially distributed in the same manner as the 3/sec spike-wave
discharge; the frontal midline maximum is quite prominent.

Six-Per-Second Spike-Wave Complexes


This is a marginal paroxysmal pattern, described by Gibbs and
Gibbs (43) as follows: “The spike in this pattern has a strong
positive component but the entire wave complex looks in gen-
eral like a miniature reproduction of the 3 per second spike and Figure 27.10 A short run of 6/ sec spike waves, posterior type,
wave of petit mal.” In view of the positive component of the recorded in a 52-year-old woman with a history of head injury 2 years
spike, Silverman (82) has stressed similarities to the 6/sec com- earlier and subsequent headache, dizziness, and memory loss. There
ponent of the 14 and 6/sec positive spike discharge; he also was computed tomography (CT) scan evidence of cortical atrophy.
Chapter 27 ■ Epilepsy in Adults and the Elderly 549

principal features of both forms by the acronyms WHAM discharges in only 0.6%. Gibbs and Gibbs (85) found 6/sec
and FOLD: spike waves in 1.3% to 2.0% of 3476 adults, peaking in the
range from 20 to 24 years, but were unable to detect any focal
W: waking record F: females
spikes or sharp waves. Focal sharp activity was reported in 1%
H: high in amplitude O: occipital
of the material of Blanc et al. (99). The reports of Lennox-
A: anterior L: low in amplitude
Buchthal et al. (104) and Kitamura and Asakura (105) show a
M: males D: drowsy record
relatively high incidence of spikes (6.4% and 4%, respectively).
An interesting sidelight of this observation is the uneven sex Zivin and Ajmone Marsan (106) and Chatrian (107) have
distribution, which is rather unique; no other EEG abnormali- contemplated the clinical significance of spikes in healthy per-
ties show any gender preference. According to Hughes (90), sons. Above all, the interpretation must take into consideration
patients with anterior 6/sec spike waves are more likely to have age. In childhood, the occurrence of central–midtemporal (also
epileptic seizures, and those with posterior discharges tend to parietal) spikes is associated with overt seizures in only 50% to
have neuroautonomic disturbances. 70% of the cases; this pertains mainly to the age from 3 to 12
Kocher et al. (91) were struck by the occurrence of 6/sec years. In occipital spikes (mainly age 3 to 5 years), the epilepto-
spike-wave complexes in the abstinence or withdrawal phase of genicity is even lower. Major studies on this subject based on
drug-dependent individuals. Tharp (92) produced the dis- large healthy populations were performed by Nekhorocheff
charge by intravenous diphenhydramine (50 mg) in normal (108), Kellaway and Fox (109), Brandt and Brandt (110), Corbin
subjects. Hecker et al. (88) found that the occipital form of the and Bickford (111), Trojaborg (112), Eeg-Olofsson et al. (113),
6/sec spike-wave complex is often related to drug dependence and Cavazzuti et al. (114). In general, “benign” focal spikes (such
(hypoanalgesics and barbiturates) and withdrawal. All this as seen in benign rolandic epilepsy) outnumber generalized-
underscores the need for a distinction of the two forms. Thus, synchronous bursts of spikes or spike waves by a slight to con-
the frontal form will have to be regarded as a truly epilepsy- siderable margin. Cavazzuti et al. obtained EEG recordings of
related paroxysmal pattern, whereas the occipital form finds 3726 children from 6 to 13 years of age without evidence of
itself on the fringe of paroxysmal discharge types. seizure or neurologic deficits. Paroxysmal discharges were found
in 131 cases (3.52%). Generalized, mainly polyspike, discharges
were found in 41 cases, midtemporal spikes in 50, centroparietal
INTERICTAL EPILEPTIFORM DISCHARGES spikes in 27, occipital spikes in 2, and bilateral (midtemporal or
IN OTHERWISE HEALTHY INDIVIDUALS parietal) spikes in 11 children. Change of spike localization
occurred in but a few subjects; shifts from focal to generalized
The occurrence of focal or generalized paroxysmal discharges spiking and vice versa were also rare. The work of Cavazzuti et
in apparently healthy individuals is a puzzling and even annoy- al. is particularly important because it includes follow-up peri-
ing finding that requires some discussion. The EEG evaluation ods of up to 9 years. In most children, the abnormalities disap-
of comparatively large healthy populations usually shows a cer- peared, usually during elementary school age or in early
tain percentage of abnormalities. The work of Thorner (93) adolescence. Only seven children developed clinical epileptic
showed paroxysmal discharges in 0.3% of 1100 flying cadets; seizures—five had generalized discharges, one had midtempo-
Gibbs et al. (94) found epileptic abnormalities in 0.9% of nor- ral, and one had centroparietal spikes. In two subjects there was
mal adult controls, whereas Harty et al. (95) observed such evidence of epileptic seizures in the family, while six children
abnormalities in 3% of candidates for medical service. “Larval had siblings with spikes, either generalized or rolandic.
epileptic disturbances” were noted in 9% of healthy controls by Both generalized-synchronous (spike wave and polyspike
Williams (96). The tumult of World War II was apparently con- wave) and rolandic (centroparieto-midtemporal) spikes in chil-
ducive to such large-scale studies, which have become more dren without epilepsy suggest a genetic predisposition if no neuro-
scarce over the past decades. logic deficit and no history of insult to the central nervous system
A study of 682 Air Force applicants showed paroxysmal (CNS) are present. In children with a history of cerebral palsy and
changes in 2.6%; about one fourth were focal and the rest were with no seizures but prominent spiking, the spike activity may
of nonfocal character (97). Further studies of this kind were herald future epileptic seizures (85). Even in perfectly healthy chil-
carried out in commercial flying personnel (98), candidates for dren with spikes, the possibility of future seizures cannot be com-
commercial airplane pilot training (99), and air cadets (100). pletely ruled out, although the chances are slim.
Some studies concentrated on the classical 3/sec spike-wave
pattern, which was found in 0.2% of 3070 normal controls (94)
and 2.7% in the subjects of Hill (101). Spike-wave complexes EPILEPTIFORM EEG FEATURES
were found in 0.9% to 1.5% of the groups studied by Dell and OF UNCERTAIN SIGNIFICANCE
Dell (98), in 1% of the subjects of Blanc et al. (99), and in 0.5% of
O’Connor’s subjects (100). No spike-wave complexes were found Small Sharp Spikes, or Benign Epileptiform
in the normative study carried out for the National Aeronautics Transients of Sleep (BETS)
and Space Administration by Baylor University in Houston, This pattern has been delineated by Gibbs and Gibbs (43,85). It
Texas (102). Bennett (103) evaluated the EEG of military and is the most inconspicuous paroxysmal discharge and hence is eas-
civil flying personnel (1332 persons) and found spike-wave ily overlooked. (Only the occipital type of the 6/sec spike-wave
550 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

et al. (120), Klass and Westmoreland (121), and Gutrecht (122)


have stressed the nonspecific nature of this pattern and do not
regard its occurrence as an abnormality. By contrast, Low et al.
(123) as well as Hughes and Gruener (124) have pointed out
that small sharp spikes are not a normal or meaningless pattern.
The data of these authors clearly show that this pattern indi-
cates “a moderate degree of epileptogenicity” (124). This has
been strongly confirmed by Saito et al. (125) and also by Molaie
et al. (126).

Needle-Like Occipital Spikes of the Blind


Spike discharges of a particularly fast and needle-like character
develop over the occipital region in most congenitally blind chil-
dren (127–129). These spikes appear to be completely innocu-
ous, are unrelated to epileptic seizure disorders, and may be due
to a state of functional deafferentation (130). The discharges dis-
appear during childhood or adolescence. The experimental basis
of visual deafferentation spikes, however, is controversial (131).
Congenitally blind children with a history of retrolental
Figure 27.11 Small sharp spikes (51-year-old patient). Note the subtle
fibroplasia often show evidence of accompanying cerebral
character and moderate voltage of the discharge; also note its predom-
impairment. Such children may show abundant and wide-
inance in the left nasopharyngeal lead. There is evidence of spread into
spread but chiefly occipital spike activity; concomitant clinical
T3, as well as into the right nasopharyngeal lead. The left section was
epileptic seizures are fairly common in this condition (85,132).
recorded in the waking state (transition to earliest drowsiness); the mid-
This type of spike activity must be differentiated from transient
dle and right sections were recorded in sleep. (Reproduced with permis-
occipital spiking in nonepileptic congenitally blind children.
sion from Clinical Electroencephalography.)
Fourteen-and-Six Positive Bursts
complex is equally small and unimpressive.) The main negative This discharge pattern has been defined as follows (6): “Bursts
and positive components are of about equally spiky character of arch shaped waves at 13–17 Hz and/or 5–7 Hz, most com-
(Fig. 27.11). The discharge is fairly widespread and is seen chiefly monly at 14 and/or 6 Hz, seen generally over the posterior tem-
over temporal and frontal areas, either shifting from side to side poral and adjacent areas of one or both sides of the head during
or synchronously firing. The pattern is almost exclusively found in sleep. The sharp peaks of its component are positive with
drowsiness and/or light non-REM sleep. It is essentially a pattern respect to other regions.” It is also pointed out in this definition
of adulthood with a peak between ages 30 and 60 years; occur- that the amplitudes are generally below 75 V, that the pattern
rence in adolescence and old age is somewhat less common. It is is best demonstrated by referential recording using contralat-
virtually absent in the first 10 years of life. This pattern is also eral ear lobes or other remote reference leads, and that the clin-
known as benign epileptiform transients of sleep (BETS). ical significance is controversial (Fig. 27.12). The synonymous
Koshino and Niedermeyer (115) found a prevalence of
1.36% with a peak of 2.9% between ages 30 and 40 years. In this
study, two thirds (67.4%) of the patients had a history of epilep-
tic seizures; this underscores the paroxysmal character of the
discharge. Small sharp spikes sometimes are precursors of typ-
ical anterior temporal spikes or sharp waves, which, in such
patients, simply occur somewhat later in drowsiness or sleep.
This association was also noted by Gibbs and Gibbs (85). In a
depth EEG study by Westmoreland et al. (116), an extensive
generator area of small sharp spikes was found.
Among those without epilepsy, the discharges may occur in
patients with cerebrovascular disorder, syncopal attacks, and
psychiatric problems. Small (87) and Small et al. (117) have
placed special emphasis on the occurrence of this spike dis-
charge in a psychiatric population and especially in patients
with manic-depressive illness. Figure 27.12 Various examples of 14/ sec and 6/ sec positive spikes
It is interesting to note that Gibbs and Gibbs (85) found a (underlined). Note posterior predominance for this pattern and shifting
rather high prevalence of small sharp spikes in presumed asymmetrics. Also note the sometimes blurred distinction between the
normal adult control subjects, reaching 7.9% in the range from 14 and 6 components, due to notch formation. The recording was
40 to 49 years. Reiher and Klass (118), Lebel et al. (119), White obtained from a 12-year-old patient; montages to ipsilateral ear.
Chapter 27 ■ Epilepsy in Adults and the Elderly 551

term ctenoids was introduced by Lombroso et al. (133) with Psychomotor Variant (Rhythmic Midtemporal
regard to the comb-like shape. The 14 and the 6 component Theta Discharges)
may be observed independently. According to Gibbs and Gibbs This pattern is widely known as “psychomotor variant dis-
(85), the 6 component prevails in early childhood and adult- charge” (43,85) because of certain similarities to rhythmic ictal
hood, while the 14 component is more prominent in older chil- activity occurring in psychomotor seizures (temporal lobe
dren and adolescents. Hughes (134,135) emphasized the seizures and complex partial seizures). This term has been dis-
harmonic character of the major frequencies and preferred the couraged by IFSECN (6), which recommended the term rhyth-
term 14 and 7/sec positive spikes. mic theta bursts of drowsiness. The term rhythmic midtemporal
This pattern was described first by Gibbs and Gibbs (136) discharge of Lipman and Hughes (158) is also widely used.
and regarded as “evidence of thalamic and hypothalamic The pattern consists of long runs of rhythmic activity in the
epilepsy.” The following 15 years produced exciting correlations range of 5 to 6.5/sec with a maximum over the midtemporal
to autonomic nervous system dysfunctions and behavioral dis- region, often with considerable spread into posterior temporal,
orders (2,137–147). Enthusiasm flagged when it became clear anterior temporal, and occipital areas (Fig. 27.13). The theta
that this pattern is often found in healthy individuals (133,148). activity shows a well-defined negative sharp component that
Little (149) even presumed that this pattern “is more likely to be apparently stresses the paroxysmal character of the discharge.
a sign of health than of disease.” These trains of sharp theta waves may occur in a unilateral
The “14 and 6 pattern” may be confined to the regions lying shifting, bilateral shifting, or synchronous distribution type.
beneath a skull defect (150) and hence could denote a “hidden The duration of a single run usually exceeds 10 seconds and
pattern” that is not detectable from the scalp unless it reaches may reach 1 minute or more. Very often, the first run is noted
major proportions. in early drowsiness; in deep drowsiness and light sleep, the pat-
The pattern occurs most commonly in children after age 4 tern tends to disappear. According to Egli et al. (159), patients
and adolescents and declines in adulthood; Gibbs and Gibbs with rhythmic theta bursts show shortened periods of REM
(85,151) recorded the 14 and 6 activity even in infants. Its sleep and also, to a lesser degree, of slow sleep in nocturnal sleep
occurrence in the waking state is exceptional; deep drowsiness studies at the expense of long drowsy periods with the rhythmic
and very light non-REM sleep are usually the ideal states for the theta pattern. Hughes (160) has emphasized that the rhythmic
documentation of the pattern, although deep sleep might be midtemporal discharge is not limited by the drowsy state and
more conducive in very young children (85). Yürüker and may be very active in wakefulness.
Menzi (152) elicited the 14 and 6 pattern with auditory stimuli The pattern occurs mainly in younger or middle-aged
presented in light sleep. Okada et al. (153) noted 14 and 6/sec adults; it is also seen in adolescents and children. It is a rare
positive spikes in association with simultaneous negative spikes pattern; Egli et al. (159) found a prevalence of 0.1% in a large
over the frontal area (suggestive of dipole formation). Depth laboratory.
recordings at the thalamic as well as the basal ganglia level (154) The clinical significance is not clear. Despite its strongly
have shown this neurophysiologic discharge, and it has been paroxysmal appearance, its epileptogenic properties seem to be
repeatedly recorded in the amygdaloid complex as well. very low; most patients have no history of clinical seizures,
Not surprisingly, the prevalence of the pattern appears to although Lipman and Hughes (158) found seizures in 36% of
increase with the recorded length of sleep. Much of the contro- their patients. Personality disorders and some autonomic nerv-
versy about this pattern has arisen from differences of record- ous system dysfunctions are common in patients with this pat-
ing techniques and the use of routine sleep tracings and, in tern (85,158,161). This view is shared by Eeg-Olofsson and
particular, the length of the sleep portion. Proven cases of
epileptic seizure disorder very seldom show 14 and 6/sec posi-
tive spikes as the only significant finding, and it appears to be a
marginal pattern of no or very little epileptologic significance.
The demonstration of 14 and 6/sec positive spikes in
advanced states of metabolic encephalopathies, especially in
hepatic coma (155), is an interesting exception. For completely
unknown reasons, this essentially innocuous discharge may
appear when slow activity becomes extremely pronounced in
such patients. A similar observation was reported by Ford and
Freeman (156). Drury (157) reported 121 children with hepatic
coma and mostly in association with Reye syndrome. Seven of
these children showed 14 and 6/sec positive spikes, occurring in
an almost periodic manner, in four children activated by stim-
ulation. In all of these cases, there was a background of severe
EEG abnormality. Repeat tracings failed to show any 14 and 6
discharges (with one exception). The “14 and 6” pattern may Figure 27.13 Psychomotor variant discharge of the right midtemporal
also occur as a transient toxic effect of diphenhydramine region (T4). Tracing was obtained from a 21-year-old woman. The
hydrochloride. record had low-voltage character; this necessitated the use of high gain.
552 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Petersén (1982; cited in Ref. 162), who also studied the genetic unique because of the beautiful cinematographic demonstration
transmission of this pattern and wondered about possible auto- of clinical and EEG features. Further work on ictal EEG patterns
somal dominant inheritance. was presented by Ralston (174), Ralston and Papatheodorou
(175), Anziska and Cracco (176), Geiger and Harner (177), and
Subclinical Rhythmic EEG Discharge Blume et al. (178,179).
of Adults (SREDA)
This pattern was originally described by Naquet et al. (163) as a Generalized Tonic–Clonic Seizures
“paroxysmal discharge of the temporo-parietooccipital junc- The ictal EEG is invariably obscured by muscle artifact; it is
tion,” facilitated or triggered by temporary hypoxic conditions. In demonstrable only in patients treated with neuromuscular block-
spite of certain paroxysmal features and its rhythmic sharp char- ers while on mechanical ventilation. Fast rhythmic spikes are pres-
acter, the possibility of an epileptic (ictal) event has been strongly ent in all leads, with some accentuation in upper frontal leads.
de-emphasized. Herranz and Lopez (164) have described this This fast spike activity characterizes the tonic stage and becomes
pattern as “subclinical paroxysmal activity” in 31 patients and discontinuous in the clonic stage. Rhythmic slow waves alternate
stressed its rarity; it has an incidence of 0.02% to 0.045%. with bursts of polyspikes synchronously with clonic jerks (poly-
Westmoreland and Klass (165,166), and Miller et al. (167) spikes) and brief relaxation (slow bursts). A period of postictal
deserve particular credit for the elucidation of this discharge electrical silence is usually fairly brief, but a longer phase of very
type. These authors disagree with the view of Naquet et al. (163) irregular slow activity follows during the ensuing minutes.
concerning the major role of cerebrovascular insufficiency and
hypoxia, although the average age of their patients was 61 years. Absence Seizures
The term “SREDA” has been proposed by Miller et al. (167). Typified by the generalized-synchronous 3/sec spike-wave com-
This pattern occurs mainly in the waking state or in light plex occurring in more prolonged bursts, absence seizures have
drowsiness. The onset may be fairly abrupt with widespread been discussed in the section “Three-Per-Second Spike-Wave
sharp rhythmic theta (4 to 7/sec) and occasionally with delta Complexes.”
activity. In other cases, the onset is gradual with a few single
sharp discharges that increasingly develop rhythmic character, Complex Partial Seizures
first at delta, and then at theta frequency. As to the spatial dis- The ictal EEG patterns of these seizures are “as variable as the
tribution, a maximum of this discharge is usually found over clinical features” (170). The astounding variability of the ictal
the centroparietal region and especially over the vertex. patterns is impressively demonstrated in the atlas of Oller-
Hyperventilation may induce this pattern. Daurella and Oller-Ferrer-Vidal (173). In early work of Gibbs et
In a “revisitation” of the SREDA pattern, O’Brien et al. (168) al. (180,181), the ictal EEG activity of complex partial seizures
analyzed this pattern in seven recent patients and only one of was described as a special type of seizure discharge characterized
them showed evidence of cerebrovascular disorder. by bursts of serrated slow waves, flat-topped 4/sec waves, and
high-voltage 6/sec waves (“seizure discharge of the psychomotor
ICTAL EEG PATTERNS type”). Much greater complexities of the ictal patterns were
stressed by Gastaut and Vigouroux (182) and Klass (183). In
Ictal EEG patterns may be a prolongation of a well-defined about 5% of the cases, a complex partial seizure may occur with-
interictal pattern that becomes ictal by virtue of its long dura- out any recordable ictal EEG activity (170).
tion (this augmenting impact on neuronal function) or com-
pletely different from preceding interictal discharges. A typical Focal Motor and Other Simple Partial Seizures
example of the former case is the absence EEG activity with Cortical focal (partial) seizures are expected to be associated
short 3/sec spike-wave bursts as interictal and prolonged 3/sec with a local discharge consisting of a sequence of repetitive
spike-wave bursts as ictal phenomena. The emergence of a spikes over the area contralateral to the motor or sensory (visual
completely different ictal pattern is exemplified by partial forms and auditory) clinical manifestations. This is correct in a large
of seizures (e.g., by cases of temporal lobe epilepsy with typical number of cases but does not pertain to all cases. According to
anterior temporal sharp waves in the interval and the sudden Gastaut and Broughton (170), there is no demonstrable ictal
appearance of completely different patterns during a seizure). EEG discharge in about 30% of the cases. This is particularly
Ictal patterns may be clinical, with the typical motor and true for circumscript motor rolandic attacks; ictal discharges
behavioral changes of a seizure of whatever category, or sub- arising from the precentral gyrus may be quite subtle and incon-
clinical with no demonstrable motor or behavioral changes. In spicuous or not demonstrable at all in the EEG (43).
such clinically silent ictal episodes, neuropsychological studies In cases with a well-documented EEG correlate of the
may demonstrate some changes. seizure, the attack is often initiated by local desynchronization,
The following discussion of the ictal EEG patterns is cursory. that is, very fast and very low voltage spiky activity, which grad-
Special studies and atlases on epileptic seizures and their EEG ually rises in amplitude with diminishing frequency (177).
correlates have been presented by Gibbs and Gibbs (43), Ajmone
Marsan and Ralston (169), Gastaut and Broughton (170), Myoclonic Seizures
Karbowski (171,172), and Oller-Daurella and Oller-Ferrer-Vidal Myoclonus is a very complex phenomenon; a considerable
(173). The atlas of Oller-Daurella and Oller-Ferrer-Vidal is number of disorders with myoclonus do not fall into the epileptic
Chapter 27 ■ Epilepsy in Adults and the Elderly 553

category (184). Epileptic myoclonus is classically characterized character of interictal seizure discharges (spikes, sharp wave,
by concomitant polyspikes or polyspike-wave discharges in the etc.). There is no satisfactory model to explain the periodicity of
EEG, of bilateral- or generalized-synchronous character, usu- the discharges.
ally with maximum over the frontal regions. A review of Periodic discharges are always of large amplitude, mostly in
myoclonus and its relationship to epilepsy has been presented the range of 100 to 300 V. These may be simple sharp waves,
by Hallett (185). but of a duration that usually exceeds 150 msec. Other periodic
discharges are compounded and polymorphic. Periodic dis-
Tonic Seizures charges may be focal, widely scattered, or generalized synchro-
The relationship between tonic seizures and massive fast spike nous. A fine tabulated review of periodic discharges has been
activity was discussed previously in this chapter. This pattern is presented by Spehlmann (188).
the typical correlate of tonic attacks occurring in patients with
the Lennox–Gastaut syndrome. Subacute Sclerosing Panencephalitis (SSPE)
Some tonic seizures are characterized by simple flattening or The periodic complexes of SSPE dominate the second state of
desynchronization of all activity during the attack (170). These this disease; this is a prolonged phase in which the clinical
authors also describe rhythmic activity around 10/sec and a diagnosis is usually made. The SSPE discharge has become
very rare diffuse slow-wave pattern (mainly delta frequency) as exceptionally rare in developed countries due to its relation-
EEG concomitants of tonic seizures. ship with measles and the excellent preventive effect of measles
immunization. This type of complex discharge almost never
Atonic Seizures occurs in other clinical conditions and is hence almost disease-
These seizures are customarily divided into a short form, such specific. It is a very reliable and highly contributory diagnostic
as simple drop attack, lasting only seconds, and a longer form, finding in this disease. These discharges were first mentioned
lasting minutes and described as “inhibitory.” The EEG shows a by Balthazar (189) and extensively described by Radermecker
few polyspike waves or spike waves of generalized distribution (190,191) and Cobb and Hill (192). The discharges show a
followed by large slow waves (short forms). More rhythmic duration from 0.5 to 3 seconds and are formed by two or more
spike activity around 10/sec and intermixed slow activity in all waves with mean amplitude of 500 V (100 to 1000 V)
leads constitute the EEG correlate of the longer lasting attacks; (Fig. 27.14) (193). In some cases, the voltage may even reach
rhythmic slow spike-wave activity (1.5 to 2/sec) may also occur 1400 V (8). The enormous height of this discharge may
(170,173). impress the inexperienced as a movement artifact; this idea is
supported by frequent associated motor events such as
Akinetic Seizures myoclonus or sudden loss of tone.
These attacks are characterized by arrest of all motion, which, The periodicity becomes manifest as the disease progresses;
however, is not caused by sudden loss of tone as in atonic in its earlier stages, which are usually dominated by diffuse 1 to
seizures. The patients, usually children, are in an absence-like 3/sec activity of fairly rhythmic character, the compounded dis-
state; the EEG correlate is a slow (mostly 1 to 2/sec) spike-wave charge may be aperiodic. Sometimes, the periodic discharge is
discharge, generalized synchronous, and often with clocklike present in the earliest stage of the disease and may persist to the
rhythmicity. This type of seizure is rather poorly understood fatal outcome; more often, this pattern disappears in the termi-
and, in recent years, the use of this term has been discouraged. nal or third stage. Vitrai et al. (194) investigated the SSPE com-
plex with the use of time functions of the auto- and
cross-correlation coefficients.
PERIODIC EPILEPTIFORM DISCHARGES The elements of the discharge are variable; a giant slow wave
ASSOCIATED WITH PARTICULAR is usually mixed with several sharp waves. The discharge is gen-
NEUROLOGIC DISORDERS eralized, with a maximum over the frontocentral areas and ver-
tex. The frequency of the complexes ranges from 4 to 16/min
Some rhythmically (or nearly rhythmically) recurring dis- (193). The discharge is more impressive in the waking state.
charges or patterns are of paroxysmal character, but are usually Accompanying myoclonus is fairly synchronous with the dis-
not associated with epileptic seizure disorders characterized by charge; the motor activity may precede or trail the discharge in
chronically recurrent seizures. Periodic discharges or periodic a range of 200 to 800 msec (195); this author showed minor
activities are most commonly an important EEG feature of a interhemispheric asynchronies (15 msec) and an earlier appear-
severe ongoing CNS disease with certain paroxysmal or even ance in parieto-occipital areas than in frontocentral areas,
overt epileptogenic properties. They are hence disease- where the discharge shows its most impressive voltage.
suggestive and sometimes virtually disease-specific, rather than The origin of the periodic SSPE discharge is controversial.
suggestive of epileptic seizure disorders in general. A review of Radermecker and Poser (196) have proposed a deep thalamic or
periodic activities was presented by Gaches (186), as well as by mesencephalic-reticular origin; this view has been supported by
Bauer and Pieber (187). Cobb (197,198), who performed depth EEG recordings, and by
The periodic character of these patterns in the presence of Lombroso (199), who studied direct current (DC) potential
severe CNS involvement remains enigmatic; such rhythmic changes during the periodic discharge. Other authors favor the
firing is markedly different from the predominantly random idea of a cortical origin (200–202).
554 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 27.14 Periodic discharges in subacute sclerosing panencephalitis (SSPE). A: At an earlier state with some preser-
vation of other activities. B: At a more advanced state (recorded from two different patients, aged 8 and 9 years).

Herpes Simplex Encephalitis charges may also occur. Predominantly occipital localization of
In this extremely severe but not invariably fatal necrotizing the periodic discharges has been reported by Bergey et al. (215).
encephalitis, slow repetitive spikes have been noted by In contrast with the complexes found in SSPE, the periodic
Radermecker (191). These observations were subsequently con- discharge of herpes simplex encephalitis is of short periodicity,
firmed by Millar and Coey (203), Perier et al. (204), Carmon et with the greatest intervals of 4 seconds or less; according to
al. (205), Rawls et al. (206), Adams and Jennett (207), Upton Gaches (186), this criterion is always met in the herpes simplex
and Gumpert (208), Gaches and Arfel (209), Elian (210), Kugler discharge. In some cases, the periodic discharge can be detected
et al. (211), and Cobb (212). The discharge was also found in only when almost daily repeat records are carried out. This
experimental disease inoculated rabbits by Gupta and Seth explains the fact that periodic discharges are not reported in all
(213). cases of herpes simplex encephalitis.
In an earlier stage, local mostly unilateral temporal poly-
morphic delta waves are the most striking feature, but soon Creutzfeldt–Jakob Disease
large sharp waves emerge over the most affected region. These The clinical and neuropathologic features of this rapidly pro-
discharges usually fire at intervals from 2 to 4 seconds. The dis- gressive dementing illness were first established by the work of
charge may be quite slow and exceed 1000 msec; consider that Creutzfeldt (216), Jakob (217), and Kirschbaum (218,219); the
a sharp wave is defined as having a duration from 70 to much later identification of pathogenic prion proteins and a
150 msec. The amplitudes are in the range from 100 to about variant form of the disorder has only occurred in recent
500 V. Similar periodic sharp discharges have been demon- decades.
strated in neonatal herpes simplex encephalitis (214). In the In the most common, sporadic form of CJD, a typical peri-
course of the disease, the originally regional discharge tends to odic discharge can be seen on EEG, consisting of a sharp wave
become generalized synchronous; asynchronous bilateral dis- or a sharp triphasic complex of 100- to 300-msec duration with
Chapter 27 ■ Epilepsy in Adults and the Elderly 555

PERIODIC LATERALIZED EPILEPTIFORM


DISCHARGES (PLEDS)
In the wake of earlier work (226–231), Chatrian et al. (42) gave
an extensive account of periodic lateralized or focal discharge
that may occur in a variety of acute neurologic conditions. This
pattern is most often associated with acute cerebral infarctions
but may also occur in neoplastic and inflammatory pathology.
Periodic discharges occurring in herpes simplex encephalitis
are discussed earlier in this chapter. PLED activity, however,
may also occur in other types of encephalitis—among others, in
infectious mononucleosis encephalitis, according to Aminoff et
al. (232). Markand and Daly (233), Dauben and Adams (234),
and Bauer et al. (235) presented further important work on this
pattern, which also plays a major role in experimental cerebral
embolism (236).
PLEDs may be simple and large sharp waves or complex
(compounded) discharges with mixed spiky and slower ele-
ments. The amplitudes usually lie around 100 to 300 V, but
occasionally may be much higher. The firing rate may be as fast
as 3/sec or as slow as 12/min (Fig. 27.16) (42).
The discharge is found over the maximally involved area,
and the local background activity is almost always severely dis-
Figure 27.15 Periodic discharges in Creutzfeldt–Jakob disease. Note ordered. The term PLED implies lateralization. It is possible,
mild lateralization of the recurrent sharp waves to the left. The 68-year- however, that PLED activity is generated independently over
old patient was demented, with psychotic features; there were long tract both hemispheres. De la Paz and Brenner (237) have investi-
signs, fasciculation, and frequent myoclonus. Myoclonic jerking did not gated the bilateral independent type of PLEDs referred to as
alter the tracing. BIPLEDs. Although strokes were found to be the leading cause
in the PLED group, CNS infection and epileptic seizure disor-
ders predominated in the group with BIPLEDs. Bilateral-
a repetition rate of 0.5 to 2/sec or intervals of 500 to 2000 msec. synchronous (generalized-synchronous) periodic activity
The discharges occur against a severely disordered background should not be listed as BIPLEDs in view of the original defini-
of activity, mostly in generalized synchrony. Some cases show tion of Chatrian et al. (42), but is often termed generalized peri-
unilateral onset with discharges over one hemisphere or lateral- odic epileptiform discharges (GPEDs). The term cerebral
ized to one hemisphere for several days or weeks (220). bigeminy has been used by Aldrich and Pugh (1984) for bilater-
According to Tariska et al. (221), the periodic discharges may ally independent PLED activity (238). Even TRI-PLEDs, arising
even remain unilateral in the course of the disease. from three different areas, have been reported (left frontal, left
The periodic activity usually shows a maximum over the posterotemporal, and right posterotemporal) in an 85-year-old
anterior region except for the Heidenhain form, which has a patient with renal encephalopathy (239).
posterior maximum (Fig. 27.15); in this special form, cortical PLEDs are often associated with simultaneous focal motor
blindness is a common feature. In sleep, the periodic dis- twitching in contralateral face or fingers, hand, arm, leg, foot,
charges tend to disappear. The absence of periodic discharges etc. This underscores the paroxysmal character of the pattern. It
after 10 weeks of illness argues against sporadic is usually a temporary pattern that changes into other abnor-
Creutzfeldt–Jakob disease as a diagnosis (222). The EEG stud- malities within 1 to 2 days. Patients with PLEDs are in most
ies of Chiofalo et al. (223) are based on the observation of 27 cases acutely ill and often show a history of a variety of mixed
cases. The differential diagnosis of the combined clinical and medical problems, such as cerebral arteriosclerosis plus renal
electroencephalographic picture of this disease can be quite insufficiency or chronic alcoholism or diabetes mellitus.
complex (224). Myoclonus is a typical clinical feature of this Patients with PLEDs are commonly older adults, although
disease, but the periodic patterns may or may not be associated this pattern occasionally occurs in young adults and children
with myoclonus (184). (240). Andriola (241) noted PLEDs as well as BIPLEDs in
It has to be emphasized that the periodic discharges of 12 children with various types of acute CNS disease; those with
Creutzfeldt–Jakob disease (i) do not occur in every sporadic bilateral activity (BIPLED) expired in the acute state. Ritaccio
case of this disease (even with numerous repeat recordings) and March (242) reported the association of BIPLEDs with
(225); (ii) are most common in the sporadic form and much complex partial status epilepticus. Silbert et al. (243) described
less commonly, if ever, seen in the familial, iatrogenic, and vari- another variant of PLEDs with independent ipsilateral periodic
ant forms of CJD; and (iii) may occasionally be found in other lateralized discharges. A clinical curiosity appeared to be the
disorders. observation of Chabolla et al. (244): a young man suffering
556 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

neuronal injury, the patient’s preexisting propensity to have


seizures, and the coexistence of any acute metabolic derange-
ments all contributing to whether seizures occur or not.”
Pohlmann-Eden et al. have strongly reemphasized the domi-
nant etiologic role of strokes in the generation of PLEDs. From
the evidence of functional neuroimaging studies, these authors
feel that PLEDs “might reflect a key pattern for focal hyperex-
citability in the penumbra zone of ischemic stroke.”
A nonlateralized variant of PLEDs is periodic epileptiform
discharges occurring in the midline (PEDIM), as cited by
Westmoreland et al. (247). These midline discharges are pre-
sumed to arise from vascular watershed areas, between the
anterior and middle and between the middle and posterior
cerebral arteries.

REFERENCES
1. Arroyo S, Lesser RP, Fisher RS, et al. Clinical and electroencephalo-
graphic evidence for sites of origin of seizures with diffuse elec-
trodecremental pattern. Epilepsia. 1994;35:974–987.
2. Kellaway P, Crawley JW, Kagawa N. A specific electroencephalo-
graphic correlate of convulsive equivalent disorders in children.
J Pediatr. 1959;55:582–592.
3. Ohtahara S, Yamatogi Y. Evolution of seizures and EEG abnormal-
ities in childhood onset epilepsy. In: Wada JA, Ellington RJ, eds.
Handbook of Electroencephalography and Clinical Neurophysiology.
Revised series. Vol 4. Amsterdam: Elsevier; 1990:457–477.
4. IFSECN. Preliminary proposal for an EEG terminology by the ter-
minology committee of the International Federation for
Electroencephalography and Clinical Neurophysiology.
Electroencephalogr Clin Neurophysiol. 1961;13:646–650.
5. IFSECN. Proposal for an EEG terminology committee of the
International Federation for Electroencephalography and Clinical
Neurophysiology. Electroencephalogr Clin Neurophysiol. 1966;30:
306–310.
6. IFSECN. A glossary of terms commonly used by clinical electroen-
cephalographers. Electroencephalogr Clin Neurophysiol. 1974;37:
538–548.
Figure 27.16 A: Periodic lateralized epileptiform discharge (PLED) in 7. Dutertre F. Catalogue of the main EEG patterns. In: Remond A,
a 70-year-old patient, widespread over left hemisphere with left editor-in-chief. Handbook of Electroencephalography and Clinical
midtemporal maximum. Note poorly developed background of activity. Neurophysiology. Vol 11A. Amsterdam: Elsevier; 1977:40–79.
Cerebrovascular accident associated with twitching of right arm. Patient 8. Kiloh LG, McComas AJ, Osselton JW. Clinical Electroencephalography.
3rd ed. London: Butterworth; 1972.
lethargic and obtunded. B: PLED over right central–midtemporal
9. Roger H, Cornil L, Paillas JE. Les Epilepsies. Paris: Flammarion;
region, sometimes also involving the right parietal area. Note well-
1949.
preserved physiologic activities of waking and light drowsiness. Also 10. Steriade M, Amzica F, Contreras D. Synchronization of fast
note the complex and multiphasic character of the discharge. Patient is (30–40 Hz) spontaneous cortical rhythms during brain attention.
68 years old with cerebrovascular disease with right centroparietal low- J Neurosci. 1996;16:392–417.
density area in CT scan (infarction). 11. Gotman J. Could interictal epileptic spikes actually be postictal
epileptic spikes? Electroencephalogr Clin Neurophysiol. 1984;58:9P
from multiple sclerosis (MS) with exacerbations ushered in by [abstract].
complex partial status epilepticus associated with an enhancing 12. Gotman J, Marciani MG. Electroencephalographic spiking activ-
right frontal MRI lesion and PLEDs in the EEG. Another report ity, drug levels and seizure occurrence in epileptic patients. Ann
Neurol. 1985;17:597–603.
on PLEDs in MS (right temporal PLEDs and MRI lesion) has
13. Pollen DA, Reid KH, Perol P. Micro-electrode studies of experi-
been presented by Gandelman-Marton et al. (245). mental wave and spike in the cat. Electroencephalogr Clin
A very thoughtful study of PLEDs deserves special attention: Neurophysiol. 1964;17:57–67.
a critical review of Pohlmann-Eden et al. (246). These authors 14. Matsuo F, Knott JR. Focal positive spikes in electroencephalogra-
consider PLEDs as “an EEG signature of a dynamic pathophys- phy. Electroencephalogr Clin Neurophysiol. 1977;42:15–25.
iologic state in which unstable neurobiologic processes create 15. Maulsby RL. Some guidelines for assessment of spikes and sharp
an ictal–interictal continuum, with the nature of the underlying waves in EEG tracings. Am J EEG Technol. 1971;11:3–16.
Chapter 27 ■ Epilepsy in Adults and the Elderly 557

16. Binnie CD, Polkey CE, Spencer S. Positive spikes over a cortical 38. Cukier F, André, N., Monod N, et al. EEG contribution to
laceration. Electroencephalogr Clin Neurophysiol. 1989;72:106P diagnosing intraventricular haemorrhage in premature
[abstract]. infants. Electroencephalogr Clin Neurophysiol. 1974;36:840
17. Bergen D. A unique EEG pattern in mucolipidosis. Electro- [abstract].
encephalogr Clin Neurophysiol. 1979;46:80. 39. Werner SS, Stockard JE, Bickford RG. Atlas of Neonatal
18. Engel J Jr, Rapin I, Giblin DR. Electrophysiological studies in two Electroencephalography. New York: Raven Press; 1977.
patients with cherry red spot-myoclonus syndrome. Epilepsia 40. Zobniw AM, Yaworth S, Niedermeyer E. Depth electroencephalo-
(New York). 1977;18:73–87. graphy. J Electrophysiol Technol. 1975;2:215–240.
19. Gregory DL, Wong PK. Topographical analysis of the centrotem- 41. Ribeiro M, Niedermyer E, Hertz S. Slow sharp waves. Clin
poral discharges in benign rolandic epilepsy of childhood. Electroencephalogr. 1999;30:114–117.
Epilepsia (New York). 1984;25:705–711. 42. Chatrian GE, Shaw CM, Leffman H. The significance of periodic
20. Wong PKH, Gregory D. Dipole fields in rolandic discharges. Am J epileptiform discharges in EEG: an electrographic, clinical and
EEG Technol. 1988;28:243–250. pathological study. Electroencephalogr Clin Neurophysiol.
21. Niedermeyer E. Clinical relevance of EEG signals in epilepsies. In: 1964;17:177–193.
Wada JA, Ellingson RJ, eds. Handbook of Electroencephalography 43. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 2.
and Clinical Neurophysiology. Revised series. Vol 4. Amsterdam: Cambridge, MA: Addison-Wesley; 1952.
Elsevier; 1990:237–261. 44. Gibbs FA, Davis H, Lennox WG. The electroencephalogram in
22. Bauer G, Markoff R. Familienuntersuchungen bei einem seltenen epilepsy and in conditions of impaired consciousness. Arch Neurol
paroxysmalen Muster. Z EEG-EMG. 1970;1:19–26. Psychiatr (Chicago). 1935;34:1133–1148.
23. Shewmon DA, Erwin RJ. The effect of focal interictal spikes on 45. Jasper HH, Kershman J. Classification of the E.E.G. in epilepsy.
perception and reaction time. I. General considerations. Electroencephalogr Clin Neurophysiol (suppl). 1949;2:123–131.
Electroencephalogr Clin Neurophysiol. 1988;69:319–337. 46. Gastaut H, Roger J, Soulayrol R, et al. Childhood epileptic
24. Shewmon DA, Erwin RJ. The effect of focal interictal spikes on encephalopathy with diffuse slow spike-waves (otherwise known
perception and reaction time. II. Neuroanatomic specificity. as “petit mal variant”) or Lennox syndrome. Epilepsia (Amsterdam).
Electroencephalogr Clin Neurophysiol. 1988;69:338–352. 1966;7:139–179.
25. Shewmon DA, Erwin RJ. Focal spike-induced cerebral dysfunction 47. Brenner RP, Atkinson R. Generalized paroxysmal fast activity: elec-
is related to the after-coming slow wave. Ann Neurol. 1988;23: troencephalographic and clinical features. Ann Neurol. 1982;11:
131–137. 386–390.
26. Niedermeyer E, Krauss GL, Pacer CE. The electroencephalogram 48. Chayasirisobhon S, Cullis P, Sack R, et al. Grand mal discharge.
and mental activation. Clin Electroencephalogr. 1989;20:215–227. Clin Electroencephalogr. 1984;15:155–158.
27. Gotman J, Gloor P, Schaul N. Comparison of traditional reading of 49. Rodin E, Smid N, Mason K. The grand mal pattern of Gibbs, Gibbs
the EEG and automatic recognition of interictal epileptic activity. and Lennox. Electroencephalogr Clin Neurophysiol. 1976;40:
Electroencephalogr Clin Neurophysiol. 1978;44:48–60. 401–406.
28. Webber WRS, Litt B, Lesser RP, et al. Automatic spike detection: 50. Niedermeyer E. The Lennox–Gastaut syndrome: a severe type of
what should the computer imitate? Electroencephalogr Clin childhood epilepsy. Dtsch Z Nervenheilk. 1969;195:263–283.
Neurophysiol. 1993;87:364–373. 51. Niedermeyer E. The Generalized Epilepsies. Springfield, IL: Charles
29. Wilson SB, Harner RN, Duffy FH, et al. Spike detection. I. C Thomas; 1972.
Correlation and reliability of human experts. Electroencephalogr 52. Niedermeyer E. Compendium of the Epilepsies. Springfield, IL:
Clin Neurophysiol. 1996;98:186–198. Charles C Thomas; 1974.
30. Webber WRS, Litt B, Wilson K, et al. Practical detection of epilep- 53. Niedermeyer E, Walker AE, Burton C. The slow spike-wave com-
tiform discharges (EDs) in the EEG using an artificial neural net- plex as a correlate of frontal and fronto-temporal posttraumatic
work: a comparison of raw and parameterized EEG data. epilepsy. Eur Neurol (Basel). 1970;3:330–346.
Electroencephalogr Clin Neurophysiol. 1994;91:194–204. 54. Niedermeyer E. The electroencephalogram in the differential diag-
31. Gambardella A, Aguglia U, Oliveri RI, et al. Negative myoclonic nosis of the Lennox–Gastaut syndrome. In: Niedermeyer E, Degen
status due to antiepileptic drug tapering: report of three cases. R, eds. The Lennox–Gastaut Syndrome. New York: Alan R. Liss;
Epilepsia. 1997;38:819–823. 1988:177–220.
32. Werhahn KJ, Noachtar S. Epileptic negative myoclonus. In: 55. Silverman D. Clinical correlates of the spike-wave complex.
Lueders HO, Noachtar S, eds. Epileptic Seizures: Pathophysiology Electroencephalogr Clin Neurophysiol. 1954;6:663–669.
and Clinical Semiology. New York: Churchill Livingstone; 56. Clark EC, Knott JR. Paroxysmal wave and spike activity and diag-
2000:473–483. nostic sub-classification. Electroencephalogr Clin Neurophysiol.
33. Fisch BJ. Interictal epileptiform activity: diagnostic and behavioral 1955;7:161–164.
implications. J Clin Neurophysiol. 2003;20:155–162. 57. Lundervold A, Henriksen GF, Fegersten L. The spike wave com-
34. Warach S, Ives JR, Schlaug G, et al. EEG-triggered echo-planar plex. A clinical correlation. Electroencephalogr Clin Neurophysiol.
functional MRI in epilepsy. Neurology. 1996;47:89–93. 1959;11:13–22.
35. Aarts JHP, Binnie CD, Smit AM, et al. Selective cognitive impair- 58. Cohn R. Spike–dome complex in the human electroencephalo-
ment during focal and generalized epileptiform EEG activity. gram. Arch Neurol Psychiatr (Chicago). 1954;71:699–706.
Brain. 1984;107:293–308. 59. Weir B. The morphology of the spike-wave complex. Electro-
36. Hughes JR. The significance of interictal spike discharge: a review. encephalogr Clin Neurophysiol. 1965;19:284–290.
Clin Electroencephalogr. 1997;28:60 [abstract]. 60. Jung R, Toennies F. Über Entstehung and Erhaltung von
37. Jasper HH. Epilepsy. In: Penfield W, Erickson TC, eds. Epilepsy and Krampfentladungen. Die Vorgänge im Reizort und die
Cerebral Localization. Springfield, IL: Charles C Thomas; Krampffähigkeit des Gehirns. Arch Psychiatr Nervenkr.
1941:380–454. 1950;1985:701–735.
558 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

61. Pollen DA. Intracellular studies of cortical neurons during thala- 86. Hughes JR, Schlagenhauff RE, Magoss M. Electro-clinical corre-
mic induced wave and spike. Electroencephalogr Clin Neurophysiol. lations in the six per second spike and wave complex.
1964;17:398–406. Electroencephalogr Clin Neurophysiol. 1965;18:71–77.
62. Weir B, Sie PG. Extracellular unit activity in cat during the spike- 87. Thomas JE, Klass DW. Six-per-second spike and wave pattern in
wave complex. Epilepsia (Amsterdam). 1966;7:30–43. the electroencephalogram: a reappraisal of clinical significance.
63. Halász P. The generalized epileptic spike-wave mechanism and the Neurology. 1968;18:587–593.
sleep–wakefulness system. Acta Physiol Acad Scient Hung. 1972;42: 88. Hecker A, Kocher R, Ladewig D, et al. Das Minatur-Spike-Wave-
293–314. Muster. Das EEG Labor. 1979;1:51–56.
64. Niedermeyer E. Immediate transition from a petit mal absence 89. Hughes JR. Two forms of the 6/sec spike and wave complex.
into a grand mal seizure. Eur Neurol (Basel). 1976;14:11–16. Electroencephalogr Clin Neurophysiol. 1980;48:535–550.
65. Dondey M. Transverse topographical analysis of petit mal 90. Hughes JR. Changes in the annual incidence of the 6/sec spike
discharges: diagnostical and pathogenic implications. and wave pattern. Clin Electroencephalogr. 1991;22:71–74.
Electroencephalogr Clin Neurophysiol. 1983;55:361–371. 91. Kocher R, Scollo-Lavizzari G, Ladewig D. Miniature-spikewave-
66. Rodin E, Ancheta O. Cerebral electrical fields during petit mal Muster: Elektroencephalographisches Korrelat in der
absences. Electroencephalogr Clin Neurophysiol. 1987;66:457–466. Abstinenzphase bei Medikamentenabhängigkeit. Z EEG-EMG.
67. Gotman J. Interhemispheric relations during bilateral spike-and- 1975;6:78–82.
wave activity. Epilepsia (New York). 1981;22:453–466. 92. Tharp BR. The 6-per-second spike and wave complex. Arch
68. Lemieux JF, Blume WT. Topographical evolution of the spike-wave Neurol (Chicago). 1966;15:533–537.
complexes. Electroencephalogr Clin Neurophysiol. 1983;56:30P 93. Thorner MW. Procurement of Electroencephalograph Tracings in
[abstract]. 1000 Flying Cadets for Evaluating the Gibbs Technique in Relation
69. Lüders H, Daube J, Johnson R, et al. Computer analysis of general- to Flying Ability. USAD School of Aviation Medical Research
ized spike-and-wave complexes. Epilepsia (New York). 1980;21:183 Report, No. 7–1; 1942.
[abstract]. 94. Gibbs FA, Gibbs EL, Lennox WG. Electroencephalographic clas-
70. Baddeley AD. Working Memory. Oxford: University Press; 1986. sification of epileptic patients and control subjects. Arch Neurol
71. Fuster JM. Memory and planning. In: Jasper HH, Riggio S, Patricia Psychiatr. 1943;50:111–128.
Goldman-Rakic PS, eds. Epilepsy and the Functional Anatomy of 95. Harty JE, Gibbs EG, Gibbs FA. Electroencephalographic study of
the Frontal Lobe. New York: Raven Press; 1995:9–19. two hundred and seventy-five candidates for military service.
72. Fuster JM. Memory in the Cerebral Cortex. Cambridge, MA: MIT War Med (Chicago). 1942;2:923–930.
Press; 1996. 96. Williams D. The nature of transient outbursts in the electroen-
73. Niedermeyer E. Frontal iobe functions and dysfunctions. Presented cephalogram of epileptics. Brain. 1944;67:10–37.
at Symposium, Lima, Peru, 1997. 97. Buchthal F, Lennox M. The EEG effect of metrazol and photic
74. Niedermeyer E. Primary (idiopathic) generalized epilepsy. Clin stimulation in 682 normal subjects. Electroencephalogr Clin
Electroencephalogr. 1996;27:1–21. Neurophysiol. 1953;5:545–558.
75. Walker A, Morello G. Experimental petit mal. Trans Am Neurol 98. Dell AR, Dell MB. L’électroencéphalogramme systématique du
Assoc. 1967;57–61. personnel navigant. Étude préliminaire portant sur 1056 cas. Méd
76. Hunter J, Jasper HH. Effects of thalamic stimulation in unanes- Aéro. 1958;13:33–47.
thetized animals. Electroencephalogr Clin Neurophysiol. 1949;1: 99. Blanc C, Lafontaine E, Laplane R. Les activités EEG paroxystiques
303–324. généralisées ou focales chez les adultes non comitiaux: Études
77. Sperling MR, Skolnick BE. Cerebral blood flow during spike-wave évolutive correlations longitudinales. Rev Neurol (Paris). 1964;111:
discharges. Epilepsia. 1995;36:156–163. 362–369.
78. Gianelli M, Cantello R, Civardi C, et al. Idiopathic generalized 100. O’Connor P. Analysis of 500 routine EEGs of R.A.F. aircrew
epilepsy: magnetic stimulation of motor cortex time-locked and cadets. Electroencephalogr Clin Neurophysiol. 1964;17:341
unlocked to 3-Hz spike-and-wave discharges. Epilepsia. 1994;35: [abstract].
53–60. 101. Hill D. EEG in episodic psychotic and psychopathic behaviour. A
79. Gibbs FA, Gibbs EL, Lennox WG. The influence of blood sugar classification of data. Electroencephalogr Clin Neurophysiol.
level on the wave and spike formation in petit mal epilepsy. Arch 1952;4:419–442.
Neurol Psychiatr (Chicago). 1939;50:111–128. 102. Baylor University College of Medicine and the Methodist
80. Lennox WG. Epilepsy and Related Disorders. Vol 1. Boston, MA: Hospital. The Normative Electroencephalographic Data Reference
Little, Brown & Co.; 1960. Library. Final Report. Washington, DC: National Aeronautics and
81. Gastaut H. Clinical and electroencephalographic correlates of gen- Space Administration; 1966.
eralized spike and wave bursts occurring spontaneously in man. 103. Bennett DR. Spike-wave complexes in “normal” flying personnel.
Epilepsia (Amsterdam). 1968;9:179–184. Aerospace Med. 1967;38:1276–1282.
82. Silverman D. Phantom spike-wave and the fourteen and six per 104. Lennox-Buchthal M, Buchthal F, Rosenfalck P. Correlation of
second positive spike pattern: a consideration of their relationship. electroencephalographic findings with crash rate of military jet
Electroencephalogr Clin Neurophysiol. 1967;23:203–217. pilots. Epilepsia (Amsterdam). 1959;1:366–373.
83. Marshall C. Some clinical correlates of the wave and spike phan- 105. Kitamura K, Asakura T. Clinical significance of spikes and sharp
tom. Electroencephalogr Clin Neurophysiol. 1955;7:633–636. waves in EEG. Electroencephalogr Clin Neurophysiol (suppl). 1958;
84. Thomas JE. A rare electroencephalographic pattern: the six per second 12:16
spike and wave discharge. Neurology (Minneapolis). 1957;7:438–442. 106. Zivin L, Ajmone Marsan C. Incidence and prognostic significance
85. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 3. of “epileptiform” activity in the EEG of nonepileptic subjects.
Reading, MA: Addison-Wesley; 1964. Brain. 1968;91:751–777.
Chapter 27 ■ Epilepsy in Adults and the Elderly 559

107. Chatrian GE. Paroxysmal pattern in “normal” subjects. In: Remond 129. Lairy GC, Harrison A, Leger EM. Foyers EEG biocciptaux asyn-
A, editor-in-chief. Handbook of Electroencephalography and Clinical chrones de pointes chez l’enfant mal voyant et aveugle d’âge sco-
Neurophysiology. Vol 6A. Amsterdam: Elsevier; 1976:114–123. laire. Rev Neurol (Paris). 1964;111:351–353.
108. Nekhorocheff MI. La stimulation lumineuse intermittante chez 130. Cannon WB, Rosenblueth A. The Supersensitivity of Denervated
l’enfant normal. Rev Neurol (Paris). 1950;83:601–602. Structures: A Law of Denervation. New York: Macmillan; 1949.
109. Kellaway P, Fox BJ. Electroencephalographic diagnosis of cerebral 131. Kellaway P, Bloxsom A, MacGregor M. Occipital spike foci associ-
pathology in infants during sleep. I. Rationale, technique and the ated with retrolental fibroplasia and other forms of retinal loss in
characteristics of normal sleep in infants. J Pediatr. 1952;41: children. Electroencephalogr Clin Neurophysiol. 1955;7:469–470.
262–287. 132. Gibbs EL, Fois A, Gibbs FA. The electroencephalogram in retro-
110. Brandt S, Brandt H. The electroencephalographic patterns in lental fibroplasia. N Engl J Med. 1955;253:1102–1106.
young healthy children from 0 to five years of life. Acta Psychiatr 133. Lombroso CT, Schwarts IH, Clark DM, et al. Ctenoids in healthy
Scand. 1955;30:77–89. youth. Controlled study of 14 and 6-per-second positive spiking.
111. Corbin HPF, Bickford RG. Studies of the electroencephalogram Neurology (Minneapolis). 1966;16:1152–1158.
of normal children: comparison of visual and automatic fre- 134. Hughes JR. The 14 and 7 per second positive spikes—a reap-
quency analysis. Electroencephalogr Clin Neurophysiol. 1955; praisal following a frequency count. Electroencephalogr Clin
7:15–28. Neurophysiol. 1960;12:495–496.
112. Trojaborg W. Focal spike discharges in children. Acta Paediatr 135. Hughes JR. A review of the positive spike phenomenon. In:
Scand (Suppl). 1966;168:1–13. Wilson WP, ed. Application of Electroencephalography in
113. Eeg-Olofsson O, Petersén I, Selldén U. The development of the Psychiatry. Durham: Duke University Press; 1965:54–101.
electroencephalogram in normal children from the age of 1 through 136. Gibbs EL, Gibbs FA. Electroencephalographic evidence of thala-
15 years. Paroxysmal activity. Neuropaediatrie. 1971;2:375–404. mic and hypothalamic epilepsy. Neurology (Minneapolis). 1951;1:
114. Cavazzuti GB, Cappella L, Nalin A. Longitudinal study of epilep- 136–144.
tiform EEG patterns in normal children. Epilepsia (New York). 137. Gibbs FA, Gibbs EL. Fourteen and six per second positive spikes.
1980;21:43–55. Electroencephalogr Clin Neurophysiol. 1963;15:353–358.
115. Koshino Y, Niedermeyer E. The clinical significance of small 138. Henry EE. Positive spike discharges in the EEG and behavior
sharp spikes in the electroencephalogram. Clin Electroencephalogr. abnormality. In: Glaser GE, ed. EEG and Behavior. New York:
1975;6:131–140. Basic Books; 1963:315–344.
116. Westmoreland BF, Reiher J, Klass DW. Recording small sharp 139. Hughes JR, Gianturco D, Stein W. Electro-clinical correlations in
spikes with depth electroencephalography. Epilepsia (New York). the positive spike phenomenon. Electroencephalogr Clin
1979;20:599–612. Neurophysiol. 1961;13:599.
117. Small JG, Small IF, Milstein V, et al. Familial associations with 140. Millen FJ, White B. Fourteen and six per second positive spike
EEG variants in manic depressive disease. Arch Gen Psychiatr. activity in children. Neurology (Minneapolis). 1954;4:541–549.
1975;32:43–48. 141. Niedermeyer E, Knott JR. Über die Bedeutung der 14 and 6/sec-
118. Reiher J, Klass DW. Two common EEG patterns of doubtful clin- positiven Spitzen im EEG. Arch Psychiatr Nervenkrankh.
ical significance. Med Clin North Am. 1968;52:933–940. 1961;202:266–280.
119. Lebel M, Reiher J, Klass D. Small sharp spikes (SSS). Reassessment 142. Poser CM, Ziegler DK. Clinical significance of 14 and 6 per second
of electroencephalographic and clinical significance. positive spike complexes. Neurology (Minneapolis). 1958;8:903–912.
Electroencephalogr Clin Neurophysiol. 1977;43:463 [abstract]. 143. Refsum S, Presthus J, Skulstad AA, et al. Clinical correlates of the
120. White JC, Langston JW, Pedley TA. Benign epileptiform tran- 14 and 6 per second positive spikes. An electroencephalographic
sients of sleep: clarification of the small sharp spike controversy. and clinical study. Acta Psychiatr Scand. 1960;35:330–344.
Neurology. 1977;27:1061–1068. 144. Schwade ED, Geiger SG. Matricide with electroencephalographic
121. Klass DW, Westmoreland BF. Nonepileptogenic epileptiform evidence of thalamic or hypothalamic disorder. Dis Nerv Syst.
electroencephalographic activity. Ann Neurol. 1985;18:627–635. 1953;14:18–20.
122. Gutrecht JA. Clinical implications of benign epileptiform transients 145. Shimoda Y. The clinical and electroencephalographic study of the
of sleep. Electroencephalogr Clin Neurophysiol. 1989;72:486–490. primary diencephalic epilepsy or epilepsy of brain stem. Acta
123. Low MD, Henrikson K, Lamont D, et al. The relationship of small Neurovegetat (Vienna). 1961;23:181–191.
sharp spikes to seizures. Electroencephalogr Clin Neurophysiol. 146. Stephenson WA. Intracranial neoplasm associated with fourteen
1984;58:11P [abstract]. and six per second positive spikes. Neurology (Minneapolis).
124. Hughes JR, Gruener, GT. Small sharp spikes revisited: further 1951;1:372–376.
data on this controversial pattern. Electroencephalogr Clin 147. Walter RD, Colbert EG, Koegler RR, et al. A controlled study of
Neurophysiol. 1984;15:208–213. the fourteen-and-six-per-second EEG pattern. Arch Gen
125. Saito F, Fukushima Y, Kubota S. Small sharp spikes: possible rela- Psychiatr. 1960;2:559–566.
tionship to epilepsy. Clin Electroencephalogr. 1987;18:144–119. 148. Long MT, Johnson LC. Fourteen and six positive spikes in
126. Molaie M, Santana HB, Cavanaugh WA. Benign epileptiform nonpatient population. Neurology (Minneapolis). 1967;17:316
transients of sleep (BETS) revisited. Electroencephalogr Clin [abstract].
Neurophysiol. 1989;72:19P [abstract]. 149. Little SC. A general analysis of the fourteen and six per second
127. Gibbs EL, Gibbs FA. Das Elektroenzephalogramm bei congeni- dysrhythmia. In: Proceedings of the 6th International Congress of
taler Anophthalmie. Z EEG-EMG. 1981;12:171–173. Electroencephalography and Clinical Neurophysiology, Vienna.
128. Gibbs FA, Gibbs EL, Gibbs TJ. Relation between specific types of Vienna: Wiener Med. Akad.; 1965:313–315.
occipital dysrhythmia and visual defects. Johns Hopkins Bull. 150. Beydoun A, Drury I. Unilateral 14 and 6 Hz positive bursts.
1968;122:343–349. Electroencephalogr Clin Neurophysiol. 1992;82:310–312.
560 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

151. Gibbs FA, Gibbs EL. Atlas of Electroencephalography. Vol 4. Menlo Observations in man. Electroencephalogr Clin Neurophysiol.
Park, CA: Addison-Wesley; 1978. 1960;12:297–304.
152. Yürüker N, Menzi W. 14 and 6/s positive-Spikes im Schlaf nach 176. Anziska B, Cracco R. Changes in frequency and amplitude in
akustischem Reiz. Z EEG-EMG. 1971;2:121–124. electrographic seizure discharges. Clin Electroencephalogr.
153. Okada S, Kato T, Miyashita K, et al. 14 and 6 Hz positive spikes: 1977;8:206–210.
relationship with a negative spiky phase in the frontal area. 177. Geiger LR, Harner RN. EEG patterns at the time of focal seizure
Electroencephalogr Clin Neurophysiol. 1990;75:S108 [abstract]. onset. Arch Neurol (Chicago). 1978;35:276–286.
154. Niedermeyer E, Ray CD, Walker AE. Depth studies in a 178. Blume WT, Lemieux JF, Young GB. EEG morphology of early
patient with fourteen and six per second positive spikes. stage epileptic seizures. Electroencephalogr Clin Neurophysiol.
Electroencephalogr Clin Neurophysiol. 1967;22:86–89. 1983;56:25P [abstract].
155. Silverman D. Fourteen and six per second positive spike pattern 179. Blume WT, Young GB, Lemieux JF. EEG morphology of partial
in a patient with hepatic coma. Electroencephalogr Clin epileptic seizures. Electroencephalogr Clin Neurophysiol. 1984;57:
Neurophysiol. 1964;16:395–398. 295–302.
156. Ford RG, Freeman AM. Positive spike bursts in a comatose adult. 180. Gibbs FA, Lennox WG, Gibbs EL. The electro-encephalogram in
Electroencephalogr Clin Neurophysiol. 1982;53:29P [abstract]. diagnosis and localization of epileptic seizures. Arch Neurol
157. Drury I. 14- and 6-Hz positive bursts in diverse encephalopathies Psychiatr (Chicago). 1936;36:1225–1235.
of childhood. Electroencephalogr Clin Neurophysiol. 1989;72:12P 181. Gibbs FA, Gibbs EL, Lennox WG. Epilepsy, a paroxysmal cerebral
[abstract]. dysrhythmia. Brain. 1937;60:377–388.
158. Lipman IL, Hughes JR. Rhythmic mid-temporal discharges. An 182. Gastaut H, Vigouroux M. Electro-clinical correlations in 500 cases
electroclinical study. Electroencephalogr Clin Neurophysiol. of psychomotor seizures. In: Baldwin M, Bailey P, eds. Temporal
1969;27:43–47. Lobe Epilepsy. Springfield, IL: Charles C Thomas; 1958:118–128.
159. Egli M, Hess R, Kuritzke G. Die Bedeutung der “rhythmic mid- 183. Klass DW. Electroencephalographic complications in complex par-
temporal discharges.” Z EEG-EMG. 1978;9:74–85. tial seizures. In: Penry JK, Daly DD, eds. Complex Partial Seizures
160. Hughes JR. The continuous rhythmic mid-temporal discharge. and Their Treatment. New York: Raven Press; 1975:113–140.
Clin Electroencephalogr. 2001;32:10–13. 184. Niedermeyer E, Fineyre R, Riley T, et al. Myoclonus and the elec-
161. Garvin JS. Psychomotor variant pattern. Dis Nerv Syst. 1968; troencephalogram. A review. Clin Electroencephalogr. 1979;
29:59–76. 10:75–95.
162. Boutros NN, Hughes JR, Weiler M. Psychiatric correlates of the 185. Hallett M. Myoclonus: relation to epilepsy. Epilepsia (New York).
rhythmic mid-temporal discharges and the 6/sec spike and wave 1985;26(suppl 1):567–577.
complexes. Biol Psychiatr. 1986;21:94–99. 186. Gaches J. Les activitiés périodiques en EEG. Rev EEG
163. Naquet R, Louard C, Rhodes J, et al. À propos de certaines décharges Neurophysiol. 1971;1:9–33.
paroxystiques du carrefour temporo-pariéto-occipital: leur activa- 187. Bauer G, Pieber R. Über periodische Komplexe im EEG. Z EEG-
tion par l’hypoxie. Rev Neurol (Paris). 1961;105:203–207. EMG. 1974;5:75–86.
164. Herranz F, Lopez S. Subclinical paroxysmal rhythmic activity. 188. Spehlmann R. EEG Primer. Amsterdam: Elsevier; 1981.
Electroencephalogr Clin Neurophysiol. 1984;4:419–442. 189. Balthazar K. Zur Kenntnis der Panencephalitis nodosa (Pette).
165. Westmoreland BF, Klass DW. A distinctive rhythmic EEG discharge Arch Psychiatr Nervenkr. 1944;117:666.
of adults. Electroencephalogr Clin Neurophysiol. 1981;51:186–191. 190. Radermecker FJ. Aspects électroencéphalographiques dans trois
166. Westmoreland BF, Klass DW. Unusual variants of subclinical rhyth- cas d’encéphalite subaigue. Acta Neurol Psychiatr Belg.
mic electrographic discharge of adults (SREDA). Electroencephalogr 1949;49:222–232. ^ ^ ^

Clin Neurophysiol. 1997;102:1–4. 191. Radermecker FJ. SystZ matique et Z lectroencZphalographie des
^ ^

167. Miller CR, Westmoreland BF, Klass DW. Subclinical rhythmic encZ phalites et encZ phalopathies. Amsterdam: Elsevier; 1956
EEG discharge of adults (SREDA): further observations. Am J (Electroencephalogr Clin Neurophysiol., suppl 5).
EEG Technol. 1985;25:217–224. 192. Cobb WA, Hill D. Electroencephalogram in subacute progressive
168. O’Brien TJ, Sharbrough FW, Westmoreland BF, et al. Subclinical encephalitis. Brain. 1950;73:392–404.
rhythmic electrographic discharges of adults (SREDA) revisited: a 193. Rabending G, Radermecker FJ. Subacute sclerosing panen-
study using digital EEG analysis. J Clin Neurophysiol. 1998;15: cephalitis (SSPE). In: Remond A, editor-in-chief. Handbook of
493–501. Electroencephalography and Clinical Neurophysiology. Vol 15A
169. Ajmone Marsan C, Ralston BL. The Epileptic Seizures. Springfield, Amsterdam: Elsevier; 1977:28–35.
IL: Charles C Thomas; 1957. 194. Vitrai J, Czobor P, Marosfi S, et al. A study of periodicity and
170. Gastaut H, Broughton R. Epileptic Seizures. Springfield, IL: structure of EEG complexes in a case of SSPE. Electroencephalogr
Charles C Thomas; 1972. Clin Neurophysiol. 1980;50:11–18.
171. Karbowski K. Das Elekroenzephalogramm im epileptischen Anfall. 195. Celesia GG. Pathophysiology of periodic EEG complexes in sub-
Bern: Huber; 1975. ^
acute sclerosing panencephalitis (SSPE). Electroencephalogr Clin
172. Karbowski K. Epileptische AnfSlle. Berlin: Springer; 1985. Neurophysiol. 1973;35:293–300.
173. Oller-Daurella L, Oller-Ferrer-Vidal L. Atlas de Crisis Epilepticas. 196. Radermecker FJ, Poser CM. The significance of repetitive paroxys-
Barcelona: Geigy Division Farmaceut; 1977. mal electroencephalographic patterns. Their specificity in suba-
174. Ralston BL. The mechanism of transitions of interictal spiking cute sclerosing leukoencephalitis. World Neurol. 1960;1:422–433.
foci into ictal seizure discharges. Electroencephalogr Clin 197. Cobb WA. The periodic events of subacute sclerosing leucoen-
Neurophysiol. 1958;10:217–232. cephalitis. Electroencephalogr Clin Neurophysiol. 1966;21:278–294.
175. Ralston BL, Papatheodorou CA. The mechanisms of transition of 198. Cobb WA. Depth recording in subacute sclerosing leucoencephali-
interictal spiking foci into ictal seizure discharges. Part II. tis. In: Kellaway P, Petersén I, eds. Clinical Electroencephalography
Chapter 27 ■ Epilepsy in Adults and the Elderly 561

of Children. New York: Grune & Stratton (Stockholm: Almquist 219. Kirschbaum WR. Jakob–Creutzfeldt’s Disease. New York:
and Wiksell); 1968:275–286. American Elsevier; 1968.
199. Lombroso CT. Remarks on the EEG and movement disorder in 220. Drury I, Beydoun A. Evolution of periodic complexes in
SSPE. Neurology (Minneapolis). 1968;18:69–75. Creutzfeldt–Jakob disease. Am J EEG Technol. 1996;36:230–234.
200. Bogacz J, Castells C, San Julian J, et al. Nonepidemic progressive 221. Tariska P, Gereby G, Majtenyi K. The clinical significance of
subacute encephalitis (type Van Bogaert). II. Serial EEG abnor- periodic lateralized epileptiform discharges (PLEDs).
malities and deep electrography. Acta Neurol Lat-Am. 1959;5: Electroencephalogr Clin Neurophysiol. 1985;61:11P–12P [abstract].
158–183. 222. Chiappa K, Young R. The EEG as a definitive diagnostic tool early
201. Lesse S, Hoefer PFA, Austin JH. The electroencephalogram in dif- in the course of Creutzfeldt–Jakob disease. Electroencephalogr
fuse encephalopathies. Arch Neurol Psychiatr (Chicago). 1958;79: Clin Neurophysiol. 1978;45:26 [abstract].
359–375. 223. Chiofalo N, Fuentes A, Galvez S. Serial EEG findings in 27 cases of
202. Storm van Leeuwen W. Electroencephalographical and neuro- Creutzfeldt–Jakob disease. Arch Neurol (Chicago). 1980;37:143–145.
physiological aspects of subacute sclerosing leuco-encephalitis. 224. Poole EW. Jakob–Creutzfeldt disease. Patients who might have
Psychiatr Neurol Neurochir (Amsterdam). 1964;67:312–322. had this. Electroencephalogr Clin Neurophysiol. 1980;49:89P.
203. Millar JHD, Coey A. The EEG in necrotizing encephalitis. 225. Bortone E, Bettoni L, Giorgi C, et al. Reliability of EEG in the
Electroencephalogr Clin Neurophysiol. 1959;11:582–585. diagnosis of Creutzfeldt–Jakob disease. Electroencephalogr Clin
204. Perier O, Parmentier B, Brihaye J, et al. A case of inclusion body Neurophysiol. 1994;90:323–330.
necrotizing encephalitis. In: Van Bogaert L, Radermecker J, Hozay 226. Alajouanine T, Lecasble R, Remond A. Eléments graphiques
J, et al., eds. Encephalitides. Amsterdam: Elsevier; 1961:235–242. paroxystiques lents de survénue périodiques. Corrélations elec-
205. Carmon A, Behar A, Beller A. Acute necrotizing encephalitis trocliniquis. Rev Neurol (Paris). 1955;93:477–478.
presenting clinically as a space-occupying lesion. A clinico- 227. Barolin G, Scherzer E, Naquet R, et al. Étude électro-clinique des
pathological study of six cases. J Neurol Sci. 1965;2:328–343. états de mal épileptiques survenants chez les apoplectiques. Rev
206. Rawls WE, Dyck PJ, Klass DW, et al. Encephalitis associated with Neurol (Paris). 1962;107:242–243.
herpes simplex virus. Ann Intern Med. 1966;64:104–115. 228. Chatrian GE. EEG pattern: pseudo-rhythmic recurrent sharp
207. Adams JH, Jennett WB. Acute necrotizing encephalitis: a prob- waves. Its relationship to local cerebral anoxia and hypoxia.
lem in diagnosis. J Neurol Neurosurg Psychiatr. 1967;30:248–260. Electroencephalogr Clin Neurophysiol. 1961;13:144 [abstract].
208. Upton A, Gumpert J. Electroencephalography in diagnosis of 229. Fischer-Williams M. Burst-suppression activity as an indication
herpes simplex encephalitis. Lancet. 1970;1:650–652. of undercut cortex. Electroencephalogr Clin Neurophysiol. 1963;
209. Gaches J, Arfel G. Certitude et suspicions d’encéphalites herpé- 15:723–724 [abstract].
tiques. Aspects électroencéphalographiques. Encéphale. 1972; 230. LeBeau FE, Dondey M. Importance diagnostique de certaines
61:510–549. activités électroencéphalographiques lateralisées périodiques ou
210. Elian M. Herpes simplex virus encephalitis and EEG. J à tendance périodique au cours des abscès de cerveau.
Electrophysiol Technol. 1975;1:161–170. Electroencephalogr Clin Neurophysiol. 1959;11:43–58.
211. Kugler J, Marltin JJ, Radermecker RJ, et al. Periodische Komplexe 231. Pagni CA, Vitale A, Cassinari V. Su alcuni casi di tracciati carat-
im EEG bei nekrotisierender Herpes-Enzephalitis. Z EEG-EMG. terizzati da elementi parassistici a ricorrenza ritmica periodica o
1976;7:63–71. pseudoperiodica. Riv Neurol. 1960;30:580–591.
212. Cobb WA. Evidence on the periodic mechanism in herpes sim- 232. Aminoff MJ, Greenberg DA, Wiekle DJ. Periodic EEG complexes
plex encephalitis. Electroencephalogr Clin Neurophysiol. 1979; in infectious mononucleosis encephalitis. Electroencephalogr Clin
46:345–350. Neurophysiol. 1982;53:28P [abstract].
213. Gupta PC, Seth P. Periodic complexes in herpes simplex 233. Markand ON, Daly DD. Pseudoperiodic lateralized paroxysmal
encephalitis. A clinical and experimental study. Electroencephalogr discharges in the electroencephalogram. Neurology (Minneapolis).
Clin Neurophysiol. 1973;35:67–74. 1971;21:975–981.
214. Fineyre R. Periodic discharge of unusually slow character in 234. Dauben RD, Adams AH. Periodic lateralized epileptiform dis-
neonatal status epilepticus caused by necrotizing encephalitis charges in EEG. A review with special attention to etiology and
(herpes simplex). Electroencephalogr Clin Neurophysiol. 1979;46: recurrence. Clin Electroencephalogr. 1974;8:116–124.
80 [abstract]. 235. Bauer G, Aichner F, Hengl W. Der diagnostische Wert periodisch
215. Bergey GK, Coyle PK, Krumholz A, et al. Herpes simplex lateralisierter Komplexe im EEG. Z EEG-EMG. 1981;12:135–141.
encephalitis with occipital localization. Arch Neurol (Chicago). 236. Naquet R, Vigouroux RA. The EEG in relation to pathology in
1982;39: 312–313. simple cerebral ischaemia. In: Remond A, editor-in-chief.
216. Creutzfeldt HG. Über eine eigenartige herdförmige Erkrankung Handbook of Electroencephalography and Clinical Neurophysiology.
des Zentralnervensystems. Z Ges Neurol Psychiatr. 1920;57:1 Vol 14A. Amsterdam: Elsevier; 1972:38–44.
(quoted after Kirschbaum (219)). 237. De la Paz D, Brenner RP. Bilateral independent periodic
217. Jakob A. Über eigenartige Erkrankungen des Zentralnervensystems lateralized epileptiform discharges—clinical significance.
mit bemerkenswerten anatomischen Befunden (spastische Electroencephalogr Clin Neurophysiol. 1982;53:27P [abstract].
Pseudosklerose, Encephalomyelopathie mit disseminierten 238. Aldrich MS, Pugh JE. Cerebral bigeminy: alternating periodic
Degenerationsbeschwerden). Dtsch Z Nervenheilk. 1921;70:132 epileptiform discharges [abstract]. Electroencephalogr Clin
(quoted after Kirschbaum (219)). Neurophysiol. 1984;58:35P.
218. Kirschbaum WR. Zwei eigenartige Erkrankungen des 239. Hughes JR, Taber J, Uppal H. TRI-PLEDs: a case report. Clin
Zentralnervensystems nach Art der spastischen Pseudosklerose Electroencephalogr. 1998;29:106–108.
(Jakob). Z Ges Neurol Psychiatr. 1924;92:157 (quoted after 240. PeBenito R, Cracco JB. Periodic lateralized epileptiform dis-
Kirschbaum (219)). charges in children. Ann Neurol. 1979;6:47–50.
562 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

241. Andriola M. PLEDS and bi-PLEDS in children. Electroencephalogr 245. Gandelman-Marton R, Rabey JM, Flechter S. Periodic lateralized
Clin Neurophysiol. 1982;53:88P [abstract]. epileptiform discharges in multiple sclerosis: a case report. J Clin
242. Ritaccio AL, March G. The significance of BIPLEDs in complex Neurophysiol. 2003;20:117–121.
partial status epilepticus. Am J EEG Technol. 1993;33:27–34. 246. Pohlmann-Eden B, Hoch DB, Cochius JI, et al. Periodic lateralized
243. Silbert PL, Radhakrishnan K, Sharbrough FW, et al. Ipsilateral epileptiform discharges—a critical review. J Clin Neurophysiol.
independent periodic lateralized epileptiform discharges. 1996;13:519–530.
Electroencephalogr Clin Neurophysiol. 1996;98:223–226. 247. Westmoreland BF, Frere RC, Klass DW. Periodic epileptiform
244. Chabolla DR, Moore JL, Westmoreland BF. Periodic lateralized discharges in the midline. J Clin Neurophysiol. 1997;14:495–498.
epileptiform discharges in multiple sclerosis. Electroencephalogr
Clin Neurophysiol. 1996;98:5–8.
CHAPTER

Convulsive Status Epilepticus


FRANK W. DRISLANE, SUSAN T. HERMAN AND PETER W. KAPLAN 28
HISTORY continued electrochemical seizure activity in the brain after ces-
sation of clinically observable convulsions, sometimes now
Status epilepticus (SE) may have been described as early as 600 referred to as “subtle” SE (8).
to 700 BC in cuneiform writings, one of which notes: “If the Relatively little progress in the management of SE was made
possessing demon possesses him many times during the middle until the 20th century. The first modern meetings on SE, the
watch of the night, and at the time of his possession his hands Marseille Colloquia of 1962 and 1964, promulgated better def-
and feet are cold, he is much darkened, keeps opening and shut- initions and classification for seizures and SE and suggested
ting his mouth, is brown and yellow as to the eye. It may go on that SE was the prolongation of many different types of epilep-
for some time, but he will die” (1,2). Caelius Aurelianus, a tic seizures. Gastaut, the organizer of the meetings, stated that
scholar from the late Roman empire, noted that “fits can recur there were “as many forms of SE as there were types of seizures.”
… even in the same day” (3). Today, most literature recognizes SE’s own particular patho-
In the 17th century England, Thomas Willis wrote, “… when physiology, not always identical to that of prolongation of an
as fits are often repeated, and every time grow more cruel, the individual seizure (9).
animal function is quickly debilitated … [and] …. the vital Convulsive SE can be characterized clinically without elec-
function is by little and little enervated, till at length, the whole trophysiologic data, but Berger’s development of the electroen-
body languishing, and the pulse loosened, and at length ceasing, cephalography (EEG) in 1929 led to valuable understanding of
at last the vital flame is extinguished” (4). the different forms of SE, especially for nonconvulsive SE (10)
Reports of SE are few through the 18th century, but in the (see Chapter 29). Since the 1960s, increasing scientific and clin-
1800s, the study of epilepsy flourished in Paris at the Salpêtrière ical attention has turned to the physiologic underpinnings,
that, with 8000 patients, was Europe’s largest asylum. Physicians neurochemistry, and pharmacology of SE, most recently with
there, including Charcot, provided extensive clinical descrip- genetic investigations and functional imaging.
tions of SE. The topic was brought to modern attention in the
university dissertation of Calmeil in Paris, who reported that DEFINITIONS
the phrase derives from the patients’ use of the term: “C’est ce
que les malades appellent entre eux l’état de mal” (5). [“It’s what Gastaut defined SE as “an epileptic seizure that is so frequently
the patients among themselves called a bad condition (of repeated or so prolonged as to create a fixed and lasting
epilepsy).”] He detailed the illness’s severity and a sequence of epileptic condition” (11). No duration was specified. The
seizures without pause that forebode a poor outcome. The use International League against Epilepsy specified “a single epilep-
in (Latinized) English of SE appeared when Bazire translated tic seizure of 30-min duration or a series of epileptic seizures
the lectures of Trousseau, who wrote, “You, however, have heard during which function is not regained between ictal events in a
of circumstances where fits have lasted two or three days, and 30-minute period” (12). Although without a definite scientific
ended in death. It is in these cases that one spoke of, at the basis, 30 minutes has been the standard criterion for duration
Salpêtrière or at Bicêtre, of an état de mal (status epilepticus)” in most clinical studies (13).
(6). Rather than simply repeated seizures, SE was then assumed SE is often a series of individual seizures, without recovery
to be a distinct entity representing the “maximum expression of between seizures. Alternatively, a single convulsion can be very
epilepsy with its own particular characteristics, although prolonged, and many convulsive seizures progress to become
restricted at the time to tonic–clonic seizures” (1). nonconvulsive SE, in which the EEG can demonstrate ongoing
In 1903, Clark and Prout described the clinical and patho- (nonconvulsive) seizure activity. If recovery occurs between
logical appearance of SE in 38 patients, detailing a fusion of individual convulsions, this is considered a clustering of
successive convulsions over 2 to 9 days, to the point of coma seizures rather than SE.
and exhaustion, along with changes in respiration, temperature, While experimental studies on SE show evidence of neuro-
and pulse (7). The presentation was “… composed of delirium, logic damage after 30 minutes, a shorter duration of seizures is
stupor or coma …. and a variety of psychic states, which have currently felt to warrant urgent treatment. Prospective clinical
for their basis cortical discharges.” They listed three stages of SE, trials must define SE as diagnosable expeditiously, and urgent
with the third described as stupor—likely corresponding to the treatment is a clinical necessity. Thus, the largest trial of differ-
nonconvulsive SE that follows generalized convulsive status ent antiepileptic drugs (AEDs) for the treatment of GCSE (14)
epilepticus (GCSE) with “electromechanical dissociation” and considered it mandatory to begin treatment (and make the

563
564 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

diagnosis) within 10 minutes. Later, an “operational” definition As SE continues (concomitant with the decay in GABA
of GCSE proposed that 5 minutes was the time by which SE receptor function), there is increasing excitatory activity of glu-
should be interrupted to avoid morbidity, mortality, or refrac- tamate receptors, especially the N-methyl- D-aspartate (NMDA)
tory SE (15). The duration considered to characterize SE has receptor, promoting and prolonging the hyperexcitable state
been (just barely) agreed upon for GCSE; it is reasonable to use (18,25,26). When glutamate binds to the NMDA receptor,
30 minutes as a clinical definition for other types of SE. membrane ion flow can still be blocked by magnesium, but
with sufficient repetitive depolarization (as occurs during
PATHOPHYSIOLOGY OF STATUS seizures), the magnesium is released and the ion channel opens
EPILEPTICUS more completely in a “use-dependent” facilitation of gluta-
matergic activity (27). NMDA receptor antagonists are not
Episodes of SE appear clinically to begin in the same way as indi- effective in blocking the initiation of SE, but as seizures con-
vidual seizures of the same type, but there are many different tinue, with repetitive depolarization, they appear to become
types of seizures and SE, and it is rare to capture the beginning of more effective anticonvulsants in the same “use-dependent”
SE on an individual EEG. Following initiation, almost half of manner (28). At that point, glutamate antagonists (such as ket-
seizures lasting from 10 to 29 minutes cease spontaneously, with- amine) may become more effective treatments (29).
out treatment (16), and most individual secondarily generalized As seizures continue, stimulation of glutamate (especially
convulsions cease on their own in under 2 minutes and the NMDA) receptors leads to excessive sodium and calcium entry
patient recovers thereafter (17). Some seizures, however, continue into neurons, and this may cause cell death. Gene activation
despite the brain’s intrinsic capacity to terminate shorter seizures, during SE may lead to the production of kinases and phos-
and they progress to the longer episodes of SE. phatases that accelerate the harmful process. Later, inflamma-
It is unclear exactly what facilitates the transition from indi- tion, glial cell changes, and neuronal sprouting or cell loss may
vidual seizures to seemingly self-sustaining prolonged seizures contribute to the refractoriness of subsequent seizures (18).
or SE, but this transition appears to depend substantially on the The potential neuronal injury from prolonged seizures or SE
relative balance of excitatory and inhibitory electrochemical may depend somewhat on the intensity (or frequency) of the
function at the time—at intracellular, membrane, extracellular, epileptiform discharges (30).
and interneuronal levels. To a large extent, SE implies a failure There is abundant experimental (if much more modest clini-
of the typical inhibitory function of the brain in terminating cal) evidence that neuronal damage results from GCSE, particu-
seizures as usual (18). larly when prolonged. A century ago, this damage was attributed
There are probably several pathophysiologic mechanisms to ischemia, but in the 1970s, Meldrum and colleagues showed
that facilitate the continuation of seizures, and in some seizures, that excessive excitatory neuronal activity was the cause of nerve
the balance of electrochemical function appears to shift, unfa- cell damage, at least in experimental animals with prolonged
vorably, toward excessive excitatory activity as seizures progress. GCSE (31–33). In baboons given bicuculline to induce SE, pro-
In experimental animals, one of the primary cellular changes in longed convulsions with continued EEG spike discharges for 1 to
the transition from prolonged seizures to refractory SE is a 5 hours caused neuronal damage in the neocortex, cerebellum,
marked and progressive impairment of gamma-amino butyric and hippocampus (31–34). By “clamping” blood flow, brain oxy-
acid (GABA) agonist–mediated inhibitory function, in turn genation, and glucose supply to preserve homeostasis during
favoring recurrent excitatory circuits (19). During seizures, neu- SE, they showed that hypoperfusion was not crucial in the devel-
rotransmitter receptor changes occur almost immediately, with opment of such damage. Neuronal injury still occurred in the
gene transcription and new peptide production after several hippocampus, and it appeared to be due to excitatory neurotrans-
minutes. GABA receptor modification occurs within 45 min- mitter activity. Thus, the prolonged ongoing cellular electrochem-
utes, showing rapid functional plasticity (20). The electrochem- ical activity of SE itself (rather than concomitant physiologic
ical activity of ongoing seizures also induces gene transcription derangements) was sufficient to cause neuronal injury, although
leading to the production of numerous molecular factors with systemic abnormalities also contributed to this injury. This hip-
excitatory or inhibiting effects on continuing seizures. Among pocampal damage may correlate with prolonged memory and
the many are galanin (which inhibits seizures and SE) (21), sub- other cognitive dysfunction in humans following SE.
stance P, neuropeptide Y, somatostatin, and dynorphin. It is very difficult to prove that SE leads to neuronal damage
Early in SE, there is a marked and maladaptive diminution in humans (as it clearly does in experimental animals exposed to
in the number of GABA-A receptors on neuronal surfaces (22), prolonged, intense electrical stimulation), in part because SE is
in large part due to progressive internalization or endocytosis of seldom fatal without causation by other severe neurologic ill-
those receptors (23). This likely contributes to the reduced nesses that themselves cause neuropathologic damage inde-
effectiveness of GABA agonists in terminating seizures. As pendent of the SE. Nevertheless, a disproportionately decreased
seizures progress, in both experimental animals and in human neuronal cell density was seen in the hippocampus of some
studies, they appear to become resistant to many AEDs (24), patients who died from GCSE (but not NCSE) (35). Forms of SE
especially those acting through the GABA system, such as ben- other than GCSE may not be as harmful and may not require
zodiazepines (BDZs) and barbiturates (20), and those drugs exactly the same treatment, but concern for the possibility of
become less useful in treating SE. such damage leads to a clear mandate for urgent treatment.
Chapter 28 ■ Convulsive Status Epilepticus 565

EPIDEMIOLOGY AND CAUSES Tabl e 2 8 . 1

Studies of the incidence of SE vary tremendously by definitions Different Forms of Status Epilepticus
and different populations studied. The corrected, age-adjusted
incidence of SE was 17.1 cases per 100,000 population per year Generalized Focal
in Germany and 54.5/100,000 in subjects greater than 60 years
Convulsive
old; the case fatality rate was 9.3% (36). This study included all
forms of SE. Most were due to remote symptomatic causes such Generalized convulsive Focal motor SE
as cerebrovascular disease; the mean age of adult patients was SE (GCSE)
65 years. SE is more common in children under 1 year and in Primary generalized Epilepsia partialis
the elderly. The incidence of SE is several times that in develop- GCSE continua (EPC)
ing countries, particularly in children (37).
Secondarily generalized
The incidence of all forms of SE was 41/100,000 in
GCSE (focal onset)
Richmond, Virginia, but 20/100,000 in the white population,
similar to that in Germany (38). The incidence was higher in Myoclonic (see Table 28.2)
African Americans and also likely higher because of ascertain- Tonic (may actually start
ment in an urban medical setting. Extrapolation from the same as focal)
database suggests an incidence of up to 50/100,000 per year,
Clonic (may start as
substantially higher than most estimates (13). In another study,
focal seizure)
mortality of SE was 19% within the first 30 days, highly associ-
ated with the acute cause of SE (39). Short-term case fatality is Atonic (very rare for SE)
generally 20% to 25% when considering GCSE alone, and in all Nonconvulsive
series it is primarily due to the underlying illness causing SE
“Classic” absence SE Complex partial SE, with
(13). In another study of patients with afebrile SE, 32% had
prolonged or repeated
recurrence of SE within 10 years of follow-up (40). Recurrence
CP seizures
is higher in patients with progressive neurologic disorders.
Causes of SE are extremely numerous. They may be grouped Other primary generalized
into larger categories similar to the causes of seizures and NCSE
epilepsy syndromes (12): “symptomatic,” that is, a consequence “Atypical” absence SE
of a known or suspected cerebral dysfunction such as head
Other generalized NCSE Other focal SE with
injury, vascular disease, infection, tumor or other mass lesion,
(often secondarily generalized) nonmotor features:
toxic or metabolic disturbances, or following surgery; “idio-
SE in coma; SE in critically for example, aphasic,
pathic,” such as in the idiopathic generalized epilepsies (IGEs),
ill patients; electrographic SE. sensory
almost always related to a known or presumed genetic-based
syndrome; or “cryptogenic,” in which there is a presumed
underlying cause, but it cannot be identified. The IGEs lead to
SE relatively infrequently, and the SE is usually nonconvulsive
(see Chapter 29), except for the unusual case of generalized which clinical syndrome a patient has. SE, like seizures, can be
convulsions upon awakening (41). organized clinically into those with a focal onset and those with
The most common cause of SE in most larger series, espe- a generalized onset—although many generalized cases cannot
cially in nonhospitalized patients, is “remote symptomatic” be divided easily into those of a primarily generalized nature
(with the cause present for more than a week), often due to ear- versus those with a focal onset and secondary generalization
lier injuries such as stroke and often with more immediate (Table 28.1). Most epidemiologic studies do not specify the type
exacerbation by some systemic problem such as infection, med- of SE, but those that do indicate that GCSE is the most com-
ications, or AED withdrawal (24,42). The next largest category mon and accounts for about half of all SE. Forms of SE with
of causes is probably an exacerbation of earlier epilepsy, also prominent motor signs, whether convulsive or myoclonic, are
commonly with an acute precipitant such as lowered AED lev- covered in this chapter; those with primarily nonmotor mani-
els, infection, or metabolic disturbance. In acute care hospitals, festations are described in the Chapter 29.
most SE patients have “acute symptomatic” causes, that is, due
to a recent (within a week) neurologic injury or illness, the most GENERALIZED CONVULSIVE STATUS
common example of which is a new stroke (43). Most have no EPILEPTICUS
history of seizures and are often older adults with acute and
severe medical or neurologic illnesses. Presentation, Clinical Observations
In order to discuss diagnosis, management, and outcome, it GCSE is the best described form of SE and has the greatest mor-
is important to recognize the tremendous variety of clinical sit- bidity, mortality, and clinical urgency for treatment (44). It may
uations that fall under the rubric of SE and to specify exactly begin as primary generalized convulsive seizures or evolve from
566 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

a focal or complex partial seizure and progress to generalized onset and may progress through a stereotyped sequence of clin-
convulsions. Most GCSE has some evidence of a focal onset or ical and electrographic manifestations (8,47) (Fig. 28.1). The
focal lesion (13), that is, about 70% to 80% is secondarily gen- initial brief interruption in behavior is often accompanied by
eralized (42). Some patients have recurrent seizures without widespread voltage attenuation with fast frequencies at 20 to 40
recovery of consciousness between seizures; others evolve from Hz, sometimes called “electrodecremental.” This may be a wide-
the first convulsion into a more prolonged seizure, with motor spread desynchronization of the background, but with
manifestations becoming less regular clonic movements and implanted EEG electrode recording, very rapid ( 200 Hz)
regular or irregular myoclonic jerking activity, in a continuous activity may be evident in the area of seizure onset (not evident
epileptic (“ictal”) state (44). Primary GCSE is relatively uncom- in routine, surface EEG), suggesting a role for pathologic organ-
mon, often occurring as a result of AED discontinuation or the ization of networks or circuits of neurons in the generation of
consequences of inappropriate AEDs or other drugs, or sys- seizures (48). Within seconds, as the initial tonic contraction
temic illness in patients with IGEs (41). occurs, generalized rhythmic electrographic seizure activity,
Clinically, GCSE is readily recognizable by its motor mani- including rapid spike discharges, gradually increases in voltage
festations, which are often dramatic. Typically, convulsive and decreases in frequency to approximately 10 Hz.
activity consists of paroxysmal or continuous bilateral, usually Not all episodes of GCSE begin this way. Some start with
rhythmic, tonic movements or clonic shaking of the limbs, early electrographic signs of an IGE that, within seconds,
commonly with facial and truncal manifestations as well, and assumes the clinical and EEG picture of a tonic and then clonic
with loss of consciousness. Convulsions may have some later- seizure (Fig. 28.2A–D). Still others begin as focal-onset seizures
alized emphasis, even though the physiologic seizure activity is and generalize rapidly. Within seconds, all three can appear very
bilateral or generalized. If the generalized convulsion is pri- similar clinically and on EEG.
mary, the patient may exhibit no movement for a second or During the clonic phase of the seizure, there is rhythmic
two, followed by the tonic phase of a seizure in which muscle slow activity, sometimes with interspersed spikes, for 10 sec-
contraction occurs throughout the whole body, including a onds or more (Fig. 28.2B). Generalized spike and slow waves or
tightening of chest muscles giving rise to the “epileptic cry.” polyspike and slow-wave activity correspond to the frequency
The tonic phase typically lasts about 10 seconds (45,46) and is of the clinical manifestations.
interrupted by the clonic phase, with particularly rapid and Many of the initial EEG rhythms of GCSE are obscured by
forceful flexion contractions of the arms. Sometimes move- the muscle activity of the convulsion, unless the patient has
ments are more subtle, and there may be a faster quivering. received paralyzing drugs. It is often only when the motor man-
Typically, there is a progressive lessening of the violence of the ifestations lessen that the EEG becomes interpretable.
convulsions, often within the first minute of SE. Eventually the Sometimes, epileptiform discharges can be seen at the vertex.
frequency of clonic contractions lessens, and they cease The superimposed electromyographic activity, however, may
(45,46). The patient is then flaccid, but there may be a large exhibit its own characteristic rhythmic appearance, synchro-
generalized myoclonic jerk at the end of the motor phase. The nous with the clinical jerking activity, and this muscle activity
tonic, and then clonic, phases of an individual seizure seldom on the EEG still indicates generalized convulsions (Fig. 28.2C).
last more than 2 minutes (17). As GCSE progresses, motor If the seizure ends (or between seizures) the discharging
manifestations may lessen and include just relatively subtle activity gives way to a slow, encephalopathic background in all
eyelid or facial or other myoclonic jerking, or motor phenom- areas, or sometimes a very suppressed background, before a
ena may disappear entirely (8). Progression to this “subtle” gradual resumption of a normal drowsy and then waking pat-
GCSE (below) is not characteristic of primary GCSE (44). tern (Fig. 28.2D). In GCSE characterized by repetitive seizures
Repeated seizures also diminish in duration and intensity over with impaired consciousness, the intervening EEG shows dif-
time (44,47). fuse slowing, voltage attenuation, and disorganization. Between
Consciousness is lost completely at the beginning of GCSE seizures, there are often persistent generalized epileptiform dis-
with the first convulsion, or earlier, and the patient is apneic charges with a rhythmic appearance, but slower than during the
during the tonic and clonic phase. The eyes are usually open convulsion (49).
and may show pupillary dilation. Tachycardia, hypertension, As GCSE becomes prolonged, the EEG becomes more dis-
and other autonomic changes are common. Following an iso- continuous, and clinical manifestations become more subtle,
lated convulsion, muscle tone and consciousness return gradu- often with relatively minimal eyelid or facial or other myoclonic
ally, usually with some improvement within minutes, but in SE jerking, nystagmoid eye movements, or even absence of motor
recovery does not occur before the next generalized convulsion movements, all while the EEG continues to show repetitive,
or progression to more subtle or nonconvulsive SE. rhythmic epileptiform discharges. When the visible motor man-
ifestations of GCSE cease, this can be termed “subtle” GCSE
THE EEG OF GENERALIZED CONVULSIVE (below) (8).
STATUS EPILEPTICUS Some have proposed that there is a characteristic sequence
of EEG changes during GCSE, based on recordings from ani-
EEG patterns in SE undergo evolution, usually paralleling the mals and humans during various stages of SE (Fig. 28.3) (8):
clinical manifestations. Generalized convulsions typically show (i) Seizures are initially discrete and followed by background
bilaterally symmetric activity (behavioral and EEG) from the slowing and attenuation until the next seizure begins. (ii)
Chapter 28 ■ Convulsive Status Epilepticus 567

Fp1-Av12
F3-Av12
C3-Av12
P3-Av12
O1-Av12
Fp2-Av12
F4-Av12

C4-Av12
P4-Av12
O2-Av12

F7-Av12
T3-Av12
T5-Av12

F8-Av12
T4-Av12
T6-Av12

Fz-Av12
Cz-Av12
Pz-Av12
RLC
LUC
75 uV 1 sec

Figure 28.1 A generalized tonic–clonic seizure in a 20-year-old woman with cryptogenic generalized epilepsy. At the
onset there is diffuse frontally maximal low-to-moderate voltage beta activity, followed by diffuse muscle artifact as low-
amplitude clonus begins.

The seizures merge gradually, with waxing and waning of volt- patients, and some patients persist in one EEG pattern
age and frequency. (iii) Eventually, seizures become continuous, throughout the course of SE. Evolution of the EEG patterns
although ictal discharges are frequently asymmetric, reflecting depends on the etiology of SE and the baseline condition of
the partial onset of many of the seizures. (iv) With time, seizures each patient, time from SE onset, type of SE, duration of
become discontinuous; there are brief (0.5 to 8 seconds) periods seizure activity, and treatment. Such variation makes classifica-
of generalized voltage attenuation or flattening. (v) In late SE, tion of SE based purely on EEG criteria difficult and adds to
generalized periodic epileptiform discharges (GPEDs) or bursts the controversy regarding which patterns are “ictal.” Stage 4
of polyspikes with frequencies from 0.5 to 4 Hz arise from a rel- above usually has subtle or no motor manifestations but has
atively flat background. “Subtle” SE often corresponds to EEG very frequent and rhythmic epileptiform discharges and is
stages 4 and 5 (8). Patients are typically comatose, with minimal usually considered to represent NCSE. Stage 5 is far more con-
or no motor manifestations at this point (8,49). troversial as to its ictal nature.
Sequential EEG changes, such as the decreased prominence There is no definite difference in clinical significance if SE is
of ictal discharges and increase in intervening background manifested on EEG by recurrent discrete electrographic
attenuation (in parallel with the diminution of clinical mani- seizures or by continuous epileptiform discharges. In one series
festations), represent progressive neuronal dysfunction, possi- of focal SE, there was no significant difference in the etiologies
bly due to metabolic depletion (8). As the EEG “deteriorates,” and outcome of patients with either continuous discharges or
motor manifestations become more subtle or disappear recurrent discrete seizures on EEG (53). In another series,
entirely. patients with continuous clinical seizure activity appeared to be
Other investigators have not found the same sequence of sicker overall than those with discrete seizures (54), but the
EEG changes (50–52). While these EEG patterns were seen fre- management and outcome does not differ much between the
quently, they often occurred out of sequence or in only some two patterns.
A
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG K2-K1
100 uV1 sec

B
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG K2-K1
100 uV1 sec

Figure 28.2 A: A generalized tonic–clonic seizure in a 43-year-old man with idiopathic generalized epilepsy and frequent
episodes of status epilepticus. The seizure begins with widespread frontally maximal alpha activity, then 5-Hz spike-and
slow-wave discharges. B: Continuation of the seizure in A. Slower rhythmic activity, with admixed spike and wave dis-
charges, gradually becomes obscured by muscle activity.
C

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG K2-K1
100 uV 1 sec

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG K2-K1
100 uV 1 sec

Figure 28.2 (Continued ) C: After a period of EEG activity nearly entirely obscured by muscle activity (not shown here),
there is evolution into generalized 1.5-Hz bursts of EMG artifact corresponding to the patient’s generalized clonic jerking.
D: The seizure ends and is followed by diffuse background voltage attenuation.
570 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 28.3 Stages of status epilepticus, illus-


trated by single-channel recordings showing EEG
evolution (see text for details). Stage 1: Discrete
seizures with intervening normal EEG. Stage 2:
Merging seizures, with some intervening normal
EEG. Stage 3: Continuous ictal activity. Stage 4:
Discontinuous ictal activity with intervening
periods of a flat background. Stage 5: Somewhat
periodic epileptiform discharges on a low-voltage
background.

COMPLICATIONS, MORBIDITY Studies (most of which are pediatric and retrospective) have
found minimal cognitive morbidity following SE. Several found
GCSE engenders major physiologic changes, several of which negative consequences, but it is unclear whether they are due to
may contribute to its significant potential for morbidity and the underlying illness or SE itself (60,61). Comprehensive neu-
mortality (42). At the beginning of GCSE (as with isolated con- rologic and neuropsychologic evaluations before and after SE
vulsions) heart rate and blood pressure increase rapidly, along are seldom available; patients with progressive illnesses worsen,
with a substantial surge of catecholamines into the blood whether or not related to the SE; and neurologic deficits may
stream. Cardiac arrhythmias, including bradyarrhythmias and fluctuate with time. It is also difficult to control for the influ-
tachyarrhythmias, are common and sometimes serious; cardiac ence of AEDs and other medications.
arrest may occur (55). Blood pressure and glucose concentra- One group studied 193 children with SE, almost all GCSE,
tions increase early but return to baseline levels after several and concluded that “the major neurologic sequelae are usually
minutes and may even decrease further (56). Hypoperfusion due to the underlying insult rather than to the prolonged
can affect the liver and pancreas. Respiratory rhythm distur- seizure itself” (60). Another group performed thorough neu-
bances are common, as is aspiration pneumonia, often leading ropsychologic testing on 15 adults with prior epilepsy (without
to fever. Prolonged treatment may be associated with infections underlying lesions or symptomatic causes) before and after
or sepsis, thrombophlebitis, and pulmonary emboli. episodes of SE, finding no significant cognitive morbidity
Metabolic complications include electrolyte disturbances accruing from the SE and suggesting that the morbidity in
such as hyponatremia. Peripheral leukocytosis and cerebrospinal other series comes from the underlying etiology, not from the
fluid (CSF) pleocytosis are common. Increased intracranial SE itself (62). Long-term cognitive complications of SE appear
pressure and cerebral edema may develop, especially in children to be relatively uncommon, occurring primarily in the most
(57). Disseminated intravascular coagulation can lead to multi- prolonged and severe cases.
organ failure. Fractures of long bones are common, particularly About 10% of chronic epilepsy presents with an episode of
with falls and trauma, as are vertebral compression fractures. SE (63). Conversely, about 30% to 40% of survivors of GCSE
Falls also cause facial, dental, skull, and many other injuries. develop subsequent epilepsy (60,64,65), four times the rate fol-
Violent muscle contractions increase lactate levels, con- lowing a single acute symptomatic seizure. SE might lead to
tributing to a systemic acidosis, and systemic temperatures epilepsy, but alternatively, patients more likely to have later
often rise significantly (42). Prolonged muscle contraction can epilepsy may present with more severe seizures at the onset, that
also lead to rhabdomyolysis, and myoglobinuria can cause renal is, SE may represent a marker for a more severe epileptic
failure. Hyperthermia may be due to continued convulsions or process that has already begun (66). While chronic epilepsy may
infection, for which the underlying cause must be sought and follow SE and SE is a risk factor for such epilepsy (67), it does
treated. Hyperthermia due to the SE itself usually resolves with not appear to be causal in most.
prompt treatment of the SE.
Imaging studies, including CT, MRI, and PET scans, have OUTCOME, MORTALITY
shown substantial changes in brain tissue with SE, although
many resolve with time, and their clinical implications are often Mortality in “the status epileptici” (with all forms taken together)
uncertain (58). There are some well-detailed cases of perma- is approximately 20% to 25% (13) with most studies consisting
nent and severe tissue damage, such as hippocampal atrophy, primarily of GCSE. Even for GCSE, outcome varies considerably
reasonably attributed to very prolonged SE alone—and accom- and depends heavily on etiology and possibly on other factors. In
panied by neuropsychologic deficits (59). one of the largest prospective, population-based studies of SE,
Chapter 28 ■ Convulsive Status Epilepticus 571

case fatality was 3% for children and 26% for adults—using a 30- The mortality of SE may be declining over the past few
minute definition and including all types of SE (13). Almost all decades—often attributed to faster recognition and treatment
studies conclude that most of the morbidity and mortality of SE (81). It is possible, however, that a wider variety of SE types
derives from the illness precipitating the SE rather than from the is now being diagnosed, including more benign cases with
SE itself (68,69). By one estimate, no more than 2% of fatalities lower morbidity and mortality, thus increasing the reported
comes from the SE itself (70). incidence of SE and improving the reported outcome. One
Etiology is overwhelmingly the most important prognostic study reported a recent decrease in mortality, at least when
factor. Short-term mortality is highest when SE occurs in the myoclonic SE (usually following cardiac arrest and anoxia) was
setting of an acute insult, especially stroke and anoxia excluded (81).
(13,39,65,71). Patients with multiple medical problems, includ-
ing sepsis, fare poorly, while those with SE due to tumors, TREATMENT OF GENERALIZED
trauma, alcohol abuse, or other drugs have an intermediate or CONVULSIVE STATUS EPILEPTICUS
lower mortality (13,42). Generally, the most favorable etiology
is epilepsy itself with some exacerbating factor (reduced AEDs, Clinical management of SE is beyond the scope of a neurophys-
fever, sleep deprivation, or an intercurrent illness or other pre- iology chapter but will be alluded to briefly. Although there is a
cipitant) prompting the SE. Older patients with a symptomatic substantial difference between the duration of SE that leads to
cause of GCSE have a mortality of 50% (43). Presentation in neuronal damage in experimental animals and the shorter
coma (vs. stupor or confusion) confers a poor prognosis (72), duration of SE now felt to warrant prompt treatment in
but this may also be related to the severity of the etiology. patients, seizures should be treated before they last 30 minutes,
Children with SE have a low mortality but the elderly a and certainly by the time of “impending status” (18). The
much higher one. In one series, SE patients over age 60 had a “operational” definition above recommended treatment within
mortality of nearly 40% and those over 80 nearly 60% (73). 5 minutes to avoid morbidity, mortality, or refractory SE (15).
Most SE types in the elderly are focal, or focal with secondary This applies primarily to GCSE.
generalization (totaling 74%; (73)), reflecting the focal pathol- As an emergency condition, GCSE warrants admission and
ogy, particularly stroke—the most common cause of SE in the intensive care unit (ICU) care in most cases, with urgent atten-
elderly. Younger patients with earlier epilepsy had a much tion to airway, breathing, circulation, and basic medical care,
lower mortality (74). Mortality of SE increases with age, but addressing the underlying cause of SE and the medical illness
this may be explained largely by the increasing incidence of causing or worsening it. There has been just one large random-
acute medical illness, such as strokes and other concomitant ized controlled trial of different AEDs for GCSE (14). Based pri-
conditions (68,75). marily on that trial, several other studies, and common practice,
The duration of SE may influence outcome. Experimentally treatment recommendations usually begin with sufficient doses
induced seizures in rodents become more refractory to treat- of intravenous (i.v.) BDZs, the most rapidly acting medications
ment with BDZs after 45 minutes (compared to those lasting (82). With its longer duration of action, lorazepam is frequently
10 minutes) (19). In clinical studies (not always controlled for preferred (82–84). This should almost always be followed by i.v.
etiology) patients with 2 or 4 hours of SE had more morbidity infusion of a longer acting AED such as phenytoin, fos-pheny-
than those with shorter episodes (24,76). In the largest epi- toin, phenobarbital, valproate, or levetiracetam (82,84–86). If
demiologic study of duration and SE, patients whose SE lasted the first agent is unsuccessful in treating SE, a second drug (often
an hour or more had a 32% mortality within 1 month versus one of the other i.v. AEDs) is used frequently (87). Often adjunc-
2.7% for shorter durations, even corrected for other factors by tive use is made of enterally administered carbamazepine, topi-
multivariate analysis (77), but there was no definite correlation ramate, and other AEDs unavailable in i.v. form.
between duration and mortality beyond 1 hour. When con- In a large series, the treatment success rate in patients with
trolled for etiology and other factors, duration of SE was not a overt GCSE was 56%, but only 15% with subtle SE (14).
prognostic factor in at least two studies (39,78), indicating that Lorazepam had the greatest success, terminating seizures in
prolonged SE should not be considered untreatable—unless 65% of cases of overt GCSE. Phenobarbital (58.2%), diazepam
the etiology is particularly ominous. Patients with shorter SE followed by phenytoin (55.8%), and phenytoin alone (43.6%)
durations do better, at least in large part, because the SE has followed, but these differences were not statistically significant
more benign causes. It has been impossible to separate dura- (except for lorazepam vs. phenytoin). If SE continues after the
tion (beyond the first hour) conclusively from the influence of first AED, a second AED succeeds in stopping SE less than 10%
etiology. of the time (and even less with subtle or purely electrographic
Appropriate treatment of SE appears to reduce morbidity SE) (88). At this point, SE is considered refractory, and most
and mortality, but it is impossible to carry out a randomized experts move on to more definitive or aggressive treatment with
trial of treatment and nontreatment. A study comparing two continuous intravenous (C-IV) AEDs.
centers found that poorer quality treatment (primarily the use Common causes of treatment failure include inadequate dos-
of inadequate BDZ doses early in SE) led to worsened outcome ing of the initial AEDs, failure in diagnosis of the cause or
(79). Another found that most treatment was inadequate and underlying etiology (which must be treated), inaccurate diagno-
correspondingly ineffective, also suggesting that adequate doses sis with psychogenic nonepileptic seizures, and failure to main-
of appropriate AEDs led to better outcomes (80). tain adequate AED therapy after the initial treatment—leading
572 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

to a relapse of SE, a common complication and often one signi- TREATMENT OF REFRACTORY STATUS
fying or predicting worsened outcome (89). EPILEPTICUS

REFRACTORY STATUS EPILEPTICUS For refractory SE, ICU admission and mechanical ventilation are
necessary. Most neurologists choose a BDZ followed by a longer
Most SE responds to appropriate use of AEDs, but over time, acting AED (106), but more definitive treatment is often neces-
with continued seizures, SE may become refractory to treat- sary. This can be provided by three commonly used i.v. medica-
ment. The primary causes of refractory status epilepticus (RSE) tions for RSE, all labeled as “coma-inducing” or anesthetic:
are similar to those of SE overall, including stroke or central midazolam, pentobarbital (or often thiopental in Europe), and
nervous system (CNS) tumor (20%), epilepsy-related (20%), propofol (82,107). In one meta-analysis, pentobarbital had a
toxic-metabolic encephalopathy (19%), CNS infection (19%), lower incidence of short-term treatment failure, breakthrough
hypoxia-ischemia (12%), and traumatic brain injury (5%) (90). seizures, and need for another treatment, but a higher incidence
The minimal duration of seizure activity to define RSE varies, of hypotension (90). The study concluded that pentobarbital was
from unspecified to 2 hours (84,91,92). Most epileptologists use more effective in short-term seizure control but was associated
the term refractory without regard to temporal duration, but with more adverse effects, without improved outcome. A few
rather for SE that has not responded successfully to treatment cases require treatment with inhaled anesthetics, usually isoflu-
with two AEDs (some require three), usually including one rane and desflurane, typically used to produce a burst-suppres-
BDZ and a longer acting AED—administered promptly and sion pattern (108). The glutamatergic antagonist, ketamine is
adequately (75,84,91–93). another option (109). Anesthetic treatment should generally be
The prevalence of RSE varies from 10% to over 30% of all continued for days, or at least for 24 hours after seizures stop,
SE, depending on definitions (84,90). In the VA Cooperative then tapered gradually, with concurrent EEG monitoring (82).
Study of GCSE treatment, RSE, defined as continued seizures Many cases of RSE are difficult to treat, but most yield even-
after two AEDs, occurred in 38% of 384 patients with overt tually to i.v. medications, at times augmented with enteral
GCSE and 82% of 134 patients with subtle GCSE (14). In a ret- AEDs. There remain infrequent cases, sometimes of focal RSE
rospective study of all types of SE, RSE (defined as lasting more from focal cortical lesions (such as cortical maldevelopments),
than 60 minutes, with failure of two AEDs) occurred in 31% of that are amenable to surgical treatment. Typically, the seizure
83 episodes of SE in 74 patients (84). RSE was more common focus is identified by neuroimaging or electrocorticography
in patients with nonconvulsive SE (88% vs. 26% with other (110), although this can be difficult. When scans are normal,
forms of SE) and focal motor SE. In children, about 40% of SE surgical localization may be guided by ictal single-photon emis-
becomes RSE, defined as 60 minutes of seizures unresponsive to sion computed tomography (SPECT) (111). In one series of 10
two AEDs (94). In one series of ICU patients (with SE identified children with focal RSE (all with focal imaging abnormalities)
and treated quickly) SE became refractory in 25% and was fatal who failed at least 2 weeks of high-dose suppressive therapy, SE
in 21%; continuous SE had a higher case fatality rate than inter- was terminated by surgery in all, and 7 of 10 became seizure-
mittent SE (95). free (112). Surgery may consist of focal resection of the epilep-
Patients with particularly prolonged SE have been labeled as togenic zone; wider, lobar or even multilobar resections, even to
having “malignant” SE, or SE persisting despite high-dose anti- the point of functional or anatomic hemispherectomy; occa-
convulsant anesthetics. Many cases occur in young adults with sional larger operations such as corpus callosotomy and hemi-
encephalitis, usually following GCSE with a continuation clini- spherectomy, this last operation used most often in cases of
cally or electrographically (96–98). They remain in RSE or devastating epilepsies of childhood, including Rasmussen
relapsing and remitting SE for up to weeks. Malignant SE has a encephalitis (RE); callostomy (113); and multiple subpial tran-
poor outcome, with mortality rates often over 30% (90,99). sections (114), with or without focal resections (115).
Similarly, the prognosis for most patients treated with pro-
longed courses of pentobarbital or other aggressive treatment EEG USE IN REFRACTORY STATUS
for RSE is dismal, with the very high mortality usually attrib- EPILEPTICUS
uted to the almost invariably severe underlying etiology
(89,92,100,101). Nevertheless, there are several reports of criti- EEG is not necessary to make a diagnosis of GCSE, but if a
cally ill patients surviving SE (most of it NCSE) lasting over a patient does not awaken quickly following treatment, an expla-
month (93,102,103), and recovery can be good, depending on nation must be sought. In the VA Cooperative study, only 17%
the etiology (93). Postanoxic SE, on the other hand, is a fatal of patients with overt GCSE (and none with “subtle” GCSE)
condition in essentially all cases, but hypothermia may help to regained normal alertness within 12 hours of treatment (14).
save some patients, especially if brainstem reflexes, somatosen- The cause may be the electrographic persistence of seizure
sory evoked potentials, and some reactivity of the EEG back- activity, that is, NCSE—which most epileptologists consider a
ground are preserved (82,104). A longer duration of SE is rarely continuation of GCSE and warranting urgent treatment.
a good sign, but it is also unwarranted to conclude that the sit- As GCSE progresses, seizure manifestations become subtle,
uation is hopeless when SE has continued for hours or days, or completely absent, and clinical assessment (e.g., cessation of
especially if the cause is relatively more benign (105). motor activity) is inadequate in determining treatment effect. It
Chapter 28 ■ Convulsive Status Epilepticus 573

is often impossible to distinguish whether GCSE has entered a in AED treatment. Most epileptologists re-treat with higher doses
postictal phase or if the lack of recovery is due to continuing SE. of definitive treatment, or for longer at doses that were successful
Both are possible. Electrographic seizures lasting under 30 min- earlier, and then have more AEDs (or higher levels of other
utes occur in about half of patients after treatment for GCSE AEDs) on board for the next attempt at tapering. This can lead to
(51). For patients with refractory SE or persistently impaired a long ICU course. Clearly, these seizures cannot be suppressed
mental status, only EEG can distinguish patients who are in unless they are detected. Isolated epileptiform discharges, how-
drug-induced coma from the 14% to 20% of patients with con- ever, do not appear to necessitate more treatment (116).
tinued nonconvulsive seizure activity (51). Patients who have Continued or recurrent SE should be suppressed for some
received long-acting neuromuscular paralyzing agents also time, usually 12 to 24 hours or more (especially during definitive
require EEG for accurate diagnosis. treatment with midazolam, pentobarbital, or propofol) to ensure
After diagnosis, and during prolonged intensive treatment the absence of electrographic (and of course, clinical) seizures.
with C-IV AEDs, continuous EEG (C-EEG) is essential to assess Nevertheless, the optimal electroclinical goal of treatment (sim-
the effectiveness of that treatment and for recognition of later ple cessation of seizures; both clinical and electrographic seizure
seizure activity and managing its re-treatment (8). C-EEG is control; or a certain degree of suppression of cerebral activity—
essential to watch for relapse of nonconvulsive seizures or often a burst-suppression pattern (Fig. 28.4)) has never been
NCSE, which is not rare, especially in the first 24 hours (51); studied well prospectively. One retrospective study of patients
this occurs in up to 68% of patients as C-IV AEDs are tapered with RSE found that patients whose EEGs were suppressed with
(99). In a retrospective study of RSE treatment, nonconvulsive pentobarbital to the point of a “flat” background did better than
seizures were found in 18% of patients within the first 6 hours those with a burst-suppression pattern, but three other patients
of i.v. midazolam infusion, and breakthrough seizures occurred whose EEGs showed simply freedom from seizures also survived
in 56% of patients later; they were clinically undetectable (i.e., (116). The study was retrospective, and patients thought more
purely electrographic) in 89% (99). likely to survive may have been treated more intensively. Another
Electrographic seizures during C-EEG predict a relapse of study found no clear difference in outcome depending on the
clinical seizures and SE (116,117) and usually warrant an increase depth of EEG suppression (118).

FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV
2 sec

Figure 28.4 Burst-suppression EEG in a 15-year-old boy with refractory status epilepticus, after treatment with pentobarbital.
574 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

In a meta-analysis, patients treated with the goal of EEG tors, can interfere with EEG recording by introducing electrical
background suppression (mostly with pentobarbital) had a 4% artifact. Also, patients are often sedated with BDZs, barbitu-
chance of breakthrough seizures versus 53% for patients treated rates, general anesthesia, and other medications, with their own
to control clinical and electrographic seizures only (mostly with effects, often obscuring EEG findings, or even suppressing EEG
midazolam or propofol) (90). Patients treated to EEG back- rhythms entirely. Head injury introduces additional challenges,
ground suppression had a 76% chance of significant hypoten- including lacerations, surgical bandages, skull defects, subgaleal
sion versus 29% for those treated to suppress seizures only. fluid collections, and edema, plus dressings, drains, and shunts,
Whatever the depth of suppression, mortality was 48%, always all of which can reduce the voltage recorded from the underly-
attributed to the severity of the underlying illness causing SE. ing brain and distort waveform morphologies. C-EEG lasting
The duration of treatment (and monitoring with C-EEG) is days require significantly more attention from the EEG technol-
also inadequately studied. Many investigators have used 12 to 24 ogist to maintaining integrity of EEG leads than does routine
hours of seizure or EEG suppression and then a taper of medica- EEG recording on cooperative patients.
tion, typically over 12 to 24 hours (92,119), but several trials of
midazolam have been shorter (90). One study of patients on
pentobarbital raised the possibility that a prolonged period of FOCAL STATUS EPILEPTICUS
seizure and EEG suppression could be beneficial (116).
During treatment of RSE, EEG should be reviewed immedi- Focal Status Epilepticus
ately until SE is terminated or a burst-suppression record is Focal status epilepticus (FSE) has impressively varied manifesta-
induced with C-IV AEDs. Use of further EEG monitoring is dic- tions, ranging from the readily recognized focal motor seizures
tated by the patient’s condition. If seizures are suppressed, less to focal sensory and aphasic seizures that remain markedly
frequent evaluation may be sufficient, while continued or recur- under-diagnosed. FSE without impairment of consciousness or
rent seizures require more intensive monitoring. Optimally, C- abnormal movements appears relatively infrequent, but it may
EEG monitoring should be performed and reviewed from the remain under-recognized due to the lack of motor findings. The
time aggressive therapy (i.e., general anesthesia) is started, until clinical manifestations of simple partial status epilepticus
seizures are stopped and the patient returns to normal con- (SPSE) reflect ictal involvement of discrete brain regions, such as
sciousness or until electrographic seizures are controlled for 24 motor, sensory, special sensory, autonomic, psychic, language, or
hours. If C-EEG is not available, EEGs might be repeated every other cognitive function; its diagnosis requires preserved con-
6 to 12 hours, until seizure activity does not return, but this may sciousness (126). The incidence of SPSE has been estimated as
miss nonconvulsive seizures. no more than 10 cases per million population per year (127).
EEG findings can also help to predict outcome after SE. After This chapter considers examples of FSE with motor manifesta-
carotid artery distribution strokes, subarachnoid hemorrhage, tions; those without motor phenomena are considered in the
meningitis, encephalitis, or closed head injury, a diagnosis of SE chapter on nonconvulsive status.
increases the mortality rate above that associated with the pre- There are many causes of FSE. Infectious (e.g., encephalitis)
cipitating event itself (120). and vascular (e.g., stroke, hemorrhage, or vasculitis) are the
There are persistent “ictal” discharges (rhythmic spikes, sharp most common, but mass lesions, trauma, multiple sclerosis, and
waves, or spike-wave discharges) lasting 10 seconds to minutes, rarely, mitochondrial or degenerative disorders can be respon-
in 48% of patients after control of GCSE (51). Discharges lasting sible (128). Occasionally, benign idiopathic (genetic-based)
longer than 30 minutes are associated with higher mortality focal epilepsies, such as rolandic epilepsy, lead to SE (129).
rates, even after controlling for age and etiology of SE. Delayed Medication and metabolic changes may precipitate an episode,
ictal discharges occurring more than 30 minutes after SE has presumably with an underlying focal lesion.
been controlled completely did not worsen outcome. Patients Many cases of focal SE occur in the setting of acute stroke—
with normal EEGs after SE had no significant morbidity or in one series causing 41% of all cases of SE—and 61% in the
mortality. elderly (71). In another series, 5% of stroke patients had new
Periodic lateralized epileptiform discharges (PLEDs) after SE seizures and 1/5 of these had SE (130). These patients had a
are associated with an increased mortality, typically attributed to high mortality rate, just over 50%. In another series, the most
more severe etiologies (52,121). The presence of GPEDs late in common cause (75%) of FSE was stroke, and most patients had
SE was associated with poor outcome in one study (52), but not a history of epilepsy; all except for one with aphasic SE had
in another (122). Burst-suppression patterns are also associated motor-predominant seizures, and about half had epilepsia par-
with worse outcomes, whether pharmacologically induced or tialis continua (EPC) (131). The median duration of SE was 4
due to hypoxia (121,123). In children over 2 years, lack of back- hours, but three patients had EPC continuing for weeks.
ground reactivity after SE portended a poorer prognosis (124). Duration did not correlate with outcome, which was deter-
Recording EEGs in the ICU is fraught with numerous diffi- mined primarily by the underlying cause.
culties (125) (see also Chapter 29). Patients are, by definition, The morbidity and mortality of focal SE are determined
critically ill and may need to be moved by nursing staff, produc- largely by etiology. Among 47 FSE patients, acute strokes caused
ing plentiful artifact during EEG, and electrodes are more likely the only poor (fatal) outcome, in four patients (131). Persistent
to become disconnected. Machinery in the ICU, including bed morbidity occurred in another 10 patients, attributed to the
equipment, ventilators, fluid infusion pumps, and other moni- underlying lesion (usually stroke) in almost all. One study found
Chapter 28 ■ Convulsive Status Epilepticus 575

a synergistic (for the worse) effect of acute stroke and SE in 83 ous or discontinuous, with discrete seizures showing evolving
patients (71). Rarely, FSE becomes extremely refractory and even frequencies of 2 to 6 Hz lasting 30 to 60 seconds or more (53).
life-threatening and necessitates surgical treatment, for example, Some EEGs show discrete electrographic seizures with wide-
some FSE due to focal cortical dysphasias (111). spread fields that are more pronounced over one hemisphere.
In a large epidemiologic study, SE with focal features had the Although clinical manifestations of a seizure may be focal, the
same mortality as generalized SE (20% to 30%) (77), but this EEG may show generalized discharges later, and an EEG initi-
large database included (relatively benign) absence SE in the ated in the middle of SE may be unable to distinguish between
generalized group and patients with secondarily GCSE in the focal and generalized SE.
focal group. For SPSE without motor manifestations, there is no
evidence of mortality or long-term morbidity. Epilepsia Partialis Continua
Focal motor SE with very prolonged jerking activity is called
EEG EPC. EPC has been defined as “clonic muscular twitching
In part because a limited volume of brain tissue is involved, repeated at fairly regular intervals in one part of the body for a
only 20% to 35% of simple partial seizures have an ictal electro- period of days or weeks” (134). It includes continuous or
graphic correlate detectable by scalp EEG (132). The EEG may nearly continuous, relatively slow (every 0.5 to 2 seconds),
be normal or show nonspecific changes such as focal slowing. often somewhat irregular, clonic jerking motions of groups of
In other patients, a variety of ictal patterns may occur, such as muscles on one side of the body (134). EPC can be remarkably
focal high-frequency discharges, rhythmic waveforms with persistent, with episodes lasting days, weeks, or even years
evolving morphology, and repetitive epileptiform discharges (135). It is readily diagnosed clinically because of its promi-
(Fig. 28.5) (53,128,133). Seizure activity may be either continu- nent and long-lasting motor manifestations. While EPC has

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

EKG
100 uV 1 sec

Figure 28.5 A 78-year-old woman with colon cancer had repetitive seizures originating in the right temporal lobe, evolv-
ing gradually to clonic jerking of the left face and neck. These seizures occurred every 5 to 10 minutes for over 12 hours,
gradually increasing in duration, becoming focal motor status epilepticus. This EEG is from a period of 3 hours of contin-
uous seizure activity. Rhythmic epileptiform activity is seen in the right posterior temporal region, with contralateral EMG
artifact from the left face and with neck muscle clonus.
576 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

become close to a synonym for focal motor SE, it is this pro- associated with progressive brain lesions, and there is a greater
longed continuous jerking activity that is EPC, while repetitive variety of seizures and occasionally, of muscle groups involved.
isolated discrete seizures are usually termed simply focal motor Type 2 EPC may have multifocal motor activity and may be
SE. The prevalence of EPC is estimated to be under one patient associated with other seizure types, more disturbance of the
per million population in the United Kingdom (136). EEG background, and a progressive neurologic deficit, such as
EPC was described by Kozhevnikov in 1895 in cases of the hemiparesis of Rasmussen syndrome (below).
Russian spring–summer encephalitis, which included spreading
focal motor seizures (137). Most cases involved children, often Treatment and Outcome
appearing months after a presumed infectious illness and with The continued focal jerking of EPC is often refractory to AED
continuation for years. Similar cases followed other encephali- treatment (82). Valproate, clonazepam, and levetiracetam are
tides. EPC in the modern era is far more often the result of common treatments, as are steroids. Intravenous immunoglob-
strokes, both hemorrhagic and ischemic, and other vascular ulin (IVIG) and plasma exchange are used increasingly (82).
lesions (including venous infarcts) in adults (134,135). Tumors Resective surgery can be curative if the lesion is small enough,
are a very uncommon cause of new-onset EPC (138). Cortical but it is often ineffective (136). Multiple subpial transections
migration abnormalities, or heterotopias, and other develop- are another possibility when the epileptogenic area is in corti-
mental abnormalities are possible causes and occasionally lead cal structures and motor or language function is at risk from
to refractory focal SE (111). Other causes include focal infec- hemispherectomy (115,149,150). Occasionally, EPC resolves
tions such as tuberculosis, syphilis, Toxoplasmosis, and HIV, without treatment (136). While EPC may remain refractory,
progressive multifocal leukoencephalopathy, tuberous sclerosis, AEDs may help prevent more dangerous secondarily general-
subacute sclerosing panencephalitis, Sturge–Weber syndrome, ized convulsions.
perinatal injury, congenital abnormalities including cortical The long-term prognosis for patients with EPC depends pri-
dysplasias, and trauma (136). Unusual causes include multiple marily on the underlying lesion or illness. It is often poor. Long-
sclerosis; mitochondrial disorders including mitochondrial term morbidity of EPC in terms of weakness, sensory and
encephalopathy with lactic acid and stroke-like episodes visual loss, and language dysfunction is substantial, and many
(MELAS) (139); and paraneoplastic syndromes (140). In chil- patients have severe cognitive deficits (136). In one series,
dren, RE is the characteristic abnormality causing EPC. nearly half the patients died over a follow-up averaging 3 years,
In addition to laboratory studies to look for infectious and usually because of the lesion causing EPC (135).
metabolic causes, all patients with new-onset EPC need MRI to
look for focal lesions. There is usually an identifiable brain EEG
lesion, although it may be small. Fluid-attenuated inversion While EPC is a subtype of SPSE characterized by continuous
recovery (FLAIR) sequences are often best at demonstrating the motor seizures, the relationship between clinical manifestations
lesion (141). A particularly important cause for recognition and and EEG findings is inconsistent. EPC is often associated with
treatment is hypernatremia and nonketotic hyperglycemia, irregular focal spikes and sharp waves and sometimes with
which may precipitate EPC especially in a patient with an ear- sharp-and-slow wave discharges with a frequency of 0.5 to 3
lier focal lesion, most often stroke (142). This EPC is usually Hz, centering on about 1 Hz (Fig. 28.6). Depending on the pop-
treatable readily with correction of the serum glucose and vol- ulation studied, epileptiform discharges on scalp recordings
ume status. EPC often occurs in patients with chronic neuro- contralateral to the motor manifestations may occur in most or
logic illness and is often relatively stable clinically and not in a minority of patients (135,136), presumably because the
always in need of urgent intervention. responsible lesions are small or, more likely, because of the
“inconvenient” orientation of discharges with respect to the
Pathophysiology cortical surface. One series reported focal discharges on scalp
Most investigators conclude that EPC results from focal cortical EEG in only 22% of patients (136), but additional electrodes
epileptic discharges, often involving a relatively small area of can show focal discharges in up to 71% (135). Discharges not
cortex near the rolandic region or motor strip. seen on scalp EEGs may be more evident on corticography or
Physiologic studies show cortical dysfunction (135,143,144), other invasive recordings (144). Back-averaging of EEG time-
with the pathophysiology of epileptogenic foci in hyperexcitable locked to clinical myoclonus can demonstrate a relationship
motor cortex (145). Focal cortical hyperexcitability can be between scalp-recorded discharges and muscle jerks in 37% to
demonstrated by correlation with focally enhanced somatosen- 45% of patients (135,136). PLEDs are seen in 22% to 71% of
sory evoked potentials (145,146). Responsible lesions, however, patients (135,151).
may be subcortical and engender hyperexcitability of the overly- The EEG background rhythm may remain normal in EPC,
ing cortex, possibly through deafferentation (134,138,147). especially in areas away from the lesion, but EPC is often asso-
EPC can be considered to take two forms (148). Type 1 arises ciated with structural lesions that produce some focal slowing
from a focal, nonprogressive lesion, for example, due to the or attenuation of the EEG background, if not epileptiform dis-
classic Russian spring–summer encephalitis. Repetitive jerking charges. Under 10% of patients have completely normal EEGs
tends to occur in a restricted group of muscles but can progress (135). Clinically, the EEG is often surprisingly unrevealing, but
to complex partial seizures or generalized convulsions. Type 2 is it is not always necessary for management.
Chapter 28 ■ Convulsive Status Epilepticus 577

Figure 28.6 A 56-year-old woman with epilepsia partialis continua following resection of a right parietal meningioma.
EEG shows periodic lateralized epileptiform discharges at approximately 2 Hz in the right hemisphere, more posteriorly,
with EMG artifact from rhythmic facial twitching over the left temporal channels.

Rasmussen Syndrome glutamate receptor antibodies (157), immune therapies such as


RE is itself uncommon, but it is a relatively common cause of high-dose corticosteroids and IVIG have been attempted, with
EPC. It was first described at the Montreal Neurologic Institute in variable success in the short-term reduction of seizures, but the
1958, usually occurring in children and adolescents and almost hemiparesis persists (158).
always restricted to one hemisphere (152). It often follows some Resective surgery becomes necessary in many patients (155)
earlier, viral infection (153). It has an inflammatory basis and is and is usually the best treatment (150), especially for seizure
posited to represent a persistent and chronic focal encephalitis of control; sometimes there is improved neurologic function post-
infectious or autoimmune basis (154,155). In most patients, RE operatively. A minority of patients may have seizures reduced
is a progressive illness with neurologic deterioration, often with or even eliminated by hemispherectomy, but this may entail
unremitting focal seizures, contralateral hemiparesis, and wors- major morbidity. Often, a partial hemispherectomy is the only
ening cognitive function. The neurologic deficit may progress for helpful treatment (159). Multiple subpial transections, with or
years and then stabilize, with persistent EPC (Fig. 28.7). For without focal resections, are a possible option (115,149,150).
unclear reasons, some cases stop progressing spontaneously, and
occasionally, the disease is self-limiting (154). On EEG, there is PERIODIC LATERALIZED EPILEPTIFORM
prominent focal slowing, correlating with the underlying and DISCHARGES
widespread inflammatory or immune lesion (156).
The EPC of RE includes jerking at 1 to 2 Hz of both agonist PLEDs were first described by Chatrian as spike or sharp-
and antagonist muscles in same area of the body. It is typically and-slow-wave complexes, with repetition rates of about 1 to 2
refractory to treatment with AEDs and persists for years. BDZs, Hz (151). PLEDs are surface-negative bi-, tri-, or polyphasic dis-
valproate, and ethosuximide may be useful in treating charges consisting of spike, sharp, or polyspike components, with
myoclonic jerks and seizures, but even high doses of AEDs usu- variable following slow-wave complexes lasting 60 to 600 msec
ally fail to suppress all seizures. Because RE appears related to (mean 200 msec), of 50 to 150 V (occasionally up to 300 V),
578 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 28.7 A: EEG showing periodic lateralized epileptiform dis-


charges in the left parietal and posterior temporal regions, following focal
status epilepticus in a 26-year-old woman with adult onset Rasmussen
encephalitis. B: MRI following status epilepticus in the same patient with
recurrent secondarily generalized seizures shows cortical T2 hyperinten-
sity and focal thickening (edema) of the cortex on the left.
Chapter 28 ■ Convulsive Status Epilepticus 579

FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

Figure 28.8 Left hemisphere periodic lateralized epileptiform discharges in a 72-year-old woman following a left middle
cerebral artery territory stroke.

usually recurring at 0.5 to 2 Hz (ranging from 0.2 to 3 Hz ); occa- the time of the EEG recording (151,163,164). PLEDs are often
sionally, intervals are up to 10 seconds (Fig. 28.8). Epileptiform considered “the terminal phase of status epilepticus” (161).
discharges are broadly distributed over most of one hemisphere Nevertheless, they are highly associated with clinical seizures
and may reflect to the opposite hemisphere as well. Between dis- and may lie somewhere along an “ictal-interictal continuum”
charges, the background activity is usually markedly attenuated (165). PLEDs may also occur on limited segments of an EEG
and slow. EEG waveform morphologies stay fairly constant for a (e.g., 20 to 30 minutes) with definite electrographic seizures later
given patient and EEG, but the discharge frequency may decline in the same recording (166) (Fig. 28.9A,B), indicating that
with time, and most PLEDs will resolve within weeks (151). PLEDs may not be “ictal” when seen but are recorded in patients
Rarely, discharges persist for months or even years (160). who have clear seizures at other times (161). Seizures typically
PLEDs occur most commonly after acute large structural begin with rhythmic fast activity, sometimes with a field distinct
destructive lesions such as stroke (the most common cause), from that of underlying PLEDs; the PLEDs may then disappear.
tumor, or infection, but also in chronic seizure disorders and static Patients with “PLEDs plus” (with lower voltage rhythmic epilep-
lesions (160). One series of 147 cases of PLEDs found the etiology tiform discharges or other rhythmic patterns between the higher
to be stroke in 27%, anoxia or old stroke in 12% each, infection voltage periodic sharp waves) (167) (Figs. 28.9 and 28.10) or
8%, tumor in 7%, and other in 34% (161). Patients with PLEDs PLEDs with intermittent electrographic seizures are more likely
are often obtunded, with focal neurologic deficits and often focal to have epileptic seizures (Fig. 28.9B) and should be considered
motor seizures. EPC is common. Clinical seizures occur in at least for more vigorous treatment with AEDS.
80% of patients with PLEDs, often before the EEG is obtained, Periodic discharges are generally considered ictal if they occur
and electrographic seizures in even higher proportions. Many had consistently with stereotyped clinical behavior that appears
prior SE (161,162). Half of patients without prior epilepsy who epileptic. More rapid discharges (at least 1 Hz and certainly 1.5
survive the acute illness develop long-term epilepsy (163). Hz) would also be interpreted as representing seizures (and if
Most electroencephalographers do not consider PLEDs to be prolonged enough, SE) by most EEG-ers. In a patient with confu-
“ictal” (i.e., a manifestation of clinical seizures or SE), at least at sion, PLEDs or BiPLEDs (bilateral-independent PLEDs) are likely
A
FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

B
Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz
EKG

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz
EKG 150 uV2 sec

Figure 28.9 A: Same patient as in Figure 28.8. Later EEG shows “PLEDs plus,” a faster repetition rate of PLEDs with inter-
vening faster frequency patterns. B: Same patient; PLEDs evolving into a focal left hemisphere seizure.
Chapter 28 ■ Convulsive Status Epilepticus 581

Figure 28.10 Another example of “PLEDs plus” (PLEDs with intervening faster components) in a 56-year-old man with
HIV infection and a left temporal ring-enhancing lesion.

to be ictal if the EEG discharges resolve and clinical symptoms over each hemisphere have different amplitudes, fields, and repe-
improve after BDZ treatment. Also, some studies have found focal tition rates (Fig. 28.11). Background activity is often severely
hyperperfusion on SPECT scans (168,169) or evidence of focally attenuated bilaterally. BiPLEDs are typically due to anoxia (28%),
increased metabolic activity on PET at the time of the PLEDs CNS infections (28%), chronic seizure disorders (22%), or vascu-
(170), suggesting that PLEDs may be ictal in those cases. lar causes and are associated more often with impaired mental
Sometimes, PLEDs are definite signs of seizures! One report status (coma in 72% vs. 24%) and higher mortality rates (61% vs.
of seven patients described recurrent confusional episodes 29%) than PLEDs, but focal seizures are less common (173).
associated with PLEDs (171). There were no structural lesions, GPEDs, also considered ictal sometimes and interictal at other
and EEG discharge intervals were as long as 4 seconds. Clinical times, are discussed further in the Chapter 29.
deficits resolved, with slowing of EEG discharges, sponta-
neously or in response to BDZs. Carbamazepine appeared to MYOCLONIC STATUS EPILEPTICUS
prevent recurrences, but patients relapsed when it was
decreased. The authors considered PLEDs an “unusual status Persistent myoclonus may or may not be epileptic in origin.
epilepticus of the elderly.” Another group recorded PLEDs dur- Without evidence of an epileptic cause, it is typically (and prob-
ing clinically well-defined SE, with an EEG evolution in fre- ably best) labeled status myoclonicus or sometimes myoclonus
quency and amplitude terminating in a more typical seizure status. It includes prolonged, but frequently nonrhythmic,
pattern—with a favorable response to BDZ treatment (172). myoclonic jerking, usually of large amplitude, often involving
They concluded that PLEDs was an ictal EEG pattern when the face, trunk, and limbs, but sometimes multifocal or asyn-
there are appropriate clinical signs. PLEDs may be seen during chronous. The cause is usually an acute and severe encephalopa-
seizures or SE, and their clinical significance differs in individ- thy, often anoxia or possibly metabolic disturbances, particularly
ual cases. EEG findings must be discussed and reinterpreted in renal failure (174). The EEG may show widespread slowing
the clinical setting. indicative of an encephalopathy and have no spikes or sharp
Similar discharges, bilateral-independent pseudoperiodic lat- waves correlating with the myoclonic jerks (Fig. 28.12). In more
eralized epileptiform discharges (BiPLEDs), occur independently severe encephalopathies, there may be a burst-suppression pat-
and asynchronously over the two hemispheres (173). Discharges tern or periodic discharges.
582 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz
50 uV 1 sec

Figure 28.11 A 68-year-old woman with herpes encephalitis and status epilepticus. EEG after control of clinical seizures
showed bilateral-independent pseudoperiodic lateralized epileptiform discharges (BiPLEDs), with different and independ-
ent discharge frequencies over the two hemispheres.

Continuing myoclonic jerks with an EEG showing persistent generalized epilepsies (IGEs) in which myoclonus is a promi-
rapid and rhythmic epileptiform discharges are persuasive for an nent manifestation both ictally and interictally, such as juvenile
epileptic origin, in which case the diagnosis is better termed myoclonic epilepsy (JME) (Fig. 28.13); childhood and juvenile
myoclonic status epilepticus (MSE) (175). MSE is characterized absence epilepsy; and grand mal seizures upon awakening
by prolonged myoclonic jerking activity, by usual definition over (Fig. 28.14) (179,180). With MSE in the IGE syndromes,
30 minutes in duration. Myoclonus may be isolated to one par- episodes may include myoclonic jerking activity up to five
ticular muscle group or, particularly in primary generalized times a second (although often every several seconds), partic-
epilepsies, appear bilaterally and symmetrically, particularly in ularly in proximal muscles, often with bilateral synchrony, and
flexor muscle groups in the arms. Myoclonus may be rhythmic, can last for hours—somewhat surprisingly without necessarily
but this is not uniform, even with generalized discharges on the affecting consciousness (179). Some patients have frequent
EEG. Pathophysiologically, the myoclonus is of the cortical reflex prolonged myoclonic seizures with normal consciousness
type (176). MSE has a remarkable variety of causes although between individual myoclonic jerks (180), even with frequent
clinical presentations may be similar from one to the next. The 3 to 6-Hz epileptiform discharges on the EEG. Myoclonus can
prognosis depends on the etiology (49,175). Although MSE is also consist primarily of eyelid myoclonia (181). The individ-
discussed in the convulsive SE chapter because movements are a ual syndrome predicts the frequency of SE (41,182).
prominent component of the SE, some myoclonic jerks are of In JME, MSE may occur upon awakening and begin with
minimal amplitude and the MSE can appear largely nonconvul- irregular and isolated myoclonus with an accelerating or
sive clinically (177). crescendo pattern, ending in SE. Patients with generalized
An insightful review of the different forms of MSE divides myoclonic epilepsies (such as JME and some other IGEs) may go
cases into those due to epilepsy itself and those symptomatic into MSE when medications are changed (175). Sleep depriva-
of other brain illnesses (178) (Table 28.2). The pure, or epilep- tion is another common precipitant, and some AEDs may lead to
tic, forms include MSE occurring in the primary “idiopathic” an exacerbation of epilepsy, sometimes to MSE (Fig. 28.14A,B)
Chapter 28 ■ Convulsive Status Epilepticus 583

Fp1-F3

F3-C3
C3-P3

P3-O1
Fp1-F7
F7-T3

T3-T5
T5-O1

Fp2-F4
F4-C4
C4-P4
P4-O2

Fp2-F8
F8-T4

T4-T6
T6-O2
Fz-Cz

Cz-Pz
EogL-A1

EogR-A2

ECG
75 uV 1 sec

Figure 28.12 Irregular generalized and multifocal sharp waves, in an 87-year-old man with acute renal failure, altered
mental status, and frequent myoclonic jerking activity. The EEG sharp waves are less rhythmic than would be considered
indicative of seizures by most electroencephalographers, and the background indicates an encephalopathy.

Tabl e 2 8 . 2

Myoclonic Status Epilepticus

“Primary,” in generalized epilepsy syndromes such as absence epilepsy, juvenile myoclonic


epilepsy, and other myoclonic epilepsies
“Secondary” in other epilepsy syndromes:
“Minor” epileptic status (of Brett)
Severe myoclonic epilepsy of infancy
MSE in myoclonic astatic epilepsy, Lennox–Gastaut syndrome
Epilepsy with myoclonic absences
SE with negative myoclonus and brief atonic episodes
“Symptomatic” of acute neurologic illness (e.g., anoxia, injuries, and encephalopathies)

Adapted from Ohtahara S, Ohtsuka Y. Myoclonic status epilepticus. Chapter 63. In: Engel J Jr, Pedley TA, eds. Epilepsy: A
Comprehensive Textbook. 2nd ed. Philadelphia: Lippincott-Raven Publishers; 2008:725–729.
584 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7

F7-T3
T3-T5

T5-O1
Fp2-F8
F8-T4
T4-T6

T6-O2
Fz-Cz
Cz-Pz
LUC
RLC
EKG
100 uV1 sec

Figure 28.13 Myoclonic status epilepticus in an 18-year-old woman with juvenile myoclonic epilepsy. EEG after a night
of sleep deprivation showed irregular bursts of generalized spike-and-wave and polyspike-and-wave discharges for nearly
one hour. Most bursts were associated with clinical myoclonic jerking activity.

(183). Overall, the IGE syndromes lead to MSE relatively infre- myoclonus is usually not the dominant feature. MSE in LGS is
quently, and the MSE is usually easy to treat and the long-term often associated with the slow spike and wave EEG patterns
outcome good (179). seen in atypical absence SE (see Chapter 29). Altered con-
MSE also occurs in other “secondary” epilepsy syndromes in sciousness may be mixed with myoclonic jerking activity,
which myoclonus is not a prominent feature of the interictal which by itself does not necessarily affect consciousness. MSE
baseline. In these syndromes, the myoclonic jerks are more often also occurs in younger children with infantile spasms and hyp-
asymmetric and asynchronous than in the IGE syndromes, and sarrhythmia.
they may be of smaller amplitude and less rhythmic (11). Severe myoclonic epilepsy of infancy (SMEI) or Dravet
Secondary MSE causes include the progressive myoclonus syndrome may include myoclonic jerking activity, but
epilepsies caused by storage diseases (Fig. 28.15) such as lipi- impairment of consciousness (“obtundation status”), some-
doses and lipofuscinosis, and Lafora disease (184), infectious ill- times prolonged, is usually the greater clinical problem (185).
nesses such as encephalitis, and severe childhood epilepsies such The irregular myoclonic activity often involves the face or
as Dravet syndrome (185). MSE is more common in the second- limbs and may persist for hours. It usually presents in infancy
ary forms of childhood myoclonic epilepsy than in the primary but may lead to NCSE around the age of 2 years. The EEG
syndromes (11), and they are frequently refractory to AEDs. may be normal at first but later includes focal or multifocal
Many episodes of MSE occur in very young children with epileptiform discharges interictally, with eventual generalized
early life genetic or structural deficits or inborn errors of spike-wave discharges on a very disturbed, slow, and irregu-
metabolism and severe baseline encephalopathies. These lar background (186). The long-term outcome is often poor,
include Lennox–Gastaut syndrome (LGS), where MSE may be reflective of the underlying illness rather than because of the
mixed with other forms of SE such as “myoclonic astatic” MSE itself.
seizures with irregular facial and limb myoclonus and epilepsy Finally, MSE may occur as a manifestation of underlying
with myoclonic absences (Fig. 28.16). SE is not rare in LGS, but encephalopathies unrelated to an earlier epilepsy syndrome,
A
FP1-F3

F3-C3
C3-P3

P3-O1
FP2-F4
F4-C4
C4-P4
P4-O2
FP1-F7
F7-T3
T3-T5

T5-O1
FP2-F8
F8-T4
T4-T6

T6-O2
Fz-Cz
Cz-Pz
LUC
RLC
EKG 200 uV
1 sec

B
Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz
LUC
RLC
EKG

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz
LUC
RLC
EKG
Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz
LUC
RLC
500 uV
EKG 2 sec

Figure 28.14 A: EEG during myoclonic status epilepticus in a 15-year-old girl with epilepsy with generalized tonic–clonic
seizures on awakening. She had no earlier history of myoclonus but was admitted with nearly continuous myoclonic jerk-
ing after being started on carbamazepine. B: Same patient as in A. After 20 minutes, the individual generalized myoclonic
jerks evolve into rhythmic generalized clonic jerking (clonic status epilepticus).
586 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz
50 uV 1 sec

Figure 28.15 Myoclonic status epilepticus in 32-year-old man with progressive myoclonus epilepsy due to
Unverricht–Lundborg disease. The background activity is slow and disorganized, and EEG shows generalized and multifo-
cal spikes and polyspikes. Clinically, the patient was more confused than at baseline and showed continuous fragmentary
myoclonus.

particularly in severe encephalopathies due to anoxia or meta- generalized, frontally predominant spikes and polyspikes with
bolic derangements (Fig. 28.17) (174,175,187). The MSE, per faster frequencies of 4 Hz or so interictally on a normal back-
se, is usually less important than the devastation caused by the ground (Fig. 28.13). Spike discharges may precede the
encephalopathy, particularly in the case of anoxia. MSE may be myoclonic jerking activity immediately (178). During seizures,
part of an acute postanoxic syndrome following cardiac arrest, there are generalized, frontally maximal irregular spike-and-
often beginning 8 to 24 hours after the insult, with continuous wave or polyspike-and-wave discharges, sometimes correlating
irregular generalized or fragmentary myoclonic jerks (188). with clinical myoclonic jerks, occurring singly or in brief clus-
This MSE is often refractory to therapy and almost always has a ters (193). In MSE, the EEG often shows intermittent 1- to 2-
poor outcome (175,189,190). Sometimes, the MSE may be second bursts of epileptiform discharges, with an intervening
abolished by AEDs, but without improvement in outcome. normal background.
Chronic postanoxic action myoclonus is the Lance–Adams syn- In secondary epilepsy forms, the EEG background is often
drome: segmental or generalized myoclonic jerks typically pre- slow, reflecting the encephalopathy (Fig. 28.15). Spike-wave dis-
cipitated by action or stimulation (191,192). EEG patterns vary charges may come in irregular bursts, often with a slower 2 to
but often include irregular centrally maximal polyspike-and- 2.5 Hz appearance. In both primary and secondary cases, there
wave discharges. is often a poor correlation between the epileptiform discharges
The EEG can be extremely helpful in distinguishing among and the myoclonic jerking activity. In symptomatic generalized
different causes of MSE, whether related to a particular primary epilepsies with MSE, such as LGS, there is usually a background
or secondary epilepsy syndrome (in which the EEG findings are encephalopathy and more prolonged irregular spike-and-wave
often an important component of the syndrome) or due to a or polyspike-and-wave discharges, but with prominent inter-
nonepileptic encephalopathy (Figs. 28.13–28.17). In the IGE vening frontally maximal delta activity (Fig. 28.16). In MSE due
syndromes, the EEG is often definitive and may show frequent to encepahalopathies, the EEG might have no simple spike-
Chapter 28 ■ Convulsive Status Epilepticus 587

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

Figure 28.16 Generalized polyspikes at approximately 2 Hz during myoclonic status epilepticus in a 16-year-old boy with
Lennox–Gastaut syndrome.

wave discharges but often shows somewhat arrhythmic, slower tions that can diminish the myoclonic jerking, but they do not
spikes on a slow background. In postanoxic MSE, the EEG alter the ultimate outcome.
background shows severe voltage attenuation or a burst-sup-
pression pattern, often with superimposed GPEDs (Fig. 28.17). Prognosis and Outcome
The characteristic suppressed (often nearly flat) background The prognosis in status myoclonicus (without evidence of
augurs poorly for prognosis. epileptic pathophysiology) follows the etiology. It is particularly
ominous after anoxia but also poor in metabolic encephalopathies
Treatment and infection (174). In one study of 107 patients with comatose
Myoclonic SE in the IGE syndromes usually responds well to after cardiac resuscitation, myoclonus status was present in 37%
AEDs (175), and episodes are often interrupted expeditiously (188). All patients died, although 70% of resuscitated patients
by BDZs (178). Intravenous valproate may also be effective without myoclonus status also died. The authors argued that
(193), and enteral ethosuximide is another possible treatment. this was a reliable sign that anoxic status myoclonicus had a
Primary generalized epilepsies with prominent myoclonic hopeless prognosis and should not be treated as epilepsy. There
components may be aggravated by use of inappropriate AEDs, have been a few reports of patients recovering from status
particularly including sodium channel-blocking agents such myoclonicus, but most of those were resuscitated almost imme-
as carbamazepine and phenytoin, as well as those with puta- diately after primarily respiratory (rather than cardiac) failure,
tive GABA-ergic mechanisms including gabapentin, tiagabine, and several had a subsequent Lance–Adams syndrome, with
and others (194,195). action myoclonus, but with a reasonably favorable cognitive
Appropriate management of other forms of MSE depends outcome (191,196,197).
overwhelmingly on etiology and can be much more difficult. The outcome of MSE is also determined strongly by the eti-
The MSE is often interruptible, but the prognosis corresponds ology. Patients with MSE in the IGE syndromes tend to
to that of the underlying syndrome or illness. For MSE in the respond to AEDs well, and the prognosis is usually good. In
setting of anoxia, BDZs and valproic acid are among medica- “secondary epileptic syndromes” such as the myoclonic
588 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2
75 uV 1 sec

Figure 28.17 A 50-year-old man with myoclonic status epilepticus after cardiac arrest. Both generalized polyspikes
and EMG artifact from myoclonic jerks can be seen. The EEG between myoclonic jerks shows severe diffuse voltage
attenuation.

epilepsies of childhood, episodes can remit and the patient other seizure types. Infrequently, TSE arises in patients with
returns to baseline—but those baselines may be quite abnor- IGEs (199).
mal. In storage diseases and progressive illness, the SE may TSE consists of maintenance of a tonic posture, particularly
stop, but the underlying disease progresses. The prognosis is of axial and often, proximal limb, musculature rather than con-
worst for patients with MSE due to an acute new illness and is vulsions (198). In clinically obvious cases, there are usually brief
particularly poor following anoxia (189). Anoxic MSE is (10 to 15 seconds) tonic contractions of the face and axial mus-
nearly always fatal—determined by the anoxic damage, rather culature, in the extremities or in combinations of these. Motor
than by any epileptic phenomenon (190). Under 5% recover activity may be very subtle, showing only mild tonic contrac-
reasonably, but hypothermia may improve the prognosis tions of paraspinal musculature or upward deviation of the
somewhat (104). eyes. Tonic seizures usually last less than 10 seconds but can
recur hundreds of times in a day. TSE can be difficult to inter-
CLONIC AND TONIC STATUS EPILEPTICUS rupt with AEDs (200), and BDZs may precipitate or worsen
TSE in some cases (201).
Clonic SE is relatively rare in adults and is seen primarily in EEG may be necessary to make the diagnosis of TSE, given
infants and children. EEG patterns are variable; some show the frequently subtle presentation. It often shows widespread
rhythmic bilateral bursts of high-amplitude delta activity with symmetric fast activity or very rapid spikes but may also include
intermixed spikes or polyspikes (11) (Fig. 28.14), often occur- periods of background suppression or attenuation, generalized
ring synchronously with clonic jerking. electrodecremental activity (voltage suppression), or brief runs
Generalized tonic status epilepticus (TSE) occurs predom- of low-voltage generalized fast activity (Fig. 28.18). The
inantly in young patients with major neurologic and cogni- amplitude of the ictal discharge gradually increases and the
tive deficits or other severe encephalopathies from birth or frequency decreases, sometimes followed by rhythmic general-
early childhood (198). TSE is rare in adults in the absence of ized spikes (47).
Chapter 28 ■ Convulsive Status Epilepticus 589

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Ecg k2-K1
100 uv 1 sec

Figure 28.18 Tonic status epilepticus in a 48-year-old man with Lennox–Gastaut syndrome. Forty-eight hours after initi-
ation of rufinamide, he developed recurrent tonic seizures every 5 minutes, without recovery to baseline between seizures.
EEG showed a high-voltage generalized slow wave, followed by a generalized electrodecremental period with superim-
posed EMG activity.

REFERENCES 7. Clark LP, Prout TP. Status epilepticus: a clinical and pathological
study in epilepsy [An article in 3 parts.] Am J Insanity. 1903/4;60:
1. Shorvon S. The concept of status epilepticus and its history. 291–306; 60:645–675; 61:81–108.
Chapter 1. In: Shorvon S, ed. Status Epilepticus: Its Clinical Features 8. Treiman DM, Walton NY, Kendrick C. A progressive sequence of
and Treatment in Children and Adults. Cambridge: Cambridge electroencephalographic changes during generalized convulsive
University Press; 1994:1–20. status epilepticus. Epilepsy Res. 1990;5:49–60.
2. XXV-XXVIth tablet of the Sakkiku cuneiform, 718-612 BC. British 9. Shorvon S. Definition, classification and frequency of status epilep-
Museum. Kinnier Wilson, JV, Reynolds EH. Translation and analy- ticus. Chapter 2. In: Shorvon S, ed. Status Epilepticus: Its Clinical
sis of a cuneirform text forming part of a Babylonian treatise on Features and Treatment in Children and Adults. Cambridge:
epilepsy. Medical History. 1990;34,185–198. Cambridge University Press; 1994:21–33.
3. Temkin O, ed. The Falling Sickness. Baltimore: The Johns Hopkins 10. Gloor P. The work of Hans Berger. In: Cobb WA, ed. Appraisal and
Press; 1971:149. Perspective of the Functional Explorations of the Nervous System.
4. Willis T. Pathologiae cerebri et nervosi generis specimen. In: quo Handbook of Electroencephalography and Clinical Neurophysiology.
agitur de morbis convulsivis et de scorbuto, 1667, translated by S. Amsterdam: Elsevier Publishing Co.; 1971:IA11–IA24.
Pordage, 1681. London: Dring. Cited in: Shorvon S. ref 1 above; 11. Gastaut H. Classification of status epilepticus. Adv Neurol.
Chapter 1. 1983;34:15–35.
5. Calmeil LF. De l’épilepsie, étudiée sous le rapport de son siege et 12. Commission on Epidemiology and Prognosis, International
de son influence sur la production de l’aliénation mentale. Paris: League Against Epilepsy. Guidelines for Epidemiologic Studies on
Thèse de Université de Paris, 1824. Epilepsy. Epilepsia. 1993;34:592–596.
6. Trousseau A. Lectures on clinical medicine delivered at the Hôtel 13. DeLorenzo RJ, Hauser WA, Towne AR, et al. A prospective, popula-
Dieu, Paris, 1868 vol. 1, translated by Bazire, PV. London: New tion-based epidemiologic study of status epilepticus in Richmond,
Sydenham Society. Virginia. Neurology. 1996;46:1029–1035.
590 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

14. Treiman DM, Meyers PD, Walton NY, et al. A comparison of four 36. Knake S, Rosenow F, Vescovi M, et al. Incidence of status epilepti-
treatments for generalized convulsive status epilepticus. N Engl J cus in adults in Germany: a prospective, population-based study.
Med. 1998;339:792–798. Epilepsia. 2001;42:714–718.
15. Lowenstein DH, Bleck T, Macdonald RL. It’s time to revise the def- 37. Sadarangani M, Seaton C, Scott JAG, et al. Incidence and outcome
inition of status epilepticus. Epilepsia. 1999;40:120–122. of convulsive status epilepticus in Kenyan children: a cohort study.
16. DeLorenzo RJ, Garnett LK, Towne AR, et al. Comparison of status Lancet Neurol. 2008;7:145–150.
epilepticus with prolonged seizure episodes lasting from 10 to 29 38. DeLorenzo RJ, Pellock JM, Towne AR, et al. Epidemiology of sta-
minutes. Epilepsia. 1999;40:164–169. tus epilepticus. J Clin Neurophysiol. 1995;12:316–325.
17. Theodore WH, Porter RJ, Albert P, et al. The secondarily general- 39. Logroscino G, Hesdorffer DC, Cascino G, et al. Short-term mor-
ized tonic-clonic seizure: a videotape analysis. Neurology. 1994;44: tality after a first episode of status epilepticus. Epilepsia. 1997;38:
1403–1407. 1344–1349.
18. Chen JWY, Wasterlain CG. Status epilepticus: pathophysiology 40. Hesdorffer DC, Logroscino G, Cascino GD, et al. Recurrence of
and management in adults. Lancet Neurol. 2006;5:246–256. afebrile status epilepticus in a population-based study in Rochester,
19. Kapur J, Macdonald RL. Rapid seizure-induced reduction of ben- Minnesota. Neurology. 2007;69:73–78.
zodiazepine and Zn 2 sensitivity of hippocampal dentate gran- 41. Shorvon S, Walker M. Status epilepticus in idiopathic generalized
ule cell GABA-A receptors. J Neurosci. 1997;17:7532–7540. epilepsy. Epilepsia. 2005;46:73–79.
20. Kapur J. Pathophysiology of nonconvulsive status epilepticus. 42. Aminoff MJ, Simon RP. Status epilepticus: causes, clinical features,
Chapter 7. In: Kaplan PW, Drislane FW, eds. Nonconvulsive Status and consequences in 98 patients. Am J Med. 1980;69:657–666.
Epilepticus. New York: Demos Medical Publishing; 2009:81–94. 43. Barry E, Hauser W. Status epilepticus: the interaction of epilepsy
21. Mazarati A, Lu X. Regulation of limbic status epilepticus by hip- and acute brain disease. Neurology. 1993;43:1473–1478.
pocampal galanin type 1 and type 2 receptors. Neuropeptides. 2005; 44. Treiman DM. Electroclinical features of status epilepticus. J Clin
39:277–280. Neurophysiol. 1995;12:343–362.
22. Naylor DE, Liu H, Wasterlain CG. Trafficking of GABA(A) recep- 45. Gastaut H, Broughton R. Epileptic seizures. Clinical and electro-
tors, loss of inhibition, and a mechanism for pharamacoresistance graphic features, diagnosis and treatment. Charles C Thomas.
in status epilepticus. J Neurosci. 2005;25:7724–7733. Springfield, IL, 1972.
23. Goodkin HP, Yeh JL, Kapur J. Status epilepticus increases the intra- 46. Niedermeyer E. Epileptic seizure disorders. Chapter 27. In:
cellular accumulation of GABA receptors. J Neurosci. 2005;25: Niedermeyer E, Lopes da Silva F, eds. Electroencephalography.
5511–5520. Basic Principles, Clinical Applications, and Related Fields. 5th ed.
24. Lowenstein DH, Alldredge BK. Status epilepticus at an urban pub- Philadelphia, PA: Lippincott, Williams & Wilkins; 2005:
lic hospital in the 1980’s. Neurology. 1993;43:483–488. 505–619.
25. Bertram EH, Lothman EW. NMDA receptor antagonists and lim- 47. Roger J, Lob H, Tassinari CA. Status epilepticus. In: Vinken P, Bruyn
bic status epilepticus: A comparison with standard anticonvul- G, eds. Handbook of Clinical Neurology, Vol 15, The Epilepsies.
sants. Epilepsy Res. 1990;5:177–184. Amsterdam: North Holland Publishing Co.; 1974:145–188.
26. Wasterlain CG, Liu H, Mazarati AM, et al. Self-sustaining status 48. Bragin A, Engel J Jr, Wilson CL, et al. Hippocampal and entorhinal
epilepticus: a condition maintained by potentiation of glutamate cortex high-frequency oscillations (100–500 Hz) in human epilep-
receptors and by plastic changes in substance P and other peptides tic brain and in kainic acid-treated rats with chronic seizures.
neuromodulators. Epilepsia. 2000;41:134–143. Epilepsia. 1999;40:127–137.
27. Fountain NB. Cellular damage and the neuropathology of status 49. Drislane FW, Schomer DL. Clinical implications of generalized
epilepticus. Chapter 9. In: Drislane FW, ed. Status Epilepticus, a electrographic status epilepticus. Epilepsy Res. 1994;19:111–121.
Clnical Perspective. Totowa, NJ: Humana Press; 2005:181–193. 50. Lowenstein D, Aminoff M. Clinical and EEG features of status
28. Williamson JM, Lothman EW. The effect of MK-801 on kindled epilepticus in comatose patients. Neurology. 1992;42:100–104.
seizures: implications for use and limitations as an antiepileptic 51. DeLorenzo RJ, Waterhouse EJ, Towne AR, et al. Persistent noncon-
drug. Ann Neurol. 1989;26:85–90. vulsive status epilepticus after the control of convulsive status
29. Mazarati AM, Wasterlain CG. N-methyl-D-aspartate receptor epilepticus. Epilepsia. 1998;39:833–840.
antagonists abolish the maintenance phase of self-sustaining sta- 52. Nei M, Lee JM, Shanker VL, et al. The EEG and prognosis in status
tus epilepticus in rat. Neurosci Lett. 1999;265:187–190. epilepticus. Epilepsia. 1999;40:157–163.
30. Lowenstein DH, Shimosaka S, So YT, et al. The relationship between 53. Drislane FW, Blum AS, Schomer DL. Focal status epilepticus: clin-
electrographic seizure activity and neuronal injury. Epilepsy Res. ical features and significance of different EEG patterns. Epilepsia.
1991;10:49–54. 1999;40:1254–1260.
31. Meldrum BS, Horton RW. Physiology of status epilepticus in pri- 54. Waterhouse EJ, Garnett LK, Towne AR, et al. Prospective popula-
mates. Arch Neurol. 1973;28:1–9. tion-based study of intermittent and continuous convulsive status
32. Meldrum BS, Brierley JB. Prolonged epileptic seizures in primates. epilepticus in Richmond, Virginia. Epilepsia. 1999;40:752–758.
Ischemic cell change and its relation to ictal physiological events. 55. Boggs JG, Painter JA, DeLorenzo RJ. Analysis of electrocardio-
Arch Neurol. 1973;28:10–17. graphic changes in status epilepticus. Epilepsy Res. 1993;14:87–94.
33. Meldrum BS, Vigouroux RA, Brierley JB. Systemic factors and 56. Lothman E. The biochemical basis and pathophysiology of status
epileptic brain damage. Arch Neurol. 1973;29:82–87. epilepticus. Neurology. 1990;40(suppl 2):13–23.
34. Lothman EW, Bertram EH, Bekenstein JW, et al. Self-sustaining 57. Brown JK, Husain IHMI. Status epilepticus. I: pathogenesis. Dev
limbic status epilepticus induced by ‘continuous’ hippocampal Med Child Neurol. 1991;35:3–17.
stimulation: electrographic and behavioral characteristics. Epilepsy 58. Cole AJ. Status epilepticus and periictal imaging. Epilepsia.
Res. 1989;3:107–119. 2004;45(suppl 4):72–77.
35. DiGiorgio CM, Tomiyasu U, Gott PS, et al. Hippocampal pyrami- 59. Pohlmann-Eden B, Gass A, Peters CAN, et al. Evolution of MRI
dal cell loss in human status epilepticus. Epilepsia. 1992;33:23–27. changes and development of bilateral hippocampal sclerosis during
Chapter 28 ■ Convulsive Status Epilepticus 591

long lasting generalised status epilepticus. J Neurol Neurosurg 82. Shorvon S, Baulac M, Cross H, et al. For the TaskForce on Status
Psychiatry. 2004;75:898–900. Epilepticus of the ILAE Commission for European Affairs.
60. Maytal J, Shinnar S, Moshe SL, et al. Low morbidity and mortality Epilepsia. 2008;49:1277–1284.
of status epilepticus in children. Pediatrics. 1989;83:323–331. 83. Leppik IE, Derivan AT, Homan RW, et al. Double-blind study of
61. Dodrill CB, Wilensky AJ. Intellectual impairment as an outcome of lorazepam and diazepam in status epilepticus. JAMA. 1983;249:
status epilepticus. Neurology. 1990;40:(suppl 2):23–27. 1452–1454.
62. Adachi N, Kanemoto K, Muramatsu R, et al. Intellectual prognosis 84. Lowenstein DH, Alldredge BK. Status epilepticus. N Engl J Med.
of status epilepticus in adult patients: analysis with Wechsler Adult 1998;338:970–976.
Intelligence Scale-Revised. Epilepsia. 2005;46:1502–1509. 85. Shaner DM, McCurdy SA, Herring MO, et al. Treatment of status
63. Shinnar S, Babb TL, Moshé SL, et al. Long-term sequelae of status epilepticus: a prospective comparison of diazepam and phenytoin
epilepticus. Chapter 65. In: Engel J, Pedley TA, eds. Epilepsy: A versus phenobarbital and optional phenytoin. Neurology. 1988;38:
Comprehensive Textbook. 2nd ed. Philadelphia: Lippincott-Raven 202–207.
Publishers; 2008:751–759. 86. Alldredge BK, Gelb AM, Isaacs SM, et al. A comparison of
64. Hesdorffer D, Logroscino G, Cascino G, et al. Incidence of status lorazepam, diazepam, and placebo for the treatment of out-of-
epilepticus in Rochester, Minnesota, 1965–1984. Neurology. 1998; hospital status epilepticus. N Engl J Med. 2001;345:631–637.
50:735–741. 87. Knake S, Hamer HM, Rosenow F. Status epilepticus: a critical
65. Eriksson KJ, Koivikko MJ. Status epilepticus in children: aetiology, review. Epilepsy Behav. 2009;15:1–14.
treatment, and outcome. Dev Med Child Neurol. 1997;39:652–658. 88. Bleck TP. Critical care of the patient in status epilepticus. In:
66. Berg AT, Shinnar S. Do seizures beget seizures? An assessment of the Wasterlain C, Treiman DT, eds. Status Epilepticus. Boston: MIT
clinical evidence in humans. J Clin Neurophysiol. 1997;14:102–110. Press; 2006:607–613.
67. Hauser WA, Rich SS, Annegers JF, et al. Seizure recurrence after a 89. Krishnamurthy KB, Drislane FW. Relapse and survival after bar-
1st unprovoked seizure: an extended follow-up. Neurology. 1990; biturate anesthetic treatment of refractory status epilepticus.
40:1163–1170. Epilepsia. 1996;37:863–867.
68. DeLorenzo RJ, Towne AR, Pellock JM, et al. Status epilepticus in 90. Claassen J, Hirsch LJ, Emerson RG, et al. Treatment of refractory
children, adults and the elderly. Epilepsia. 1992;33(suppl 4):15–25. status epilepticus with pentobarbital, propofol, or midazolam: a
69. Oxbury JM, Whitty CWM. Causes and consequences of status systematic review. Epilepsia. 2002;43:146–153.
epilepticus in adults. A study of 86 cases. Brain. 1971;94:733–744. 91. Jagoda A, Riggio S. Refractory status epilepticus in adults. Ann
70. Hauser WA. Status epilepticus: epidemiologic considerations. Emerg Med. 1993;22:1337–1348.
Neurology. 1990;40(suppl 2):9–13. 92. Prasad A, Worrall BB, Bertram EH, et al. Propofol and midazo-
71. Waterhouse EJ, Vaughan JK, Barnes TY, et al. Synergistic effect of lam in the treatment of refractory status epilepticus. Epilepsia.
status epilepticus and ischemic brain injury on mortality. Epilepsy 2001;42:380–386.
Res. 1998;29:175–183. 93. Sahin M, Menache CC, Holmes GL, et al. Outcome of severe refrac-
72. Rossetti AO, Hurwitz S, Logroscino G, et al. Prognosis of status tory status epilepticus in children. Epilepsia. 2001;42:1461–1467.
epilepticus: role of aetiology, age, and consciousness impairment 94. Lambrechtsen FACP, Buchhalter JR. Aborted and refractory
at presentation. J Neuro Neurosurg Psychiatry. 2006;77:611–615. status epilepticus in children: a comparative analysis. Epilepsia.
73. DeLorenzo RJ. Clinical and epidemiologic study of status epilepticus 2008;49:615–625.
in the elderly. Chapter 15. In: Rowan AJ, Ramsay RE, eds. Seizures and 95. Legriel S, Mourvillier B, Bele N, et al. Outcomes in 140 critically
Epilepsy in the Elderly. Boston: Butterworth-Heinemann; 1997: ill patients with status epilepticus. Intensive Care Med. 2008;34:
191–205. 476–480.
74. Logroscino G, Hesdorffer DC, Cascino GD, et al. Long-term mor- 96. Sahin M, Menache CC, Holmes GL, et al. Prolonged treatment
tality after a first episode of status epilepticus. Neurology. 2002;58: for acute symptomatic refractory status epilepticus: outcome in
537–541. children. Neurology. 2003;61:398–401.
75. Mayer SA, Claassen J, Lokin J, et al. Refractory status epilepticus. 97. Holtkamp M, Othman J, Buchheim K, et al. A “malignant” vari-
Frequency, risk factors, and impact on outcome. Arch Neurol. ant of status epilepticus. Arch Neurol. 2005;62:1428–1431.
2002;59:205–210. 98. Kalita J, Nair PP, Misra UK. Status epilepticus and encephalitis. A
76. Scholtes FB, Renier WO, Meinardi H. Generalized convulsive sta- study of clinical findings, magnetic resonance imaging and
tus epilepticus: causes, therapy, and outcome in 346 patients. response to antiepileptic drugs. J Neurovirol. 2008;14:412–417.
Epilepsia. 1994;35:1104–1112. 99. Claassen J, Hirsch LJ, Emerson RG, et al. Continuous EEG mon-
77. Towne AR, Pellock JM, Ko D, et al. Determinants of mortality in itoring and midazolam infusion for refractory nonconvulsive sta-
status epilepticus. Epilepsia. 1994;35:27–34. tus epilepticus. Neurology. 2001;57:1036–1042.
78. Drislane FW, Blum AS, Lopez MR, et al. Duration of refractory 100. Yaffe K, Lowenstein DH. Prognostic factors for pentobarbital
status epilepticus and outcome: loss of prognostic utility after sev- therapy for refractory generalized status epilepticus. Neurology.
eral hours. Epilepsia. 2009;50:1566–1571. 1993;43:895–900.
79. Vignatelli L, Rinaldo R, Baldin E, et al. Impact of treatment on the 101. Osorio I, Reed RC, Peltzer JN. Refractory idiopathic status
short-term prognosis of status epilepticus in two population- epilepticus: a probable paradoxical effect of phenytoin and car-
based cohorts. J Neurol. 2008;255:197–204. bamazepine. Epilepsia. 2000;41:887–894.
80. Cascino GD, Hesdorffer D, Logroscino G, et al. Treatment of non- 102. Krishnamurthy KB, Drislane FW. Phenobarbital and benzodi-
febrile status epilepticus in Rochester, Minn, from 1965 through azepine assisted withdrawal of prolonged pentobarbital treatment
1984. Mayo Clin Proc. 2001;76:39–41. for refractory status epilepticus. J Epilepsy. 1997;10:211–214.
81. Logroscino G, Hesdorffer DC, Cascino G, et al. Time trends in 103. Mirski MA, Williams MA, Hanley DF. Prolonged pentobarbital
incidence, mortality, and case-fatality after first episode of status and phenobarbital coma for refractory generalized status epilep-
epilepticus. Epilepsia. 2001;42:1031–1035. ticus. Crit Care Med. 1995;23:400–404.
592 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

104. Rossetti AO, Oddo M, Liaudet L, et al. Predictors of awakening 126. Ferrie CD, Caraballo R, Covanis A, et al. Autonomic status epilep-
from postanoxic status epilepticus after therapeutic hypother- ticus in Panayiotopoulos syndrome and other childhood and
mia. Neurology. 2009;72:744–749. adult epilepsies: a consensus view. Epilepsia. 2007;48:1165–1172.
105. Cooper AD, Britton JW, Rabinstein AA. Functional and cognitive 127. Wieser HG, Chauvel P. Simple partial status epilepticus and
outcome in prolonged refractory status epilepticus. Arch Neurol. epilepsia partialis continua of Kozhevnikov. Chapter 61. In: Engel
2009;66:1505–1509. J, Pedley TA, eds. Epilepsy: A Comprehensive Textbook. 2nd ed.
106. Claassen J, Hirsch LJ, Mayer SA. Treatment of status epilepticus: Philadelphia: Lippincott-Raven Publishers; 2008:705–723.
a survey of neurologists. J Neurol Sci. 2003;211:37–41. 128. Schomer DL. Focal status epilepticus and epilepsia partialis
107. Holtkamp M. The anaesthetic and intensive care of status epilep- continua in adults and children. Epilepsia. 1993;34(suppl 1):
ticus. Curr Opin Neurol. 2007;20:188–193. S29–S36.
108. Mirsattari SM, Sharpe MD, Young GB. Treatment of refractory 129. Fejerman N, DiBlasi AM. Status epilepticus of benign partial
status epilepticus with inhalational anesthetic agents isoflurane epilepsies in children: Report of two cases. Epilepsia. 1987;28:
and desflurane. Arch Neurol. 2004;61:1254–1259. 351–355.
109. Sheth RD, Gidal BE. Refractory status epilepticus: response to 130. Rumbach L, Sablot D, Berger E, et al. Status epilepticus and
ketamine. Neurology. 1998;51:1765–1766. stroke. Report on a hospital-based stroke cohort. Neurology. 2000;
110. Ng Y, Kim HL, Wheless JW. Successful neurosurgical treatment of 54:350–354.
childhood complex partial status epilepticus with focal resection. 131. Scholtes FB, Renier WO, Meinardi H. Simple partial status
Epilepsia. 2003;44:468–471. epilepticus: causes, treatment, and outcome in 47 patients. J
111. Desbiens R, Berkovic SF, Dubeau F, et al. Life-threatening focal Neurol Neurosurg Psychiatry. 1996;61:90–92.
status epilepticus due to occult cortical dysplasia. Arch Neurol. 132. Devinsky O, Kelley K, Porter RJ, et al. Clinical and electroen-
1993;50:695–700. cephalographic features of simple partial seizures. Neurology. 1988;
112. Alexopoulos A, Lachhwani DK, Gupta A, et al. Resective surgery 38:1347–1352.
to treat refractory status epilepticus in children with focal epilep- 133. Grand’Maison F, Reiher J, Leduc CP. Retrospective inventory of
togenesis. Neurology. 2005;64:567–570. EEG abnormalities in partial status epilepticus. Electroencephalogr
113. Lhatoo SD, Alexopoulos AV. The surgical treatment of status Clin Neurophysiol. 1991;79:264–270.
epilepticus. Epilepsia. 2007;48(suppl 8):61–65. 134. Juul-Jensen P, Denny-Brown D. Epilepsia partialis continua. A
114. D’Giano CH, Garcia MDC, Pomata H, et al. Treatment of refrac- clinical, electroencephalographic, and neuropathological study
tory partial status epilepticus with multiple subpial transaction: of nine cases. Arch Neurol. 1966;15:563–578.
case report. Seizure. 2001;10:382–385. 135. Thomas JE, Reagan TJ, Klass DW. Epilepsia partialis continua. A
115. Spencer SS, Schramm J, Wyler A, et al. Multiple subpial transac- review of 32 cases. Arch Neurol. 1977;34:266–275.
tion for intractable partial epilepsy: an international meta-analy- 136. Cockerell OC, Rothwell J, Thompson PD, et al. Clinical and phys-
sis. Epilepsia. 2002;43:141–145. iological features of epilepsia partialis continua. Cases ascer-
116. Krishnamurthy KB, Drislane FW. Depth of EEG suppression and tained in the UK. Brain. 1996;119(Pt 2):393–407.
outcome in barbiturate anesthetic treatment for refractory status 137. Kozhevnikov A. Eine besondere Form von corticaler Epilepsie.
epilepticus. Epilepsia. 1999;40:759–762. Neurol Centralbl. 1895;14:47–48.
117. Kaplan PW. Assessing the outcomes in patients with nonconvul- 138. Botez MI, Brossard L. Epilepsia partialis continua with well-
sive status epilepticus: nonconvulsive status epilepticus is under- delimited subcortical frontal tumor. Epilepsia. 1974;15:39–43.
diagnosed, potentially overtreated, and confounded by 139. Feddersen B, Bender A, Arnold S, et al. Aggressive confusional
comorbidity. J Clin Neurophysiol. 1999;16:341–352. state as a clinical manifestation of status epilepticus in MELAS.
118. Rossetti AO, Logroscino G, Bromfield EB. Refractory status Neurology. 2003;61:1149–1150.
epilepticus: effect of treatment aggressiveness on prognosis. Arch 140. Shavit YB, Graus F, Probst A, et al. Epilepsia partialis continua: a
Neurol. 2005;62:1698–1702. new manifestation of anti-Hu-associated paraneoplastic
119. Rashkin MC, Youngs C, Penovich P. Pentobarbital treatment of encephalomyelitis. Ann Neurol. 1999;45:255–258.
refractory status epilepticus. Neurology. 1987;37:500–503. 141. Ruggieri PM, Najm I, Bronen R, et al. Neuroimaging of the cor-
120. Knake S, Rochon J, Fleischer S, et al. Status epilepticus after stroke tical dysplasias. Neurology. 2004;62(suppl 3):S27–S29.
is associated with increased long-term case fatality. Epilepsia. 142. Singh BM, Strobos RJ. Epilepsia partialis continua associated
2006;47:2020–2026. with nonketotic hyperglycemia: clinical and biochemical profile
121. Jaitly R, Sgro JA, Towne AR, et al. Prognostic value of EEG mon- of 21 patients. Ann Neurol. 1980;8:155–160.
itoring after status epilepticus: a prospective adult study. J Clin 143. Kuroiwa Y, Tohgi H, Takahashi A, et al. Epilepsia partialis con-
Neurophysiol. 1997;14:326–334. tinua: active cortical spike discharges and high blood flow in the
122. Husain AM, Mebust KA, Radtke RA. Generalized periodic motor cortex and enhanced transcortical long loop reflex. J
epileptiform discharges: etiologies, relationship to status epilep- Neurol. 1985;232:162–166.
ticus, and prognosis. J Clin Neurophysiol. 1999;16:51–58. 144. Wieser HG, Graf HP, Bernoulli C, et al. Quantitative analysis of
123. Claassen J, Mayer SA, Kowalski RG, et al. Detection of electro- intracerebral recordings in epilepsia partialis continua.
graphic seizures with continuous EEG monitoring in critically ill Electroencephalogr Clin Neurophysiol. 1978;44:14–22.
patients. Neurology. 2004;62:1743–1748. 145. Watanabe K, KuroiwaY, Toyokura A. Epilepsia partialis continua.
124. Jette N, Claassen J, Emerson RG, et al. Frequency and predictors Epileptogenic focus in motor cortex and its participation in
of nonconvulsive seizures during continuous electroencephalo- transcortical reflexes. Arch Neurol. 1984;41:1040–1044.
graphic monitoring in critically ill children. Arch Neurol. 2006; 146. Cowan JMA, Rothwell JC, Wise RJS, et al. Electrophysiological
63:1750–1755. and positron emission studies in a patient with cortical
125. Hirsch LJ. Continuous EEG monitoring in the intensive care myoclonus, epilepsia partialis continua and motor epilepsy. J
unit: an overview. J Clin Neurophysiol. 2004;21:332–340. Neurol Neurosurg Psychiatr. 1986;49:796–807.
Chapter 28 ■ Convulsive Status Epilepticus 593

147. Kristiansen K, Kaada BR, Henriksen GF. Epilepsia partialis con- association with seizures. Electroencephalogr clin Neurophysiol.
tinua. Epilepsia. 1971;12:263–267. 1991;78:12–17.
148. Bancaud J, Bonis A, Trottier S, et al. L’épilepsie partielle continue: 168. Ali II, Pirzada NA, Vaughn BV. Periodic lateralized epileptiform
syndrome et maladie. Rev Neurol (Paris). 1982;138:803–814. discharges after complex partial status epilepticus associated with
149. Morrell F, Whisler WW, Bleck TP. Multiple subpial transection: a increased focal cerebral blood flow. J Clin Neurophysiol.
new approach to the surgical treatment of focal epilepsy. J 2001;18:565–569.
Neurosurg. 1989;32:553–559. 169. Assal F, Papazyan JP, Slosman DO, et al. SPECT in periodic later-
150. Morrell F, Whisler WW, Smith MC. Multiple subpial transaction alized epileptiform discharges (PLEDs): a form of partial status
in Rasmussen’s encephalitis. Chapter 20. In: Andermann F, ed. epilepticus? Seizure. 2001;10:260–265.
Chronic Encephalitis and Epilepsy. Rasmusen’s Syndrome. Boston: 170. Handforth A, Cheng JT, Mandelkern MA, et al. Markedly
Butterworth-Heinemann; 1991:219–233. increased mesiotemporal lobe metabolism in a case with PLEDs:
151. Chatrian GE, Shaw CM, Leffman H. The significance of periodic further evidence that PLEDs are a manifestation of partial status
lateralized epileptiform discharges in EEG: An electrographic, epilepticus. Epilepsia. 1994;35:876–881.
clinical and pathological study. Electroenceph clin Neurophysiol. 171. Terzano MG, Parrino L, Mazzucchi A, et al. Confusional states
1964;17:177–193. with periodic lateralized epileptiform discharges (PLEDs): a
152. Rasmussen T, Olszewski J, Lloyd-Smith DL. Focal seizures due to peculiar epileptic syndrome in the elderly. Epilepsia. 1986;27:
chronic localized encephalitis. Neurology. 1958;8:435–445. 446–457.
153. Oguni H, Andermann F, Rasmussen TB. The natural history of 172. Garzon E, Fernandes RMF, Sakamoto AC. Serial EEG during
the syndrome of chronic encephalitis and epilepsy. A study of the human status epilepticus. Evidence for PLED as an ictal pattern.
MNI series of forty-eight cases. In: Andermann F, ed., Chronic Neurology. 2001;57:1175–1183.
Encephalitis and Epilepsy. Boston: Butterworth-Heinemann; 173. de la Paz D, Brenner RP. Bilateral independent periodic lateral-
1991:7–35. ized epileptiform discharges. Clinical significance. Arch Neurol.
154. Aguilar MJ, Rasmussen T. Role of encephalitis in pathogenesis of 1981;38:713–715.
epilepsy. Arch Neurol. 1960;2:663–676. 174. Celesia GG, Grigg MM, Ross E. Generalized status myoclonicus in
155. Rasmussen T. Further observations on the syndrome of chronic acute anoxic and toxic-metabolic encephalopathies. Arch Neurol.
encephalitis and epilepsy. Appl Neurophysiol. 1978;41:1–12. 1988;45:781–784.
156. So NK, Gloor P. Electroencephalographic and electrocortico- 175. Jumao-as A, Brenner RP. Myoclonic status epilepticus: a clinical and
graphic findings in chronic encephalitis of the Rasmussen type. electroencephalographic study. Neurology. 1990;40:1199–1202.
In: Andermann F, ed. Chronic Encephalitis and Epilepsy. Boston: 176. Hallett M. Myoclonus: relation to epilepsy. Epilepsia. 1985;
Butterworth-Heinemann; 1991:37–46. 26(suppl 1):67–77.
157. Rogers SW, Andrews PI, Gahring LC, et al. Autoantibodies to 177. Wakamoto H, Nagao H, Manabe K, et al. Nonconvulsive status
glutamate receptor GluR3 in Rasmussen’s encephalitis. Science. epilepticus in eyelid myoclonia with absences—evidence of
1996;46:242–246. provocation unrelated to photosensitivity. Neuropediatrics.
158. Hart YM, Cortez M, Andermann F, et al. Medical treatment of 1999;30:149–150.
Rasmussen’s syndrome (chronic encephalitis and epilepsy): effect 178. Ohtahara S, Ohtsuka Y. Myoclonic status epilepticus. Chapter 63.
of high-dose steroids or immunoglobulins in 19 patients. In: Engel J Jr, Pedley TA, eds. Epilepsy: A Comprehensive Textbook.
Neurology. 1994;44:1030–1036. 2nd ed. Philadelphia: Lippincott-Raven Publishers; 2008:725–729.
159. Villemure JG, Vernnet O, Delalande O. Hemispheric disconnec- 179. Asconape J, Penry JK. Some clinical and EEG aspects of benign
tion: callosotomy and hemispherectomy. Adv Tech Stand juvenile myoclonic epilepsy. Epilepsia. 1984;25:108–114.
Neurosurg. 2000;26:25–78. 180. Kimura S, Kobayashi T. Two patients with juvenile myoclonic
160. Westmoreland BF, Klass DW, Sharbrough FW. Chronic periodic epilepsy and nonconvulsive status epilepticus. Epilepsia. 1996;
lateralized epileptiform discharges. Arch Neurol. 1986;43: 37:275–279.
494–496. 181. Adachi M, Inoue T, Tsuneishi S, et al. Eyelid myoclonia with
161. Snodgrass SM, Tsuburaya K, Ajmone-Marsan C. Clinical signifi- absences in monozygotic twins. Pediatr Int. 2005;47:343–347.
cance of periodic lateralized epileptiform discharges: Relationship 182. Wheless JW. Acute management of seizures in the syndromes
with status epilepticus. J Clin Neurophysiol. 1989;6:159–172. of idiopathic generalized epilepsies. Epilepsia. 2003;44(suppl 2):
162. Baykan B, Kinay D, Gokyigit A, et al. Periodic lateralized epilepti- 22–26.
form discharges: association with seizures. Seizure. 2000;9:402–406. 183. Genton P, Gelisse P, Thomas P, et al. Do carbamazepine and
163. Schraeder, PL, Singh N. Seizure disorders following periodic lat- phenytoin aggravate juvenile myoclonic epilepsy? Neurology.
eralized epileptiform discharges. Epilepsia. 1980;21:647–653. 2000;55:1106–1109.
164. Schwartz MS, Prior PF, Scott DR. The occurrence and evolution 184. Berkovic SF, Andermann F, Carpenter S, et al. Progressive
in the EEG of a lateralized periodic phenomenon. Brain. 1973; myoclonus epilepsies: specific causes and diagnosis. N Engl J
96:613–622. Med. 1986;315:296–305.
165. Chong DJ, Hirsch LJ. Which EEG patterns warrant treatment in 185. Dravet C, Bureau M, Oguni H, et al. Severe myoclonic epilepsy in
the critically ill? Reviewing the evidence for treatment of periodic infancy: Dravet syndrome. Adv Neurol. 2005;95:71–102.
epileptiform discharges and related patterns. J Clin Neurophysiol. 186. Korff CM, Laux L, Kelley Kr, et al. Dravet syndrome (severe
2005;22:79–91. myoclonic epilepsy in infancy): a retrospective study of 16 patients.
166. Jirsch J, Hirsch LJ. Nonconvulsive seizures: developing a rational J Child Neurol. 2007;22:185–194.
approach to the diagnosis and management in the critically ill 187. Simon RP, Aminoff MJ. Electrographic status epilepticus in fatal
population. Clin Neurophysiol. 2007;118:1660–1670. anoxic coma. Ann Neurol. 1986;20:351–355.
167. Reiher J, Rivest J, Grand'Maison F, et al. Periodic lateralized 188. Wijdicks EF, Young GB. Myoclonus status in comatose patients
epileptiform discharges with transitional rhythmic discharges: after cardiac arrest. Lancet. 1994;343:1642–1643.
594 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

189. Krumholz A, Stern BJ, Weiss HD. Outcome from coma after car- 196. Arnoldus EPJ, Lammers GJ. Postanoxic coma: good recovery
diopulmonary resuscitation. Neurology. 1988;38:401–405. despite myoclonus status. Ann Neurol. 1995;38:697.
190. Young GB, Gilbert JJ, Zochodne DW. The significance of 197. Morris HR, Howard RS, Brown P. Early myoclonic status and out-
myoclonic status epilepticus in postanoxic coma. Neurology. come after cardiorespiratory arrest. J Neurol Neurosurg Psychiatry.
1990;40:1843–1848. 1998;64:267–268.
191. Brown P, Thompson PD, Rothwell JC, et al. A case of postanoxic 198. Gastaut H, Roger J, Ouahchi S, et al. An electroclinical study of
encephalopathy with cortical action and brainstem reticular generalized epileptic seizures of tonic expression. Epilepsia. 1963;
reflex myoclonus. Mov Disord. 1991;6:139–144. 4:15–44.
192. Sheth RD, Gidal BE. Intravenous valproic acid for myoclonic sta- 199. Kobayashi E, Thomas P, Andermann F. Tonic status epilepticus in
tus epilepticus. Neurology. 2000;54:1201. patients with idiopathic generalized epilepsy. Epileptic Disord.
193. Genton P. When antiepileptic drugs aggravate epilepsy. Brain 2005;7:327–331.
Dev. 2000;22:75–80. 200. Somerville ER, Bruni J. Tonic status epilepticus presenting as a
194. Perucca E, Gram L, Avanzini G, et al. Antiepileptic drugs as a confusional state. Ann Neurol. 1983;13:549–551.
cause of worsening seizures. Epilepsia. 1998;39:5–17. 201. Tassinari CA, Dravet C, Roger J, et al. Tonic status epilepticus pre-
195. Lance JW, Adams RD. The syndrome of intention or action cipitated by intravenous benzodiazepine in five patients with
myoclonus as a sequel to hypoxic encephalopathy. Brain. 1963; Lennox-Gastaut syndrome. Epilepsia. 1972;13:421–435.
86:111–136.
CHAPTER

Nonconvulsive Status Epilepticus


FRANK W. DRISLANE, PETER W. KAPLAN, AND SUSAN T. HERMAN 29
HISTORY NCSE. Clinical descriptions of confusional states (even with
signs of epilepsy such as twitching of facial or limb muscles)
Although generalized convulsive status epilepticus has been were insufficient to prove that NCSE was a proper diagnosis.
recognized for millennia, nonconvulsive status epilepticus Encephalopathy, psychogenic unresponsiveness, or postictal
(NCSE) is a much more recent concept. Perhaps by association behavior might appear very similar.
with other forms of epilepsy with altered consciousness, some Berger’s 1929 invention and development of the electroen-
speculation about the possible existence of NCSE emerged by cephalogram (EEG) made it possible to record the previously
1800 or so. Trousseau, at Hotel Dieu in Paris, noted that petit suspected electrical activity of the brain (9). He also began
mal seizures might recur with sufficient frequency “that one studies correlating EEG activity, as a measure of brain electrical
seizure would become confused with the next, simulating a function, with psychologic and behavioral abnormalities, and
continuous seizure which might persist for two or three days eventually, epileptic seizures (10). Thus, earlier historical
millennia” thus anticipating a diagnosis of absence status descriptions of what might have been NCSE were thrust into a
epilepticus (1–3). Nonconvulsive spells also attracted the atten- provable domain. Just over a decade later, clinical-EEG correla-
tion of Jules Falret (1824 to 1902) who said that a patient with tions showed beyond doubt that NCSE was part of epilepsy and
“petit mal intellectuel … might leave home or work, with not, as some had suspected, from hysterical or nonepileptic
clouded mind … [and] … complete lapses of memory (2,4).” fugue states. The stage was set for modern electroclinical corre-
Samuel Wilks in London in the late 1800s described a patient lation, including video-EEG monitoring, various forms of
who was: “… in the condition which is popularly called ‘lost;’ imaging, and refinement of treatment.
he is scarcely conscious of acts and conversation going on Lennox detailed one of the first cases of “petit mal status”
around him, yet he may continue walking in a given direction, with an electrographic correlate in 1945 (11). In 1954, Penfield
showing that his movements must still, in a measure, be guided and Jasper described simple partial status epilepticus (SPSE) or
by his senses … in much the same state as a somnambulist. This aura continua in the form of recurrent sensory phenomena,
condition … is called the status epilepticus, although the term commenting that they were “at least as common as continuing
is more usually applied to [a] patient … who, after a succession circumscribed movements (12).” In the first thorough descrip-
of fits, lay for hours in a state of lethargy. In the milder forms it tion of complex partial status epilepticus (CPSE) in 1956,
is one of great interest from a physiology point-of-view and Gastaut and Roger described a nurse whose seizures may have
seems to point to the possibility of a subconscious state, in lasted for months (13). In the past half century, there have been
which the brain is sufficiently active to control the spinal system numerous clinical series detailing the electroclinical correlates
and yet not awake [for] consciousness (1,5).” and nature of NCSE.
Also in the late 1800s, Charcot and others described cases
suggestive of NCSE. Charcot postulated that a state of somnam- DEFINITION
bulism derived from ongoing seizures. He presented a patient
with a nonconvulsive form of long-lasting seizures, several It is difficult to state a definition of NCSE, primarily because
times in his “Leçons du Mardi” at the Saltpêtrière—a delivery the clinical manifestations are protean and because the EEG
man who wandered about Paris and even to the coast, at Brest, findings, even while crucial for diagnosis, are often ambiguous
being arrested on one occasion and released only on the cog- or controversial. The significance of epileptiform waveforms
nizance of Charcot—who treated him with bromides (6). In and other abnormalities on the EEG is often unclear, and even
Britain, Gowers speculated that similar states rather than being the role of clearly epileptiform EEG abnormalities in causing
ictal, occurred after the seizure: “After epileptic fits of moderate clinical deficits can be uncertain or impossible to determine.
severity, the patient may pass into a condition of mental Because NCSE is considered an epileptic process without con-
automatism, in which various acts are performed in an appar- vulsions (or no more than minimal motor manifestations),
ently conscious manner, but of which no recollection is after- almost all definitions insist on EEG evidence of continuing or
ward retained (7).” very frequent epileptiform activity.
Charcot’s observations fostered speculation about the exis- The definition of generalized convulsive status epilepticus
tence of NCSE, but without an appreciation of the role of electric- (GCSE) can be complicated and controversial (Chapter 28), but
ity in the nervous system [starting to emerge in the 17th century that for NCSE is much more so. It has been defined as a “range
(8)], it would be hard to conceive a modern understanding of of conditions in which electrographic seizure activity is prolonged

595
596 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

and results in nonconvulsive clinical symptoms. [It is … ] prima- abnormalities. The other illnesses may obscure the diagnosis.
rily as a form of epileptic cerebral response which is dependant Many patients with NCSE have had their compromised cogni-
largely on the level of cerebral development and integrity, the pres- tion attributed to problems such as electrolyte imbalance,
ence or absence of encephalopathy, the type of epilepsy syndrome, hyperglycemia, pneumonia, or earlier seizures (24). Many
and the anatomical location of the epileptic activity (14).” This patients have dementia, mental retardation, or psychiatric dis-
definition reflects the pleomorphic character of NCSE and orders, making it difficult for unfamiliar observers, including
alludes to important insights into its biology, but diagnosis of physicians, to notice a new perturbation in cognition or behav-
individual cases can remain challenging. While there have been ior (19,24–28). It is often difficult to determine if the patient is
several proposals to change a temporal definition of GCSE from “more confused than usual,” “confused beyond what might be
30 to 5 or 10 minutes (15), it is probably most reasonable to use attributable to an infection or metabolic disturbance,” or “inap-
a 30-minute definition for NCSE (16). propriately lethargic after a convulsion.”
Altered alertness or behavior may be ascribed to an
CLINICAL PRESENTATION encephalopathy or a postictal state, delaying diagnosis (19,24).
Patients with chronic psychiatric illness are at risk of delayed
NCSE has remarkably varied and often very subtle presenta- diagnosis of NCSE because of their neuroleptic burden and
tions (17,18). At times, it follows generalized seizures and propensity for starting and stopping benzodiazepines
GCSE, that is, GCSE continues in a more subtle form; the (BDZs)—triggers for de novo NCSE (29). Especially in the eld-
epileptiform discharges of a seizure continue on the EEG after erly, NCSE may present with confusion, suggesting medication
the motor manifestations (convulsions) have ceased. In this side effects or dementia (30,31). Many sick, hospitalized
case, it is best to consider NCSE as a later stage of GCSE, in patients have altered mental status due to medications, infec-
terms of its pathophysiology, clinical implications, and man- tions, and metabolic encephalopathies from their underlying
date for treatment. Any patient who has not recovered from a illnesses, and the same illness may cause or precipitate NCSE
seizure within 20 to 30 minutes or so should be considered as (32,33). Diagnostic clues of NCSE include an earlier history of
possibly in NCSE. epilepsy or risk factors for epilepsy.
NCSE can also occur without clinically evident seizures, that NCSE is typically discovered in the emergency department,
is, purely nonconvulsive events. Even on its own (not following intensive care units (ICUs), and on neurology and psychiatry
GCSE), NCSE has pleomorphic presentations. Its behavioral services, although it can occur during other hospital services
correlates in different types of NCSE arise from—or are at least and even in ambulatory patients. In the emergency department,
associated with—different areas of maximal involvement of the diagnosis of NCSE may be missed when there is lethargy or
seizure activity in the brain. confusion attributed to a postictal state; ictal confusion mis-
NCSE typically encompasses an ictal impairment of mental taken for a metabolic encephalopathy; unresponsiveness and
status, cognition, or behavior compared with a baseline state; catalepsy presumed to be psychogenic; obtundation thought to
automatisms; or change in sensory perception (e.g., auditory, be due to alcohol or drug intoxication; hallucinations and agi-
visual, somatosensory, or psychic) (19). Anterograde and retro- tation mistaken for psychosis or delirium; lethargy presumed
grade memory may be affected; affects can be altered (agitation, due to hyperglycemia; mutism attributed to aphasia; and laugh-
sadness, weeping); speech and language may be impaired (with ing and crying ascribed to emotional lability (24). In hospital-
mutism or paraphasic errors); and confusional states may ized or institutionalized patients, NCSE can present primarily
occur. There can be psychomotor retardation or behavioral as an impaired level of consciousness or responsiveness.
changes, beyond the patient’s baseline. Sometimes, there are Drugs precipitating some cases of NCSE include neuroleptics,
admixed, but generally minimal, motor manifestations, such as cephalosporins, radiologic contrast agents (34), and gamma
subtle facial or limb jerking, blinking, eye deviation with nys- amino butyric acid (GABA) agonists, including baclofen (35),
tagmus (20), or perioral and eyelid myoclonus—highly sugges- tiagabine, and vigabatrin (36,37). Occasionally, refractory NCSE
tive of NCSE in the appropriate clinical setting (21). can be caused by mitochondrial disorders, including mitochon-
Most epidemiologic studies have been focused on the inci- drial encephalomyopathy with lactic acidosis and stroke-like
dence of GCSE. All forms of NCSE may constitute one-fourth episodes (MELAS) (38). Unusual forms of NCSE may result from
of all cases of SE (22), and thus have an incidence of about paraneoplastic syndromes, especially in those likely caused by cell
10/100,000 persons per year in the general population (16). surface antibodies (39). In the anti-NMDA-receptor-associated
About half of all cases in a German study of SE were noncon- encephalitis, patients are particularly likely to be young women
vulsive, implying about 8.5 incident cases per 100,000 popula- with ovarian teratomas, presenting with psychiatric or memory
tion per year (23). Given the difficulties in diagnosis, these disorders, transferred to ICUs with stupor, refractory NCSE, and
estimates of the incidence of NCSE may be far too low. bizarre orofacial dyskinesias; many respond well to immunosup-
pressive treatment (and tumor removal) but may take many
DIFFERENTIAL DIAGNOSIS months to return to normal function (40).
Mimics of NCSE include migraine, transient global amnesia,
NCSE often occurs in patients with underlying chronic medical or sleep disorders, including cataplexy. Others include psychi-
and neurologic illnesses, and also with acute and subacute med- atric disorders, metabolic derangements, paroxysmal cardiovas-
ical problems such as infections, vascular disease, or metabolic cular or autonomic dysfunction, endocrine dysfunction, and
Chapter 29 ■ Nonconvulsive Status Epilepticus 597

limbic encephalitis—whether paraneoplastic or related to volt- Frequently, the response to AEDs is inconclusive, and the diag-
age-gated potassium channel antibodies (39), and confusional nosis must be made on clinical and EEG grounds alone.
states induced by many drugs, such as lithium, other psy- Importantly, lack of a prompt improvement with AEDs does
chotropic medications, or alcohol (24,41). not refute a diagnosis of NCSE. Even in patients with definite
NCSE, a rapid clinical response is uncommon, especially in
OVERVIEW OF DIAGNOSIS obtunded or comatose patients (48). In many, the response is
equivocal or substantially delayed (24,49), and sedating BDZs
While generalized convulsions are usually apparent clinically, may also impair a clinical response. Following a clinical seizure,
nonconvulsive seizures (probably the majority in adults) are if the patient improves on EEG, but not clinically, continuous
harder to recognize. Paradoxically, NCSE may be even harder to EEG (C-EEG) monitoring should be considered to look for evi-
recognize than individual seizures because there may be no sud- dence of subsequent recurrent seizures.
den behavioral change at onset; the patient’s condition may fluc- There have also been a few cases of focal NCSE diagnosed
tuate little over the day; the significance of many EEG patterns is clearly by neuroimaging, for example, with 18F-fluo-
controversial; and the response to antiepileptic drugs (AEDs) rodeoxyglucose (FDG)-positron emission tomography (PET)
may be subtle or delayed. scans (50,51). Some patients in these reports had clear areas of
The diagnosis of NCSE derives from two major elements: an persistent focal hypermetabolism during clinical episodes of
alteration in baseline cognition, behavior, or other neurologic definite aphasia or apparent epileptic amnesia (with an unre-
function, and concurrent epileptiform seizure patterns on the markable EEG, or focal slowing) and an appropriate and quick
EEG (42,43). Satisfying these criteria is often complicated and response to intravenous BDZs. Occasionally, hyperperfusion on
difficult. To recognize the clinical features, one must maintain a ictal single-photon emission computed tomography (SPECT)
high index of suspicion. The sign of NCSE is not merely the may help to make, or confirm, the diagnosis (52).
abnormal behavior, but its change from a baseline state.
To diagnose NCSE, epileptologists have required impaired PATHOPHYSIOLOGY
consciousness for 30 to 60 minutes and an EEG with continuous
(44), or at least some form of, seizure activity (42). EEG wave- The pathophysiology of convulsive status epilepticus is dis-
form morphologies may include rhythmic slowing, sharp waves, cussed in Chapter 28. That of NCSE is substantially less well
spikes, and mixtures of these features (45). Discharges may be understood, both in the generation of neural activity that pro-
continuous, persistent with brief pauses of a few seconds, or duces and sustains NCSE, and in the possibility of its inducing
intermittent. Many EEGs are more ambiguous (see section “EEG subsequent neuronal damage and longer-lasting epilepsy on its
Diagnosis of Nonconvulsive Status Epilepticus”), with more own.
blunted waveforms slower than 1.5 Hz, sometimes resembling Most NCSE, at least in adults, is either CPSE or secondary
triphasic waves (TWs) (43,46). Over time, NCSE patterns tend to generalized NCSE—both with underlying focal onset, whether
evolve in morphology, amplitude and frequency, and wax or evident clinically or not. This NCSE usually begins at the same
wane. EEG ictal discharges should be continuous or recurrent for focus of epileptogenesis as for focal seizures that do not become
more than 30 minutes, without the patient’s return to a normal prolonged to the point of SE. How do these focal seizures reach
clinical state or resumption of the preictal EEG pattern between a self-sustaining state?
seizures. Specific EEG features of individual types of NCSE are In a rodent model of limbic SE produced by repetitive, rapid
detailed in the sections below describing those types of NCSE. direct electrical stimulation of the hippocampus for 30 to 90
Although the EEG is the most reliable diagnostic test for minutes, seizures progress from self-limited to much more pro-
NCSE, an AED challenge may also be useful. Some patients longed, and eventually self-sustaining SE, persisting for 12 to 24
with less “classic” EEG findings show a clinical and electro- hours after the stimulation has ceased (53,54). The induced
graphic response to BDZs or other AEDs (42). In the appropri- seizures appear similar to those of humans with CPSE—
ate clinical setting, a rapid response to AEDs can be persuasive although it is still unclear whether an analogous process occurs
for a diagnosis of NCSE. Lorazepam may be given in small in humans. Seizures beginning in the hippocampus appear to
sequential doses, while monitoring blood pressure, respiratory promote the propagation of yet more seizure activity, which is
rate, and oxygenation. To be diagnostic of ongoing seizure maintained in a circuit of structures including the entorhinal
activity, there should be prompt improvement in both the cortex, dentate gyrus, and parts of the hippocampus and
patient’s clinical state and the EEG, or complete cessation of subiculum (55,56). The initiation of SE in these models was
electrographic ictal activity with return of normal EEG back- similar to that for individual seizures, but inhibitory or termi-
ground activity (43,46) (see Figs. 29.6, 29.14, 29.15, 29.19, and nating mechanisms failed (see Chapter 28). Generalized
29.25). Intravenous BDZs may abolish electrographic seizures, seizures, or seizures with onset in other areas, can also precipi-
but they can also suppress nonepileptic EEG patterns, such as tate focal temporolimbic seizures and focal SE by way of sec-
TWs (47), so improvement in the EEG alone upon AED admin- ondary involvement of the hippocampus or other mesial
istration does not prove that a particular EEG pattern was a temporal structures (57).
seizure. A controversial EEG pattern is confirmed as ictal only The activity of certain neurotransmitters can facilitate or
when marked clinical improvement accompanies the EEG inhibit the activation of complex circuits of limbic neuronal
improvement—but often such an improvement does not occur. activity, prolonging or interrupting nonconvulsive seizures in a
598 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

(somewhat similar) model of self-sustaining SE produced by SE, neurologic damage in the amygdala and pyriform cortex
stimulation of the perforant pathway in rats (58). For example, correlated strongly with prolonged “high-frequency” (10 Hz)
injection of galanin (a peptide that inhibits glutamate release discharges, but there was no damage following discharges
presynaptically) into the hippocampus attenuates seizure activ- slower than 1 Hz (68). Damage was directly related to the dura-
ity (59). Galanin production can decline during SE, while sub- tion and intensity of electrographic seizure activity. It is not
stance P (which may enhance SE by promoting glutamate clear that such prolonged intense stimulation, or the resultant
release) may increase (60), facilitating the prolongation of 10-Hz spike discharges, are an accurate model of human NCSE.
seizures. As with GCSE, the balance of excitatory and inhibitory Also, in the NCSE model with continuous hippocampal stimu-
function seems to shift toward excitation as NCSE progresses. lation, neuronal loss did not occur when the interval between
The pathophysiology of absence SE and other forms of pri- nonconvulsive seizures was increased (69). Currently, little is
marily generalized SE, however, is starkly different from that of known about the pathologic effects of NCSE in humans.
focal-onset NCSE (61). These generalized forms of NCSE Clinical consequences of NCSE are discussed in later sections.
involve excessively prolonged synchronization in widespread
thalamocortical circuits as part of the generation, maintenance, CLASSIFICATION
and reinforcement of aberrant excitatory electrical rhythms in
a neuronal system involving both cortical and subcortical struc- No classification system for NCSE will be satisfactory to all
tures (55,62). These abnormal electrical rhythms are mani- basic investigators, electroencephalographers (EEG-ers), clini-
fested by rhythmic generalized, anteriorly predominant, spike cians, and scholars. Gastaut stated that there were “as many
and polyspike and slow-wave discharges on the EEG. types of status as there are types of epileptic seizures” (70), but
Experimentally, these 3-Hz generalized cortical spike-and the correspondence is not strict. For example, different focal
slow-wave discharges can be produced by electrical stimulation seizures emanate from different brain areas, with different signs
of midline intralaminar nuclei of the thalamus (63), and the and symptoms, but many can go on to generalized convulsions
same thalamic discharges occur during absence seizures in and GCSE, and to subsequent NCSE after convulsions—with
humans (64). The discharges are dependent on low-threshold all later stages appearing similar despite the different origins.
T-type calcium channel activity (55). Thalamic injections of Others reject a classification of NCSE according to seizure
GABA-B agonists increase the spike-and slow-wave discharges, types, pointing out that epilepsy syndromes such as juvenile
and this activity can be suppressed by specific AEDs (55). myoclonic epilepsy (JME) consist of more than seizures types
Little experimental evidence is available on long-term conse- alone, and also that NCSE is not always simply the prolongation
quences of NCSE. Models of repetitive limbic seizures and SE of individual seizures (71). Different NCSE syndromes include
have been produced by local tissue damage caused by injections EEG, clinical, and genetic bases; developmental aspects; age-
of kainic acid, but chemical and electrical methods of inducing specific presentations; varied concomitant structural brain abnor-
SE may damage neurologic tissue independent of the subse- malities or encephalopathies; quite varied responses to treatment;
quent seizures, that is, it may be the local electrical or chemical and different longer-term outcome. Thus, NCSE can also be clas-
precipitant or toxin that damages neurons. sified based primarily on age and the state of cerebral develop-
There are a few clinical reports of patients with hippocam- ment or maturation, presence or absence of an encephalopathy,
pal edema during episodes of NCSE, with subsequent hip- occurrence within an epilepsy syndrome, and, for NCSE of focal
pocampal atrophy in the same area (65,66), but not all studies origin, understanding by anatomic localization (Table 29.1) (14).
show this progression to atrophy, possibly depending in part on This scheme, organized primarily by age of onset, aids in under-
how prolonged the episodes of NCSE were. These results also standing NCSE in the context of the brain’s development and
contrast with the very minimal clinical morbidity attributable prior condition. Many of the syndromes included occur in
to NCSE directly. Lothman has summarized the many physio- neonates, infants, and young children; some continue into adoles-
logic changes that occur during SE, but most apply primarily to cence in individuals, or occur in adults.
GCSE; some may apply to NCSE (67). Boundary syndromes are mostly those with markedly abnor-
It is very difficult to prove that any neurologic damage mal baseline neurologic conditions and epileptiform abnormali-
accrues from NCSE. Pathologic studies of the effects of GCSE in ties on EEG—in which it is difficult or impossible to determine
humans are relatively few, in part because fatal SE cases are often what role the epileptiform discharges have in causation of the
associated with acute, severe brain-injuring illnesses that may clinical manifestations. Most are pediatric NCSE syndromes
cause damage independently. Episodes of NCSE are even less that occur within a broad range of illnesses such as the
often fatal unless they occur in association with GCSE or other Lennox–Gastaut syndrome (LGS), and include electrical status
acute, severe neurologic illness—in which case it is difficult to epilepticus in sleep (ESES), described briefly here but more in
sort out the cause of any neuronal damage. For the most part, Chapters 25 and 26. Boundary syndromes may also include
pathologic studies of patients with pure forms of NCSE remain epileptiform EEG patterns of uncertain significance such as
unavailable. “triphasic” waves or periodic discharges (lateralized or general-
The experimental models described in the chapter on GCSE ized), where there is substantial uncertainty in differentiating
included SE durations of well over 30 minutes (typically 1 to 2 whether this represents seizure activity or not (see next section)
hours of convulsions and electrical discharging activity) to (41,72). “Subtle” SE often includes prominent myoclonus, usually
produce neuronal damage in animals. In one rodent model of occurring in seriously ill patients in stupor or coma.
Chapter 29 ■ Nonconvulsive Status Epilepticus 599

Tabl e 2 9 . 1

Classification of Nonconvulsive Status Epilepticus Syndromes

1. Neonatal and infantile syndromes (including):


• West syndrome, Ohtahara syndrome
• Severe myoclonic epilepsy of infancy (SMEI; i.e., Dravet syndrome)
2. Childhood syndromes (including):
• Genetic-based illnesses such as ring chromosome 20 syndrome; Angelman syndrome, Rett syndrome
• Benign childhood epilepsies without a discrete genetic origin, for example, Panayiotopoulos syndrome
• Childhood epileptic encephalopathies such as Landau–Kleffner syndrome and electrical status epilepticus in sleep (ESES)
Febrile SE
3. Childhood and adult forms of NCSE:
(a) With epileptic encephalopathy:
(i) For example, within the Lennox–Gastaut syndrome, with atypical absence SE or tonic SE, and the continuation of
LGS into adulthood
(ii) Other epileptic encephalopathies
(b) Without epileptic encephalopathy:
• Typical absence SE of idiopathic generalized epilepsies
• Focal NCSE, sometimes referred to as “aura continua” with sensory, autonomic, or cognitive symptoms
• Complex partial SE: limbic or nonlimbic
• NCSE that follows the convulsions of generalized seizures or generalized convulsive SE
• “Subtle” SE with myoclonic SE following convulsions
4. Later adult NCSE: including de novo absence SE of late onset
5. “Boundary syndromes”:
• Some “atypical absence” SE of childhood
• Epileptic behavioral disturbances or psychosis
• Other forms of epileptic encephalopathy and coma due to brain injury (e.g., anoxia) with epileptiform discharges,
including “subtle” status.
• Drug or metabolic confusional states with epileptiform discharges (generally not considered true NCSE)

Modified from Shorvon (14).

EARLY LIFE, AGE-RELATED morphology, or spatial distribution, but many show little or no
NONCONVULSIVE STATUS such evolution (76). Morphologies include sharply contoured,
EPILEPTICUS SYNDROMES sinusoidal, or rounded waveforms in the alpha, theta, and delta
frequency range, but the frequency and morphology may vary
Many neonatal seizures and NCSE have minimal clinical mani- within a single seizure and from seizure to seizure (Fig. 29.1A–C).
festations and remain underdiagnosed if EEG monitoring is not Many neonatal seizures last 2 to 3 minutes but recur frequently;
utilized (73,74) (see also Chapter 25), especially in neonates with prolonged continuous seizures are less frequent (77).
severe brain injury (75) and in pharmacologically paralyzed Conversely, not all abnormal-appearing neonatal movements
infants. Electrographic patterns of neonatal seizures also differ are seizures (74). Proper use of EEG and EEG monitoring can
from those of older patients, often remaining localized to rela- help to avoid inappropriate AED treatment of movements such
tively small brain areas, likely due to incomplete myelination and as some swimming or pedaling motions, and stimulus-sensitive
neuronal migration (76). Multifocal seizures (more than three clonus or myoclonus, when they are not epileptic in origin. In
foci) are common, especially with multifocal or diffuse brain infants, particular attention must be paid to exclude artifacts
injury. Some seizures show evolution in frequency, amplitude, mimicking electrographic seizures.
600 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

A
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG
50 uV 2 sec

B
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG
50 uV 2 sec
Chapter 29 ■ Nonconvulsive Status Epilepticus 601

C
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

ECG
50 uV 2 sec

Figure 29.1 Neonatal status epilepticus in a 36-week conceptional age neonate with multifocal infarcts due to a placen-
tal abruption following a maternal motor vehicle accident. Multifocal seizures had various electrographic patterns. A:
Monomorphic sharply contoured delta activity. B: Continuation of the same seizure, showing evolution into rhythmic
spike-and slow-waves at 2.5 Hz, maximal in the right posterior quadrant. C: Later in same seizure, now with rhythmic
spike-and-wave over the left hemisphere and independent lower voltage ictal delta activity in the right hemisphere.

Several neonatal seizures and SE syndromes have had genetic syndrome, continuous spikes and waves during slow sleep
characterization. Dravet syndrome (severe myoclonic epilepsy of (CSWS), is an age-related pediatric epilepsy syndrome, usually
infancy) was originally described as being manifested clinically starting late in the first decade of life in children who were neu-
by prolonged obtundation, referred to as “obtundation status” rologically normal before seizures began (84). Seizures include
(78,79). Infantile epileptic encephalopathy, or Ohtahara syn- generalized convulsions and often evolve to become atypical
drome, typically presents with greater flexor, extensor, or tonic absence and atonic seizures with dangerous falls (81,85). EEG
spasms and prominent suppression bursts on the EEG (80). during wakefulness and REM sleep shows infrequent focal or
Many of the most prominent pediatric NCSE syndromes generalized epileptiform discharges. Non-REM sleep prompts
associated with underlying developmental delay or regression continuous generalized or bilaterally synchronous (if some-
demonstrate activation of epileptiform abnormalities during times asymmetric) spike-and-wave complexes at 1.5 to 3.5 Hz
sleep (81,82). ESES implies activation by sleep of persistent (Fig. 29.2). Continuous spike-wave patterns occupying at least
epileptiform activity suggestive of SE, although not all children 85% of slow-wave sleep are required for the diagnosis of CSWS
with these EEGs have clinical seizures. The waking EEG usually (Fig. 29.2) (85,86). Most children with CSWS have focal or gen-
shows infrequent focal or occasionally generalized epileptiform eralized seizures, global behavioral problems, and regression of
discharges, with the spike component usually more prominent cognitive function, more than in language alone (85,87). The
than the slow wave. During rapid eye movement (REM) sleep, epilepsy is often severe and can include many seizure types;
epileptiform discharges are similar to those on the waking EEG. some children have prolonged atypical absence SE (81).
Some children have an associated ESES syndrome, with CSWS may last for years, but the typical EEG pattern usually
ESES on the EEG along with progressive cognitive dysfunction, occurs between ages 5 and 15 years. The EEG eventually nor-
often referred to as an epileptic encephalopathy (83). One such malizes, including during sleep, and almost all seizures resolve,
602 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

Figure 29.2 Continuous spike waves during slow-wave sleep in a 9-year-old girl with mild developmental delay.

but cognitive and behavioral problems persist in most cases sleep-activated bursts of diffuse or multifocal spike-and-wave
(81,85). In LGS, the CSWS syndrome may include a prominent discharges at 1.5 to 3.5 Hz (ESES), most commonly in anterior
component of negative myoclonus or asterixis, at times explain- and middle temporal regions, but also in temporoparietooccip-
ing apparent atonic seizures and dangerous falls (27). These ital regions, some with a very broad, nearly generalized field
syndromes are covered more extensively in Chapter 26. (Fig. 29.3) (86,93). In contrast to CSWS, epileptiform discharges
The Landau–Kleffner syndrome (LKS) can be considered are present in less than 85% of slow-wave sleep, and may occur
another “boundary” syndrome and may overlap with CSWS and during REM sleep (94). The EEG manifestations usually appear
other “benign” focal epilepsy syndromes. LKS is characterized by between 3 and 5 years of age and resolve at about 15. The EEG
acquired aphasia, seizures, and a behavioral disorder presenting may improve over time, with or without treatment (90).
in children with previously normal language development, LKS and other ESES syndromes are often treated with AEDs,
between the ages of 2 and 4 years (88). A gradual deterioration, especially BDZs, with a goal of eliminating epileptiform dis-
specifically in language and speech output, may begin with charges and the idea (not entirely proved) that the epileptic
apparent word deafness or a form of auditory agnosia (89), but process contributes to the cognitive deterioration. Although
there can also be hyperkinetic behavior disorders. LKS exhibits episodes of ESES may recur frequently, the prognosis for seizure
clinical seizures in most cases, though seldom overt SE. It is control is good, but CSWS has a poor prognosis for cognitive
unclear whether the deterioration in language function is the function. LKS and CSWS are rare syndromes, each accounting
result of epileptiform activity or if the language deterioration for about 0.2% of all pediatric epilepsy patients (95).
and epileptic activity (both clinical and EEG) result from a com- In ring chromosome 20 syndrome, particularly in patients
mon underlying pathologic condition. Most LKS cases are of from Japan between the ages of 13 and 31 years, NCSE consists
unknown origin; a few cases are due to symptomatic lesions of prolonged confusional states, with or without motor phe-
(90). Corticosteroids are the most common treatment for LKS nomena, and prominent bilateral or generalized high-voltage
(91), but most children have persistent language deficits (89,92). slow waves with intermixed spikes on the EEG (96). The
The waking EEG in LKS may be normal or show unilateral epilepsy is characterized by intermittent motor seizures with
or bilateral temporal spike-and-wave discharges. There are confusion, staring, head turning, mutism and meaningless
Chapter 29 ■ Nonconvulsive Status Epilepticus 603

Fp1-F3
F3-C3

C3-P3
P3-O1
Fp2-F4
F4-C4

C4-P4
P4-O2
Fp1-F7
F7-T3

T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6

T6-O2
Fz-Cz
Cz-Pz
LUC
RLC
EKG
75 uV 1 sec

Figure 29.3 A 4-year-old boy with language regression, Landau–Kleffner syndrome. EEG during sleep shows bilateral
1-Hz centrotemporal sharp waves, more prominent over the right hemisphere.

utterances, facial flushing, and limb shaking. Impulsive behav- psychic, or autonomic symptoms (97). Focal SE with motor
ior, inappropriate responses, and myoclonus occur in episodes components is described in Chapter 28. Nonmotor (noncon-
up to several times daily. vulsive) forms of focal SE include persistent sensory distur-
bances (e.g., visual hallucinations) and autonomic and
cognitive deficits, including affective, amnestic, and language
disturbances such as aphasia (42).
Although NCSE in children is perhaps best considered in the Overall, focal onset of symptoms and electrographic abnor-
context of age and brain development, the descriptions of NCSE malities is more common than SE with a generalized onset
in adults in this chapter follow more familiar International (98). Focal NCSE is seen frequently after strokes or other acute
League Against Epilepsy (ILAE) seizure types and syndromes, brain injuries, and should be suspected when patients do not
focusing on the type of seizure apparently persisting and caus- stabilize or improve as expected. In some critically ill patients,
ing the patient’s clinical deficit. They are organized into focal NCSE consists of brief cyclical electrographic seizures occur-
and generalized forms, acknowledging that seizures can progress ring every 5 to 10 minutes over several hours (99). Focal lesions
from focal onset to generalized clinical and EEG manifestations. from vascular causes, trauma, developmental abnormalities
Myoclonic, tonic, and clonic SE are detailed further in the con- (such as heterotopias and other migration abnormalities),
vulsive SE chapter because of their motor manifestations. and mitochondrial disorders are among many possible causes
(100,101).
FOCAL NONCONVULSIVE STATUS
Focal Sensory Nonconvulsive Status Epilepticus
EPILEPTICUS
Persistent sensory seizures (without motor accompaniment) have
Focal status epilepticus may be either SPSE with no impairment been called “aura continua” (102,103). Infrequently, they may last
of consciousness, or CPSE with consciousness impaired. SPSE for hours. Simple partial sensory status epilepticus is identical to
presents with ictal symptoms reflecting the involvement of dis- the prolonged continuation of an isolated aura continua. In these
crete regions of brain, such as motor, sensory, special sensory, cases, local inhibitory function is sufficient to prevent spread of
604 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3
F3-C3
C3-P3

P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2

Fp1-F7
F7-T3
T3-T5
T5-O1

Fp2-F8
F8-T4
T4-T6

T6-O2
Fz-Cz
Cz-Pz

LUC
RLC
EKG 75 uV
1 sec

Figure 29.4 An 8-year-old boy with simple partial status epilepticus arising in the right occipital region. Seizures con-
sisted of seeing pulsating green and red circles in the left visual field.

the seizure more broadly in the brain. If, however, these seizures of color and light (110) (Figs. 29.4 and 29.5). There may be sco-
spread to involve areas of the brain necessary for responsiveness tomata or simple visual hallucinations predominating in the cen-
(for a long period), this becomes CPSE. Occasionally, benign tral visual field, but complex hallucinations are also possible
rolandic epilepsy, in which there is speech arrest, drooling, dys- (111). Slightly more anteriorly, involvement of the temporopari-
phagia, facial weakness, head deviation, and mild confusion, may etooccipital junction may produce nystagmus with contraversive
be prolonged enough to constitute NCSE (104,105). eye deviation (20).
Pure sensory SE is generally considered rare, but this may The “positive” sensory symptom of pain during SPSE appears
reflect a detection bias as an EEG may show little at the time of even more rare, although shorter painful seizures have been
SE (97), and persistent sensory disturbances are often attrib- reported (103,112). Some are parietal in origin, in the somatosen-
uted to transient ischemia (106) or other nonepileptic causes. sory area. Parietal NCSE can also be manifested solely by persist-
In one report, four patients had focal sensory NCSE lasting up ent paresthesias (51). Other unpleasant epileptic sensory seizures
to several days, without motor or behavioral symptoms or alter- may include nausea, ictal fear, anorexia, poorly described visceral
ation in responsiveness (107). Some were frontal or temporal in sensations, and the distress of “abdominal epilepsy” originating
origin. They appeared to be less common than auras progress- in mesial temporal areas, especially involving the amygdala
ing to more obvious seizures, and far less common than CPSE. (112–114). Autonomic symptoms are reported increasingly fre-
Focal lesions in temporal, parietal, and occipital cortices may quently, particularly in children (115).
produce SE with persistent cognitive or sensory symptoms, with Some focal SE involve a larger brain area than a typical simple
hallucinations of any sensation, including visual, auditory, olfac- focus, and could be called “regional.” The most common (usually
tory, gustatory, somatosensory, or vertiginous perception (103). pediatric) form of regional SE is the Panayiotopoulos syndrome,
Olfactory hallucinations from temporal epilepsy may be the in which an exact focus is often difficult to determine (116). This
most common. Some SPSE consist of auditory sensations alone, syndrome is an age-dependent epileptic susceptibility that is usu-
with epileptiform discharges confined to Heschl’s gyrus (108). ally idiopathic and benign, although perhaps 15% are due to
Transient cortical blindness or visual field loss (sometimes focal lesions. It affects about 13% of all children aged 3 to 6 with
referred to as status epilepticus amauroticus) can be a manifesta- nonfebrile seizures. Manifestations are primarily autonomic,
tion of occipital SPSE (109), although simple visual hallucina- with ictal emesis and occasional syncope, usually arising from
tions and ictal blindness are usually brief (110). Occipital seizures sleep. Half of the episodes last longer than 30 minutes and thus
can also include macropsia, micropsia, misperception of spatial constitute autonomic SE. Most children have few episodes and
orientation, hallucinations of faces or animals, or simple patterns typically do better without treatment, and the long-term risk of
Chapter 29 ■ Nonconvulsive Status Epilepticus 605

A
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

B
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

Figure 29.5 Focal status epilepticus in a 14-year-old girl with right occipitoparietal cortical dysplasia. A: Seizure begins
with rhythmic beta activity over both occipital regions, more prominent on the right. B: Seizure evolves into high-voltage
3- to 4-Hz spikes in the left occipital region, and spikes with slower frequencies in the right occipital region. Seizures
recurred every 2 to 3 minutes for several hours before presentation to the ER.
606 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

A
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
50uV 1 sec

B
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
50 uV 1 sec

Figure 29.6 A: A 49-year-old woman with mild developmental delay who presented with an anterior aphasia. She was
able to follow commands. EEG during aphasia shows repetitive sharp waves in the left temporal region at 2 Hz. B: Same
patient after 2 mg IV lorazepam. EEG shows resolution of ictal activity and a left midtemporal spike. The aphasia resolved.
Chapter 29 ■ Nonconvulsive Status Epilepticus 607

epilepsy is small (116). Earlier described in correlation with diagnose and can be confused with many other illnesses (see
occipital spikes, this syndrome may have discharges elsewhere or Differential Diagnosis above). The EEG is extremely useful in
multifocally and is probably better considered a regional (but not establishing a diagnosis, but not all patients have diagnostic
quite generalized) form of seizures and SE. findings. In one report of six patients with prolonged neu-
rocognitive deficits and evidence of focal-onset NCSE, all had
Amnestic and Aphasic Status Epilepticus rapid rhythmic epileptiform discharges on EEG and eventual
Persistent memory dysfunction often follows complex partial resolution of all symptoms with treatment (122).
seizures in the postictal period, and it may follow, and be due to,
an episode of status epilepticus (117,118). Episodes of pure EEG
amnesia (with preserved cognitive function in other realms) can Surface EEGs, even when recorded during the symptoms, have
also be due to individual epileptic seizures, referred to as “tran- limited sensitivity for detection of ictal discharges in SPSE, at
sient epileptic amnesia (TEA)” (119). TEA almost always occurs least in part because the seizures may involve a small volume of
in patients with earlier epilepsy and is often associated with brain tissue. The EEG may be normal or show nonspecific
additional persistent memory deficits. Seizures preferentially changes (129). One report described 87 well-characterized sim-
affecting memory may require involvement of medial temporal ple partial seizures in 14 patients, and just 20% of seizures had
structures bilaterally, as shown by depth electrode recording identifiable epileptiform abnormalities on scalp EEG (97). Two
(120). TEA typically occurs in older patients, thus overlapping thirds of seizures had primarily sensory symptoms, and these
clinically, and often difficult to distinguish from (nonepileptic) showed ictal EEG changes in just 15%. In another study, only
transient global amnesia. Many episodes of TEA are prolonged 20% to 35% of simple partial seizures had ictal correlates on
beyond 30 minutes, constituting NCSE with amnesia as the sole surface EEG (130). Sometimes, SPSE can remain undiagnosed
clinical manifestation (119). Other amnestic syndromes can be until invasive EEG monitoring is performed—usually carried
caused by frontal or generalized NCSE (121); the frontal dis- out because of (other) refractory seizures (131,132).
charges suggest an inability to access extant memory during the When SPSE is visible on EEG, it usually shows ictal onset
seizures. Various “psychic” phenomena, including apraxia and with a single topographic EEG maximum, followed by evolution
acalculia, as well as prolonged mood changes, such as depression in frequency, voltage, and distribution. Focal fast-frequency dis-
and panic attacks, can also occur in NCSE (122). charges, rhythmic waveforms with evolving morphology, repeti-
Aphasic SE, with preserved responsiveness and a clinical tive epileptiform discharges, or regional or lateralized alterations
deficit restricted to language dysfunction alone, is a focal or in EEG background activities may be seen (129,130). If SE is dis-
regional NCSE (while CPSE implies a disturbance in respon- continuous, the background activity between seizures is typically
siveness) (123–127). Careful language testing is necessary to slow and disorganized, with occasional focal interictal epilepti-
determine that the deficit is truly an aphasia (125,128). form discharges.
Speech arrest alone can result from seizures in many areas. A Electrographic focal SE is seen in some stuporous or coma-
focal neurologic deficit in speech production may be mistaken tose patients with no clear clinical signs of seizure activity
for other syndromes such as transient ischemia or psychiatric (133). Seizures may be either continuous or repetitive, and EEG
illness. patterns are similar to those in SPSE and CPSE. Such partial SE
A wide variety of aphasic NCSE types has been characterized is not rare after strokes or other acute brain injuries and should
by appropriate and thorough language testing—with preserved be suspected when patients in those settings do not stabilize or
responsiveness and other cognition except for the aphasia improve as expected. EEG should be performed for any ICU
(128). Often, the ongoing epileptiform discharges on EEG cor- patient in whom mental status changes are unexplained or
respond anatomically to the area associated with a particular appear out of proportion to the degree of acute neurologic
aphasia (Fig. 29.6), such as posterotemporal focal discharges injury. Patients with suspected SPSE also warrant an MRI scan
in patients with clinically evident Wernicke’s-type aphasia to look for an underlying focal lesion.
(124,126). Those with more anterior aphasias (some with fron-
totemporal discharges on EEG) are often unable to speak but Complex Partial Status Epilepticus
may retain verbal memory and respond appropriately to verbal The first definite case of CPSE was reported by Gastaut and Roger
commands, giving evidence of relatively preserved comprehen- in 1956 (13). In the 1970s, CPSE was considered rare and was the
sion (122,123). Some have alexia and reduction in spontaneous subject of isolated case reports (117,118), and by 1985 only 17
speech. Still, the correlation to classical (usually stroke-defined) clearly identified cases had been published (134). With the realiza-
areas of clinical involvement for different types of aphasia is far tion that absence SE is a separate category rather than simply any
from precise (128). NCSE with generalized discharges, CPSE was eventually recog-
nized as more common (17,18,135), although it remains under-
Diagnosis of Focal Nonconvulsive diagnosed (136). Other terms for CPSE include temporal lobe SE
Status Epilepticus (often not an accurate localization), prolonged epileptic fugue
Focal NCSE is usually more subtle in presentation than either state, and psychomotor SE (134,137–140).
focal motor SE or generalized forms of SE. SPSE manifested by The definition of SE as a seizure or series of seizures without
sensory or cognitive symptoms alone can be very difficult to recovery for 30 minutes (141) allows for the possibility that
608 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

Figure 29.7 “Psychic” status epilepticus in a 41-year-old woman with right mesial temporal sclerosis. She presented with
mild confusion but could follow most commands. She did not answer questions and had bizarre speech, repeating “you
are my Norm, you are my one true love” continuously.

CPSE can consist of a single very long complex partial seizure CPSE may present as abnormal behavior or diminished
or a series of recurrent complex partial seizures without clinical responsiveness associated with lateralized epileptiform seizure
normalization between seizures (Fig. 29.10) (118). CPSE is activity. It may begin with a simple partial seizure, or conscious-
probably the most common form of NCSE in adults who are ness may be impaired at the onset. Impairment of consciousness,
ambulatory or not critically ill. which may cycle or fluctuate, ranges from almost nondiscernible
CPSE begins with a focal-onset seizure, progressing to clouding of higher cortical function to coma (44,99,118,145).
involve more of the brain such that responsiveness is impaired, CPSE may include an “epileptic twilight state” with a lack of
and then with prolongation or recurrence. CPSE may last responsiveness or confusion, and bizarre, and particularly fluctu-
hours, days, or even months (17,142). The most common ating, behavior or paranoia (17,117,118,134,140) (Fig. 29.7). The
causes are new acute vascular disease, or old strokes with a extensive variety of clinical presentations of CPSE makes EEG
more recent precipitant such as infection or metabolic derange- essential to distinguish CPSE from other causes of altered mental
ment (44,143). Other infections (e.g., encephalitis), tumors, status.
congenital developmental abnormalities, and vascular malfor- Lip-smacking, other oroalimentary automatisms, lateralized
mations are possible. The diagnosis of CPSE should be consid- limb automatisms and dystonic posturing, eye deviation, and
ered in patients who are partially or completely unresponsive, nystagmus are typical of CPSE; myoclonus is rare (44,140,146).
especially those with earlier epilepsy or vascular disease. Fear, aggressivity, irritability and anxiety, and stereotyped, com-
While most patients have earlier epilepsy (144), CPSE may plex automatisms are more frequent with CPSE than with
arise in patients without prior seizures. CPSE is seldom absence SE (146), but when the automatisms or movements are
reported in children, but it may be significantly underrecog- minimal, CPSE can be difficult to distinguish from absence SE.
nized and occur in those with substantial developmental delay
and neurologic deficits, and even in those with other seizure EEG
types (145). From the incidence of SE in different populations In contrast to SPSE, most CPSE shows discernible surface EEG
with epilepsy, it has been estimated that CPSE occurs in 35 cases changes. The EEG patterns of CPSE are variable, reflecting
per million population per year, but possibly five times as often differences in the location of ictal onset zones and propaga-
in developmentally delayed patients (144). tion pathways, but they are often very similar to those seen in
Chapter 29 ■ Nonconvulsive Status Epilepticus 609

Figure 29.8 Quantitative EEG showing complex partial status epilepticus with recurrent seizures originating independ-
ently from both hemispheres, in a 45-year-old woman with multiple sclerosis and malfunction of an intrathecal baclofen
pump. The bottom two lines of the compressed spectral array (CSA) show peaks (see arrows) of rhythmic activity of alpha
and theta frequencies indicative of seizures. Expanded “usual” EEG below shows, (A) right hemisphere seizures, corre-
sponding to the thick arrows on the CSA, and (B) left hemisphere seizures corresponding the thin arrows on the CSA above.
(See color insert)

isolated complex partial seizures. CPSE may be characterized involve a greater area of the cortex (Fig. 29.9) and result in dif-
by either repetitive complex partial seizures, each showing fuse or generalized epileptiform discharges difficult to distin-
focal onset, with background slowing between seizures, or by guish from those of absence SE (18,44,45). As CPSE progresses,
continuous ictal rhythms, corresponding to cycling or contin- the epileptiform discharges often increase in voltage, slow in
uous clinical manifestations (70,99,134,147,148). Rarely, frequency, and broaden in distribution. Later, seizures may
ictal patterns arise independently from both hemispheres become fragmentary, interrupted by brief periods of back-
(Fig. 29.8). Waveform morphologies include repetitive epilep- ground attenuation (152). Between discrete seizures, the back-
tiform spikes; spike-and-slow wave discharges; rhythmic theta, ground shows focal slowing, focal epileptiform discharges, or
delta, or alpha frequencies; or rhythmic low-voltage fast activ- periodic lateralized epileptiform discharges (PLEDs) (152).
ity (45). Occasionally, the EEG can be normal or obscured by
artifact (149). Temporal Versus Frontal Complex
Often, early EEG abnormalities are focal or lateralized Partial Status Epilepticus
throughout the episode (130). Focal onset is usually followed by CPSE may arise from many brain regions. Many episodes rep-
evolution in distribution, voltage, and frequency, in commonly resent prolonged temporal lobe seizures, as demonstrated
recognized patterns of electrographic seizures (110,150,151). clearly by depth electrode recording (153), but in another study
Epileptiform discharges may merge gradually to produce con- of eight patients with depth electrode-recorded episodes of
tinuous focal seizure activity. Many have subsequent spread to CPSE, all had frontal origins for the seizures (145). At least on
610 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz 400 uV
Cz-Pz 2 sec

Figure 29.9 Left temporal seizure evolution in a 21-year-old woman with intractable epilepsy admitted with confusion
and aphasia. She had complex partial status epilepticus consisting of frequent discrete electrographic and clinical seizures.
EEG in three horizontal sections, of 18 channels each. Top section: one of frequent discrete electrographic seizures arises
in the left frontotemporal region, evolving with higher voltage, slightly lower frequency sharp waves in the left temporal
region; with spread to the right side in the middle section; eventual slowing of discharges and interruption by brief nondis-
charging periods in the bottom section. Just after this, there was generalized, 1.5 Hz slowing for 5 seconds and then a very
suppressed background in all areas as the seizure ended.

the scalp EEG, parietal CPSE is usually nonlocalizing or falsely Temporal Complex Partial Status Epilepticus
localizing (154). Overall, extratemporal onset of CPSE may be The clinical manifestations of temporolimbic CPSE are exten-
more common, including in children (115). sive. “Experiential” sensations involving memories of percep-
Extratemporal foci may produce visual, auditory, somes- tions or experiences, often with an emotional overtone, indicate
thetic, or vestibular hallucinations; a perception of warmth; limbic structure involvement of seizures (155) (Table 29.2). For
changes in facial color; nausea; or unilateral arm automatisms the seizure to be “experienced,” or remembered, there should
or posturing. When seizures spread to involve the amygdala and also be involvement of both medial temporal and neocortical
hippocampus, there may be chewing movements, lip-smacking, structures (102). Seizures involving limbic areas of the hip-
and gesticulatory automatisms (118). pocampus and amygdala interfere with responsiveness, becom-
Frontal CPSE generally has less impairment of conscious- ing “dyscognitive” seizures and CPSE (102). In temporal CPSE,
ness and fewer fluctuations than does temporal lobe CPSE dyscognitive focal SE may overlap with aura continua or evolve
(146). Frontal CPSE may recur frequently, but it is misdiag- from it (153).
nosed often. Temporal CPSE includes more fear, anxiety, anger, Temporal CPSE may have a sudden or gradual onset (102).
irritability, aggressiveness, agitation, and simple and complex Hippocampal, amygdalar, and amygdalohippocampal CPSE
automatisms (146). Psychomotor slowing is more frequent in often present with cyclical changes in behavior (99,102,118). An
frontal than in temporal lobe CPSE, but about the same as in early report described a “twilight state” with partial responsive-
absence SE, and CPSE with a frontal origin may be very difficult ness and intermittent speech arrest (118). Some patients were
to distinguish from absence SE. completely unresponsive, with motionless staring. Others had
Chapter 29 ■ Nonconvulsive Status Epilepticus 611

Tabl e 2 9 . 2 CPSE originating in the temporal lobes may have different


manifestations from seizures starting elsewhere and spreading
Symptoms of Temporal Lobe Complex Partial to temporal structures. Opercular, frontal, and occipital origin
Status Epilepticus seizures can all spread secondarily to involve mesial temporal
regions. Some seizures with temporal lobe-related manifesta-
“Experiential” seizures combining perceptual and memory tions actually originate in frontal lobes, with the initial behav-
abnormalities, and mood or affect disturbances, including ioral features representing frontal dysfunction.
depression and anxiety.
EEG
Memory abnormalities, including déjà vu, jamais vu, and so
CPSE arising from mesial temporal regions usually begins with
on or gaps in memory.
unilateral temporal 5 to 7-Hz rhythmic activity, which appears
“Psychic” auras, including forced thinking, and behavior within 30 seconds of the initial clinical symptoms or signs (150)
mimicking psychiatric disorders. (Figs. 29.9 and 29.10). Interictally, there is focal slowing or
Emotional symptoms, including fear (if involving the attenuation on the side of seizure onset. In neocortical tempo-
amygdala), sadness, and distress. ral seizures, discharges are more widely distributed, sometimes
over the entire hemisphere. As compared to the EEG findings in
Alterations in perception of reality, including
medial temporal CPSE, the seizures of neocortical temporal
depersonalization or out of body experience.
CPSE often begin with slower frequencies, show less stable fre-
quency and voltage, and appear later after clinical signs (151).
Adapted from articles by Wieser (102,153) and Gloor (155).
Frontal Complex Partial Status Epilepticus
oroalimentary or more complex automatisms, vocalizations, Thomas and colleagues described two types of frontal lobe (FL)
and perseverative gesticulations. A more anterior seizure origin NCSE (139). Type 1 had continuous mood and behavioral dis-
or spread may induce bizarre limb automatisms or wandering, turbances, with mild hypomania, affective disinhibition (or
as in a fugue state (poriomania) (118). Behavior may fluctuate conversely, indifference), and subtle impairment of cognition,
and become very strange. Memory for the event is often oblit- but no overt confusion. Patients could exhibit a blank facial
erated or impaired. expression, lack of spontaneous emotion, and decreased verbal

FP1-F3
F3-C3
C3-P3

P3-O1
FP2-F4

F4-C4
C4-P4
P4-O2
FP1-F7
F7-T3
T3-T5
T5-O1
FP2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz

Cz-Pz
LUC
RLC
EKG 75 uV 1 sec

Figure 29.10 Right temporal complex partial seizure in a 32-year-old man with mesial temporal sclerosis, presenting with
status epilepticus consisting of repetitive seizures. The early EEG before this seizure shows widespread postictal slowing
following the previous seizure.
612 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 29.11 Frontal lobe complex partial status epilepticus in a 56-year-old woman with a history of traumatic brain
injury. EEG shows irregular 5- to –8-Hz spikes and polyspikes over the left frontal region.

fluency. Some could recall events that occurred during the spell discharges on EEG, indistinguishable from those of absence SE,
and could carry out routine activities, such as eating. There and often with a very similar clinical appearance (138,156).
were simple gestural automatisms such as scratching, rubbing,
or picking at clothes, but complex bipedal or bimanual automa- Complex Partial Status Epilepticus:
tisms were not observed. Type 1 FL-CPSE led to no morbidity Treatment and Outcome
in later cognitive function. Usually by the time of diagnosis of CPSE, seizures have been
The rarer type 2 FL-CPSE had greater impairment of con- present for at least 30 minutes, so treatment should begin
sciousness, often with cyclic fluctuations. There was marked promptly with intravenous AEDs. Possibly because of the asso-
behavioral disturbance, temporospatial disorientation, confu- ciation with vascular disease and other lesions, and prior focal
sion, prominent perseveration, and occasional catatonic stupor. epilepsy, CPSE is often harder to treat than primary generalized
Some patients required restraints because of aggressiveness. SE and is more likely to recur (17). Fortunately, patients with
There was occasional head and eye deviation, and perioral CPSE in the setting of earlier epilepsy are very likely to survive
myoclonia. Patients were universally amnestic for the episode. and generally warrant a less aggressive approach than does
GCSE (157). CPSE is usually treated in the same way as focal-
EEG onset GCSE, but somewhat less aggressively if there is an earlier
While ictal EEG is nonlocalizing or normal in many patients history of epilepsy, and CPSE is usually more easily treated than
with frontal lobe complex partial seizures (149), scalp EEGs is GCSE (158,159).
eventually show bilateral sharp-and-slow-wave activity in most The outcome from episodes of CPSE depends primarily on
(145) (Fig. 29.11). Some patients have focal rhythmic alpha or the etiology, and morbidity often derives from the unresponsive
beta activity at the onset of seizures originating near scalp elec- state and from complications of the underlying illness causing
trodes (149), but the scalp EEG often shows no changes until the the SE (particularly encephalitis and strokes), rather than from
seizure spreads to medial temporal structures, producing ictal the CPSE itself. In one study, there was no difference in mortal-
patterns typical for medial temporal seizures. In type 1 FL-CPSE ity between patients with CPSE (with focal discharges on EEG)
(above), the EEG showed a unilateral frontal focus; in type 2, there and those with GCSE, but the CPSE included patients with sec-
were bilateral asymmetric frontal foci with an abnormal back- ondary generalization (160). With CPSE, mostly of frontal ori-
ground (139). Spread of epileptiform activity from a unilateral gin, episodes of longer duration had no worse an outcome than
frontal focus may lead to bilaterally synchronous or generalized those with shorter durations (145).
Chapter 29 ■ Nonconvulsive Status Epilepticus 613

Early reports on CPSE included very few patients. Almost all Precipitants include sleep deprivation, alcohol ingestion,
returned to normal or “baseline cognitive function” (134,139, metabolic abnormalities, and intercurrent illnesses. Many med-
161), with resolution of all neurologic deficits over time, but ications have been implicated in the precipitation of absence
this may take weeks or months (117). In one CPSE series, none SE, especially neuroleptics, tricyclic antidepressants, lithium,
of the 20 patients had lasting cognitive deterioration, and 5 had and BDZ withdrawal. AEDs such as phenytoin and carba-
meticulous neuropsychologic assessment (17). There are mazepine may contribute (171). A generalized convulsion may
reports of prolonged memory and other cognitive deficits, for set off an episode of absence SE or end it. In the evaluation of
weeks or months after NCSE (51,117,118,122,143), but many patients with absence SE, MRI scans are almost invariably nor-
resolve eventually. In some cases, both clinical and imaging mal; any focal abnormalities are likely incidental.
abnormalities can resolve completely over a period as long as a Typical absence SE starts abruptly, without warning, and
year (17,139). consists of individual seizures in rapid succession without
Long-term clinical effects of CPSE could also include wors- recovery over a long period or, more commonly, a single pro-
ened cognitive function or increased subsequent seizure fre- longed episode with continuous epileptiform discharges and
quency. Most outcome studies, however, are of GCSE, and it impaired consciousness (172). The clinical presentation can
correlates with worsened seizure control, but this might be be very subtle, with cognitive deficits discernible only on for-
attributed to the underlying illness rather than to the episode of mal neuropsychologic testing. There may be just a relatively
SE itself (162). hard-to-diagnose change in responsiveness, with preserved
alertness (165). Cognition may fail when more complex
GENERALIZED STATUS EPILEPTICUS planning is needed. Patients may remain reactive to external
stimuli, and some can walk or carry on relatively complex
Absence Status Epilepticus activities without interruption, but usually with slowness and
Although “petit mal intellectual” was postulated in the 19th cen- diminished insight and complexity of activities (164,172).
tury, Lennox demonstrated in 1945 that altered mental status There is often a fluctuating diminution of responsiveness or
could be caused by absence seizures. He suspected that a series overt confusion. Language use may be relatively preserved, but
of individual absence seizures and EEG discharges could persist with slowness, paucity of speech, and monosyllabic answers
for much longer than was generally recognized (11). Absence (156), and often an inability to discuss more complex con-
SE was first described as “status epilepticus in petit mal” by cepts. Language may be reduced to simple phrases or less, and
Schwab in 1953 (163), recognizing the same EEG patterns seen patients may follow only the simplest of instructions, or none.
in petit mal epilepsy by Lennox earlier. The prolonged confu- Infrequently, absence SE progresses to complete unrespon-
sion and slowed behavior were labeled “spike-wave stupor” by siveness (164).
Niedermeyer and Khalifeh (164). Typical absence SE has also Motor abnormalities are minimal. There may be brief spo-
been called petit mal status, minor epileptic status, absence con- radic myoclonic eyelid jerks and eye blinking, especially in the
tinue, epilepsia minoris continua, and status pyknoepilepticus. IGE syndromes (146). Limb myoclonus is common. Gestural and
Early reports and classification of NCSE (i.e., in the 1970s ambulatory automatisms occur (166,173), but automatisms are
through the 1990s [!]) generally divided it into two types: usually less complex than in CPSE. Gait tends to be preserved if
“absence SE” (all NCSE with generalized epileptiform discharges the patient can stand. Sometimes there are bradykinesia, delayed
on the EEG) and “complex partial SE” (all others felt to have a reactions, and withdrawal (166). Patients may appear to have
focal seizure onset). CPSE could have focal EEG discharges or a
clear focal onset clinically (44). The oversimplification of the
generalized forms is no longer tenable. Tabl e 2 9 . 3
Absence SE is a pure form of primary generalized epilepsy
with clinical episodes lasting over 30 minutes. In its classic Status Epilepticus in the Idiopathic Generalized
appearance, it occurs in patients with earlier absence epilepsy or Epilepsies
other idiopathic generalized epilepsies (IGEs) (165–167).
Absence epilepsy (see Chapter 26) begins anywhere from age 3 Generalized convulsive tonic–clonic SE, especially upon
years until early adulthood, but usually in childhood or adoles- awakening
cence; seizures usually decrease markedly in the third decade. The Myoclonic SE
incidence of absence SE is about one case per million population
per year (144), and about 10% of adults who still have absence Nonconvulsive forms:
epilepsy will have at least one episode of absence SE (168). • Typical absence SE
Absence SE occurs in nearly all IGE syndromes, including juve- • Atypical absence SE
nile absence epilepsy (Fig. 29.12), JME, perioral myoclonia with
absences, eyelid myoclonus and absence, and IGEs with phantom • “De novo” absence SE
absences (169)—even when those syndromes are usually mani- • Autonomic SE (perhaps better characterized as regional)
fested by other types of seizures, for example, myoclonus in JME
(167) (Table 29.3). NCSE is uncommon in JME (170). Adapted from Shorvon and Walker (167).
614 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3

F3-C3
C3-P3
P3-O1

Fp2-F4
F4-C4
C4-P4

P4-O2

Fp1-F7
F7-T3
T3-T5

T5-O1
Fp2-F8
F8-T4

T4-T6
T6-O2
Fz-Cz

Cz-Pz
LUC
RLC

EKG
100 uV1 sec

Figure 29.12 Absence status epilepticus with generalized, frontally predominant, 3-Hz spike-and slow-wave discharges.

diminished initiative or motivation. Perseveration in motor depression, or hysterical behavior (166,172); or from posttrau-
activity or on performance tests is typical. matic amnesia or metabolic encephalopathies (24). Medication
Patients’ experiential descriptions are vivid. There may be side effects can appear very similar to absence SE clinically.
visual hallucinations, a dreamy state, and inappropriate man- Myoclonus is not rare in absence SE, but it is rare in CPSE (146).
ner. Some patients are aware, themselves, of slowing or diffi-
culty with thought and speech, sometimes noticeable only to EEG
those closest to them. Occasionally, patients show hostility and EEG is often required for the accurate diagnosis of absence SE.
aggression, agitation, and impulsive behavior (13,172). The characteristic EEG of absence SE is very similar to those of
Behavior mistaken for psychosis is more likely with CPSE, and absence epilepsy: frequently recurrent or prolonged uninter-
confabulation was seen in CPSE only (146). Absence SE often rupted seizures for over 30 minutes, with extremely rhythmic,
induces relatively little amnesia or postictal confusion (172). bilaterally symmetric runs of spike-and-slow-wave discharges;
Absence SE may last anywhere from 30 minutes (by defini- polyspikes may be admixed (Figs. 29.12 and 29.13) (174,175).
tion) to months (24). It may cease abruptly—spontaneously Other EEG patterns include generalized rhythmic slowing with
or upon treatment with BDZs, with rapid return to normal intermixed spike-and-slow-wave complexes, irregular sharp-
mentation. Some episodes end with a generalized convulsion. and-slow-wave discharges, or diffuse background slowing with
Most reported cases have been isolated, but some recur fre- superimposed bursts of fast activity (172). The epileptiform
quently (172). discharges are bilaterally synchronous and usually of maximal
Recently described is a syndrome in which episodes of voltage over frontal and central regions. Characteristic-appearing
absence SE are the primary manifestation of epilepsy, with few generalized epileptiform discharges may have some asymmetry,
other seizures (169). All patients (adults) had normal neuro- but this should not be overinterpreted (138).
logic examinations and imaging and no family history of Discharge frequencies may reach up to 4 Hz at the onset of
epilepsy. Episodes of absence SE lasted from 30 minutes up to a the episode but typically slow quickly to the usual 3-Hz pattern
few weeks, and many recurred several times a year. within seconds. Discharges are characteristically regular, at 3 to
Clinically, absence SE may appear remarkably similar to CPSE 3.5 Hz. If seizures are prolonged, the discharges may slow to
of frontal or temporal origin; and all of these can be difficult to less than 3 Hz and may become irregular (176). More rapid dis-
differentiate from a postictal confusion; from nonepileptic causes, charges and polyspikes suggest other IGE syndromes. Discharges
including psychiatric conditions such as dissociative states, are occasionally interrupted by brief bursts of normal EEG
Chapter 29 ■ Nonconvulsive Status Epilepticus 615

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
200 uV
2 sec

Figure 29.13 Absence SE in a 17-year-old boy with juvenile absence epilepsy. Brief absence seizures (during an episode
of SE) demonstrate generalized frontally maximal spike-wave activity at 3 Hz. Frequent 5- to –10-second absence seizures
recur every 5 to 10 seconds over 20 minutes until administration of IV lorazepam. Clinical manifestations were staring,
unresponsiveness, eyelid fluttering, and subtle limb automatisms.

background activity. Interictal EEGs may show the same dis- NCSE in IGE syndromes. Nevertheless, this conclusion rests on
charges in brief bursts, almost always on a normal background. clinical impressions in moderate-sized series rather than on
confirmation by neuropsychologic testing. Still, the prognosis
Treatment and Outcome of absence SE is superb.
Typical absence SE that occurs within absence epilepsy or other
IGE syndromes is almost always readily interrupted (clinically De novo Absence Status Epilepticus
and on EEG) by relatively low doses of BDZs or other AEDs, Typical absence SE is unlikely in older patients without an ear-
preferably intravenously, but sometimes effective even orally lier history of an IGE, but occasional episodes have been
(Fig. 29.14). Interruption by intravenous BDZs is so often effec- reported in patients older than 50 years with no prior epilepsy
tive that it is a useful diagnostic test. Lorazepam doses under 0.1 (de novo absence SE of late onset) (29). Most have moderate
mg/kg will often suffice (45). Intravenous valproic acid at 20 impairment of consciousness. The most common setting is that
mg/kg is a reasonable alternative (177) and is also effective in withdrawal of BDZs used for sleep or psychiatric reasons. Other
patients with recurrent absence SE, particularly in the IGE syn- triggers include toxins or metabolic dysfunction and the use of
dromes (178). Lamotrigine is also helpful. Typically, the EEG neuroleptic and other psychotropic medications (179). Some
will resume a normal background quickly, and the patient may “de novo” patients may have had primary generalized epilepsies
emerge from the confusion within minutes. Phenytoin and car- earlier in life without diagnosis, and others may have been mis-
bamazepine are not ideal treatments as they may exacerbate or diagnosed. It is unclear that all of these cases are truly “de novo.”
even precipitate absence SE (171). Fortunately, episodes of de novo SE are relatively uncommon
For typical absence SE, the outcome and consequences and usually easy to interrupt quickly (167). Especially when
depend on the etiology—in this case, the epilepsy syndrome, precipitated by some identifiable cause such as BDZ with-
which is benign. Absence SE has produced no mortality and drawal, most patients do not need maintenance AEDs.
minimal morbidity in reported series, and no significant long- The EEG in de novo absence SE tends to be characteristic of
term morbidity can be attributed to it (29,172) or to any other absence SE, showing generalized spike-and-wave activity at
616 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

A
FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

B
FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
75 uV 1 sec

Figure 29.14 A: A 75-year-old woman with a history of generalized tonic–clonic seizures (probably a long-standing IGE),
now with absence status epilepticus causing confusion. EEG shows semirhythmic high-voltage 3-Hz sharp-and-slow-wave
discharges, alternating with periods of diffuse delta activity and slower epileptiform discharges. B: Same patient with com-
plete cessation of ictal activity after IV lorazepam, likely causing the widespread faster beta activity.
Chapter 29 ■ Nonconvulsive Status Epilepticus 617

FP1-F3

F3-C3

C3-P3

P3-O1

FP2-F4

F4-C4

C4-P4

P4-O2

FP1-F7

F7-T3

T3-T5

T5-O1

FP2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz
75 uV 1 sec

Figure 29.15 A 58-year-old woman with no history of epilepsy, admitted with confusion. EEG shows generalized 2.5-Hz
spike-and slow-wave activity. A few minutes after 2 mg IV lorazapam, the EEG showed mild diffuse slowing with plenti-
ful beta activity. The confusion improved concurrently with the EEG improvement, about 4 minutes after the lorazepam
administration.

frequencies from 0.5 to 4 Hz (180) (Fig. 29.15). The spike-and- The clinical manifestation of AASE is often a further slowing
wave component can be less prominent and more irregular or reduction in cognitive activity—from a poor baseline.
than in typical absence SE but it still shows a similar frontal or Patients are more confused or less responsive than before. They
centrally maximal distribution (29). may become mute or immobile, but some children are able to
follow simple commands (181). Consciousness may fluctuate
Atypical Absence Status Epilepticus and become markedly impaired. Patients exhibit poor control
Atypical absence status epilepticus (AASE) is a relatively rare of movements and frequent falls (pseudoataxia) and cognitive
type of SE that occurs primarily in children with cryptogenic difficulties with near stupor (pseudodementia) (27). AASE can
and secondarily generalized epilepsy, such as in the LGS with go on for days, without a precise onset or ending. There is typ-
substantial neurologic deficits, developmental delay, and severe ically a gradual onset and resolution of altered behavior in pro-
encephalopathies (70,181,182). These patients often have longed episodes, but automatisms and other motor activities
mixed epilepsies, with multiple seizure types and episodes of (such as walking) may continue, even while responsiveness is
tonic, atonic, and myoclonic seizures (Fig. 29.16). The inci- reduced (184). Often, there are no overt clinical seizures, and it
dence of atypical absence SE is anywhere from 100 to 200 can be very difficult to distinguish AASE from the encephalo-
episodes per million per year (144). pathic, developmental, or psychiatric behavioral baseline of the
Atypical absence seizures resemble typical absence seizures (often institutionalized) patients.
in their occurrence primarily in young patients, with general- LGS is the most common setting for AASE. With LGS and
ized spike-and-slow-wave discharges, and with the same favor- “myoclonic-astatic” seizures, there may be myoclonus or stupor
able (and unfavorable) responses to medications, for example, (182). When the AASE of LGS continues into adulthood, psy-
exacerbation with AEDs such as carbamazepine, and treatment chiatric features may predominate, including “regressive behav-
with ethosuximide and BDZs (183). Nevertheless, AASE is ior” or stubbornness; episodes of obtundation may point to
more complicated than typical absence SE. It presents at an ear- NCSE (28). Other etiologies include several genetic syndromes
lier age, typically before adolescence, with significant back- associated with mental retardation (181). Angelman syndrome
ground encephalopathies, both clinically and on EEG (181). can cause atypical absence seizures, with high-voltage bursts of
618 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz
Cz-Pz

Fp1-F3
F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4
P4-O2
Fp1-F7
F7-T3
T3-T5
T5-O1
Fp2-F8
F8-T4
T4-T6
T6-O2
Fz-Cz 300 uV
Cz-Pz
2 sec

Figure 29.16 Atonic status epilepticus in a 12-year-old girl with new-onset epilepsy. Generalized high-voltage slow waves
with irregular generalized spikes correlated with atonic episodes of the limbs and trunk. Ninety seconds of EEG show
recurrent atonic seizures; a similar pattern persisted for more than 30 minutes.

slowing on the EEG, evolving to spike and wave discharges later herald or terminate AASE. Long-term morbidity is essentially
in childhood (185). always considered due to the underlying neurologic condition.
Diagnosis of AASE is difficult, primarily because most patients
have severe underlying encephalopathies and neurologic deficits, EEG
along with a markedly abnormal mental status, even when not in The EEG of AASE is characterized by generalized spike or poly-
SE. AASE includes impaired responsiveness, prominent epilepti- spike and slow-wave discharges with a dominant frequency
form discharges, and electrographic seizures. To qualify as SE, below 3 Hz (slow-spike-and-wave), with somewhat less rhyth-
there should be a clear change from baseline, both clinically and micity and less perfect symmetry than seen in absence seizures
electrographically, but because patients often progress in and out (Figs. 29.17 and 29.18) (184). Discharge frequencies usually
of AASE, electrographic seizures may not correlate well with clin- range from 1 to 2.5 Hz and are often irregular, usually superim-
ical signs, and it is unclear what role the epileptiform discharges posed on a slow background. Interictally, the background is
play in the clinical syndrome. It could thus be considered another usually quite abnormal, including generalized or focal slowing
“boundary syndrome.” Radiologic scans are often unremarkable, with generalized spike and sharp waves, in isolation or in brief
but some show evidence of cortical dysplasia or hematomas (181). bursts, and admixed low-voltage generalized paroxysmal alpha
Episodes of AASE can recur frequently and remain refractory or beta frequency activity (70,182).
to treatment. Intravenous BDZs may interrupt the EEG dis- In a model of AASE with the same response to AEDs as in
charges without changing behavior substantially, but BDZs may human AASE, there was a gradual onset of reduced movement,
also precipitate tonic seizures or tonic SE in these patients (186). and epileptiform discharges were slower (183). Whereas in typi-
The underlying mental retardation and the difficulty of differen- cal absence seizures the epileptiform spike and wave discharges
tiating AASE from an interictal state has often led to less vigorous appear more restricted to thalamocortical circuitry, in experi-
treatment. When episodes end, the baseline neurologic func- mental models of atypical absence, epileptiform discharges also
tion is often still significantly impaired. Occasionally, convulsions involve the hippocampus and other deep limbic structures (187).
Chapter 29 ■ Nonconvulsive Status Epilepticus 619

Figure 29.17 Atypical absence status epilepticus (AASE) in a 49-year-old man with Lennox–Gastaut syndrome who pre-
sented with drooling and lethargy. Interictal EEG shows slow-spike-and-wave-activity at 1 Hz. EEG during AASE showed
2- to 2.5-Hz generalized spike-and-wave activity.

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

Fz-Cz

Cz-Pz

EKG
100 uV1 sec

Figure 29.18 Atypical absence status in a 26-year-old woman with tuberous sclerosis and Lennox–Gastaut syndrome.
Clinically, there were frequent episodes of staring and blinking lasting 20 to 30 seconds followed by confusion.
620 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Correspondingly, clinical AASE in humans appears to involve focal predominance. Among pediatric ICU patients, focal or
more of deeper structures, correlating with the more persistent multifocal NCSE (65%) was more common than generalized SE
memory and other cognitive problems in AASE, as opposed to an (35%), often reflecting underlying acute focal brain lesions (189).
apparent lack of residua following typical absence SE. The epilep- This ongoing electrographic (nonconvulsive, and usually sec-
tiform discharges of AASE also appear more likely to engender ondarily generalized) seizure activity is very common among
(pathologic) hippocampal synchronization than that occurs in critically ill ICU patients. Many have had earlier generalized
typical absence subjects, perhaps because of earlier cerebral dam- convulsions or GCSE, often in the setting of serious medical ill-
age that may have occurred in patients with LGS or other causes ness with cerebrovascular disease, sepsis, hypoxia, or severe
of severe childhood epilepsies (187). metabolic derangements. Those with minimal movement (typi-
cally nystagmus or myoclonus) after GCSE have been called
Secondarily Generalized Nonconvulsive “subtle” SE (190). Some are referred to as electrographic status
(Electrographic) Status Epilepticus epilepticus (ESE) (Fig. 29.21) (33). Others refer to “status epilep-
Most generalized NCSE in adults without IGE syndromes are not ticus in coma,” but not all patients with ongoing electrographic
truly absence SE, but rather secondary from a focus (16,49,139), seizures are comatose. Some label these patients with severe
even though the EEG and clinical signs are often generalized at the medical illness as having “epileptic encephalopathies,” indicating
time of diagnosis and management. Although secondarily gener- that the underlying disease causing the discharges is key, and
alized, these episodes of SE may appear very similar clinically (and that the epileptic component is not primary and may not
on EEG) to cases of primary generalized NCSE and it may be respond to AEDs. This term, however, is probably best reserved
impossible to tell the difference at the time of presentation. for childhood conditions such as those associated with ESES
Secondarily generalized NCSE is the true diagnosis of most cases (see section on Early Life, Age-related Nonconvulsive Status
described earlier in the literature as “absence SE,” but they have lit- Epilepticus Syndromes).
tle to do with typical absence epilepsy. In the IGE syndromes, The term “subtle” SE often describes the later stages of
absence SE is relatively uncommon, typically easy to treat, and has GCSE, in which initially overt motor seizures become subtle
a superb prognosis. Secondarily generalized NCSE usually has gradually, often in comatose patients (Chapter 28). The EEG
underlying (and sometimes severe) lesions, is much harder to may show discontinuous ictal activity with brief bursts of gen-
treat, and has the prognosis of the underlying illness. Secondarily eralized polyspikes or generalized periodic discharges (33,190).
generalized NCSE is the most common type of SE in ICUs. Some consider these EEG patterns to indicate ongoing SE only
if the patient had previous clinical seizures or SE (32). Many
EEG consider this NCSE as a natural progression from GCSE (190).
Often, generalized NCSE cases cannot be separated easily into Most agree that the urgency of treatment continues during the
those of a primarily generalized nature versus those with focal persistent electrographic seizures, at least when preceded by
onset spreading to a broader or even generalized distribution. generalized convulsions. Nevertheless, abolition of electro-
[Experimentally, generalized epileptiform discharges can result graphic seizures may not lead to a clinical improvement, and
from focal electrical stimulation in frontal regions (188)]. Rarely these patients have high morbidity and mortality (32,190–192).
does one capture the onset of SE on EEG except during continu- Despite the prominent epileptiform activity on EEG, the diag-
ous monitoring, and an EEG begun after the clinical onset of nosis of NCSE is not always clear (see sections below on EEG
NCSE may not allow distinction between focal and generalized Diagnosis of Nonconvulsive Status Epilepticus, and Controversial
SE. Nevertheless, the EEG can still be helpful in deducing whether EEG Patterns). In clinical practice, however, ESE (rhythmic, rela-
there was a focal onset to the seizures. Focal epileptiform dis- tively rapid epileptiform discharges, typical for SE on the EEG)
charges, especially with a consistent location, help point to a focal without obvious clinical manifestations should be considered
onset. Some episodes of apparent absence SE include prominent SE rather than simply an encephalopathy with epileptiform dis-
focal features on the EEG and clinically (138), possibly because the charges—even when treatment is unsuccessful in effecting a clin-
generalized epileptiform discharges, regular as they appear, are ical improvement. The EEGs are very similar to those in many
secondarily generalized (Figs. 29.19 and 29.20). Some of these published NCSE studies. Also, patients with ESE upon emergence
EEGs include discharges with more irregularity or asymmetry from pentobarbital or midazolam infusion treatment usually go
than are found on EEGs of typical absence SE. Also, some appar- on to have clinically evident seizures (193,194), indicating that
ently generalized epileptiform activity in NCSE evolves toward ESE is not just an EEG aberration. Finally, some patients with
unilateral discharges with treatment, suggesting a focal onset (45). these EEGs do respond well to AEDs (195).
Finally, some seizures have minimal or no clinical or EEG features
suggesting a focal onset but are associated with known unilateral NONCONVULSIVE STATUS EPILEPTICUS
structural lesions and are very likely to have focal origins. IN THE INTENSIVE CARE UNIT
In one review of EEG patterns during NCSE, there were three
main groups: (1) epileptiform discharges generalized at the The diagnosis of NCSE in acutely ill patients with abnormal
onset; (2) discharges that begin focally, with or without second- mental status is often complicated and difficult, and many other
ary generalization; and (3) patterns with both focal and general- conditions mimic NCSE. Among the many reasons for altered
ized features or otherwise poorly classified (45). In that series of behavior and impaired consciousness are severe toxic, metabolic,
85 episodes of NCSE in 78 patients, 69% showed a generalized and infectious encephalopathies; sedating medications; postictal
pattern, 13% a focal pattern, and 18% a generalized pattern with or postoperative encephalopathies (many due to medications);
Chapter 29 ■ Nonconvulsive Status Epilepticus 621

Figure 29.19 A: A 31-year-old woman with metastatic colon cancer, admitted with catatonia. EEG initially showed rhyth-
mic 1.5-Hz delta activity over the right hemisphere. B: Several hours later, the EEG shows generalized sharp-and slow-wave
discharges at 1.5 Hz, mimicking generalized status epilepticus. (Ten minutes after 4 mg IV lorazepam, the EEG showed
resolution of all rhythmic and sharp activity, and the patient became more responsive clinically.)

BDZ withdrawal states or toxicity; neuroleptic malignant and questioned and should be confirmed by EEG prior to more
serotonin syndromes; and other systemic illnesses. Several of aggressive treatment. It is sometimes necessary to exclude psy-
these conditions are accompanied by abnormal EEGs, sometimes chogenic nonepileptic SE (a conversion disorder), which does
even with epileptiform features such as sharp waves. not usually respond to AEDs (196,197). Pseudostatus can mimic
There are also patients treated for SE with the wrong diag- NCSE, with prolonged staring, blinking, and unresponsiveness,
nosis! When clinical manifestations are atypical and appear and even expert clinicians may be unable to distinguish it from
refractory to first-line AEDs, some diagnoses of SE should be SE. Often, psychogenic SE can be diagnosed definitively by EEG
622 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 29.20 A: A 46-year-old man with a right frontal cavernous malformation, admitted with bizarre behavior. Ictal
EEG during an episode of status epilepticus shows initial bilateral frontal (right left) polyspike and wave activity. B:
Within 10 seconds of onset, the EEG evolves to show generalized symmetric polyspike and wave activity at 4 Hz, sugges-
tive of primary generalized epilepsy. He had 32 seizures in a 4-hour period.

alone. The correct diagnosis can prevent inappropriate, aggressive the EEG (197). Following true epileptic seizures, postictal slow-
treatment with IV AEDs, general anesthesia, and intubation— ing is the rule, and the presence of a normal alpha rhythm
and their complications (198,199). immediately following apparent convulsions (or during brief
In pseudostatus, the EEG background activity is usually nor- pauses in motor activity) provides strong evidence for pseu-
mal (Fig. 29.22), but muscle and movement artifact may obscure dostatus (Fig. 29.23).
Chapter 29 ■ Nonconvulsive Status Epilepticus 623

Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

EogL-A1

EogR-A2

EKG
50 uV 1 sec

B
Fp1-F3

F3-C3

C3-P3

P3-O1

Fp2-F4

F4-C4

C4-P4

P4-O2

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp2-F8

F8-T4

T4-T6

T6-O2

EogL-A1

EogR-A2

EKG
75 uV 1 sec

Figure 29.21 A: Electrographic status epilepticus in a 50-year-old man with multiorgan dysfunction after anoxic insult.
The patient was comatose, but there were no other clinical manifestations. B: Same patient after treatment with IV leve-
tiracetam, now showing subtle generalized electrographic status epilepticus.
624 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 29.22 Nonepileptic psychogenic status epilepticus. A 51-year-old woman with history of epilepsy and left tempo-
ral lobectomy, admitted with more than 30 episodes per hour characterized by speech arrest, unresponsiveness, and closed
eyelids during the episode. EEG shows left hemisphere breach artifact from temporal lobe surgery, but no ictal changes.

On the other hand, ICU patients with impaired responsive- history of epilepsy, and remote symptomatic CNS insults (e.g.,
ness are not rarely in NCSE (98,140,200,201), especially when stroke, neurosurgical intervention, tumor, etc.), especially when
they have had earlier seizures or clinically evident SE (33,49, the patient is encephalopathic (21). Other risks for NCSE are
195,202). Of 164 patients who had EEGs after apparent control intercurrent electrolyte abnormalities, infections such as pneu-
of clinical SE in one series, 42% had continued seizure dis- monia or urinary tract infections, or glucose dysregulation.
charges, and 14% were considered in NCSE (98). Similarly, 8% of The patients at highest risk for unrecognized NCSE are those
patients with coma of all causes and similar monitoring (without who were in GCSE and were thought to have been treated suc-
clinical signs of seizures) had NCSE (200). With C-EEG monitor- cessfully—but were not! A prolonged “postictal” state after a
ing for the question of seizures, or to evaluate coma, another convulsion should raise concern for the possibility of ongoing
group found that 19% of patients monitored in a neurologic ICU NCSE, and EEG is usually the only way to make the diagnosis
over a 6-year period had seizures recorded, and 92% of these (33,49). After generalized convulsions, most patients begin to
were strictly nonconvulsive (203). Coma, prior epilepsy, age below recover responsiveness within 20 to 30 minutes, although there
18 years, and earlier convulsive seizures were risks for nonconvul- is a broad range. All patients with seizures or SE who do not
sive seizures. return to a normal level of consciousness should have EEGs to
There are many causes of NCSE in the ICU. Patients with see if treatment was adequate or the patient is still seizing.
acute neurologic injuries are at particular risk, especially those One group determined that nonconvulsive seizures and
with intracerebral or subarachnoid hemorrhage, CNS infection, NCSE should be suspected in the ICU when (1) a prolonged
brain tumors, severe head trauma, or following neurosurgery— encephalopathy follows generalized convulsions, an operation,
in each of which 20% to 30% of monitored patients have had or a neurologic insult; (2) there is acutely impaired or fluctuat-
nonconvulsive seizures in different studies (201,203–206). With ing consciousness interrupted by normal alertness; (3) there is
C-EEG monitoring of patients with intracerebral hemorrhage, impaired consciousness with facial myoclonus or nystagmus;
about one-third had seizures (although patients were selected in (4) staring, aphasia, or automatisms (e.g., limb or facial) occur
part due to prior seizures or a suspicion of seizures) (206). Half episodically; or (5) other acutely altered behavior has no obvi-
of seizures detected were purely nonconvulsive. ous cause (191).
Coma is a particular risk for NCSE in the ICU, especially if How long should C-EEG be continued in the search for non-
the cause is unclear. In one study, more than half of 97 coma- convulsive seizures and NCSE in patients with reduced respon-
tose patients undergoing C-EEG had electrographic seizures siveness? In one study, a routine 30-minute EEG identified
(203). Other risks include recent generalized convulsions, a electrographic seizures in 11% of 105 critically ill patients, while
Chapter 29 ■ Nonconvulsive Status Epilepticus 625

Figure 29.23 Psychogenic generalized status epilepticus. This 22-year-old woman had prolonged episodes of generalized
on–off tremulous limb movements. The EEG shows repetitive EMG artifact, with normal background EEG activity evident
in the areas between the tremor artifact.

the subsequent C-EEG (for a median of 2.9 days) found seizures EEG may miss them, so many modern ICUs utilize C-EEG to
in 27% (207). In another series, only half of 110 patients with detect seizures and other changes in neurophysiologic function.
seizures seen eventually on EEG had them during the first hour The diagnosis of seizures and NCSE on an EEG relies on the
of recording (208). In that series, 95% of noncomatose patients determination by a clinical neurophysiologist that certain pat-
had their seizures found in the first 24 hours, versus 80% for terns are “ictal” in nature, that is, having to do with epileptic
comatose patients. After 48 hours, this increased to 98% and seizures, but controversy exists regarding which EEG patterns are
87%, respectively. Among children with seizures, half had the diagnostic of, or consistent with, NCSE. Sometimes, seizures and
first seizure detected in the first hour of EEG recording, and 80% NCSE are readily apparent on the EEG, with characteristic or
within 24 hours (209). The authors concluded that C-EEG mon- even “classic” features. Other EEGs show disorganized slowing,
itoring was appropriate for 24 hours in patients without coma without rhythmicity or epileptiform abnormalities, more sugges-
and for 48 hours in comatose patients, in those with periodic tive of encephalopathies than seizures. Very similar EEG findings
discharges, or when AEDs are being withdrawn (209). can be seen in NCSE and in metabolic encephalopathies, and
many EEG patterns are neither pathognomonic nor easy to inter-
EEG DIAGNOSIS OF NONCONVULSIVE pret. Even after the diagnosis of NCSE, the clinical significance of
STATUS EPILEPTICUS the epileptiform discharges can be unclear—that is, the electro-
graphic seizure activity may or may not be the cause of the clini-
Some unresponsive patients in ICUs (and elsewhere) are having cal abnormalities.
seizures or NCSE, and others are not. It is often impossible to tell Also, the orderly temporal progression of EEG patterns
the difference solely by clinical examination. EEG is far superior described for GCSE (Chapter 28) usually does not occur in NCSE
to any other technique in detecting nonconvulsive seizures and is (32,210). In one study of 40 patients, 53% had discrete seizures
crucial in the assessment of unresponsive patients, particularly alone, without any other ictal patterns; 28% had discrete seizures
those who have had earlier convulsions. Nevertheless, seizures with periodic epileptiform discharges (PEDs) between seizures;
(including nonconvulsive) can be intermittent, and an individual and 8% had continuous ictal activity (210). Occurrence of
626 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 2 9 . 4

Tentative Research Criteria for Nonconvulsive


Seizures [and if 30 minutes, Nonconvulsive
Status Epilepticus]

1. Repetitive generalized or focal spikes, polyspikes, sharp


waves, spike-and-wave or sharp-and-slow wave complexes,
or other rhythmic waveforms at 2.5/sec, lasting longer
than 10 seconds.
2. Same waveforms as above, with discharges 2.5/sec, with
clear clinical ictal phenomena, such as facial twitching,
nystagmus, or limb myoclonus.
3. Same waveforms as above, with discharges 2.5/sec, with
an unequivocal evolution of the rhythmic pattern,
including increase or decrease in frequency (by 1 Hz)
and change in discharge morphology or in location
(gradual spread of rhythmic activity into or out of a region
involving at least two electrodes). Changes in discharge
amplitude or “sharpness” alone are not sufficient.
4. Rhythmic theta or delta waves at 1/sec, with additional
criterion of unequivocal clinical improvement or
improvement on EEG (such as resolution of epileptiform
discharges and reappearance of previously absent normal
background rhythms and reactivity) or both, following
quickly after acute administration of rapidly acting AEDs,
typically benzodiazepines. (Resolution of discharges
leaving a slow background alone, without clinical
improvement, would not suffice.)

Figure 29.24 Various EEG waveform morphologies seen in NCSE.

patterns, and on electrographic definitions of NCSE. A commit-


continuous seizures, discrete seizures, or PEDs could occur at any tee of the American Clinical Neurophysiology Society (ACNS)
time after SE, and no predictable pattern of evolution was iden- has recommended standardization of terminology for periodic
tified. EEG evolution probably depends on the etiology and type and potentially epileptiform EEG findings in order to facilitate
of SE, baseline condition of the patient, duration of seizure activ- multicenter clinical research (211). They recommend that EEG
ity, and type and duration of treatment. The variability in evolu- findings be described by morphology, location, frequency, and
tion adds to the controversy regarding which patterns are “ictal.” periodicity of waveforms—without using clinical connotations,
Seizures captured in their entirety typically show progression but with subsequent interpretation in the clinical context. The
from low-voltage, high-frequency spikes to high-voltage, low-fre- committee also suggested tentative criteria for the EEG diagno-
quency spike-and-slow wave activity, before stopping abruptly sis of seizures and, if persistent enough, NCSE, to be tested
and being replaced by background slowing or suppression (Fig. empirically, similar to those in Table 29.4 (72,191,204).
29.9). Usual morphologic features include typical rhythmic, gen- Seizures are generally indicated by (1) repetitive focal or gen-
eralized, symmetric spike-and-waves or polyspikes and waves at eralized epileptiform activity or rhythmic waveforms faster than
2 to 3.5 Hz; atypical spike and wave with lower frequency and 2.5 Hz, lasting longer than 10 seconds; or (2) similar discharges
less symmetry; multiple spike-and-wave (repetitive complexes of slower than 2.5 Hz with (a) unequivocal evolution in frequency,
two or more spikes followed by a slow wave); and high-voltage, morphology, or field; (b) clinical motor manifestations (e.g.,
repetitive, rhythmic, focal or generalized delta activity with inter- limb or facial twitching); or (c) clear clinical and EEG improve-
mixed spikes, sharp waves, or sharp components (Fig. 29.24) ment after administration of rapid-acting intravenous AEDs,
(45). Diagnosis is more difficult when the seizure (or SE) pre- typically BDZs. “Evolution” of the electrographic discharges of
cedes the beginning of the tracing and continues beyond its end. seizures on the EEG over time includes an increase or decrease
In such cases, rhythmic sharp features, typically faster than 1 Hz, in frequency by at least 1 Hz, significant change in waveform
may be seen, often with variability. morphology, or spread to at least two adjacent EEG electrodes
There is a lack of consensus on the terms and descriptors for (72,191) (Fig. 29.9). Diagnostic criteria for invariant patterns
electrographic seizures and periodic and other abnormal EEG and discharges slower than 2.5 Hz are more controversial (212).
Chapter 29 ■ Nonconvulsive Status Epilepticus 627

Figure 29.25 Generalized periodic epileptiform discharges (GPEDs) in a 62-year-old woman with intractable epilepsy.
Variable GPED repetition rate up to 3 Hz, with intervening periods including rhythmic fast activity and variable morphol-
ogy, which may represent ictal activity. After treatment with IV lorazepam, the background showed mild diffuse slowing
but no further ictal activity. The patient became less confused and was able to answer questions.

Slower discharges are more likely to signify epileptic seizures if rhythmic midtemporal theta activity, subclinical rhythmic EEG
they evolve in frequency or follow clinical SE. discharges of adults (SREDA), and wicket spikes (204) (see
Straightforward examples of NCSE on EEG conform to the also Chapter 14, on normal variants). Other patterns generally
patterns described above and continue for over 30 minutes. not considered to represent NCSE include periodic discharges
Whatever the generalized or focal pattern, if the patient returns in patients with acute cerebral injuries but without prior
rapidly to a baseline state (after AED treatment or sponta- clinical seizures, and discharges in patients with epileptic
neously) and the EEG becomes normal, then this very likely was encephalopathies in which the periodic discharges appear sim-
NCSE (Fig. 29.25). Resolution of the epileptiform discharges on ilar to those on the baseline EEG (72,213).
EEG and resumption of a posterior alpha frequency back- Frequently, the EEG determination of seizures is not a
ground suggestive of wakefulness are also considered diagnos- blinded exercise. The interpreter may be aware of the patient’s
tic of an earlier seizure. staring, facial twitching, or subtle myoclonus, and be predis-
These criteria may provide for greater certainty in the diagno- posed to diagnose ambiguous patterns as NCSE. For example,
sis of electrographic seizures and NCSE, but they were designed the appearance of periodic discharges at 1 Hz in a patient with
as guides for research purposes, and some patients whose EEGs focal twitching would favor a diagnosis of seizure over that of
do not meet these strict criteria may also be having seizures or be an interictal periodic pattern (43,46). Often, EEG determina-
in NCSE (204). For example, in well-established clinical cases of tion of seizures needs re-interpretation in the clinical setting.
NCSE, stereotypic spike-and-wave activity at 3 Hz was seen in
only 7% of patients; discharge frequency was usually 1 to 2.5 Hz CONTROVERSIAL EEG PATTERNS
(45). Similar rhythmic and sharp EEG patterns are still strongly
suggestive of seizure activity in the appropriate clinical setting, Periodic Discharges
especially following earlier generalized convulsions or GCSE, Sharp and rhythmic EEG features vary remarkably and range
or with severe medical illness. Many are associated with mini- from the nonictal “irritative” (post-, inter-, or preictal) to the
mal or no motor convulsive activity, as in the ESE described “actively seizing”—along an “ictal–interictal continuum”
above in the section on Secondarily Generalized Nonconvulsive (43,46,72,214). They include PEDs with various repetition rates
(Electrographic) Status Epilepticus. and morphologies, including PLEDs and bilateral independent
Some epileptiform EEG findings generally agreed upon to periodic lateralized epileptiform discharges (BiPLEDs) (see
not represent seizures include midtemporal theta of drowsiness, Chapter 28), and generalized periodic epileptiform discharges
628 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 29.26 EEG of a 72-year-old man following cardiac arrest. On weaning of propofol, continuous electrographic
seizures began, with repetitive spike-and-slow-wave discharges at 2.5 to 3 Hz.

(GPEDs). Periodic discharges are more likely to be “ictal” when Focal or generalized periodic and quasi-periodic discharges
they are rapid ( 2 Hz) (Fig. 29.26), or show clear evolution in can also be elicited in stuporous or comatose patients upon
frequency (increase or decrease by at least 1 Hz), morphology, or stimulation—“stimulus-induced rhythmic, periodic, or ictal
field (72,204). Periodic discharges with intervening fast and discharges” or SIRPIDs (Fig. 29.29) (220). SIRPIDs usually abate
admixed sharp components (e.g., “PLEDs-plus”) are also more after the stimulus recedes. Whether they represent seizures or an
likely to be “ictal” than simple isolated discharges (see also abnormal arousal process is often unclear. Some are definitely
Chapter 28, Fig. 29.9A,B) (215). The presence of PEDs should seizures (221). When elicited along with facial or limb jerking
prompt consideration of C-EEG monitoring to look for electro- (as in a minority of cases), they are more likely to be ictal phe-
graphic seizures arising out of that background. nomena. Many appear to lie along the “ictal–interictal contin-
GPEDs are continuous generalized spikes, polyspikes, sharp- uum,” especially when there is no clinical motor correlate.
and-slow waves, or TWs, often with a repetition rate of about 1
Hz, typically arising from a diffusely slow or attenuated back- Triphasic Waves
ground (Figs. 29.25 through 29.28) (213). They often occur in Another controversial area in EEG diagnosis is that of triphasic
obtunded patients, typically after anoxia or other catastrophes, waves (TW)—bursts of moderate- to high-voltage (100 to 300
metabolic insults, recent overt seizures, or in the late stages of V) rhythmic complexes, often recurring at 1 to 2 Hz (Fig.
GCSE (190). GPEDs are highly associated with clinical seizures 29.30A,B). TWs are named for their multiphasic morphology,
but not necessarily indicative of seizures proceeding at the same consisting of an initial blunted, low-voltage negative spike-like
time (210,216). Some consider GPEDs to be reliably ictal and phase; a second phase consisting of a slowly rising high-voltage
recommend aggressive AED therapy (217), whereas others positive component; and third phase of a lower voltage, broad
believe that they are the sign of neuronal injury, but not actual negative slow wave. TWs are broadly distributed, usually with
seizures and not requiring aggressive treatment (Figs. 29.27 and a frontal voltage maximum. They frequently occur in clusters,
29.28) (216,218). One study did not find any features that could but may be continuous at 0.5 to 2 Hz (47). TWs are commonly
distinguish clearly between GPEDs after anoxia and GPEDs state responsive, increasing with stimulation (Fig. 29.30B) and
after SE (219), even though those conditions have markedly dif- attenuating with deeper levels of stupor or coma, and may be
ferent implications for treatment. GPEDs tend to persist even abolished or regress after administration of IV drugs or other
with aggressive therapy, and it is not known whether patients AEDs (47). TWs are often associated with diminished levels
benefit from treatment of these EEG findings. of consciousness, and occasionally with myoclonus. There is
Chapter 29 ■ Nonconvulsive Status Epilepticus 629

Figure 29.27 Generalized periodic epileptiform discharges (GPEDs) at the relatively slow repetition rate of 1 to 1.5 Hz
are usually not considered ictal activity.

Figure 29.28 Generalized periodic epileptiform discharges (GPEDs) in a 77-year-old man following cardiac arrest. Note
the absence of almost all EEG activity other than the discharges.
Figure 29.29 Stimulus-induced rhythmic, periodic, or ictal discharges (SIRPIDs) in a 17-year-old girl with encephalitis.
Despite high-dose thiopental, she continued to have frequent electrographic seizures, both spontaneously and induced by
stimulation as in this example. Some SIRPIDs were brief and seen only on EEG, but others evolved into clinical general-
ized convulsions.

Figure 29.30 A: A 65-year-old woman with uremia and intermittent episodes of unresponsiveness. There are occasional 1
Hz frontally maximal triphasic waves (TWs). The inset shows the characteristic anterior to posterior gradient of TWs—the
first negative component in channel 1 (frontal) precedes the negative component in channel 3 (occipital) by about 50 msec.
Chapter 29 ■ Nonconvulsive Status Epilepticus 631

Figure 29.30 (Continued) B: With stimulation, the TWs increase in amplitude, field, and frequency, mimicking electro-
graphic seizure activity.

often an underlying toxic or metabolic state, including uremia, waking pattern are more suggestive of NCSE, whereas resolu-
hepatic insufficiency, hypothyroidism; toxicity from lithium, tion of TWs leaving an encephalopathic slow background sug-
baclofen, ifosfamide, tiagabine, cefapime, neuroleptics, and gests a nonepileptic encephalopathy. Still, some true NCSE do
other medications; or the neuroleptic malignant or serotonin not respond to AEDs or may also give way to an encephalo-
syndromes (41,47,222). pathic pattern without clinical improvement.
TWs have long been a source of confusion because of their
“epileptiform” appearance on EEG even while they occur in both TECHNOLOGY
encephalopathies and NCSE. Typical TWs are generally not con-
sidered epileptic by most investigators, but they may be identical Rapid detection of nonconvulsive seizures, in the ICU or else-
to the discharges that occur in definite NCSE, especially when where, requires near “real-time” review of EEG. Linking contin-
they have a frequency exceeding 1 Hz (223). TWs are usually of a uous digital EEG recordings to a hospital computer network
broader, more blunted appearance than typical epileptiform dis- allows EEG technologists and EEG-ers to access ongoing EEG
charges. Epileptiform discharges in generalized NCSE usually from a variety of locations, including offsite over the Internet.
have higher frequencies (mean 2.4 Hz vs. 1.8 Hz), more poly- It also facilitates postrecording adjustment of montages, sensi-
spikes (69% vs. 0%), sharper morphologies, and less concomitant tivity, and filters; acquisition and storage of large amounts of
background slowing than TWs associated with encephalopathies data; quantitative data analysis; and automated spike and
(224). TWs also have a relative lack of phase reversal, longer seizure detection. For C-EEG to be useful for clinical decisions,
duration, and a lesser slope of incline in the third phase of the records should be reviewed and interpreted by trained person-
waveform. A phase lag from anterior to posterior channels is nel several times a day. In one study, only 40% of nonconvulsive
often seen with TWs (Fig. 29.30A), but not usually with NCSE. seizures were detected visually online at the time of the seizure,
Facial or limb myoclonus is more suggestive of SE. Table 29.5 lists with the remainder found at later review (225).
characteristics that can help to distinguish NCSE from TWs. C-EEG produces more data than an individual clinical neu-
Resolution of TWs in response to administration of BDZs rophysiologist can review, at least in its raw form; for example,
may suggest a diagnosis of NCSE, but BDZs may also abolish following an episode of SE, long periods of C-EEG must be
typical, even rhythmic, TWs from an underlying encephalopa- reviewed to ensure that breakthrough or recurrent seizures have
thy, usually without clinical improvement (41,46,47). not occurred. Several digital analysis methods and graphical
Resolution of abnormalities and resumption of a normal alpha displays of quantitative EEG (QEEG) are commonly used to
632 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 2 9 . 5

Triphasic Waves versus Periodic Epileptiform Discharges

Triphasic Waves:
Surface-negative, blunted triphasic complexes with (a) low-amplitude, blunted, negative first phase (often wide-based); (b)
dominant, steep positive second phase; and (c) slow rising third “slow-wave” component. No polyspike components.
Complex duration: 400–600 msec
Amplitude: 100–300 V on referential montage; smaller on bipolar.
Frequency: 1.0–2.5 Hz (typically 1.8 Hz).
Persistence: wax and wane, but 10% of a standard 20-minute recording.
Evolution/reactivity: decrease with sleep, drowsiness, or after benzodiazepines; increase and reappear with arousal or noxious
stimulation.
May exhibit phase lag, seen best on referential montage.
Periodic Epileptiform Discharges:
Surface-negative bi-, tri-, or polyphasic discharges with spike, sharp, or polyspike components; slow-wave complexes; or
combinations of these.
Complex duration: 60–600 msec (mean 200 msec)
Amplitude: 50–300 V (usually up to 150 V)
Frequency: 0.2–3 Hz (usually 0.5–2.0 Hz)
Persistence: minimum of 10 minutes in an EEG recording.
Evolution: static—with only minor variability in the above characteristics.

reduce the enormous quantity of data produced by C-EEG. currently available software has been “trained” and validated
These methods allow rapid detection of even subtle changes in primarily on samples from EMUs and ambulatory epilepsy
EEG activity, obviating review of thousands of pages or screens patients (229,231) and is inadequate for detection of ictal pat-
of raw EEG data (226). terns in patients with critical neurologic illnesses. New soft-
Automated detection programs identify EEG segments that ware trained on these patients, and better suited to C-EEG in
include events of interest, such as epileptiform spikes or patterns ICUs, should increase the utility of these algorithms in the ICU
suggestive of seizures, flagging events for later review by EEG-ers (212,226,227,232).
(Fig. 29.31). Several commercially available systems have algo- One commonly employed technique involves creation of
rithms designed to identify paroxysmal rhythmic activity with graphical displays of QEEG data. QEEG uses fast Fourier trans-
increased voltage or frequency compared to background activity formation of digital EEG signals into their component frequen-
(227–229). The sensitivity of variables such as frequency (usu- cies and amplitudes. Compressed spectral array (CSA) is the
ally 3 to 20 Hz), field, and voltage can be adjusted to refine detec- most commonly used graphical display for QEEG. EEG data for
tion. Alternatively, artificial neural networks can be trained to single channels or combinations of channels over each hemi-
detect events by presenting many samples representative of sphere, displayed as color graphs of power (e.g., total power;
seizures and nonseizures, with the network trained to distin- power in certain frequency bands, usually theta and alpha; ratios
guish between the two patterns (227,230). of power in certain bands to total power; etc.) versus time can be
A practical concern in using detection algorithms is that used to detect electrographic seizures (Fig. 29.32) (212). QEEG
nonconvulsive seizures and NCSE exhibit very different elec- methods can help to select episodes for review by EEG-ers (209).
trographic patterns in critically ill patients from those in CSA displays can also assist with an initial, bedside EEG inter-
patients in standard epilepsy monitoring units (EMUs) being pretation by ICU personnel and physicians not trained in EEG
evaluated for epilepsy surgery or for a question of seizure ver- (212,233), who can then notify EEG technologists or clinical
sus pseudoseizure (209). Seizures and NCSE in critically ill neurophysiologists to review the EEG sections in question.
patients often have poorer organization, slower frequencies, Still, automatic detection algorithms do not detect all seizures,
longer duration, and indistinct onsets and endings. The differ- and they often select artifacts as possible seizures; the sensitivity
ence in electrographic seizure morphology makes it harder for and specificity of these techniques for NCSE in the ICU have not
standard algorithms to detect seizures in ICUs. Diffuse back- been determined well. Power changes can be caused by seizures,
ground slowing, focal slowing or attenuation, and breach arti- periodic patterns, changes in sleep–wake states, arousal, or arti-
fact may also alter the sensitivity of detection algorithms. Most facts, and the specific cause cannot be determined by reviewing
Chapter 29 ■ Nonconvulsive Status Epilepticus 633

FFT Power 6–14 Left

FFT Power 6–14 Right

FFT Spectrogram 1–20Hz Left


FFT Power 6-14 Left

FFT Spectrogram 1–20Hz Right

Figure 29.31 CSA shows total power in 6 to 14 Hz band in the left hemisphere (top line) and right hemisphere (second
line). Third and fourth lines show color density spectrogram in the right and left hemispheres, respectively. Peaks (in blue,
top two arrays; white, bottom two arrays) indicate seizures. Recurrent generalized seizures are eventually controlled by IV
thiopental (thick arrow), but recur soon as brief seizures (thin arrow), which gradually increase in duration (white arrow).
(See color insert)

the CSA display. The associated original, raw EEG data must be ICU patients who are restless or agitated produce substantial
available for review to exclude state changes and artifacts altering movement artifact on EEG. Nonepileptic but rhythmic or parox-
the QEEG, and to confirm that electrographic seizures are not ysmal movements such as dystonic posturing and (nonepileptic)
missed (Fig. 29.32B). Computer analysis can aid in data compres- myoclonus can produce artifacts as well. Comatose ICU patients
sion, but it cannot currently replace expert clinical neurophysiol- may have clinical episodes of twitching, tremors, posturing, or
ogist review. sudden changes of heart rate or blood pressure that may appear to
be seizures clinically, and by video. This is not diagnostic unless
ARTIFACTS correlated with simultaneous EEG evidence of seizure activity
(221). The EEG may show some such activity to be very unlikely
C-EEG in the ICU is particularly susceptible to contamination to be epileptic, and inappropriate to treat as seizures. Accurate
with a variety of artifacts, hindering accurate interpretation (234). interpretation is also facilitated by staff assessment of the patient
Electrode integrity can be difficult to maintain, necessitating use and recording nursing interventions on the EEG, and by video and
of needle electrodes or disk electrodes affixed with collodion. ICU audio recording obtained simultaneously with the EEG (which
equipment, bed machinery, hemoperfusion pumps, and ventila- also helps in diagnosing pseudoseizures). Findings that appear
tors may produce 60-Hz line noise. Artifacts include ECG, ballis- “epileptiform” may be due to artifact, and others may be seizures
tocardiographic, and pacemaker effects. They can also result from when they appear at first to be artifact (221,234). Video–EEG cor-
skull defects and scalp edema. Nursing care, stimulation, sponta- relation also aids in clarifying the clinical and behavioral correlates
neous arousals, chewing artifact, and chest percussion during of nonconvulsive seizure activity. Similar behavior might other-
physical therapy can all produce artifact. Some artifacts are peri- wise be ascribed to postictal or interictal confusion or psychosis.
odic or rhythmic and mimic epileptiform discharges or electro- Many groups use video now even with routine EEG recordings, to
graphic seizures (Figs. 29.33 and 29.34). look for artifacts and evidence of their causes.
A
Seizure detections

Seizure probability

Rhythmic run detector, 1–24 Hz, left

Rhythmic run detector, 1–24 Hz, right

FFT spectrogram, 1–24 Hz, left

FFT spectrogram, 1–24 Hz, right

Figure 29.32 A: A 30-year-old woman with mild mental retardation and intractable epilepsy admitted with lethargy and
intermittent head and eye deviation to the right. Continuous EEG monitoring with automated seizure detection shows fre-
quent right hemisphere seizures over a 2-hour period. Seizures are apparent both on rhythmic run detection and display
(black arrows), and color spectrogram (white arrows). EEG showed frequent electrographic seizures beginning with rhyth-
mic beta activity in the right occipital region (seen on traditional EEG display in B). B: Individual episode of seizure activ-
ity (corresponding to large black arrow on rhythmic run detection and color spectrogram in Fig. 29.31A). EEG showed
frequent electrographic seizures beginning with rhythmic beta activity in the right occipital region, gradually increasing in
amplitude and spreading to the right posterior temporal region and left occipital region. (See color insert)
Chapter 29 ■ Nonconvulsive Status Epilepticus 635

Figure 29.33 Rhythmic artifact from chest percussion obscures much of the EEG.

Fp1-F7
F7-T3
T3-T5
T5-O1

Fp2-F8
F8-T4
T4-T6
T6-02

Fp1-F3
F3-C3
C3-P3
P3-O1

Fp2-F4
F4-C4
C4-P4
P4-O2

Fz-Cz 75 uV 1 sec
Cz-Pz

Figure 29.34 Bed percussion artifact.


636 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

TREATMENT OF NONCONVULSIVE STATUS may occur shortly thereafter (Figs. 29.35 and 29.36; see also Fig.
EPILEPTICUS IN THE ICU 28.9A,B). Without C-EEG, the seizures will not be detected.
When NCSE recurs, it should be re-treated while following
There have been no randomized controlled trials evaluating the the EEG, but the best method, intensity, and duration of treat-
treatment of NCSE. Treatment decisions are extrapolated from ment have not yet been determined (see the section “EEG Use
trials of GCSE treatment or tailored to the perceived urgency and in Refractory Status Epilepticus” of Chapter 28). For refractory
morbidity of the type of NCSE encountered (235,236). The like- NCSE, many neurologists will use additional standard AEDs
lihood of successful treatment is strongly related to the etiology and then a trial of aggressive treatment (often with midazolam,
and type of NCSE. Treatment and outcome of absence, primarily pentobarbital, or propofol) to at least suppress all electro-
generalized NCSE (i.e., NCSE in the IGE syndromes), and CPSE graphic and clinical seizures (239,240).
are detailed earlier in the discussions of the several individual C-EEG aids in the recognition of nonconvulsive, electro-
types of NCSE. graphic seizures and may also provide important prognostic
Absence and de novo absence SE are usually treated success- information (see Chapter 28). Earlier diagnosis and EEG-guided
fully and easily and seldom require aggressive treatment. treatment may improve the care of patients with SE, but its actual
Atypical absence SE is much harder to diagnose and treat. effect on outcome has not yet been established (237,241). No
Except for occasional AASE and some cases of CPSE (also diffi- prospective studies have evaluated the use of C-EEG compared to
cult to treat at times), they are seldom seen in ICUs. Other routine EEG or other clinical measures for SE in the ICU.
forms of NCSE, particularly secondarily generalized NCSE, are
more often refractory to treatment (24). MORBIDITY OF NONCONVULSIVE STATUS
Patients with NCSE in the setting of earlier epilepsy are usually EPILEPTICUS
treated successfully and generally warrant a less aggressive
approach than does GCSE—unless the NCSE is the continuation Outcome for specific NCSE types was detailed earlier along
of GCSE (157). Treatment is usually similar to that for GCSE with the clinical features of simple partial SE, absence SE, and
(see the section “Treatment of Refractory Status Epilepticus” in well-defined cases of CPSE, but many clinical series describe
Chapter 28), but the most aggressive therapies, often those caus- NCSE without specifying the type. The prognosis for these
ing respiratory compromise, are usually avoided (158,159), unless many different illnesses depends heavily on the type of NCSE,
NCSE is refractory (236). Even with NCSE following GCSE, or in as identified clinically and by EEG, and on their causes and pre-
the setting of acute medical illness with severe encephalopathy, cipitants, and may also be influenced by age, concomitant med-
much of the diminished level of consciousness and consequent ical illness, duration of SE, and the effectiveness and
neurologic morbidity stems from the underlying cause of seizures complications of treatment. Etiology is by far the most impor-
rather than from the seizures themselves (195,237). tant prognostic factor. Some types of NCSE have an excellent
While risks of overtreatment exist, most patients with NCSE prognosis, without evidence of morbidity in any case, but oth-
should be treated quickly with AEDs because they are having ers have a high likelihood of a fatal outcome. For example, there
ongoing seizures with impaired consciousness and other neuro- is essentially no mortality with simple partial sensory SE (107).
logic deficits that are potentially reversible. There is also the Longer-term risks of NCSE should consider both experi-
attendant morbidity of incidental trauma, aspiration pneumo- mental and clinical data—but most basic studies on the damage
nia, and so on, and many episodes of NCSE begin, and may due to SE have used models of GCSE. While GCSE poses a def-
end, with potentially harmful generalized convulsions. inite threat to health and warrants vigorous treatment (Chapter
28), there is very little evidence of lasting harm due to NCSE
EEG USE IN THE ICU MANAGEMENT alone (242). The treatment imperative is less, but not negligible.
OF NONCONVULSIVE STATUS EPILEPTICUS NCSE is potentially “underdiagnosed and overtreated” (136).
In non-ICU patients, clinical reports of NCSE show few
Once nonconvulsive seizures or NCSE have been diagnosed, long-term sequelae of the NCSE itself (25,26,44,243), but most
most patients still need EEG monitoring during prolonged had limited follow up. Cognitive deficits caused by NCSE,
treatment in the ICU. Because clinical signs of NCSE are non- including language dysfunction, can persist for months (51).
specific, subtle, or nonexistent, C-EEG is usually necessary to One study of 65 NCSE patients found good outcomes in all but
guide the use of intensive treatment and to assess the response to one (244). Another reported neuropsychologic testing on 15
therapy (Chapter 28). NCSE persists in 14% to 20% of patients epilepsy patients (without underlying lesions or “symptomatic”
after cessation of clinically evident seizure activity (98). EEG also causes) before and after episodes of SE (half nonconvulsive)
helps to determine if NCSE recurs after treatment. Relapse of (245) demonstrating that no significant cognitive morbidity
NCSE is not rare, especially in the first 24 hours of treatment accrued from the SE. Long-term cognitive complications of
(98) and also when AEDs are being tapered, but it portends a NCSE appear uncommon, occurring primarily in the most pro-
worsened outcome and should be avoided if possible (238). longed and severe cases.
Individual seizures can be missed if EEG monitoring is inter- Outcome is not nearly as favorable for patients with second-
mittent. A given EEG may show a “postictal” encephalopathy, arily generalized NCSE in the setting of serious medical and neu-
drug effect, isolated or periodic discharges, or a burst suppres- rologic illness, especially in the ICU. The physiologic effects of
sion tracing at one point, and nonconvulsive seizures or NCSE persistent NCSE in critically ill patients can include an increased
Chapter 29 ■ Nonconvulsive Status Epilepticus 637

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Fp1-F7

F7-T3

T3-T5

T5-O1

Figure 29.35 Evolution of an electrographic seizure in a comatose 55-year-old woman with a left temporal intracerebral
hemorrhage. EEG shows left temporal periodic lateralized epileptiform discharges (PLEDs) evolving into faster frequency,
sharply contoured delta activity. Evolution of the seizure includes spread in the left temporal region, and then left parasagit-
tal region, and eventual end of the seizure. The patient remained comatose, without clinical manifestation of the seizure.

Fp1-F3

F3-C3
C3-P3
P3-O1
Fp2-F4
F4-C4
C4-P4

P4-O2
Fp1-F7

F7-T3
T3-T5
T5-O1

Fp2-F8
F8-T4

T4-T6
T6-O2
Fz-Cz

Cz-Pz
LUC
RLC
EKG
75 uV 1 sec

Figure 29.36 A focal seizure arises from the left hemisphere after an EEG showing PLEDs (see Fig. 28.7A) in the left pari-
etal and posterior temporal regions in a 26-year-old woman with adult onset Rasmussen encephalitis and earlier focal sta-
tus epilepticus.
638 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

cerebral blood flow, intracranial pressure, oxygen demands, and 9. Berger H. Über das Elektrenkephalogramm des Menschen. Arch
the lactate–pyruvate ratio, even when clinical signs are absent Psychiatr Nervenkr. 1929;87:527.
(246,247). NCSE might exacerbate neuronal damage in patients 10. Gloor P. The work of Hans Berger. In: Cobb WA, ed. Appraisal and
with acute insults such as traumatic brain injury (TBI) or stroke Perspective of the Functional Explorations of the Nervous System.
Handbook of Electroencephalography and Clinical Neurophysiology.
(201). Patients with both intracerebral hemorrhage and noncon-
Amsterdam: Elsevier Publishing Co.; 1971:IA11–IA24.
vulsive electrographic seizures had a worse outcome in two
11. Lennox WG. The treatment of epilepsy. Med Clin North Am. 1945;
reports, but it was not possible to determine if this was due to 29:1114–1128.
seizures or (more likely) the underlying illness (205,206). Among 12. Penfield W, Jasper HH. Epilepsy and Functional Anatomy of the
94 consecutive patients with moderate-to-severe TBI, 22% had Human Brain. Boston: Little Brown; 1954.
electrographic seizures on EEG and 29% were in NCSE, with 13. Gastaut H, Roger A. Sur la signification de certain fugues épilep-
higher mortality in these patients than in similar trauma patients tiques: état de mal temporal. Rev Neurol. 1956;94:298–301.
without seizures (201). It remains unclear whether treatment of 14. Shorvon SD. What is nonconvulsive status epilepticus, and what
nonconvulsive seizures would have changed this (248). In one are its subtypes? Epilepsia. 2007;48(Suppl 8):35–38.
series of 49 patients with nonconvulsive seizures, mortality cor- 15. Lowenstein DH, Bleck T, Macdonald RL. It’s time to revise the def-
related with etiology, age, and length of ICU stay, but only seizure inition of status epilepticus. Epilepsia. 1999;40:120–122.
16. DeLorenzo RJ, Hauser WA, Towne AR, et al. A prospective, popula-
duration and delay-to-diagnosis were significant predictors upon
tion-based epidemiologic study of status epilepticus in Richmond,
multivariate analysis (191).
Virginia. Neurology. 1996;46:1029–1035.
One series of patients with NCSE found that patients with 17. Cockerell OC, Walker MC, Sander JWAS, et al. Complex partial sta-
minimal obtundation had a mortality of 7% versus 39% for tus epilepticus: a recurrent problem. J Neurol Neurosurg Psychiatry.
those with deep lethargy or coma (157). Patients with anoxia or 1994;57:835–837.
multiple medical problems, including sepsis, fare poorly, while 18. Tomson T, Svanborg E, Wedlund JE. Nonconvulsive status epilep-
those with strokes, tumors, trauma, infection, alcohol abuse, or ticus: high incidence of complex partial status. Epilepsia. 1986;
other drugs have intermediate results (16). Generally, the most 27:276–285.
favorable etiology is epilepsy itself with some relatively benign 19. Kaplan PW. Behavioral manifestations of nonconvulsive status
precipitant, such as reduced AEDs. epilepticus. Epilepsy Behav. 2002;3:122–139.
In the setting of severe medical or neurologic disease, the 20. Kaplan PW, Tusa RJ. Neurophysiologic and clinical correlations of
epileptic nystagmus. Neurology. 1993;43:2508–2514.
morbidity and mortality of NCSE are substantial (157),
21. Husain AM, Horn GJ, Jacobson MP. Non-convulsive status epilep-
although it is not possible to dissect out that portion of the
ticus: usefulness of clinical features in selecting patients for urgent
long-term harm done by epileptiform discharges, seizures, or EEG. J Neurol Neurosurg Psychiatry. 2003;74:189–191.
SE from the damage caused by the underlying illness 22. Celesia GG, Grigg MM, Ross E. Generalized status myoclonicus in
(32,33,195,202). Little of this morbidity is usually attributed to acute anoxic and toxic-metabolic encephalopathies. Arch Neurol.
the NCSE itself, but complications of treatment can also con- 1988;45:781–784.
tribute to morbidity (237). Overaggressive therapy can result in 23. Knake S, Rosenow F, Vescovi M, et al. Incidence of status epilepti-
hypotension or prolonged mechanical ventilation. C-EEG can cus in adults in Germany: a prospective, population-based study.
help guide treatment of SE and help to avoid undertreatment or Epilepsia. 2001;42:714–718.
overtreatment. 24. Kaplan PW. Nonconvulsive status epilepticus in the emergency
room. Epilepsia. 1996;37:643–650.
25. Guberman A, Cantu-Reyna G, Stuss D, et al. Nonconvulsive
REFERENCES
generalized status epilepticus: clinical features, neuropsychological
1. Temkin O, ed. The Falling Sickness. Baltimore: The Johns Hopkins testing, and long-term follow-up. Neurology. 1986;36:1284–1291.
Press; 1971. 26. Dunne JW, Summers QA, Stewart-Wynne EG. Non-convulsive
2. Shorvon S. The concept of status epilepticus and its history. In: status epilepticus: a prospective study in an adult general hospital.
Shorvon S, ed. Status Epilepticus: Its Clinical Features and Treatment Q J Med. 1987;62:117–126.
in Children and Adults. Cambridge, UK: Cambridge University Press; 27. Stores G, Zaiwalla Z, Styles E, et al. Non-convulsive status epilep-
1994:1–20. ticus. Arch Dis Child. 1995;73:106–111.
3. Trousseau A. Lectures on Clinical Medicine Delivered at the Hôtel 28. Brodtkorb E, Sand T, Kristiansen A, et al. Non-convulsive status
Dieu, Paris. Vol 1, translated by Bazire PV. London: New Sydenham epilepticus in the adult mentally retarded. Seizure. 1993;2:115–123.
Society; 1868. 29. Thomas P, Beaumanoir A, Genton P, et al. “De novo” absence sta-
4. Falret J. De l’état mental des épileptiques. Arch Gén Méd, Fifth tus of late onset: report of 11 cases. Neurology. 1992;42:104–110.
Series. 1860;16:661–679; 1861;17:461–449. 30. Tatum WO, Ross J, Cole AJ. Epileptic pseudodementia. Neurology.
5. Wilks S. Lectures on Diseases of the Nervous System. London: 1998;50:1472–1475.
Churchill; 1878. 31. Chicarro AV, Kanner AM. Psychiatric manifestations of noncon-
6. Goetz CG. Charcot the Clinician: The Tuesday Lessons. New York, vulsive status epilepticus. In: Kaplan PW, Drislane FW, eds.
NY: Raven Press; 1987. Nonconvulsive Status Epilepticus. New York, NY: Demos Medical
7. Gowers WR. Epilepsy and Other Chronic Convulsive Disorders. Publishing; 2009:203–215.
London: Churchill; 1901. 32. Lowenstein D, Aminoff M. Clinical and EEG features of status
8. Zimmer C. Soul made Flesh. The Discovery of the Brain—and How epilepticus in comatose patients. Neurology. 1992;42:100–104.
it Changed the World. New York, NY: Free Press, Simon & Schuster; 33. Drislane FW, Schomer DL. Clinical implications of generalized
2004. electrographic status epilepticus. Epilepsy Res. 1994;19:111–121.
Chapter 29 ■ Nonconvulsive Status Epilepticus 639

34. Pritchard PB III, O’Neal DB. Nonconvulsive status epilepticus fol- 57. Lothman EW, Bertram EH. Epileptogenic effects of status epilep-
lowing metrizamide myelography. Ann Neurol. 1984;16:252–254. ticus. Epilepsia. 1993;34(Suppl 1):59–70.
35. Zak R, Solomon G, Petito F, et al. Baclofen-induced generalized 58. Wasterlain CG, Mazarati AM, Naylor D, et al. Short-term plastic-
nonconvulsive status epilepticus. Ann Neurol. 1994;36:113–114. ity of hippocampal neuropeptides and neuronal circuitry in exper-
36. Panayiotopoulos CP, Agathonikou A, Sharoqi IA, et al. Vigabatrin imental status epilepticus. Epilepsia. 2002;43(Suppl 5):20–29.
aggravates absences and absence status. Neurology. 1997;49:1467. 59. Mazarati AM, Halaszi E, Telegdy G. Anticonvulsive effects of
37. Shinnar S, Berg AT, Treiman DT, et al. Status epilepticus and galanin administered into the central nervous system upon the
tiagabine therapy: review of safety data and epidemiologic com- picrotoxin-kindled seizure syndrome in rats. Brain Res. 1992;
parisons. Epilepsia. 2001;42:372–379. 589:164–166.
38. Feddersen B, Bender A, Arnold S, et al. Aggressive confusional state as 60. Liu H, Mazarati AM, Katsumori H, et al. Substance P is expressed
a clinical manifestation of status epilepticus in MELAS. Neurology. in hippocampal principal neurons during status epilepticus and
2003;61:1149–1150. plays a critical role in the maintenance of status epilepticus. Proc
39. Dalmau J, Rosenfeld MR. Paraneoplastic syndromes of the CNS. Natl Acad Sci USA. 1999;96:5286–5291.
Lancet Neurol. 2008;7:327–340. 61. Hughes JR. Absence seizures: a review of recent reports with new
40. Dalmau J, Gleichman AJ, Hughes EG, et al. Anti-NMDA-receptor concepts. Epilepsy Behav. 2009;15:404–412.
encephalitis: case series and analysis of the effects of antibodies. 62. Gloor P, Fariello RG. Generalized epilepsy: some of its cellular
Lancet Neurol. 2008;7:1091–1098. mechanisms differ from those of focal epilepsy. Trends Neurosci.
41. Kaplan PW, Birbeck G. Lithium-induced confusional states: non- 1988;11:63–68.
convulsive status epilepticus or triphasic encephalopathy? Epilepsia. 63. Jasper HH, Droogleever-Fortuyn J. Experimental studies on the
2006;47:2071–2074. functional anatomy of petit mal epilepsy. Res Publ Ass Nerv Ment
42. Kaplan PK. Nonconvulsive status epilepticus. Semin Neurol. 1996; Dis. 1947;26:272–298.
16:33–40. 64. Williams D. The thalamus and epilepsy. Brain. 1965;88:539–556.
43. Kaplan PW. The EEG of status epilepticus. J Clin Neurophysiol. 65. Tien RD, Felsberg GJ. The hippocampus in status epilepticus:
2006;23:221–229. demonstration of signal intensity and morphologic changes with
44. Tomson T, Lindbom U, Nilsson BY. Nonconvulsive status epilepti- sequential fast spin–echo MR imaging. Radiology. 1995;194:249–256.
cus in adults: thirty-two consecutive patients from a general hos- 66. Wieshmann UC, Woermann FG, Lemieux L, et al. Development of
pital population. Epilepsia. 1992;33:829–835. hippocampal atrophy: a serial magnetic resonance imaging study
45. Granner MA, Lee SI. Nonconvulsive status epilepticus: EEG analy- in a patient who developed epilepsy after generalized status epilep-
sis in a large series. Epilepsia. 1994;35:42–47. ticus. Epilepsia. 1997;38:1238–1241.
46. Kaplan PW. The EEG criteria for NCSE. Epilepsia. 2007;48:39–41. 67. Lothman E. The biochemical basis and pathophysiology of status
47. Fountain NB, Waldman WA. Effects of benzodiazepines on tripha- epilepticus. Neurology. 1990;40(Suppl 2):13–23.
sic waves: implications for nonconvulsive status epilepticus. J Clin 68. Lowenstein DH, Shimosaka S, So YT, et al. The relationship between
Neurophysiol. 2001;18:345–352. electrographic seizure activity and neuronal injury. Epilepsy Res.
48. Treiman DM, Meyers PD, Walton NY, et al. A comparison of four 1991;10:49–54.
treatments for generalized convulsive status epilepticus. Veterans 69. Bertram EH, Lothman EW, Lenn NJ. The hippocampus in experi-
Affairs Status Epilepticus Cooperative Study Group. N Engl J Med. mental chronic epilepsy: a morphometric analysis. Ann Neurol.
1998;339:792–798. 1990;27:43–48.
49. Fagan KJ, Lee SI. Prolonged confusion following convulsions due 70. Gastaut H. Classification of status epilepticus. Adv Neurol. 1983;
to generalized non-convulsive status epilepticus. Neurology. 1990; 34:15–35.
40:1689–1694. 71. Shorvon S. The classification of nonconvulsive status epilepticus.
50. Dong C, Sriram S, Delbeke D, et al. Aphasic or amnestic status epilep- In: Kaplan PW, Drislane FW, eds. Nonconvulsive Status Epilepticus.
ticus detected on PET but not EEG. Epilpesia. 2009;50:251–255. New York, NY: Demos Medical Publishing; 2009:11–22.
51. van Paesschen W, Porke K, Fannes K, et al. Cognitive deficits dur- 72. Chong DJ, Hirsch LJ. Which EEG patterns warrant treatment in
ing status epilepticus and time course of recovery: a case report. the critically ill? Reviewing the evidence for treatment of periodic
Epilepsia. 2007;48:1979–1983. epileptiform discharges and related patterns. J Clin Neurophysiol.
52. Kutluay E, Beattie J, Passaro EA, et al. Diagnostic and localizing 2005;22:79–91.
value of ictal SPECT in patients with nonconvulsive status epilep- 73. Olmos-Garcia de Alba G, Udaeta Mora E, Malagon Valdez J, et al.
ticus. Epilepsy Behav. 2005;6:212–217. Neonatal status epilepticus II: electroencephalographic aspects.
53. Lothman EW, Bertram EH, Bekenstein JW, et al. Self-sustaining Clin Electroencephalogr. 1984;15:197–201.
limbic status epilepticus induced by 'continuous’ hippocampal 74. Mizrahi EM, Kellaway P. Characterization and classification of
stimulation: electrographic and behavioral characteristics. Epilepsy neonatal seizures. Neurology. 1987;37:1837–1844.
Res. 1989;3:107–119. 75. Knauss TA, Carlson CB. Neonatal paroxysmal monorhythmic alpha
54. van Landingham KE, Lothman EW. Self-sustaining limbic status activity. Arch Neurol. 1978;35:104–107.
epilepticus. I. Acute and chronic cerebral metabolic studies: limbic 76. Scher MS. Controversies regarding neonatal seizure recognition.
hypermetabolism and neocortical hypometabolism. Neurology. Epileptic Disord. 2002;4:139–158.
1991;41:1942–1949. 77. Clancy RR, Legido A. The exact ictal and interictal duration of elec-
55. Kapur J. Pathophysiology of nonconvulsive status epilepticus. In: troencephalographic neonatal seizures. Epilepsia. 1987;28:537–541.
Kaplan PW, Drislane FW, eds. Nonconvulsive Status Epilepticus. 78. Dravet C, Bureau M, Oguni H, et al. Severe myoclonic epilepsy in
New York, NY: Demos Medical Publishing; 2009:81–94. infancy: Dravet syndrome. Adv Neurol. 2005;95:71–102.
56. van Landingham KE, Lothman EW. Self-sustaining limbic status 79. Korff CM, Laux L, Kelley KR, et al. Dravet syndrome (severe
epilepticus. II. Role of hippocampal commissures in metabolic myoclonic epilepsy in infancy): a retrospective study of 16 patients.
responses. Neurology. 1991;41:1950–1957. J Child Neurol. 2007;22:185–194.
640 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

80. Ohtahara S, Yamatogi Y. Ohtahara syndrome: with special refer- 100. Iizuka T, Sakai F, Suzuki N, et al. Neuronal hyperexcitability in
ence to it developmental aspects for differentiating from early stroke-like episodes of MELAS syndrome. Neurology. 2002;59:
myoclonic encephalopathy. Epilepsy Res. 2006;70(Suppl 1):58–67. 816–824.
81. Bureau M. ‘Continuous spikes and waves during slow sleep’ 101. Taylor I, Scheffer IE, Berkovic SF. Occipital epilepsies: identifica-
(CSWS): definition of the syndrome. In: Beaumanoir A, Bureau tion of specific and newly recognized syndromes. Brain. 2003;
M, Deonna T, et al., eds. Continuous Spikes and Waves During Slow 126:753–769.
Sleep. London: John Libbey and Co.; 1995:17–26. 102. Wieser HG, Fischer M. Temporal lobe nonconvulsive status
82. Riviello JJ Jr. Status epilepticus in children. In: Drislane FW, ed. epilepticus. In: Kaplan PW, Drislane FW, eds. Nonconvulsive
Status Epilepticus: a Clinical Perspective. Totowa, NJ: Humana Status Epilepticus. New York, NY: Demos Medical Publishing; 2009:
Press; 2005:313–338. 119–137.
83. Hadjiloizou S, Riviello JJ. Epileptic and epileptiform 103. Seshia SS, McLachlan RS. Aura continua. Epilepsia. 2005;46:
encephalopathies. Neurology, eMedicine. WebMD. Available at: 454–455.
http://www.emedicine.com/neuro/topic547.htm. Accessed on 104. Fejerman N, DiBlasi AM. Status epilepticus of benign partial
May 15, 2006. epilepsies in children: report of two cases. Epilepsia. 1987;28:
84. Patry G, Lyagoubi S, Tassinari CA. Subclinical “electrical status 351–355.
epilepticus” induced by sleep in children. A clinical and elec- 105. Colamaria V, Sgro V, Caraballo R, et al. Status epilepticus in
troencephalographic study of six cases. Arch Neurol. 1971;24: benign rolandic epilepsy manifesting as anterior operculum syn-
242–252. drome. Epilepsia. 1991;32:329–334.
85. Tassinari CA, Bureau M, Dravet C, et al. Epilepsy with continuous 106. Scott JS, Masland RL. Occurrence of “continuous symptoms” in
spikes and waves during slow sleep—otherwise described as ESES epilepsy patients. Neurology. 1953;3:297–301.
(epilepsy with electrical status epilepticus during slow sleep). In: 107. Manford M, Shorvon SD. Prolonged sensory or visceral symp-
Roger J, Bureau M, Dravet C, et al., eds. Epileptic Syndromes in toms: an under-diagnosed form of non-convulsive focal (simple
Infancy, Childhood, and Adolescence. 2nd ed. London: John Libbey partial) status epilepticus. J Neurol Neurosurg Psychiatry. 1992;
and Co.; 1992:245–256. 55:714–716.
86. De Negri M. Electrical status epilepticus during sleep (ESES). 108. Blair HT. Temporal lobe or psychomotor status epilepticus. A case
Different clinical syndromes: towards a unifying view? Brain Dev. report. Electroencephalogr Clin Neurophysiol. 1980;48:558–572.
1997;19:447–451. 109. Barry E, Sussman NM, Bosley TM, et al. Ictal blindness and sta-
87. Guilhoto LMFF, Morrell F. Electrophysiological differences between tus epilepticus amauroticus. Epilepsia. 1985;26:577–584.
Landau-Kleffner Syndrome and other conditions showing the 110. Salanova V, Andermann F, Olivier A, et al. Occipital lobe epilepsy:
CSWS electrical pattern. Epilepsia. 1994;35(Suppl 8):126. Abstract. electroclinical manifestations, electrocorticography, cortical
88. Landau WM, Kleffner FR. Syndrome of acquired epileptic aphasia stimulation and outcome in 42 patients treated between 1930
with convulsive disorder in children. Neurology. 1957;7:523–530. and 1991. Brain. 1992;115:1655–1680.
89. Bishop DVM. Age of onset and outcome in acquired aphasia with 111. Sowa MV, Pituck S. Prolonged spontaneous complex visual hal-
convulsive disorder (Landau-Kleffner Syndrome). Dev Med Child lucinations and illusions as ictal phenomena. Epilepsia. 1989;
Neurol. 1985;27:705–712. 30:524–526.
90. Riviello JJ Jr. Nonconvulsive status epilepticus in children. In: 112. Young G, Blume WT. Painful epileptic seizures. Brain. 1983;106:
Kaplan PW, Drislane FW, eds. Nonconvulsive Status Epilepticus. 537–554.
New York, NY: Demos Medical Publishing; 2009:227–237. 113. Peppercorn M, Herzog AG. The spectrum of abdominal epilepsy
91. Lerman P, Lerman-Sagie T, Kivity S. Effect of early corticosteroid in adults. Am J Gastroenterol. 1989;84:1294–1296.
therapy for Landau-Kleffner Syndrome. Dev Med Child Neurol. 114. Eschle D, Siegel AM, Wieser H-G. Epilepsy with severe abdomi-
1991;33:257–260. nal pain. Mayo Clin Proc. 2002;77:1358–1360.
92. Shinnar S, Rapin I, Arnold S, et al. Language regression in child- 115. Kikumoto K, Yoshinaga H, Kobayashi K, et al. Complex partial
hood. Pediatr Neurol. 2001;24:183–189. status epilepticus in children with epilepsy. Brain Dev. 2009;31:
93. McVicar KA, Shinnar S. Landau-Kleffner syndrome, electrical sta- 148–157.
tus epilepticus in slow wave sleep, and language regression in chil- 116. Panayiotopoulos CP. Autonomic seizures and autonomic status
dren. Ment Retard Dev Disabil Res Rev. 2004;10:144–149. epilepticus peculiar to childhood: diagnosis and management.
94. Jayakar PB, Seshia SS. Electrical status epilepticus during slow- Epilepsy Behav. 2004;5:286–295.
wave sleep: a review. J Clin Neurophysiol. 1991;8:299–311. 117. Engel J, Ludwig BI, Fetell M. Prolonged partial complex status
95. Kramer U, Nevo Y, Neufeld MY, et al. Epidemiology of epilepsy in epilepticus: EEG and behavioral observations. Neurology. 1978;
childhood: a cohort of 440 consecutive patients. Pediatr Neurol. 28:863–869.
1998;18:46–50. 118. Treiman DM, Delgado-Escueta AV. Complex partial status epilep-
96. Inoue Y, Fujiwara T, Matsuda K, et al. Ring chromosome 20 and ticus. In: Delgado-Escueta AV, Wasterlain CG, Treiman DM, et al.,
nonconvulsive status epilepticus. A new syndrome. Brain. 1997;120: eds. Status Epilepticus (Advances in Neurology, Vol 34). New York,
939–953. NY: Raven Press; 1983:69–81.
97. Devinsky O, Kelley K, Porter RJ, et al. Clinical and electroen- 119. Butler CR, Graham KS, Hodges JR, et al. The syndrome of tran-
cephalographic features of simple partial seizures. Neurology. 1988; sient epileptic amnesia. Ann Neurol. 2007;61:587–598.
38:1347–1352. 120. Palmini AL, Gloor P, Jones-Gotman M. Pure amnestic seizures in
98. DeLorenzo RJ, Waterhouse EJ, Towne AR, et al. Persistent noncon- temporal lobe epilepsy. Definition, clinical symptomatology and
vulsive status epilepticus after the control of convulsive status epilep- functional anatomical considerations. Brain. 199;115:749–769.
ticus. Epilepsia. 1998;39:833–840. 121. Vuilleumier P, Despland PA, Regli F. Failure to recall (but not to
99. Friedman DE, Schevon C, Emerson RG, et al. Cyclic electrographic remember): pure transient amnesia during nonconvulsive status
seizures in critically ill patients. Epilepsia. 2008;49:281–287. epilepticus. Neurology. 1996;46:1036–1039.
Chapter 29 ■ Nonconvulsive Status Epilepticus 641

122. Profitlich T, Hoppe C, Reuber M, et al. Ictal neuropsychological 144. Shorvon S. Definition, classification and frequency of status
findings in focal nonconvulsive status epilepticus. Epilepsy Behav. epilepticus. In: Shorvon S, ed. Status Epilepticus: Its Clinical
2008;12:269–275. Features and Treatment in Children and Adults. Cambridge, UK:
123. Hamilton NG, Matthews T. Aphasia: the sole manifestation of Cambridge University Press; 1994:21–33.
focal status epilepticus. Neurology. 1979;29:745–748. 145. Williamson PD, Spencer DD, Spencer SS, et al. Complex partial
124. Racy A, Osborn MA, Vern BA, et al. Epileptic aphasia. First onset status epilepticus: a depth-electrode study. Ann Neurol. 1985;18:
of prolonged monosymptomatic status epilepticus in adults. 647–654.
Arch Neurol. 1980;37:419–422. 146. Rohr-le-Floch J, Gauthier G, Beaumanoir A. États confusionelles
125. Gilmore RL, Heilman KM. Speech arrest in partial seizures: d’origine épileptique: interêt de l’EEG fait en urgence. Rev Neurol
evidence of an associated language disorder. Neurology. 1981;31: (Paris). 1988;144:425–436.
1016–1019. 147. Markand ON, Wheeler GL, Pollack SL. Complex partial status
126. Knight RT, Cooper J. Status epilepticus manifesting as reversible epilepticus (psychomotor status). Neurology. 1978;28:189–196.
Wernicke’s aphasia. Epilepsia. 1986;27:301–304. 148. Mayeux R, Lueders H. Complex partial status epilepticus: case
127. Kirshner HS, Hughes T, Fakhoury T, et al. Aphasia secondary to report and proposal for diagnostic criteria. Neurology. 1978;28:
partial status epilepticus of the basal temporal language area. 957–961.
Neurology. 1995;45:1616–1618. 149. Salanova V, Morris HH, Van Ness P, et al. Frontal lobe seizures:
128. Benatar M. Ictal aphasia. Epilepsy Behav. 2002;3:413–419. electroclinical syndromes. Epilepsia. 1995;36:16–24.
129. Schomer DL. Focal status epilepticus and epilepsia partialis 150. Williamson PD, French JA, Thadani VM, et al. Characteristics of
continua in adults and children. Epilepsia. 1993;34(Suppl 1): medial temporal lobe epilepsy: II. Interictal and ictal scalp elec-
S29–S36. troencephalography, neuropsychological testing, neuroimaging,
130. Grand’Maison F, Reiher J, Leduc CP. Retrospective inventory of surgical results, and pathology. Ann Neurol. 1993;34:781–787.
EEG abnormalities in partial status epilepticus. Electroencephalogr 151. Foldvary N, Lee N, Thwaites G, et al. Clinical and electrographic
Clin Neurophysiol. 1991;79:264–270. manifestations of lesional neocortical temporal lobe epilepsy.
131. Hirsch LJ, Emerson RG, Pedley TA. Prolonged “postictal” aphasia: Neurology. 1997;49:757–763.
demonstration of persistent ictal activity with intracranial elec- 152. Nowack WJ, Shaikh IA. Progression of electroclinical changes in
trodes. Neurology. 2001;56:134–136. complex partial status epilepticus: filling in the blanks. Clin
132. Jobst BC, Roberts DW, Williamson PD. Occipital lobe noncon- Electroencephalogr. 1999;30:5–8.
vulsive status epilepticus. In: Kaplan PW, Drislane FW, eds. 153. Wieser HG. Temporal lobe or psychomotor status epilepticus: a
Nonconvulsive Status Epilepticus. New York, NY: Demos Medical case report. Electroencephalogr Clin Neurophysiol. 1980;48:558–572.
Publishing; 2009:139–150. 154. Williamson PD, Boon PA, Thadani VM, et al. Parietal lobe
133. Drislane FW, Blum AS, Schomer DL. Focal status epilepticus: clin- epilepsy: diagnostic considerations and results of surgery. Ann
ical features and significance of different EEG patterns. Epilepsia. Neurol. 1992;31:193–201.
1999;40:1254–1260. 155. Gloor P, Olivier A, Quesney LF, et al. The role of the limbic sys-
134. Ballenger CE, King DW, Gallagher BB. Partial complex status tem in experiential phenomena of temporal lobe epilepsy. Ann
epilepticus. Neurology. 1983;33:1545–1552. Neurol. 1982;12:129–144.
135. Williamson PD. Complex partial status epilepticus. In: Engel JE Jr, 156. Kudo T, Sato K, Yagi K, et al. Can absence status epilepticus be of
Pedley T, eds. Epilepsy. A Comprehensive Text. 2nd ed. Philadelphia, frontal lobe origin? Acta Neurol Scand. 1995;92:472–477.
PA: Lippincott Williams & Wilkins; 2008:677–692. 157. Shneker BF, Fountain NB. Assessment of acute morbidity and
136. Kaplan PW. Assessing the outcomes in patients with nonconvulsive mortality in nonconvulsive status epilepticus. Neurology. 2003;
status epilepticus: nonconvulsive status epilepticus is underdiag- 61:1066–1073.
nosed, potentially overtreated, and confounded by comorbidity. J 158. Shorvon S, Baulac M, Cross H, et al. The drug treatment of sta-
Clin Neurophysiol. 1999;16:341–352. tus epilepticus in Europe: consensus document from a workshop
137. Mikati MA, Lee WL, DeLong GR. Protracted epileptiform at the first London Colloquium on Status Epilepticus. Epilepsia.
encephalopathy: an unusual form of partial complex status epilep- 2008;49:1277–1284.
ticus. Epilepsia. 1985;25:563–571. 159. Meierkord H, Holtkamp M. Non-convulsive status epilepticus in
138. Niedermeyer E, Fineyre F, Riley T, et al. Absence status (petit mal adults: clinical forms and treatment. Lancet Neurol. 2007;6:329–339.
status) with focal characteristics. Arch Neurol. 1979;36:417–421. 160. Towne AR, Pellock JM, Ko D, et al. Determinants of mortality in
139. Thomas P, Zifkin B, Migneco O, et al. Nonconvulsive status status epilepticus. Epilepsia. 1994;35:27–34.
epilepticus of frontal origin. Neurology. 1999;52:1174–1183. 161. McBride MC, Dooling EC, Oppenheimer EY. Complex partial sta-
140. Belafsky MA, Carwille S, Miller P, et al. Prolonged epileptic twi- tus epilepticus in young children. Ann Neurol. 1981;9:526–530.
light states: continuous recordings with nasopharyngeal elec- 162. Berg AT, Shinnar S. Do seizures beget seizures? An assessment of the
trodes and videotape analysis. Neurology. 1978;28:239–245. clinical evidence in humans. J Clin Neurophysiol. 1997;14:102–110.
141. Commission on Epidemiology and Prognosis, International 163. Schwab RS. A case of status epilepticus in petit mal.
League Against Epilepsy. Guidelines for epidemiologic studies on Electroencephalogr Clin Neurophysiol. 1953;5:441–442.
epilepsy. Epilepsia. 1993;34:592–596. 164. Niedermeyer E, Khalifeh R. Petit-mal status (“spike-wave stu-
142. Shorvon S. Clinical forms of status epilepticus. In: Shorvon S, ed. por”). An electro-clinical appraisal. Epilepsia. 1965;6:250–262.
Status Epilepticus: Its Clinical Features and Treatment in Children 165. Wheless JW. Acute management of seizures in the syndromes of
and Adults. Cambridge, UK: Cambridge University Press; 1994: idiopathic generalized epilepsies. Epilepsia. 2003;44(Suppl 2):
34–138. 22–26.
143. Krumholz A, Sung GY, Fisher RS, et al. Complex partial status 166. Agathonikou A, Panayiotopoulos CP, Giannakodimos S, et al.
epilepticus accompanied by serious morbidity and mortality. Typical absence status in adults: diagnostic and syndromic con-
Neurology. 1995;45:1499–1504. siderations. Epilepsia. 1998;39:1265–1276.
642 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

167. Shorvon S, Walker M. Status epilepticus in idiopathic generalized 190. Treiman DM, Walton NY, Kendrick C. A progressive sequence of
epilepsy. Epilepsia. 2005;46:73–79. electroencephalographic changes during generalized convulsive
168. Thomas P, Snead OC. Absence status epilepticus. In: Engel J, status epilepticus. Epilepsy Res. 1990;5:49–60.
Pedley TA, eds. Epilepsy: A Comprehensive Textbook. 2nd ed. 191. Young GB, Jordan KG, Doig GS. An assessment of nonconvulsive
Philadelphia, PA: Lippincott-Raven Publishers; 2008:693–703. seizures in the intensive care unit using continuous EEG moni-
169. Genton P, Ferlazzo E, Thomas P. Absence status epilepsy: delin- toring: an investigation of variables associated with mortality.
eation of a distinct idiopathic generalized epilepsy syndrome. Neurology. 1996;47:83–89.
Epilepsia. 2008;49:642–649. 192. Barry E, Hauser W. Status epilepticus: the interaction of epilepsy
170. Kimura S, Kobayashi T. Two patients with juvenile myoclonic and acute brain disease. Neurology. 1993;43:1473–1478.
epilepsy and nonconvulsive status epilepticus. Epilepsia. 1996;37: 193. Krishnamurthy KB, Drislane FW. Depth of electroencephalo-
275–279. gram suppression and outcome in pentobarbital coma for refrac-
171. Osorio I, Reed RC, Peltzer JN. Refractory idiopathic status tory status epilepticus. Epilepsia. 1999;40:759–762.
epilepticus: a probable paradoxical effect of phenytoin and car- 194. Claassen J, Hirsch LJ, Emerson RG, et al. Continuous EEG mon-
bamazepine. Epilepsia. 2000;41:887–894. itoring and midazolam infusion for refractory nonconvulsive sta-
172. Andermann F, Robb JP. Absence status: a reappraisal following tus epilepticus. Neurology. 2001;57:1036–1042.
review of thirty-eight patients. Epilepsia. 1972;13:177–187. 195. So EL, Ruggles KH, Ahmann PA, et al. Clinical significance and
173. Niedermeyer E, Ribeiro M. Considerations of nonconvulsive sta- outcome of subclinical status epilepticus in adults. J Epilepsy.
tus epilepticus. Clin EEG. 2000;31:192–195. 1995;8:11–15.
174. Baykan B, Gokyigit A, Gurses C, et al. Recurrent absence status 196. Walker M. Status epilepticus: an evidence based guide. Br Med J.
epilepticus: clinical and EEG characteristics. Seizure. 2002;11: 2005;331:673–677.
310–319. 197. Holtkamp M, Othman J, Buchheim K, et al. Diagnosis of psy-
175. Berkovic SF, Bladin PF. Absence status in adults. Clin Exp Neurol. chogenic nonepileptic status epilepticus in the emergency set-
1983;19:198–207. ting. Neurology. 2006;66:1727–1729.
176. Treiman DM. Electroclinical features of status epilepticus. J Clin 198. Wilner AN, Bream PR. Status epilepticus and pseudostatus
Neurophysiol. 1995;12:343–362. epilepticus. Seizure. 1993;2:257–260.
177. Hovinga CA, Chicella MF, Rose DF, et al. Use of intravenous val- 199. Howell SJ, Owen L, Chadwick DW. Pseudostatus epilepticus. Q J
proate in three pediatric patients with nonconvulsive or convul- Med. 1989;71:507–519.
sive status epilepticus. Ann Pharmacother. 1999;33:579–584. 200. Towne AR, Waterhouse EJ, Boggs JG, et al. Prevalence of noncon-
178. Berkovic SF, Andermann F, Guberman A, et al. Valproate prevents vulsive status epilepticus in comatose patients. Neurology.
the recurrence of absence status. Neurology. 1989;39:1294–1297. 2000;54:340–345.
179. Fernandez-Torre JL. De novo absence status of late onset following 201. Vespa PM, Nuwer MR, Nenov V, et al. Increased incidence and
withdrawal of lorazepam: a case report. Seizure. 2001;10:433–437. impact of nonconvulsive and convulsive seizures after traumatic
180. D’Agostino MD, Andermann F, Dubeau F, et al. Exceptionally brain injury as detected by continuous electroencephalographic
long absence status: multifactorial etiology, drug interactions and monitoring. J Neurosurg. 1999;91:750–760.
complications. Epileptic Disord. 1999;1:229–232. 202. Privitera M, Hoffman M, Moore JL, et al. EEG detection of non-
181. Nolan M, Bergazar M, Chu B, et al. Clinical and neurophysiologic tonic-clonic status epilepticus in patients with altered conscious-
spectrum associated with atypical absence seizures in children ness. Epilepsy Res. 1994;18:155–166.
with intractable epilepsy. J Child Neurol. 2005;20:404–410. 203. Claassen J, Mayer SA, Kowalski RG, et al. Detection of electro-
182. Roger J, Lob H, Tassinari CA. Status epilepticus. In: Vinken P, Bruyn graphic seizures with continuous EEG monitoring in critically ill
G, eds. Handbook of Clinical Neurology, Vol 15, The Epilepsies. patients. Neurology. 2004;62:1743–1748.
Amsterdam: North Holland Publishing Co.; 1974:145–188. 204. Jirsch J, Hirsch LJ. Nonconvulsive seizures: developing a rational
183. Cortez MA, McKerlie C, Snead OC. A model of atypical absence approach to the diagnosis and management in the critically ill
seizures. EEG, pharmacology, and developmental characteriza- population. Clin Neurophysiol. 2007;118:1660–1670.
tion. Neurology. 2001;56:341–349. 205. Vespa PM, O’Phelan K, Shah M, et al. Acute seizures after intrac-
184. Holmes GL, McKeever M, Adamson M. Absence seizures in chil- erebral hemorrhage: a factor in progressive midline shift and
dren: clinical and electroencephalographic features. Ann Neurol. outcome. Neurology. 2003;60:1441–1446.
1987;21:268–273. 206. Claassen J, Jette N, Chum F, et al. Electrographic seizures and
185. Matsumoto A, Kumagai T, Miura K, et al. Epilepsy in Angelman periodic discharges after intracerebral hemorrhage. Neurology.
syndrome associated with chromosome 15q deletion. Epilepsia. 2007;69:1356–1365.
1992;33:1083–1090. 207. Pandian JD, Cascino GD, So EL, et al. Digital video-electroen-
186. Gastaut H, Roger J, Ouahchi S, et al. An electroclinical study of cephalographic monitoring in the neurological–neurosurgical
generalized epileptic seizures of tonic expression. Epilepsia. 1963;4: intensive care unit: clinical features and outcome. Arch Neurol.
15–44. 2004;61:1090–1094.
187. Perez Velazquez JL, Huo JZ, Dominguez LG, et al. Typical versus 208. Jette N, Claassen J, Emerson RG, et al. Frequency and predictors
atypical absence seizures: network mechanisms of the spread of of nonconvulsive seizures during continuous electroencephalo-
paroxysms. Epilepsia. 2007;48:1585–1593. graphic monitoring in critically ill children. Arch Neurol. 2006;
188. Bancaud J, Talairach J, Morel P, et al. “Generalized” epileptic 63:1750–1755.
seizures elicited by electrical stimulation of the frontal lobe in 209. Jirsch J, Hirsch LJ. Nonconvulsive status epilepticus in critically
man. Electroencephalogr Clin Neurophysiol. 1974;37:275–282. ill and comatose patients in the intensive care unit. In: Kaplan
189. Abend NS, Dlugos DJ. Nonconvulsive status epilepticus in a PW, Drislane FW, eds. Nonconvulsive Status Epilepticus. New
pediatric intensive care unit. Pediatr Neurol. 2007;37:165–170. York, NY: Demos Medical Publishing; 2009:175–186.
Chapter 29 ■ Nonconvulsive Status Epilepticus 643

210. Nei M, Lee JM, Shanker VL, et al. The EEG and prognosis in sta- 230. Wilson SB. Algorithm architectures for patient dependent seizure
tus epilepticus. Epilepsia. 1999;40:157–163. detection. Clin Neuophysiol. 2006;117:1204–1216.
211. Hirsch LJ, Brenner RP, Drislane FW, et al. The ACNS subcommit- 231. Wilson SB. A neural network method for automatic and incre-
tee on research terminology for continuous EEG monitoring: pro- mental learning applied to patient-dependent seizure detection.
posed standardized terminology for rhythmic and periodic EEG Clin Neurophysiol. 2005;116:1785–1795.
patterns encountered in critically ill patients. J Clin Neurophysiol. 232. Qu H, Gotman J. Improvement in seizure detection performance
2005;22:128–135. by automatic adaptation to the EEG of each patient.
212. Scheuer ML. Continuous EEG monitoring in the intensive care Electroencephalogr Clin Neurophysiol. 1993;86:79–87.
unit. Epilepsia. 2002;43(Suppl 3):114–127. 233. Abend NS, Dlugos D, Herman S. Neonatal seizure detection
213. Brenner RP. Is it status? Epilepsia. 2002;43(Suppl 3):103–113. using multichannel display of envelope trend. Epilepsia. 2008;
214. Pohlmann-Eden B, Hoch DB, Cochius JI, et al. Periodic lateralized 49:349–352.
epileptiform discharges—a critical review. J Clin Neurophysiol. 234. Hirsch LJ. Continuous EEG monitoring in the intensive care
1996;13:519–530. unit: an overview. J Clin Neurophysiol. 2004;21:332–340.
215. Reiher J, Rivest J, Grand’Maison F, et al. Periodic lateralized epilep- 235. Bleck TP. Intensive care unit management of patients with status
tiform discharges with transitional rhythmic discharges: associa- epilepticus. Epilepsia. 2007;48(Suppl 8);59–60.
tion with seizures. Electroencephalogr Clin Neurophysiol. 1991;78: 236. Walker M, Cross H, Smith S, et al. Nonconvulsive status epilepti-
12–17. cus: epilepsy research foundation workshop reports. Epileptic
216. Brenner RP, Schaul N. Periodic EEG patterns: classification, clin- Disord. 2005;7:253–296.
ical correlation, and pathophysiology. J Clin Neurophysiol. 1990;7: 237. Litt B, Wityk RJ, Hertz SH, et al. Nonconvulsive status epilepticus
249–267. in the critically ill elderly. Epilepsia. 1998;39:1194–1202.
217. Treiman DM. Status epilepticus. Baillieres Clin Neurol. 1996;5: 238. Krishnamurthy KB, Drislane FW. Relapse and survival after bar-
821–839. biturate anesthetic treatment of refractory status epilepticus.
218. Krumholz A. Epidemiology and evidence for morbidity of noncon- Epilepsia. 1996;37:863–867.
vulsive status epilepticus. J Clin Neurophysiol. 1999;16:314–322. 239. Walker MC. Diagnosis and treatment of nonconvulsive status
219. Husain AM, Mebust KA, Radtke RA. Generalized periodic epilepticus. CNS Drugs. 2001;15:931–939.
epileptiform discharges: etiologies, relationship to status epilep- 240. Claassen J, Hirsch LJ, Emerson RG, et al. Treatment of refractory
ticus, and prognosis. J Clin Neurophysiol. 1999;16:51–58. status epilepticus with pentobarbital, propofol, or midazolam: a
220. Hirsch LJ, Claassen J, Mayer SA, et al. Stimulus-induced rhyth- systematic review. Epilepsia. 2002;43:146–153.
mic, periodic, or ictal discharges (SIRPIDs): a common EEG 241. Drislane FW, Lopez MR, Blum AS, et al. Detection and treatment
phenomenon in the critically ill. Epilepsia. 2004;45:109–123. of refractory status epilepticus in the intensive care unit. J Clin
221. Hirsch LJ, Pang T, Claassen J, et al. Focal motor seizures induced Neurophysiol. 2008;25:181–186.
by altering stimuli in critically ill patients. Epilepsia. 2008;49: 242. Drislane FW. Evidence against permanent neurologic damage
968–973. from nonconvulsive status epilepticus. J Clin Neurophysiol. 1999;16:
222. Fisch BJ, Klass DW. The diagnostic specificity of triphasic wave 323–331.
patterns. Electroencephalogr Clin Neurophysiol. 1988;70:1–8. 243. Lee SI. Nonconvulsive status epilepticus. Ictal confusion in later
223. Sheridan PH, Sato S. Triphasic waves of metabolic encephalopathy life. Arch Neurol. 1985;42:778–781.
versus spike-wave stupor. J Neurol Neurosurg Psychiatry. 1986;49: 244. Scholtes FB, Renier WO, Meinardi H. Non-convulsive status
108–109. epilepticus: causes, treatment, and outcome in 65 patients. J Neurol
224. Boulanger JM, Deacon C, Lecuyer D, et al. Triphasic waves versus Neurosurg Psychiatry. 1996;61:93–95.
nonconvulsive status epilepticus: EEG distinction. Can J Neurol 245. Adachi N, Kanemoto K, Muramatsu R, et al. Intellectual
Sci. 2006;33:175–180. prognosis of status epilepticus in adult patients: analysis with
225. Jordan KG. Continuous EEG and evoked potential monitoring in Wechsler Adult Intelligence Scale-Revised. Epilepsia. 2005;46:
the neuroscience intensive care unit. J Clin Neurophysiol. 1993; 1502–1509.
10:445–475. 246. Nevander G, Ingvar M, Auer R, et al. Status epilepticus in well-
226. Scheuer ML, Wilson SB. Data analysis for continuous EEG mon- oxygenated rats causes neuronal necrosis. Ann Neurol. 1985;18:
itoring in the ICU: seeing the forest and the trees. J Clin 281–290.
Neurophysiol. 2004;21:353–378. 247. Vespa PM, Miller C, McArthur D, et al. Nonconvulsive electro-
227. Agarwal R, Gotman J. Long-term EEG compression for inten- graphic seizures after traumatic brain injury result in a delayed,
sive-care settings. IEEE Eng Med Biol Mag. 2001;20:23–29. prolonged increase in intracranial pressure and metabolic crisis.
228. Gotman J. Automatic seizure detection: improvements and eval- Crit Care Med. 2007;35:2830–2836.
uation. Electroencephalogr Clin Neurophysiol. 1990;76:317–324. 248. Ruegg SJ, Dichter MA. Diagnosis and treatment of nonconvul-
229. Gotman J. Automatic detection of seizures and spikes. J Clin sive status epilepticus in an intensive care unit setting. Curr Treat
Neurophysiol. 1999;16:130–140. Options Neurol. 2003;5:93–110.
CHAPTER

Anticipating Seizures Based on EEG


FERNANDO H. LOPES DA SILVA AND STILIYAN KALITZIN 30
INTRODUCTION dynamical systems. In the third phase, since about 2000, more
sophisticated studies are appearing particularly with respect to
In this chapter, we deal with the possibility of anticipating theoretical models of seizure dynamics, to statistical methods of
seizures based on the analysis of electroencephalogram (EEG) evaluating clinical application studies, and to proposals of new
recordings. Nonetheless it is interesting to note that there exist strategies. In any case the development of reliable methods to
many reports stating that epileptic patients may feel an anticipate seizures is the aim of a wide range of researchers hav-
approaching seizure (1), but most of these are based on rela- ing in view the desideratum of being able to control the occur-
tively small patient populations. It is relevant to assess this issue rence of seizures, ideally avoiding their onset.
critically since it may be important in order to determine the Considering the relevance of the mathematical approaches,
value of any automatic method based on EEG recordings. however, we start by presenting a sketch of the analytical tools
Schulze-Bonhage et al. (2) carried out a comprehensive analysis used in this context. Before entering into the main question of
of subjective seizure anticipation reported by 500 consecutively anticipation of epileptic seizures, we consider how these mathe-
recruited outpatients with focal and generalized epilepsy. The matical analyses have been applied to characterize the dynamics
patients were asked to report any premonitory symptoms, with of the neuronal networks during seizures. This is useful to under-
respect to, at least 30 minutes prior to a seizure. No auras were stand better what happens in the dynamics of the corresponding
included. These authors found that 6.2% of patients were able EEG signals in the course of the transition from the interictal to
to report anticipating seizures. The median estimated time the ictal or seizure state. We finish this chapter with the discus-
interval between occurrence of premonitory symptoms and sion of alternative strategies specially in relation to theoretical
seizure onset was 90 minutes. More recently, Scaramelli et al. modeling of seizure dynamics. In this context, it is interesting to
(3) reported a study where the existence of premonitory or pro- note that the question of predicting seizures is related to similar
dromal signs (PS) was investigated in a randomly selected pop- problems in other fields of science. Recently, Scheffer et al. (5)
ulation of 100 adult epileptic patients both with focal and reviewed this issue in a general way and concluded that there may
generalized epilepsies. Using a semistructured protocol and exist generic early-warning signals that may indicate for a wide
personal interviews they found evidence for PS (behavioral, class of systems with multiple attractors if a critical threshold is
cognitive and mood changes, excluding auras) in 39% of approaching, such as in the case of an approaching epileptic
patients, mostly in those having complex partial and general- seizure. The authors assume in this chapter, however, that transi-
ized tonic-clonic seizures. In this study PS were reported to tions occur due to crossing that threshold. In reality internal or
have an insidious onset and a duration ranging from 30 min- external fluctuations, or “noise,” can trigger a transition even
utes to several hours prior to seizure onset. We should note that away from the critical threshold in a multi-attractor system.
these results are certainly relevant although it is not yet possible Another aspect that should be noted is that in order to define
to estimate the corresponding sensitivity and specificity with effectively a precursor, or early-warning signal, it is necessary to
respect to the pre-ictal period. The fact that no EEG recordings have a specific model of critical transitions.
were generally carried out during the period where the patients
reported PS raises also the question as to whether EEG signal
changes might be associated with the presence of PS. In assess- CHARACTERIZATION OF EEG SIGNALS
ing the clinical significance of EEG automatic methods of DURING EPILEPTIC SEIZURES AS
analysis, it is important to keep these results on subjective expe- REALIZATIONS OF LOW-DIMENSION
riences of epileptic patients in mind. NONLINEAR COMPLEX SYSTEMS
In their thoughtful account of the “state of seizure predic-
tion,” Zaveri et al. (4) considered that three phases can be distin- In the last decade, the interest for the characterization of epilep-
guished in this research field in the course of the last decades. In tic seizures based on EEG recordings using analytical tools
the first phase in the seventies, a variety of attempts were tried, derived from the mathematical theory of nonlinear complex
from measuring epileptiform spike rates to estimating time- systems has grown steadily. A large number of approaches have
varying EEG spectral features. In the second phase, particularly been tried out with this aim in view.
in the late eighties and beginning nineties, the question of antic- A very abbreviated explanation of the corresponding basic
ipating epileptic seizures was approached within the frame of concepts is appropriate here. Traditionally EEG analysis employs
mathematical methods derived from the theory of nonlinear mathematical tools used in the analysis of linear systems as

645
646 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

discussed in detail in Chapter 54. More recently nonlinear meth- a sudden decrease in the value of the largest Lyapunov expo-
ods of analysis of time series have received special attention, par- nent, an indication of deterministic chaos, at seizure onset in
ticularly since Takens (6) proposed the mathematical basis for subdural electrocorticogram (ECoG) recordings in seizures
the reconstruction of dynamical systems using time series and with a temporal lobe epilepsy (TLE) onset, initially in those
Grassberger and Procaccia (7,8) introduced methods to estimate channels close to the epileptogenic focal area, followed by other
invariants of such systems from a single time series, such as the channels. In an animal model of epilepsy (limbic kindling in
correlation dimension (D2), Lyapunov exponents, and entropy (for the rat) with a focal onset in the hippocampus, Pijn et al. (12)
mathematical definitions see, for example, Ref. 9). The most demonstrated a very clear decrease of the value of the correla-
important aspects of nonlinear time series analysis are described tion dimension D2, occurring at seizure onset. The drop in the
in Grassberger et al. (10), and a tutorial review of nonlinear value of D2 was most pronounced in the “focal area.” The
dynamical EEG analysis is that of Pritchard and Duke (9). The process of seizure spreading could be followed by showing the
basic principle is to reconstruct the state space of the system progressive decrease of D2 for signals recorded from various
using delay-vectors derived from the EEG time series. A funda- brain areas at increasing distances from the “focal area.”
mental concept is that the dynamics of a system must be studied In this experimental animal study (12) and in a subsequent
in a phase space; a point in this space characterizes the state of study in patients with temporal lobe epilepsy (21,22) we found
the system at any moment of time. The phase space is a recon- that most EEG signals recorded during interictal periods could
struction procedure that starts from raw data and builds vectors not be distinguished from random time series, while most
by iterations of a time delay (delay-vectors). In this way invari- epochs recorded during seizures were clearly distinguishable
ants of the signal dynamics, namely the correlation integral and from random noise. These results demonstrate that EEG signals
the correlation dimension D2, or the Lyapunov exponents can be during epileptic seizures have different dynamics with respect
estimated to characterize the signal dynamics. to the interictal EEGs and are characterized by low-correlation
Given an EEG signal one can put in evidence nonlinear dimension. This implies that the dynamical state of the neu-
deterministic dynamics, by testing whether the null hypothesis ronal networks underlying an epileptic seizure can be described
that the signal is a realization of a Gaussian random process can by a relatively small number of degrees of freedom, in contrast
be rejected. This can be tested in various ways; one of the most with the interictal EEG. Whether this corresponds to a state
common tests consists in making a comparison of the invari- characterized by a chaotic attractor cannot be concluded with-
ants of the signal, such as the correlation dimension D2 or the out further evidence.
Lyapunov exponents, with those of surrogate signals (random- The estimation of correlation dimension D2 in the cases
ized signals) as proposed by Theiler et al. (11), Pijn et al. (12), reported by Pijn et al. (21) yields excellent results in terms of
Prichard and Theiler (13), and Schreiber and Schmitz (14). This delineating the epileptogenic zone in cases of TLE, as well as
methodology has been applied by several authors to the analy- tracing ictal spread throughout the brain. These studies, how-
sis of EEG signals recorded during different behavioral states, ever, did not reveal any value of D2 able to predict reliably the
including sleep (15) and alpha rhythms (16), by computing the onslaught of a complex partial seizure. In fact these findings
correlation dimension (D2) of EEG epochs and the correspon- indicate that a behavior reminiscent of a low-dimensional
ding surrogates. chaotic attractor becomes manifest only well into the seizure.
With respect to the study of EEG signals recorded from the In any case we should stress that these kind of measurements
brain of epileptic patients, the basic question is whether differ- can be influenced by a number of factors that have to be taken
ences in the dynamics of neuronal networks, in terms of invari- into account in interpreting the results, namely the following:
ants such as the correlation dimension (D2) and/or Lyapunov the activity of the reference electrode may be of considerable
exponents, could be found in these signals during the period influence on estimates of D2; bipolar montage reconstruction is
preceding an epileptic seizure, with respect to reference periods the method of choice, whereas in case of monopolar recordings,
far from seizure occurrence. one should always consider the possibility that extraneous
This concept stems from previous studies where it was “noisy” contamination through the reference electrode may
shown that EEG signals recorded during epileptic seizures have influence the estimation of D2. In addition the presence of
different properties in terms of nonlinear dynamics compared epileptiform spikes in the EEG signals, recorded interictally or
to normal ongoing EEG activity. Hallmarks of such studies were pre-ictally, may yield a very low D2, close to 0. EEG signals dis-
those of Babloyanz and Destexhe (17) who estimated nonlinear playing seizure activity always yielded D2 values different from
dynamical parameters of absence seizures, namely the correla- surrogate signals. The differences, however, varied from being
tion dimension and the largest Lyapunov exponent, and subtle to very large. Indeed there is usually a marked degree of
reported a considerably lower value for the seizure periods in nonstationarity in most EEG signals during the course of a
comparison with normal EEG, what these authors interpreted seizure. Therefore, it is inappropriate to allocate a single value
as indicating that the EEG during absence seizures has low- for D2 for a whole seizure and thus only an analysis that takes
dimensional chaotic nature. These authors claimed the “exis- into account the evolution of D2 in the course of time might
tence of a chaotic attractor” during the absence seizure. reveal a correct picture. Another limiting factor in the nonlin-
Although this claim has been rejected by controlled studies per- ear EEG analysis is related to the nature of the acquisition tech-
formed later, this study has set the tone for many later investi- nique. EEG signals are amplified and band-pass filtered during
gations. Along the same line Iasemidis et al. (18–20) described recording, and this may influence the values of calculated D2 or
Chapter 30 ■ Anticipating Seizures Based on EEG 647

the Laypunov exponents. As those calculations are based on synchronization. Prediction times of 1 to 6 seconds were found
defining neighborhoods in the reconstructed phase space of the in several seizures from five patients. These methods, however,
system, excluding low-frequency variations of the signals may were not applied in general in view of the short prediction win-
create spurious “closeness” of states and vice versa, taking a dow and the uncertainty about the precise moment of starting
broader band signal may separate states in the phase space of of these seizures. These techniques were not tested thoroughly
the system. in a general clinical setting.
Although many investigators have considered the possibility Efforts to solve the problem of anticipation in a more gen-
that dynamical processes in the brain considered in general may eral context got a considerable impulse with the application of
be of a low-dimensional chaotic nature (17,23,24), such has mathematical concepts developed for the analysis of nonlinear
never reliably been clearly proven above any doubt for “nor- complex dynamical systems. Having passed above in review evi-
mal” ongoing EEG activity recorded from the scalp. dence that during epileptic seizures EEG signals display low-
dimension correlation dimension values, the question is
whether this is also the case before seizure occurrence, that is, in
ANTICIPATING EPILEPTIC SEIZURES USING the pre-ictal period. In this respect memorable contributions
SPONTANEOUS EEG SIGNALS were those of Iasemidis et al. (18–20), Lehnertz and Elger (29),
and Martinerie et al. (30).
First we should have an operational definition of what is meant As indicated above, a seizure onset, in general, represents an
by anticipating or predicting epileptic seizures. Predictability may abrupt transition from a high-dimensional to a low-dimen-
be defined operationally as “the demonstration that there exists sional state. This can be described as a phase transition in the
a statistically significant association between the outcomes of a dynamics of the neuronal networks characteristic of the non-
set of measurements preceding a seizure, and the time to the first linear nature of the underlying system. The question is thus
seizure following these measurements.” The main point that has whether phase transitions occur already in the pre-ictal period?
to be added is what kind of measurement is appropriate for the Iasemidis et al. (18–20) analyzed long stretches of EEG sig-
required association. Here we pass in review a number of meas- nals, starting many hours before a seizure, recorded from sub-
ures that have been proposed and explored. dural and implanted electrodes in the hippocampus of TLE
An early and simple idea was to use as a measure the rate of patients. They reconstructed for each EEG epoch the phase
occurrence of epileptiform spikes, that is, to check whether space and estimated the corresponding maximum Lyapunov
there exist any consistent association between variations in the exponent (Lmax). The values of Lmax of EEG epochs recorded
rate of occurrence of epileptiform spikes and the time of seizure before the seizure showed fluctuations over time. Noteworthy,
onset. While there were some early reports indicating a possible the Lmax values of EEG signals recorded from different cortical
relationship, a well controlled study by Gotman and Marciani sites started to converge (phase of entrainment) several minutes
(25) in 44 patients with focal epilepsy clarified this issue. They before a seizure, and seconds prior to a seizure these values were
monitored continuously EEG and behavior of these patients, locked together and showed an abrupt transition to a more
quantified spiking rate by an automatic detection method, and ordered state. These authors noted that a critical number of
demonstrated that spiking rates do not change before seizures sites should display entrainment over a certain time period for
but may increase markedly after them. Thus neither high nor a seizure to occur. In this way these authors consider that this
low spiking rates are associated with the occurrence of seizures, analysis methodology allows the definition of a pre-ictal transi-
and spike rating cannot be applied in order to predict seizure tion state and thus it may be used to detect an impending
onset. Other EEG statistical properties have been used with the seizure. Based on these data Iasemidis et al. (31) proposed an
aim of detecting hidden information in the pre-ictal period that adaptive seizure prediction algorithm to analyze continuous
might be used to predict the impending occurrence of a seizure. EEG signals for the prediction of TLE seizures.
Thus Viglione and Walsh (26) using pattern recognition ana- Lehnertz and Elger (29), applied the concept of EEG signal
lytic procedures of spectral data reported that EEG changes “complexity,” defined as the variability of a set of consecutive
characteristic of preseizure states could be detected a few sec- D2 estimates, in order to determine whether seizure onset could
onds before seizure onset. Using linear autoregressive models be anticipated. They found that “complexity” was reduced in
fitted to ongoing EEG signals in 12 epileptic patients suffering EEG epochs recorded close to seizure onset and proposed that
from “absences,” Rogowski et al. (27) showed that the location “neuronal complexity loss” could be used to anticipate seizure
of the poles in the z- and s-planes, as a function of time, pre- occurrence.
sented a specific pattern linked with the occurrence of the Martinerie et al. (30) analyzed 19 seizures from a homoge-
seizure. These authors noted further that the trajectory of the neous group of 11 patients with MTLE associated with
“most mobile pole” during the preseizure period could aid in hippocampal sclerosis, using indwelling electrodes, by recon-
the prediction of the seizure by several seconds. Later, Salant et structing intracranial EEG signals as trajectories in phase space.
al. (28) used multivariate spectral estimation based on para- They introduced nonlinear indicators to characterize the signal
metric modeling (determination of pole trajectories and coher- dynamics taking into account the EEG signals both as functions
ence functions) in order to detect EEG changes preceding the of time and space. In this way they were able to pinpoint the
outbreak of a seizure. Prediction of oncoming primary general- brain sites where seizure apparently started and showed that in
ized seizures was based on the detection of increased pre-ictal most cases seizure onset could be anticipated 2 to 6 minutes
648 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

frequency flows, and the problem of studying synchronization


becomes the problem of tracking frequency flows. This method
was applied to simulated signals and real EEG or MEG signals
recorded before and during epileptic seizures (35); it appeared
to be relevant in tracking transient collective dynamics fluctu-
ating in time, frequency, and space.
The reader may be overwhelmed by the large amount of
methods that we have passed in review in this short chapter.
This considerable series of methods is the result of the very
inventive work of several research groups pursuing the still eva-
sive objective of developing “the” optimal EEG analysis method
with respect to detecting relevant features of EEG signals in the
pre-ictal phase. An excellent critical description of the various
methods used in this field is given by Mormann et al. (41).
Figure 30.1 Nonlinear analysis of an intracranial EEG signal showing Recently Wendling et al. (42) made a critical comparison of
changes in the “similarity index” computed between a reference period the performance of three families of methods regarding their
and successive time windows preceding the seizure (D). The statistical capacity to assess the degree of coupling between EEG signals,
significance is indicated by color code (blue indicates significance lev- namely linear and nonlinear regression, phase synchronization,
els up to 5 standard deviations). The anticipation time with respect to and generalized synchronization. This was evaluated using a
seizure onset for EEG channel PC1 (Precuneus gyrus) is 13.5 minutes model where the relationships (coupling parameters) between
in this case. Channels with and without pre-ictal changes are depicted signals were precisely known. The main result was that there
in red and blue, respectively, on a diagram showing the locations of was no “ideal” method, that is, none of the methods performed
intracranial electrodes (E). The first channel where seizure onset is better than all the other in all conditions. They pointed out,
detected is indicated by a yellow star. AT, anterior temporal lobe; PT, however, that linear and nonlinear regression methods showed
posterior temporal lobe; PC, precuneus gyrus; IC, infracalcarine; SC, sensitivity to the coupling parameter in all tested models with
supracalcarine. (From Navarro V, Martinerie J, Le Van Quyen M, et al. average or good performances. Accordingly they concluded that
Seizure anticipation in human neocortical partial epilepsy. Brain. the strategy of choice with respect to the analysis of coupling of
2002;125(Pt 3):640–655.)(See color insert) EEG signals in epileptic brains is to first apply the regression
methods in order to characterize functional connectivity before
using more sophisticated methods that require more stringent
(Fig. 30.1). Confronted with the difficulties of applying this assumptions. We concur with this advice and stress that a prac-
methodology in practice particularly because of the nonsta- tical strategy is to use more than one method applied to the
tionarity of EEG signals that may cause many spurious detec- same EEG data, including always linear and nonlinear regres-
tions, this group developed alternative methods choosing, sion (21,43).
instead of a continuous sliding EEG window, a time window far
from the seizure as reference and compared the dynamical
properties of successive epochs of EEG signals to those of the ASSESSING THE PERFORMANCE
reference window. In this way a new measure was introduced: OF ALGORITHMS IN ANTICIPATING
the “dynamical similarity index,” which quantified changes in EPILEPTIC SEIZURES
dynamics relative to the reference window. These authors
reported a decrease of the dynamical similarity index several The early studies were followed by a series of investigations
minutes before seizures in both intracranial (32–37) and even where researchers tried a variety of modifications of the
scalp EEG recordings (38). Chávez et al. (39) analyzed changes approaches described above and proposed additional algo-
in phase synchrony and in the degree of association between rithms like the “accumulated energy” algorithm, based on wide
EEG signals recorded from different sites (using nonlinear cor- frequency band spectral energy (44), and presented critical
relation coefficient) and found a significant decrease of syn- studies with the aim of evaluating the results obtained by means
chrony in the focal area several minutes before seizures ( 30 of different methods in a rigorous way. In the context of a crit-
minutes), mainly in the frequency band of 10 to 25 Hz. ical evaluation of the performance of the “dynamical similarity
More recently the same group (40) introduced a new index” Navarro et al. (37) analyzed the dynamics of long EEG
method for analyzing multivariate phase synchronization in epochs recorded from patients with indwelling electrodes
EEG signals, which basically tracks multivariate phase synchro- undergoing monitoring for presurgical evaluation of refractory
nization in space and time–frequency domains; these authors partial epilepsy at the Montreal Neurological Institute. The
called this method frequency flow analysis (FFA). This method similarity index detected pre-ictal changes considered relevant
is particularly suited to analyze nonstationary time–frequency in two third of the seizures, these detections preceded a seizure
dynamics, since it allows the estimation of continuous phase on average by about 12 minutes, but the index could display
differences as function of time. Using this approach, groups fluctuations that were not directly followed by seizures. A large
of EEG signals can be identified as belonging to common number of these fluctuations were related to changes of vigilance
Chapter 30 ■ Anticipating Seizures Based on EEG 649

or behavior. These authors carefully note that the dynamic using “control tests” with artificial signals and also applying this
changes detected pre-ictally may “represent physiologic changes method to real ECoG signals and concluded that the use of
acting as facilitating factors or pathologic changes reflecting a Lyapunov exponents for seizure prediction is “practically
network dysfunction.” impossible as the brain dynamical system generating the ECoG
An important assessment study in this context is that of signals is more complicated than low-dimensional chaotic sys-
Aschenbrenner-Scheibe et al. (45). These authors investigated tems, and is noisy.”
the sensitivity and specificity of Lehnertz and Elger’s method This accumulation of control studies reporting a rather weak
(29,46,47) based on estimating a “dimension complexity drop” performance of the initially proposed algorithms for seizure
of sufficient amplitude and duration as indicative of the pre- prediction damped the expanding optimistic mood of this
ictal state. In invasive 24-hour long EEG recordings from 21 research field into what Mormann et al. called (41) “the rise of
patients with medically refractory partial epilepsies, who skepticism.”
underwent invasive presurgical monitoring, they found that It is recommendable that new algorithms in the field of
only one out of 88 seizures was preceded by a significant pre- seizure prediction, possibly after a first stage to present a “proof-
ictal dimension drop. They were not able to detect significant of-principle,” must be tested in a large EEG data sample.
differences of mean length and amplitude of dimension drops Although a “learning set” may initially be appropriate for algo-
between interictal and pre-ictal states. Furthermore they ana- rithm development, the results of interest must be obtained in a
lyzed the results of the same algorithm applied to EEG signals “test set” recorded from nonselected patients undergoing EEG-
comprising 50 minutes of pre-ictal and 24 hours of interictal video monitoring in the course of preparation for brain surgery
periods. Using this data set they found that this algorithm which are recorded for many hours or days continuously.
resulted in a sensitivity of 38% in hippocampal seizures and Notwithstanding the difficulties in developing reliable algo-
33% in neocortical seizures, considering that a false-positive rithms all studies support the idea that deterministic nonlinear
rate of 0.1/hr was allowed. Regarding the clinical applicability processes are involved in pre-ictal neural reorganization in
of this algorithm Aschenbrenner-Scheibe et al. (45) concluded transition to seizure.
with the comment that “used as a pure warning system, a pre-
diction method of this quality would probably be ignored after
a short time.” THE SEARCH FOR NOVEL BIOMARKERS
This study brought clear evidence that it is not sufficient to OF EPILEPTIC NETWORKS
study EEG signals during relatively short periods, namely 1
hour preceding a seizure in order to estimate the sensitivity and In addition to the endeavor of applying analytical methods to
specificity of automatic analysis methods. These estimates have ongoing EEG signals in general, in the last decade special atten-
to be made on long-term EEG recordings lasting a large num- tion was given also to particular EEG features that appear to be
ber of hours or preferably several days and nights including the specifically associated with epilepsy, whether directly related to
natural variations in wakefulness and sleep state (48,49). More seizure occurrence. This is the case of the fast oscillations that
refined methods of assessing the statistical validity of algo- were described in the hippocampus and entorhinal cortex of
rithms applied in this kind of data have been proposed (50,51). rats with spontaneous seizures (59) that were called fast ripples
Also with respect to other algorithms, subsequent tests (FRs) because they have a higher frequency range (250 to 500
under controlled conditions in practice showed that results Hz) than the normal ripple oscillations (100 to 200 Hz)
reported earlier were difficult to reproduce (52). This was the described previously in normal brain (60). Similar FRs were
case for the results obtained using the “accumulated energy” also encountered in the epileptic zones of patients with tempo-
algorithm of Litt et al. (44) that were not reproduced by ral lobe epilepsy (61). It should be noted that quite frequently
Harrison et al. (53). Maiwald et al. (54) introduced a criterion focal seizures begin with high-frequency oscillations (61,62).
called the “seizure prediction characteristic” that incorporates The interpretation is that these FRs result from the activity of
the assessment of sensitivity and false prediction rate to evalu- clusters of interconnected neurons capable of sustaining syn-
ate the performance of seizure prediction algorithms (55). chrony of discharges, surrounded by a strong inhibitory ring;
Applying this criterion, three algorithms were evaluated on a overcoming interneuron feedback inhibition may elicit the for-
large EEG data set comprising seizures of 21 patients. The mation of similar clusters in target areas, ultimately leading to
“dynamical similarity index” (37) yielded 1 to 3.6 false predic- seizures (63).
tions per day and had a sensitivity between 21% and 42%; the Thus FRs could be considered biomarkers of epileptogenic
“accumulated energy” (44) yielded a sensitivity between 18% networks, although we should add that the fact that many of these
and 31% for the extended, prospective version; and values events are restricted to tiny volumes of brain tissue makes it more
between 13% and 30% were found for the “effective correlation difficult to detect FRs with standard electrophysiologic techniques
dimension” (46,56). Performances at this level preclude clinical in the human brain. Jirsch et al. (64) showed that FRs can be
applications in a general setting. Similarly the results obtained recorded from human epileptic brain using depth macroelec-
using the Lyapunov exponent (18) could not be reproduced in trodes and concluded that these “macro-FRs” reflect the partial
the study of Lai et al. (57) on account of finite-time statistical synchronization of very local oscillations as recorded by means of
fluctuations and noise. Furthermore Lai et al. (58) examined microwires. Jacobs et al. (65) evaluated whether the occurrence of
more in detail the predictive power of Lyapunov exponents FRs (250 to 500 Hz) showed any association with the pre-ictal
650 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

period in seven consecutive MTLE patients. Total rates of FRs values to which a dynamical system settles some time after being
were significantly higher in the seizure-onset zone than outside, perturbed or in mathematical terminology an asymptotically sta-
but they did not change in a systematic way in the pre-ictal period. ble manifold in the phase space of the system. Topologically, an
Thus we may conclude that the occurrence and rate of FRs does attractor can be a point, a curve-like a limit cycle, a manifold, or
not appear to be associated with pre-ictal EEG periods. even a complicated set with a fractal structure known as a strange
attractor; the latter is typical of a chaotic dynamical system. With
respect to epilepsy in general we proposed (69,70) the following
THEORETICAL AND EXPERIMENTAL general scheme:
MODELS OF THE TRANSITION BETWEEN
Model 1: Bifurcation Model
INTERICTAL AND SEIZURE STATES
“Bi”- or “multi”-stable systems are systems where jumps
A basic problem subjacent to the difficulties in developing reli- between two or more coexisting attractors can take place,
able methods of anticipating seizures is that, at present, rela- caused by stochastic fluctuations, such as noise, of any input. In
tively little is known about the dynamics of the neuronal this case the transition between interictal state and a seizure
processes leading to seizures. Nonetheless some theoretical con- may occur at random due to some fluctuations in input condi-
cepts have been formulated that are essential in order to gain tions or in some system’s parameters, or in other words seizures
insight into these processes, as well as to put in perspective can be generated autonomously due to internal instability caus-
experimental findings in animals and the human. ing intermittent behavior (72) (Fig. 30.2).
A discussion about general principles of brain dynamics is rel-
evant in this context, since it is difficult to develop and interpret Model 2: Deformation Model
EEG data obtained in epileptic patients without having a general Systems with deformable parameters such that the dynamics
theoretical framework of the dynamics of brain activity that may may evolve from one attractor that represents the normal or the
account both for normal and epileptic neuronal activities. In this interictal state to another attractor or to a series of attractors.
context, we assume that in the epileptic brain, some neuronal The parametric deformation may be caused by the fact that
networks possess an abnormal set of control parameters that can some parameters may be, at a biologic level, unstable or may be
produce different kinds of parallel dynamical states. In other sensitive to some endogenous modulating influences, as for
words, these networks may have “bi”- or “multi”-stable proper- example, chemical neurotransmitters, hormones, and composi-
ties. This means that, in addition to a normal steady state, they tion of extracellular medium (pH), or may be induced by an
also have an abnormal one characterized by widespread synchro- external stimulus, for example, sensory stimulus in reflex
nous activity and that the transition between these two states may epilepsies. An interesting observation in this respect was pre-
occur more or less abruptly. This accounts for the two main char- sented by Baumgartner et al. (73) who performed pre-ictal
acteristics of epilepsy: (i) that an epileptic brain can function SPECT scans fortuitously while video-EEG monitoring was
apparently normally between seizures, that is, during the interic- being carried out in two TLE patients. These authors found in
tal state; and (ii) that the seizures occur in a paroxysmal way, pre-ictal SPECT a significant increase in rCBF in the epileptic
thereby impairing brain functioning to a lesser or greater extent. temporal lobe, which was not associated with significant
In this sense, epileptic disorders may be considered special cases
of the large class of dynamical diseases, meaning those patho-
physiologic states that can be characterized by intermittent
occurrence of abnormal dynamics, a theoretical concept pro-
posed by Glass and Mackey (66), and Bélair et al. (67), which we
(68–70) and others (71) have used in the context of epilepsy.
The theory of nonlinear dynamics offers the possibility to
understand, in formal terms, how the occurrence of the manifes-
tations of dynamical diseases takes place. In the case of epilepsy,
the basic question is how changes in the dynamics of a neuronal
network may occur such that paroxysmal widespread synchro-
nous oscillations abruptly emerge. It is often difficult to study
these changes in the human brain due to the inherent limita- Figure 30.2 Phase portraits of the thalamo-cortical system in two
tions of obtaining detailed neuronal data from recordings in the cases: normal and epileptic brain. Sample trajectories (thin lines) from
human brain. Therefore it is important to investigate these phe- normal (left) and epileptic model (right) projected onto a two-dimen-
nomena in experimental animals that provide reliable models of sional slice of state space. The axes are values of two state variables:
some types of epilepsy and in appropriate computer models. cortical excitatory and inhibitory activity. The thick line is a separatrix
In general, we can distinguish (69,70) two basic models that separating two attractors. (From Lopes da Silva FH, Blanes W, Kalitzin
can account for transitions to an epileptic seizure. Above we have SN, et al. Dynamical diseases of brain systems: different routes to
already presented some of the properties of complex nonlinear epileptic seizures. IEEE Trans Biomed Eng. 2003;50(5):540–548 and
dynamical systems, but a further element should be put forward. Lytton WW. Computer modelling of epilepsy. Nat Rev Neurosci.
It is a fact that these systems possess attractors, that is, sets of 2008;9(8):626–637.)
Chapter 30 ■ Anticipating Seizures Based on EEG 651

changes of the ongoing EEG. They interpreted this finding as constructed and proved to have not only face value but also pre-
indicating that rCBF changes observed on peri-ictal SPECT dictive value. Without entering here in details (see for detailed
may reflect a change in neuronal activity precipitating the tran- model Refs. 82–85) we may state that the model studies revealed
sition from the interictal to the ictal state. Also a study by that (i) SWDs result from dynamical bifurcations that occur in
Federico et al. (74) attempted to determine whether changes in a bistable neuronal network; (ii) the durations of interictal
fMRI (BOLD signal) could be detected in the preseizure period. epochs have exponential distributions, indicating that transi-
The results in three patients indeed showed changes in pre-ictal tions between these two stable states occur randomly over time
BOLD signal before seizure onset but these were rather com- with constant probabilities (see Ref. 86 for statistical distribu-
plex and not easy to interpret. tions of seizures in epileptic patients); (iii) the probabilistic
In addition we can also consider systems displaying a combi- nature of the onset of paroxysmal activity implies that it is not
nation of these two processes, that is, scenarios where the system possible to predict its occurrence; (iv) the bistable nature of the
“deforms” from single attractor to multistable dynamics (bifur- dynamical system allows that an ictal state may be aborted by a
cation). One feature of both the deformation and bifurcation single counter-stimulus, even by a single stimulation pulse.
models is that they are not, strictly speaking, autonomous. They Kalitzin et al. (75) have developed this concept further including
need external input or endogenous noise in order to allow for continuously operating external modules designed to constantly
transitions. This is not, however, a major conceptual problem monitor and react to epileptic transitions.
(75). The living brain is hardly ever a closed dynamic system, An example of Model 2 is the occurrence of partial seizures
since it is always subjected to external influences. The so-called in MTLE. This model is in accordance with experimental stud-
spontaneous seizures, for example, simply indicate our lack of ies. One of these in particular is relevant. This is the investigation
knowledge about the precipitating factor or event. carried out by Cohen et al. (87) in hippocampal slices, in vitro,
Recently the possibility of a third class of models of that shed light into the basic question, namely whether in this
autonomous epileptic seizure generation has been investigated. kind of epileptic seizures a pro-ictal state can be identified, in
In these models the system possesses a feature called dynamic contrast to what was observed with respect to absence seizures.
intermittency (76–78). In its essence such systems are character- This study showed that before a full-blown seizure may be
ized by complex attractors and partially unstable dynamics. detectable in local field potentials, there is a gradual change in
Areas in the phase space (around the so-called saddle points) electrophysiological properties that becomes manifest as a
can deflect the system into various types of behavior. In those build-up of neuronal firing frequency variance. This implies that
systems the transitions to epileptiform activity can occur pure a pro-ictal state with special properties can be identified at least
autonomously, without the help from any endogenous or exoge- in this experimental condition. Thus these observations show
nous factor. Although intermittency has been suggested as a pos- that the evolution of the excitability state of the neuronal popu-
sible mechanism for various occurrences of seizure generation lation in these hippocampal slices differs sharply from what was
(79), a realistic neuronal model of intermittency expressed in seen in the case of SWDs in absence epilepsy. Assuming that
terms of changes in neuronal dynamics is yet to be built. these in vitro observations are representative of the in vivo situ-
An example of Model 1 is the occurrence of spike-and-wave ation in patients with MTLE, and taking these findings together,
discharges (SWDs) in the thalamocortical system, typical of we may draw the conclusion that it should be possible, in prin-
absence seizures. This model is in agreement with experimental ciple, to identify a pro-ictal state in this and similar cases.
observations carried out in order to understand how SWDs are Within the framework of these theoretical and experimental
generated. Typical SWDs start and end abruptly. We found in a models we can state that the transition to a seizure can occur
rat genetic model of absences, the so called WAG/Rij rat (43) according to two scenarios: (i) as a sudden leap; in this case the
that SWDs are initiated in the facial somatosensory cortex and transition may be unpredictable as explained in detail by the
from there propagate to other cortical areas and to the thalamus. model studies shortly described above; (ii) as a continuous
This was confirmed in another experimental rat model of sequence of changes in the dynamics of neuronal networks. In
absences, the Genetic Absence Epilepsy Rats of Strasbourg this type of ictal transition, it is conceivable that the seizure may
(GAERS), where Polack et al. (80) found that the display of be anticipated in its early, pre-ictal (preclinical) phase. Whether
SWDs in the local ECoG coincides with rhythmic membrane these dynamical changes at the neuronal level may be detectable
depolarizations superimposed on a tonic hyperpolarization of at the ongoing EEG level is, of course, the crux of all the discus-
layer IV neurons, which occurs abruptly. Furthermore it was sions concerning the algorithms developed to anticipate
demonstrated in a detailed microelectrophysiolgic study (81), seizures based on the analysis of ongoing EEG signals, which we
also in GAERS, that layer VI corticothalamic neurons act as “dri- discussed above. An important advance in this respect is the
vers” in the generation of SWDs in the somatosensory thalamo- model of Wendling et al. (88,89) that shows how the deforma-
cortical system. These observations reveal also that SWDs occur tion of certain parameters can cause the neuronal network
abruptly in this animal model. A general conclusion that may be dynamics to evolve though a succession of phases and ulti-
drawn from these experimental studies is that the SWDs, char- mately to display a full-blown seizure (Fig. 30.3).
acteristic of absence seizures, tend to occur without a clear pro- An important clue to understand this basic question was
ictal electrophysiologic ECoGraphic pattern. In order to obtained when we investigated the process leading to an absence
understand in more general terms how this abrupt change of seizure induced by intermittent photic stimulation (IPS) (90,91).
neuronal dynamics takes place a computational model has been This investigation revealed that the phase clustering of harmonically
652 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Figure 30.3 Computational model (left-hand side) based on a lumped approach of a hippocampal network (based on
the representation of neuronal subpopulations of pyramidal neurons and two populations of interneurons) and a real
neuronal network (right-hand side, based on the explicit representation of cells and interconnections) approach.
Simulated activity (local field potentials) can be quantitatively compared to real activity using information processing
techniques. This quantitative analysis allows for identifying the parameters settings for which models best reproduce
real data. Performing this identification over a sliding time window can trace the evolution of model parameters, pro-
viding information into the mechanisms responsible for the transition between normal and seizure activity. (From
Wendling F. Computational models of epileptic activity: a bridge between observation and pathophysiological interpre-
tation. Expert Rev Neurother. 2008;8(6):889–896.)(See color insert)

related frequency components of a subject’s MEG/EEG responses pre-ictal; indeed pre-ictal denotes just the activity immediately
evoked by IPS increases significantly seconds before a seizure preceding in time the seizure, which is, of course, always pres-
occurs. This led to the introduction of a quantity—the phase clus- ent, whether or not it may be causally related to the transition
tering index (PCI, see Appendix for definition)—by means of to a seizure; pro-ictal implies that the activity reflects a state of
which this enhancement can be quantified. We may argue that this excitability of the underlying neuronal networks that can lead
quantity reflects the degree of excitability of the underlying dynam- to a seizure. If the system is detected to be in a pro-ictal state
ical system, and it can indicate the presence of nonlinear dynamics. then an impending seizure may be anticipated. Here we must
Applying this form of analysis we found that the patients who add that such a state itself does not determine for 100% the
develop seizures during IPS present an enhancement of the PCI at occurrence of the seizure, only the predisposition of the system
the gamma frequency band, compared with that at the driving fre- for a transition to an epileptic state.
quency; this is why the index is called relative PCI or rPCI. An
important lesson to be drawn from this finding is that the change in
dynamics of EEG or MEG signals preceding the transition to the NEW APPROACHES IN ANTICIPATING
seizure may be more readily detected by using an active probe, that SEIZURES: “ACTIVE” VERSUS
is, a stimulus to provoke a response of the system than by a passive “PASSIVE” ALGORITHMS
analysis of the ongoing signals. These observations led to test
whether the use of active probes could be also used in other forms The findings of photosensitive epilepsy described above
of epilepsy according to Model 2 scenario, such as in MTLE, as (90,91) prompted us to investigate whether rPCI of EEG sig-
described in the section “New Approaches in Anticipating Seizures.” nals recorded in patients with MTLE could also have predictive
As a footnote we may add that it may be more appropriate value with respect to the anticipation of seizures. This implies
to use the term pro-ictal than to use the more common term the application of a stimulus, since the estimation of rPCI is
Chapter 30 ■ Anticipating Seizures Based on EEG 653

based on the EEG activity modulated by an external “carrier” Although this method requires brain electrical stimulation, it
signal. In the case of photosensitive epilepsy we used, intermit- is not more complex than electrical stimulation paradigms
tent light. In the case of patients with MTLE we had to resort used for clinical purposes during intracranial EEG investiga-
to deep brain electrical stimulation using the available elec- tions to delineate epileptogenic areas and seizure-onset zones
trodes that were implanted for long-term EEG monitoring and by determining thresholds for eliciting afterdischarges (ADs)
recording seizures. Thus we developed an active electrical or by triggering seizures. In fact our method was also success-
direct-stimulation paradigm. With this objective we stimulated ful in identifying the seizure-onset zone as the location pro-
patients with indwelling electrodes in the hippocampal forma- ducing increased rPCI values. This let us to conclude that the
tion using short, low-intensity (up to 1 mA) bursts of electri- rPCI quantity reflects the ability of the neuronal system to gen-
cal pulses, 5 seconds long, at the frequency of 20 Hz, repeated erate epileptic discharges. Remarkably, the measurement of
at intervals of 20 seconds over long period of hours, and even rPCI does not involve provoking those discharges, unlike other
days at which time the intracranial EEG was sampled in paral- stimulation-based techniques. To understand the mechanisms
lel. We call this stimulation paradigm a carrier frequency mod- that allow for such effectiveness we have resorted again to real-
ulation probe (92). The relative phase clustering index (rPCI) istic computer models.
computed for all signals and all stimulated epochs of six The emphasis on “active paradigms” for testing changes of
patients was found to be larger for electrode sites near to the excitability of neuronal networks that according to either the
seizure-onset site. Even more interesting was the finding that it “deformation model” or the “combined model” will occur in
was possible to forecast the probability of a seizure occurring the pro-ictal period as the onset of a seizure approaches has also
within a certain time based on the values of rPCI estimated at been recently corroborated by simulations, using a computa-
a given moment in time (Fig. 30.4). For example in the patient tional model of seizure generation. Suffczynski et al. (84) inves-
population analyzed in this study, a value of rPCI 0.6 pre- tigated (i) how changes in model parameters that lead to a
dicts that a seizure will occur in less than 2 hours, with an transition from the normal ongoing EEG to an ictal pattern are
accuracy 80%. This study has still to be considered as a reflected in the properties of the simulated EEG output signals
“proof-of-principle” since no further controlled studies were and (ii) how the evolution of neuronal excitability towards
published so far. Nonetheless the application of “active probes” seizures can be reconstructed from EEG data using an “active”
opens a novel strategy in the field of seizure anticipation. paradigm, rather than the traditional “passive” algorithms that
use only ongoing EEG signals. The simulations indicate that an
“active” paradigm combined with appropriate analytical tools,
such as rPCI, yields more readily relevant information about
the change in excitability that characterizes the pre-ictal, or bet-
ter the pro-ictal, period than algorithms applied to the ongoing
EEG. These findings give strong support to the development
and application of “active” paradigms with the aim of charac-
terizing the change of excitability during the pro-ictal period
and thus anticipating the occurrence of a transition to an
epileptic seizure.
A quite different study gives support to the concept that
“active paradigms” may yield relevant information regarding
the characterization of the pre-ictal state. This is the investiga-
tion of Badawy et al. (93) who used TMS in patients with dif-
ferent forms of epilepsy (generalized, focal or focal with
Figure 30.4 Results of the analysis of intracranial EEG signals from secondary generalization) to determine the cortical motor
temporal lobe epileptic patients. The relative Phase Clustering Index threshold and paired pulse recovery curves (at short, 2 to 15
(rPCI) was computed for several EEG channels of six patients at vari- msec, and long, 50 to 400 msec interstimulus intervals) at 24
ous times before seizure onset. Upper image: median values and 95% hours before a seizure (also postseizure). They found
confidence intervals for the six patients are shown along the horizontal decreased motor threshold, increased intracortical facilitation,
axis; along the vertical axis the time interval to the next seizure is plot- and decreased intracortical inhibition, indicative of an increase
ted. Note that the values of rPCI increase with a decrease in the dura- in cortical excitability and/or a decrease in cortical inhibitory
tion of this interval. Lower image: plot of the rPCI value (vertical axis) processes, at short and long interstimulus intervals, in the 24
against the prediction horizon, that is, the time interval to seizure hours before a seizure, and the opposite changes in the 24
onset. In color the error rate of the prediction is indicated, from a low hours after a seizure. These effects were bilateral in tonic-
error (dark blue) to a large error (red, brown). Note that for values of clonic seizures in idiopathic generalized epilepsy and also in
rPCI 0.6 there is a probability of a seizure occurring in less than 2 secondarily generalized seizures in patients with focal epilepsy.
hours with an accuracy of 80% . (From Kalitzin SN, Velis DN, Similar changes were seen in the hemisphere ipsilateral to the
Suffczynski P, et al. Electrical brain-stimulation paradigm for estimating seizure focus in focal seizures that did not secondarily general-
the seizure onset site and the time to ictal transition in temporal lobe ize. It appears that by means of TMS it is possible to detect
epilepsy. Clin Neurophysiol. 2005;116(3):718–728.)(See color insert) changes in cortical excitability preceding seizure by hours.
654 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Nonetheless these results are based on measurements of aver- nine chronically implanted patients with mesial temporal lobe
ages of the TMS responses obtained in experimental sessions epilepsy who underwent almost continuous high-frequency
that lasted for 60 to 90 minutes; these measurements were not hippocampal electrical stimulation (97) no memory deficits
followed along time as done by Kalitzin et al. (94), so it is not could be identified on neuropsychological testing with a fol-
known whether these changes covary with the progressive low-up of up to 7 years.
approximation of a seizure. Therefore we can assume that This is a hard, though challenging field that requires the
those findings corroborate our own findings and our hypoth- convergence of a variety of scientific research approaches to
esis of existence of a pro-ictal state that precedes in a nondeter- achieve results that may have practical usefulness in clinical
ministic way the transition to epileptic seizure. epilepsy.

CONCLUDING REMARKS APPENDIX


The road toward the development of strategies aiming at a reli- We introduce here the phase clustering index (PCI) (91) and
able detection of a pro-ictal state has been full of exciting the- the associated relative PCI (rPCI) as a measure of the ability of
oretical challenges and cunning experimental approaches. the system to phase-lock its response to an external stimulus.
Despite the fact that the scientific community learned a lot We assume that the system is exposed to the same stimulus
about the dynamics of neuronal networks in the context of at discrete time points ts H5t1, p , tN s6 , where N s is the number
epileptic seizures in these last decades, we are still far from hav- of stimuli. If we denote the system’s response (this can be any
ing a generally accepted and robust strategy. A positive devel- measured multidimensional physiologic signal such as EEG,
opment is that the community has set up a number of MEG, etc.) as Sc(t), c = 1, p , N c , N c indicating the number of
Workshops on Seizure Prediction and a digital database that channels, then the stimulus-triggered response (STR) function
can be used to develop and test suitable algorithms in a real is defined as
collaborative effort. The need of new strategies is becoming
Rsc(t) S c(t ts) (30.1)
even more pressing since the access to deep brain stimulation
techniques for a variety of brain diseases is now also open to In the particular case of periodic stimulations where all con-
epileptic patients, particularly to those who are pharmacologi- secutive stimuli are separated by a constant period T ts 1
cally refractory and yet have either been rejected for a surgical ts, s 1, p , N s, it is natural to select the response interval t H
intervention or have had failed epilepsy surgery. Thus it would [0,T) in Eq. (30.1). Our further definitions are not restricted to
be desirable to have reliable methods capable of seizure antic- this choice but the previous applications of PCI and the exam-
ipation so that a dedicated “closed-loop” stimulation proce- ples given below assume this simple nonredundant representa-
dure might be developed to avoid the occurrence of seizures on tion. The general theory is also perfectly applicable to general
the basis of continuous and reliable analysis of the running nonperiodic stimuli. Below we also omit the channel index c for
EEG record. The success of such procedures depends on the notation simplicity.
possibility to either foresee an impending seizure or to detect Next we perform a discrete Fourier transform (30.1) along
its occurrence at the earliest possible stage and only then to the t-axis:
apply a counter-stimulus in order to arrest it. A few “closed-
Z fs fft(Rs(t) (30.2)
loop” approaches are starting to be explored, namely using
properties of the measured EEG signal to determine the exact where f denotes the discrete frequency associated with the com-
timing of the counter-stimulation (95) or generating an appro- plex coefficients Z. In our applications we assume f H{1,2, p
priate seizure suppression stimulation waveform (96). We may N f}f0, where f0 is the fundamental frequency, typically the fre-
refer to the latter as “state-reactive control.” This matter is dis- quency of the stimulation in case of periodic stimuli and N f is
cussed theoretically in more detail by Kalitzin et al. (75) who the highest harmonic frequency considered. We will also omit
give arguments to justify different scenarios for seizure control, the f0 notation, unless otherwise stated, and we shall refer to the
required according to different dynamical models of seizure frequency as an integer number.
generation. Multistable and possibly some intermittent sys- Averaging either Eq. (30.1) or (30.2) over the stimuli yields
tems may be controlled by an acute intervention that aborts the classic averaged evoked response (or evoked potential if the
the transition to the seizure and resets the system to its normal signal represents local field potentials). We introduce here the
state. Parametric deformation of the system’s attractor can be definition of the PCI:
compensated by continuously stimulating the system and
Zfs s PCIf Z fs s PCIf Af (30.3)
reducing the ictal attractor to a stable steady state. All of the
aforementioned approaches require indwelling systems that Here and later s means averaging over all periods (stimuli)
deliver electrical current to selected targets in the brain. and denotes taking the magnitude of a complex number. In
Although a considerable body of evidence exists on the basis of essence the factorization in Eq. (30.3) represents the spectral-
which intermittent high-frequency deep brain stimulation in evoked response as a product of an averaged amplitude spec-
extrapyramidal disorders is deemed safe, the long-term effects trum and the PCI. In this way one can obtain one more degree
of chronic simulation administered to the human hippocam- of information related to the evoked responses. Explicitly the
pus are still largely unknown. In a review of the experience in factors are
Chapter 30 ■ Anticipating Seizures Based on EEG 655

Z fs s 7. Grassberger P, Procaccia I. Measuring the strangeness of strange


PCIf ` ` attractors. Physica D. 1983;9:189–208.
Z fs s (30.4)
8. Grassberger P, Procaccia I. Characterization of strange attractors.
Af Z fs s Phys Rev Lett. 1983;50(5):236–349.
9. Pritchard WS, Duke DW. Measuring chaos in the brain. A tutorial
From Eqs. (30.3) and (30.4) it is clear that in Fourier repre- review of nonlinear dynamical EEG analysis. Int J Neurosci.
sentation the STR is a product of two factors. One of the factors 1992;67:31–80.
represents the averaged spectral power of the response and the 10. Grassberger P, Schreiber T, Schaffrath C. Nonlinear time sequence
second, the PCI, accounts for the phase of the response relative analysis. Int J Bifurc Chaos. 1991;1:521–547.
11. Theiler J, Eubank S, Longtin A, et al. Testing for nonlinearity in
to the stimulus.
time series: the method of surrogate data. Physica D. 1992;58:
We can easily see that the PCI is exactly equal to 1 if the 77–94.
response complex amplitudes are of the same phase, irrespec- 12. Pijn JP, van Neerven DN, Noest A, et al. Chaos or noise in EEG sig-
tive of the values of the magnitudes. If on the contrary, the nals: dependence on state and brain site. Electroenceph Clin
phases of the individual responses are randomly scattered and Neurophysiol. 1991;79:371–381.
the magnitudes are equal, the PCI quantity is exactly 0. 13. Prichard D, Theiler J. Generating surrogate data for time series
We have shown previously (90,91) that a particular combi- with several simultaneously measured variables. Phys Rev Lett.
nation of the PCI spectrum, that we called rPCI, emerged as a 1994;73(7):951–954.
good identifier of an impending transition to seizures both in 14. Schreiber T, Schmitz A. Improved surrogate data for nonlinearity
patients with photosensitive epilepsy submitted to periodic tests. Phys Rev Lett 1996;77(4):635–638.
visual stimulation and in TLE patients suffering from sponta- 15. Achermann P, Hartmann R, Gunzinger A, et al. Correlation
dimension of the human sleep electroencephalogram: cyclic
neous seizures (Fig. 30.4). We define the rPCI as
changes in the course of the night. Eur J Neurosci. 1994;6(3):
rPCI maxf (PCIf ) PCI1 (30.5) 497–500.
16. Stam CJ, Pijn JP, Suffczynski P, et al. Dynamics of the human alpha
In this definition the spectral maximum of PCI at frequency rhythm: evidence for non-linearity? Clin Neurophysiol.
f is compared to the phase consistency of the first Fourier com- 1999;110(10):1801–1813.
ponent or “the base driving.” 17. Babloyantz A, Destexhe A. Low-dimensional chaos in an instance
Our final remark in this section concerns the statistical signif- of epilepsy. Proc Natl Acad Sci USA. 1986;83(10):3513–3517.
icance test of the measured PCI values. Using the same magni- 18. Iasemidis LD, Sackellares JC, Zaveri HP, et al. Phase space topogra-
tudes Af as defined in Eqs. (30.3) and (30.4) but generating a phy and the Lyapunov exponent of electrocorticograms in partial
large number of surrogate phases for each frequency component seizures. Brain Topogr. 1990;2(3):187–201.
of the response, we can compute a set of PCI values in the 19. Iasemidis LD, Olson LD, Savit RS, et al. Time dependencies in the
occurrences of epileptic seizures. Epilepsy Res. 1994;17(1):81–94.
absence of phase-locking to the stimulus. Then the 95th per-
20. Iasemidis LD, Principe JC, Czaplewski JM, et al. Spatiotemporal
centile PCI value defines the threshold for P 0.05 significant transitions to epileptic seizures: a nonlinear dynamical analysis
PCI. This “surrogate phases test” will exclude falsely high PCI val- of scalp and intracranial EEG recordings. In: Lopes da Silva F,
ues that are generated by a single complex amplitude in which a Principe JC Almeida LB, eds. Spatiotemporal Models in
very high magnitude dominates the averaging in Eq. (30.4). Biological and Artificial Systems. Amsterdam: IOS Press;
1996:81–88.
21. Pijn JP, Velis DN, van der Heyden MJ, et al. Nonlinear dynamics of
REFERENCES epileptic seizures on basis of intracranial EEG recordings. Brain
Topogr. 1997;9(4):249–270.
1. Hughes J, Devinsky O, Feldmann E, et al. Premonitory symptoms
22. van der Heyden MJ, Velis DN, Hoekstra BP, et al. Non-linear analy-
in epilepsy. Seizure. 1993;2:201–203.
sis of intracranial human EEG in temporal lobe epilepsy. Clin
2. Schulze-Bonhage A, Kurth C, Carius A, et al. Seizure anticipation Neurophysiol. 1999;110(10):1726–1740.
by patients with focal and generalized epilepsy: a multicentre 23. Skarda CA, Freeman WJ. How brains make chaos in order to make
assessment of premonitory symptoms. Epilepsy Res. 2006;70(1): sense of the world. Behav Brain Sci. 1987;10:161–195.
83–88. 24. Freeman WJ. Neural networks and chaos. J Theor Biol.
3. Scaramelli A, Braga P, Avellanal A, et al. Prodromal symptoms in 1994;171(1):13–18.
epileptic patients: clinical characterization of the pre-ictal phase. 25. Gotman J, Marciani MG. Electroencephalographic spiking activ-
Seizure. 2009;18(4):246–250. ity, drug levels, and seizure occurrence in epileptic patients. Ann
4. Zaveri HP, Frei MG, Osorio I. State of seizure prediction: a report Neurol. 1985;17(6):597–603.
on informal discussions with participation of the 3rd 26. Viglione SS, Walsh GO. Proceedings: epileptic seizure prediction.
International Workshop on seizure prediction. In: Schelter B, Electroencephalogr Clin Neurophysiol. 1975;39(4):435–436.
Timmer J, Schulze-Bonhage A, eds. Seizure Prediction in Epilepsy. 27. Rogowski Z, Gath I, Bental E. On the prediction of epileptic
Weinheim: John Wiley – VCH Verlag GmbH & Co. KGaA; seizures. Biol Cybern. 1981;42:9–15.
2008:325–330. 28. Salant Y, Gath I, Henriksen O. Prediction of epileptic seizures from
5. Scheffer M, Bascompte J, Brock WA, et al. Early-warning signals two-channel EEG. Med Biol Eng Comput. 1998;36(5):549–556.
for critical transitions. Nature. 2009;461(7260):53–59. 29. Lehnertz K, Elger CE. Spatio-temporal dynamics of the primary
6. Takens F. Detecting strange attractors in turbulence. In: Rand DA, epileptogenic area in temporal lobe epilepsy characterized by neu-
Young LS, eds. Lecture Notes in Mathematics. Vol. 898. Berlin:
Springer-Verlag; 1981:366–381.
656 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

ronal complexity loss. Electroencephalogr Clin Neurophysiol. tic seizure prediction. Chaos. 2006;16(1):013108.
1995;95(2):108–117. 51. Schelter B, Feldwisch-Drentrup H, Timmer J, et al. A common
30. Martinerie J, Adam C, Le Van Quyen M, et al. Epileptic seizures can strategy and database to compare the performance of seizure pre-
be anticipated by non-linear analysis. Nat Med. 1998;4(10): diction algorithms. Epilepsy Behav. 2010;17(2):154–156.
1173–1176. 52. Harrison MA, Osorio I, Frei MG, et al. Correlation dimension and
31. Iasemidis LD, Shiau DS, Chaovalitwongse W, et al. Adaptive integral do not predict epileptic seizures. Chaos. 2005;15(3):33106.
epileptic seizure prediction system. IEEE Trans Biomed Eng. 53. Harrison MA, Frei MG, Osorio I. Accumulated energy revisited.
2003;50(5):616–627. Clin Neurophysiol. 2005;116(3):527–531.
32. Le Van Quyen M, Martinerie J, Baulac M, et al. Anticipating 54. Maiwald T, Winterhalder M, Aschenbrenner-Scheibe R, et al.
epileptic seizures in real time by a non-linear analysis of similarity Comparison of three nonlinear seizure prediction methods by
between EEG recordings. Neuroreport. 1999;10(10):2149–2155. means of the seizure prediction characteristic. Physica D.
33. Le Van Quyen M, Adam C, Martinerie J, et al. Spatio-temporal 2004;194(3–4):357–368.
characterizations of non-linear changes in intracranial activities 55. Winterhalder M, Maiwald T, Voss HU, et al. The seizure prediction
prior to human temporal lobe seizures. Eur J Neurosci. characteristic: a general framework to assess and compare seizure
2000;12(6):2124–2134. prediction methods. Epilepsy Behav. 2003;4(3):318–325.
34. Le Van Quyen M. Anticipating epileptic seizures: from mathemat- 56. Lehnertz K, Elger CE. Neuronal complexity loss in temporal lobe
ics to clinical applications. C R Biol. 2005;328(2):187–198. epilepsy: effects of carbamazepine on the dynamics of the epilepto-
35. Le Van Quyen M, Amor F, Rudrauf D. Exploring the dynamics of genic focus. Electroencephalogr Clin Neurophysiol. 1997;103(3):
collective synchronizations in large ensembles of brain signals. 376–380.
J Physiol Paris. 2006;100(4):194–200. 57. Lai YC, Harrison MA, Frei MG, et al. Inability of Lyapunov expo-
36. Navarro V, Martinerie J, Le Van Quyen M, et al. Seizure anticipa- nents to predict epileptic seizures. Phys Rev Lett. 2003;91:068102.
tion in human neocortical partial epilepsy. Brain. 2002;125 58. Lai YC, Harrison MA, Frei MG, et al. Controlled test for predictive
(Pt 3):640–655. power of Lyapunov exponents: their inability to predict epileptic
37. Navarro V, Martinerie J, Le Van Quyen M, et al. Seizure anticipa- seizures. Chaos. 2004;14:630–642.
tion: do mathematical measures correlate with video-EEG evalua- 59. Bragin A, Engel J Jr, Wilson CL, et al. Electrophysiologic analysis of
tion? Epilepsia. 2005;46(3):385–396. a chronic seizure model after unilateral hippocampal Epilepsia KA
38. Le Van Quyen M, Martinerie J, Navarro V, et al. Anticipation of injection. Epilepsia. 1999;40(9):1210–1221.
epileptic seizures from standard EEG recordings. Lancet. 60. Buzsáki G, Horváth Z, Urioste R, et al. High-frequency network
2001;357(9251):183–188. oscillation in the hippocampus. Science. 1992;256(5059):
39. Chávez M, Le Van Quyen M, Navarro V, et al. Spatio-temporal 1025–1027.
dynamics prior to neocortical seizures: amplitude versus phase 61. Bragin A, Engel J Jr, Wilson CL, et al. High-frequency oscillations
couplings. IEEE Trans Biomed Eng. 2003;50(5):571–583. in human brain. Hippocampus. 1999;9(2):137–142.
40. Rudrauf D, Douiri A, Kovach C, et al. Frequency flows and the 62. Traub RD, Draguhn A, Whittington MA, et al. Axonal gap junctions
time-frequency dynamics of multivariate phase synchronization between principal neurons: a novel source of network oscillations,
in brain signals. Neuroimage. 2006;31(1):209–227. and perhaps epileptogenesis. Rev Neurosci. 2002;13(1):1–30.
41. Mormann F, Andrzejak RG, Elger CE, et al. Seizure prediction: the 63. Bragin A, Wilson CL, Staba RJ, et al. Interictal high-frequency
long and winding road. Brain. 2007;130(Pt 2):314–333. oscillations (80–500 Hz) in the human epileptic brain: entorhinal
42. Wendling F, Ansari-Asl K, Bartolomei F, et al. From EEG signals to cortex. Ann Neurol. 2002;52(4):407–415.
brain connectivity: a model-based evaluation of interdependence 64. Jirsch JD, Urrestarazu E, LeVan P, et al. High-frequency oscillations
measures. J Neurosci Methods. 2009;183(1):9–18. during human focal seizures. Brain. 2006;129(Pt 6):1593–1608.
43. Meeren HK, Pijn JP, Van Luijtelaar EL, et al. Cortical focus drives 65. Jacobs J, Zijlmans M, Zelmann R, et al. Value of electrical stimula-
widespread corticothalamic networks during spontaneous absence tion and high frequency oscillations (80–500 Hz) in identifying
seizures in rats. J Neurosci. 2002;22(4):1480–1495. epileptogenic areas during intracranial EEG recordings. Epilepsia.
44. Litt B, Esteller R, Echauz J, et al. Epileptic seizures may begin hours 2010;51(4):573–582.
in advance of clinical onset: a report of five patients. Neuron. 66. Glass L, Mackey MC. (1988). The Rhythms of Life. New Jersey:
2001;30(1):51–64. Princeton University Press; 1988:248.
45. Aschenbrenner-Scheibe R, Maiwald T, Winterhalder M, et al. How 67. Bélair J, Glass L, van der Heiden U, et al. Dynamical disease: iden-
well can epileptic seizures be predicted? An evaluation of a nonlin- tification, temporal aspects and treatment strategies of human ill-
ear method. Brain. 2003;126(Pt 12):2616–2626. ness. Chaos. 1995;5(1):1–7.
46. Elger CE, Lehnertz K. Seizure prediction by non-linear time series 68. Lopes da Silva FH, Pijn JPM, Gorter JA, et al. Rhythms of the
analysis of brain electrical activity. Eur J Neurosci. 1998;10(2): brain: between randomness and determinism. In: Lehnertz K,
786–789. Arhold J, Grassberger P, et al., eds. Chaos in Brain. Singapore:
47. Lehnertz K, Andrzejak RG, Arnhold J, et al. Nonlinear EEG analy- World Scientific; 2000:63–76.
sis in epilepsy: its possible use for interictal focus localization, 69. Lopes da Silva FH, Blanes W, Kalitzin SN, et al. Epilepsies as
seizure anticipation, and prevention. J Clin Neurophysiol. dynamical diseases of brain systems: basic models of the transition
2001;18(3):209–222. between normal and epileptic activity. Epilepsia. 2003;44(s12):
48. Litt B, Lehnertz K. Seizure prediction and the preseizure period. 72–83.
Curr Opin Neurol. 2002;15(2):173–177. 70. Lopes da Silva FH, Blanes W, Kalitzin SN, et al. Dynamical diseases
49. Litt B, Echauz J. Prediction of epileptic seizures. Lancet Neurol. of brain systems: different routes to epileptic seizures. IEEE Trans
2002;1(1):22–30. Biomed Eng. 2003;50(5):540–548.
50. Schelter B, Winterhalder M, Maiwald T, et al. Testing statistical sig- 71. Andrzejak RG, Widman G, Lehnertz K, et al. The epileptic process
nificance of multivariate time series analysis techniques for epilep- as nonlinear deterministic dynamics in a stochastic environment:
Chapter 30 ■ Anticipating Seizures Based on EEG 657

an evaluation on mesial temporal lobe epilepsy. Epilepsy Res. absence epilepsy simulating spike and wave discharges after viga-
2001;44:129–40. batrin in WAG/Rij rats. Eur J Neurosci. 2007;25(9):2783–2790.
72. Lytton WW. Computer modelling of epilepsy. Nat Rev Neurosci. 86. Milton JG, Gotman J, Remillard GM, et al. Timing of seizure
2008;9(8):626–637. recurrence in adult epileptic patients: a statistical analysis.
73. Baumgartner C, Serles W, Leutmezer F, et al. Preictal SPECT in Epilepsia. 1987;28:471–478.
temporal lobe epilepsy: regional cerebral blood flow is increased 87. Cohen I, Huberfeld G, Miles R. Emergence of disinhibition-
prior to electroencephalography-seizure onset. J Nucl Med. induced synchrony in the CA3 region of the guinea pig hippocam-
1998;39(6):978–982. pus in vitro. J Physiol. 2006;570(Pt 3):583–594.
74. Federico P, Abbott DF, Briellmann RS, et al. Functional MRI of the 88. Wendling F, Bartolomei F, Bellanger JJ, et al. Epileptic fast activity
pre-ictal state. Brain. 2005;128:1811–1817. can be explained by a model of impaired GABAergic dendritic
75. Kalitzin SN, Velis DN, Lopes da Silva FH. Stimulation-based antic- inhibition. Eur J Neurosci. 2002;15(9):1499–1508.
ipation and control of state transitions in the epileptic brain. 89. Wendling F. Computational models of epileptic activity: a bridge
Epilepsy Behav. 2010;17(3):310–323. between observation and pathophysiological interpretation.,
76. Ohayon EL, Kwan HC, McIntyre Burnham W, et al. Adaptable Expert Rev Neurother. 2008;8(6):889–896.
intermittency and autonomous transitions in epilepsy and cogni- 90. Parra J, Kalitzin SN, Iriarte J, et al. Gamma-band phase clustering
tion, advances in cognitive neurodynamics. ICCN Proceedings. and photosensitivity: Is there an underlying mechanism common
Springer; 2007;485–490. to photosensitive epilepsy and visual perception? Brain.
77. Ohayon EL, Kalitzin S, Suffczynski P, et al. Charting epilepsy by 2003;126(Pt 5):1164–1172.
searching for intelligence in network space with the help of evolv- 91. Kalitzin SN, Parra JG, Velis DN, et al. Enhancement of phase cluster-
ing autonomous agents. J Physiol. 98(4–6):507–529. ing in the EEG/MEG gamma frequency band anticipates transitions
78. Zalay OC, Bardakjian BL. Mapped clock oscillators as ring devices to paroxysmal epileptiform activity in epileptic patients with known
and their application to neuronal electrical rhythms. IEEE Trans visual sensitivity. IEEE Trans BioMed Eng. 2002;49(11):1279–1285.
Neural Syst Rehabil Eng. 2008;16(3):233–244. 92. Kalitzin SN, Velis DN, Suffczynski P, et al. Electrical brain-
79. Velazquez JL, Khosravani H, Lozano A, et al. Type III intermittency stimulation paradigm for estimating the seizure onset site and the
in human partial epilepsy. Eur J Neurosci. 1999;11(7):2571–2576. time to ictal transition in temporal lobe epilepsy. Clin Neurophysiol.
80. Polack PO, Guillemain I, Hu E, et al. Deep layer somatosensory 2005;116(3):718–728.
cortical neurons initiate spike-and-wave discharges in a genetic 93. Badawy R, Macdonell R, Jackson G, et al. The peri-ictal state: cor-
model of absence seizures. J Neurosci. 2007;27(24):6590–6599. tical excitability changes within 24 h of a seizure. Brain. 2009;132:
81. Pinault, D, O’Brien TJ. Cellular and network mechanisms of 1013–1021.
genetically-determined absence seizures. Thalamus Relat Syst. 94. Kalitzin SN, Parra J, Velis DN, et al. Quantification of unidirec-
2007;03(03):217–226. tional nonlinear associations between multidimensional signals.
82. Suffczynski P, Kalitzin S, Lopes Da Silva FH. Dynamics of non- IEEE Trans Biomed Eng. 2007;54(3):454–461.
convulsive epileptic phenomena modeled by a bistable neuronal 95. Osorio I, Frei MG. Real-time detection, quantification, warning,
network. Neuroscience. 2004;126(2):467–484. and control of epileptic seizures: the foundations for a scientific
83. Suffczynski P, Lopes da Silva FH, Parra J, et al. Dynamics of epilep- epileptology. Epilepsy Behav. 2009;16(3):391–396.
tic phenomena determined from statistics of ictal transitions. IEEE 96. Ullah G, Schiff SJ. Tracking and control of neuronal Hodgkin-
Trans Biomed Eng. 2006;53(3):524–532. Huxley dynamics. Phys Rev E Stat Nonlin Soft Matter Phys.
84. Suffczynski P, Kalitzin S, Lopes da Silva F, et al. Active paradigms 2009;79(4 Pt 1):040901.
of seizure anticipation: computer model evidence for necessity of 97. Velasco AL, Velasco F, Velasco M, et al. Electrical stimulation of the
stimulation. Phys Rev E Stat Nonlin Soft Matter Phys. 2008;78 hippocampal epileptic foci for seizure control: a double-blind,
(5 Pt 1):051917. long-term follow-up study. Epilepsia. 2007;48(10):1895–1903.
85. Bouwman BM, Suffczynski P, Lopes da Silva FH, et al. GABAergic
mechanisms in absence epilepsy: a computational model of
CHAPTER

Nonepileptic Attacks
MEGAN SELVITELLI, TRUDY PANG, DONALD L. SCHOMER, AND ERNST NIEDERMEYER 31
S
udden, brief loss of consciousness, sudden behavior and noted myoclonus in 90% of subjects, including multifocal
changes, or strange subjective sensations are not neces- and generalized myoclonus (3). Other involuntary movements
sarily epileptic. This should be a truism, but there is a were also witnessed, including head turning, lip-smacking,
present trend to ascribe too many transient conditions to an chewing, and fumbling movements (79%); vocalizations
epileptic mechanism. Here the differential diagnosis of epilep- (40%); or righting movements (76%) (3). Similar involuntary
tic seizures is discussed briefly. Topics covered will include: movements were also described in 14/15 patients undergoing
tilt table testing for unexplained syncope (4). In these patients,
1. Circulatory insufficiency giving rise to acute cerebral
rigid flexed postures were observed with associated eye rolling
ischemia
and head extension, with an additional 30% of patients experi-
2. Psychogenic alterations
encing bilateral myoclonic rhythmic jerks and 27% experienc-
3. Movement disorders imitating epileptic seizures (often asso-
ing urinary incontinence. Thus, clinical description of events
ciated with loss of consciousness)
may be insufficient for accurate diagnosis of either syncope or
4. Paroxysmal disturbances in infants and children imitating
seizure. For a summary of these features, see Table 31-1.
epileptic seizures
Given the difficulties in clinically differentiating convulsive
5. Toxic–metabolic disturbances
syncope from epileptic convulsions, Sheldon et al. utilized
6. Disturbances related to sleep disorders (such as
symptom-based questionnaires to identify features more com-
narcolepsy–cataplexy complex and parasomnias)
mon in patients with diagnosed syncope or epilepsy. Epilepsy
7. Cerebrovascular disorders
was the more frequent etiology of loss of consciousness if the
8. Migraines
events occurred often, were triggered by stressors, or were asso-
The first four topics will be discussed in this chapter, while ciated with head deviation or unresponsiveness (5). Conversely,
toxic–metabolic disturbances, sleep disorders, cerebrovascular syncope was more frequently associated with diaphoresis before
disorders, and migraines are covered in Chapters 21, 41, 19, and or after the event or associated with events triggered by pro-
24, respectively. longed sitting or standing (5).
Despite clinical features that may help differentiate between
SYNCOPAL ATTACKS seizures and syncope, these two entities may rarely co-occur. In
this rare entity, rhythmic ictal discharges give rise to bradycar-
Syncope has been defined as a sudden brief loss of conscious- dia, leading to decreased cardiac output and syncope. Schuele et
ness due to a discrete episode of generalized cerebral ischemia; al. identified 10 patients with ictal asystole from a cohort of
it is hence a manifestation of acute insufficiency of the cerebral 6825 patients undergoing video-electroencephalography (EEG)
circulation (1). Syncope is frequently encountered and encom- monitoring for medically refractory partial seizures (6). These
passes up to 3% of emergency room evaluations and 6% of hos- 10 patients represented 0.27% of patients monitored and
pital admissions per year in the United States (2). As syncope demonstrated an initial ictal onset of 29.9 seconds prior to the
may result from benign causes such as vasovagal syncope to development of asystole (6). In these patients, loss of postural
more serious etiologies such as cardiac arrhythmias and auto- tone was more commonly seen late in the seizure. Schuele et al.,
nomic dysfunction, a thorough understanding of the symp- as well as other investigators, have not been able to convincingly
toms, pathophysiology, evaluation, and differentiation from identify a significant laterality to ictal asystole, although
seizures is warranted. involvement of the temporal lobes, particularly the insular cor-
Classic descriptions of syncope note prodromal symptoms, tex, and the orbitofrontal cortex has been suggested (7). The
followed by loss of consciousness and rapid resumption of exact frequency of ictal asystole is not known and is presumed
awareness shortly thereafter. Prodromal symptoms may include to be underreported, as the identification requires concomitant
blurry vision, decreased hearing, warmth, nausea, diaphoresis, EEG and EKG recording. Identification of ictal asystole is
pallor, lightheadedness, and generalized weakness. important, however, as it may contribute to sudden unex-
If cerebral perfusion is prolonged, as may occur with artifi- plained syncope in epilepsy. Treatment more commonly
cial maintenance of an upright posture, abnormal involuntary includes placement of a permanent pacemaker and medication
movements may be observed that may be misinterpreted as with antiepileptic drugs, with care taken to avoid antiepileptic
epileptic. Lempert et al. induced syncope in healthy volunteers medications that may contribute to arrhythmias.

659
660 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 31 . 1

List of Historical and Event Features that Differentiate Syncope and Seizure

Syncope Seizure
Preictal features
Diaphoresis More common Uncommon
Ictal features
Loss of consciousness Common Common, depends on
seizure type
Myoclonus Possible Possible
Frequency of ictus Rare Common
Precipitation by stress Uncommon More common
Head turning Uncommon More common
Occurrence after prolonged
sitting or standing More common Less common
Postictal features
Diaphoresis More common Uncommon

Etiologies output may be seen with obstructive cardiac disease, such as


Any decrease in cerebral perfusion may result in syncope. Various hypertrophic obstructive cardiomyopathy or aortic stenosis,
etiologies may include neurally mediated syncope, orthostasis, and may also be seen with low ejection fraction from prior
cardiac arrhythmias, or cardiac outflow obstructions. myocardial infarctions.
Vasovagal faints, carotid sinus hypersensitivity, and situa-
EEG in Syncopal Attacks
tional faints all are examples of neurally mediated syncope,
resulting from inappropriate bradycardia and hypotension. If clinical features alone do not help differentiate between syn-
Pressure on hypersensitive carotid sinus baroreceptors increases cope and seizures, an EEG and simultaneous EKG during the
vagal tone, causing hypotension or bradycardia, or both, thereby event may help clarify the diagnosis. Numerous EEG studies of
leading to cerebral hypoperfusion. Situational faints may occur syncope have revealed the following sequence: initial alpha
with physical pain, receipt of frightening news, defecation, mic- depression, followed by low-voltage fast activity, generalized
turition, or cough; these typically occur in healthy individuals high-amplitude theta, and then delta slowing occurring con-
with normal blood pressure and frequently consistent triggers. comitantly with the loss of consciousness. In general, a period of
Orthostasis may also cause syncope, due to either autonomic 10 seconds elapses before high-voltage delta waves appear. If the
dysfunction or decreased blood volume due to dehydration or loss of consciousness is prolonged ( 15 seconds), the EEG may
blood loss. Autonomic dysfunction may be seen with neurode- demonstrate electrocerebral flattening. The patient may experi-
generative disorders, such as Shy–Drager syndrome, Parkinson ence myoclonic jerks, with no corresponding epileptiform dis-
disease, or postural orthostatic syndrome. Alternatively, sys- charges (4,8). After the event, the EEG demonstrates recovery to
temic diseases such as diabetes, alcoholism, or amyloidosis may the normal background rhythms. Conversely, epileptic loss of
cause an autonomic neuropathy that results in orthostasis. In consciousness may reveal epileptiform discharges consistent
addition, medications that cause peripheral vasodilatation (e.g., with the seizure type, either focal or generalized.
nitroglycerin, beta blockers) may cause a relative decrease in
blood volume. BREATH-HOLDING ATTACKS OF EARLY
Finally, cardiac disease may also contribute to syncope, CHILDHOOD AS A SPECIAL FORM OF
through either arrhythmias or decreased cardiac output. SYNCOPE
Bradyarrhythmias, such as atrial fibrillation with slowed ven-
tricular response, atrioventricular block, and long QTc syn- Breath-holding attacks are related to syncope and occur rather
drome, and tachyarrhythmias, such as supraventricular frequently in older infants and small children (9). Emotional
tachycardia, ventricular tachycardia, or torsades de pointes, may stress, frustration, minor trauma, and crying trigger these
decrease cardiac output and thereby decrease cerebral perfusion attacks. Cyanotic spells typically begin at 6 to 18 months, result-
episodically. Conversely, a more persistent decrease in cardiac ing in breath-holding during expiration, with subsequent loss
Chapter 31 ■ Nonepileptic Attacks 661

of consciousness and flaccidity. These may be accompanied by NES is associated with improved outcomes and better NES event
increased tonic and clonic movements. In contrast, pallid spells control. Nonetheless, the differentiation between these entities is
occur in a similar age group, resulting in decreased tone, loss of often difficult based on historical features alone and is best diag-
pulse, and pallor, with recovery in less than 1 minute. Pallid nosed with video-EEG monitoring. Complicating the discrimi-
breath-holding is felt to be secondary to bradycardia or asys- nation between NES and epileptic seizures is their common
tole, resulting in decreased cerebral perfusion. Of note, breath- co-occurrence.
holding spells may be associated with iron deficiency and
respond to supplementation (10). The children almost always Differentiation of Nonepileptic
have normal EEG findings. Khurana et al. demonstrated that an Seizures from Epileptic Seizures
increased RR interval from baseline of 0.5 seconds during 10 Numerous researchers have attempted to find a diagnostic test
seconds of ocular compression with simultaneous EEG and that may easily differentiate between NES and epileptic seizures.
EKG recording helps differentiate breath-holding spells and Excellent reviews by Cragar and colleagues, as well as by Reuber,
syncope from epileptic events in children (11). provide a summary of differentiating features based on demo-
graphic variables, medical history, and seizure semiology (24,25).
PSYCHOGENIC ATTACKS Of note, patients with NES are more likely to be women, and
have a prior history of psychiatric treatment and a prior history
Concept of Nonepileptic Seizures of abuse or sexual abuse; however, men with NES frequently have
The symptomatology of the hysterical attack has been well a history of work-related problems or posttraumatic stress disor-
known since the work of Briquet (12) and Charcot (13). der. NES patients are typically older at the onset of the events and
Briquet’s approach to this problem has proved to be a more had been treated with fewer anticonvulsants than patients with
solid foundation for modern theories than the work of Charcot, epilepsy. With regard to seizure semiology, NES patients had
which is beset with iatrogenic artifacts. greater variability in behaviors during the events, greater fre-
Other concepts were essentially based on psychodynamic quency of pelvic thrusting and eye closure during the event.
mechanisms such as dissociation (14) and conversion (15–18). Reuber also cited commonly held beliefs that NES events often
The concept of Kretschmer with emphasis on “hyponoic” and manifest with gradual onset, excessive limb movements, often
“hypobulic” mechanisms warrants particular attention (19,20). asynchronous and asymmetric, with possible opisthotonus, side-
Hysterical seizures are associated with loss of impulse, usu- to-side head shaking, crying, resistance to eye opening, preserva-
ally in stressful situations. These attacks used to be attributed tion of pupillary light responses, and purposeful movements
solely (and by definition) to females; this clearly has been (25). Interestingly, there were no significant differences between
proved to be untrue, although female adolescents and young patients with NES or epileptic seizures with regards to age, years
adults are most often affected. of education, seizure frequency, or family history of epilepsy or
The term pseudoseizures denotes a conglomeration of non- psychiatric disorders. Similarly, there were no significant differ-
cerebral or nonepileptic attacks. It was introduced by Liske and ences between these groups regarding presence of urinary incon-
Forster, who were aware of the fact that many, but not all, of the tinence or tongue biting during the ictus. Epileptic seizures were
patients had psychogenic (hysterical) attacks (21). Unfortunately, typically of shorter duration, more commonly associated with
this term is not conducive to a differentiation of the nonepileptic injuries during the event, more frequently occurred during sleep,
attacks with certain distinctive features. and were associated with postictal confusion.
The term pseudoseizures has been more or less equated with More recent studies have similarly looked for historical and
psychogenic attacks. This may have been the result of a search event behaviors that may predict whether the events represent
for a euphemistic term since older terms such as hysterical or NES or epileptic seizures. Benbadis established that patients
psychogenic attacks have been thought to have a derogatory con- evaluated in an epilepsy clinic for refractory seizures were more
notation. Interestingly, the term pseudoseizures has been fading likely to exhibit NES if they had a history of fibromyalgia or
since 1990, while the term psychogenic seizures once again is chronic pain or had experienced a typical event in the waiting
being used more frequently. or examining room (26). Opherk and Hirsch found that during
By definition, nonepileptic seizures (NES) connote paroxys- staring spells, patients with NES had no change in heart rate,
mal events that behaviorally or symptomatically mimic epileptic while patients with complex partial seizures had a marked
seizures but are not associated with concomitant epileptiform tachycardia; a similar pattern was seen comparing generalized
activity on EEG. Presumably, these events are secondary to a tonic–clonic seizures with nonepileptic convulsive seizures
psychologic mechanism, with conversion disorder often being (27). Oliva and colleagues noted a low sensitivity (8%) but high
cited. The incidence of NES is variable and estimated between specificity (100%) for epileptic seizures when both oral lacera-
0.91 and 3.03/100,000 in the general population (22,23). Correct tions and urinary incontinence were observed during inpatient
diagnosis is often delayed in these patients, leading to increased video-EEG monitoring compared to their occurrence in NES
risk for iatrogenic complications of anticonvulsant use, delayed events (28). Postictally, whispering responses, partial motor
referral to psychiatric treatment, and potential exposure to responses to commands, and shallow, irregular, rapid breathing
unnecessary emergency therapies, including intubation and are more commonly seen with NES events (29,30). For a sum-
CNS-suppressant medications. Furthermore, earlier diagnosis of mary of these features, see Table 31-2.
662 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

Tabl e 31 . 2

List of Historical and Paroxysmal Event Characteristics That Discriminate Between Nonepileptic
Attacks and Epileptic Seizures

Nonepileptic Attacks Epileptic Seizures


Historical features
Age of onset Older Younger
Gender predominance Female None
History of abuse Common Less common
History of pain syndromes Common Uncommon
or event in waiting
or examining room
History of prior psychiatric treatment Common Less common
History of anticonvulsant therapy Typically fewer medication Typically more medication
trials and less polytherapy trials and greater polytherapy
Event features
Duration of event Longer (typically 2 minutes) Shorter (typically 1–2 minutes)
Sleep occurrence Uncommon Common
Ictal behaviors (pelvic thrusting, Common Uncommon
side-to-side head movements,
gradual onset, asymmetric/
asynchronous limb movements,
variable amplitude, crying)
Ictal heart rate Stable Tachycardia
Ictal urinary incontinence Uncommon Common
and tongue bite
Ictal eye closure Common Uncommon
Postictal confusion Uncommon Common
Postictal whisper or partial Common Not seen
motor responses
Postictal breathing patterns Often rapid, shallow, soft, Deep and prolonged inspiratory/
and irregular expiratory phases, regular and
loud, often with snoring

Electroencephalographic Observations sharply contoured normal background alpha rhythms, wicket


Given the variable clinical manifestations of NES, video-EEG spikes, hypnagogic hypersynchrony, and hyperventilation-
monitoring is considered the gold standard for diagnosis of induced slowing. In a separate study, Reuber et al. performed
NES. However, video-EEG monitoring is a labor-intensive and both a retrospective analysis of interictal EEGs and a blinded,
costly procedure and numerous investigators have evaluated controlled investigation of interictal routine EEG abnormalities
alternative options for diagnosis of NES. in video-EEG-confirmed NES compared to age- and sex-
Interictal EEGs are the more commonly used modality in the matched healthy controls (32). In the retrospective analysis,
initial evaluation of patients with suspected seizures. In video- 53.8% of NES patients demonstrated nonspecific EEG abnor-
EEG-confirmed NES patients, up to 32% of patients had a prior malities while 12.3% demonstrated epileptiform discharges. In
history of an abnormal “epileptiform” EEG in one study (31). the prospective study, 9/50 NES patients demonstrated inter-
On visual inspection of 37% of these outside EEGs, none had mittent slow rhythms and 1 patient had sharp and slow-wave
epileptiform discharges. Normal EEG features that were over- activity. Comparatively, 5/50 controls also demonstrated inter-
stated as epileptiform abnormalities included fluctuations of mittent slow activity, with 1 patient demonstrating isolated left
Chapter 31 ■ Nonepileptic Attacks 663

temporal sharp waves. Thus, in the prospective study, there did similar ictal semiology with both events, making clinical dis-
not appear to be a difference in the frequency of interictal non- tinction challenging in these patients (38).
specific slowing or epileptiform discharges between the NES Thus, continued studies are needed to determine whether a
patients and control group. These studies would suggest that combination of historical variables, ictal and postictal behav-
the incidence of epileptiform discharges and nonspecific slow- iors, and interictal EEG abnormalities is helpful in the differen-
ing in NES patients is similar to that found in the normal pop- tiation of NES and epileptic seizures, while the gold standard of
ulation and implies that a normal EEG is more commonly seen diagnosis currently remains video-EEG demonstration of typi-
in patients with NES compared to epileptic seizures. cal ictal behaviors with absence of epileptiform abnormalities.
As interictal EEGs may also be normal in many patients with
epileptic seizures, the gold standard for diagnosis is video-EEG MOVEMENT DISORDERS IMITATING
monitoring. As inpatient monitoring involves high labor costs, EPILEPTIC SEIZURES
procedures to decrease monitoring time have been devised.
Benbadis et al. found that 1 to 2 hours of outpatient video-EEG Hyperkinetic movement disorders can often mimic focal motor
monitoring confirmed the diagnosis in suspected NES in 69% seizures and present a diagnostic dilemma in the clinical set-
of patients, when induction techniques including prolonged ting. However, the history and phenomenology of the abnormal
hyperventilation, photic stimulation, and verbal description of movements play a critical role in helping to differentiate the two
the events were used (33). Wassmer and colleagues were able to conditions. Clearly identifiable triggers and movements that are
induce NES with saline infusion and verbal suggestion in 62% not stereotyped are more suggestive of a movement disorder,
of patients with clinically suspected NES and 24 hours of non- rather than epilepsy. In addition, EEG recording while the
diagnostic video-EEG testing (34). Ethical considerations have patient is symptomatic is helpful as it is normal in movement
arisen with the use of induction tests, although the use of disorders. A variety of conditions and their distinction from
hyperventilation and photic stimulation, which are known to partial seizures are discussed below, but see a recent publication
induce seizures, appears to remove the deceit inherent in some for a more complete description of several of the disorders (40).
induction techniques (e.g., saline infusion, alcohol pad place- Chorea is derived from the Greek word chorea, meaning “a
ment, and use of tuning fork). dance.” It is a hyperkinetic disorder characterized by brief, jerky,
Nonetheless, video-EEG monitoring, without the use of nonrhythmic movements that involve various body parts in a
induction techniques, was demonstrated to confirm the diag- random fashion. Bilateral involvement is typically encountered.
nosis of either NES or epileptic seizures within 2 days of inpa- A wide variety of conditions affecting the basal ganglia may
tient monitoring in 88% of patients (35). result in chorea, but it is most often associated with Huntington
However, a small number of patients cannot be diagnosed by disease, Sydenham chorea after streptococcal infections, or
video-EEG monitoring due to ictal semiology (partial symp- strokes. Certain systemic disorders may also be associated with
toms or symptoms suggesting frontal lobe seizures that may chorea, such as hyperthyroidism, systemic lupus erythemato-
manifest without EEG alterations), absence of events during sus, primary antiphospholipid antibody syndrome, or paraneo-
monitoring, or obscuration of the EEG due to movement arti- plastic syndromes. Oral contraceptive use and pregnancy result
fact (36). Vinton et al. describe a technique in which the domi- in hormonal changes that have been recognized as a cause of
nant EEG frequency for 1- to 2-second epochs throughout the chorea. While choreiform movements are nonrhythmic, jerky,
episode was evaluated during NES and epileptic seizures (37). and brief, focal motor seizures are stereotyped, rhythmic move-
Interestingly, there was little variability of the dominant EEG ments and can affect a single body part or spread to other body
frequency during NES, suggesting that movement artifact asso- parts in a progressive fashion. When the pattern of progression
ciated with NES is constant throughout the event. Furthermore, follows the motor homunculus, this is known as the
the authors noted that NES events were associated with an “Jacksonian march.”
“on–off” pattern of intermittent and sudden variation between Athetosis consists of slow, continuous writhing movements
the dominant alpha rhythm and the movement artifact. involving the limbs or face (unilaterally or bilaterally), trunk,
Conversely, ictal EEG dominant frequencies evolved through- head, or tongue. It often results from lesions of the basal gan-
out the seizures in two dominant patterns. Thus, despite move- glia, due to stroke, tumor, demyelination, and infection, or it
ment artifact obscuring the underlying EEG abnormalities, may be drug-induced. Perinatal injury to the basal ganglia and
assessment of the dominant EEG frequency over time can dif- thalamus has been recognized as an etiology in the pediatric
ferentiate between NES and epileptic seizures. population. In many cases, athetosis occurs in conjunction with
chorea and this is known as choreoathetosis. The slow and
Co-occurrence of NES and Epilepsy writhing nature of the movements is quite distinct from focal
Aggravating the difficulties discriminating between NES and motor seizures that are generally fast and rhythmic.
epileptic seizures is their frequent co-occurrence. Various esti- Ballismus is another hyperkinetic movement disorder.
mates are suggested, 5% to 47%, with lower estimates seen in Ballismus or ballism is derived from the Greek word meaning
studies requiring video-EEG-confirmed NES and epilepsy and “to throw.” In contrast to focal seizures that are stereotyped,
higher estimates seen when epilepsy was presumed on the basis rhythmic, and predictable, ballismus is characterized by
of clinical history (38,39). Of note, Mari and associates noted rapid, violent, high-amplitude flinging or flailing limb move-
64% of patients with concomitant NES and epilepsy had ments that are unpredictable. The proximal muscles are
664 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

predominantly affected and the movements are usually unilat- drome demonstrate clear epileptiform discharges. On occasion,
eral, resulting in hemiballismus. It can be caused by a variety electromyography (EMG) may also be helpful in that the EMG
of conditions affecting the striatum or subthalamic nucleus, burst due to epileptic myoclonus is usually short, lasting less
including systemic processes, such as hyperglycemia, or than 50 msec, compared to a much longer duration of 200 to
structural lesions, such as strokes and tumor (41,42). 300 msec as seen in nonepileptic myoclonus (47). Tics represent
Medications have been known to cause ballismus as well, and an extremely heterogeneous group of disorders and consist of
the commonly described agents include phenytoin, levodopa, brief, intermittent, involuntary movements (motor tics) or
and oral contraceptives. sounds (phonic tics) that may be exacerbated by anxiety. They
Dystonia is another condition that may be confused with may also occur in normal individuals. Classically, patients expe-
partial seizures, and the correct diagnosis is important in rience an urge that builds and is relieved by performance of the
directing treatment. It is characterized by paroxysmal, sustained tic. Although tics can be temporarily suppressed, the urge can
co-contractions of the agonist and antagonist muscles of a body become so overwhelming with time that ultimately the tic can-
part resulting in twisting, repetitive movements, or abnormal not be suppressed. The myriad of etiologies include structural
postures (43). Although the movements are usually slow, they lesions, genetic abnormalities, psychiatric disorders, or strepto-
can also be quick and jerky, and may even have a rhythmic com- coccal infections (48,49). The disorder commonly begins in
ponent. The limbs are most often involved, but facial involve- childhood and peaks in adolescence. Complex motor tics such
ment has also been seen. Anxiety or stress can trigger or worsen as eye blinking may occasionally be confused with absence
the dystonia, but some patients have developed “sensory tricks,” seizures; however, patients do not have any alteration of aware-
which are maneuvers or positions that improve or minimize it. ness. Other complex motor tics such as head shaking, jumping,
Dystonia is caused by abnormal basal ganglia function and is and obscene gestures are less likely to be confused with seizures,
felt to be associated with dysfunction of the inhibitory in that they are usually not stereotyped and can be briefly sup-
interneuronal circuits in the brainstem and spinal cord, as well pressed by suppressing the urge to tic. The EEG during the
as thalamic control of cortical motor planning and execution attacks is normal.
(44). Patients can present with primary dystonia or secondary Hemifacial spasm is characterized by irregular contractions
dystonia, due to stroke, tumor, neurodegenerative processes, of the facial muscles and may be mistaken for focal motor
medications, and various toxic substances. seizures. It is typically on one side of the face, although bilateral
Myoclonus is one of the most frequently encountered move- involvement has also been reported (50). It results from spon-
ment disorders, characterized by sudden, brief involuntary taneous firing of the facial nerve, which may occur sporadically
movements, due to either muscle contractions, resulting in pos- or with vascular compression of the nerve after it exits from the
itive myoclonus, or sudden muscle inhibition, resulting in neg- brainstem, and, very infrequently, with a demyelinating brain-
ative myoclonus. Myoclonus is referred to as segmental when it stem plaque due to multiple sclerosis. An association with a past
is restricted to certain muscle groups or body segments, but it history of Bell’s palsy or injury to the facial nerve has also been
can be diffuse and involve the entire body. Since there are reported, in which synkinesis of the facial muscles due to aber-
numerous causes of myoclonus, it is commonly classified into rant regeneration of the facial nerve may sometimes mimic
the following four major groups: (1) physiologic, such as hic- focal motor seizures. Differentiating between hemifacial spasm
cups; (2) essential, also known as hereditary or idiopathic; (3) and focal motor seizures based on EEG alone may be difficult
symptomatic, with an underlying abnormality (toxic, meta- since focal motor seizures involving a small area of the cerebral
bolic, infectious, anoxic causes); (4) epileptic, with myoclonus cortex may not have a surface EEG correlate. Thus, a detailed
as part of an epileptic syndrome such as juvenile myoclonic history and brain imaging will play an important role in the
epilepsy. Physiologic myoclonus is considered to be the most diagnostic process.
benign form. Essential myoclonus, also known as benign essen- Paroxysmal kinesigenic dyskinesia (PKD), a hyperkinetic
tial myoclonus, is an inherited, autosomal dominant disorder in disorder, is characterized by recurrent attacks of any combina-
which myoclonic jerks affect the arms or trunk and there may tion of movements, including dystonic posture, chorea, atheto-
be associated dystonia (45,46). In symptomatic myoclonus, sis, and ballism (51). The movements may be unilateral or
there is an identifiable underlying cause, such as a systemic dis- bilateral, and are usually brief, lasting seconds, and always less
turbance, as seen in infections and toxic or metabolic derange- than 5 minutes. A number of triggers have been reported,
ments, or it may be a manifestation of a variety of neurologic including sudden movement, hyperventilation, and startle. The
conditions, including neurodegenerative disorders, strokes, or primary form carries both an autosomal dominant and an
injuries to the spinal cord (spinal segmental myoclonus). In sys- autosomal recessive pattern of inheritance and the locus has
temic disturbances or neurodegenerative disorders, been mapped to chromosome 16 (52,53). The secondary form
encephalopathy is the predominant feature and seizures may may result from other neurologic disorders such as multiple
occur. Finally, last category of myoclonus, epileptic myoclonus, sclerosis (54). There is an association with infantile generalized
is a feature that may be seen in a wide variety of seizure types, convulsions in patients with familial PKD (55), while some
for example, juvenile myoclonic epilepsy. To distinguish reported cases by Hirata et al. and Lombroso demonstrated
between epileptic and nonepileptic myoclonus, EEG is an rhythmic discharges during attacks (56,57). Sadamatsu et al.
essential tool since nonepileptic myoclonus does not have any performed video-EEG monitoring in two patients with classic
EEG correlate, whereas those with an underlying epilepsy syn- PKD and did not find ictal discharges during attacks (58). The
Chapter 31 ■ Nonepileptic Attacks 665

clinical cases presented by Lombroso and Sadamatsu may be startle response followed by tonic contraction (usually asym-
distinct as the former case involved a patient with mental retar- metric); patients may fall and have clonic jerks as well. These
dation and the attacks were precipitated loud noises and men- patients typically have a neurologic deficit such as hemiparesis
tal stress in addition to movement, whereas the latter cases by and the weak side is typically involved in the seizure. The EEG
Sadamatsu involved developmentally normal patients who is very helpful in distinguishing the two disorders as the EEG in
never developed generalized convulsions after PKC attacks; hyperekplexia is normal, while the EEG abnormality in startle
thus, Sadamatsu concluded that PKC in their patients is distinct epilepsy usually involves the supplementary motor area; thus,
from reflex epilepsy. Paroxysmal nonkinesigenic dyskinesia frontal, central, or frontocentral spikes can be seen (69).
(PNKD) is similar to PKD, but as the name implies, it is not Startle disease should be distinguished from another syn-
triggered by movement. Coffee, alcohol, hunger, fatigue, and drome known as the “Jumping Frenchmen of Maine” in which
tobacco have been reported as precipitating factors (59). It typ- excessive startle reflex is a key feature. It was first described by
ically lasts longer, on the order of minutes to hours rather than Beard, in a group of French-Canadian lumbermen in the
seconds to minutes and consciousness is preserved. A number Moosehead Lake region of Maine (70). Patients with this disor-
of etiologies have been implicated, including psychogenic cere- der have the characteristic startle, but have additional features,
brovascular disease, multiple sclerosis, encephalitis, cerebral including echolalia and echopraxia, which are thought to be tic
trauma, peripheral trauma, migraine, and kernicterus (60). related, and again the EEG does not demonstrate epileptiform
Tardive dyskinesia and tardive dystonia are two movement activity. The underlying molecular basis is unknown.
disorders commonly associated with dopamine antagonists,
such as antipsychotics, antiemetics, and even antidepressants.
While tardive dyskinesia refers to choreiform movements of the PAROXYSMAL DISTURBANCES IN
mouth, tongue, head, neck, and limbs, tardive dystonia refers to INFANTS AND CHILDREN IMITATING
dystonic posturing or twisting of body parts as a result of sus- EPILEPTIC SEIZURES
tained muscle contractions (61). The first step in the treatment
of these conditions is removing the offending agent; however, Hyperekplexia, also known as startle disease or stiff baby syn-
sometimes additional pharmacologic treatment is required to drome, is described in the section “Movement Disorders
control or improve the symptoms. The history, clinical appear- Imitating Epileptic Seizures.”
ance of the movements, and normal EEG help to distinguish Shuddering attacks in children are brief, fast tremors of the
the two disorders from epilepsy. head, shoulder, or trunk, lasting seconds, and often occur dur-
Hyperekplexia (startle disease) is a rare hereditary condi- ing eating. The condition begins in infancy or childhood and
tion, caused by an abnormal gene for the subunit of glycine resolves spontaneously by the teenage years. Neurologically and
receptor on chromosome 5q31.2 (62,63), and follows an auto- developmentally, the affected children are normal, and the EEG
somal dominant pattern of inheritance. Less commonly, auto- is normal during these attacks (71). There is an apparent asso-
somal recessive cases have also been reported (64). Excessive ciation with a family history of essential tremor (72).
and persistent startle to sudden auditory, visual, or sensory Opisthotonos consists of intermittent generalized body stiff-
stimuli is the hallmark feature of this condition (65,66). It typ- ening or extension posturing. This may be due to a gastroe-
ically begins in infancy causing stiff baby syndrome, as sudden sophageal reflux disease known as Sandifer syndrome, which
stimuli can trigger leg or body stiffening that are sometimes may be mistaken for tonic seizures. These spells tend to occur
severe enough to cause apnea (67). Affected babies demonstrate within 30 minutes of a feeding and children may exhibit apnea,
the head retraction reflex, whereby gentle tapping of the nose or staring, and minor jerking of the extremities during them. The
forehead results in brisk retraction of the head that does not clear association with feedings is a good indicator of the diag-
habituate with repetition. In children, sudden stimuli or strong nosis and the EEG during these spells is normal (73).
emotions such as excitement or fear can trigger a typical attack, Repetitive movements known as stereotypies refer to various
resulting in falls with preserved awareness. At night, patients stereotypic movements such as head banging, body rocking,
may also experience spontaneous clonus lasting a few minutes and hand flapping in children. Although these may be seen in
in lower extremities (65,66). normal children, they are commonly encountered in children
The clinical features of hyperekplexia may resemble startle with neurologic deficits, such as mental retardation and autism.
epilepsy, which also can present with falls and body stiffening. The behaviors are thought to be “self-stimulatory” for the pur-
Thus, it is important to differentiate hyperekplexia from the pose of relaxation and can be present during the waking state or
epileptic disorder. To compound the confusion, treatment of during transition to sleep.
hyperekplexia consists of clonazepam or valproate that has Spasmus nutans consists of a triad of clinical symptoms of
been shown to be effective for symptomatic relief (65,68). recurrent monocular nystagmus, head nodding, and head tilt,
However, as hyperekplexia is typically an inherited condition, which may be mistaken for seizures. The typical age of onset is
particularly in infants and children, a positive family history is 4 months and often resolves within a few years. The exact
often present and examination of the parents aids the diagno- underlying cause is still unknown and the EEG during these
sis. Startle epilepsy is characterized by brief seizures, typically spells is normal.
less than 30 seconds, triggered by unexpected auditory or tactile Daydreaming is a common phenomenon in children which
stimuli, usually a sudden sound (69). The seizures consist of a may be mistaken for absence or complex partial seizures. Unlike
666 Part IV ■ Clinical EEG in Epilepsy and Related Disorders

seizures, during these spells, children can respond to questions 23. Szaflarski JP, Ficker DM, Cahill WT, et al. Four-year incidence of
appropriately when their attention is redirected and there are psychogenic nonepileptic seizures in adults in Hamilton county,
no postictal symptoms such as confusion or lethargy. OH. Neurology. 2000;55:1561–1563.
24. Cragar DE, Berry DTR, Fakhoury TA, et al. A review of diagnostic
techniques in the differential diagnosis of epileptic and nonepilep-
REFERENCES tic seizures. Neuropsychol Rev. 2002;12:31–64.
25. Reuber M. Psychogenic nonepileptic seizures: answers and ques-
1. Naquet R, Vigouroux RA. Acute cerebral anoxia and syncopal
tions. Epilepsy Behav. 2008;12:622–635.
attacks. In: Remond A, ed. Handbook of Electroencephalography
26. Benbadis SR. A spell in the epilepsy clinic and a history of “chronic
and Clinical Neurophysiology. Vol 14A. Amsterdam: Elsevier;
pain” or “fibromyalgia” independently predict a diagnosis of psy-
1972:68–71.
chogenic seizures. Epilepsy Behav. 2005;6:264–265.
2. Sarasin FP, Louis-Simonet M, Carballo D, et al. Prospective evalu-
27. Opherk C, Hirsch LJ. Ictal heart rate differentiates epileptic from
ation of patients with syncope: a population-based study. Am
non-epileptic seizures. Neurology. 2002;58:636–638.
J Med. 2001;111:177–184.
28. Oliva M, Pattison C, Carino J, et al. The diagnostic value of oral
3. Lempert T, Bauer M, Schmidt D. Syncope: a videometric analysis
lacerations and incontinence during convulsive “seizures”.
of 56 episodes of transient cerebral hypoxia. Ann Neurol.
Epilepsia. 2008;49:962–967.
1994;36:233–237.
29. Chabolla DR, Shih JJ. Postictal behaviors associated with psy-
4. Ammirati F, Colivicchi F, Di Battista G, et al. Electro -
chogenic nonepileptic seizures. Epilepsy Behav. 2006;9:307–311.
encephalographic correlates of vasovagal syncope induced by
30. Azar NJ, Tayah TF, Wang L, et al. Postictal breathing pattern distin-
head-up tilt testing. Stroke. 1998;29:2347–2351.
guishes epileptic from nonepileptic convulsive seizures. Epilepsia.
5. Sheldon R, Rose S, Connolly S, et al. Diagnostic criteria for vaso-
2008;49:132–137.
vagal syncope based on a quantitative history. Eur Heart J. 2006;27:
31. Benbadis SR, Tatum WO. Overinterpretation of EEGs and misdi-
344–350.
agnosis of epilepsy. J Clin Neurophysiol. 2003;20(1):42–44.
6. Schuele SU, Bermeo AC, Alexopoulos AV, et al. Video-electrographic
32. Reuber M, Fernandez G, Bauer J, et al. Interictal EEG abnormali-
and clinical features in patients with ictal asystole. Neurology. 2007;
ties in patients with psychogenic nonepileptic seizures. Epilepsia.
69:434–441.
2002;43(9):1013–1020.
7. Carinci V, Barbato G, Baldrati A, et al. Asystole induced by partial
33. Benbadis SR, Seigrist K, Tatum WO, et al. Short-term outpatient
seizures: a rare cause of syncope. Pacing Clin Electrophysiol. 2007;30:
EEG with induction in the diagnosis of psychogenic seizures.
1416–1419.
Neurology. 2004;63:1728–1730.
8. Gastaut H, Fischer-Williams M. Electro-encephalographic study
34. Wassmer E, Wasmer SR, Donati F. Saline infusion: a diagnostic and
of syncope; its differentiation from epilepsy. Lancet. 1957;
therapeutic tool in nonepileptic attacks? Epilepsy Behav. 2003;4:
273(7004):1018–1025.
500–506.
9. Fejerman N. Nonepileptic disorders imitating generalized idio-
35. Lobello K, Morgenlander JC, Radtke RA, et al. Video/EEG moni-
pathic epilepsies. Epilepsia. 2005;46(suppl 9):80–83.
toring in the evaluation of paroxysmal behavioral events: dura-
10. Mackay M. Fits, faints and funny turns in children. Aust Fam
tion, effectiveness and limitations. Epilepsy Behav. 2006;8:261–266.
Physician. 2005;34:1003–1008.
36. Benbadis SR. The EEG in nonepileptic seizures. J Clin
11. Khurana DS, Valencia I, Kruthiventi S, et al. Usefulness of ocular
Neurophysiol. 2006;23:340–352.
compression during electroencephalography in distinguishing
37. Vinton A, Carino J, Vogrin S, et al. “Convulsive” nonepileptic
breath-holding spells and syncope from epileptic seizures. J Child
seizures have a characteristic pattern of rhythmic artifact distin-
Neurol. 2006;21:907–910.
guishing them from convulsive epileptic seizures. Epilepsia.
12. Briquet P. Traité de l’Hystérie. Paris; 1859.
2004;45:1344–1350.
13. Charcot JM. Leçons du Mardi à la Salpêtrière, Policlinique Paris:
38. Mari F, Di Bonaventura C, Vanacore N, et al. Video-EEG study of
Bureaux du Progrès Médical. Paris: Lecroisnier and Babe;
psychogenic nonepileptic seizures: differential characteristics in
1887–1889.
patients with and without epilepsy. Epilepsia. 2006;47(S5):64–67.
14. Janet P. L’état Mental des Hystériques. Paris; 1893/1894.
39. Martin R, Prasad BA, Powell T, et al. Frequency of epilepsy in
15. Breuer J. Studien über Hysterie. 2nd ed. Vienna: Deuticke; 1895.
patients with psychogenic seizures monitored by video-EEG.
16. Breuer J, Freud S. Studies on hysteria. In: Standard Edition of the
Neurology. 2003;61:1791–1792.
Complete Psychological Works of Sigmund Freud. Vol 7. London:
40. Schmitz B, Tettenborn B, Schomer DL. The Paroxysmal Disorders.
Hogarth; 1895, 1955.
2nd ed. Cambridge: Cambridge University Press; 2010.
17. Chodoff P, Lyons H. Hysteria, the hysterical personality and “hys-
41. Lee MS, Marsden CD. Movement disorders following lesions of
terical” conversion. Am J Psychiatry. 1958;114:734.
the thalamus or subthalamic region. Mov Disord. 1994;9:493–507.
18. Ziegler F, Imboden J, Meyer E. Contemporary conversion reac-
42. Lai PH, Tien RD, Chang MH, et al. Chorea–ballismus with nonke-
tions: a clinical study. Am J Psychiatry. 1960;116:901.
totic hyperglycemia in primary diabetes mellitus. AJNR Am
19. Kretschmer E. Hysterie, Reflex und Instinkt. 6th ed. Stuttgart:
J Neuroradiol. 1996;17:1057–1064.
Thieme; 1923, 1958.
43. Jankovic J, Tolosa E. Dystonic disorders. In: Parkinson’s Disease and
20. Kretschmer E. Medizinische Psychologie. 11th ed. Stuttgart:
Movement Disorders. 4th ed. Philadelphia, PA: Lippincott Williams
Thieme; 1956.
& Wilkins; 2002:14,721.
21. Liske E, Forster FM. Pseudoseizures: a problem in the diagnosis
44. Berardelli A, Rothwell JC, Hallett M, et al. The pathophysiology of
and management of epileptic patients. Neurology. 1964;14:41–49.
primary dystonia. Brain. 1998;121:1195–1212.
22. O’Sullivan SS, Spillane JE, McMahom EM, et al. Clinical characteris-
45. Mahloudji M, Pikielny RT. Hereditary essential myoclonus. Brain.
tics and outcome of patients diagnosed with psychogenic nonepilep-
1967;90:669–674.
tic seizures: a 5-year review. Epilepsy Behav. 2007;11:77–84.
Chapter 31 ■ Nonepileptic Attacks 667

46. Vidailhet M, Tassin J, Durif F, et al. A major locus for several phe- 61. Burke RE, Fahn S, Jankovic J, et al. Tardive dystonia: late-onset and
notypes of myoclonus-dystonia on chromosome 7q. Neurology. persistent dystonia caused by antipsychotic drugs. Neurology.
2001;56:1213–1216. 1982;32:1335–1346.
47. Hallett M. Myoclonus: relation to epilepsy. Epilepsia. 1985;26: 62. Ryan SG, Sherman SL, Terry JC, et al. Startle disease, or hyperek-
S67–S77. plexia: response to clonazepam and assignment of the gene (STHE)
48. Kumar R, Lang AE. Tourette syndrome. Secondary tic disorders. to chromosome 5q by linkage analysis. Ann Neurol. 1992;31:
Neurol Clin. 1997;15:309–331. 663–668.
49. Swedo SE, Leonard HL, Garvey M, et al. Pediatric autoimmune 63. Shiang R, Ryan SG, Zhu YZ, et al. Mutations in the alpha 1 subunit
neuropsychiatric disorders associated with streptococcal infec- of the inhibitory glycine receptor cause the dominant neurologic
tions: clinical description of the first 50 cases. Am J Psychiatry. disorder, hyperekplexia. Nat Genet. 1993;5:351–358.
1998;155:264–271. 64. Rees MI, Andrew M, Jawad S, et al. Evidence for recessive as well as
50. Tan EK, Jankovic J. Bilateral hemifacial spasm: a report of five dominant forms of startle disease (hyperekplexia) caused by
cases and a literature review. Mov Disord. 1999;14:345–349. mutations in the alpha 1 subunit of the inhibitory glycine recep-
51. Jankovic J, Demirkiran M. Classification of paroxysmal dyskine- tor. Hum Mol Genet. 1994;3:2175–2179.
sias and ataxias. Adv Neurol. 2002;89:387–400. 65. Andermann F, Keene DL, Andermann E, et al. Startle disease or
52. Bennett LB, Roach ES, Bowcock AM. A locus for paroxysmal kine- hyperekplexia: further delineation of the syndrome. Brain.
sigenic dyskinesia maps to human chromosome 16. Neurology. 1980;103:985–997.
2000;54:125–130. 66. Kurczynski TW. Hyperekplexia. Arch Neurol. 1983;40:246–248.
53. Goodenough DJ, Fariello RG, Annis BL, et al. Familial and 67. Cioni G, Biagioni E, Bottai P, et al. Hyperekplexia and stiff-baby
acquired paroxysmal dyskinesias. A proposed classification with syndrome: an identical neurological disorder? Ital J Neurol Sci.
delineation of clinical features. Arch Neurol. 1978;35:827–831. 1993;14:145–152.
54. Tranchant C, Bhatia KP, Marsden CD. Movement disorders in 68. Dooley JM, Andermann F. Startle disease or hyperekplexia: adoles-
multiple sclerosis. Mov Disord. 1995;10:418–423. cent onset and response to valproate. Pediatr Neurol. 1989;5:
55. Swoboda KJ, Soong B, McKenna C, et al. Paroxysmal kinesigenic 126–127.
dyskinesia and infantile convulsions: clinical and linkage studies. 69. Aguglia U, Tinuper P, Gastaut H. Startle-induced epileptic
Neurology. 2000;55:224–230. seizures. Epilepsia. 1984;25:712–720.
56. Hirata K, Katayama S, Saito T, et al. Paroxysmal kinesigenic 70. Beard GM. Remarks upon ‘jumpers or jumping Frenchmen’. J Nerv
choreoathetosis with abnormal electroencephalogram during Ment Dis. 1878;5:526.
attacks. Epilepsia. 1991;32:492–494. 71. Holmes GL, Russman BS. Shuddering attacks. Evaluation using
57. Lombroso CT. Paroxysmal choreoathetosis: an epileptic or non- electroencephalographic frequency modulation radiotelemetry
epileptic disorder? Ital J Neurol Sci. 1995;16:271–277. and videotape monitoring. Am J Dis Child. 1986;140:72–73.
58. Sadamatsu M, Masui A, Sakai T, et al. Familial paroxysmal kinesi- 72. Vanasse M, Bedard P, Andermann F. Shuddering attacks in chil-
genic choreoathetosis: an electrophysiologic and genotypic analy- dren: an early clinical manifestation of essential tremor. Neurology.
sis. Epilepsia. 1999;40:942–949. 1976;26:1027–1030.
59. Bruno MK, Lee HY, Auburger GW, et al. Genotype–phenotype 73. Kabakus N, Kurt A. Sandifer syndrome: a continuing problem of
correlation of paroxysmal nonkinesigenic dyskinesia. Neurology. misdiagnosis. Pediatr Int. 2006;48:622–625.
2007;68:1782–1789.
60. Demirkiran M, Jankovic J. Paroxysmal dyskinesias: clinical fea-
tures and classification. Ann Neurol. 1995;38:571–579.
Part V Complementary and Special Techniques
CHAPTER
Nasopharyngeal, Anterotemporal,
and Sphenoidal Electrodes
ANDRES M. KANNER, TRAVIS STOUB, AND STEVE BILD
32
INTRODUCTION discharges that were not recorded with the standard temporal
electrodes included in the 10-20 system. The noninvasive and
Temporal lobe epilepsy (TLE) is the most frequent type of user-friendly placement of ATE led to their use in most epilepsy
epilepsy of focal origin affecting adults. It is a heterogeneous centers. Yet, in the last two decades an ongoing debate has been
type of epilepsy as the epileptogenic zone can be restricted to taking place on whether SE and NPE add any additional data to
mesial temporal structures and/or involve temporal lateral neo- those yielded by ATE. The purpose of this chapter is to review
cortex. Accordingly, the yield of detection of epileptiform activ- the conditions in which SE, NPE, and ATE should be used in
ity with scalp electrodes depends on the following variables: (i) patients suspected of suffering from TLE of mesial-temporal
the source and extent of the epileptogenic area relative to the origin.
position of the scalp electrodes, (ii) the electric field generated
by the epileptiform activity and the orientation of the electric
vectors, and (iii) the extent of propagation of the epileptiform TECHNICAL ASPECTS
activity from mesial to temporal lateral regions. For example,
epileptiform discharges originating in temporal lateral neocor- Sphenoidal Electrodes
tex would be readily recorded with the standard 10-20 midtem- Jones was the first to report the use of SE in 1950 (5). At that
poral scalp electrodes. On the other hand, such electrodes may time, the electrodes consisted of a fine insulated needle electrode
often fail to detect epileptiform activity of mesial temporal ori- inserted under local anesthesia. The point of entry was under the
gin. For one, the amygdala nucleus is not a laminar structure zygomatic arch through the mandibular notch and targeted a
and, therefore, it generates very restricted electrical field poten- position under the base of the skull lateral to the foramen ovale
tials. Likewise, the hippocampus, with its ‘‘double C shape,’’ (FO). In 1957, Marshall introduced SE made of flexible wire ini-
appears as a closed loop electrically and generates epileptiform tially made of stainless steal to allow for longer periods of recor-
discharges with a vertical dipole that could only be detected by ding (6), particularly during presurgical EEG monitoring studies,
frontotemporal and temporal lateral electrodes if the epilepti- which over the following two decades underwent further modifi-
form activity were to propagate to those regions. cations. More recently, SE consisted of multistranded silver and
Several studies have shown that the standard 10-20 interna- platinum wires, the latter to avert its lateral displacement (7).
tional system of scalp electrode placement may often fail to Typically, the insertion of SE is carried out at the bedside,
identify epileptiform activity of mesial-temporal lobe origin under local anesthesia with lidocaine. The SE wire is mounted
(1–4). Indeed, the overall findings of these studies suggest that on a #22-gauge lumbar puncture needle and the recording end
the lateral and anterior portion of the temporal lobe is not ade- is inserted into the lumen of the needle. The needle is inserted
quately covered using the standard 10-20 system. In the 10-20 through the skin at a point 3 cm anterior to the external audi-
system, electrodes F7, F8, T7, T8 (also known as T3-T4), and tory canal, beneath the zygomatic arch, and through the
A1-A2 have been used to record anterior temporal electrical mandibular notch oriented in a 10 posterior direction target-
activity. Yet, electrodes F7 and F8 are positioned over the infe- ing the lateral border of the FO (Fig. 32.1). The tip of the nee-
rior frontal gyrus and the rostral pars triangularis (1), while dle is advanced to a depth of 4 to 5 cm beneath the skin’s surface
electrodes T7 and T8 are positioned over the middle and supe- or until the patient reports mandibular pain (8).
rior temporal gyri posterior to the rolandic fissure (1). Hence, Yet, the blind insertion of SE can have several problems that
the angle subtended by these electrodes relative to the mesial- may negate any advantage of their use. First, when SE are
temporal source of the epileptiform discharges may often be inserted blindly, the recording tips may end up in a position at
inadequate (lest it propagates to temporal lateral regions). a distance from the FO, either anterior or posterior to it.
This problem was recognized in the first half of the 20th cen- Recordings from SE positioned posterior to the FO are ham-
tury, which led to the development of sphenoidal electrodes (SE) pered by recording through the thick (2 cm) petrous pyramid,
and nasopharyngeal electrodes (NPE). Later on, anterotemporal while a position anterior or lateral to the foramen has the dou-
(ATE), minisphenoidal (MSE), and basal-temporal (scalp) elec- ble disadvantage of recording through bone and at an increased
trodes (BTE) started to be used in an attempt to avert the incon- distance from target mesial-temporal structures. To overcome
venience and discomfort associated with the insertion of SE. this potential problem, Kanner et al. suggested the insertion of
Studies carried out in the 1980s and 1990s reported that SE SE under fluoroscopic guidance (Fig. 32.2, left and right panels)
and ATE identified a significantly greater number of epileptiform (9,10). This technique ensured that the recording tip is

669
670 Part V ■ Complementary and Special Techniques

Figure 32.1 Phantom image of the head showing a


22-gauge carrier needle entering the skin (concentric target
lines, S) in front of the condylar process of the mandible
passing under the zygomatic arch (Z) and through the
mandibular notch (MN), en route to V3 emerging from the
foramen ovale (FO, seen on the edge). PP denotes the
pterygoid plate. The SE that is mounted on the superior
surface of the needle is not shown. (Reprinted with permis-
sion from Kanner AM, Ramirez L, Jones JC. The utility of
placing electrodes under the foramen ovale with fluoro-
scope guidance. J. Clin. Neurophysiol. 1995;12:72–81.)

positioned inferior to FO (Fig. 32.3). Furthermore, the variable Minisphenoidal Electrodes


shape of the sphenoid wing among patients can result in an MSE were introduced by Laxer in 1984 as an alternative to SE (11)
obstacle to the insertion of the needle when done blindly, which in a study of 100 patients, 34 of whom had epileptiform activity of
in turn results in additional discomfort to patients. The inser- temporal lobe origin. In 21 of these patients, the epileptiform
tion under fluoroscopic guidance averts this problem. On the activity was only detected by MSE. MSE consist of stainless steel
other hand, the higher costs associated with the insertion of SE 0.5 cm needles inserted under the zygomatic arch, yet the inser-
under fluoroscopic guidance is a disadvantage of this insertion tion of these electrodes is associated with less discomfort than that
technique. of SE (12). In a study of 100 patients, Buchhalter et al. found that

Figure 32.2 Left panel: Submental-vertex radiograph shows the normal relationship of the FO (open arrow)
to the foramen spinosum (arrowhead). The electrode/ needle system (solid arrow) is positioned anterior and
slightly lateral to the FO. (Reprinted with permission from Kissani N, Alarcon G, Dad M, et al. Sensitivity of
recordings at sphenoidal electrode site for detecting seizure onset: evidence from scalp, superficial and deep
foramen ovale recordings. Clin Neurophysiol. 2001;112:232–240.) Right panel: Submental-vertex radiograph
shows the position of both SE wires (open arrows) anterior and lateral to their respective FO (solid arrows).
(Reprinted with permission from Fenton DS, Geremia GK, Dowd AM, et al. Precise placement of sphenoidal
electrodes via fluoroscopic guidance. Am J Neuroradiol. 1997;18:776–778.)
Chapter 32 ■ Nasopharyngeal, Anterotemporal, and Sphenoidal Electrodes 671

Figure 32.3 Coronal CT through FO, showing the rela-


tion between the SE (E) and the inferomesial temporal
lobe. M, mandible; A, amygdala; P, parahypocampal
gyrus; OT, occipitotemporal gyrus; I, inferior temporal
gyrus; V, third ventricle; NP, nasopharynx; L, lentiform
nucleus.

MSE and SE yielded comparative data in nine patients; however, electrical activity of anterotemporal origin (17). These ATE,
SE identified interictal epileptiform activity not detected by MSE labeled T1 and T2, are positioned “one centimeter above a point
in six patients (12). On the other hand, other investigators, how- one third of the distance along a line from the external auditory
ever, failed to find any advantage of MSE to ATE (3,13). meatus to the outer canthus of the ipsilateral eye.” A disadvan-
tage of T1 and T2 is that their locations are not proportional to
Nasopharyngeal Electrodes 10-20 or 10-10 system positions and thus display dispropor-
In 1948, Roubicek and Hill (14) introduced NPE, and their tionate voltages when utilized in bipolar derivations (Fig. 32.5).
increased yield of localization of epileptiform activity of mesial Accordingly, referential derivations may be preferable when
temporal origin was confirmed by several authors (3,14). using these electrodes (Figs. 32.6 and 32.7).
These electrodes consist of a solid silver rod with a small silver Homan et al. reported a study of 80 patients in whom a total
ball at the tip (Fig. 32.4) of 0.67 and 0.055 inches in diameter of 58 epileptiform foci were found. ATE significantly improved
for use in adults and children, respectively (15,16). The shaft is
flexible and malleable and is insulated up to the ball tip with
six or seven coats of insulex varnish. The distal 1 inch is angled
about 20 , but such angle can be changed to fit any nasal pas-
sage. Unlike SE, NPE can be inserted by EEG technologists
through the nasal cavity to target the posterior pharyngeal
wall. When positioned optimally, the ball tip is expected to lie
in an area beneath the foramen lacerum.

Scalp Anterotemporal and Basal-Temporal


Electrodes
In 1960, Silverman suggested a position for scalp electrodes
outside of the 10-20 international system aimed at recording

Figure 32.5 Position of ATE and BTE within the 10-10 electrode
Figure 32.4 Picture of nasopharyngeal electrodes. placement system.
672 Part V ■ Complementary and Special Techniques

Figure 32.6 Interictal recordings


of a patient with a partial seizure
disorder of right mesial temporal
origin. Interictal spikes are detected
only at SP2 in the bipolar montage,
while in the referential montages
sharp transients can be identified at
T2 and T4 electrodes in the first
two discharges.

Figure 32.7 Ictal recordings of a


patient with a partial seizure disorder
of right mesial temporal origin. The
ictal onset was identified 6 seconds
at SP2 before it was recorded at ATE.
Chapter 32 ■ Nasopharyngeal, Anterotemporal, and Sphenoidal Electrodes 673

detection of the epileptiform activity of mesial-temporal origin of longer electrode–generator distances and the effect of bone
compared to the standard temporal electrodes (2). Other attenuation. In such circumstances, recordings obtained with SE
authors confirmed these observations (3). positioned at a distance from the FO would look the same as
In addition to T1 and T2 electrodes, scalp BTE that are part ATE/BTE recordings. In addition, SE placed anterior to the FO
of the 10-10 system have been used with increasing frequency and ATE/BTE yield comparable subtended angles to mesial-basal
to record epileptiform activity of mesial-temporal origin. Each generators and, hence, epileptiform activity would not differ at
BTE is located in the coronal plane defined by their superior either electrode, no matter the extent of its electric field.
counterparts (i.e., F9 and F10 are in the plane defined by F7, F3, Thus, the question that needs to be asked is not “whether SE
Fz, F4, and F8 [Fig. 32.4]). Interelectrode distances within the are superior to ATE or BTE in recording epileptiform activity of
coronal plane are determined by the superior electrodes as well mesial-temporal origin” but “in what circumstances should SE
(i.e., F9–F7 is 2.5 cm when F7–F3 is 5 cm). Distances between be used?” Data from three studies carried out by one of the
basal electrodes should be as equidistant as possible (T9 and authors (AMK) may provide answers to these questions
T10 will usually have to be placed either anterior or posterior to (9,10,28). The first study included 17 patients with TLE of
their ideal position to avoid the ear auricles). mesial-temporal origin in which SE inserted blindly (SEB) and
ATE failed to identify a focal ictal onset (9). Recordings with SE
CHOICE OF ELECTRODES inserted under fluoroscopy (SEF) detected a unilateral
anterotemporal interictal focus in four patients in whom
Several studies have compared the localizing yield of ATE, NPE, SEB/ATE had failed to record any interictal spikes and detected
SE, and MSE over the 10-20 scalp temporal electrodes in the bitemporal independent interictal foci in one patient where
recording of interictal and ictal epileptiform activity of mesial- SEB/ATE had only identified unilateral spikes. In nine other
temporal origin. These studies have confirmed that any of these patients, the spike count obtained with SEF recordings
electrodes are superior to the standard 10-20 temporal elec- increased by more than 100%, relative to the spike count
trodes in the recording of epileptiform activity of mesial-tem- obtained with SEB/ATE recordings. In addition, SEF recorded
poral origin. seizures with an anterotemporal focal onset pattern in 10
Studies that have compared the effectiveness of spike detec- patients where SEB/ATE had failed to do so. These data raised
tion between NPE and ATE showed that ATE are as good as or the hypothesis that the detection of interictal and ictal activity
better at detecting epileptiform activity than NPE (4,17,18). In with only SEF was related to a restricted electric field of the
fact, one of these studies showed NPE detecting significantly activity recorded with SEF (Figs. 32.6 and 32.7).
fewer spikes (4). In addition, most of these studies report To test this hypothesis, a second study was carried out
patient discomfort with NPE and more muscle and movement using the interictal data of the previous study. The study con-
artifact associated with NPE than ATE. Given these data, ATE sisted of a comparison of spike voltages at SEF, SEB, and ATE
have replaced the use of NPE in most epilepsy centers. in sets of five randomly selected spikes per interictal focus,
When only two ATE are being placed, T1 and T2 may be recorded in the course of the two monitoring studies (10).
preferable to 10-10 system electrodes because of their ease of The voltage differences were expressed as ratios, VATE/SEF and
placement. The closest counterparts to T1 and T2 in the 10-10 VATE/SEB and for each spike set, a mean ratio was calculated.
system are electrodes FT9 and FT10. Locating FT9 and FT10 The spike voltages were almost identical at SEB and ATE
requires the calculation of at least four other coordinates in the (mean VATE/SEB = 0.94), while they were significantly higher at
10-10 system. SEF than at ATE (mean VATE/SEF = 0.66; P 0.001). In the SEF
ATE and scalp BTE are included today in EEG and video- recordings, the narrowest electric field contours were found
EEG studies in patients with presumed epilepsy of mesial tem- among interictal foci that had only been recorded with SEF,
poral origin in all major epilepsy centers worldwide. A big which, in turn, were significantly smaller than those of inter-
debate remains on whether SE add any localizing yield to the ictal foci in which SEF yielded an increment of more than
interictal and ictal data obtained with ATE and BTE. Table 32.1 100% in spike counts (relative to those obtained with
summarizes the data from studies that compared the localizing SEB/ATE). These, in turn, had a significantly smaller contour
yield of interictal and ictal foci with SE and ATE (9,10,19–28). than those of the interictal foci where SEF failed to yield any
As seen from the table, some studies show some advantage of advantage over SEB/ATE (P 0.001). In addition, VATE/SEF of
SE over ATE, while others do not. interictal foci in which SEF recorded seizures with a focal
How can we explain such discrepant findings? Most epilepsy onset pattern were significantly lower than those of foci where
centers insert SE under blind conditions, in which cases the SEF failed to do so (P = 0.016). Finally, VATE/SEF did not differ
recording tips commonly may end up in a position at a distance from VATE/SEB among interictal foci where SEF failed to yield
from the FO, either anterior or posterior to it. As stated above, the any advantage over SEB/ATE in either interictal (P = 0.240) or
petrous pyramid is interposed between the SE recording tip and ictal (P = 0.311) recordings. The data of the second study con-
mesial-temporal structures when it ends up in a posterior posi- firmed our hypothesis and proved that SEF yield additional
tion to the FO, while a position anterior or lateral to the foramen localizing data when recording epileptiform activity with a
has the disadvantage of recording through bone and at an restricted field, provided that its recording tip is positioned
increased distance from the presumed epileptogenic area. A large below the FO. When distant from the FO, SE can be expected
electric field of interictal and ictal activity minimizes the impact to yield comparable data to those obtained with ATE.
674 Part V ■ Complementary and Special Techniques

Tabl e 3 2 . 1

Comparison of Interictal and Ictal Recordings among ATE, SE, and NPE

Authors Electrodes Compared Results


Sperling and Engel (19) Interictal discharges at sphenoidal, 99% of discharges were detected with sphenoidal,
earlobe, and nasopharyngeal electrodes 57% with nasopharyngeal, and 54% with
earlobe electrodes
Binnie et al. (20) Interictal discharges at multipolar Sphenoidal electrodes offer no improvement in
sphenoidal and surface electrodes sensitivity compared with surface electrodes
(near current T1 and T2 sites)
Sperling et al. (21) Interictal discharges at sphenoidal and Sphenoidal electrodes were synchronous with
invasive electrodes invasive electrodes in the neocortical areas but
not in the amygdala or hippocampus
Binnie et al. (22) Interictal discharges at sphenoidal 4% of discharges were seen at sphenoidal electro-
electrodes stereotactically placed into des only, while 3% were seen at surface only
the foreman ovale and surface electrodes
Sperling and Engel (23) Ictal onset at sphenoidal and scalp 19% of the seizure onset was seen only at
electrodes sphenoidal electrodes. The sphenoidal electro-
des led the scalp by more than 5 seconds in
70% of the seizure onsets
Fernández Torre et al. (24) Ictal onset at sphenoidal, infrazygomatic, Ictal onset was seen earliest at the
anterior temporal, and surface temporal sphenoidal and then at infrazygomatic and
electrodes anterior temporal leads
Ives et al. (25) Ictal onset at sphenoidal and T1/T2 40% of the seizures were seen first at sphenoidal
electrodes electrodes and 60% of the seizures were seen at
both recording sites
Wilkus et al. (26) Ictal onset at sphenoidal, T1/T2, No difference was seen between electrodes
and anterior temporal electrodes
Pacia et al. (27) Ictal onset at foreman ovale–placed Improved identification and timing at the
sphenoidal electrodes and anterior sphenoidal electrodes 7% of the time
temporal electrodes
Kanner et al. (9) Interictal and ictal activity with Fluoroscopically placed sphenoidal electrodes
sphenoidal electodes placed with added additional information such as increased
fluoroscopic guidance just below the spike count and better seizure onset
foramen ovale and standard localization
blind–placed sphenoidal electrodes
Kanner and Jones (10) Interictal and ictal activity at sphenoidal Significantly higher voltage spikes and significan-
electodes placed with fluoroscopic tly narrower electric field contour were seen
guidance just below the foramen ovale, during ictal and interictal activity recorded
standard blind–placed sphenoidal from fluoroscopic-guided sphenoidal
electrodes, and anterior temporal electrodes adding additional data
electrodes
Mintzer et al. (28) Ictal onset at sphenoidal electodes placed Fluoroscopically placed sphenoidal electrodes
with fluoroscopic guidance just below increased rater reliability, increased the number
the foramen ovale and anterior temporal of seizures correctly localized, and detected
electrodes seizures 5 seconds before they were seen at
the anterior temporal electrodes in 43% of
the seizures
Chapter 32 ■ Nasopharyngeal, Anterotemporal, and Sphenoidal Electrodes 675

The question of when should SE be inserted was addressed THE ADVANTAGE OF JOINT USE
in a third study by determining the frequency with which SEF OF SE AND ATE/ BTE
detected localizing data not obtained with ATE/BTE recordings
in a group of 40 unselected patients whose presurgical evalua- As stated above, epileptic seizures of TLE may originate in
tion had demonstrated a unilateral (n = 32) seizure focus or mesial temporal structures or temporal lateral neocortex. With
bilateral independent (n = 8) seizure foci in anterotemporal the widespread availability of digital EEG systems, a spatial-
regions (28). The study consisted of a comparison of the ictal temporal analysis of epileptiform discharges with SE and ATE
onset pattern of 156 seizures obtained with SEF and ATE/BTE may help establish the source in mesial structures by demon-
by four electroencephalographers who reviewed the 312 ictal strating the initial negativity at the SE. On the other hand, a
recordings independently, and blind to the patients’ identity, to synchronous discharge at SE and ATE/BTE does not exclude a
any presurgical data, and to whether the recordings’ montage mesial source.
included ATE or SEF. Inter-rater agreement among the four
raters was significantly greater with SEF than ATE recordings CONCLUDING REMARKS
(P 0.0001). The number of seizures correctly localized by at
least three raters was significantly greater with SEF (n = 144) The recordings of epileptiform activity of presumed mesial
than ATE (n = 99; P 0.0001). All the seizures (n = 36, or 23%) temporal origin should always include ATE/BTE. The available
originating from 14 ictal foci (29%) in 11 patients (27.5%) were data do not appear to support the use of NPE, they do not
only localized with SEF. This was a significantly more common appear to yield any advantage over ATE/BTE, and they are asso-
occurrence in patients with bilateral independent ictal foci (P = ciated with some discomfort. SE should be considered in special
0.04). The ictal onset was detected at SEF five or more seconds circumstances, and their insertion should be done under fluo-
earlier than at ATE in 67 seizures (43%) originating from 33 roscopic guidance.
(69%) foci in 30 patients (75%). In addition, SEF had a signifi-
cant advantage over ATE in the recording of seizures of patients REFERENCES
with bilateral independent foci and with a normal MRI, but not 1. Homan RW, Herman J, Purdy P. Cerebral localization of the 10-20
in seizures of patients with a structural lesion. These data sug- system electrode placement. Electroencephalogr Clin Neurophysiol.
gested that SEF facilitate the localization of anterotemporal ictal 1987;66:373–382.
onset and in about one-fourth of patients SEF identify ictal data 2. Homan RW, Jones MC, Tawat S. Anterior temporal electrodes in
not localized or mislocalized with ATE recordings. The follow- complex partial seizures. Electroencephalogr Clin Neurophysiol.
ing conclusions can be derived from these data: 1988;70:105–109.
3. Goodin DS, Aminoff MJ, Laxer KD. Detection of epileptiform
(1) If SE are to be inserted, they should be placed under fluo- activity by different noninvasive EEG methods in complex partial
roscopic guidance. epilepsy. Ann Neurol. 1990;27:330–334.
(2) SE need not be inserted in all patients with suspected TLE 4. Sadler RM, Goodwin J. Multiple electrodes for detection of spikes
of mesial temporal origin. in partial complex seizures. Can J Neurol Sci. 1989;16:326–329.
(3) The indications to insert SEF are as follows. 5. Jones DP. Recording of the basal electroencephalogram with sphe-
noidal electrodes. Electroencephalogr Clin Neurophysiol.
In case of hippocampal atrophy, the decision to insert SEF can be 1951;3:100.
delayed until the first seizure is recorded and the interictal 6. Marshall C. Sphenoidal electrodes: an effort toward their popular-
recordings of the first 24 hours have been reviewed. SEF will not ization. Electroencephalogr Clin Neurophysiol. 1957;9:379–382.
be necessary in the case of patients with unilateral atrophy if 7. Rovit RL, Gloor P, Rasmussen T. Sphenoidal electrodes in the elec-
ATE recordings demonstrate: trographic study of patients with temporal lobe epilepsy: an eval-
uation. J Neurosurg. 1961;18:151–158.
• A majority of spikes ipsilateral to the atrophic hippocampus. 8. Ives JR, Gloor P. Update: chronic sphenoidal electrodes.
• The first seizure has a focal or regional onset pattern (pro- Electroencephalogr Clin Neurophysiol. 1978;44:789–790.
vided that all of the patient’s seizures are identical by history). 9. Kanner AM, Ramirez L, Jones JC. The utility of placing electrodes
under the foramen ovale with fluoroscope guidance. J Clin
However, SEF should be inserted:
Neurophysiol. 1995;12:72–81.
• If ATE interictal recordings reveal bilateral independent spike 10. Kanner AM, Jones JC. When do sphenoidal electrodes yield addi-
foci with a relative frequency of 50/50 to 60/40. tional data to that obtained with antero-temporal electrodes?
• If ATE recordings reveal an ictal onset pattern that is lateral- Electroencephalogr Clin Neurophysiol. 1997;102:12–19.
ized or not localized. 11. Laxer KD. Mini-sphenoidal electrodes in the investigation of
• In case the patient’s seizures are not identical by history seizures. Electroencephalogr Clin Neurophysiol. 1984;58:127–129.
(excluding complex partial seizures that also evolve to sec- 12. Buchhalter J, Schomer DL, Schachter S. A comparative study on
ondarily generalized tonic–clonic seizures). the sensitivity of the mini and standard sphenoidal electrodes.
• In case of bilateral hippocampal atrophy. J Epilepsy. 1991;4:29–32.
13. Chu NS. Surface sphenoidal electrode for recording anterior tem-
In case of a unilateral anterotemporal structural lesion, the poral spikes. Clin Electroencephalogr. 1992;23(4):190–195.
same criteria as outlined above can be used. 14. Roubicek J, Hill D. Electroencephalography with pharyngeal elec-
In case of a normal brain MRI, SEF should be inserted. trodes. Brain. 1948;71:77–87.
676 Part V ■ Complementary and Special Techniques

15. deJesus PV Jr., Marsland WS. The role of nasopharyngeal elec- 25. Ives JM, Drislane FW, Schachter SC, et al. Comparison of coronal
trodes in clinical electroencephalography. Neurology. 1970;20: sphenoidal versus standard anteroposterior temporal montage in
869–878. the EEG recording of temporal lobe seizures. Electroencephalogr
16. Kashing DM, Celesia GG. Nasopharyngeal electrodes in the diag- Clin Neurophysiol. 1996;98:417–421.
nosis of partial seizures with complex symptoms. Arch Neurol. 26. Wilkus RJ, Vossler DG, Thompson PM. Comparison of EEG
1976;35:519–520. derived from sphenoidal, infrazygomatic, anterior temporal, and
17. Silverman D. The anterior temporal electrode and the ten-twenty midtemporal electrodes during complex partial seizures.
system. Electroencephalogr Clin Neurophysiol. 1960;12:735–737. J Epilepsy. 1993;6:152–161.
18. Nowack WJ, Janati A, Metzer WS, et al. The anterior temporal elec- 27. Pacia SV, Jung WJ, Devinsky O. Localization of mesial temporal
trode in the EEG of the adult. Clin Electroencephalogr. 1988;4: lobe seizures with sphenoidal electrodes. J Clin Neurophysiol.
199–204. 1998;15:256–261.
19. Sperling MR, Engel J Jr. EEG from the temporal lobes: a compari- 28. Mintzer S, Nicholl JS, Stern JM, et al. Relative utility of sphenoidal
son of ear, anterior temporal and nasopharygeal electrodes. and temporal surface electrodes for the localization of ictal onset
Neurology. 1985;17:510–513. in temporal lobe epilepsy. Clin Neurophysiol. 2002;113:911–916.
20. Binnie CD, Dekker E, Smit A, et al. Practical considerations in the 29. McClean PD. A new nasopharyngeal lead. Electroencephalogr Clin
positioning of EEG electrodes. Electroencephalogr Clin Neuro- Neurophysiol. 1949;1:110–112.
physiol. 1982;53:453–458. 30. Bach-Y-Rita G, Lion J, Reynolds J, et al. An improved nasopharyn-
21. Sperling MR, Mendiuys JR, Engel J Jr. Mesial temporal spikes: a geal lead. Electroencephalogr Clin Neurophysiol. 1969;26:220–221.
simultaneous comparison of sphenoidal, nasopharyngeal and ear 31. Kissani N, Alarcon G, Dad M, et al. Sensitivity of recordings at
electrodes. Epilepsy. 1986;27:81–86. sphenoidal electrode site for detecting seizure onset: evidence
22. Binnie CD, Marston D, Polkey CE, et al. Distribution of temporal from scalp, superficial and deep foramen ovale recordings. Clin
spikes in relation to the sphenoidal electrode. Electroencephalogr Neurophysiol. 2001;112:232–240.
Clin Neurophysiol. 1989;73:403–409. 32. Kanner AM, Parra J, Gil-Nagel A, et al. The localization yield of
23. Sperling MR, Engel J. Sphenoidal electrodes. J Clin Neurophysiol. sphenoidal and anterior temporal electrodes in ictal recordings: a
1986;3:67–73. comparison study. Epilepsia. 2002;43:1–8.
24. Fernández Torre JL, Alarcón G, Binnie CD, et al. Comparison of 33. Fenton DS, Geremia GK, Dowd AM, et al. Precise placement of
sphenoidal, foramen ovale, and anterior temporal placements for sphenoidal electrodes via fluoroscopic guidance. Am J Neuroradiol.
detecting interictal epileptiform discharges in presurgical assess- 1997;18:776–778.
ment for temporal lobe surgery. Clin Neurophysiol. 1999;110:
890–895.
CHAPTER
Intracranial Monitoring: Depth,
Subdural, and Foramen Ovale Electrodes
MARGITTA SEECK, DONALD L. SCHOMER, AND ERNST NIEDERMEYER
33
INTRODUCTION monitoring” or “no intracranial monitoring.” However, the
yield of precisely mapping the cortex in terms of focus localiza-
In most Western societies, epilepsy affects around 0.7% of the tion and vital cortex is evident even without formal studies.
population, that is, 2.1 million in the United States, 3.5 million Most patients who are candidates for IEEG fall into an inter-
in Europe, and 47 million worldwide are affected. In about 20% mediate category of operability: a focal onset is suspected or at
to 30%, drug treatment cannot control seizure occurrence and least not completely ruled out on the basis of the results of non-
the possibility of surgical treatment is then considered. Although invasive EEG exams, but immediate resective surgery is consid-
the optimal profiles of surgical candidates continue to be ered too risky in terms of neurologic complications. Statistically
defined, the precise localization of a focal onset remains a cor- speaking, the need for IEEG is an unfavorable predictor for
nerstone of successful surgical treatment. In many patients, postoperative success (1).
localization requires EEG recording directly from the cortex. The definition of candidacy for IEEG has changed over the
This is done either by peroperative corticography, that is, directly past decades, due in large part to more sophisticated brain
from the brain in the operating room (see Chapter 34), or by imaging. Today, in most centers, a patient with a glioma in the
chronically implanted electrodes for a variable duration lasting anterior mesial temporal lobe and consistent EEG findings
from a few days to several weeks. The major goals of intracranial would not be considered an IEEG candidate, even if he/she had
EEG (IEEG) recordings are to determine the seizure onset and contralateral interictal spikes. In contrast, a patient with multi-
distinguish the focus from vital cortex (Table 33.1). There are no ple lesions and discordant EEG findings, or with no MRI abnor-
true evidence-based studies on the yield of chronic IEEG moni- mality at all, would be considered an appropriate IEEG
toring, that is, studies assigning subjects randomly to “intracranial candidate. However, to the best of our knowledge, there are no
published consensus guidelines regarding the utility of IEEG.
The need for invasive recordings has been discussed extensively
Tabl e 3 3 . 1 for different clinical scenarios for patients with temporal lobe
epilepsy (TLE). It has been proposed that the concordance of
Requirements and Goals of Intracranial Monitoring unilateral hippocampal atrophy and interictal EEG is sufficient
to proceed to surgery, that is, even without (scalp) ictal record-
• Noninvasive exams are inconclusive concerning the site ings, which may be sometimes misleadingly showing bilateral
and extent of a surgical intervention or even contralateral epileptic discharges (2). However, some
• The clinical status of the patient is compatible with centers would envision not only ictal recordings but even IEEG
prolonged intracranial recordings, for example, no major in order to determine unambiguously EEG onset (3). In any
behavioral disturbances, absence of psychosis, and case, careful evaluation of each individual patient is mandatory,
bleeding disorder including functional imaging and neuropsychological data.
• The patient accepts the procedure and the risk of Ictal scalp recordings help to better identify the patient’s seizure
inconclusive results after intracranial monitoring semiology, verify the presumed ictal onset by analysis of the
clinical picture, and estimate the burden of epilepsy as well as
• The precise focus identification requires the following following up on postoperative persistent seizures. Apart from
information: persistent epileptic seizures, postoperative nonepileptic (psy-
• Lateralization of onset (e.g., left or right temporal lobe chogenic) seizures have been described in 2% to 8%. These can
epilepsy) be difficult to diagnose if the preoperative semiology is not
• Identification of lobe of onset (e.g., temporal vs. frontal known (4).
lobe epilepsy) Despite improvements to our understanding about who ben-
efits most from this otherwise costly and labor-intensive proce-
• Precise region of onset (e.g., left anterior vs. posterior dure, each center has its own strategies, which depend on the
temporal onset) experience of the epilepsy surgery team, the availability and qual-
• Localization of vital or eloquent cortex in adjacent cortex ity of the noninvasive exams, and overall national health care sys-
is mandatory (e.g., determination of language cortex in a tems that facilitate (or not) the access to the highest level epilepsy
patient with left posterior temporal lobe seizure onset) surgery centers and IEEG. Requirements for the equipment and
staffing of such centers have been published (5). The diversity in
677
678 Part V ■ Complementary and Special Techniques

presurgical epilepsy evaluation across nations or continents BASIC CONCEPTS


became evident in the recent survey. The Pediatric Epilepsy
Surgery Subcommittee of the International League Against IEEG should be considered if it is felt that surgery can cure or
Epilepsy conducted a survey in Europe, the United States, and at least significantly ameliorate the epilepsy disorder, but perti-
Australia (6). It appeared that in the United States children are nent localizing information is still missing from the noninva-
more often evaluated with intracranial electrodes, even in the sive evaluation. This implies that every patient should first
presence of an MRI lesion. There is also more access to functional undergo a noninvasive evaluation in order to understand the
imaging in the United States compared to their European and size and nature of the problem. A clear clinical hypothesis is
Australian counterparts. However, more patients are treated with mandatory before implantation, because implanted electrodes
a vagal nerve stimulator, a palliative procedure, in the United record only from a limited cortical volume. Although the exact
States compared to Europe and Australia. Although other charac- volume is not known, there are estimates indicating that a cor-
teristics of the patient populations were not different (underlying tical area of 10 to 20 cm 2 is necessary to generate a scalp spike
pathology, affected lobe, etc.), the reasons for these differences are (24). Detailed analysis of scalp discharges and the simultane-
not entirely clear but are probably of multiple origins, which ously recorded subdural EEG showed that only the early com-
include different national “schools” with different algorithms of ponents of spikes and seizures arise from a limited volume,
epilepsy care or different regulations from health care providers. which is, however, still larger than 10 cm 2. With further propa-
However, while sophisticated epilepsy centers exist today in gation, the active brain areas attain 30 cm 2 or more (25). In this
all Western countries, it should not be forgotten that there are context, imaging methods that suggest point sources of the
still countries where epilepsy surgery is not done or is only underlying spikes represent rather a simplification than the true
rarely available, which prevents patients from obtaining a estimation of the size of epileptogenic cortex (26).
potentially very effective epilepsy treatment (7). Moreover, the surgical act and the fact that the electrodes
In the past 10 to 20 years, the development of new noninva- remain in place for several days or weeks are associated with a
sive imaging techniques was readily embraced in the presurgical significant risk of morbidity but a relatively small risk for mor-
epilepsy evaluation (8–13), starting with the entry of high-def- tality (see below), requiring careful elaboration of the goal of
inition MRI and special imaging protocols for epilepsy patients the investigation and the sites of implantation.
(14). Indeed, the knowledge obtained from these techniques has Several concepts need to be recognized and defined as much
given us patient populations and helped to specify patient pop- as possible before any epilepsy surgical procedure is proposed
ulations where IEEG is more likely to be diagnostic. For exam- (27). The epileptogenic area, that is, the area whose resection is
ple, 10 years ago, invasive monitoring was considered in necessary and sufficient to abolish further seizures, needs to be
patients with hippocampal sclerosis, but bilateral temporal clearly identified. This zone is not to be confounded with
spikes or seizures. However, the presence of an MRI abnormal- • the lesional zone, that is, the area that appears morphologi-
ity is a good prognostic indicator for seizure-free outcome post- cally altered but may be smaller or even remote to the epilep-
operatively (15), and patients with bilateral spikes or even togenic zone
seizures may very well benefit from surgery on the sclerotic hip- • the irritative zone, that is, the area of interictal spikes, which
pocampus (16,17), even without IEEG. However, truly bitem- can be more widespread than the epileptogenic zone and
poral lobe epilepsy, as determined by IEEG and MRI, is include even contralateral structures
associated with a lower success rate and only 50% became • the symptomatic zone, that is, the area whose activation pro-
seizure-free according to a recent study (18). On the other duces the clinical symptoms, such as nausea or dystonic
hand, severe unilateral hippocampal atrophy, the so-called posturing
burned-out hippocampus, may present with aberrant, even • the functional deficit zone, that is, area whose activation/
contralateral, scalp ictal discharges. Those patients nevertheless deactivation leads to neurologic or neuropsychological
benefit from surgery (19). In some of these patients, IEEG was deficits, for example, verbal memory impairment or Todd
used to verify ictal onset in the affected hippocampus. paresis
More recently, it has become evident that the presence of a
localized or lateralized temporal hypometabolism equals the Overall, IEEG data look different compared to scalp EEG data
presence of an MRI lesion. Both MRI lesional and nonlesional and appear to be more “epileptogenic,” due to lack of the filter
TLE have the same postoperative prognosis, that is, a chance of qualities of the skull, although the same rhythms known from
seizure control of approximately 80%, if the temporal hypome- scalp EEG can also be retrieved in the IEEG. The determination
tabolism in the PET is obvious and unilateral (20). Thus, if this of ictal EEG pattern is usually at the center of the investigation;
finding is confirmed in more patients, nonlesional TLE patients however, careful analysis of interictal epileptogenic discharges is
may not need IEEG anymore if they underwent good-quality probably more important than previously appreciated.
PET-imaging and its results are concordant with the remainder The overall yield of IEEG depends on the criteria that deter-
of the evaluation. Over the next few years, other investigative mine at which degree of inconsistency intracranial evaluation is
measures such as studies on other pathologies, for example, offered. There are only very few data on this subject. A recent
dysplasia or tuberous sclerosis, identifying reliable noninvasive study, with access to modern imaging tools, described the
markers of the epileptogenic zone may further reduce the over- results in 77 patients with mesial temporal (39%), lateral tem-
all number of patients who need IEEG (21–23). poral (12%), and extratemporal (43%) epilepsy (12). In the
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 679

remaining five patients (6%), seizures could not be captured Today, depth and subdural electrodes are the most estab-
and they remained unclassified. Localizing results were lished electrodes types. Most centers have a preference for one
obtained in 74%, which corresponds to numbers from other or the other electrode type, which depends on personal experi-
studies (28,29) and to the authors’ experience. Thus, despite the ence or technical equipment in the neurosurgical clinic. Around
invasive nature of the procedure and the physical and psycho- the same time of the arrival of subdural electrodes, foramen
logical investment by the patient, it is not always certain that ovale electrodes (FOE) (36) and epidural peg electrodes were
IEEG will be followed by a resective procedure. In around 25%, described by the Zürich group of Wieser et al. and the
surgery cannot be proposed, which needs to be addressed and Cleveland Clinic group of Lüders et al. (37), respectively. The
reiterated during preparatory talks with the patients. use of the latter two types remains relatively limited and is often
used as an intermediate step in determining if, and which,
HISTORY larger set of intracranial electrode setups will be more adequate
or as complementary electrode type, if depth or subdurals alone
IEEG focus evaluation has a relatively brief history, starting in do not appear sufficient.
Germany with Berger’s discovery of the EEG published in 1929
(30). Otfrid Foerster used preoperative EEG during epilepsy
surgery and became a pioneer in using EEG for the determina- DETERMINATION OF PRECISE
tion of the epileptogenic zone. Foerster worked with Wernicke ELECTRODE PLACEMENT
and published a brain atlas, and as an aside he was the treating
Independent of the chosen electrode type, knowledge of the
neurologist for Lenin when he suffered a stroke in 1922. The ris-
exact position of the electrodes with respect to the individual
ing Nationalist government basically destroyed Germany’s sci-
brain anatomy is of utmost importance for successful surgery in
entific culture for the next decades and many researchers left
terms of complete focus removal while sparing eloquent cortex.
the country. Dr Foerster was the mentor of Wilder Penfield who
Previously, only radiographs of the skull were possible. These
himself became widely known for the corticography of the
provided only a gross idea of the electrode position (38,39).
human neocortex. Thanks to his research on patients, the corti-
Now 3D reconstruction of CT and MRI with coregistration of
cal representations of the motor cortex (homunculus) became
X-rays provides superior electrode localization results (39).
standard knowledge. Dr Penfield was a neurosurgeon and
Coregistration of CT, MRI before and after electrode place-
founded the Montreal Neurological Institute. Herbert Jasper
ment, or digital photos taken intraoperatively with the patient’s
joined him as a neurologist and clinical neurophysiologist, and
MRI (40–45) provides excellent results in terms of electrode
together they trained numerous neurosurgeons and neurolo-
recording sites localization. Mean mismatch between real and
gists in the field of epilepsy surgery. They probably represent
virtual electrode placement was found between 2 and 4 mm
the first example of a successful relationship between a neurol-
(42,44). Additional external fiduciary markers improve this pre-
ogist and a neurosurgeon. This combination remains a prereq-
cision (44). Coregistration of different MR sequences, notably
uisite for any successful epilepsy surgery team (31).
Flair and T2, allow the visualization of smaller lesion inside the
Wires and a type of “strip” electrode, placed in the epidural
OR (46). Implantable fiduciary markers were also used, which
space, were the first to be used in epilepsy patients by Jasper and
led to an even smaller localizing error (~0.5 mm) (47). Other
Penfield. Later, subdural electrodes and other electrode types
difficulties inherent to all methods of coregistration between a
were developed and applied (31). Bancaud and Talairach, another
preimplantation and a postimplantation image are the transi-
successful team of neurologist and neurosurgeon from Hôpital
tory changes of the brain anatomy due to cerebral edema,
Sainte-Anne in Paris, introduced multicontact depth electrodes
hematoma, and brain shift. In most algorithms, the electrode
penetrating directly into the cerebral tissue (32,33), allowing a
artifacts are used to identify their exact localization, on the basis
systematic investigation of deep structures. This was a major step
of either a CT or an MRI. In a second step, the extracerebral tis-
forward in epilepsy surgery investigations, since at that time MRI
sue (skull, scalp) is stripped away, and brain reconstructed in a
was not available and the evaluation of surgical candidates relied
3D fashion, showing the surface with the individual gyri and
more on neurophysiologic methods. Ten years later, Wyler devel-
sulci. In Figure 33.1, coregistration algorithm is proposed.
oped and introduced another electrode type into the field: the
The exact localization of depth electrodes is simpler, given
subdural electrode, which was being placed onto the brain’s sur-
that there is less of brain shift. Otherwise, similar algorithms to
face without the risks of an intracerebral insertion (34). Initially,
reconstruct the position of the electrodes are employed.
strips with four electrodes were described, evolving later to more
complex and larger arrangements (see below).
Although the number of inserted electrodes steadily DEPTH ELECTRODES
increased, or at least became technically possible, simultaneous
recording of 100 electrodes was difficult for EEG systems, Technical Aspects
requiring the selection of recorded electrodes and even changes Taking all patient studies together, depth electrodes are proba-
during the evaluation. Upgrading of established systems and bly the most frequently used electrode types (Fig. 33.2), given
innovative setups has finally overcome this obstacle (35), and at that they were already in use in 1960s and 1970s in the medical
the time of the writing of this text, EEG systems with up to 256 field. They are inserted through Burr holes by stereotactic
channels are on the market. methods, using guidance from previously acquired brain
680 Part V ■ Complementary and Special Techniques

Figure 33.1 Coregistration of int -


racranial electrodes and the
patient’s individual MRI. Propo -
sition for a workflow aiming fusion
of the patient’s preimplantation
MRI with the postimplantation MRI
or postimplantation high-resolution
CT. Courtesy of L. Spinelli, PhD.
The preimplantation is normalized
and skull stripped, then fusioned
with the postimplantation MRI,
which underwent the same compu-
tation. In this example, particular
care is taken to compensate for the
brain shift due to compression
by subdural grids. This is less of
an issue in patients with depth
electrodes.

imaging. Depth electrodes are built as wires containing evenly electrodes. The electrodes themselves can be rigid or flexible,
spaced electrode contacts of conductive material. The insertion but most neurosurgeons prefer flexible electrodes, which may
is done by a mandarin whose trajectory is determined by the lead to less precise electrode placement (due to involuntary
site of the target(s) through CT and/or MRI of the patient’s deviation from the initial target), but they have had a lower
brain. Sometimes additional arteriography or MR-angiography risk of bleeding. Most companies that produce intracranial
is required in order to identify crucial vessels and diminish the electrodes allow the choice between platinum–iridium and
risk of hemorrhage. In many centers today, a stereotactic setup nickel–chrome alloys. Only the first combination is MR com-
is utilized using the patient’s MRI. More recently, a frameless patible, but they are somewhat more expensive. The elec-
robot-guided insertion has been reported (48), and the future trodes have multiple contacts (usually between 8 and 12) and
will show if the application of such sophisticated methods will can be inserted orthogonally, that is, perpendicular to the lat-
decrease the risk of depth electrode placement and/or shorten eral surface or longitudinal-parasagittally. Oblique insertions
operation time significantly. into the frontal lobe toward the basal ganglia have also been
Electrode spacing is most often between 5 and 10 mm, but employed. However, in all cases, there is only very limited
any configuration, distance, and number of electrodes can be sampling of the neocortical–lateral cortex, which may require
chosen, that is, either at regular distances or only at sites that additional subdural or scalp electrodes if the cortex needs
cover the gray substance. Most electrodes contain 6 to 15 larger coverage.

Figure 33.2 Depth electrodes. Left: Coregistration of the


orthogonally inserted electrodes and the patient’s MRI.
The insertion sites of depth electrodes in both hemispheres
are marked in white. Upper right: Same patient as shown
on left, showing his depth electrode inserted in the left
amygdala. In this coregistration, each contact is identified
separately (light gray balls). Lower right: Longitudinal inser-
tion of depth electrodes, indicated in light gray.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 681

Given that this electrode type is the only one that goes into epilepsy in 1939 (53), he can be probably considered the first to
the brain parenchyma, the major advantage of depth electrode have used this electrode type. Subdural or epidural electrodes
recordings is the opportunity to record from deep, buried cor- consist of disk-shaped electrodes, embedded in transparent
tex, such as hippocampus, amygdala, or mesial frontal struc- silastic or Teflon material, arranged in rows or arrays of all
tures. Furthermore, the simultaneous sampling of mesial and shapes (Fig. 33.3). As with depth electrodes, they are made of
lateral cerebral structures, associated to an excellent EEG qual- platinum–iridium and nickel–chrome alloys, allowing MR
ity, is a favorable aspect of depth electrodes. They also permit imaging only if they are made of platinum. The contacts are put
bilateral and multilobar sampling, but there is limited spatial freehand on the cortex, subdurally on the pia mater. If neces-
sampling in a given region, for example, identification of lan- sary, they can be reduced with sterile scissors or along perfo-
guage cortex and its extent is not possible with depth electrodes rated lines to smaller arrays directly in the OR. There are recent
alone. Another significant advantage is that they can be easily reports that make use of neuronavigation and stereotactic
removed, which can be accomplished without any anesthesia. methods to improve the precision of placement, in particular
They can be removed even at bedside. for strips, or for merging imaging data sets that help to take all
preoperative imaging information into consideration (54,55).
Risks Similar to depth electrodes, determination of electrode posi-
The major risk of using depth electrodes is intracranial or tions by MRI or CT imaging is an important issue because
intracerebral hemorrhage, which occurs in 1% to 4% (49). With smaller or larger shifts may occur after the implantation, in par-
modern imaging, the rate seems to be lower. In a study of 50 ticular with small arrays. The contacts are usually 5 to 10 mm
patients with depth and subdural strip electrodes (50), no apart and can be arranged as one line (strip) or in a larger array,
deaths, no infections, and no new neurologic deficits were for example, 8 8 contacts (grids of 64 electrodes) (Fig. 33.3).
reported. Ross et al. provided a review on previously reported Strip electrodes arranged in a single row can be inserted
series of depth electrode patients, and when considering the through Burr holes. However, a large grid, for example, array of
whole group of 1656 patients together, the relative risk was high- 4 4 or 8 8 electrodes, needs a full craniotomy. This is usu-
est for intracranial hemorrhage (1.1%), followed by infection ally a more invasive procedure than the stereotactic procedure
(0.8%). In a recent review of 259 patients, a risk of symptomatic employed for the placement of depth electrodes.
intracranial hemorrhage of 1.2% and 0.7% for permanent neu- The main indication for subdural electrodes is the precise
rologic deficit is reported (51). The Montreal group reported the localization of a seizure focus within a suspicious area. When
absence of major complications in a total of 6415 electrode using grid arrays, two to three lobes can be covered with a single
implantations (491 patients) between 1976 and 2006, that is, a grid, for example, frontal posterior, temporal, and parietal. This
time span before and after introduction of MRI. Hematoma and requires that noninvasive evaluation narrows down the hypoth-
infection were noted in 0.8% and 1.8%, respectively (52). esis of the site of the focus to one or two lobes. Subdural elec-
Overall, in this study, complications were more frequent in the trodes also allow for preoperative corticography, with the aim to
frontal lobe, perhaps due to the higher number of implanted determine vital cortex. This procedure is described below in
electrodes. Mortality was basically zero in the series after 1990. section Special Considerations in Pediatric Patients and in a
Among the hemorrhages, only very few were life threatening. In study by Lesser et al. (118). By stimulation with short (2 to 5
the authors’ experience, it should be less than 0.1%.
Patient counseling on the avoidance of antiplatelet drugs as
well as the implementation of preoperative coagulation param-
eters is recommended. There is no agreement on the use of pro-
phylactic antibiotic treatment in patients during depth
electrode investigations. If the contact between the intra- and
extracranial milieu is minimized with sterile bandages and the
patient is inhibited from scratching under the bandage, the risk
of infection is even less than 0.8%. Even years after depth elec-
trode investigations, discrete gliotic changes along the insertion
channel are seen in MRI images, but, to date, there is no evi-
dence that these changes become themselves epileptogenic or
are related to new neurologic or cognitive deficits.

SUBDURAL ELECTRODES
Technical Aspects
These electrodes were introduced somewhat later into the field Figure 33.3 Intraoperative subdural grid placement. Each contact
of epilepsy surgery than the depth electrodes and are used for (platinum disk, 0.8 mm diameter) is recorded through a wire, which is
epidural recordings. When Penfield recorded epidurally from led out of the crane through a larger white cable (in the lower left of the
the left parietotemporal region in a patient with posttraumatic image; in this grid, bundles of eight wires each).
682 Part V ■ Complementary and Special Techniques

seconds) intermittent electrical currents, sensations or involun- persistent neurologic deficits after subdural grid placement,
tary movements are produced, which supposedly reflect the not explained by brain swelling (61). IEEG in this patient
characteristics of the underlying cortex. Primary cortex can group may be related to more severe complications than in
thus be reliably identified. However, higher order cortical phe- patients without this history.
nomena, like language, are not always easily identified as shown Experience is also an important factor. The Cleveland Clinic
by Seeck et al. (56) group reported 33% of minor and more serious complications
These electrodes do not necessarily require knowledge of at the beginning of their epilepsy program, which dropped to
stereotactic procedures and/or a neuronavigation device and, as 19% in more recent years. Overall, the use of more than 100
such, can be performed in neurosurgic centers without experi- electrodes, longer recording sessions, that is, more than 2 weeks,
ence in stereotactic methods. Subdural electrodes record from a and more than one cable exit are related to an increase in com-
larger cortical surface and capture incompletely signals from plication rates (62,63). It is probably safe to say that complica-
the sulci as well as from radial sources. It is a well-established tions between 10% and 20% of the cases can be expected,
finding that only one third of the brain is accessible from the requiring close monitoring by specialized personnel and in a
surface, which also means only one third is accessible in terms center with 24 hours access to a scanner and an ICU. Regular
of recording. Thus, most brain tissue underneath a subdural neurologic exams and monitoring for infectious diseases are
grid will not be directly recorded, but may be involved only after mandatory (Table 33.2).
propagation to these more superficial cortical regions has If bleeding complications are present, it is safer to remove
occurred. the electrodes as soon as possible. If there are signs of local
infections, and if enough seizures are recorded, removal of
Risks electrodes is recommended. The rising of C-reactive protein
The morbidity is low for strip electrodes and includes infec- (CRP), after attaining postoperatively normal or near-normal
tions, around 1%, or bleeding when bridge veins are damaged values, is a valuable marker. If this is not the case, that is, no or
(57). Larger grid arrays are more likely to be associated to more only too few seizures are recorded, introduction or reinforce-
significant complications. The infection rate is higher than with ment of the antibiotic treatment with prolongation of the
depth or strip electrodes alone, and is reported to be around recording period, perhaps with more rigorous drug and sleep
8%. This risk can be diminished if the cables exit through a tun- withdrawal, should be considered. Since intracranial electrode
nel via a second incision a few centimeters away from the orig- implantation is a costly and intensive intervention, in terms of
inal incision. As for depth electrodes, no consensus exists manpower and, more importantly, physical and psychological
whether or not prophylactic antibiotic therapy should be given strain on the patients themselves, reimplantation at another
(57,63), albeit this issue is more relevant with subdural elec- moment is not easily done or recommended. General guide-
trodes. Based on personal experience, prophylactic antibiotic lines are difficult to establish since every center has its own
treatment in patients with large arrays of subdural electrodes resources, and patients differ in their willingness to cope with
was related to lower incidence of infectious problems, for exam- unforeseen clinical situations.
ple, Cefazolinum 4 500 mg IV.
Most patients with grids experience local edema of the
underlying cortex, leading to headache and photophonopho-
bia. This complication occurs less often with strips. Rigorous Tabl e 3 3 . 2
symptomatic treatment with antipain drugs as well as position-
ing of the head at a 30 to 45 elevation are helpful. Major or Proposed Surveillance Algorithm for Patients
near-fatal raised intracranial pressure is extremely rare, but if Admitted for Subdural Intracranial EEG Monitoring
discovered, may need immediate surgical decompression.
Consequently, a neurosurgeon on call nearby is a necessity. • 24 hours supervision by nurses, technicians, and/or other
In a recent study, delayed subdural hematoma was noted persons, for example, medical students or family members
to be the most frequent complication (8%) (58). They • Postimplantation 24-hour surveillance in the ICU
occurred up to 3 days postimplantation. This “fragile” period • When admitted for EEG monitoring
is probably somewhat longer and, as for most brain surgeries,
spans up to 5 days. A recent study in a pediatric group noted • CRP, blood: every day or every second day
postimplantation problems in 20% of their patients, with • Temperature: two times per day
predominance of hematoma, but this may be due to the fact • Brief neurologic exam (pupils, responsiveness, motor
that 43% of their patients had already undergone a prior functions) every 2–4 hours for the first 2 days, then
epilepsy surgery procedure (59). Another pediatric series every 4–6 hours for the next 3–4 days
reported a lower complication rate, including hematoma
(6%) (60). However, implantation of larger arrays of sub- • Optional: prophylactic antibiotic treatment (e.g.,
dural electrodes remains a relatively delicate procedure, cefazolin) starting from the day of the implantation
requesting unambiguous agreement on the indication, in through the monitoring period
particular in children. Patients with a history of high-dose • Optional: dexamethasone during the first 3– 4 days
radiotherapy and chemotherapy may develop significant
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 683

Despite higher complication rates with subdural electrodes, mainly of discomfort when the electrodes are inserted through
the rates of permanent neurologic deficits do not seem to differ the temporalis muscle and some superficial wound infections.
from the numbers for depth electrodes and are situated around Their main indication is the verification of the absence of
1% to 2% (63). The removal can be done at bedside, if strips epileptogenic discharges and/or recording of selected sites
were used, but will require another craniotomy during which where scalp electrodes would be too contaminated by muscle
the surgical resection of the focus is carried out as well, if indi- artifacts.
cated, if grids are used.
SURVEILLANCE
FORAMEN OVALE ELECTRODES
AND EPIDURAL ELECTRODES There is no consensus guidelines concerning the degree of med-
ical and paramedical staff surveillance and concomitant treat-
FOE, initially developed in 1985 by the Zürich group of Wieser ment in patients admitted for EEG monitoring. However, a
et al., are used somewhat less frequently in most centers (36). “semi-intensive” care unit with a lower nurse–patient ratio
FOE record epidurally and extracerebrally from the mesial tem- compared to normal care units appears the most appropriate
poral lobe structures. They contain four to six contacts, and can setting. There are still many centers that cannot offer profes-
be inserted under local anesthesia (Fig. 33.4). They can also be sional surveillance at night or on weekends, leading to seizure-
easily removed at bedside, similar to depth electrodes. Major related complications, for example, autonomous electrode
complications are rare, but they are not “semi-invasive” as ini- removal during the postictal confusional state.
tially advocated. Subarachnoidal hemorrhage and transient The choice on whom to rely on during the patient’s monitor-
brainstem symptoms, for example, trigeminal neuralgia, are ing session is influenced by budget and legal considerations.
observed. Complication rates of 1% to 9% are reported, mostly Although we know relatively well the risk of IEEG complications
transient, that is, similar to rates reported for depth or subdural such as bleeding or infection, there is disagreement across surgi-
electrodes. FOE yield good recordings from the mid and poste- cal epilepsy centers on who watches the patient: parent/close
rior portions of the hippocampus, but less well from the ante- family member; nonmedical staff, for example, out-of-work
rior aspect and the amygdala and/or small subcompartments of people recruited on through a placement agency; EEG techni-
the mesial temporal structures. The classical indication for FOE cians; or nurses. Patients who are recorded after drug with-
is the lateralization of temporal lobe onset. They may be com- drawal carry a risk for more prolonged or more intense seizures
bined with subdural or scalp electrodes for more extensive sam- (i.e., more often generalized tonic–clonic seizures) resulting in
pling from lateral temporal or extratemporal structures, and trauma and/or prolonged postictal confusion. Pulling out the
consequently the indication may be extended to other indica- electrodes is also encountered. In a recent review, 3 out of 491
tions. patients (0.6%) pulled out their electrodes during a seizure or
Epidural peg electrodes share configuration aspects with sub- during the early postimplantation period (52). CSF leakage,
dural electrodes and are inserted through burr holes or twist bleeding, or infection may result from such an act, particularly if
drill holes (37). They can be placed bilaterally, similar to scalp larger subdural electrodes arrays are removed.
electrodes, but cannot be used for cortical mapping. Electrical Family members or nonmedical people may not be enough,
stimulation through epidural electrodes is painful because the since they are often unable to judge the clinical context or sever-
rest of the dura is innervated and irritated by electrical current. ity of a given situation. There are no studies on the guidelines
Precise sublobar focus localization is not possible with epidural on the size and composition of the surveillance staff necessary
electrodes alone since their spatial sampling is not sufficient. to minimize these complications and to reduce the recording
However, they have a low morbidity ( 0.5%), consisting time. For obvious reasons, professional surveillance by trained

Figure 33.4 Foramen ovale electrodes. Left: Insertion


procedure through the base of the skull (i.e., through the
foramen ovale). (Courtesy of CCND Winnipeg.) Right:
Foramen ovale electrodes. (Courtesy of Dixi electronics.)
684 Part V ■ Complementary and Special Techniques

nurses and technicians, in collaboration with an experienced temporal focus. However, if they are placed or moved too later-
epileptologist or neurologist, is the optimal setting. Legal ally, in particular lateral to the collateral sulcus, false localization
aspects also need to be considered in order to be protected and lateralization may occur due to propagation to orbitofrontal
against “under-surveillance,” and these differ markedly or contralateral temporal cortex before recruitment of the
between countries. In some countries, like the United States, affected mesial temporal structure becomes visible (69).
legal action against the hospital is more readily taken if compli- Mesial extratemporal (i.e., interhemispheric) structures are
cations are encountered, and therefore, different solutions and explored well with both depth and subdural electrodes.
their legal implications need to be more thoroughly studied. Subdural electrodes are more easily placed given the larger cor-
The clinical observation of patients with IEEG should tical surface compared to mesial temporal structures. However,
include close monitoring of the neurologic status, cognitive comparative studies do not exist.
functions, and blood values depicting infections or bleeding. In extratemporal lobe epilepsy, subdural electrodes seem to
The “algorithm” proposed in Table 33.2 stems from personal be slightly better than depth electrodes (68), if convexity struc-
experiences. Close monitoring for the first 5 days is recom- tures need to be recorded. Since they have the advantage to
mended by most of the neurosurgeons, due to an increased risk allow cortical mapping, subdurals may be preferred if focus
of intracranial bleeding during this period. From then on, daily localization includes mainly extratemporal cortex where there
neurologic surveillance may be sufficient, aiming particularly at is a question of the involvement and extent of eloquent cortex.
the detection of infectious complications. Combination of depth and subdural electrodes is also more and
Since the introduction of chronic IEEG, there is ongoing more frequently used. However, comparative studies on the
debate if, when, and how long antibiotic therapy or corticos- yield of combined depth and subdural electrodes compared to
teroids are administered. Informal review of the protocols of each electrode type alone are missing.
the different centers shows that changes of such protocols have Comparisons between FOE and other electrode types have
emerged. The most frequent pattern had the regular use of also been performed. In seven patients with FOE and depth
steroids during the initial period. This pattern has been aban- electrodes, interictal and ictal discharges were picked up by FOE
doned for most patients. In most centers, steroids and antibi- if amygdala, hippocampus, and parahippocampal gyrus dis-
otics are considered with subdural implants, but rarely charged simultaneously, but less reliably if only a subcompart-
considered with depth electrodes. The placement of large grids ment of the mesial temporal structures was active (70).
is almost always followed by focal cerebral edema of variable Intracranial electrodes served also as “gold standard” to
degree and clinical symptoms, which usually improves sponta- determine the accuracy of scalp and sphenoidal electrodes.
neously within 3 to 5 days postoperatively, or with steroid use. Compared to discharges retrieved in FOE, scalp electrodes over
Regarding the use of steroids, there are two studies on the the anterior temporal lobe (1 cm anterior to T1/T2) retrieved
use of dexamethasone in patients undergoing monitoring with 95.6% of the discharges but midtemporal scalp electrodes
subdural electrodes. The prophylactic use in children was felt retrieved only 77.7% (T3/T4) (71). Rarely (3%), the interictal
efficacious for brain swelling, as determined by CT changes discharges showed up in the anterior temporal scalp electrode
manifest by midline shift, decrease of ventricular size, and so but not in the FOE. Discharges noted in sphenoidal electrodes
on, although it led to slightly longer (~1 day) seizure-monitor- arose mainly in mesial temporal cortex, but not exclusively,
ing time (64). The benefit of steroids during a limited period of since in the group of 21 patients studied, 3 also had spikes ori-
3 days in adults seems to be less clear (65). In a randomized, ginating from the orbitofrontal and temporal neocortex (72).
placebo-controlled study of 30 patients, less pain and nausea
were reported, but the effect was limited over time. INTERICTAL INTRACRANIAL EEG
There is no study on the yield of antibiotics during IEEG.
Most centers consider prolonged antibiotics more with sub- All normal physiologic rhythms, for example, alpha, theta (dur-
dural recordings as compared to depth electrodes. Other centers ing drowsiness), and mu, which can be seen in scalp recordings,
give single or short-term period during and shortly after the appear also in IEEG at the corresponding cortical sites, that is,
operation, while others use them also in patients with depth mu over sensory–motor cortex. However, due to the absence of
electrodes. Up till now, no study supporting the view of better scalp and skull, all EEG patterns look more “spiky.” Scalp and
protection against nosocomial infections with antibiotic ther- skull filter out in particular high frequencies, including spikes,
apy exists. whereas slow frequencies pass. Thus, only a small fraction of
intracranial spikes are visible in scalp EEG. If an area of less
COMPARISON OF YIELD BETWEEN than 6 cm 2 is active, no recognizable spike activity is found on
DIFFERENT ELECTRODE TYPES the scalp. Discharges from areas between 6 and 10 cm 2 resulted
in 10% spikes, and those from areas 10 cm 2 resulted in 90%
Depth and subdural electrodes are the intracranial electrodes of spikes that were retrievable in scalp EEGs (24).
that are most frequently used. However, each center has its own It has been reiterated that IEEG has the advantage of being
preferences and experiences that may influence the choice of devoid of muscle artifacts; however, this might not be entirely
one over the other. true as indicated by a recent case report (73). In this 17-year-old
Recordings from deep temporal structures, in particular from boy, muscle artifacts appeared at inferolateral temporal
the amygdala, yield better results with depth electrodes (66–68). subdural electrodes, using another intracranial electrode as ref-
Subdural electrodes are also helpful in identifying a mesial erence, while eating or during involuntary facial movement as
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 685

part of the seizure semiology. Thus, muscle activity may be The intracranial interictal EEG correlates of dysplastic tissue
picked up by intracranial recordings, similar to any other inter- have been investigated, since dysplasia and/or its extent are
fering electrical activity, albeit its appearance may be different often invisible in the MRI. Rhythmic spike-wave complexes in a
from muscle artifacts in scalp recordings. continuous or quasi-continuous fashion are most likely to be
If high frequencies remain in the EEG, everything appears associated with cortical dysplasia (79,80). In fact, in patients
“epileptogenic,” which occasionally has led to incorrect EEG with dysplasia, the analysis of “interictal” anomalies seemed to
interpretation. This may be the reason why most authorities in be more promising than that of ictal intracranial recordings
the field recommend relying mainly on the ictal data, and less on (81). Other studies did not find any difference in interictal spik-
the interictal EEG. Consequently, in intracranial recordings ing pattern between dysplastic and nondysplastic cortex (82).
interictal discharges appear more often multifocal and more fre- Continuous discharge pattern are also found with severely gli-
quently than in surface recordings. To determine the relevance of otic tissue (83), but the difference between dysplasia and severe
the interictal IEEG spikes in a given location, several approaches gliosis should be obvious already from the patient’s MRI.
are conceivable. First, the determination of the underlying area In conclusion, disregard for interictal discharges in the EEG
(see above): if the spikes are seen over an area of 10 cm 2 or more and concentration only on ictal recordings is an obsolete con-
(as estimated by the number of simultaneously active elec- cept if only “true” interictal spikes are considered. Careful
trodes), they arise most likely from the epileptogenic zone (see analysis of the localization and frequency of interictal spikes in
above). Second, a quantitative analysis, regarding the relative fre- IEEG can significantly add to the detection of the epileptogenic
quency of spikes from different regions, may help to determine zone. Recently, studies were presented that aimed at the brain
their relevance. This could be done by visual analysis or through “current source” reconstruction from intracranial recordings
automatic or semiautomatic detection algorithms. The latter (84), similar to algorithms for scalp EEG. Future studies will
were used in a study of IEEG in 32 patients (24 TLE and 8 determine their feasibility and correctness in a clinical environ-
extratemporal lobe epilepsy), with mostly combined depth and ment (see also Chapter 57). If sophisticated analysis of interictal
subdural electrode recordings (74). Spikes with highest fre- IEEG provides sufficient data on the seizure-onset area and
quency, highest amplitude, and shorter duration originated resection leads to seizure freedom in most cases, recording time
most often from the ictal onset zone, that is, 2 cm from the site could be significantly shorted and result in lower morbidity and
of seizure origin. Another study in 13 children, including also costs. Electric source localization of interictal scalp EEG corre-
patients with cortical dysplasia, came to similar conclusions. In sponds most often to the ictal onset zone, and its resection leads
that study, subdural electrodes were implanted. Electrodes with to seizure freedom in the majority of patients (21,85).
the highest spike frequency and amplitude were closest to the
area of seizure onset in most of the patients (75), and 11 out of ICTAL INTRACRANIAL EEG PATTERN
13 patients were seizure-free (Engel’s class I (76)). The yield of AND THEIR RELEVANCE TO OUTCOME
careful interictal analysis was studied in patients with exclusively
extratemporal lobe epilepsy, including cortical dysplasia. No Despite promising results from interictal intracranial studies in
quantification for different locations of interictal spikes was pro- terms of localization of the epileptogenic zone, ictal recording
vided, except mapping of the extension of the interictal epilep- still remains the chief goal of IEEG. Till date, there is no consen-
togenic zone including depth and subdural electrodes. If the sus of how many seizures are required to be confident enough to
resection volume included the area of dominating interictal recommend surgery. Most centers try to obtain a minimum of
spikes, good surgical outcome was significantly more likely (77). five seizures or more if multifocal origin is suspected. The goal is
The definition of IEEG spikes is mostly qualitative and to determine a principal seizure-onset focus. However, strict
adopted from scalp EEG. Criteria include a morphologic pat- rules are probably not possible, given that intracranial record-
tern that has (a) triangular waveform, with or without a follow- ings cannot be extended indefinitely due to medical concerns.
ing slow wave, (b) has duration of 70 msec, and (c) stands out As a general rule, the chances of seizure control are markedly
clearly from the background with its amplitude and steep com- better if the EEG onset precedes the clinical onset. The reasons
ponent. Given the difficulties to determine unambiguously the are obvious. If the seizure is already ongoing clinically, without
presence or absence of spikes, spike detection programs for focal EEG changes, there is a strong risk that the electrodes do
scalp EEGs were developed and applied also for IEEG (78). not cover the zone of seizure generation, and any conclusion of
Three of those algorithms were tested for IEEG from seven the true area of epilepsy onset remains obscure. Furthermore,
patients (six with TLE) and compared to analysis by visual the chance of postoperative seizure control is higher if there is
inspection from two human reviewers. The agreement between only one type of seizure semiology. This is true for ictal scalp,
the two EEG experts was relatively small with less than 50% of and is also true for ictal intracranial workups. However, since
all spikes identified by both reviewers, a number that compares the noninvasive “phase 1” workup usually precedes the invasive
similarly to scalp EEG. Compared to the human reviewers, the “phase 2” workup, the information on how many different
automatic algorithms were concordant in only 24% to 32%. seizure types are present is usually already provided.
However, despite the low number of agreement between the A large body of studies looked into a possible relationship
human experts and computer, this affected rather the number between intracranial ictal EEG pattern, its location, and surgical
of detected spikes but not the location: both reviewers and outcome. Although most early studies included patients with TLE
automatic systems identified the spikes in the same anatomical using depth electrode recordings, later observations were extended
localizations. to subdural electrode recordings and/or extratemporal epilepsies.
686 Part V ■ Complementary and Special Techniques

Concerning iEEG, the impact of focal versus regional or dif- Tabl e 3 3 . 3


fuse onset has also been examined in several studies. A higher
chance of postoperative seizure freedom is associated with onset Intracranial Ictal Onset Pattern
in only few electrodes (86–88). Focal onset, defined as ictal activ-
ity in less than four to five electrode contacts, appears to be a Low-voltage fast activity (LVFA; 13–20 Hz)
sign of proximity of the recording contacts to the epileptogenic • Beta range (13–30 Hz)
zone, so its resection is likely to result in seizure control or major
• Gamma range ( 30 Hz)
seizure reduction in up to 85% of patients with fast frequency
onset (88,89). This relationship was less evident in another study Rhythmic spiking 1–2 Hz
of neocortical, mostly extratemporal epilepsy (90), which Rhythmic sinusoidal pattern in alpha–theta range (around
reported no difference between focal versus nonfocal onset. 5–9 Hz)
Concerning intracranial ictal EEG pattern, onset rhythms were
Rhythmic spiking in the alpha–theta range
classified either in terms of frequency bands (92) or as certain
morphologic patterns (88). Since fast rhythms of cortical origin Semirhythmic slow waves 5 Hz
are not filtered by skull and scalp, ictal onset, often characterized High-amplitude beta spike activity
by low-amplitude rapid beta or gamma frequencies, are seen eas-
ier but most importantly earlier than on scalp EEG. In fact, fast
EEG onset frequencies (in the beta or gamma range) are the most
frequently reported EEG patterns at onset (91,92), significantly
related to class I and II outcomes (89,93,94). According to Lee et indicate multiple seizure origins. In a study of 18 patients with
al., extratemporal onset is faster and found in the gamma range TLE, more than one ictal EEG onset pattern was found in most
( 30 Hz), whereas temporal lobe onset was seen in the beta-range patients (87). In an earlier study, it has been underlined that the
frequency (88). These initial fast EEG patterns are also often asso- variability of the onset frequency is a characteristic of TLE and
ciated with focal onset, and therefore, evocative of closeness of the rarely found in extratemporal cases (92). However, in both
electrodes to the seizure-onset zone. Slower EEG onset frequencies studies, surgical outcome was not reported, that is, it remains
( 5 Hz), probably indicate that there is already propagation from unknown if variability of onset EEG pattern holds prognostic
a focus elsewhere (88,95). During slow iEEG patterns, the patient significance.
may show loss of responsiveness, automatisms, and other behav- Apart from onset IEEG pattern, the further evolution of the
ioral signs of already ongoing seizure activity (95). Consequently, seizure pattern and its prognostic significance has been addressed.
better surgical outcome is reported with focal, faster ictal EEG The speed of propagation represents an important factor. Fast
onset, although other evidence indicated that slow onset is not a propagation, that is, within one to a few seconds, to remote
bad sign in all circumstances. If slow-onset ictal IEEG contains regions is related with a less favorable surgical result (90,100). This
poor prognosis, then it is associated with developmental pathol- appears to be equally true for temporal and extratemporal lobe
ogy, regional distribution, or extratemporal location (only 1 in 16 epilepsies. Interhemispheric propagation time in patients with
become seizure free) (88). TLE was analyzed, and a delay of only 0.5 to 5 seconds was asso-
Other ictal IEEG patterns include sinusoidal pattern, preictal ciated with a poor prognosis (101). It is assumed that rapid or
spiking, or high-amplitude beta spiking. Sinusoidal pattern was even almost simultaneous EEG onset at two distant sites such as
only seen with mature pathologic substrates. Its prognosis is both temporal lobes points to a pacemaker elsewhere with rapid
unclear. It was felt to be a sign of propagation (94), although implication to the two sites. Alternatively, rapid propagation could
this was less clear in another report (88). suggest diffuse damage of remote areas, each with significant
Next to low-voltage, fast beta activity, periodic spiking before epileptogenic properties of its own.
seizure onset was frequently observed. It represents another ictal Propagation may occur through different EEG patterns. Two
onset pattern and was found to be related to seizures of hip- propagation patterns at distant sites were identified by Schiller
pocampal and amygdala onset (92,96–98). Ictal spiking at et al. in temporal and extratemporal lobe epilepsies (94): first,
seizure onset is correlated to the presence of hippocampal scle- rhythmic theta–delta activity correlating with the activity at the
rosis and/or neuronal decrease in the CA1 region of the hip- seizure-onset site, and second, continuation of the seizure pat-
pocampus, that is, which is in itself a favorable sign with respect tern of the seizure-onset site. No significant correlation
to postoperative outcome (98). Consequently, ictal onset spiking between onset and propagation EEG pattern and outcome was
is related to good prognosis in terms of seizure control. Finally, found. The visual analysis of iEEG during propagation is prob-
high-amplitude beta spike activity has also been described (99) ably less relevant in the clinical context, and may even differ
in TLE, but it is rare and its prognosis is still unclear. within a patient from one seizure to the other. It remains to be
Although the intracranial ictal onset pattern was not predic- shown if coherence-based measurements are more informative,
tive of the underlying pathology, low-voltage fast activity and that is, if they help to identify nonfocal epilepsies (102).
rhythmic alpha–theta spike-wave were more common in devel- Finally, the end of the intracranial ictal EEG pattern (offset)
opmental substrates (e.g., dysplasia) (Table 33.3) (88). was studied in order to determine if offset outside the generat-
Another study subject concerns the variability of ictal onset. ing structures is related to worse outcome. Theoretically, it
The rational behind it is that variable onset patterns could could indicate the presence of epileptogenic tissue in remote
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 687

sites, ready to pick up seizure activity from elsewhere. However, low-pass filter, instead of the usual 200 to 250 Hz sampling rate
there was no prognostic difference between patients with an and 70 to 100 Hz low-pass filter used for the routine iEEG.
ipsilateral and contralateral offset (103), but this is challenged HFOs are recorded with the commercially available subdural or
by reports that find that patients with contralateral offset have depth electrodes or with microwires that are inserted together
poorer surgical outcome (104,105). However, as for the onset, with macroelectrodes (Figs. 33.5 and 33.6).
the offset pattern can also vary within one patient, and more HFOs are short bursts of high-frequency pattern that
studies are probably needed to determine the prognostic rele- become visible if the EEG is sampled with a wide bandwidth of
vance of the offset pattern. up to 5000 Hz. Bradin and coworkers differentiated ripples (60
Examples of ictal EEGs, exemplifying the different EEG to 160 Hz) and fast ripples (FRs) (250 to 500 Hz) (108), a
onset and offset patterns described above, are found at the end nomenclature that has been taken up by similar studies, albeit
of the main text. the definition of lower and upper limits of the ripple frequen-
Although there have been many studies on the value of ictal cies is slightly different from study to study. Both ripple types
IEEG, many questions remain on the prognostic significance of increase during slow-wave sleep (109,110), which may explain
a given EEG pattern. It appears that several variables influence the propensity of seizures to occur during periods of decreased
the creation of an ictal IEEG pattern, and these variables vigilance or sleep. However, there is evidence that HFO of ripple
include the affected lobe, underlying histopathology, the type of frequency is also found in the nonepileptogenic temporal lobe,
electrode and sites of contacts, age of patient, and speed of whereas FRs are equally predominant in wakefulness and sleep,
medication withdrawal. So many variables exist that most stud- that is, they do not appear to be a physiologic event (see review
ies are underpowered. Multicenter studies making use of mod- of subject in Chapter 37).
ern 3D localization of all contacts could help to better identify Across several studies and groups, it was claimed that FRs are
the significance of ictal IEEG pattern. more often pinpointing to the ictal epileptogenic zone than
HFO of lower frequency (111,112,115); however, not all studies
HIGH-FREQUENCY OSCILLATIONS came to the same conclusion (113). They may occur alone or
superposed on the spike (107). Overall, FRs appear to be a more
In recent years, the research on the markers of epileptogenic tis- localized event compared to HFOs of lower ripple frequency
sue in the interictal EEG have been restarted, given that the (112–114). They may also precede the seizure onset for up to
new-generation recording systems with higher EEG sampling several seconds to 20 minutes (109,115), but are not present in
rates can capture very fast rhythms (30 to 500 Hz). Probably the all patients (115). If HFOs indeed indicate the zone of seizure
first study on that subject was published by R. Fisher and his onset, longer EEG recordings requiring the capture of several
colleagues (106) who found increased high-frequency activity, seizures and confirming the presence of a unifocal ictal onset
40 Hz, in the seizure-onset zone. Many more studies followed, zone would become obsolete. However, the issue is far from
supporting the notion that high-frequency oscillations (HFOs) resolved. Recordings from both macro- and microelectrodes
are more frequently colocalized with the seizure-onset zone, showed that the morphology of HFOs of both types, FRs and
albeit the exact shape and frequency band of what is truly ripples, also occur together, that is, the burst starts with ripples
evocative of the epileptic focus still needs to be defined (107). and evolves to FRs, or inverse (112). FR HFOs were often
They are believed to reflect inhibitory field potentials, which recorded from a single microwire (108,112), indicating a very
facilitate information transfer by synchronizing neuronal activ- local event from a neuronal assembly. How and when such a
ity over long distances. These recordings require a high sam- local and short discharge (20 to 50 msec) evolves to clinical
pling rate, that is, 2000 to 5000 Hz and a 500 to 1000 Hz seizures of several minutes remains to be elucidated.

Figure 33.5 Microelectrode arrays.


Left: So-called Utah microarray
made of multiple silicon spikes of
1.0 or 1.5 mm length. A pressure
applicator is used to insert the sili-
con array into the cortex across the
pia. Right: Microelectrodes as a
bundle at the end of a depth elec-
trode. (From Turner DA, Patil PG,
Nicolelis MAL. Conceptua l a nd
Technical Approaches to Human
Neura l En semble Recordings.
Methods for Neura l Ensemble
Recordings, Frontiers in Neuro-
science. CRC Press; 2008.)
688 Part V ■ Complementary and Special Techniques

Figure 33.6 High-frequency oscil-


lations recorded from macro- and
microelectrodes from the left mesial
temporal cortex, harboring also the
seizure-onset zone. Note the coinci-
dence of the spike in the macroelec-
trodes, and ripples recorded in
the microelectrodes. (From Worell
et al., Brain, 2008.)

Research on human HFOs is relatively recent. Most of the for intracranial monitoring if the exact focus localization is not
studies are done in mesial temporal lobe structures, given that yet evident from noninvasive monitoring. Like in adults, addi-
TLE patients are more frequent in the surgical patient popula- tional electrocorticography may be necessary to identify elo-
tion and/or more frequently investigated with intracranial and quent cortex.
microwire electrodes together. The existence of ripples or FRs In older children, the corticography procedure is similar to
in extratemporal lobe epilepsy is less well known up till now, that in adults (118). Trains of 50 Hz, alternating polarity
but preliminary data suggest that they are also present in square-wave pulses of fixed pulse duration (0.3 msec) are most
extratemporal areas (113). Thus, the most important question often used, starting with low currents, for example, 0.5 or 1 mA.
to be resolved concerns which type of HFO is truly pathologic The stimulation intensity is gradually augmented in 1-mA steps
and potentially serves as biomarker (107). It is of interest to until a sensation or movement is elicited or the maximal value
note that recent studies found paroxysmal fast activity even in has been reached (up to a maximum of 10 to 15 mA, without
scalp recordings, mainly in pediatric patients in the beta and any perceptible sensation or movement). However, in very
gamma frequency bands (15 to 70 Hz) (116,117). young children, this approach is often ineffective, since motor
responses are virtually unobtainable under 1 year of age, and
SPECIAL CONSIDERATIONS with difficulties until 4 to 5 years of age (119). The Miami
IN PEDIATRIC PATIENTS group described a different stimulation procedure in children
younger than 5 years, which results more often in motor move-
Invasive procedures are rare in infants younger than 3 years, but ments: they propose dual increment of pulse duration and
if they are indicated, they should be done at specialized epilepsy stimulus intensity instead of increment of only stimulus inten-
surgery centers. Close collaboration with the (neuro) pediatri- sity (120). The minimal current intensity (in milliamperes)
cians is mandatory if the center is not already located in a pedi- required to elicit a response at very long pulse duration (in mil-
atric department. In contrast to previous attitudes, children liseconds) is called rheobase. The double of rheobase is called
with epilepsy should be investigated as soon as pharmacoresis- chronaxie, and corresponds to the pulse duration required to
tance is documented. To wait until the epilepsy “grows” out is elicit a response twice as intense as the rheobase. In nonmyeli-
no more appropriate unless the underlying syndrome is well nated fibers, the chronaxie is longer than that of myelinated
determined and surgical treatment is clearly not indicated, for fibers. In very young children, myelination is still incomplete,
example, Dravet syndrome. Thorough workup helps to gradually increases with age and is also site dependent; that is,
determine the underlying syndrome and potential usefulness of myelination starts in the posterior cortices. Jayakar et al. inves-
surgical therapy. Not all children with early-onset difficult tigated the dual-increment approach in six patients between 1
epilepsy are surgical candidates, but if they are, any year of per- and 10 years using pulse duration between 0.3 and 1 msec and
sistent seizures has deleterious effects on their development. If increasing intensity starting at 1 mA up to 15 mA. With dual
the indication for IEEG is given, the same implantation strate- increment, that is, alternating 1 to 2 mA and 0.1 to 0.2 msec
gies, electrode types, and most importantly, the clinical reason- increments, motor responses were obtained in three patients of
ing regarding the goals apply as for adults. 1, 3, and 4.5 years, respectively, and afterdischarges in 12/14
Pediatric patients suffer more frequently from extratemporal electrodes, but no response with the adult paradigm. In three
lobe epilepsy. Consequently, they are considered as candidate older children, aged 5.5, 7, and 10 years, the adult paradigm,
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 689

that is, with fixed pulse duration of 0.3 msec, as well as the dual- possible brain regions can be covered, and more importantly, the
increment paradigm, both elicited responses and afterdis- complication risk increases and may outweigh the benefit of the
charges in 16/16 contacts. IEEG procedure. To obtain a valid hypothesis, coregistration and
An alternative approach in children (or adults) who are not multimodal imaging may help to increase the yield of the inva-
able to describe their feelings or collaborate with the examiner sive phase (124). Whole-head scalp EEG (and MEG) arrays of
is evoked potentials. In pre- or peroperative monitoring, 200 or more electrodes/sensory, together with EEG source local-
somatosensory or motor evoked potentials identified the local- ization algorithms, were successfully employed in pediatric and
ization of the central sulcus (121,122). adult epileptic patients with temporal and extratemporal lobe
epilepsy for focus localization (125,126).
OUTLOOK IEEG may be used for other purposes in addition to determin-
ing the seizure focus. There are a number of studies, albeit limited,
Evaluation of patients with intracranial electrodes has under- that try to make use of IEEG recordings to determine neuropsy-
gone changes in terms of defining the indication for their use. chological functionality of distinct brain structures. For example,
Most patients with mesial temporal epilepsy and hippocampal ongoing seizure activity was used as “electrical Wada test,” (127)
sclerosis do not need IEEG monitoring if they have access to presence or absence of N400 components, as well as HFOs as indi-
modern imaging techniques. Even patients with nonlesional cator of (healthy) mnestic processes (128–131) or event-related
epilepsy may not always need intracranial evaluation (123). gamma activity during naming to localize language cortex (132).
Despite advances in brain imaging, the rate of seizure-free Intracranial monitoring imposes a certain burden on the
patients is still lower in patients with extratemporal compared to patient and his/her family, both on the psychological and phys-
TLE. The precision and yield of noninvasive imaging for patients ical levels, and the indication must be carefully weighted against
with extratemporal lobe epilepsy is still under evaluation and the possible benefit. Across most centers and studies, the num-
will further improve. However, in the meantime patients with ber of patients who cannot be offered surgery, despite intracra-
conflicting results from the different exams in the noninvasive nial monitoring, is around 25% to 30%. Future studies will
phase (phase 1) are still candidates for intracranial monitoring. show if with the addition of sophisticated noninvasive brain
As stated in the beginning of this chapter, each intracranial eval- imaging this percentage can be lowered, either because non-
uation needs a solid hypothesis as to where the seizures might surgical candidates can be more easily identified or because
have their origin. IEEG should not be a “fishing expedition,” the electrodes are placed closer to the epileptogenic zone. See
because even with EEG systems of 200 or more channels, not all Figures 33.7 to 33.21.

Figure 33.7 A: Dissociation between seizure-onset zone and symptomatic zone. Patient CA, 22-year-old male with left hip-
pocampal sclerosis but nonlocalizing ictal scalp pattern. Continuous discharges in the left hippocampus (depth electrodes;
HAG1, HPG1, HPG2) as interictal EEG. AD, right amygdala/ anterior temporal lobe; HD, right hippocampus/ midtemporal
lobe; G-contacts, subdural grid electrodes overlying the lateral temporoparietal cortex.
(Continued on next page)
Figure 33.7 (Continued) B: Position of depth elec-
trodes in the left and right temporal and frontal cortex,
and subdural grids over temporoparietal cortex.
Coregistration of high-resolution CT (with the elec-
trodes) and the patient’s MRI. C: The interictal rhythm
changes to rhythmic appearance of bursts of rapid 30 to
40 Hz pattern, appearing every 6 seconds. D: The hip-
pocampal bursts stop and are replaced by widespread 10
Hz rhythms in the lateral fronto-orbital cortex (FOG7)
and lateral temporal subdural contacts (contacts GA,
GB, GC, GD), coinciding with the onset of clinical symp-
toms (*), that is, lip smacking, manual automatisms,
loss of contact.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 691

Figure 33.7 (Continued) E: The extrahippocampal discharges persist while the left hippocampus remains silent. This case
represents an example of a dissociation between areas of onset and the symptomatic propagation, which implicates rather
lateral neocortical contacts.

Figure 33.8 A: Slow ictal onset EEG pattern ( 5 Hz). Patient VA, 12-year-old female with nonlesional left hemispheric
epilepsy with dysesthesia in the right foot. Slow onset (2 Hz waves) in the left superior parietal region (→), corresponding
to the dark gray circled contacts. Five seconds later: rhythmic spikes in the adjacent region (**). IA and IP, anterior and
posterior temporal subdural strips (not shown).

(Continued on next page)


692 Part V ■ Complementary and Special Techniques

Figure 33.8 (Continued) B: Continuous rhythmic 5-Hz spiking in the contacts close to the sensory foot area, related to
the patient’s typical aura. However, persistent seizures postoperatively after a limited superior parietal resection suggest
that the site of ictal onset does not coincide with the epileptogenic zone. Slow-onset rhythms ( 5 Hz) are considered to
be less likely associated to the true epileptogenic zone. This is true even when the slow onset pattern evolves to rhythmic
discharges. Seizures continued after limited resection in the postcentral region.

Figure 33.9 A: Focal onset spiking. Patient AF, 18-year-old with nonlesional left frontal epilepsy. Localized onset as rhyth-
mic spiking (→) in four selective subdural contacts (C3, C4, D3, D4) over the lateral anterior left frontal cortex. Within
5 seconds spreads to other adjacent contacts.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 693

Figure 33.9 (Continued) B: (Right) Localization of subdural electrodes coregistered with the patient MRI. Contacts of
seizure onset (see Figure 33.8) are shown as big, dark gray circles. The IEEG localization of the ictal focus was concordant
with the localization obtained by noninvasive electric source localization (left; see also Chapter 55). C: Persistent rhythmic
spike-wave pattern continued. No clinical symptoms were seen or noted by the patient. A topectomy of the cortex under-
lying the four contacts resulted in marked improvement of the epilepsy disorder (i.e., one to two GTCS per year if AEDs
were forgotten). Focal onset, that is, onset in less than five contacts is more likely related to good outcome, as it is the case
here. Independently, ictal onset spiking has been described as another favorable sign in hippocampal seizures, it may also
be true for extratemporal lobe epilepsy.
694 Part V ■ Complementary and Special Techniques

Figure 33.10 A: Low-voltage beta activity at ictal onset. Patient CR, 20-year-old, right-handed, with transmantle dysplasia
in the left posterior temporal cortex. Onset was characterized by rapid rhythms ( 25 Hz) in two distinct contacts (D4 C4).
Left: Schema of electrodes coregistered to the patient’s brain. Contacts corresponding to early ictal activity in black. B:
Propagation to other contact over other temporal lateral cortex is noted. Discharges remain relatively well localized, but
become associated with clinical symptoms (*).
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 695

Figure 33.10 (Continued) C: Discharges continue, implicating now the whole temporal lobe, and to a lesser degree also
frontal regions. The onset contacts remain the most active—low-amplitude beta activity onset is considered a favorable
sign in terms of seizure control. Indeed, resection of this area resulted in persistent seizure freedom ( 5 years).
Comprehension was found to be localized in the contralateral hemisphere.

Figure 33.11 A: Diffuse slow ictal onset. Patient FS, 42-year-old male, with right hippocampal sclerosis and multiple right
hemispheric heterotopia. Poorly defined ictal onset: diffuse onset with 1 to 2 Hz sharp slow waves in right frontal (CD7-8),
right lateral temporal (AD6-8), right parietal periventricular heterotopia (HPPD6-8). Left: Depth electrode position coregis-
tered with the patient’s brain.

(Continued on next page)


696 Part V ■ Complementary and Special Techniques

Figure 33.11 (Continued) B: No further localization, but synchronous 1 to 2 Hz discharges of a network comprising
fronto-orbital (FOD), right frontal lateral (CD), lateral temporal (AD), both right periventricular temporal (HPPTD), and
parietal (PAD) heterotopia. C: Recording of another seizure, with simultaneous onset of the temporal lobe, heterotopias,
and frontal structures, preceded by a brief hippocampal burst (HPD) of 6 Hz sharp waves. Resection of the hippocampus
and partially the anterior heterotopia brought only partial seizure relief (decrease of 50% ). Diffuse onset, even in the pre -
sence of structural lesions, is less likely related to good postoperative seizure control. Moreover, variable seizure-onset pat-
terns were noted, another unfavorable prognostic factor.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 697

Figure 33.12 Gamma activity at onset. Patient MS, 15-year-old female with tuberous sclerosis. Largest tuber in the left
posterior temporal lobe. Seizure onset presents as rapid gamma activity ( 100 Hz; GG5-8; GH1-2). Incterictally (before
seizure onset) more widespread discharges over the left posterior cortex.

Figure 33.13 A: Patient MS: 80 to 100 Hz ripples at the site of seizure onset (GG3). Left: Coregistration of the patient’s
subdural electrodes position with her own brain. Site of ripple is indicated by dark gray circle.

(Continued on next page)


698 Part V ■ Complementary and Special Techniques

Figure 33.13 (Continued) B: Patient MS: Diffuse semirhythmic sharp waves. Close analysis reveals the presence of rapid
ripples riding on the discharges (max GH2). Resection of the tissues underlying these ripples resulted in seizure freedom
(follow-up 2 years), despite more diffuse spike and sharp wave discharges. Not only focal beta but also gamma activity at
seizure onset is a good prognostic factor, despite the presence of multifocal lesions.

Figure 33.14 A: Preictal spiking. Patient VT, 28-year-old, with left hippocampal sclerosis. Monitoring was advised due
to nonlateralizing seizure onset in ictal scalp EEGs. Depth electrodes were inserted orthogonally in both hemispheres.
(Right: FOD, fronto-orbital; CD, frontal lateral cortex/ anterior cingulate gyrus; AD, anterior temporal lobe/ amygdala;
HAD/ HPD, anterior/ posterior hippocampus. Left: FOG, CG, AG, HAG, HPG). Before the seizure, preictal spiking is noted
in the sclerotic left hippocampus (HAG2-3, HPG2-3).
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 699

Figure 33.14 (Continued) B: Inhabitual propagation pattern. Patient VT: Preictal spiking is replaced by rhythmic spiking
in the left hippocampus (HAG2, HPG2, →). Recruitment occurs first in the lateral frontal cortex (FOG7-8, *). Small ampli-
tude discharges in the left amydala become more rhythmic (AG1-2, 2-3, **). The smaller amplitude is explained by the closed
electrical field of the amygdala as compared to the hippocampus. C: Recruitment in the lateral frontal cortex (FOG7-8)
continues. Close analysis of the temporal pattern reveals that the discharges in the fronto-orbital cortex are 50 to 100 msec
later than the large hippocampal spikes, indicating that the hippocampus drives the fronto-orbital discharges. No propaga-
tion to lateral temporal contacts (HAG7,8; HPG7,8), explaining retrospectively why the scalp EEG was unrevealing.
Propagation pattern can show significant variability between patients and may involve even structures outside the lobe of
onset, including the contralateral hemisphere.
700 Part V ■ Complementary and Special Techniques

Figure 33.15 Low-amplitude gamma activity in extratemporal lobe epilepsy. Patient SC, 17-year-old, with nonlesional
right frontal epilepsy with daily nocturnal generalized tonic-clonic seizures (GTCS). Seizure onset is characterized by a very
focal beta activity (GA7), which diffuses to other subdural contacts. Resection of the epileptogenic zone including the cor-
tex underlying GA7 resulted in disappearance of GTCS (except once after sleep withdrawal), but persistence of short simple
partial seizures (sensory–motor symptoms of the tongue). Focal low-amplitude rapid ictal onset activity is favorable sign
also in extratemporal lobe epilepsy.

Figure 33.16 A: Significance of different onset patterns. Patient BE, 43-year-old, with bitemporal lobe epilepsy with left-
sided predominance, began after vaccination at the age of 7 years, MRI: bilateral atrophy. Onset is characterized by beta
activity in the left parahippocampal gyrus (LH4-5 and adjacent contacts, →). RF/ LF, right/ left fronto-orbital cortex; RA,
toward right amygdala (LA, left amygdala); RH/ LH, toward right/ left hippocampus.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 701

Figure 33.16 (Continued) B: Offset pattern of this seizure in the same contacts as the seizure onset (parahippocampal
gyrus, LH3-4, LH4-5). C: Another seizure that also started with focal beta activity but is now in the left amygdala and hip-
pocampus (LH1-2, LA2-3, LA1-2, →), leading to recruitment of right mesial temporal structures (RA1-2, RA2-3, RH-1-2, *);
variable onset pattern are rather less favorable signs. Indeed, selective resection of left mesial temporal structures did not
lead to persistent seizure freedom, albeit reduction of approximately 70% .
702 Part V ■ Complementary and Special Techniques

Figure 33.17 Diffuse EEG onset. Patient SG, 20-year-old male, with epilepsy onset at the age of 3. The scalp EEG showed
almost continuous discharges over the left frontal lateral cortex. 8 8 subdural grid was implanted over the left frontal
and temporal cortex. Seizure onset (→) was noted simultaneously over frontal and temporal regions (B5–6, 6–7, 7–8;
H5–6, 6–7, 7–8). Limited resection of left frontal cortex due to the presence of language cortex (histopathology: severe cor-
tical dysplasia) did not control seizures persistently.

Figure 33.18 A: Ictal recording with foramen ovale electrodes (FOE). Patient ES: 21-year-old, left-handed, known for
seizures since the age of 9. Left hippocampal sclerosis. Scalp ictal EEG showed left, right, or bitemporal onset. Interictal
spikes were found over left and right temporal lobe with variable predominance. FOE were inserted bilaterally (left: L1-6;
right: R1-6), and recorded together with scalp electrodes. Ictal recording with first changes in the left FOE.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 703

Figure 33.18 (Continued) B: The seizure evolves, with few changes in the corresponding scalp electrodes. C: A low-
amplitude beta activity starts (*).

(Continued on next page)


704 Part V ■ Complementary and Special Techniques

Figure 33.18 (Continued) D: Beta activity persists, with still few changes in the scalp EEG and no clinical symptoms.
Thus, FOE is capable of recording focal hippocampal ictal activity, with higher sensitivity as scalp EEG.

Figure 33.19 A: Subclinical seizures. Patient VT, 28-year-old, with left hippocampal sclerosis. Subclinical seizure in the
left sclerotic hippocampus ( 30 Hz beta activity) with very focal onset (in one contact, HPG2). No further recruitment is
observed. Clinical symptoms are limited to deficient verbal memory.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 705

Figure 33.19 (Continued) B: Subclinical seizure pattern gradually stops and discharges are of lower frequency.
Multichannel IEEG needs to be carefully scrutinized, because they may involve only one channel. If frequent, they usually
indicate a high epileptogenicity of the recorded structure.

Figure 33.20 A: Ictal beta activity, less focal. Patient RP, 24-year-old, diagnosed with nonlesional right temporal epilepsy.
Noninvasive workup found right anterior and posterior temporal discharges. After ESI localizing the focus rather to poste-
rior temporal regions, careful review of the MRI showed focal polymicrogyria in the temporo-occipital junction. A subdural
grid was placed there, as well as depth electrodes in the left and right temporal (right: AD, HAD, HPD; left: AG, HMG)
and frontal regions (CD, FOD).

(Continued on next page)


706 Part V ■ Complementary and Special Techniques

Figure 33.20 (Continued) B: Beta activity persists, becoming more rhythmic and organized. Around 10 seconds after EEG
onset in the temporo-occipital cortex, recruitment of the right hippocampus (→). C: Persistence of right-sided seizure activ-
ity, with left hippocampal recruitment (→).
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 707

Figure 33.20 (Continued) D: Posterior and bihippocampal discharges persist. This recruitment pattern may explain why
the patient is amnesic of the entire event. Seizures came back 2 years after resection of the polymicropgyric cortex, but
subsided again after re-adaptation of drug treatment. Less focal ( 5 Hz) EEG onset, together with a less organized rhythm
at the beginning, may reflect more widespread epileptogenic zone.

Figure 33.21 A: Focal onset with late recruitment and contralateral offset. Patient SS, 37-year-old, right-handed, with non-
lesional right temporal lobe epilepsy. Depth electrodes were implanted in both temporal lobes and right frontal lobe. Onset
is characterized by high-amplitude spiking, mixed with beta activity. Onset is restricted to four contacts in the right mesial
temporal lobe (HAD1,2, HPD1,2; AG and HAG, left anterior and midtemporal lobe, respectively; FOD, fronto-orbital; CD,
right frontal lateral).

(Continued on next page)


708 Part V ■ Complementary and Special Techniques

Figure 33.21 (Continued) B: Focal discharges persist, somewhat more recruitment of the right amygdala (AD1-3). Please
note that each page contains 20 seconds of EEG. C: After 29 seconds recruitment of lateral temporal and fronto-orbital
contacts.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 709

Figure 33.21 (Continued) D: Only very late ( 40 seconds) recruitment of the contralateral hippocampus (*). E: While
the right-sided discharges subside, ictal spiking in the left hippocampus persists until the end of the seizure.

(Continued on next page)


710 Part V ■ Complementary and Special Techniques

Figure 33.21 (Continued) F: The patient received a selective right antero-mesial temporal resection and is seizure-free
since (histopathology: cortical dysplasia). Focal onset ( 5 contacts) and persistence focality is an advantageous finding.
However, offset in the contralateral hemisphere is obviously less predictive of postoperative outcome.

ACKNOWLEDGMENTS 8. Seeck M, Lazeyras F, Michel CM, et al. Non-invasive epileptic focus


localization using EEG-triggered functional MRI and electromag-
Supported by the Swiss National Science Foundation (Grant netic tomography. Electroencephalogr Clin Neurophysiol. 1998;106:
Nos. 320030-113766 and 33CM30-124089). MS is grateful to 508–512.
Carlo Schaller for helpful comments on the neurosurgical 9. O’Brien TJ, Miles K, Ware R, et al. The cost-effective use of 18F-
aspects of intracranial EEG. Dr Vitalo Chiosa and Dr Laurent FDG PET in the presurgical evaluation of medically refractory
Spinelli provided invaluable help in the preparation of figures. focal epilepsy. J Nucl Med. 2008;49:931–937.
10. O’Brien TJ, So EL, Mullan BP, et al. Subtraction ictal SPECT co-
registered to MRI improves clinical usefulness of SPECT in local-
REFERENCES izing the surgical seizure focus. Neurology. 1998;50:445–454.
11. Laxer KD. Clinical applications of magnetic resonance spec-
1. Tonini C, Beghi E, Berg AT, et al. Predictors of epilepsy surgery troscopy. Epilepsia. 1997;38(suppl 4):S13–S17.
outcome: a meta-analysis. Epilepsy Res. 2004;62:75–87. 12. Knowlton RC, Elgavish RA, Limdi N, et al. Functional imaging: I.
2. Cendes F, Li LM, Watson C, et al. Is ictal recording mandatory in Relative predictive value of intracranial electroencephalography.
temporal lobe epilepsy? Arch Neurol. 2000;57:497–500. Ann Neurol. 2008;64(1):25–34.
3. Diehl B, Lüders HO. Temporal lobe epilepsy: when are invasive 13. Duncan JS. The current status of neuroimaging for epilepsy. Curr
recordings are needed? Epilepsia. 2000;41(suppl 3):S61–S74. Opin Neurol. 2009;22:179–184.
4. Reuber M, Kral T, Kurthen M, et al. New-onset psychogenic seizures 14. Briellmann RS, Pell GS, Wellard RM, et al. MR imaging of epilepsy:
after intracranial neurosurgery. Acta Neurochir (Wien). 2002;144: state of the art at 1.5 T and potential of 3 T. Epileptic Disord. 2003;
901–907. 5:3–20.
5. Fourth level-epilepsy surgical center for epilepsy. Epilepsia. 1990; 15. Sirven JI, Malamut BL, Liporace JD, et al. Outcome after temporal
31(suppl 1):S7–S8. lobectomy in bilateral temporal lobe epilepsy. Ann Neurol. 1997;
6. Harvey S, Cross JH, Shinnar S, et al. and ILAE Pediatric Epilepsy 42:873–878.
Surgery Survey Taskforce. Defining the spectrum of internation 16. Holmes MD, Dodrill CB, Ojemann GA, et al. Outcome following
practice in pediatric epilepsy surgery patients. Epilepsia. 2008;49: surgery in patients with bitemporal interictal epileptiform pat-
146–155. terns. Neurology. 1997;48:1037–1040.
7. Srikijvilaikul T, Locharernkul C, Deesudchit T, et al. The first inva- 17. Hirsch LJ, Spencer SS, Spencer DD, et al. Temporal lobectomy in
sive EEG monitoring for surgical treatment of epilepsy in Thailand. patients with bitemporal epilepsy defined by depth electroen-
J Med Assoc Thai. 2006;89:527–532. cephalography. Ann Neurol. 1991;30:347–356.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 711

18. Boling W, Aghakhani Y, Andermann F, et al. Surgical treatment of 40. Silberbusch MA, Rothman MI, Bergey GK, et al. Subdural grid
independent bitemporal lobe epilepsy defined by invasive record- implantation for intracranial EEG recording: CT and MR appear-
ings. J Neurol Neurosurg Psychiatr. 2009;80:533–538. ance. Am J Neuroradiol. 1998;19:1089–1093.
19. Mintzer S, Cendes F, Soss J, et al. Unilateral hippocampal sclerosis 41. Kovalev D, Spreer J, Honegger J, et al. Rapid and fully automated
with contralateral temporal scalp ictal onset. Epilepsia. 2004;45: visualization of subdural electrodes in the presurgical evaluation
792–802. of epilepsy patients. Am J Neuroradiol. 2005;26:1078–1083.
20. Carne RP, O’Brien TJ, Kilpatrick CJ, et al. MRI-negative PET- 42. Sebastiano F, Di Gennaro G, Esposito V, et al. A rapid and reliable
positive temporal lobe epilepsy: a distinct surgically remediable procedure to localize subdural electrodes in presurgical evaluation
syndrome. Brain. 2004;127:2276–2285. of patients with drug-resistant focal epilepsy. Clin Neurophysiol.
21. Michel CM, Lantz G, Spinelli L, et al. 128-channel EEG source 2006;117:341–347.
imaging in epilepsy: clinical yield and localization precision. J Clin 43. Mahvash M, König R, Wellmer J, et al. Coregistration of digital
Neurophysiol. 2004;21:71–83. photography of the human cortex and cranial magnetic resonance
22. Brodbeck V, Lascano A, Spinelli L, et al. Accuracy of electrical source imaging for visualization of subdural electrodes in epilepsy sur-
imaging of epileptic spikes in patients with large brain lesions. J Clin gery. Neurosurgery. 2007;61:340–344.
Neurophysiol. 2009;120:679–685. 44. Tao JX, Hawes-Ebersole S, Baldwin M, et al. The accuracy and reli-
23. Gavaret M, Trébuchon A, Bartolomei F, et al. Source localization of ability of 3D CT/MRI co-registration in planning epilepsy surgery.
scalp-EEG interictal spikes in posterior cortex epilepsies investi- Clin Neurophysiol. 2009;120:748–753.
gated by HR-EEG and SEEG. Epilepsia. 2009;50:276–289. 45. Seeck M, Spinelli L. Intracranial monitoring [Review]. Suppl Clin
24. Tao JX, Ray A, Hawes-Ebersole S, et al. Intracranial EEG substrates Neurophysiol. 2004;57:485–493.
of scalp EEG interictal spikes. Epilepsia. 2005;46:669–676. 46. Mahvash M, König R, Urbach H, et al. FLAIR-/T1-/T2-co-
25. Ray A, Tao JX, Hawes-Ebersole SM, et al. Localizing value of scalp registration for image-guided diagnostic and resective epilepsy
EEG spikes: a simultaneous scalp and intracranial study. Clin surgery. Neurosurgery. 2008;62(suppl 2):482–488.
Neurophysiol. 2007;118:69–79. 47. Maurer CR Jr, Fitzpatrick JM, Wang MY, et al. Registration of head
26. Ebersole JS, Hawes-Ebersole S. Clinical application of dipole mod- volume images using implantable fiducial markers. IEEE Trans
els in the localization of epileptiform activity. J Clin Neurophysiol. Med Imaging. 1997;16:447–462.
2007;24:120–129. 48. Spire WJ, Jobst BC, Thadani VM, et al. Robotic image-guided
27. Rosenow F, Lüders H. Presurgical evaluation of epilepsy. Brain. depth electrode implantation in the evaluation of medically
2001;124:11683–1700. intractable epilepsy. Neurosurg Focus. 2008;25(3):E19.
28. Wong CH, Birkett J, Byth K, et al. Risk factors for complications 49. Van Buren JM. Complications of surgical procedures in the diag-
during intracranial electrode recording in presurgical evaluation nosis and treatment of epilepsy. In: Engel J, ed. Surgical Treatment
of drug resistant partial epilepsy. Acta Neurochir. 2009;151:37–50. of the Epilepsies. New York: Raven Press; 1987:465–475.
29. Burneo JG, Steven DA, McLachlan RS, et al. Morbidity associated 50. Ross DA, Brunberg JA, Drury I, et al. Intracerebral depth electrode
with the use of intracranial electrodes for epilepsy surgery. Can J monitoring in partial epilepsy: the morbidity and efficacy of place-
Neurol Sci. 2006;33:223–227. ment using magnetic resonance image-guided stereotactic surgery.
30. Berger H. Über das Elektrenkephalogramm des Menschen. Arch Neurosurgery. 1996;39:327–334.
Psychiatr. 1929;87:527–570. 51. Sansur CA, Frysinger RC, Pouratian N, et al. Incidence of sympto-
31. Penfield W, Jasper H. Epilepsy and the Functional Anatomy of the matic hemorrhage after sterotactic electrode placement. JNeurosurg.
Human Brain. Boston: Little, Brown; 1954. 2007;107:998–1003.
32. Talairach J, Bancaud J. Stereotaxic exploration and therapy in 52. Tanriverdi T, Ajlan A, Poulin N, et al. Morbidity in epilepsy sur-
epilepsy. In: Vinken PJ, Bruyn GW, eds. The Epilepsies. Handbook gery: an experience based on 2449 epilepsy surgery procedures
of Clinical Neurology. Vol 15. Amsterdam: North Holland; 1974: from a single institution. J Neurosurg. 2009;110:1111–1123.
758–782. 53. Almeida AN, Martinez V, Feindel W. The first case of invasive EEG
33. Talairach J, Bancaud J. Stereotactic approach to epilepsy. Prog monitoring for the treatment of epilepsy: historical significance
Neurol Surg. 1973;5:297–354. and context. Epilepsia. 2005;46:1082–1085.
34. Wyler AR, Ojemann GA, Lettich E, et al. Subdural strip electrodes 54. Eröss L, Bagó AG, Entz L, et al. Neuronavigation and fluoroscopy-
for localizing epileptogenic foci. J Neurosurg. 1984;60:1195–1200. assisted subdural strip electrode positioning: a simple method to
35. Ives JR, Mainwaring NR, Gruber LJ, et al. 128-channel cable- increase intraoperative accuracy of strip localization in epilepsy
telemetry EEG recording system for long-term invasive monitor- surgery. J Neurosurg. 2008. DOI: 10.3171/2008.6. JNS17611 [Epub
ing. Electroencephalogr Clin Neurophysiol. 1991;79:69–72. ahead of print].
36. Siegfried J, Wieser HG, Stodieck SR. Foramen ovale electrodes: a new 55. Chamoun RB, Nayar VV, Yoshor D. Neuronavigation applied to
technique enabling presurgical evaluation of patients with mesiobasal epilepsy monitoring with subdural electrodes. Neurosurg Focus.
temporal lobe seizures. Appl Neurophysiol. 1985;48(1–6):408–417. 2008;25:E21.
37. Barnett GH, Burgess RC, Awad IA, et al. Epidural peg electrodes 56. Seeck M, Pegna A, Ortigue S, et al. Speech arrest with stimulation
for the presurgical evaluation of intractable epilepsy. Neurosurgery. may not reliably predict language deficit after epilepsy surgery.
1990;27:113–115. Neurology. 2006;66:592–594.
38. Cordova JE, Rowe RE, Furman MD, et al. A method for imaging of 57. Wyler AR, Walker G, Somes G. The morbidity of long-term seizure
intracranial EEG electrodes using magnetic resonance imaging. monitoring using subdural strip electrodes. J Neurosurg. 1991;74:
Comput Biomed Res. 1994;27:337–341. 734–737.
39. Winkler PA, Vollmar C, Krishnan KG, et al. Usefulness of 3-D recon- 58. Lee WS, Lee JK, Lee SA, et al. Complications and results of sub-
structed images of the human cerebral cortex for localization of sub- dural grid electrode implantation in epilepsy surgery. Surg Neurol.
dural electrodes in epilepsy surgery. Epilepsy Res. 2000;41:169–178. 2000;54:346–351.
712 Part V ■ Complementary and Special Techniques

59. Onal C, Otsubo H, Araki T, et al. Complications of invasive sub- 78. Dümpelmann M, Elger CE. Visual and automatic investigation of
dural grid monitoring in children with epilepsy. J Neurosurg. 2003; epileptiform spikes in intracranial EEG recordings. Epilepsia.
98:1017–1026. 1999;40:275–285.
60. Musleh W, Yassari R, Hecox K, et al. Low incidence of subdural 79. Palmini A, Gambardella A, Andermann F, et al. Intrinsic epilepto-
grid-related complications in prolonged pediatric EEG monitor- genicity of human dysplastic cortex as suggested by corticography
ing. Pediatr Neurosurg. 2006;42:284–287. and surgical results. Ann Neurol. 1995;37:476–487.
61. Jobst BC, Williamson PD, Coughlin CT, et al. An unusual compli- 80. Gambardella A, Palmini A, Andermann F, et al. Usefulness of focal
cation of intracranial electrodes. Epilepsia. 2000;41:898–902. rhythmic discharges on scalp EEG of patients with focal cortical dys-
62. Hamer HM, Morris HH, Mascha EJ, et al. Complications of invasive plasia and intractable epilepsy. Electroencephalogr Clin Neurophysiol.
video-EEG monitoring with subdural grid electrodes. Neurology. 1996;98:243–249.
2002;58:97–103. 81. Hirabayashi S, Binnie CD, Janota I, et al. Surgical treatment of
63. Wiggins GC, Elisevich K, Smith BJ. Morbidity and infection in epilepsy due to cortical dysplasia: clinical and EEG findings.
combined subdural grid and strip electrode investigation for J Neurol Neurosurg Psychiatr. 1993;56:765–770.
intractable epilepsy. Epilepsy Res. 1999;37:73–80. 82. Turkdogan D, Duchowny M, Resnick T, et al. Subdural EEG pat-
64. Araki T, Otsubo H, Makino Y, et al. Efficacy if dexamethasone on terns in children with Taylor-type cortical dysplasia: comparison
cerebral swelling and seizures during subdural grid EEG recording with nondysplastic lesions. Clin Neurophysiol. 2005;22:37–42.
in children. Epilepsia. 2006;47:176–180. 83. Guerreiro MM, Quesney LF, Salanova V, et al. Continuous elec-
65. Sahjpaul RL, Mahon J, Wiebe S. Dexamethasone for morbidity trocorticogram epileptiform discharges due to brain gliosis.
after subdural electrode insertion—a randomized controlled trial. J Clin Neurophysiol. 2003;20:239–242.
Can J Neurol Sci. 2003;30:340–348. 84. Zhang Y, van Drongelen W, Kohrman M, et al. Three-
66. Sperling MR, O’Connor MJ. Comparison of depth and subdural elec- dimensional brain current source reconstruction from intra-
trodes in recording temporal lobe seizures. Neurology. 1989;39:1497. cranial ECoG recordings. Neuroimage. 2008;42:683–695.
67. Spencer SS, Spencer DD, Williamson PD, et al. Combined depth 85. Sperli F, Spinelli L, Seeck M, et al. EEG source imaging in pedi-
and subdural electrode investigation in uncontrolled epilepsy. atric epilepsy surgery: a new perspective in presurgical workup.
Neurology. 1990;40:74–79. Epilepsia. 2006;47:981–990.
68. Brekelmans GJF, Van Emde Boas W, Velis DN, et al. Comparison of 86. Jung WY, Pacia SV, Devinsky O. Neocortical temporal lobe
combined versus subdural or intracerebral electrodes alone in epilepsy: intracranial EEG features and surgical outcome. J Clin
presurgical focus localization. Epilepsia. 1998;39:1290–1301. Neurophysiol. 1999;16:419–425.
69. Eisenschenk S, Gilmore RL, Cibula JE, et al. Lateralization of tem- 87. Wennberg R, Arruda F, Quesney LF, et al. Preeminence of
poral lobe foci: depth versus subdural electrodes. Clin Neurophysiol. extrahippocampal structures in the generation of mesial tempo-
2001;112:836–844. ral seizures: evidence from human depth electrode recordings.
70. Wieser HG, Elger CE, Stodieck SR. The “foramen ovale electrode”: Epilepsia. 2002;43:716–726.
a new recording method for the preoperative evaluation of 88. Lee SA, Spencer DD, Spencer SS. Intracranial EEG seizure-onset
patients suffering from mesio-basal temporal lobe epilepsy. patterns in neocortical epilepsy. Epilepsia. 2000;41:297–307.
Electroencephalogr Clin Neurophysiol. 1985;61:314–322. 89. Wetjen N, Marsh WR, Myer FB, et al. Intracranial electroen-
71. Fernandez Torre JL, Alarcon G, Binnie CD, et al. Comparison of cephalography seizure onset patterns and surgical outcomes in
sphenoidal, foramen ovale and anterior temporal placements for non-lesional extratemporal epilepsy. J Neurosurg. 2009;110:
detecting interictal epileptiform discharges in presurgical assess- 1147–1152.
ment for temporal lobe epilepsy. Clin Neurophysiol. 1999;110: 90. Kutsy RL, Farrell DF, Ojemann GA. Ictal patterns of neocortical
895–904. seizures monitored with intracranial electrodes: correlation with
72. Marks DA, Katz A, Booke J, et al. Comparison and correlation of surgical outcome. Epilepsia. 1999;40:257–266.
surface and sphenoidal electrodes with simultaneous intracranial 91. McGonigal A, Bartolomei F, Régis J, et al. Stereoelectro -
recording: an interictal study. Electroencephalogr Clin Neurophysiol. encephalography in presurgical assessment of MRI-negative
1992;82:23–29. epilepsy. Brain. 2007;130:3169–3183.
73. Otsubo H, Ochi A, Imai K, et al. High-frequency oscillations of 92. Spencer SS, Guimaraes P, Katz A, et al. Morphological patterns of
ictal muscle activity and epileptogenic discharges on intracranial seizures recorded intracranially. Epilepsia. 1992;33:537–545.
EEG in a temporal lobe epilepsy patient. Clin Neurophysiol. 2008; 93. Fisher R, Weber W, Lesser R, et al. High-frequency EEG activity
119:862–868. at the start of seizures. J Clin Neurophysiol. 1992;83:229–235.
74. Hufnagel A, Dümpelmann M, Zentner J, et al. Clinical relevance of 94. Schiller Y, Cascino GD, Busacker NE, et al. Characterization and
quantified intracranial interictal spike activity in presurgical eval- comparison of local onset and remote propagated electrographic
uation of epilepsy. Epilepsia. 2000;41:467–478. seizures recorded with intracranial electrodes. Epilepsia. 1998;
75. Asano E, Muzik O, Shah A, et al. Quantitative interictal subdural 39:380–388.
EEG analyses in children with neocortical epilepsy. Epilepsia. 2003; 95. Blumenfeld H, Rivera M, McNally KA, et al. Ictal neocortical slow-
44:425–434. ing in temporal lobe epilepsy. Neurology. 2004;63:1015–1021.
76. Engel J. Outcome with respect to epileptic seizures. In: Engel J Jr, 96. Spencer SS, Kim J, Spencer DD. Ictal spikes: a marker of specific
ed. Surgical Treatment of the Epilepsies. New York, NY: Raven Press; hippocampal cell loss. Electroencephalogr Clin Neurophysiol.
1987:553–571. 1992;83:104–111.
77. Bautista RE, Cobbs MA, Spencer DD, et al. Prediction of surgical 97. Townsend JB, Engel J Jr. Clinicopathological correlation of
outcome by interictal epileptiform abnormalities during intracra- low voltage fast and high amplitude spike and wave mesial
nial EEG monitoring in patients with extrahippocampal seizures. temporal stereoencephalographic ictal onsets. Epilepsia. 1991;
Epilepsia. 1999;40:880–890. 32(suppl 3):21.
Chapter 33 ■ Intracranial Monitoring: Depth, Subdural, and Foramen Ovale Electrodes 713

98. Schuh LA, Henry TR, Ross DA, et al. Ictal spiking patterns 116. Yamazaki M, Chan D, Tovar-Spinoza Z, et al. Interictal epilepto-
recorded from temporal depth electrodes predict good outcome genic fast oscillations on neonatal and infantile EEGs in
after anterior temporal lobectomy. Epilepsia. 2000;41:316–319. hemimegalencephaly. Epilepsy Res. 2008 [Epub ahead of print].
99. Faught E, Kuzniecky RI, Hurst DC. Ictal EEG wave forms from 117. Wu JY, Koh S, Sankar R, et al. Paroxysmal fast activity: an interic-
epidural electrodes predictive of seizure control after temporal tal scalp EEG marker of epileptogenesis in children. Epilepsy Res.
lobectomy. Electroencephalogr Clin Neurophysiol. 1992;83:229–235. 2008;82:99–106.
100. Adam C, Saint-Hilaire JM, Richer F. Temporal and spatial charac- 118. Lesser RP, Lüders H, Klem G, et al. Extraoperative cortical func-
teristics of intracerebral seizure propagation: predictive value in tional localization in patients with epilepsy. J Clin Neurophysiol.
surgery for temporal lobe epilepsy. Epilepsia. 1994;35:1065–1072. 1987;4:27–53.
101. Lieb JP, Engel J Jr, Babb TL. Interhemispheric propagation time 119. Duchowny M, Jayakar P. Functional cortical mapping in chil-
of human hippocampal seizures. I. Relationship to surgical out- dren. In: Devinsky O, Beric A, Dogali M, eds. Electrical and
come. Epilepsia. 1986;27:286–293. Magnetic Stimulation of the Brain and Spinal Cord. New York, NY:
102. Zaveri HP, Pincus SM, Goncharova II, et al. Localization-related Raven Press, Ltd; 1993:149–154.
epilepsy exhibits significant connectivity away from the seizure- 120. Jayakar P, Alvarez LA, Duchowny MS, et al. A safe and effective
onset area. Neuroreport. 2009;20:891–895. paradigm to functionally map the cortex in childhood. J Clin
103. Brekelmans GJF, Velis DN, van Veelen CWM, et al. Intracranial Neurophysiol. 1992;9:288–293.
EEG seizure off-set termination patterns: relation to outcome of 121. Allison T, McCarthy G, Luby M, et al. Localization of functional
epilepsy surgery in temporal lobe epilepsy. Epilepsia. 1998;39: regions of human mesial cortex by somatosensory evoked poten-
259–266. tial recording and by cortical stimulation. Electroencephalogr Clin
104. Spencer SS, Spencer DD. Implication of seizure termination loca- Neurophysiol. 1996;100:126–140.
tion in temporal lobe epilepsy. Epilepsia. 1996;37:455–458. 122. Neuloh G, Schramm J. Motor evoked potential monitoring for
105. Verma A, Lewis D, Van Landingham KE, et al. Lateralized seizure the surgery of brain tumours and vascular malformations. Adv
termination: relationship to outcome following anterior tempo- Tech Stand Neurosurg. 2004;29:171–228.
ral lobectomy. Epilepsy Res. 2001;47:9–15. 123. Carne RP, O’Brien TJ, Kilpatrick CJ, et al. MRI-negative PET-
106. Fisher RS, Webber WR, Lesser RP, et al. High-frequency EEG activ- positive temporal lobe epilepsy: a distinct surgically remediable
ity at the start of seizures. J Clin Neurophysiol. 1992;9:441–448. syndrome. Brain. 2004;127:2276–2285.
107. Engel J Jr, Bragin A, Staba R, et al. High-frequency oscillations: 124. Kurian M, Spinelli L, Delavelle J, et al. Multimodality imaging for
what is normal and what is not? Epilepsia. 2009;50:598–604. focus localization in pediatric pharmacoresistant epilepsy.
108. Bragin A, Wilson CL, Staba RJ, et al. Interictal high-frequency Epileptic Disord. 2007;9:20–31.
oscillations (80–500 Hz) in the human epileptic brain: entorhinal 125. Michel CM, Grave de Peralta R, Lantz G, et al. Spatiotemporal
cortex. Ann Neurol. 2002;52(4):407–415. EEG analysis and distributed source estimation in presurgical
109. Staba RJ, Wilson CL, Bragin A, et al. High-frequency oscillations epilepsy evaluation. J Clin Neurophysiol. 1999;16:239–266.
recorded in human medial temporal lobe during sleep. Ann 126. Lantz G, Grave de Peralta R, Spinelli L, et al. Epileptic source
Neurol. 2004;56:108–115. localization with high density EEG: how many electrodes are
110. Worrell GA, Parish L, Cranstoun SD, et al. High-frequency oscil- needed? Clin Neurophysiol. 2003;114:63–69.
lations and seizure generation in neocortical epilepsy. Brain. 127. Regard M, Cook ND, Wieser HG, et al. The dynamics of cerebral
2004;127(pt 7):1496–1506. dominance during unilateral limbic seizures. Brain. 1994;117:
111. Urrestarazu E, Chander R, Dubeau F, et al. Interictal high- 91–104.
frequency oscillations (100–500 Hz) in the intracerebral EEG of 128. Seeck M, Mainwaring N, Cosgrove R, et al. Neurophysiologic cor-
epileptic patients. Brain. 2007;130:2354–2366. relates of implicit face memory in intracranial visual evoked
112. Worrell GA, Gardner AB, Stead SM, et al. High-frequency oscil- potentials. Neurology. 1997;49:1312–1316.
lations in human temporal lobe: simultaneous microwire and 129. Grunwald T, Lehnertz K, Heinze HJ, et al. Verbal novelty detec-
clinical macroelectrode recordings. Brain. 2008;131:928–937. tion within the human hippocampus proper. Proc Natl Acad Sci
113. Jacobs J, Levan P, Chander R, et al. Interictal high-frequency U S A. 1998;95:3193–3197.
oscillations (80–500 Hz) are an indicator of seizure onset areas 130. Axmacher N, Elger CE, Fell J. Ripples in the medial temporal lobe
independent of spikes in the human epileptic brain. Epilepsia. are relevant for human memory consolidation. Brain. 2008;131:
2008;49(11):1893–1907 1806–1817.
114. Jirsch JD, Urrestarazu E, LeVan P, et al. High-frequency oscilla- 131. Vannucci M, Pezer N, Helmstaedter C, et al. Hippocampal
tions during human focal seizures. Brain. 2006;129:1593–1608. response to visual objects is related to visual memory functioning.
115. Khosravani H, Mehrotra N, Rigby M, et al. Spatial localization Neuroreport. 2008;19:965–968.
and time-dependant changes of electrographic high frequency 132. Sinai A, Bowers CW, Crainiceanu CM, et al. Eletrocorticographic
oscillations in human temporal lobe epilepsy. Epilepsia. 2008 high gamma activity versus electrical cortical stimulation map-
[Epub ahead of print]. ping of naming. Brain. 2005;128:1556–1570.
CHAPTER

Electrocorticography
MARC R. NUWER 34
E
lectrocorticography (ECoG) is intraoperative electroen- place chronically in the subdural space for days or weeks to
cephalography (EEG) recorded directly from the exposed catch epileptic seizures in an epilepsy monitoring unit, grid
cerebral cortex. The technique is used for localizing sizes up to 8 8 are available.
epileptic discharges and regions of impaired cortex prior to When used in surgery, ECoG grids and strips tend to curl up
resection. Together with direct cortical stimulation, the tech- or pull slightly away from the cortex especially in sulci. This
niques are used to identify motor, language, and sensory cortex results in poor electrical connection to some contacts. To pre-
during tumor resection. The goal of ECoG is to separate regions vent corners pulling up, the surgeon should place saline-wetted
suitable for resection from those that should be left intact. surgical Cottonoid over each strip or grid to weigh it down.
ECoG was the first application of intraoperative neurophys- These strips and grids can be moved around to record from var-
iology, a field that has since evolved to include many other tech- ious regions of exposed cortex, or even under the dura or skull
niques (1). Even as far back as the 1940s, ECoG has been used edges, to record from as many regions as needed.
to localize epileptogenic tissue prior in the neurosurgical treat- Montages are either bipolar among the adjacent contacts or
ment of partial epilepsy (2–7). The equipment for ECoG has referential to a distant site. A clip electrode on muscle or a disc
evolved since then, but the clinical goal remains relatively scalp electrode can serve as a distant neutral reference. Some
unchanged. users prefer the mastoid site. Because the amplitude of ECoG
The French group (8–10) developed a method called stere- is several-fold higher in amplitude than EEG picked up by
oencephalography (SEEG) to record from deep structures and electrodes on the muscle or scalp, referential recordings are
buried cortex that are not readily accessible by subdural or cor- often used without excessive channel contamination by an
tical methods of recording. In this way, ECoG can be obtained active reference.
simultaneously from superficial and deep brain structures. A A low-frequency filter of 0.5 Hz and a high-frequency filter
review (11) of the SEEG technique in relation to other methods of 70 Hz ensure adequate recording of epileptiform discharges
of ECoG notes that SEEG exploration is well tolerated by the and background activity. The amplitude of EEG recorded
majority of patients and overall complication rates of SEEG are directly from the cortex is several-fold higher than EEG
reported as being of the order of 5% (12,13). This is similar to recorded from the scalp. This allows for a relatively low sensitiv-
complication rates of subdural strips (14) and is somewhat less ity of 30 to 50 µV/mm to be used for ECoG recordings. A col-
than those of subdural grids (15). Although acute depth elec- lection of 20 to 40 cortical electrodes is recommended, which
trodes appear to be “more invasive” than subdural recordings, often are composed of signals from more than one grid or strip.
the complication rate is not higher. Acute multicontact depth electrodes are used by some cen-
ters. This has been reported as very helpful for an acute explo-
ELECTRODES AND METHODS ration of deep limbic and neocortical structures during
temporal lobectomy (16,17).
OF RECORDING DIRECTLY FROM
CEREBRAL CORTEX
DIRECT CORTICAL STIMULATION
ECoG recording from the exposed cerebral cortex most often TECHNIQUE
uses strip and grid electrodes. These electrodes are arrays of
3-mm metal discs embedded in a flexible silicon rubber matrix. Direct cortical stimulation is used to localize functional cortex.
Each disc is 1 cm apart measured center to center. Discs are This technique requires special instrumentation and precau-
stainless steel or platinum alloy. Fine flexible wires connect to tions. Generally equipment specifically designed for this pur-
each disc. These wires course through the silicon rubber matrix pose is used. Contact with the cortical surface usually is made
and are gathered together to exit at one end of the strip or grid. using a bipolar stimulation wand or probe held against the cor-
Together they form a ribbon cable or braided wire connector tex. Alternately, the stimulation may be delivered through the
cable leading to a multipin connector. A matched multipin con- grid or strip electrodes; the latter stimulation technique is used
nector with grounded heavier wiring transmits to the standard more often for the subacute subdural studies on epilepsy mon-
EEG machine electrode junction box. Strips and grids come in itoring units. The wand stimulation technique is more conven-
many different sizes. Sizes used more often in surgery are 1 8 ient during surgery where the cortex is directly exposed. The
strips and 4 5 grids. For patients in whom these are left in typical stimulating wand or probe is 30 cm long, with two

715
716 Part V ■ Complementary and Special Techniques

round metal ball tips separated by a few millimeters. Such a Recordings above and below the sylvian fissure would show
wand can move among cortical locations quickly. This allows more fast activity above and less below. ECoG background
for testing many different locations in a short time. activity is more sharply contoured than typically seen on scalp
Stimuli are bipolar and biphasic. Bipolar means that electric EEG recordings; this sharply contoured EEG is similar to the
current runs between the two adjacent active contacts. Biphasic scalp EEG over a skull breach. This sharply contoured back-
means that the current switches polarity halfway through each ground needs to be distinguished from epileptic activity during
stimulus pulse. Biphasic stimulation is preferred in the ECoG interpretation. Some ECoG epileptiform spikes may not be
setting so as not to leave a net electrical polarization at either apparent on concurrent scalp-recorded EEG (20). Areas of
metal–cortex contacts. Polarization could allow metal ions to impaired cortex may show relatively decreased fast activity,
move onto the brain, which is undesirable especially for iron increased slow activity, or epileptic spikes. A knowledgeable and
ions that would serve as a source of future cortical irritation. experienced physician neurophysiologist identifies these
Polarization alters the electrical sensitivity of the underlying changes and thereby identifies regions with impaired function.
cortex to subsequent stimulus pulses. Bipolar biphasic stimula- ECoG is used for certain patients during epilepsy surgery.
tion equipment is available, but most stimulation equipment Most often this is when the epileptogenic zone was inadequately
for routine intraoperative monitoring is not biphasic. The latter localized by other methods. ECoG is used to further define the
routine stimulation equipment is unsuitable for direct cortical resection limits. For patients with very frequent seizures during
stimulation studies. either scalp or invasive EEG monitoring, intraoperative ictal
Typical direct cortical stimuli are 100 to 300 sec (0.1 to ECoG may be useful to demarcate the electrographic ictal onset
0.3 msec) pulses delivered in trains of 50 pulses per second for zone more precisely. An interictal ECoG is much more com-
several seconds. A 5- to 7-second stimulation train is used for monly obtained. Interictal spike discharges usually arise from
language testing, whereas shorter trains are used for motor test- the epileptogenic zone but also extend to cortex beyond it. Those
ing. Stimulus pulses are delivered using currents up to 14 mil- regions are defined and discussed with the surgeon to tailor the
liamperes (mA) for the usual wand or probe designs. This is resection to the needs of the individual patient.
sufficient to disrupt function at the cortex underlying the wand’s Sometimes ECoG is performed after resection. Epileptiform
location. Often one starts with a lower intensity, for example, at discharges observed at the resection margins may just represent
4 to 6 mA, increasing gradually to higher intensities if no desired “injury potentials” or “injury spikes,” not the location of addi-
or adverse effects are seen at the lower intensities at each site. tional epileptic tissue. Epileptic spikes distant from the resection
Either constant current or constant voltage stimulation may be margin are more concerning, and frontal lobe epilepsy patients
used. Duration longer than 8 seconds should be avoided. with epileptiform discharges three or more gyri away from the
During direct cortical stimulation, ECoG is recorded from resection site indicated likelihood of seizure recurrence.
nearby sites. This is carried out to check whether epileptic-like To avoid anesthetic effects, the anesthesia is lightened or
discharges occur with the stimulations. When epileptic-like dis- minimized during the ECoG. Many anesthetics can alter the
charges last longer than the stimulation, they are referred to as presence of epileptiform discharges sought in ECoG. The
afterdischarges. Afterdischarges appear as rhythmic repetitive effects of the anesthetic on the ECoG are clearly less important
high-voltage spikes or polyspike–wave complexes lasting sec- than the anesthetic’s clearance time. Propofol clears adequately
onds or occasionally minutes. Such prolonged afterdischarges to allow good ECoG in most patients. Nitrous oxide and nar-
are usually subclinical or associated with subtle clinical behav- cotics may be used during the ECoG to avoid pain or con-
ioral manifestations (18). When afterdischarges occur, stimula- sciousness during the procedure, although these may affect the
tion may have spread to other cortical regions. Therefore those epileptiform discharges seen on the recording.
stimulation trials should not be used to localize cortical func- Direct cortical stimulation testing is interpreted by the pres-
tions because they include activation of areas away from the ence of movement or sensation or the disruption of language.
wand’s tip. Afterdischarges may herald epileptic seizures occur- Neuromuscular blockade needs to be avoided. For motor test-
ring with further stimulation at that site or intensity. This may ing, anesthesia cannot be deep. Observations of responses can
prompt moving the stimulating wand to a different site or to be made visually, or EMG electrodes can be placed in various
reducing the stimulus intensity. muscles to record movements under the surgical drapes.
Cortical stimulation can provoke epileptic seizures. Cold Stimulations then can identify various face, arms, and leg
Ringer’s lactate solution can treat seizures provoked by cortical regions. Finding stimulation effects at some cortical areas
stimulation. Such irrigation rapidly stops partial seizures (19). shows that the anesthetic depth is adequate and the technique
is working properly. Considerable individual variability exists
INTERPRETATION OF ECOG AND DIRECT in location of eloquent cortex regions for language, motor, and
CORTICAL STIMULATION sensory function (21–23). Anatomic locations need to be deter-
mined for each eloquent function for each individual patient.
The goal of ECoG is to determine locations of epileptic or oth- Stimulation mapping allows for best resection of pathologic tis-
erwise impaired cortex. The recording usually shows EEG fea- sue while preserving function (24,25). Functional neuroimag-
tures similar to those seen with scalp EEG. Frontocentral fast ing navigation methods are usually combined with stimulation
activity, low-amplitude generalized slowing, and frontopolar mapping; such a dual approach is the current best guide for the
triangular slow waves are seen during many types of anesthesia. limits of a resection (26,27).
Chapter 34 ■ Electrocorticography 717

The patient is awake during surgery for sensory and language GENERAL ANESTHESIA AND CORTICAL
testing. The conscious patient is extubated while restrained on EEG ACTIVITY
the table (28). The primary sensory cortex can be mapped using
such stimulation in the awake patient. For some awake epilepsy Anesthetic drugs tend to either excite or depress the EEG and
patients, stimulation near a seizure focus will cause the patient’s follow a pattern summarized by Winters (38). The activating or
typical aura. exciting anesthetic agents are propofol and etomidate. They
For language testing, a neuropsychologist or other trained lan- inhibit brainstem arousal mechanisms through GABAA receptor
guage specialist administers language tasks for different aspects of mechanisms (39,40). The resulting EEG patterns resemble slow-
auditory and visual language processing and short-term memory. wave sleep including vertex sharp waves and spindles. Despite
The clinical neurophysiologist delivers the stimulations and early reports, propofol does not produce EEG epileptiform
observes for afterdischarges while the surgeon holds the wand and activity (41–43). Propofol is a popular agent for ECoG recording
the language specialist administers oral and visual testing. Most and before direct cortical stimulation. Because of its rapid
adult patients tolerate this awake craniotomy language testing metabolism, propofol can induce unconsciousness during por-
adequately. Stimulation at a series of cortical locations can iden- tions of the craniotomy. Yet it is eliminated rapidly enough to
tify functional language areas. Regions without noticeable disrup- allow intraoperative awakening, a major advantage for ECoG
tion at 12 to 14 mA are considered candidates for resection. (44,45). Etomidate is another activating or exciting anesthetic
The asleep–awake–asleep technique requires cooperative agent. Like propofol, it enhances epileptic activity at low doses
patients (28). Initial anesthesia is often propofol or sodium (0.1 mg/kg). It may produce seizures in patients with epilepsy
thiopental with endotracheal intubation. Local anesthetic is infil- (46). The paradoxical excitatory effect of low-dose propofol may
trated around the operative site margins. The patient is awakened result from a membrane-level interaction between the GABAA
and extubated once the craniotomy exposure is achieved. current and an intrinsic membrane slow potassium current (M-
Consistent intraoperative language performance baseline is current) according to a proposal by McCarthy et al. (47). In that
needed before stimulation testing. Without baseline consistency, theory, a potential network mechanism underlying the genera-
any observed language disruption might be due to inconsistent tion of propofol-induced paradoxical excitation consists of an
performance rather than specific anatomic disruption due to interneuron antisynchrony, one that also produces an EEG beta
stimulation. After mapping is completed, the patient is reintu- rhythm at low drug concentrations.
bated and remains under general anesthesia for the remainder of The commonly used volatile inhalation anesthetic agents are
the procedure. sedating anesthetics. They include halothane, desflurane, isoflu-
Language mapping includes several types of tasks. Visual rane, and sevoflurane. They cause immobility through multiple
object naming involves the naming of objects presented as line molecular targets predominantly in the spinal cord, including
drawings from the Boston Naming Test (29). Drawings are pre- GABAA receptors, glycine receptors, glutamate receptors, and
sented that the patient could name quickly in preoperative test- two-pore-domain potassium channels (48). These agents cause
ing (21). Naming is a useful screening task for language during rhythmic frontocentral alpha activity. This rhythmic fast activ-
cortical stimulation mapping because naming deficits are fre- ity can be abolished by nitrous oxide (49). Hyperventilation
quently part of aphasia syndromes. Speech arrest or anomia may with high concentrations of these agents, except for desflurane,
occur upon stimulating a cortical region important for lan- can produce epileptiform spikes or even electrographic non-
guage, although not all cortical language sites produce naming convulsive seizures. These agents can reduce (50) or increase
deficits (30,31). Different cortical sites may reveal stimulation (51) epileptiform discharges at standard anesthetic levels. They
deficits in sentence reading (32). Another easily accomplished have little effect at low levels (52).
stimulation language test is word generation in which patients Barbiturates are similar to inhalational agents in producing
generate lists of words that begin with a certain letter or are of a activation and fast activity at low doses and a depressant effect
certain category (e.g., animals). In auditory responsive naming, leading to burst suppression at higher doses. These bursts may
the patient names objects, that is, the answer for “tall pink bird” have very sharp components that could be misinterpreted as
would be “flamingo.” Auditory responsive naming and word epileptiform. At low dose, the short-acting barbiturate metho-
generation tasks activate frontal language areas (33,34). Visual hexital (0.5 mg/kg) does activate epileptic spike activity during
responsive naming differs from auditory responsive naming ECoG, and so it has been used deliberately to identify seizure
because the question is written on a flash card. foci (46,53–55). Methohexital also can produce seizures.
Stimulation mapping effects are well localized. Moving the Benzodiazepines produce frontal beta activity with a decrease in
stimulating electrodes 0.5 to 2 cm, even along the same gyrus, alpha activity at low doses. Some patients show epileptiform
yields different functional effects (32,35). A site with consistent spike activity in that frontal beta activity. Slowing of higher fre-
language function can be 2 cm from a site on the same gyrus quencies through inhibitory mechanisms may produce the bar-
without language disruption (21). Language function often is biturate beta activity (56). Higher benzodiazepine doses
localized to small 1 to 2 cm 2 regions (32,36). Postoperative lan- produce generalized slowing. As an anticonvulsant, benzodi-
guage deficits occur when resection margins are within 1.5 to azepines are avoided if ECoG is planned for identification of
2.0 cm of language function sites. No permanent language seizure foci. Nitrous oxide produces a frontocentral fast activity
deficits were reported if the surgical margins remain 2.0 cm when used by itself. When used with inhalational agents, it pro-
from language sites (32,36,37). duces variable results. Nitrous oxide is proconvulsant when
718 Part V ■ Complementary and Special Techniques

combined with some agents. It may increase or suppress epilep- spike locations are to be resected. Interictal epileptic distur-
tic spike activity during ECoG depending on which other bances recorded at ECoG in patients with temporal lobe
agents are used (57,58). Droperidol and dexmedetomidine are epilepsy (TLE) often involve (Fig. 34.1) adjacent regions
other adjunctive agents sometimes used during ECoG proce- (18,64–77). Some reports suggest that interictal epileptic dis-
dures. These seem compatible with ECoG. Narcotic drugs can turbance recorded intraoperatively should guide surgical resec-
increase epileptiform spiking, including spiking or seizures tion (18,68,78–83)). Others have emphasized that standardized
from sites outside the epileptogenic area (59–61). Remifentanil lobectomy could be performed without ECoG control for
may increase epileptiform discharges at higher doses (62) but patients with typical mesial temporal epilepsy (84–86). ECoG
not with lower-dose continuous infusions (63). recording after a temporal resection is controversial. Some
Overall, light anesthesia enhances the ability to find epilep- authors believe that the persistence of spike activity after surgi-
tiform spiking during ECoG. Propofol is an example of an cal excision is of little or no prognostic value (2,87–89). Other
anesthetic agent that can be reduced sufficiently quickly to investigators advocate that persistent spike activity after surgi-
achieve a light anesthesia, and which does not suppress intra- cal removal predicts an unsatisfactory outcome (16,79,90,91).
operative epileptiform abnormalities. Methohexital or other Recent reports (92) showed that intraoperative ECoG activ-
drugs sometimes are used to enhance epileptiform abnormali- ities can be analyzed with respect to more complex spike pat-
ties during ECoG. terns, particularly in TLEs; namely, in addition to focal spiking
with or without propagation, focal slowing in the theta or delta
APPLICATION IN EPILEPSY SURGERY range and ictaform ECoG patterns were found. Furthermore,
high-frequency oscillations (HFOs; (93)) in the range of 250 to
Spikes occur not only in the epileptogenic zone but also in 600 Hz (fast ripples) have been identified in the ECoGs
other cortical areas. Even the most active spiking area may not recorded from the hippocampus and parahippocampal areas of
coincide with the location of the lesion (6). The goal in ECoG patients with TLE, which may be used to identify brain epilep-
is to localize the region to be resected. Not all ECoG epileptic togenic areas. Rates and durations of HFOs were found (94) to

Figure 34.1 ECoG recording showing spontaneous multifocal interictal spiking involving the second temporal gyrus ante-
riorly (1–2), electrode 6 in the first temporal gyrus, and the mesial temporal lobe structures synchronously (A1–2, P1–2).
In addition, widespread interictal epileptiform bursts involving the mesial temporal lobe structures as well as the tempo-
ral neocortex were recorded. (Reproduced from Stefan, H., Quesney, L.F., Abou-Khalil, B., and Olivier, A. 1991.
Electrocorticography in temporal lobe epilepsy surgery. Acta Neurol Scand. 83:65–72. )
Chapter 34 ■ Electrocorticography 719

Figure 34.2 Preexisting electrocorticogram in a patient with a right frontocentral cortical dysplastic lesion on MRI. The
tracing shows electrographic seizure activity recorded multifocally. A 11>2 to 2 Hz rhythmic activity was seen at electrode 3;
rhythmic polyspike activity at 10 Hz recorded maximally at electrodes 1 and 7; rhythmic 2 to 3 Hz spike-and-wave activ-
ity recorded at electrode 10. (Reproduced from Palmini, A., Gambardella, A., Andermann, F., et al. 1995. Intrinsic epilep-
togenicity of human dysplastic cortex as suggested by corticography and surgical results. Ann Neurol. 37:476–487.)

be significantly higher in the seizure onset zone than outside, (Fig. 34.2) corresponding to MRI evidence for cortical dysplasia.
and occurred to a large extent independently of spikes. A good surgical outcome in these patients is proportional to the
In extratemporal surgery, interictal epileptiform spiking is completeness of the lesion’s surgical removal (103) (Fig. 34.3).
often widespread (95–100) even when the patient has a circum- Afterdischarge locations during direct cortical stimulation
scribed lesion. In neocortical epilepsy, ECoG mapping of the do not correlate with impaired function or pathology and often
interictal epileptogenic area is useful to tailor the surgical resec- arise at normal cortex. Stimulation at certain anatomical sites,
tion (101). The distribution and abundance of interictal spiking for example, hippocampus, is more predisposed to elicit after-
recorded at ECoG is a prognostic indicator of postsurgical discharges (104–106). Electrical stimulation thresholds are
seizure control outcome (100). Both preexcision epileptic often increased in damaged areas (107,108), so that impaired
abnormality recorded from less than two gyri away and absence regions tend not to have afterdischarges (16,107).
of postexcision epileptic activity distant to the resection border
strongly predicted a favorable outcome. Residual spiking lim- SUMMARY
ited to the resection border did not significantly correlate with
outcome. The presence of a circumscribed frontal lobe lesion ECoG is a method to detect cortical regions with substantial
was also significantly correlated with favorable outcome. In epileptiform interictal discharges during surgery. Direct corti-
frontal lobe epilepsy, postexcision residual spiking indicates a cal stimulation studies provide a means to identify language,
poor outcome in terms of seizure control (102). motor, and sensory regions during a craniotomy. These tech-
ECoG is useful for localizing cortical dysplasia. The region of niques can help to determine the limits for a resection. They are
the lesion shows rhythmic electrographic seizure discharges used during epilepsy and tumor surgery. They can be adversely
720 Part V ■ Complementary and Special Techniques

Figure 34.3 A: Baseline preexcision ECoG prior to methohexital injection showing active limbic epileptiform activity and
a single independent temporal neocortical spike. B: Preexcision ECoG 1 minute after injection of 40 mg methohexital show-
ing activation of epileptiform activity recorded independently from limbic and temporal neocortical structures. A slight
burst-suppression pattern developing over neocortex.
Chapter 34 ■ Electrocorticography 721

Figure 34.3 (continued) C: Postexcision ECoG showing activation of temporal neocortical epileptiform activity and burst-
suppression pattern. (Reproduced from Wennberg, R., Quesney, F., Olivier, A., and Dubeau, F. 1997b. Induction of burst-
suppression and activation of epileptiform activity after methohexital and selective amygdalo-hippocampectomy.
Electroencephalogr Clin Neurophysiol. 102:443–451.)

affected or limited by the anesthetic agents used. Amount of 10. Chauvel P, Buser P, Badier J, et al. La “zone épileptogène” chez
spiking from diffuse or remote cortical regions ECoG can pre- l’homme: représentation des événements intercritiques par
dict postoperative seizure control. cartes spatio-temporelles. Rev Neurol (Paris). 143:443–550.
11. McGonigal A, Bartolomei F, Régis J, et al. Stereoelectro -
encephalography in presurgical assessment of MRI-negative
REFERENCES epilepsy. Brain. 2007; 130(pt 12):3169–3183. [Epub September 12,
2007].
1. Nuwer, MR. Intraoperative Monitoring of Neural Function: 12. Guenot M, Isnard J, Ryvlin P, et al. Neurophysiological monitoring
Handbook of Clinical Neurophysiology. Amsterdam: Elsevier; 2008. for epilepsy surgery: the Talairach SEEG method. Stereo -
2. Ajmone Marsan C, Baldwin M. Electrocorticography. In: Baldwin electroencephalography Indications, results, complications and
M, Bailey P, eds. Temporal Lobe Epilepsy Springfield, IL: Charles C therapeutic applications in a series of 100 consecutive cases.
Thomas; 1958: 368–395. Stereotact Funct Neurosurg. 77:29–32.
3. Jasper, HH. Electrocorticogram in man. Electroencephalogr Clin 13. Cossu M, Cardinale F, Castana L, et al. Stereoelectro -
Neurophysiol. 1949;2(suppl):16–29. encephalography in the presurgical evaluation of focal epilepsy: a
4. Marshall, C, Walker, AE. Electrocorticography. Bull John Hopkins retrospective analysis of 215 procedures. Neurosurgery. 2005;57:
Hosp. 1949;85:344–359. 706–718.
5. Meyer, HR., Knott R., Hayne RA et al and Sweeney, D.B. The sur- 14. Alarcon G, Valentin A, Watt C, et al. Is it worth pursuing surgery
gery of epilepsy. Limitations of the concept of the corticoelectro- for epilepsy in patients with normal neuroimaging? J Neurol
graphic “spike” as an index of the epileptogenic focus. Neurosurg Psychiatry. 2006;77:474–480.
J Neurosurg. 1950;7:337–346. 15. Hamer HM, Morris HH, Mascha MS, et al. Complications of inva-
6. Penfield W, Jasper HH Epilepsy and the Functional Anatomy of the sive video-EEG monitoring with subdural grid electrodes.
Human Brain. Boston, MA: Little, Brown; 1954. Neurology. 2002;58:97–103.
7. Walker AE, Marshall C, Beresford EM Electrocorticographic char- 16. Gloor P. ntributions of electroencephalography and electrocor-
acteristics of the cerebrum posttraumatic epilepsy. Assoc Res Nerv ticography to the neurosurgical treatment of the epilepsies. In:
Men Dis. 1947;26:502–515. Purpura DP, Penry JK, Walker RD, eds. Neurosurgical
8. Bancaud J, Talairach J, Bonis A, et al. La stéréoélectroencephalo- Management of Epilepsies. New York: Raven Press; 1975: 59–105.
graphie dans l’épilepsie: informations neurophysiopathologiques 17. Wennberg R, Quesney F, Olivier A et al. Mesial temporal versus lat-
apportées par l’investigation fonctionelle stéréotaxique. Paris: eral temporal interictal epileptiform activity: comparison of
Masson & Cie; 1965. chronic and acute intracranial recordings. Electroencephalogr Clin
9. Talairach J, Bancaud J, Szickla G, et al. Approche nouvelle de la Neurophysiol. 1997;102:486–494.
chirurgie de l’épilepsie: méthodologie stéréotaxique et résultats 18. Stefan H, Quesney LF, Abou-Khalil, B., et al Electrocorticography in
thérapeutiques. Neurochirurgie. 1974;20:1–240. temporal lobe epilepsy surgery. Acta Neurol Scand. 1991;83:65–72.
722 Part V ■ Complementary and Special Techniques

19. Sartorius CJ, Berger MS. Rapid termination of intraoperative 40. Rudolph U, Antkowiak B. Molecular and neuronal substrates for
stimulation-evoked seizures with application of cold Ringer’s lac- general anaesthetics. Nat Rev Neurosci. 2004;5:709–720.
tate to the cortex. Technical note. J Neurosurg. 1998;88:349–351. 41. Ebrahim ZY, Schubert, Van Ness et al The effect of propofol on the
20. Tao JX, Ray, A., Hawes-Ebersole, S, et al Intracranial EEG sub- electroencephalogram of patients with epilepsy. Anesth Analg.
strates of scalp EEG interictal spikes. Epilepsia. 2005;46:669–676. 1994;78:275–279.
21. Ojemann GA. Individual variability in cortical localization of lan- 42. Samra SK, Sneyd JR, Ross DA, et al and Henry, T.R. Effects of
guage. J Neurosurg. 1979;50:164–169. propofol sedation on seizures and intracranially recorded epilepti-
22. Uematsu S, Lesser R, Fisher RS, et al. Motor and sensory cortex in form activity in patients with partial epilepsy. Anesthesiology.
humans: topography studied with chronic subdural stimulation. 1995;82:843–851.
Neurosurgery. 1992;31:59–71; discussion 71–72. 43. Cheng MA, Tempelhoff R, Silbergeld DL, et al. Large-dose propo-
23. Branco DM, Coelho TM, Branco BM, et al. Functional variability fol alone in adult epileptic patients: electrocorticographic results.
of the human cortical motor map: electrical stimulation findings Anesth Analg. 1996;83(1):169–174.
in perirolandic epilepsy surgery. J Clin Neurophysiol. 2003;20: 44. Skucas, AP Artru A. Anesthetic complications of awake cran-
17–25. iotomies for epilepsy surgery. Anesth Analg. 2006;102:882–887.
24. Duffau, H, Lopes M, Arthuis F, et al. Contribution of intraopera- 45. Soriano SG, Eldredge EA, Wang FK, et al. The effect of propofol on
tive electrical stimulations in surgery of low grade gliomas: a com- intraoperative Electrocorticography and cortical stimulation during
parative study between two series without (1985–96) and with awake craniotomies in children. Paediatr Anaesth. 2000;10: 29–34.
(1996–2003) functional mapping in the same institution. J Neurol 46. Rampil IJ. Electroencephalogram. In: Albin MA, ed. Textbook of
Neurosurg Psychiatry. 2005;76:845–851. Neuroanesthesia with Neurosurgical and Neuroscience Perspectives .
25. Duffau H, Capelle L, Sichez J, et al. Intra-operative direct electrical New York: McGraw-Hill; 1997: 193–220.
stimulations of the central nervous system: the Salpetriere experi- 47. McCarthy MM, Brown, EN Kopell N. Potential network mecha-
ence with 60 patients. Acta Neurochir (Wien). 1999;141: nisms mediating electroencephalographic beta rhythm changes
1157–1167. during propofol-induced paradoxical excitation. J Neurosci.
26. Duffau H. Intraoperative direct subcortical stimulation for identi- 2008;28:13488–13504.
fication of the internal capsule, combined with an image-guided 48. Grasshoff C, Rudolph U Antkowiak, B. Molecular and systemic
stereotactic system during surgery for basal ganglia lesions. Surg mechanisms of general anaesthesia: the ‘multi-site and multiple
Neurol. 2000;53:250–254. mechanisms’ concept. Curr Opin Anaesthesiol. 2005;18:386–391.
27. Kamada K, Todo T, Masutani Y, et al. Combined use of tractogra- 49. Yli-Hankala A. The effect of nitrous oxide on EEG spectral power
phy-integrated functional neuronavigation and direct fiber stimu- during halothane and isoflurane anaesthesia. Acta Anaesthesiol
lation. J Neurosurg. 2005;102:664–672. Scand. 1990;34:579–584.
28. Huncke K, Van De Wiele B, Fried I et al The asleep-awake-asleep 50. Asano E, Benedek K, Shah A, et al. Is intraoperative electrocor-
anesthetic technique for intraoperative language mapping. ticography reliable in children with intractable neocortical
Neurosurgery. 1998;42:1312–1316; discussion 1316–1317. epilepsy? Epilepsia. 2004;45(9):1091–1099.
29. Kaplan E, Goodglass, H Weintraub, S. Boston Naming Test. 51. Endo T, Sato K, Shamato H, Effects of sevoflurane on electrocor-
Philadelphia, PA: Lea and Febiger; 1976. ticography in patients with intractable temporal lobe epilepsy. J
30. Ojemann G, Mateer, C. Human language cortex: localization of Neurosurg Anesthesiol. 14:59–62.
memory, syntax, and sequential motorphoneme identification sys- 52. Fiol ME, Boening JA, Cruz-Rodriguez R, Effect of isoflurane
tems. Science. 1979;205:1401–1403. (Forane) on intraoperative electrocorticogram. Epilepsia. 1993;34:
31. Ojemann GA. Functional mapping of cortical language areas in 897–900.
adults. Intraoperative approaches. Adv Neurol. 1993;63:155–163. 53. Ajmone Marsan C, O’Connor M. Electrocorticography. In:
32. Ojemann GA. Brain organization for language from the perspec- Remond A, ed. Handbook of Electroencephalography and Clinical
tive of electrical stimulation mapping. Behav Brain Sci. 1983;2: Neurophysiology. Vol. 10C. Amsterdam: Elsevier; 1973: 3–49.
189–230. 54. Musella L, Wilder BJ, Schmidt RP. Electroencephalographic activa-
33. Bookheimer SY, Zeffiro TA, Blaxton, et al T Regional cerebral tion with intravenous methohexital in psycho-motor epilepsy.
blood flow during auditory responsive naming: evidence for cross- Neurology (Minneapolis). 1971:21:594–602.
modality neural activation. Neuroreport. 1998;9:2409–2413. 55. Wennberg R, Quesney F, Olivier A, et al Induction of burst-sup-
34. Cuenod CA, Bookheimer SY, Hertz-Pannier L, et al Functional pression and activation of epileptiform activity after methohexital
MRI during word generation, using conventional equipment: a and selective amygdalo-hippocampectomy. Electroencephalogr
potential tool for language localization in the clinical environ- Clin Neurophysiol. 1997;102:443–451.
ment. Neurology. 1995;45:1821–1827. 56. Jensen O, Goel P, Kopell N, et al. On the human sensorimotor-cor-
35. Ojemann, GA, Whitaker HA. Language localization and variabil- tex beta rhythm: sources and modeling. Neuroimage. 2005;26:
ity. Brain Lang. 1978;6:239–260. 347–355.
36. Ojemann A, Cortical organization of language. J Neurosci. 57. Artru AA, Lettich E, Colley PS, et al. Nitrous oxide: suppression of
1991;11:2281–2287. focal epileptiform activity during inhalation, and spreading of
37. Haglund MM, Berger MS, Shamseldin M, et al Cortical localiza- seizure activity following withdrawal. J Neurosurg Anesthesiol.
tion of temporal lobe language sites in patients with gliomas. 1990;2(3):189–193.
Neurosurgery. 1994;34:567–576; discussion 576. 58. Hosain S, Nagarajan L, Fraser R, et al. Effects of nitrous oxide on
38. Winters WD. Effects of drugs on the electrical activity of the brain: electrocorticography during epilepsy surgery. Electroencephalogr
anesthetics. Annu Rev Pharmacol Toxicol. 1976;16:413–426. Clin Neurophysiol. 1996;102:340–342.
39. Nelson LE, Guo TZ, Lu, J., JCBNP et al The sedative component of 59. Tempelhoff R, Modica PA, Bernardo KL, et al.I Fentanylinduced
anesthesia is mediated by GABA(A) receptors in an endogenous electrocorticographic seizures in patients with complex partial
sleep pathway. Nat Neurosci. 2002;5:979–984. epilepsy. J Neurosurg. 1992;77:201–208.
Chapter 34 ■ Electrocorticography 723

60. Cascino GD, So EL, Sharbrough FW, et al. Alfentanil induced 80. Fiol ME, Gates JR, Torres F, et al. The prognostic value of residual
epileptiform activity in patients with partial epilepsy. J Clin spikes in the postexcision electrocorticogram after temporal lobec-
Neurophysiol. 1993;10(4):520–525. tomy. Neurology. 1991;41:512–516.
61. Manninen PH, Burke SJ, Wennberg R, et al. Intraoperative local- 81. Jennum P, Dhuna A, Davies K, et al. Outcome of resective surgery
ization of an epileptogenic focus with alfentanil and fentanyl. for intractable partial epilepsy guided by subdural electrode
Anesth Analg. 1999;88:1101–1106. arrays. Acta Neurol Scand. 1993;87:434–437.
62. Wass CT, Grady RE, Fessler AJ, et al. The effects of remifentanil on 82. Ojeman GA. Basic mechanisms implicated in surgical treatments
epileptiform discharges during intraoperative electrocorticogra- of epilepsy. In: Lockard JS, Ward AA Jr, eds. Window to Brain
phy in patients undergoing epilepsy surgery. Epilepsia. Mechanisms. New York: Raven Press; 1980:261–277.
2001;42:1340–1344. 83. Primrose DC, Ojeman GA. Outcome of resective surgery for tem-
63. Herrick IA, Craen RA, Blume WT, et al. Sedative doses of remifen- poral lobe epilepsy. In: Lüders H, ed. Epilepsy Surgery. New York:
tanil have minimal effect on ECoG spike activity during awake Raven Press; 1992:601–611.
epilepsy surgery. J Neurosurg Anesthesiol. 2002;14(1):55–58. 84. Engel J Jr, ed. Approaches to the localization of the epileptogenic
64. Cendes F, Dubeau F, Olivier A, et al. Increased neocortical spiking lesion. Surgical Treatment of the Epilepsies. New York: Raven Press;
and surgical outcome after selective amygdalo- hippocampectomy. 1987:75–100.
Epilepsy Res. 1993;16:195–206. 85. Spencer DD, Inserni J. Temporal lobectomy. In: Lüders HO, ed.
65. Devinsky O, Canevini MP, Sato S, et al. Quantitative electrocor- Epilepsy Surgery. New York: Raven Press; 1992:533–545
ticography in patients undergoing temporal lobectomy. 86. Doyle WK, Spencer DD. Anterior temporal resections. In: Engel
J Epilepsy. 1992;5:178–185. J Jr, Pedley T, eds. Epilepsy: A Comprehensive Textbook. Vol. 2.
66. Engel J Jr, Driver MV, Falconer MA. Electrophysiological correlates Philadelphia, PA: Lippincott–Raven; 1997:1807–1817
of pathology and surgical results in temporal lobe epilepsy. Brain. 87. Chatrian GE, Quesney LF. Intraoperative electrocorticography. In:
1975;98:129–156. Engel J Jr, Pedley TA, eds. Epilepsy: A Comprehensive Textbook. New
67. Gastaut H, Naquet R, Vigouroux R, et al. Étude électrographique York: Raven Press; 1997
chez l’homme et chez l’animal des décharges épileptiques dites 88. Gibbs FA, Amador L, Rich C. Electro -encephalographic findings
“psychomotrices.” Rev Neurol. 1953;88:310–354. and therapeutic results in surgical treatment of psychomotor
68. Graf M, Niedermeyer E, Schiemann J, et al. Electrocorticography: epilepsy. In: Baldwin M, Bailey P, eds. Temporal Lobe Epilepsy.
information derived from intraoperative recordings during seizure Springfield, IL: Charles C Thomas; 1958:358–367
surgery. Clin Electroencephalogr. 1984;15:83–91. 89. Walker AE, Lichtenstein S, Marshall C. A critical analysis of elec-
69. Kajtor F, Hullay J, Farago L, et al. Electrical activity of the hip- trocorticography in temporal lobe epilepsy. Arch Neurol.
pocampus of patients with temporal lobe epilepsy. AMA Arch 1960;2:172–182.
Neurol Psychiatry. 1958;80:25–38. 90. Jasper HH, Arfel-Capdevielle G, Rasmussen T. Evaluation of EEG
70. Niedermeyer, E. 1982. Electrocorticography. In Electro- and cortical electrographic studies for prognosis of seizures fol-
encephalography, E. Niedermeyer and F. Lopes da Silva, Eds., pp. lowing surgical excision of epileptogenic lesions. Epilepsia.
537–541. Munich: Urban & Schwarzenberg. 1961;2:130–137.
71. Niedermeyer E. Electrocorticography. In: Niedermeyer E, Lopes da 91. McBride MC, Binnie CD, Janota I, et al. Predictive value of intra-
Silva F, eds. Electroencephalography, Basic Principles, Clinical operative electrocorticograms. Electroencephalogr. Clin Neurol.
Applications and Related Fields, 2nd ed. Baltimore, MD: Urban and 1991;30:526–532.
Schwarzenberg; 1987:613–617 92. Stefan H, Hopfengärtner R, Kreiselmeyer G, et al. Interictal triple
72. Quesney LF, Niedermeyer E. Electrocorticography. In: ECoG characteristics of temporal lobe epilepsies: an intraoperative
Niedermeyer E, Lopes da Silva F, eds. Electroencephalography: Basic ECoG analysis correlated with surgical outcome. Clin
Principles, Clinical Applications, and Related Fields, 3rd ed. Neurophysiol. 2008;119:642–652.
Baltimore, MD: Williams & Wilkins; 1993:695–699. 93. Engel J Jr, Bragin A, Staba R, et al. High-frequency oscillations:
73. Rasmussen TB. Surgical treatment of complex partial seizures: what is normal and what is not? Epilepsia. 2009;50(4):598-604.
results, lessons and problems. Epilepsia. 1983;24(suppl 1):S65–S76. 94. Jacobs J, LeVan P, Chander R, et al. Interictal high-frequency oscil-
74. Tuunainen A, Nousiainen U, Mervaala E, et al. Postoperative EEG lations (80–500 Hz) are an indicator of seizure onset areas inde-
and electrocorticography: relation to clinical outcome in patients pendent of spikes in the human epileptic brain. Epilepsia.
with temporal lobe surgery. Epilepsia. 1994;35:1165–1173. 2008;49:1893–1907.
75. Walker AE. Temporal lobectomy. J Neurosurg. 1967;26: 642–649. 95. Quesney LF, Constain M, Fish DR, et al. Frontal lobe epilepsy—
76. Walker AE. Surgery for epilepsy. In: Magnus O, Lorentz de Haas field of recent emphasis. Am J EEG Technol. 1990;30:177–193.
AM, eds. Handbook of Clinical Neurology, Vol. 15: The Epilepsies. 96. Quesney LF, Constain M, Rasmussen T, et al. How large are frontal
Amsterdam: North Holland; 1974:739–757 lobe epileptogenic zones? EEG, ECoG and SEEG evidence. In:
77. Quesney LF, Abou-Khalil B, Cole A, et al. Pre-operative extracra- Chauvel P, Delgado-Escueta AV, Halgren E, et al., eds. Advances in
nial and intracerebral EEG investigation in patients with temporal Neurology, Vol. 57. New York: Raven Press; 1992
lobe epilepsy: trends, results and review of pathophysiologic 97. Salanova V, Andermann F, Olivier A, et al. Occipital lobe epilepsy:
mechanisms. Acta Neurol Scand. 1988;78:52–61. electroclinical manifestations, electrocorticography, cortical stimu-
78. Ajmone Marsan C. Depth electrography and electrocorticography. lation and outcome in 42 patients treated between 1930 and 1991.
In: Aminoff MJ, ed. Electrodiagnosis in Clinical Neurology. New Brain. 1992;115:1655–1680.
York: Churchill Livingstone; 1980:167–196. 98. Salanova V, Andermann F, Rasmussen T, et al. Parietal lobe
79. Bengzon ARA, Rasmussen T, Gloor P, et al. Prognostic factors in epilepsy. Clinical manifestations and outcome in 82 patients
surgical treatment of temporal lobe epilepsy. Neurology treated surgically between 1929 and 1988. Brain. 1995;118:
(Minneapolis). 1968;18:717–731. 607–627.
724 Part V ■ Complementary and Special Techniques

99. Salanova V, Andermann F, Rasmussen T, et al. Tumoural parietal 104. Jasper HH. Electrocorticography. In: Penfield W, Jasper HH, eds.
lobe epilepsy. Clinical manifestations and outcome in 34 patients Epilepsy and the Functional Anatomy of the Human Brain. Boston,
treated between 1934 and 1988. Brain. 1995;118:1289–1304. MA: Little, Brown; 1954:692–738
100. Wennberg R, Quesney LF, Villemure JG. Epileptiform and non- 105. Walker AE. Electrocorticography in epilepsy. A surgeon’s
epileptiform paroxysmal activity from isolated cortex after func- appraisal. In: Fischgold H, ed. Electroencephalographie et
tional hemispherectomy. Electroencephalogr Clin Neurophysiol. Electrocorticographie de l’epilepsie, Vol. 2. 1949:30–37.
1997;102:437–442 106. Ajmone Marsan C. Focal electrical stimulation. In: Purpura DP,
101. Quesney LF, Wennberg R, Olivier A, et al., eds. EcoG findings in Penry JK, Tower DB, et al., eds. Experimental Models of Epilepsy:
extratemporal epilepsy: the MNI experience. Electrocorticography: A Manual for the Laboratory Worker. New York: Raven Press;
Current Trends and Future Perspectives, (EEG Suppl. 48). 1972:147–172
Amsterdam: Elsevier Science; 1998. 107. Bernier GP, Richer F, Giard N. Electrical stimulation of the
102. Wennberg R, Quesney LF, Olivier A, et al. Electrocorticography human brain in epilepsy. Epilepsia. 1990;31:513–520.
and outcome in frontal lobe epilepsy. Electroencephalogr Clin 108. Cherlow DG, Dymond AM, Crandall PH, et al. Evoked response
Neurophysiol. 1998;106:357–368. and after-discharge thresholds to electrical stimulation in tempo-
103. Palmini A, Gambardella A, Andermann F, et al. Intrinsic epilep- ral lobe epileptics. Arch Neurol. 1977;34:527–531.
togenicity of human dysplastic cortex as suggested by corticogra-
phy and surgical results. Ann Neurol. 1995;37:476–487.
CHAPTER
Principles and Techniques for Long-Term
EEG Recording (EMU, ICU, Ambulatory)
JEAN GOTMAN, MARC NUWER, AND RONALD G. EMERSON
35
INTRODUCTION even erroneous: patients are often not aware of their seizures or
of parts of their symptoms and relatives are frequently poor
Most standard EEG recordings last 20 to 30 minutes and some historians and poor observers. In some cases, it is important to
are extended to 1 or 2 hours. It is also possible to record the EEG know the EEG manifestations of the seizures in order to make a
over long periods of time, from a few hours to days or weeks. correct diagnosis. For example, behavioral arrest can occur with
This is the domain of “long-term monitoring” (LTM) and this primary generalized epilepsy and generalized spike and wave
chapter will discuss when such long recordings should be per- and with a temporal lobe seizure. One can refine the indications
formed and in particular the special recording and analysis tech- for LTM as follows (3):
niques that are available to facilitate the collection and review of
these data in the most efficient way, while providing the patient • The diagnosis of epilepsy needs to be made or rejected, as the
with minimum discomfort. Most such LTM is performed in the events cannot be unambiguously classified from historical
context of episodic disorders and the characterization of the information. Some patients have epileptic and nonepileptic
behavioral component of these disorders is an integral part of seizures.
the diagnosis process. Video recording of the patient, synchro- • The diagnosis of epilepsy has been made but the type of
nized with the EEG, is therefore an integral part of LTM. seizure or syndrome is not clear.
Long-term EEG and video recording were born in the 1970s • Epilepsy is medically intractable and surgery is considered.
and became a mature field in the early 1980s (1). The technol- This requires a full characterization of the electroclinical
ogy has considerably improved since then but fundamental manifestations of seizures, at first with scalp EEG, and possi-
aspects have not really changed, with the possible exception of bly subsequently with intracranial electrodes in case the scalp
LTM in the intensive care unit (ICU), which has only developed EEG does not provide sufficient information.
in the last few years. It is interesting to note that the combined • Seizures need to be quantified to assess response to medical
recording of EEG and behavior has a long history in the context treatment, as may be the case in brief but frequent seizures.
of epilepsy (2); at that time the “long-term” part was missing and In addition, LTM allows recording throughout the
only the “EEG video” (albeit on film) was possible for a few min- sleep–wake cycle. Some patients have predominantly or exclu-
utes. The recording of behavior only became practically feasible sively seizures during sleep, others predominantly on awaken-
over extended periods in the 1970s with time-lapse video ing. Interictal activity in focal epilepsy is activated by non-REM
recorders, recording one or just a few frames per second. Major sleep and in some patients is only present during that stage.
leaps occurred with the availability of VCRs and more recently
with the digital recording of video signals. With respect to the Equipment and Personnel
ability to record EEG signals over the long term, the storage The basic components of a LTM system are the electrodes and
capacity of computers has steadily increased, such that it electrode wires, the amplifier, the EEG transmission system, the
was possible to record discontinuous samples in the 1970s and camera, and the recording computer. We will examine the char-
1980s and then it became possible to record continuously for acteristics of these different components.
24 hours in the 1990s, at the standard sampling rate of 200 Hz.
Today the storage requirements for the long-term EEG signal Electrodes
have become trivial, except in the context of large numbers of Electrode attachment is a particularly important aspect of
channels (100 or more) and very high sampling rates (2000 Hz). obtaining good-quality EEG during LTM and while the patient
experiences the many movements caused by the activities (e.g.,
LONG-TERM MONITORING FOR EPILEPSY eating) present during a recording that last several days and
while the patient has seizures, which can include violent move-
Indications ments. Electrode problems are the most common cause of poor
The International League Against Epilepsy has published guide- quality of a recording. The scalp electrodes must be placed in
lines for the use of LTM in epilepsy (3). In order to make a solid such a way that they provide a good contact for a long period,
diagnosis of epilepsy and of the type of epilepsy, a description typically at least 24 hours, sometimes longer if technicians are
of the behavioral and cognitive manifestations of the seizures is not available every day or during weekends. Collodion or other
necessary. This is most often obtained from the patients and durable electrode-scalp adhesive is therefore recommended. It
their relatives. In many cases, the description is incomplete or is important to minimize artifacts caused by the movement of
725
726 Part V ■ Complementary and Special Techniques

electrode wires, which are particularly common during or to a connector in the patient room must be light and comfort-
seizures. This can be achieved by wrapping a bandage or plac- able, as well as very robust to withstand the stress of large
ing elastic netting over the patient’s head, thus preventing the seizures. The functions of the cable are to transmit the EEG sig-
wires from moving. This also limits the patient’s access to elec- nal and to provide power to the amplifier. Recent technology
trodes (e.g., to prevent dislodging electrodes during scratch- allows wireless transmission of the EEG, which gives much free-
ing). All wires can be bundled together until the input of the dom of movement and added comfort to the patient. The
amplifier: the bundle has more rigidity than individual wires drawback of wireless transmission is that the equipment carried
and will also limit movement artifact. by the patient must have a battery to power the amplifier as well
If a patient undergoing LTM requires an MRI, the electrodes as the wireless transmission. This significantly increases the
must be removed and then replaced after the study. This can weight of the patient-worn system and runs the risk of battery
require a considerable time. Electrodes that can be kept during depletion, thus interrupting monitoring. Wireless transmission
MRI studies have been developed recently. They include MR- must therefore be considered for relatively short periods, with
compatible material for the electrodes and the wires and an cable transmission used most of the time.
MR-compatible connector system. Such electrodes include
gold, silver–silver chloride, subdermal silver wire, and conduc- Video Recording
tive plastic electrodes (4,5). Some of these electrodes are also Digital video cameras are most commonly used. The usual
CT-compatible, greatly facilitating EEG monitoring of patients color camera can be combined with an infrared illuminator and
in the ICU. an automatic switching to black and white for low light condi-
It has been shown in multiple studies that electrodes record- tions, thus providing an excellent quality image even in dark-
ing from regions lower than the standard 10/20 electrodes are ness. Allowing darkness increases patient comfort at night. The
important in the characterization of temporal and inferior digital video signal is recorded continuously on the computer,
frontal discharges. A minimum configuration for LTM should synchronized with the EEG. The EEG amplifier and the video
therefore include 25 electrodes: the 19 electrodes of the 10/20 camera have independent clocks for sampling the EEG and for
system plus three inferior electrodes on each side (e.g., F9, T9, timing video frames. This makes the synchronization of the two
P9, F10, T10, P10 of the 10/10 system). Recordings are most signals a complex operation and it is important to verify that
often referential and it is therefore particularly important to this synchronization is maintained.
have a reference of good quality. Display with any other refer-
ence can be subsequently calculated. The EKG must also be Recording Computer, Display, and Network
recorded since cardiac changes can be an important component The EEG and the video signals are transmitted to the computer,
of seizures or seizure-like events. It may be useful in some situ- which may be at a distance from the patient room, either by
ations to use additional electrodes in order to obtain better spa- dedicated cable or more and more commonly through a stan-
tial sampling of a region. We will not discuss here the various dard computer network (either the hospital network or a net-
types of intracerebral electrodes as this is addressed elsewhere work dedicated to the LTM and EEG laboratories). Given the
in this volume. storage capacity of modern computer disks, there is no problem
in storing several days of EEG recording, even sampled at high
EEG Amplification and Transmission rates, and several days of video recording. Large monitors allow
It is most convenient if the amplifier is sufficiently small to be the easy display of 15 or 20 seconds of EEG on the screen.
carried by the patient, usually in a pouch placed on the chest or Characteristics of the EEG display, gain, time scale, and filters,
belt. After the amplifier, the transmitted signal is subject to can easily be changed. Since EEGs are usually recorded referen-
much less artifact because it has been amplified. The amplifier tially, alternate montages can be viewed (bipolar, changed refer-
should have minimal characteristics of a standard EEG ampli- ence, average reference, etc.). Network facilities can be used to
fier. These are typically a low filter at 0.3 Hz and high filter at 60 view the EEG from any computer connected to the computer
or 70 Hz and sampling at 200 or 250 Hz. There is recent evi- on which the EEG is stored, either during the recording or after
dence that high frequencies in the intracerebral EEG (6–8; see it is completed. This includes viewing from locations outside
also Chapter 37) and low frequencies in newborns (9) may also the hospital, such as at home, provided security barriers are
be of interest. Optimum characteristics may include low fre- respected.
quencies (low filter at 0.1 Hz) and high frequencies (high filter Archiving the data is a challenge because it is not practical to
at 600 Hz and sampling at 2000 Hz). Digital filters can be used archive all recorded data on permanent storage media such as
when reviewing the EEG to eliminate unwanted low frequen- DVDs, particularly the video, because of size. It is likely that
cies if they have been recorded. Recording with a high sampling complete archiving will become possible in a few years, but at
rate has the disadvantage of increasing memory requirements present, the data that are to be archived must be selected by a
and slowing down display. Twelve-bit digitizing is common and person familiar with EEG interpretation to make sure all
most likely to be sufficient, although 16-bit digitizing is also important information is retained.
available and covers more than sufficiently the dynamic range
of the intracerebral EEG. Personnel and EEG Interpretation
Since the patient is monitored for extended periods, the cable Trained observers need to be near the patients to provide close
system connecting the patient-worn amplifier to the computer clinical observation and interaction with the patient during
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 727

seizures, as well for ensuring patient safety. To assist with con- in spiking (17,18) have shown that spiking does not increase
stantly observing patients on monitoring, patients can press an following drug withdrawal per se. Spiking does increase follow-
alarm button to report any auras or seizures they notice. ing seizures. When drugs are reduced, seizures occur and spik-
Observation for seizures may be enhanced with automatic ing increases as a result of increased seizures. It may therefore
seizure detection systems (see below). It is also useful if a rela- appear that drug reduction results in increased spiking, but this
tive or companion can stay in the room with the patient and is an indirect effect mediated by seizures. In the absence of
alert staff when seizures occur. seizures, spiking does not change. These observations were con-
Experienced EEG technologists should be used so as to firmed in a recent study using more sophisticated analysis tools
obtain the best information from an LTM session. They are crit- (19). This study also demonstrated that spiking could even
ical to secure electrodes well and in the proper locations. They decrease as a result of drug withdrawal in the absence of
review the preliminary recorded data and seek to achieve suit- seizures.
able recording quality. In some centers they make a first review
of the 24-hour session, marking the seizures and any section of Considerations for Children
interest. Only the marked sections are reviewed subsequently by or Infants and for Adults
the clinical neurophysiology physician. In other centers, the full LTM can be performed at all ages including during the new-
recording is reviewed by the physician, but this is often consid- born period (20). In newborns and young infants, a subset of
ered too time consuming. Automatic seizure and spike detection the standard 10/20 electrodes can be used although one has to
may be helpful in selecting the sections to review (see below). In be careful because seizures can be very focal. Collodion should
addition to selecting what data need review, it is necessary to not be used in young children, as their skin is very delicate. As
decide what subset of the original recording needs to be perma- seizures and interictal activity are more frequent in young
nently archived. (The full original recording is usually too large patients than in adults, the duration of monitoring is usually
for storage, particularly the video.) The selection of what needs shorter, often limited to 1 or 2 days. In children, a full syn-
to be retained results from review and interpretation of the dromic definition often requires that different seizure patterns
record. It typically consists of all seizures and some interictal be recorded, as well as the interictal activity (3). In adults but
spikes, as well as some sample of the EEG background. more particularly for children, it is important to have a parent
in the room during monitoring to help identify the events being
Duration of Monitoring and Drug Withdrawal investigated.
The duration of monitoring depends on the purpose. In gen-
eral, it is best to try to be generous with the duration of moni- Automatic Seizure and Spike Detection
toring because it is frequent that unexpected events are The size of computer disks is such that it is simple to perform a
discovered. Patients may report one seizure type but not be continuous 32-channel, 7-day recording on a standard com-
aware that they also have brief seizures of which they are not puter, storing EEG and video. A computer-based recording
aware or nocturnal seizures nobody has witnessed. Some allows a flexible review procedure, with random access to any
patients have more than one type of seizure, so a sample of only part of the recording and the availability of numerous methods
two to four seizures may be too few to use for major clinical of data manipulation and analysis. In most 24-hour recordings,
decisions such as surgery. The clinical description reported by 95% to 99% of the recording offers little useful information in
the patient or family is often incomplete or erroneous and it the evaluation of an epileptic disorder. Automatic detection
may require the recording of several seizures to fully document methods, despite their imperfections, can facilitate the marking
a seizure disorder that is different from what was expected. In and thus the review of valuable information.
the context of presurgical evaluation, it has been estimated that
five seizures with the same region of onset and no contradictory Automatic Seizure Detection
information provide a reasonable sample to decide that the During LTM, patients are usually asked to press a button when
onset is indeed in that region (10). they feel a seizure coming. If patients could always feel their
Antiepileptic medications are frequently reduced or discon- seizure coming, there would be no need for automatic seizure
tinued during LTM in order to allow seizures to occur more fre- detection. It is frequent, however, that patients are not aware of
quently. In the context of presurgical evaluation, the question of their seizures, and observers are not always present or able to
whether seizures occurring after medication withdrawal are recognize a seizure. Furthermore, some seizures have very min-
similar to habitual seizures has been raised. Symptomatology imal clinical signs—and sometimes no visible signs at all.
and the EEG pattern at onset are similar after medication with- Finding all seizures may require the review of days and days of
drawal (11–14). There is no evidence that seizures spread more EEG, a process that is long and tedious—and not always effec-
rapidly from one hemisphere to the other after drug with- tive in as much as short seizures can be missed, particularly if
drawal. Seizures are more often secondarily generalized after there are many channels as in intracerebral recordings.
drug reduction (15,16), so monitoring unit staff should be pre- Automatic seizure detection can be helpful in this context, since
pared to respond in case of repeated generalized seizures. it provides a means for marking the EEG sections that are likely
It is also often said that the reduction in antiepileptic med- to include seizure patterns. For this purpose, the detection can
ication results in increased spiking activity. Careful studies of take place at any time during the seizure (not necessarily early)
the timing of drug withdrawal, seizure occurrence, and changes because the EEG will be reviewed a posteriori.
728 Part V ■ Complementary and Special Techniques

Seizure detection could also be used in real time, to warn the also on the respective position of the electrodes and the gener-
patient or observer that a seizure has just started. In this case, ator. In some cases, no seizure discharge may be visible because
the detection must occur early to allow the patient to take pro- the electrodes are too far from the generator. Similarly, the
tective measures (if he or she is still able to do so), to assist an seizure discharge may appear only as a mild modification to the
observer in providing protective measures in a timely fashion, ongoing EEG. It is therefore important to distinguish the
and to facilitate the observer’s attempts to question the patient seizure itself from its manifestations (i.e., from the vantage
so as to improve our understanding of clinical signs (e.g., can point of the recording electrodes). Since seizure detection is
the patient follow commands, talk, remember?). Such an early performed from analyzing the EEG, it is only the seizures that
seizure detection system can be used during monitoring in the have a clear manifestation in the EEG that can be detected.
clinic (21,22), but could also be incorporated in an implantable Figures 35.1 and 35.2 show examples of seizures recorded from
device that would provide a warning (e.g., auditory signal) to the scalp, and Figure 35.3 shows a seizure recorded from intrac-
the patient during normal everyday activities. erebral electrodes. In Figures 35.1 and 35.3, the specific
Seizure prediction is an area of active investigation but, moment of onset is not easily identified. In Figure 35.2, the
unlike seizure detection and the warning provided by the onset onset is clear, consisting of a few seconds of rhythmic discharge
of the seizure, it is an area that has proven difficult and without followed by irregular slow waves.
clear results. Despite initially promising results, it is not yet There is no formal definition of what a seizure consist of in
clear if it is possible to predict seizures (23; see also Chapter 30). the EEG. Qualitative definitions usually include the terms
“paroxysmal, rhythmic, evolving”— but an unambiguous defini-
Detection Methods tion has not been formulated. Automatic detection methods have
Our current understanding is that seizures consist of paroxys- therefore relied on trying to encode these qualitative characteris-
mal rhythmic activity generated by a large number of synchro- tics into algorithms, and improving the algorithms by trial and
nized neurons. This activity usually evolves in its spatial error in an effort to increase detection sensitivity and decrease
distribution, frequency, and amplitude. The patterns that are the false detection rate. The first general-purpose seizure detec-
actually recorded depend not only on the discharge itself, but tion method validated on a large data sample, characterized each

Cz-C3
---

C3-T3
---

T3-Zy1
---

Zy1-Zy2
---

Zy2-T4
---

T4-C4
---

C4-Cz
---

Fp1-F7
---

F7-T3
---

T3-T5
---

T5-O1
---

Fp2-F8
---

F8-T4 ---

T4-T6 ---

T6-O2 ---

150 uV
1s

Figure 35.1 Scalp-recorded right temporal seizure occurring in the context of muscle and eye movement artifacts. This
early part of the seizure consists of a discharge in the theta range of frequencies, commonly seen in temporal lobe seizures.
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 729

Fp1-F7
---

F7-T3
---

T3-T5
---

T5-O1
---

Fp2-F8
---

F8-T4
---

T4-T6
---

T6-O2
---

Fp1-F3
---

F3-C3
---

C3-P3
---

P3-O1 ---

Fp2-F4 ---

F4-C4 ---

C4-P4 ---

P4-O2 ---

100 uV
1s

Figure 35.2 Scalp-recorded seizure with a generalized and abrupt onset. A few seconds of rhythmic theta frequency
activity are followed by irregular slow waves in the delta range.

2-second epoch by measures of frequency, rhythmicity, and An extension to the above concept of context is to incorpo-
amplitude change, and compared these measures to a constantly rate a particular patient’s seizure characteristics in the context.
updated background (24,25). A set of empirical rules combined In this case, the method is aimed at detecting seizures in that
the findings in multiple epochs and multiple channels to reach a particular patient, and only seizures that look like the sample
decision on detection. The concept of a “constantly updated seizures (that have been incorporated in the context) are
background” plays an important role in the analysis of long-term detected. The method can then be labeled “patient-specific”—
data, since EEG is nonstationary, that is, the background content or more correctly, “seizure-specific”—since a patient can have
changes considerably due to factors such as states of vigilance. It several types of seizure. Better detection performance can thus
is also important that the results not be dependent on the selec- be obtained if it is acceptable to detect only a specific seizure
tion of a particular background epoch, a dependence that would pattern in a given patient (21,22,31,32).
introduce arbitrariness into the process. More recent methods Seizure patterns are quite different in the newborn EEG than
that have followed a similar empirical approach include those of in older children and adults: the discharges are often much
Grewal and Gotman (26), and Saab and Gotman (27). slower and sometimes very focal (limited to one electrode). For
Artificial neural networks (ANNs) are an interesting these reasons, methods specific to the newborn have been
approach to seizure detection (28–30). The ANN is presented developed, with the particular aim of detecting the very slow
with a large number of training examples that comprise both discharges often present during newborn seizures (33–35).
seizure and nonseizure data, and gradually learns to differenti-
ate seizures from nonseizure EEG. A set of features characteriz- Validation of Detection Methods
ing the EEG needs to be specified in advance (e.g., amplitude, A method must be tested on a data set independent from that
rhythmicity, dominant frequency, etc.), but it is not necessary to on which it was developed. The testing data must be represen-
explicitly describe seizure patterns. The ANN automatically tative of the context in which the method will be used: they
determines which combinations of features are characteristic of should include a large variety of seizures, and a much larger
seizures by adapting its internal structure to reflect the training amount of nonseizure data than of seizure data (in order to
data. The performance of ANN methods depends on an appro- ensure that all types of interictal data are well represented—
priate set of features and on exposure to a sufficiently large quiet and active wakefulness, different sleep stages, different
training set containing a variety of seizure patterns. types of artifacts common during LTM). The best way to avoid
730 Part V ■ Complementary and Special Techniques

RH2-RH3
---

RH4-RH5
---

RH5-RH6
---

RH6-RH7
---

RH7-RH8
---

RH8-RH9
---

RH9-RH10
---

RE1-RE2 ---

RE2-RE3 ---

RE3-RE4 ---

RE5-RE6 ---

RE6-RE7 ---

500 uV

5s

Figure 35.3 Seizure recorded from intracerebral electrodes placed on the temporal lobe. The RH contacts are along an
orthogonal multicontact depth electrode having its deepest contacts (lower numbers) in the hippocampus and most superfi-
cial contacts in the second temporal convolution. The RE electrodes are epidural electrodes placed along the first temporal
convolution. The seizure onset consists of mixed frequency activity, followed by a more rhythmic discharge. Note compressed
time scale. For the purpose of clarity, this EEG illustrates only a fraction of the electrodes implanted in this patient’s brain.

selection bias is probably to select all data between two given the intracerebral EEG often includes many paroxysmal bursts of
dates. An important issue in validating a method is the defini- uncertain significance, which can also contribute to false detec-
tion of a seizure itself. Whereas there is no ambiguity with a tions; and seizure patterns are often more prominent in intrac-
clear seizure lasting 30 seconds or 1 minute, significant ques- erebral EEG, thus resulting in better sensitivity (26).
tions arise for short events: What is the minimum duration of a
seizure? Should a seizure having clinical manifestations, but no Automatic Spike Detection
change in the EEG, be counted as an event to detect? What if the Many epileptic patients present with interictal abnormalities in
EEG changes are very minor, such as a brief flattening or a short their EEG. These consist of abnormalities of the background,
burst of slow waves? When presenting validation results, these which will not be discussed here, and paroxysmal events such as
issues should be addressed. spikes, polyspikes, sharp waves, and bursts of spike and wave.
Methods that were evaluated on extensive data sets (hun- These paroxysmal events occur most often unpredictably and
dreds of hours, tens of patients) report detection sensitivities sometimes rarely. They play an important role in defining some
from 75% to 90%. Sensitivity is often lower in newborns than epileptic syndromes and they contribute to localizing the
in adults, as some newborn seizure patterns are very subtle. The source of seizure discharges. Although used primarily for these
false detection rate, the most common measure of failure, is in purposes, it is important to remember that they, by themselves,
the range of 0.3 to 1 per hour in recent studies. Few studies disturb normal brain function (36,37). Automatic spike detec-
report results with intracerebral electrodes, but these recordings tion methods started to be developed when computer analysis
have important distinguishing characteristics: scalp artifacts of the EEG was in its infancy, in the 1960s. Despite this long his-
(electromyogram [EMG], movement, eye blinks) are absent, tory and despite the tremendous increase in the power of com-
thus reducing false detections; the dynamic range of sponta- puters over this period, automatic detection today is far from
neous fluctuations in background is much larger in intracere- perfect. There are probably two reasons for this: the first is that
bral than in scalp EEG—thus resulting in more false detections; spikes are not a well-defined pattern, as illustrated by the low
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 731

inter-rater agreement demonstrated in the study of Wilson et can determine whether a critically ill patient is having seizures,
al. (38); it is an almost impossible task to detect a pattern for including nonconvulsive seizures, that cause a decreased level of
which there is so little agreement. The second reason may be consciousness (41,42). Nonconvulsive seizure EEG patterns
that, despite its apparent simplicity, spike detection is quite a may be frequent to constant epileptic spiking, or more discrete
difficult task that the human brain performs rapidly but that cycles of ictal discharges separated by interictal attenuation and
takes into account a complex analysis of temporal and spatial slowing. Spike morphology can be the traditional single spike
features as well as context, an analysis that is not easy to repro- or sharp wave events, or more organized trains of spike wave
duce in a computer program. seen in focal or generalized epilepsy (43–52). EEG monitoring
During routine EEG examinations or short recordings, auto- of critically ill patients is indicated for patients with decreased
matic spike detection is of little use for an expert because it is level of consciousness not otherwise explained.
easy to find all the spikes visually. It is sometimes said, however, ICU EEG monitoring can separate movements from seizures.
that nonexperts overinterpret the EEG, defining as epileptiform For example, pseudoperiodic lateralized epileptiform discharges
transients that are artifacts or normal variants (39). This can (PLEDs) can be seen at the site of focal lesions, such as ischemic
result in overtreatment of patients. In such a context a reliable stroke (53,54). They may be accompanied by myoclonic jerks,
automatic detection method would be useful. Generally, auto- usually seen as temporally separate rather than synchronous
matic detection becomes valuable mostly when analyzing long with the PLED discharges. EEG recordings with EMG channels
recordings, particularly during long-term epilepsy monitoring, accompanying the myoclonic jerks can demonstrate the lack of
when hours or days of recording have to be examined. It is also temporal relationship of jerks to EEG discharges, evidence that
in this context that automatic detection is most difficult observed movements are myoclonic rather than epileptic. Some
because the recording includes multiple transients that are dif- medications, for example, tacrolimus, are associated with tics or
ficult to differentiate from epileptiform transients: eye blinks, other repetitive movements for which EEG with an EMG chan-
movement and muscle artifacts, electrode artifacts, sleep tran- nel or video-EEG ICU monitoring can help determine whether
sients (vertex waves, spindles, K complexes). movements are epileptic.
An excellent review of spike detection methods for scalp Monitoring can grade the degree of encephalopathy. Slowing
EEG has recently been published by Halford (40) and we refer in routine EEGs can be either continuous or intermittent. A
the reader to this comprehensive review. No spike detection fixed structural deficit or anoxic injury tends to produce con-
method has addressed specifically the problem of detecting tinuous slowing. A metabolic encephalopathy tends to produce
spikes in the intracerebral EEG. Although in appearance it is an waxing and waning of background, that is, intermittent slow-
easier problem than detection in the scalp EEG because of the ing. For both the continuous and intermittent slowing, the
absence of most of the artifacts, it remains a difficult problem degree of EEG slow activity corresponds in a general way with
because the issue mentioned above regarding the definition of the degree of impairment. For example, alpha coma from
spikes is worse in the intracerebral EEG. Some channels show hypoxic injury is characterized by monotonously continuous
almost continuous sharp activity and it is very difficult to iden- frontocentral alpha activity (55–61) that is nonreactive, that is,
tify isolated transients. The morphology of sharp transients is does not change when the patient is stimulated. EEG monitor-
also highly variable. In other words, the problem is even more ing can evaluate the EEG not only for reactivity to stimulation
ill-defined than for scalp spikes. and moment to moment monotony, but also for the sponta-
neous variability that occurs more normally over many minutes
LONG-TERM ICU MONITORING to hours. Presence of such variability is a favorable sign, and
absence is unfavorable. Shorter routine EEGs may miss these
ICU EEG monitoring is used for several clinical purposes. In longer duration fluctuations in EEG content because the sam-
coma EEG can search for possible presence of seizures, some of ple is too short. ICU EEG monitoring can identify and measure
which are nonconvulsive. For patients at risk for deterioration, the variability in long-term EEGs.
EEG can monitor continuously for adverse signs. For patients Burst suppression carries a grave prognosis when associated
in a deliberate burst suppression state, EEG can monitor that with anoxia or diffuse brain injury. It also is the desired state
the induced coma is sufficiently deep. when a patient is in a deliberately heavily sedated state. EEG
A primary goal of ICU EEG monitoring is identification of monitoring can monitor the extent of burst suppression. That
acute adverse events early enough to intervene to prevent or ame- can be used as a gauge of prognosis in hypoxic–ischemic burst
liorate long-term adverse clinical sequellae. Examples of acute suppression. Or, it can be a feedback measurement for adequacy
events in neurologic critical care patients include subarachnoid of therapy in deliberate burst suppression. ICU EEG monitor-
hemorrhage (SAH), intracranial bleeding, vascular thrombosis ing can measure and trend the level of burst suppression.
or embolus, convulsive or nonconvulsive seizures, cerebral
edema, or vasospasm. Some are beyond meaningful intervention. Incidence and Impact of Seizures and Status Epilepticus
Many are amenable to various medical or surgical therapies. Seizures occurred in 29% to 35% of ICU patients who had
stroke, intracerebral hemorrhage, seizures, metabolic coma,
Typical Indications for ICU EEG Monitoring brain tumors, and brain trauma as reasons for hospitalization
Monitoring can identify subtle or nonconvulsive seizures. (41,62,63). Table 35-1 gives additional rates of ICU seizures. Two
Seizures can manifest in various ways and circumstances. EEG thirds to three fourths of patients had nonconvulsive seizures or
732 Part V ■ Complementary and Special Techniques

Tabl e 3 5 . 1

The Incidence of Seizures Using ICU EEG Monitoring in a Neurologic Critical Care Illness Population

Percentage with
ICU EEG Studies N Seizures Principal Diagnosis
Jordan (62) 124 35 Neurologic critical illness
Vespa et al. (64) 91 22 Brain trauma
Vespa et al. (144) 300 21 Subarachnoid
hemorrhage/brain trauma
Vespa et al. (65) 65 28 Primary intracerebral hemorrhage
Claassen et al. (66) 204 17 Subarachnoid hemorrhage
Pandian et al. (67) 105 42 Neurologic critical illness
Young and Doig (145) 55 20 Neurologic critical illness

nonconvulsive status epilepticus. Such seizures are not readily seizures (78). The increase in glutamate concentration exceeds
diagnosed by motor manifestations. The diagnosis required EEG the concentration known to induce cell death and swelling, and
monitoring. Clinical manifestations of the seizures included persists for days. This excitotoxic secondary injury in turn
persistent stupor and coma. Many EEG patterns were typical
ictal events, whereas others had various kinds of continuous
repetitive sharp waves, polyspikes, or sharply contoured rhyth-
mic slowing. The variety of EEG presentations in critically ill
patients differs from the presentation of ictal events in an
epilepsy monitoring unit. Among these patients, 21% were
initially misdiagnosed with nonepileptic conditions. EEG mon-
itoring sometimes was not initiated at the beginning of the ICU
stay. An average of 43 hours passed before EEG monitoring was
used, the correct diagnosis made, and appropriate therapy
undertaken. Other reports are similar (63,68–70).
Changes in patient care commonly included initiating or
modifying anticonvulsants, obtaining neuroimaging, or adjust-
ing blood pressure. EEG monitoring was decisive in initiating
or prompting changes in patient care in 51% of patients and
made a significant contribution in 31%. Several studies assessed
the incidence of nonconvulsive seizures and status in ICU
patients (66,71–73). The incidence ranged from 11% (74) to
55% (66). After moderate to severe traumatic brain injury, 22%
of 91 patients had seizures, a majority (57%) of which were Figure 35.4 EEG detection of four nonconvulsive seizures. EEG mon-
nonconvulsive (Fig. 35.4) (64). The incidence of overt clinical itoring over 5 hours in a 50-year-old hypertensive man 3 days after a
seizures after an ischemia stroke was 5% to 17% (75–77), 3-cm left thalamic hemorrhagic stroke. There were no outward signs of
whereas the incidence of subtle or electrographic nonconvul- seizures on examination of this comatose patient. EEG monitoring
sive seizures is twice as high (62). After intracerebral hemor- detected three nonconvulsive seizures during this 5-hour period.
rhage the incidence of seizures was 28% (65), many of which Monitoring trends from six EEG channels are shown. Time is displayed
were nonconvulsive. Compared to studies of overt clinical along the horizontal axis, from 11 AM to 4 PM. For each channel, the
seizures, ICU EEG monitoring detects more than twice as many amount of alpha activity is displayed as vertical bars. Alpha activity is
seizures, with the increase being mainly nonconvulsive seizures. expressed as a percentage of the overall EEG activity from 1 to 30 Hz.
Some convulsive seizures also were found that had been over- The percent-alpha values range here from approximately 150% to 50% .
looked by nursing and medical staff. Seizures are suspected when an abrupt jump in percent-alpha is seen,
followed by a suppression of alpha activity. The three suspected events
Seizures in the ICU Damage the Brain are noted here with arrows. Review of the stored polygraph EEG
Seizures after brain injury or hemorrhage provoke a pathophys- showed that these were generalized epileptic EEG discharges. These
iologic response at a time when the brain is very vulnerable. repetitive nonconvulsive seizures were then treated with phenytoin.
Levels of extracellular glutamate increase after posttraumatic (Figure courtesy of UCLA EEG Lab.)
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 733

causes lipid peroxidation, membrane disruption, and increases stroke. Often the patient is sent for an MRI or CT to evaluate
in extracellular lipid breakdown product (e.g., glycerol) levels the acute change, since EEG is not specific enough to localize or
(79). Nonconvulsive seizures in critically ill patients increase confirm the cause of a change. The amount of EEG slowing
glucose metabolism and intracranial pressure. These mecha- parallels perfusion-weighted MRI lesion volume (90). EEG also
nisms most likely underlie the observation that nonconvulsive correlates well with clinical control of hypertension and
seizures after brain hemorrhage lead to increased cerebral hypotension. Six of 11 patients showed EEG improvement dur-
edema and midline shift (65). Compared with brain hemor- ing hypertension, and 12 of 18 patients showed EEG worsening
rhage patients without seizures, those with seizures had a sub- during periods of hypotension (91). An acute EEG index
stantial increase in midline shift and increased mortality. reflected the therapeutic effects of tPA (90,92). The progression
Seizures were an independent factor predicting mortality. These of adverse change to isoelectric can be regional rather than gen-
lines of evidence show that nonconvulsive seizures should not eralized, a phenomenon labeled by Schneider and Jordan (93)
be considered benign in critically ill patients, and that they as regional attenuation without delta (RAWOD).
should be detected and treated.
Vasospasm After Subarachnoid Hemorrhage
Variability, Reactivity, and Sleep Cycles During Coma SAH causes EEG slowing (94) and disrupts the posterior dom-
Certain EEG features are associated with better prognoses. In inant rhythm (95). Among 116 consecutive SAH patients with
the comatose or deeply encephalopathic patient, these favorable EEG monitoring (96), 16% showed periodic epileptiform dis-
features include reactivity, variability, and sleep cycles. charges, 6% nonconvulsive seizures, and 4% nonconvulsive sta-
Unfavorable findings are monotonous slow activity without tus epilepticus. The EEG was nonreactive to external stimuli in
reactivity or variability (80–82). Reactivity is seen over short 14%, showed no state changes in 14%, and stage II sleep tran-
times in response to verbal, visual, or tactile stimulation. sients were absent in 85%. Other EEG findings included frontal
Variability is a spontaneous change in the EEG seen over longer rhythmic delta activity (FRDA; 8%) and stimulus-induced
periods. Sleep cycles are one form of variability. EEG monitor- rhythmic, periodic, or ictal discharges (SIRPIDs; 8%) (96,97).
ing is the only way to detect these changes that occur over long Clinical deterioration or delayed cerebral ischemia after SAH is
periods. Sleep–wake cycles are particularly favorable. The find- a major complication. Angiographic vasospasm is detected in
ing of a less regulated variability, cycling, or alternating EEG 50% to 70% of SAH patients (98) and delayed cerebral ischemia
pattern also carries a favorable prognosis. In postanoxic and in 19% to 46% (99–104) and up to 54% with worse SAH grades
traumatic coma, 95% of patients had unfavorable outcomes (105). Because treatments for vasospasm may prevent infarc-
when their EEG showed a slow monotonous pattern, whereas tion if started early, timely warning is the key. Angiography and
only 30% of patients had unfavorable neurologic outcomes transcranial Doppler are not monitoring techniques, whereas
when their EEG showed variable or sleep–wake cycle EEG pat- EEG monitoring is continuous. EEG also may help guide effec-
terns (83). In a further 500 patients with clinical evidence of tiveness of therapy. Several EEG changes herald the onset of
upper brainstem impairment, the mortality rate was 93% vasospasm: total power trends (106), relative alpha variability
among patients whose EEG showed a slow monotonous pat- (107), and poststimulation alpha/delta ratio (66). EEG in one
tern, 46% among those with a variable pattern, and 28% among study was 100% sensitive for angiographically defined
those patients with a sleep cycle pattern (84). Among patients vasospasm (107). Focal ischemia from vasospasm may produce
with a cortical or subcortical injury from head trauma, the bilateral EEG changes (66,107). EEG correctly predicted a
mortality rates were 63% for a slow monotonous EEG pattern, vasospasm-free course among 73% of 151 SAH patients.
40% for a variable pattern, and 9% for a sleep cycle pattern
(84). EEG monitoring can detect and measure these different Neonatal Applications
patterns, but a routine EEG in the ICU may miss the long-term The simple amplitude integrated trending technique has
changes of variability and sleep cycles. These EEG changes are received attention in the neonatal ICU setting (108). The tech-
not noticeable on physical examination. nique has been applied in recent years as a method for identify-
ing or monitoring a variety of critical care neonatal conditions.
Cerebral Ischemia A greater number of total power bursts per hour were favorable
EEG is altered by cerebral blood flow (CBF) below 18 to for preterm infants with intraventricular hemorrhage
25 mL/100 g/min, whereas ischemic injury occurs at lower CBF (109–111). Features could differentiate preterm infants with
levels of 12 to 18 mL/100 g/min (85–87). EEG changes begin normal neurologic status from those with brain injury
with loss of fast activity and progress at lower CBF to polymor- (112–116). Criteria included voltage, continuity, variability,
phic delta and eventually to isoelectric (88). EEG may demon- bursts, and sleep cycles.
strate brain function recovery before the clinical exam (89). Limitations arise in the use of power trends without EEG
These observations make the EEG a useful tool to monitor tracings. There is considerable expertise and literature built over
brain perfusion. decades on the implications of spikes, sharp waves, symmetry,
Acute ischemic stroke is best visualized on diffusion- and and content of premature neonatal EEG. That literature and
perfusion-weighted MRI. Those techniques are not well suited clinical application depends on interpretation of EEG tracings
for continuous ICU monitoring. EEG can serve a monitoring by knowledgeable electroencephalographers. The use of trends
role, screening for changes suspicious for new acute ischemic by themselves lacks the detail to allow clinical application of that
734 Part V ■ Complementary and Special Techniques

extensive knowledge. As a result, the field of neonatal ICU EEG EEG is feasible (120), as is standard for intraoperative moni-
finds itself where adult EEG was several decades ago when com- toring. Since hook ups are needed at any time 24 hours daily,
pressed spectral array (CSA) (117,118) was the commonly used a process for applying electrodes and initiating monitoring
technique for adult ICU EEG monitoring. requires either a knowledgeable technologist or an allied
health professional. Such health professionals, such as ICU
Methods nurses, might use preconfigured head nets or caps to locate
The principal goal is rapid identification of new complications electrode sites.
in time to intervene, thereby lessening or avoiding long-term Some have suggested that single-channel frequency analysis
neurologic complications. ICU monitoring must be automated systems, such as the BIS monitor (Aspect Medical, Newton,
because nurses cannot devote much time to EEG problem solv- MA) or the SNAP monitor (Everest Biomedical, Chesterfield,
ing. Monitoring should identify quickly abrupt changes as well MO), can be used to monitor patients with status epilepticus.
as gradual changes. However, these limited quantitative methods have great diffi-
Methods for ICU EEG monitoring are borrowed from other culty monitoring seizure activity because changes are not spe-
EEG applications. Continuous recording is taken from epilepsy cific enough. These limitations are similar to the drawbacks that
monitoring. Observing for complications, deterioration, or led to replacement of CSA by modern generation ICU EEG
improvement is taken from intraoperative monitoring. technology.
Trending of features is taken from polysomnography and ear-
lier generation CSA techniques. Conclusion
Prior to 1985, ICU EEG monitoring favored two- or four- ICU EEG monitoring is a method for detecting acute problems
channel CSAs (117,118). CSA monitors were too difficult to in critical care patients in time to intervene and prevent serious
understand and use. In 1985, modern generation EEG monitor- long-term sequellae. It allows for identification of complica-
ing was introduced using innovations that went beyond CSA tions so as to reduce ICU stay and improve clinical outcome.
(119). Those innovations include recording from large number Beginning in 1985 the modern generation ICU EEG monitor-
of EEG channels and storage of the EEG for review. Simpler ing technique has used large numbers of EEG channels, remote
trend displays are easier to understand. Remote access allows access to read the tracings, saving of entire EEG files for review,
for expert review as needed. Both frequency trends and EEG and simplified trending for ease of nurses and nonspecialists.
tracings are continuously displayed at the bedside. Automated Trending also helps to identify gradual changes over hours and
seizure detection is borrowed from epilepsy monitoring tools. days. These and other tools have moved the field well beyond
Nurses are taught simple rules for calling attention to EEG the initial CSA techniques of the 1970s.
changes. More recently, video-EEG monitoring has been added Now EEG ICU monitoring is used to identify seizures and
for seizures (67). status epilepticus, focal impairment, and measure gradual
ICU EEG requires a trained electroencephalographer to improvement or deterioration. Those changes can suggest the
interpret and to review changes frequently. Nurses can be onset of such important clinical complications as hemorrhage
taught the general principles of when changes need attention, or vasospasm. Monitoring helps to show the adequacy of treat-
but it is difficult to teach most ICU nurses sufficient skills to ment with blood pressure control to achieve adequate cerebral
understand many findings. For that reason, a clinical neuro- perfusion, or medications to induce deliberate burst suppres-
physiology physician’s involvement usually is needed several sion. The tools can help with prognosis. The field is better
times daily and on request to interpret the EEG when changes defined and advanced in the adult ICU. In the neonatal ICU,
occur. There are two clinical paradigms for this involvement. In monitoring tools still try to apply trending alone.
one, a critical care neurologist is trained as an electroencephalo- In much of the same way that electrocardiographic monitor-
grapher and includes the EEG among the tests monitored ing is a required element of caring for the myocardial ischemia
throughout the ICU stay. In the second paradigm, an electroen- patient, ICU EEG monitoring is becoming the standard of care
cephalographer interprets the EEG repeatedly during each day required for neurologic critical patient care and others with
and is available for further reviews as needed, and conveys the coma of uncertain cause.
findings to the patient’s critical care physician.
Staff and physician availability are needed to connect AMBULATORY MONITORING
patients, to maintain and interpret recordings. Frequency
analysis trending can improve identification of change and Ambulatory EEG (AEEG) fills a useful niche between routine
reduce reading time but trends should never be interpreted in laboratory-based EEGs and inpatient video-EEG monitoring.
isolation. The original EEG tracings should be the basis for AEEG provides the ability to extend the duration of EEG sur-
definitive interpretation, which requires training in electroen- veillance from the 30 minutes typical of a routine EEG to sev-
cephalography. A 24-hour coverage by an electroencephalog- eral days without requiring hospitalization. In the past, AEEG
rapher in neurologic ICUs is impractical, so trained ICU staff devices offered limited fidelity and flexibility, and could
or specially trained technicians akin to cardiac telemetry tech- record relatively few channels. Those limitations no longer
nicians could screen the trends and tracings and refer ques- apply, and current AEEG systems can produce recordings
tionable events for definitive interpretation remotely by a equivalent in quality to recordings obtained using standard
trained electroencephalographer. Remote access to real-time laboratory systems.
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 735

Clinical Utility of Ambulatory EEG Indications for Ambulatory EEG


The ability to record EEG over extended periods increases the Given these considerations, the principal indications for AEEG
likelihood of capturing interictal discharges and seizures. This monitoring are (128):
information is often critical for accurate diagnosis and clini-
1. Diagnosis: In patients with suspected epilepsy, AEEG may
cal management. In one study, the yield of AEEG monitoring
help to establish the diagnosis of epilepsy by recording inter-
for providing evidence to support the diagnosis of epilepsy
ictal discharges and seizures, and may help to distinguish
was 2.5 times that of a standard EEG, and 83% that of inpa-
epilepsy from nonepileptic seizures and other behavior dis-
tient video-EEG monitoring (121). Importantly, the chance of orders, syncope, cardiac arrhythmias, and disturbances of
capturing seizures is far greater on AEEG than standard EEG; sleep.
in a study comparing sleep-deprived EEGs and AEEGs in 2. Classification: The electrographic patterns and scalp distri-
patients with normal or nondiagnostic standard EEGs, sleep- butions of interictal discharges and seizures, their temporal
deprived EEGs captured no seizures while seizures were patterns of occurrence including precipitating events and
recorded in 15% of patients on AEEG, including in several stimuli, as well as the EEG background activity may provide
patients who were unaware of them (122). Underreporting of useful information for refining the definition of seizure type
seizures is well recognized, in part because amnesia for the and the epilepsy diagnosis.
ictus is common; in one study, patients were unaware of 63% 3. Quantification: AEEG may provide important information
of all seizures occurring in an inpatient epilepsy monitoring regarding the frequency of discharges and seizures, as well as
unit (123). documentation of response to medication. This may be par-
While the ability to record EEG as the patient goes about ticularly important in patients with frequent seizures, of
normal activity is an advantage, daytime recordings are likely which they may not be aware.
to contain substantially more movement- and muscle-related
artifacts than recordings made in an EEG laboratory. Ambulatory EEG Equipment
Nighttime recordings are more likely to be artifact free, and Since the early days of galvanometers and photographic record-
indeed the ability to obtain prolonged sleep recording is an ing devices, clinical neurophysiology has progressed in lockstep
attractive feature of AEEG. Perhaps the greatest limitation of with technology. The first commercial devices recorded four
AEEG is that it is not possible for a technician to monitor channels of EEG (or three EEG plus a time code), and used a
recording quality and electrode integrity, and to adjust elec- four-channel analog 1/8-in. cassette tape recorder designed for
trodes if necessary. Failure of electrode contacts and occasion- general industrial applications (129). A standard tape cassette
ally of ambulatory equipment compromises the quality of could store 24 hours of EEG. EEG signals were amplified using
AEEG recordings. specially designed small single-channel preamplifiers that were
Although AEEG can provide days of continuous EEG mon- affixed, along with electrodes, to the scalp with collodion. A
itoring, it is generally not a substitute for inpatient video-EEG, video display reader station permitted rapid visual review, as
because simultaneous time-locked video provides substantial fast as 1 hour of EEG/minute.
diagnostic information not provided by EEG alone. Most com- In the early 1980s, a nine-channel analog device was intro-
mercial AEEG systems are not designed to record time-locked duced that used standard 1/4-in. recording tape. An important
video along with EEG. Although some AEEG systems do sup- advantage was that eight-channel EEG proved easier to inter-
port video, the requirement that the patient remains “on- pret than three- or four-channel EEG; with eight channels,
camera” defeats one advantage of AEEG, the capability of montages could be constructed that more closely resembled
recording EEG as the patient goes about routine activities. traditional montages. While sensitivity for identification of
Further, inpatient video-EEG monitoring allows trained per- epileptiform abnormalities was similar given three- or eight-
sonnel to observe, to interact with, and to test patients during channel data, review of eight-channel data afforded a lower
seizures or seizure-like episodes, acquiring important informa- false-positive rate (130). Additionally, these devices offered
tion regarding semiology that is not captured by unsupervised greater flexibility for adjustment of filters and gain, printouts of
video recording. The inpatient setting makes it possible to safely selectable segments, and capability to easily locate the EEG
withdraw medication to increase the likelihood of recording recorded at specific times during the study. A useful new feature
seizures, and, of course, the inpatient setting is uniquely suited was the ability to convert one or several EEG channels to audio,
to invasive EEG recording (3). allowing seizures to be identified by listening to high-speed
These limitations notwithstanding, one group has reported playback (128).
the successful use of outpatient monitoring as part of the Attachment of eight on-head preamplifiers to scalp with col-
presurgical evaluation of a group of patients with temporal lodion proved cumbersome and time consuming. Further,
lobe epilepsy (124). Further, seizure frequency is often although in principle on-head amplification can reduce artifact
reported to decrease on hospitalization (125,126), perhaps related to electrode lead length and movement, it became
related to medication compliance or environment stressors apparent that on-head amplification could not reduce artifact
(124). Some have suggested a role for AEEG in conjunction from some important sources, including EMG and eye move-
with inpatient video-EEG monitoring, either prior to or ment. Further, fixing a standard EEG lead wire to the scalp with
following hospitalization (127). collodion next to the electrode cup effectively mitigates
736 Part V ■ Complementary and Special Techniques

electrode artifacts produced by movement at the electrode limiting factor, and present-day systems retain all EEG recorded.
cup–scalp interface (128). With the introduction of eight- When the study is complete, data are easily transferred to a desk-
channel AEEG devices, off-head amplifiers for use with stan- top computer for review, and if desired, processing by automatic
dard EEG electrodes became available. The trend toward spike and seizure detection software (27,30). As with inpatient
off-head amplification has continued; nonetheless, on-head LTM recordings, the physician has the option of exhaustively
amplification may be superior under conditions of vigorous reviewing the entire recording, or relying on periodic samples,
movement, for example, during a convulsive seizure. Direct “push button events,” and automated detections.
bipolar recordings can provide signal quality superior to that
produced by referential recordings reformatted into bipolar Future Directions
montages (128,131). AEEG has the potential to provide accurate information about
During the 1980s, 16-channel digital AEEG devices were the seizure frequency that, in some patients, may be difficult to
introduced, still using tape for storage. Initially, the trade-off for obtain otherwise (134). Underreporting of seizures by patients
16 channels was limited storage capabilities; instead of retaining is well recognized (123), in part because reporting requires the
the complete recording, intermittent samples, for example, 15 patient to remember an event that itself commonly produces
seconds every 10 minutes, as well as sections before and after amnesia. When seizures go undetected and underreported,
manual activation of a push button by the patient, were stored. medical management may be compromised because the effi-
Subsequently hybrid “computer-assisted” AEEG systems com- cacy of treatment can be difficult to ascertain. The apparent
bined a 16-channel waist-worn device with a portable com- relationship between seizure frequency and the risk of sudden
puter, connected by a wire tether, that processed EEG in real unexplained death in patients with epilepsy underscores the
time using automated spike and seizure detection software importance of accurate measurement (135).
(132,133). Data available for clinical review consisted of inter- Proof of concept for chronic AEEG has been effectively pro-
mittent samples, manually recognized events, and automatic vided by Neuropace Inc.’s investigational responsive neurostim-
detections, in much the same manner as the inpatient ulation (RNS) device (136,137). Implanted within the skull and
video/monitoring systems in use at the time. connected to subdural or depth electrodes, the device is
Current AEEG devices represent an important departure designed to detect seizures and deliver electrical stimulation
from past designs. Present-day digital technology makes possible intended to abort the seizure (138). The device also contains
portable EEG devices that are not much larger than a PDA or a memory sufficient to store about 30 minutes of four-channel
cell phone and are able to record EEG with fidelity and number EEG corresponding to seizure detections, allowing it to func-
of channels equal to standard laboratory EEG machines. Indeed, tion as a chronic AEEG recorder.
the designs of AEEG systems and standard EEG systems have Technology is presently within reach that should make the
largely merged. In a typical EEG machine, amplification and design of a device optimized for chronic AEEG recording and
analog to digital conversion are performed in the “headbox”; the monitoring feasible. High-capacity, dense, solid-state storage is
computer performs control, data storage, and display functions. now in widespread use, in standard AEEG systems as well as
An AEEG system simply adds battery power and solid-state consumer electronic devices. Newly developed power harvesting
memory to the headbox. Since some headboxes, particularly techniques may provide a solution to the problems of power
those intended for use in inpatient monitoring units, often con- consumption and battery size and life that have, until now,
tain memory and rechargeable batteries, permitting the patient plagued implantable devices (139). In contrast to standard near-
to disconnect from the wired system for several hours, real dif- field inductive coupling, used to recharge consumer devices and
ferences between AEEG systems and other EEG systems relate to which requires that the power source and powered device be in
packaging, quantity of storage, battery life, and marketing. close proximity, power can be transmitted using radio frequency
The typical modern AEEG recorder provides for 21 or more (RF) energy over somewhat greater distances. Holleman et al.
referential channels of EEG that can be reformatted in the same recently described a low-power (20 A at 1.9 V) prototype bat-
manner as standard laboratory EEG. It samples EEG with 16-bit tery-free neural interface, which counted action potentials/sec-
resolution at typically 200 or 256 Hz/channel. It also has several ond, and was powered by a 900-MHz radio signal at a distance
channels for inputs such as pulse oximetry and other of 1 m (140). Proprietary RF harvesting techniques claim to be
polysomnographic devices, EMG and EKG. It connects to a desk- capable of safely providing milliwatt-range power over even
top computer using USB or Ethernet for startup, and for down- greater distances (141). It is easy to envision a device implanted
load and review of data. It weighs just over 1 lb, is powered by a in the skull that records full-fidelity EEG through epidural peg
several AA or rechargeable batteries, and has 2 to 4 GB of flash electrodes. Epidural electrodes would provide high-quality sta-
memory capable of storing 2 to 4 days of continuous data. AEEG ble EEG signals, relatively immune from artifacts. At night, while
systems are also available that support time-synchronized video. the patient sleeps, the device would recharge while data were
Real-time processing of EEG for spike and seizure detection automatically downloaded to a computer that would run seizure
was important to the design of earlier computer-assisted AEEG detection software, logging the results or transmitting them,
systems that had insufficient storage capacity to retain entire 16- along with raw data if desired, back to the physician.
channel day-long EEG recordings, and instead stored only sam- An attractive alternative to wired connections between mul-
ples and automatic detections. Storage capacity is no longer a tiple transcranial passive peg electrodes and an implanted AEEG
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 737

recording device could be a wireless local network, consisting of 14. Theodore WH, Porter RJ, Albert P, et al. The secondarily generalized
small active peg electrodes (142) and the AEEG recorder, per- tonic–clonic seizure: a videotape analysis. Neurology. 1994;44(8):
haps implanted in the chest or abdominal wall. Each peg would 1403–1407.
record a channel of EEG and transmit it back to the recorder, 15. Spencer SS, Spencer DD, Williamson PD, et al. Ictal effects of anti-
convulsant medication withdrawal in epileptic patients. Epilepsia.
where the data would be stored for subsequent download. The
1981;22(3):297–307.
network could use the Medical Implant Communication Service
16. Marciani MG, Gotman J, Andermann F, et al. Patterns of seizure
band, 402 to 405 kHz, recently established by the FCC for radio activation after withdrawal of antiepileptic medication. Neurology.
transmission to implanted medical devices (143). This concept 1985;35(11):1537–1543.
is readily expanded to include continuous recording of other 17. Gotman J, Marciani MG. Electroencephalographic spiking activ-
physiologic signals, such as EKG, respiration, and blood oxygen ity, drug levels, and seizure occurrence in epileptic patients. Ann
saturation. The relatively liberal power budget afforded by RF Neurol. 1985;17(6):597–603.
power harvesting might permit the functionality of implanted 18. Gotman J, Koffler DJ. Interictal spiking increases after seizures but
AEEG systems to include not only data storage, but also real- does not after decrease in medication. Electroencephalogr Clin
time seizure detection and, if effective methodologies emerge, Neurophysiol. 1989;72(1):7–15.
seizure prediction and warning (23). 19. Spencer SS, Goncharova II, Duckrow RB, et al. Interictal spikes on
intracranial recording: behavior, physiology, and implications.
Epilepsia. 2008;49(11):1881–1892.
REFERENCES 20. Mizrahi EM. Pediatric electroencephalographic video monitoring.
J Clin Neurophysiol. 1999;16(2):100–110.
1. Gotman J. Seizure recognition and analysis. In: Gotman J, Ives JR, 21. Qu H, Gotman J. A patient-specific algorithm for the detection of
Gloor P, eds. Long-Term Monitoring in Epilepsy. Amsterdam: Elsevier; seizure onset in long-term EEG monitoring: possible use as a
1985:133–145. warning device. IEEE Trans Biomed Eng. 1997;44(2):115–122.
2. Hunter J, Jasper HH. A method of analysis of seizure pattern and elec- 22. Shoeb A, Edwards H, Connolly J, et al. Patient-specific seizure
troencephalogram; a cinematographic technique. Electroencephalogr onset detection. Epilepsy Behav. 2004;5(4):483–498.
Clin Neurophysiol. 1949;1(1):113. 23. Mormann F, Andrzejak RG, Elger CE, et al. Seizure prediction: the
3. Velis D, Plouin P, Gotman J, et al. ILAE DMC Subcommittee on long and winding road. Brain. 2007;130:314–333.
Neurophysiology. Recommendations regarding the requirements 24. Gotman J. Interhemispheric relations during bilateral spike-and-
and applications for long-term recordings in epilepsy. Epilepsia. wave activity. Epilepsia. 1981;22(4):453–466.
2007;48(2):379–384. 25. Gotman J. Automatic seizure detection: improvements and
4. Mirsattari SM, Tapsell LM, Ives JR, et al. MR imaging-compatible evaluation. Electroencephalogr Clin Neurophysiol. 1990;76(4):
electroencephalography electrode system for an epilepsy monitor- 317–324.
ing unit. Am J Neuroradiol. 2008;29(9):1649–1651. 26. Grewal S, Gotman J. An automatic warning system for epileptic
5. Mirsattari SM, Davies-Schinkel C, Young GB, et al. Usefulness of a seizures recorded on intracerebral EEGs. Clin Neurophysiol.
1.5 T MRI-compatible EEG electrode system for routine use in the 2005;116(10):2460–2472.
intensive care unit of a tertiary care hospital. Epilepsy Res. 2009; 27. Saab ME, Gotman J. A system to detect the onset of epileptic
84(1):28–32. seizures in scalp EEG. Clin Neurophysiol. 2005;116(2):427–442.
6. Jirsch JD, Urrestarazu E, LeVan P, et al. High-frequency oscillations 28. Gabor AJ, Leach RR, Dowla FU. Automated seizure detection using a
during human focal seizures. Brain. 2006;129:1593–1608. self-organising neural network. Electroencephalogr Clin Neurophysiol.
7. Urrestarazu E, Chander R, Dubeau F, et al. Interictal high- 1996;99:257–266.
frequency oscillations (100–500 Hz) in the intracerebral EEG of 29. Webber WRS, Lesser RP, Richardson RT, et al. An approach to
epileptic patients. Brain. 2007;130:2354–2366. seizure detection using an artificial neural network. Electro-
8. Jacobs J, LeVan P, Chander R, et al. Interictal high-frequency oscil- encephalogr Clin Neurophysiol. 1996;98:250–272.
lations (80–500 Hz) are an indicator of seizure onset areas inde- 30. Wilson SB, Scheuer ML, Emerson RG, et al. Seizure detection:
pendent of spikes in the human epileptic brain. Epilepsia. evaluation of the Reveal algorithm. Clin Neurophysiol. 2004;
2008;49:1893–1907. 115(10):2280–2291.
9. Vanhatalo S, Voipio J, Kaila K. Full-band EEG (fbEEG): a new stan- 31. Qu H, Gotman J. A seizure warning system for long-term epilepsy
dard for clinical electroencephalography. Clin EEG Neurosci. monitoring. Neurology. 1995;45(12):2250–2254.
2005;36(4):311–317. 32. Wilson SB. A neural network method for automatic and incre-
10. Blum D. Prevalence of bilateral partial seizure foci and implica- mental learning applied to patient-dependent seizure detection.
tions for electroencephalographic telemetry monitoring and Clin Neurophysiol. 2005;116(8):1785–1795.
epilepsy surgery. Electroencephalogr Clin Neurophysiol. 1994; 33. Gotman J, Flanagan D, Rosenblatt B, et al. Evaluation of an auto-
91(5):329–336. matic seizure detection method for the newborn EEG. Electro-
11. Marciani MG, Gotman J. Effects of drug withdrawal on location of encephalogr Clin Neurophysiol. 1997;103(3):363–369.
seizure onset. Epilepsia. 1986;27(4):423–431. 34. Celka P, Colditz P. A computer-aided detection of EEG seizures in
12. So N, Gotman J. Quantitative EEG analysis of carbamazepine infants: a singular-spectrum approach and performance compari-
effects on amygdaloid kindled seizures in cats. Electroencephalogr son. IEEE Trans Biomed Eng. 2002;49(5):455–462.
Clin Neurophysiol. 1990;76(1):63–72. 35. Aarabi A, Wallois F, Grebe R. Automated neonatal seizure detec-
13. Zhou D, Wang Y, Hopp P, et al. Influence on ictal seizure semiol- tion: a multistage classification system through feature selection
ogy of rapid withdrawal of carbamazepine and valproate in based on relevance and redundancy analysis. Clin Neurophysiol.
monotherapy. Epilepsia. 2002;43(4):386–393. 2006;117(2):328–340.
738 Part V ■ Complementary and Special Techniques

36. Aarts JH, Binnie CD, Smit AM, et al. Selective cognitive impair- 60. Sorensen K, Thomassen A, Wernberg M. Prognostic significance of
ment during focal and generalized epileptiform EEG activity. alpha frequency EEG rhythm in coma after cardiac arrest. J Neurol
Brain. 1984;107(pt 1):293–308. Neurosurg Psychiatry. 1978;41:840.
37. Shewmon DA, Erwin RJ. Transient impairment of visual percep- 61. Westmoreland B, Klass OW, Sharbrough FW, et al. Alpha-coma:
tion induced by single interictal occipital spikes. J Clin Exp electroencephalographic, clinical, pathologic, and etiologic corre-
Neuropsychol. 1989;11(5):675–691. lations. Arch Neurol. 1975;32:713.
38. Wilson SB, Harner RN, Duffy FH, et al. Spike detection. I. 62. Jordan KG. Neurophysiologic monitoring in the neuroscience
Correlation and reliability of human experts. Electroencephalogr intensive care unit. Neurol Clin. 1995;13:579–626.
Clin Neurophysiol. 1996;98(3):186–198. 63. Archibald JE, Drazkowski JF. Clinical applications of compressed
39. Benbadis SR, Lin K. Errors in EEG interpretation and misdiagno- spectral analysis (CSA) in OR/ICU settings. Am J EEG Technol.
sis of epilepsy. Which EEG patterns are overread? Eur Neurol. 1985;25:13–36.
2008;59:267–271. 64. Vespa PM, Nuwer MR, Nenov V, et al. Increased incidence and
40. Halford JJ. Computerized epileptiform transient detection in the impact of nonconvulsive and convulsive seizures after traumatic
scalp electroencephalogram: obstacles to progress and the example brain injury as detected by continuous EEG in the intensive care
of computerized ECG interpretation. Clin Neurophysiol. 2009; unit. J Neurosurg. 1999;91:750–760.
120(11):1909–1915. 65. Vespa PM, O’Phelan K, Mirabelli J, et al. Acute seizures after
41. Jordan KG. Continuous EEG and evoked potential monitoring in intracerebral hemorrhage: a factor in progressive midline shift and
the neuroscience intensive care unit. J Clin Neurophysiol. 1993; outcome. Neurology. 2003;60:1441–1446.
10:445–475. 66. Claassen J, Hirsch LJ, Kreiter KT, et al. Quantitative continuous EEG
42. Jordan KG. Nonconvulsive seizures and nonconvulsive status for detecting delayed cerebral ischemia in patients with poor-grade
epilepticus detected by continuous EEG monitoring in the neuro- subarachnoid hemorrhage. Clin Neurophysiol. 2004;115:2699–2710.
ICU. Neurology. 1992;42(suppl 3):194. 67. Pandian JD, Cascino GD, So EL, et al. Digital video-electroen-
43. Ballenger CE III, King OW, Gallagher B. Partial complex status cephalographic monitoring in the neurological–neurosurgical
epilepticus. Neurology. 1983;33:1545–1552. intensive care unit: clinical features and outcome. Arch Neurol.
44. Bauer G, Aichner F, Mayr U. Nonconvulsive status epilepticus fol- 2004;61(7):1090–1094.
lowing generalized tonic–clonic seizures. Eur Neurol. 1982;21: 68. Maynard D, Prior PF, Scott DF. Device for continuous monitoring
411–419. of cerebral activity in resuscitated patients. Br Med J. 1969;4:
45. Doose H. Nonconvulsive status epilepticus in childhood: clinical 545–546.
aspects and classification. Adv Neurol. 1984;34:115–125. 69. Prior P. The EEG in Acute Cerebral Anoxia. Amsterdam: Excerpta
46. Ellis JM, Lee SI. Acute prolonged confusion in later life as an ictal Medica; 1973.
state. Epilepsia. 1978;19:119–128. 70. Stidham GL, Nugent SK, Rogers MC. Monitoring cerebral electri-
47. Engel J Jr, ed. Seizures and Epilepsy. Philadelphia, PA: F.A. Davis; cal function in the ICU. Crit Care Med. 1980;8:519–523.
1989. 71. Young GB, Jordan KG, Doig GS. An assessment of nonconvulsive
48. Escueta AV, Boxley J, Stubbs N, et al. Prolonged twilight state and seizures in the intensive care unit using continuous EEG monitoring:
automatisms: a case report. Neurology. 1974;24:331–339. an investigation of variables associated with mortality. Neurology.
49. Porter RJ, Penry JK. Petit mal status. Adv Neurol. 1983;34:61–67. 1996;47(1):83–89.
50. Pritchard PB III, O’Neal DB. Nonconvulsive status epilepticus 72. Grand’Maison F, Reiher J, Laduke CP. Retrospective inventory of
following metrizamide myelography. Ann Neurol. 1984;16: EEG abnormalities in partial status epilepticus. Electroencephalogr
252–254. Clin Neurophysiol. 1991;79:264–270.
51. Rumpl E, Hinterhuber H. Unusual “spike-wave stupor” in a 73. Scholtes FB, Renier WO, Meinardi H. Nonconvulsive status epilep-
patient with manic-depressive psychosis treated with amitripty- ticus: causes, treatment, and outcome in 65 patients. J Neurol
line. J Neurol. 1981;226:131–135. Neurosurg Psychiatry. 1996;61:93–95.
52. Rumpl E, Lorenzi E, Hackl J, et al. The EEG at different stages of 74. Litt B, Dizon L, Ryan D. Fatal nonconvulsive status epilepticus in
acute secondary traumatic midbrain and bulbar brain syndromes. the elderly. Epilepsia. 1994;35:10–17.
Electroencephalogr Clin Neurophysiol. 1981;46:487–497. 75. Shinton RA, Gill JS, Melnick AK. The frequency, characteristics,
53. Chatrian GE, Shaw CM, Leffman H. The significance of periodic and prognosis of epileptic seizures at the onset of stroke. J Neurol
lateralized epileptiform discharges in EEG: an electrographic, clin- Neurosurg Psychiatry. 1988;51:273–276.
ical and pathological study. Electroencephalogr Clin Neurophysiol. 76. Olsen TS, Hogenhaven H, Thage O. Epilepsy after stroke.
1964;17:177. Neurology. 1987;37:1209–1211.
54. Markland ON, Daly DO. Pseudoperiodic lateralized paroxysmal 77. Lancman ME, Crolinstock A, Morseini J, et al. Risk factors for
discharges in electroencephalogram. Neurology. 1971;21:975. developing seizures after a stroke. Epilepsia. 1993;34:141–143.
55. Austin EI, Wilkus RJ, Longstreth WT. Etiology and prognosis of 78. Vespa PM, Ronne-Engström E, Smith M, et al. Increase in extracel-
alpha coma. Neurology. 1988;38:773. lular glutamate caused by reduced cerebral perfusion pressure and
56. Carroll WM, Mastaglia FL. Alpha and beta coma in drug intoxica- seizures after human traumatic brain injury: a microdialysis study.
tion uncomplicated by cerebral hypoxia. Electroencephalogr Clin J Neurosurg. 1998;89:971–982.
Neurophysiol. 1979;46:95. 79. Vespa PM, Martin NA, Nenov V, et al. Delayed increase in extracel-
57. Chokroverty S. “Alpha like” rhythms in electroencephalograms in lular glycerol with post-traumatic electrographic epileptic activity:
coma after cardiac arrest. Neurology. 1975;25:655. support for the theory that seizures induce secondary injury. Acta
58. Grindal AB, Suter C, Martinez AJ. Alpha-pattern coma: 24 cases Neurochir Suppl. 2002;81:355–357.
with 9 survivors. Ann Neurol. 1977;1:371. 80. Bergamasco B, Bergamini L, Doriguzzi T. Clinical value of the
59. Iragui VJ, McCutchen CB. Physiologic and prognostic significance sleep electroencephalographic patterns in post-traumatic coma.
of “alpha coma.” J Neurol Neurosurg Psychiatry. 1983;46:632. Acta Neurol Scand. 1968;44:495–511.
Chapter 35 ■ Principles and Techniques for Long-Term EEG Recording (EMU, ICU, Ambulatory) 739

81. Bergamasco B, Bergamini L, Doriguzzi T, et al. EEG sleep pat- 101. Charpentier C, Audibert G, Guillemin F, et al. Multivariate analy-
terns as a prognostic criterion in post-traumatic coma. Electro- sis of predictors of cerebral vasospasm occurrence after aneurys-
encephalogr Clin Neurophysiol. 1968;24:374–377. mal subarachnoid hemorrhage. Stroke. 1999;30:1402–1408.
82. Bricolo A, Turella G, Ore GD, et al. A proposal for the EEG eval- 102. Hop JW, Rinkel GJ, Algra A, et al. Initial loss of consciousness and
uation of acute traumatic coma in neurosurgical practice. risk of delayed cerebral ischemia after aneurysmal subarachnoid
Electroencephalogr Clin Neurophysiol. 1973;34:789. hemorrhage. Stroke. 1999;30:2268–2271.
83. Bricolo A, Turazzi S, Faccioli F, et al. Clinical application of 103. Qureshi AI, Sung GY, Razumovsky AY, et al. Early identification
compressed spectral array in long-term EEG monitoring of of patients at risk for symptomatic vasospasm after aneurismal
comatose patients. Electroencephalogr Clin Neurophysiol. 1978; subarachnoid hemorrhage. Crit Care Med. 2000;28:984–990.
45:211–225. 104. Claassen J, Bernardini GL, Kreiter K, et al. Effect of cisternal and
84. Bricolo A, Turazzi S, Faccioli F. Combined clinical and EEG exam- ventricular blood on risk of delayed cerebral ischemia after sub-
inations for assessment of severity of acute head injuries: electro- arachnoid hemorrhage: the Fisher scale revisited. Stroke. 2001;
physiological methods. Acta Neurochir Suppl. 1979;28:35–39. 32:2012–2020.
85. Astrup J, Siesjö BK, Symon L. Thresholds in cerebral ischemia— 105. Kirmani JF, Qureshi AI, Hanel RA, et al. Silent cerebral infarction
the ischemic penumbra. Stroke. 1981;12:723–725. in poor-grade patients with subarachnoid hemorrhage. Neurology.
86. Sundt TM Jr, Sharbrough FW, Piepgras DG, et al. Correlation of 2002;58(7):159.
cerebral blood flow and electroencephalographic changes during 106. Labar DR, Fisch BJ, Pedley TA, et al. Quantitative EEG monitor-
carotid endarterectomy: with results of surgery and hemody- ing for patients with subarachnoid hemorrhage. Electro-
namics of cerebral ischemia. Mayo Clin Proc. 1981;56:533–543. encephalogr Clin Neurophysiol. 1991;78:325–332.
87. Baron JC. Perfusion thresholds in human cerebral ischemia: his- 107. Vespa PM, Nuwer MR, Juhasz C, et al. Early detection of
torical perspective and therapeutic implications. Cerebrovasc Dis. vasospasm after acute subarachnoid hemorrhage using continu-
2001;11:2–8. ous EEG ICU monitoring. Electroencephalogr Clin Neurophysiol.
88. Nuwer MR, Jordan SE, Ahn SS. Evaluation of stroke using EEG 1997;103:607–615.
frequency analysis and topographic mapping. Neurology. 1987; 108. Hellström-Westas L, De Vries LS, Rosén I. Neonatal ICU moni-
37:1153–1159. toring. In: Marc R, Nuwer MR, eds. Intraoperative Monitoring of
89. Jordan KG. Continuous EEG monitoring in the neuroscience Neural Function. Handbook of Clinical Neurophysiology.
intensive care unit and emergency department. J Clin Neuro- Amsterdam: Elsevier; 2008.
physiol. 1999;16:14–39. 109. Greisen G, Hellström-Westas L, Lou H, et al. EEG depression and
90. Finnigan SP, Rose SE, Walsh M, et al. Correlation of quantita- germinal layer haemorrhage in the newborn. Acta Paediatr Scand.
tive EEG in acute ischemic stroke with 30-day NIHSS score: 1987;76:519–525.
comparison with diffusion and perfusion MRI. Stroke. 110. Hellström-Westas L, Rosen I, Svenningsen NW. Cerebral function
2004;35:899–903. monitoring during the first week of life in extremely small low
91. Suzuki A, Yoshioka K, Yasui N. Clinical application of EEG birthweight (ESLBW) infants. Neuropediatrics. 1991;22:27–32.
topography in cerebral ischemia: detection of functional 111. Hellström-Westas L, Klette H, Thorngren-Jerneck K, et al. Early
reversibility and hemodynamics. Brain Topogr. 1990;3:167–174. prediction of outcome with aEEG in preterm infants with large
92. Van Putten MJ, Tavy DL. Continuous quantitative EEG monitor- intraventricular hemorrhages. Neuropediatrics. 2001;32:319–324.
ing in hemispheric stroke patients using the brain symmetry 112. Olischar M, Klebermass K, Kuhle S, et al. Reference values for
index. Stroke. 2004;35:2489–2492. amplitude-integrated electroencephalographic activity in preterm
93. Schneider AL, Jordan KG. Regional attenuation without delta infants younger than 30 weeks’ gestational age. Pediatrics. 2004;
(RAWOD): a distinctive EEG pattern that can aid in the diagno- 113:e61–e66.
sis and management of severe acute ischemic stroke. Am J 113. Sisman J, Campbell D, Brion LP. Amplitude integrated EEG in
Electroneurodiagn Technol. 2005;45:102–117. preterm infants: maturation of background pattern and amplitude
94. Daly DD, Markand ON. Focal brain lesions. In: Daly DD, Pedley voltage with postmenstrual age and gestational age. J Perinatol.
TA, eds. Current Practice of Clinical Electroencephalography. 2nd 2005;25:391–396.
ed. New York, NY: Raven Press; 1990:335–370. 114. Al Naqeeb N, Edwards AD, Cowan F, et al. Assessment of neona-
95. Niedermeyer E. Cerebrovascular disorders and EEG. In: tal encephalopathy by amplitude integrated electroencephalogra-
Niedermeyer E, Lopes da Silva F, eds. Electroencephalography. 5th phy. Pediatrics. 1999;103:1263–1271.
ed. Baltimore, MD: Urban & Schwarzenberg Inc.; 2005:339–362. 115. Hellström-Westas L, Rosen I, Svenningsen NW. Predictive value
96. Claassen J, Hirsch LJ, Frontera JA, et al. Prognostic significance of of early continuous amplitude integrated EEG recordings on
continuous EEG monitoring in patients with poor-grade sub- outcome after severe birth asphyxia in full term infants. Arch Dis
arachnoid hemorrhage. Neurocrit Care. 2006;4:103–112. Child. 1995;72:F34–F38.
97. Hirsch LJ, Claassen J, Mayer SA, et al. Stimulus-induced rhyth- 116. Toet MC, Hellström-Westas L, Groenendaal F, et al. Amplitude
mic, periodic, or ictal discharges (SIRPIDs): a common EEG integrated EEG 3 and 6 hours after birth in full term neonates
phenomenon in the critically ill. Epilepsia. 2004;45:109–123. with hypoxic-ischaemic encephalopathy. Arch Dis Child Fetal
98. Weir B, Grace M, Hansen J, et al. Time course of vasospasm in Neonatal Ed. 1999;81:F19–F23.
man. J Neurosurg. 1978;48:173–178. 117. Bickford RG, Fleming N, Billinger T. Compression of EEG data.
99. Hijdra A, Van Gijn J, Nagelkerke NJ, et al. Prediction of delayed Trans Am Neurol Assoc. 1971;96:118–122.
cerebral ischemia, rebleeding, and outcome after aneurysmal 118. Karnaze DS, Marshall LF, Bickford RG. EEG monitoring of
subarachnoid hemorrhage. Stroke. 1988;19:1250–1256. clinical coma: the compressed spectral array. Neurology. 1982;32:
100. Murayama Y, Malisch T, Guglielmi G, et al. Incidence of cerebral 289–292.
vasospasm after endovascular treatment of acutely ruptured 119. Nuwer MR. EEG and evoked potentials: monitoring cerebral func-
aneurysms: report on 69 cases. J Neurosurg. 1997;87:830–835. tion in the neurosurgical ICU. In: Martin NA, ed. Neurosurgery
740 Part V ■ Complementary and Special Techniques

Clinics of North America, Vol. 5, No. 4, Neurosurgical Intensive Care. 135. Tomson T, Nashef L, Ryvlin P. Sudden unexpected death in
Philadelphia, PA: W.B. Saunders; 1994:647–659. epilepsy: current knowledge and future directions. Lancet Neurol.
120. Scheuer ML, Wilson SB. Data analysis for continuous EEG mon- 2008;7(11):1021–1031.
itoring in the ICU: seeing the forest and the trees. J Clin 136. Spanaki M, Smith B, Burdette D, et al. Chronic measurement of
Neurophysiol. 2004;21:353–378. increased epileptiform activity during menses using the responsive
121. Bridgers SL, Ebersole J. Ambulatory cassette EEG in clinical practice: neurostimulator system (RNS) in a patient with catamenial
experience with 500 patients. Neurology. 1985;35(12): 1167–1168. seizures; 2005. Available at: http://www.neuropace.com/resources/
122. Liporace J, Tatum W 4th, Morris GL 3rd, et al. Clinical utility of publications/0512.html#8. Accessed May 19, 2010.
sleep-deprived versus computer-assisted ambulatory 16-channel 137. Vossler DG, Tcheng T. Ambulatory intracranial ictal electroen-
EEG in epilepsy patients: a multi-center study. Epilepsy Res. cephalogram patterns recorded chronically using the first
1998;32:357–362. implanted, self-contained ECOG recording and analysis instru-
123. Blum DE, Eskola J, Bortz JJ, et al. Patient awareness of seizures. ments; 2005. Available at: http://www.neuropace.com/resources/
Neurology. 1966;47:260–264. publications/0512.html#9. Accessed May 19, 2010.
124. Chang BS, Ives JR, Schomer DL, et al. Outpatient EEG monitor- 138. Fischell RE, Rischell DR. Integrated system for EEG monitoring
ing in the presurgical evaluation of patients with refractory tem- and electrical stimulation with a multiplicity of electrodes.
poral lobe epilepsy. J Clin Neurophysiol. 2002;19(2):152–156. United States patent no. 6,320,049 B1. 2001.
125. Nuwer MR, Engel J Jr, Sutherling WW, et al. Monitoring at the 139. Sauer C, Stanacevic M, Cauwenberghs G, et al. Power harvesting
University of California, Los Angeles. Electroencephalogr Clin and telemetry in CMOS for implanted devices. IEEE Trans
Neurophysiol. 1985;37:385–402. Circuits Syst I Regular Pap. 2005;52(10):2605–2613.
126. Riley T, Porter RJ, White BG, et al. The hospital experience and 140. Holleman J, Yeager D, Prasad R, et al. NeuralWISP: an energy-
seizure control. Neurology. 1981;31:912–915. harvesting wireless neural interface with 1-m range. In: IEEE
127. Schomer DL. Ambulatory EEG telemetry: how good is it? J Clin Biological Circuits and Systems (BioCAS) Conference, November
Neurophysiol. 2006;23(4):294–305. 2008. Available at: http://web.media.mit.edu/~jrs/ neuralwisp.pdf.
128. Ebersole J. Ambulatory cassette EEG. J Clin Neurophysiol. 1985; Accessed May 19, 2010.
2(4):397–418. 141. Powercast I; 2008. Available at: http://powercastco.com/life-
129. Ives JR, Woods J. 4-Channel 24 hour cassette recorded for long- time_power_app.html. Accessed May 19, 2010.
term EEG monitoring of ambulatory patients. Electroencephalogr 142. Bhutani A, McGarvey Z, Frye J, et al. Remote monitoring of EEG
Clin Neurophysiol. 1975;39:88–92. signals through wireless sensor networks. In: Multi-Disciplinary
130. Ebersole JS, Bridgers SL. Direct comparison of 3- and 8-channel Engineering Design Conference; 2008. Rochester Institute of
ambulatory cassette EEG with intensive inpatient monitoring. Technology. Available at: http://edge.rit.edu/content/P08050/
Neurology. 1985;35:846–854. pu blic/ PDF%20Docu m en t at ion / P08050_Tech Paper.pdf.
131. Schomer DL, Ives JR, Schachter SC. The role of ambulatory EEG in Accessed May 19, 2010.
the evaluation of patients for epilepsy surgery. J Clin Neurophysiol. 143. Yuce MR, Ng SW, Myo NL, et al. Wireless body sensor network
1999;16(2):116–129. using medical implant band. J Med Syst. 2007;31:467–474.
132. Gotman J. Automatic recognition of epileptic seizures in the 144. Vespa PM, Nenov V, Nuwer MR. Continuous EEG monitoring in
EEG. Electroencephalogr Clin Neurophysiol. 1982;54:530–540. the intensive care unit: early findings and clinical efficacy. J Clin
133. Morris GL III, Galezowska J, Leroy R, et al. The results of Neurophysiol. 1999b;16:1–13.
computer-assisted ambulatory 16-channel EEG. Electro- 145. Young GB, Doig GS. Continuous EEG monitoring in comatose
encephalogr Clin Neurophysiol 1994;91:229–231. intensive care patients: epileptiform activity in etiologically dis-
134. Tatum WO, Winters L, Gieron M, et al. Results of 502 patients tinct groups. Neurocrit Care. 2005;2(2):5–10.
using computer-assisted ambulatory EEG. J Clin Neurophysiol.
2001;18(1):14–19.
CHAPTER

Infraslow EEG Activity


SAMPSA VANHATALO, JUHA VOIPIO, AND KAI KAILA 36
INTRODUCTION Hence, instead of the old “DC-EEG” nomenclature, we pre-
fer using the term Full-band EEG (FbEEG). This conveys the
The conventional way of recording EEG is more than half a cen- idea that the full, physiologically and clinically relevant EEG
tury old. The early pioneers were faced with the challenge to bandwidth can be readily examined without favoring any fre-
design routine EEG techniques that enabled recordings of the quency band at the expense of another. The term DC shift is
most salient features in the human EEG known at that time. A used here to indicate slow shifts in FbEEG baseline that would
major obstacle was encountered in attempts to faithfully record go unnoticed in conventional EEG. DC-coupled amplifiers are
slow events. Electrode drifts that produced artifactual slow sig- devices that record electrode signals without elimination of the
nals tended to contaminate the recordings and to saturate the FbEEG baseline by inbuilt high-pass filters.
amplifier’s dynamic range, and they also pushed the polygraph
recorder pens out of scale. In order to circumvent these prob-
EVENTS VERSUS OSCILLATIONS
lems, the EEG amplifiers became furnished with an inbuilt
high-pass filter. Hence, all kinds of slow signals, whether physi- In the context of infraslow activity, it is especially important to
ologic or artifactual, were eliminated. acknowledge the distinction between events and oscillations (6).
In practice, the technical compromise described above A variety of EEG phenomena with a duration of seconds are,
means that the conventional EEG has a poor low-frequency indeed, not genuine oscillations but discrete events. As will be
response, which results in attenuation and distortion of signals shown below, spontaneous activity transients (SATs) in
at <0.5 Hz. This is in sharp contrast with a wealth of data show- preterm babies or arousal shifts in older subjects are very slow
ing that the physiologic frequency range of human EEG signals transient events nesting complex patterns of high-frequency
ranges from infraslow (0.01 Hz or even less; this chapter) to EEG activity, whereas ictal DC shifts during epileptic seizures
ultrafast (up to several hundreds of hertz; Chapter 37) frequen- are seen as slow deflections in the recording baseline. Although
cies. In this chapter, several lines of evidence will be presented the infraslow activity seen during sleep or cognitive tasks
to demonstrate that, at the present stage of EEG technology, sta- clearly reflects cyclic changes, a well-defined oscillation fre-
bility is not an issue that would require any trade-off at the quency is not discernible in the spectrograms. Hence it has
expense of low-frequency response. Notably, filtering off the been called either infraslow oscillation (ISO) (7) or infraslow
lower end of the EEG spectrum by the standard high pass at fluctuation (ISF) (8).
around 0.5 Hz can lead to a situation where the most salient
features of the EEG are effectively deleted. This kind of a mas- TECHNICAL ASPECTS
sive loss of relevant data is clearly evident, for instance, in stan-
dard EEG recordings of the immature human brain and during Recording of a genuine FbEEG signal sets requirements both to
epileptic events. the amplifier and to the electrode–skin interface. It is important
Because of historical reasons, the terminology related to to note that these requirements are not only related to the DC
recording slow EEG events is somewhat confusing. The term stability already mentioned above, but addition of infraslow
direct current EEG (DC-EEG) has been widely used in older lit- and ultrafast frequency ranges introduces new sources of noise
erature to imply a (ideal) frequency response of the EEG with a that are of technical or of biologic origin (e.g., baseline drifts
minimum at 0 Hz. However, the term “DC” has at least two and electrical interference from other devices or skin potential,
connotations, which are misleading. First, it is impossible to respectively). Practical solutions to overcome these problems
have a per definition 0 Hz response, since at 0 Hz the signal level are, however, well established, often trivial, and widely available
is constant and never changing. Second, the use of the term in several modern higher end installations.
DC-EEG puts an a priori emphasis on the low-frequency part Regarding the amplifier, detection of the infraslow events
of the EEG spectrum but, as we will show below, looking at low requires a genuine DC-coupled amplifier with a sufficiently
frequencies does not compromise any simultaneous analyses high input impedance and DC stability as well as a wide-
carried out at fast or even ultrafast frequencies. The results enough dynamic range (preferably ± 100 mV or higher),
described here show that EEG responses, with a time course of whereas detection of the ultrafast frequencies requires a high-
several minutes to achieve their peak levels, can be easily enough sampling rate and a large data storage capacity. Several
recorded if attention is paid to trivial aspects of EEG electrode clinically suitable, DC-coupled amplifiers of this kind have
and amplifier design and characteristics (1–5). recently become commercially available.

741
742 Part V ■ Complementary and Special Techniques

Regarding the electrode–skin interface, detection of the transepithelial potential is left intact under the electrode.
infraslow events requires a DC-stable electrode–skin interface Another compromise that is sometimes encountered is the
(Fig. 36.1). This is achieved by using electrode materials that are use of polarizable (DC unstable) electrodes in combination
nonpolarizable (in practice Ag/AgCl) together with a gel con- with high-pass filtering at an extremely low frequency. While
taining chloride, which is an absolute requirement for obtain- such recordings may be used to detect genuine DC shifts of a
ing a stable electrode potential (3). In addition, electrodes need foreseeable duration, they may equally give spurious results if
to be firmly fixed on the skin to minimize artifacts caused by gel the evident technical pitfalls are ignored. Unlike reversible
leakage or drying. Drying of the gel gives rise to an increase in electrodes that maintain their electrode potential and provide
its electrolyte content, and, for example, a gradual 10% increase a primarily resistive coupling of slow signals across the elec-
in the Cl concentration seen by the Ag/AgCl electrode will trode interface, capacitive signal coupling plays a major role
generate a negative drift of 2.5 mV. Finally, skin-borne signals in polarizable electrodes. Therefore, it is possible that the
(9) caused by electrode movements or galvanic skin reactions low-frequency cutoff is not set by the selected high-pass filter
(GSR/SSR) can be strongly suppressed or eliminated by short- but rather by the circuit formed by the electrode and the
circuiting the epithelium at the recording site (various tech- amplifier input, resulting in unpredictable distortion of slow
niques are available for this purpose, see also Refs. 10, 11). signals. Furthermore, in this kind of recordings slow fluctua-
Sufficient short-circuiting of the skin is painless enough to be tions of the drifting electrode potential (3) will appear as
performed in even sleeping neonates (12). infraslow noise on an apparently stable baseline.
Compromises in recording setup are sometimes necessary, The challenges of interpretation of FbEEG data are mostly
especially under clinical conditions (13,14). For instance, if similar to those of conventional EEG data. Lack of high-pass fil-
the electrode–skin interface is not optimized as described tering in FbEEG changes the visual appearance of slow artifacts
above (Fig 36.1), most slow events are hard if not impossible (e.g., eye and tongue movements) (16–18), but their visual
to be distinguished from noncerebral artifacts. The situation recognition in FbEEG recordings is easy with little experience.
is, of course, different if the subjects are calm and cooperative Median filters and/or spatial filters, for near-complete remov-
(as in research labs) as opposed to recording restless patients ing of many artifacts, are now readily available in pertinent
(as in intensive care units). Our own experience has shown analysis software packages. Skin-borne potentials (“sweat arti-
that a suboptimal electrode–skin interface may still give rea- facts”), in turn, do not cause problems in FbEEG recordings if
sonably good clinical recordings in calm and/or sleeping they are excluded by short-circuiting (see above).
patients (4,5,15) if the EEG events of interest are phasic with
relatively short duration (such as SATs in preterm babies, see
section "Slow Activity in the Preterm Human EEG"). SLOW ACTIVITY IN THE
However, recordings of longer responses (such as ISOs dur- PRETERM HUMAN EEG
ing sleep) or phasic events accompanied with changes in the
autonomic nervous system (such as arousal shifts, which may In FbEEG recordings from preterm neonates the most salient
include GSRs) will no longer be reliable. Movement artifacts frequency range that dominates the total spectral power is
and slow baseline drifts will also be very much larger if the much below the conventional EEG frequency band (Fig. 36.2B)
(12,15,19). This is consistent with previous well-known obser-
vations that an abundance of slow activity is a major feature of
EEG activity in the immature brain (20,21) (see also Chapters 9
and 25). In babies born 2 or more months before term age, the
spectral power peaks at frequencies as low as 0.01–0.1 Hz. It is
obvious that when recorded using conventional EEG, this kind
of activity must be highly attenuated and distorted by filtering.
Hence, only faint echoes of the slow neonatal EEG activity can
Figure 36.1. A schematic drawing illustrating the requirements for be detected at around the lower edge of the conventional
a stable electrode–skin interface. A: A nonpolarizing electrode recording bandwidth.
element (Ag| AgCl) is sealed in an insulating plastic housing that is Most notably, FbEEG showed that the preterm human EEG
filled with chloride-containing electrode gel and tightly attached exhibits very slow (up to 5 seconds) SATs. Furthermore, most
to the skin in order to avoid gel leakage and drying. The epithelium of the faster EEG activity is nested within these slow events,
under the electrode is scratched or punctured in order to short circuit and hence, practically all of the EEG activity of the most
skin-borne potentials. The housing of the electrode may have a immature cortex is associated with SATs (Fig. 36.2A) (12,15).
small opening for applying gel and scratching/ puncturing the skin. This is an extremely interesting finding in itself and in light of
B: A simplified equivalent circuit of the skin where the resistance and animal experiments that have demonstrated that endoge-
potential difference across intact skin epithelium are represented nously driven, spontaneous activity is crucial in shaping neu-
by Re and Ee , and the subepithelial resistance by Rd . Short- ronal connectivity in the developing brain wiring already at an
circuiting the epithelium introduces a low-resistance shunt pathway immature stage where sensory input has little or no role at all
Rsh , which causes the transepithelial potential difference to fall (reviewed in Refs. 19, 22, 23). Moreover, it has been shown that
to Ee (Rsh )>(Re Rsh ) . sensory input to the as yet poorly wired brain is able to trigger
Chapter 36 ■ Infraslow EEG Activity 743

for the identification and diagnostics of activity-dependent dis-


eases and malfunctions of the immature brain as proposed by
Shatz et al. (26).

INFRASLOW EEG OSCILLATIONS


AND VOLTAGE SHIFTS DURING SLEEP
Using FbEEG, ISOs that take place at a wide range of
frequencies (0.02–0.2 Hz) with an amplitude of up to several
tens of microvolts are readily observed during non-REM
sleep (7). Interestingly, the phase of ISO shows a robust corre-
lation with higher frequency EEG activities (Fig. 36.3) as well as
with phasic brain events, such as the K-complex or interictal
epileptiform activity. ISOs recorded by FbEEG in humans may
thus reflect slow cyclic modulation of cortical excitability under
both physiologic and pathophysiologic conditions. This idea
fits well with results obtained in in vivo experiments on
rats (27), rabbits (28), and monkeys (29).
Transient arousals during sleep are associated with a vertex-
positive DC shift with an amplitude that may exceed hundred
microvolts and a duration of several tens of seconds (Fig. 36.4).
A comparable, very slow positive DC shift is also seen during
awakening or change in sleep stage to a more superficial one,
whereas a vertex-negative DC shift takes place when a person
falls asleep or shifts to deeper sleep stages (30).

INFRASLOW EEG OSCILLATIONS


AND PSYCHOPHYSICAL PERFORMANCE
Recently, we addressed the question whether infraslow EEG
oscillations in awake subjects (31) are linked to behavioral
performance. Prominent infraslow EEG fluctuations were
Figure 36.2. Slow activity in the perinatal human cortex. Specimen observed during a somatosensory detection task experiment,
recording of FbEEG activity (at O1 vs. Cz) of a premature neonate at 33 with a log–log linear increase in the power spectrum down to
weeks of conceptional age shows prominent spontaneous activity tran- below 0.01 Hz (Fig. 36.5B) (8). The subject’s ability to detect
sients. FbEEG data (A, lower trace) was high-pass filtered off-line at the stimulus was strongly correlated with the phase but not
0.5 Hz (upper trace) in order to demonstrate the pronounced loss with the amplitude of the infraslow EEG fluctuations in the
(attenuation, distortion) of slow activity. This is also clearly seen in the 0.01 to 0.1 Hz band (Fig. 36.5A). In addition, the phase of the
power spectrum of FbEEG data from the same experiment (B). Note the infraslow EEG was strongly correlated with higher frequency EEG
small fraction of the total EEG power above the conventional cutoff fre- activity. These results and the observation that EEG activity at
quency of 0.5 Hz (dashed line). FFT was obtained from three 3-minute
segments, using 60-second Hanning window. (C) Tactile stimulation
(asterisk) to palm area in a preterm baby (30 weeks conceptional age)
results in very prominent EEG events in the contralateral electrode (C3)
above somatosensory cortex. These events are characterized by a slow-
wave nesting faster activity, and they have striking similarities to some
SAT events generated spontaneously (i.e., without apparent stimuli) in
the same cortical area (19).
Figure 36.3. Infraslow oscillation during sleep. A 2-minute epoch of
self-organized network events of this kind (Fig. 36.2C) FbEEG activity recorded during slow-wave sleep (midfrontal derivation
(reviewed in Ref. 24; see also Ref. 25). against a calculated linked mastoid reference, adult subject). Data are
FbEEG recordings do not only unravel a fully novel type of shown as FbEEG and following band-pass filtering at 0.02–0.2 Hz to visu-
activity in the immature human cortex in the low-frequency alize the prominent infraslow oscillation and its phase locking to activity
bandwidth. In addition, it is obvious that these data call for a at higher frequencies. (Modified from Vanhatalo S, Palva JM, Holmes MD,
revised nomenclature for the perinatal EEG (19,22). Future et al. Infraslow oscillations modulate interictal epileptic activity in the
studies will show whether work of this kind might set the stage human cortex during sleep. Proc Natl Acad Sci USA. 2004;191:5053–5057.)
744 Part V ■ Complementary and Special Techniques

Figure 36.5. Infraslow fluctuation in EEG and cognitive performance


in an awake subject. A specimen FbEEG recording at 0 to 200 Hz dur-
ing a somatosensory detection task experiment in which threshold
level stimuli were delivered at random 1.5- to 4.5-second intervals
(A). Successful detections of stimuli are indicated with vertical lines,
Figure 36.4. Direct current (DC) shifts associated with arousals during and they show pronounced clustering and phase locking to the rising
sleep. Large positive DC shifts are seen at vertex during transient phase of the infraslow EEG fluctuation. The mean power spectrum
arousals. One event is shown on an expanded time scale in the middle of FbEEG data from 10 subjects shows a log–log linear increase in
and lower traces to illustrate how only the faster activity that is associ- power from 5 to 0.005 Hz (B). (Modified from Monto S, Palva S,
ated with the ascending phase of the positive shift can be seen when Voipio J, et al. Very slow EEG fluctuations predict the dynamics of
using a conventional EEG bandwidth. Unpublished observations of stimulus detection and oscillation amplitudes in humans. J Neurosci.
S. Vanhatalo, J.M. Palva, J. Voipio, and K. Kaila. 2008;28: 8268–8272.)

conventional frequencies correlates with <0.1 Hz blood oxy- shifts usually begin within seconds after the initiation of elec-
genation level-dependent (BOLD) signal fluctuations in func- trographic seizure activity, continuing throughout the seizure
tional magnetic resonance imaging (32,33) strongly support the with slow fluctuation of the amplitude that can reach a level of
idea that FbEEG provides a powerful means for the monitoring over 100 V (Fig. 36.6). While the polarity of the ictal DC shifts
of slow fluctuations in the functional state of cerebral networks. has been invariably negative when recording directly on the
cortical surface above the ictal focus, it may vary when record-
SLOW EEG SIGNALS GENERATED ing seizures from the scalp.
Using simultaneous intracranial and FbEEG recordings, we
BY SEIZURE ACTIVITY
found that the onset of the scalp-recorded DC shift can often
Invasive recordings in experimental animals (34–37) and disclose the side (lateralization) of seizure initiation (Fig. 36.6),
humans (38–40) have established that seizures are associated even in cases where other noninvasive studies (such as video-
with very slow EEG responses. Early pioneering work in the EEG and neuroimaging) are equivocal (16, 47). These findings
1960s (41–43) demonstrated that ictal DC shifts may be strongly suggest that in noninvasive recordings of ictal EEG
recorded also from the scalp during generalized seizures, but activity (48), the bandwidth should be extended to the low
technical difficulties have limited noninvasive, ictal recordings of frequencies that are readily seen in FbEEG.
focal seizures until recently (11,16,44). In addition, some more
recent invasive studies using DC-incompatible electrodes (3) NONNEURONAL EEG SIGNALS ASSOCIATED
and long-time constant AC amplifiers have demonstrated vari- WITH CHANGES IN RESPIRATION
able low-frequency fluctuations at the seizure focus (44–46). AND HEMODYNAMICS
Our recent studies (11,16) using FbEEG recordings on
epilepsy patients undergoing presurgical evaluation demon- While it is evident that fast EEG activity has a neuronal origin,
strated that focal seizures are associated with DC shifts that slow EEG signals may arise from a variety of sources, including
confine to the area with seizure activity (Fig. 36.6). The ictal DC both neuronal and nonneuronal generators. Below, we use the

Figure 36.6. Lateralized direct current (DC)-shift during a


seizure. FbEEG recording from scalp during a subclinical par-
tial seizure from an adult patient with temporal lobe epilepsy
reveals an unilateral negative DC shift with a clear spatiotem-
poral correlation with spiking activity. All traces are shown
against a linked Cz + Oz reference. (Modified from Vanhatalo
S, Holmes MD, Tallgren P, et al. Very slow EEG responses lat-
eralize temporal lobe seizures: an evaluation of noninvasive
DC-EEG. Neurology. 2003; 60:1098–1102.)
Chapter 36 ■ Infraslow EEG Activity 745

In light of the available data, it is clear that much more work


is needed to elucidate the relative contributions and interactions
of nonneuronal and neuronal/glial generators to DC shifts seen
in both invasive and noninvasive recordings of brain activity.

SLOW EEG ACTIVITY, COGNITIVE STATES,


AND EVENT-RELATED POTENTIALS
During the last decades, much of the research on low-frequency
Figure 36.7. A large nonneuronal direct current (DC) shift caused by EEG has focused on slow potentials that are associated with var-
hyperventilation. Voluntary hyperventilation gives rise to a progressively ious kinds of cognitive tasks and states (49,55), such as contin-
increasing negative DC shift that has a millivolt-scale amplitude at ver- gent stimulation (contingent negative variation, CNV), motor
tex (mastoid reference, healthy adult subject). The DC shift is closely movements (Bereitschaftspotential), and the orienting para-
paralleled by a fall in end-tidal partial pressure of CO2 (PCO2; continu- digm. Subjects may also learn to deliberately induce slow scalp-
ous capnograph signal). (Modified from Voipio J, Tallgren P, Heinonen recorded potentials, which has been used in attempts to
E, et al. Millivolt-scale DC shifts in the human scalp EEG: evidence for construct functional brain–computer interfaces (56). All these
a nonneuronal generator. J Neurophysiol. 2003;89: 2208–2214.) potentials have a duration of up to several seconds and often an
amplitude in the order of only a few microvolts, hence requir-
ing an FbEEG as well as electrodes with an optimal DC per-
term nonneuronal when referring to generators of EEG signals formance (3) for their accurate recording. In addition, the fact
that do not arise from the brain tissue parenchyma, that is, that nonneuronal EEG signals are readily elicited by changes in
neurons and glial cells (2). respiration patterns (see section above) or tongue movements
In order to study the contribution of nonneuronal potential (18) call for caution in the interpretation of slow EEG signals
shifts to human FbEEG signals, we have used manipulations related to cognitive tasks and preparatory states (2,18).
known to induce pH shifts at the level of the blood–brain bar- In addition to slow EEG responses seen in cognitive studies
rier (BBB) as well as large hemodynamic changes in healthy sub- of the kind referred above, careful analysis of long-latency com-
jects (2,17) (Fig. 36.7). Voluntary hyperventilation for a few ponents (up to ~2500 milliseconds) of sensory-evoked poten-
minutes induced a progressive negative shift with an astonish- tials may possess significant potential for clinical monitoring of
ingly high peak value of up to 2 mV at vertex, whereas hypoven- brain functions. For instance, it has been shown that long-
tilation caused positive shifts (2). As discussed in detail latency components of auditory-evoked potential correlate
elsewhere (2), the high amplitude and duration (several min- nicely with deepening of anesthesia as well as with emergence
utes) of these scalp-recorded signals cannot be explained on the from anesthesia (57), thus raising the possibility that they could
basis of any neuronal generator mechanism, such as a change in be effectively utilized in anesthesia monitoring. In addition,
the state of tonic excitation of apical dendrites in cortical pyram- large slow potential shifts, which may share a number of char-
idal neurons (36,49). Furthermore, bilateral jugular vein com- acteristics with the ISOs described above, have also been
pression, head-up and head-down tilt, and Valsalva and Mueller reported during anesthesia (58).
maneuvers (i.e., exhaling and inhaling against a load, respec-
tively) all resulted in pronounced DC shifts that were highest at OTHER INFRASLOW MECHANISMS
midline derivations and were paralleled by hemodynamic
changes monitored using near-infrared spectroscopy (17). The evidence for the presence of infraslow brain activity does
The results described above are in agreement with early works not only come from direct demonstration of infraslow EEG sig-
(50–52, see also Ref. 53) that have demonstrated large shifts in the nals. Several lines of indirect evidence have recently emerged to
potential of brain tissue that correlate strikingly well with support the idea that an underlying infraslow modulation is a
changes in cerebral blood flow and/or CO2/pH and are generated widespread mechanism in brain functions. Many different phys-
across the BBB. Such nonneuronal changes in the potential of iological and pathological EEG activities exhibit infraslow peri-
brain parenchyma are readily conducted to scalp, which fully odicity in humans (7,8,28,59–61) as well as in animals (29,62).
accounts for the large DC shifts seen in FbEEG recordings (2). In addition, fundamental studies on temporal dynamics of
The fact that slow EEG signals can arise at the level of the human cognition have shown that cognitive performance (such
BBB has a number of implications. For instance, it is easy to as reaction time) fluctuates at infraslow frequencies (63), and,
envisage that epileptic activity, known to cause a large change in strikingly, this fluctuation in performance is tightly linked to the
local blood flow (54), may produce a DC shift (see above) that temporal phase of an underlying, scalp-recorded infraslow EEG
is at least to some extent generated by the BBB in the region activity (8) (Fig. 36.5). These observations are not only theoret-
associated with neuronal hyperactivity. Also, slow signals ically interesting, but they might provide completely novel clin-
related to cognitive paradigms may be prone to “contamina- ical paradigms in diagnostics of patients, such as attention
tion” by BBB-generated potentials that arise in response to deficit disorders, where infraslow mechanisms underlying cog-
uncontrolled changes in respiration and systemic CO2 during nitive processes are disturbed (64). Finally, widespread mecha-
the cognitive tasks (see below) (2). nisms of infraslow brain activity are implicated by the presence
746 Part V ■ Complementary and Special Techniques

of the so-called resting state (or default mode) networks that are 7. Vanhatalo S, Palva JM, Holmes MD, et al. Infraslow oscillations
shown to produce distinct, infraslow activity in magnetic reso- modulate interictal epileptic activity in the human cortex during
nance imaging (32,65,66). sleep. Proc Natl Acad Sci USA. 2004;191:5053–5057.
8. Monto S, Palva S, Voipio J, et al. Very slow EEG fluctuations pre-
dict the dynamics of stimulus detection and oscillation amplitudes
CONCLUSIONS in humans. J Neurosci. 2008;28:8268–8272.
9. Picton TW, Hillyard SA. Cephalic skin potentials in electroence-
It is obvious that FbEEG with its capability to record slow activ- phalography. Electroencephalogr. Clin Neurophysiol. 1972;33:
ity of the human brain has a wide range of potential applica- 419–424.
tions in both basic science and in the clinic. Above, we have 10. Julkunen P, Pääkkönen A, Hukkanen T, et al. Efficient reduction of
shown that FbEEG is mandatory for faithful, nondistorted, and stimulus artifact in TMS-EEG by epithelial short circuiting by
nonattenuated recording of the most salient aspects of EEG mini-punctures Clin Neurophys. 2008;119:475–481.
activity seen under a wide variety of circumstances. These 11. Miller JW, Kim W, Holmes MD, et al. Ictal localization by source
include recordings of immature human cortical activity and analysis of infraslow activity in DC-coupled scalp EEG recordings.
NeuroImage. 2007;35:583–597.
epileptic seizures as well as EEG studies of sleep and of various
12. Vanhatalo S, Tallgren P, Andersson S, et al. DC-EEG unmasks very
kinds of cognitive tasks and states.
slow activity patterns during quiet sleep in preterm infants. Clin
It is likely that the scope of FbEEG measurements will Neurophysiol. 2002;113:1822–1825.
strongly expand in the near future. For instance, animal studies 13. Vanhatalo S, Metsäranta M, Andersson S. High fidelity recording
have shown that large waves of spreading depression take place of brain activity in the extremely preterm babies: feasibility study
in ischemic brain tissue, giving rise to high-amplitude, infra- in the incubator. Clin Neurophysiol. 2008;119:439–445.
slow electrical responses (67). It is likely that such events can be 14. Vanhatalo S, Kaila K. Generation of ‘positive slow waves’ in the
readily measured from the human scalp during early phases of preterm EEG: by the brain or by the EEG setup? Clin Neurophysiol.
ischemia. It will also be of interest to use FbEEG in a reexami- 2008;119:1453–1454.
nation of the hitherto unresolved question whether slow DC 15. Vanhatalo S, Palva M, Andersson S, et al. Slow endogenous activity
shifts related to cortical spreading depression take place during transients and developmental expression of K-Cl cotransporter 2 in
the immature human cortex. Eur J Neurosci. 2005c;22:2799–2804.
migraine attacks in humans (68–71).
16. Vanhatalo S, Holmes MD, Tallgren P, et al. Very slow EEG
The technical problems related to FbEEG recording of slow
responses lateralize temporal lobe seizures: an evaluation of non-
activity can be readily solved, as described above. Hence, it is invasive DC-EEG. Neurology. 2003; 60:1098–1102.
likely that FbEEG will become a standard technique in various 17. Vanhatalo S, Tallgren P, Becker C, et al. Scalp-recorded slow EEG
fields of research including neurophysiology, neurology as well responses generated in response to hemodynamic changes in the
as experimental psychology. human brain. Clin Neurophysiol. 2003;114:1744–1754.
18. Vanhatalo S, Voipio J, Dewaraja A, et al. Topography and elimina-
tion of slow EEG responses related to tongue movements.
ACKNOWLEDGMENTS NeuroImage. 2003;20:1419–1423.
19. Vanhatalo S, Kaila K. Spontaneous and evoked activity in the early
Our research is supported by the Academy of Finland, by the
human brain. In: Lagercrantz H, Hanson M, Evrard P, et al., ed.
Sigrid Jusélius Foundation, by Arvo and Lea Ylppö Foundation,
The Newborn Brain: Neuroscience & Clinical Applications. 2nd ed.
and by European IP “EPICURE” EFP6-037315 (KK). New York: Cambridge University Press; 2009: 229–243.
20. Watanabe K, Hayakawa F, Okumura A. Neonatal EEG: a powerful
REFERENCES tool in the assessment of brain damage in preterm infants. Brain
Dev. 1999;21:361–372.
1. Bauer H, Korunka C, Leodolter M. Technical requirements for 21. Lamblin MD, Andre M, Challamel MJ, et al. Electroencephalography
high-quality scalp DC recordings. Electroencephalogr Clin of the premature and term newborn. Developmental features and
Neurophysiol. 1989;72:545–547. glossary. Neurophysiol Clin. 1999;29:123–219.
2. Voipio J, Tallgren P, Heinonen E, et al. Millivolt-scale DC shifts in 22. Vanhatalo S, Kaila K. Ontogenesis of EEG activity: from phenom-
the human scalp EEG: evidence for a nonneuronal generator. enology to physiology. Semin Fetal Neonatal Med.2006;11:
J Neurophysiol. 2003;89:2208–2214. 471–478.
3. Tallgren P, Vanhatalo S, Kaila K, et al. Evaluation of commercially 23. Sipilä ST, Kaila K. GABAergic Control of CA3-driven network
available electrodes and gels for recording of slow EEG potentials. events in the developing hippocampus. In: Darlison M, ed.
Clin Neurophysiol. 2005;116:799–806. Inhibitory Regulation of Excitatory Neurotransmission. Results
4. Vanhatalo S, Voipio J, Kaila K. Full-Band EEG (FbEEG): an emerg- Probl Cell Differ. Heidelberg: Springer-Verlag; 2008;44:99–121.
ing standard in electroencephalography. Clin Neurophys. 24. Vanhatalo S, Lauronen L. Neonatal SEP: back to bedside with basic
2005;116:1–8. science. Semin Fetal Neonatal Med. 2006; 11:464–470.
5. Vanhatalo S, Voipio J, Kaila K. Full-Band EEG (FbEEG): a new 25. Milh M, Kaminska A, Huon C, et al. Rapid cortical oscillations and
standard in clinical electroencephalography. Clin EEG and early motor activity in premature human neonate. Cereb Cortex.
Neurosci. 2005;36:311–317. 2007;17:1582–1594.
6. Bullock TH, Mcclune MC, Enright JT. Are the electroencephalo- 26. Penn AA, Shatz CJ. Brain waves and brain wiring: the role of
grams mainly rhythmic? Assessment of periodicity in wide-band endogenous and sensory-driven neural activity in development.
time series. Neuroscience. 2003;121:233–252. Pediatr Res. 1999;45:447–458.
Chapter 36 ■ Infraslow EEG Activity 747

27. Penttonen M, Nurminen N, Miettinen R, et al. Ultra-slow oscilla- 49. Birbaumer N, Elbert T, Canavan AG, et al. Slow potentials of the
tion (0.025 Hz) triggers hippocampal afterdischarges in Wistar cerebral cortex and behavior. Physiol Rev. 1990;70:1–41.
rats. Neuroscience. 1999;94:735–743. 50. Besson JM, Woody CD, Aleonard P, et al. Correlations of brain d-
28. Prince DA. Cyclical spike driving in chronically isolated cortex. c shifts with changes in cerebral blood flow. Am J Physiol.
Epilepsia. 1965;6:226–242. 1970;218:284–291.
29. Leopold DA, Murayama Y, Logothetis NK. Very slow activity fluc- 51. Woody CD, Marshall WH, Besson JM, et al. Brain potential shift
tuations in monkey visual cortex: implications for functional brain with respiratory acidosis in the cat and monkey. Am J Physiol.
imaging. Cereb. Cortex. 2003;13:422–433. 1970;218:275–283.
30. Marshall L, Molle M, Fehm HL, et al. Scalp recorded direct current 52. Sorensen E, Olesen J, Rask-Madsen J, et al. The electrical potential
brain potentials during human sleep. Eur J Neurosci. difference and impedance between CSF and blood in unanes-
1998;10:1167–1178. thetized man. Scand J Clin Lab Invest. 1978;38:203–207.
31. Girton DG, Benson KL, Kamiya J. Observation of very slow poten- 53. Tétrault S, Chever O, Sik A, et al. Opening of the blood-brain bar-
tial oscillations in human scalp recordings. Electroencephalogr Clin rier during isoflurane anaesthesia. Eur J Neurosci. 2008;28:
Neurophysiol. 1973;35:561–568. 1330–1341.
32. Mantini D, Perrucci MG, Del Gratta C, et al. Electrophysiological 54. Duncan JS. Imaging and epilepsy. Brain. 1997;120:339–377.
signatures of resting state networks in the human brain. Proc Natl 55. Cui RQ, Huter D, Egkher A, et al. High resolution DC-EEG map-
Acad Sci USA. 2007;104:13170–13175. ping of the Bereitschaftspotential preceding simple or complex
33. Fox MD, Raichle ME. Spontaneous fluctuations in brain activity bimanual sequential finger movement. Exp Brain Res. 2000;
observed with functional magnetic resonance imaging. Nat Rev 134:49–57.
Neurosci. 2007;8:700–711. 56. Hinterberger T, Kubler A, Kaiser J, et al. A brain-computer inter-
34. Gumnit RJ, Takahashi T. Changes in direct current activity during face (BCI) for the locked-in: comparison of different EEG classifi-
experimental focal seizures. Electroencephalogr Clin Neurophysiol. cations for the thought translation device. Clin Neurophysiol.
1965;19:63–74. 2003;114:416–425.
35. Mayanagi Y, Walker AE. DC potentials of temporal lobe seizures in 57. Fitzgerald RD, Lamm C, Oczenski W, et al. Direct current auditory
the monkey. J Neurol. 1975;209:199–215. evoked potentials during wakefulness, anesthesia, and emergence
36. Caspers H, Speckmann E-J, Lehmenkuhler A. DC potentials of the from anesthesia. Anesth Analg. 2001;92:154–160.
cerebral cortex: seizure activity and changes in gas pressures. Rev 58. Roughan JV, Laming PR. Large slow potential shifts occur during
Physiol Biochem Pharmacol. 1987;106:127–178. halothane anaesthesia in gerbils. J Comp Physiol. 1998;182:
37. Nita DA, Vanhatalo S, Lafortune FD, et al. Nonneuronal origin of 839–848.
CO 2-related DC EEG shifts: an in vivo study in the cat. 59. Bullock TH, McClune MC, Achimowicz JZ, et al. Temporal fluctu-
J Neurophysiol. 2004;92:1011–1022. ations in coherence of brain waves. Proc Natl Acad Sci USA.
38. Goldring S. Negative steady potential shifts which lead to seizure 1995;92:11568–11572.
discharge. UCLA Forum Sci. 1963;1:215–236. 60. Parrino L, Halasz P, Tassinari CA, et al. CAP, epilepsy and motor
39. Wieser HG, Elger CE, Stodieck SR. The ‘foramen ovale electrode’: events during sleep: the unifying role of arousal. Sleep Med Rev.
a new recording method for the preoperative evaluation of 2006;10:267–285.
patients suffering from mesio-basal temporal lobe epilepsy. 61. Nir Y, Mukamel R, Dinstein I, et al. Interhemispheric correlations
Electroencephalogr Clin Neurophysiol. 1985;61:314–322. of slow spontaneous neuronal fluctuations revealed in human sen-
40. Bragin A, Claeys P, Vonck K, et al. Analysis of initial slow waves sory cortex. Nature Neurosci. 2008;11:1100–1108.
(ISWs) at the seizure onset in patients with drug resistant tempo- 62. Wichmann T, Kliem MA, Soares J. Slow oscillatory discharge in the
ral lobe epilepsy. Epilepsia. 2007;48:1883–1894. primate basal ganglia. J Neurophysiol. 2002;87:1145–1148.
41. Bates JAV. The unidirectional potential changes in petit mal 63. Gilden DL. Cognitive emissions of 1/f noise. Psychol Rev.
epilepsy. UCLA Forum Sci. 1963;1:237–279. 2001;108:33–56.
42. Cohn R. DC recordings of paroxysmal disorders in man. 64. Castellanos FX, Sonuga-Barke EJ, Scheres A, et al. Varieties of
Electroencephalogr Clin Neurophysiol. 1964;17:17–24. attention-deficit/hyperactivity disorder-related intra-individual
43. Chatrian GE, Somasundaram M, Tassinari CA. DC changes variability. Biol Psychiatry. 2005;57:1416–1423.
recorded transcranially during “typical” three per second spike 65. Fransson P, Skiöld B, Horsch S, et al. Resting-state networks in the
and wave discharges in man. Epilepsia. 1968;9:185–209. infant brain. Proc Natl Acad Sci USA. 2007;104:15531–15536.
44. Ikeda A, Taki W, Kunieda T, et al. Focal ictal direct current shifts in 66. Raichle ME, Snyder AZ. A default mode of brain function: a brief
human epilepsy as studied by subdural and scalp recording. Brain. history of an evolving idea. NeuroImage. 2007;37:1083–1090.
1999;122:827–838. 67. Nedergaard M. Mechanisms of brain damage in focal cerebral
45. Gross DW, Gotman J, Quesney LF, et al. Intracranial EEG with very ischemia. Acta Neurol Scand. 1988;77:81–101.
low frequency activity fails to demonstrate an advantage over con- 68. Lauritzen M. Cortical spreading depression in migraine.
ventional recordings. Epilepsia. 1999;40:891–898. Cephalalgia. 2001;21:757–760.
46. Rodin E, Modur P. Ictal intracranial infraslow EEG activity. Clin 69. Eikermann-Haerter K, Ayata C. Cortical spreading depression and
Neurophysiol. 2008;119:2188–2200. migraine. Curr Neurol Neurosci Rep. 2010;10:167-173.
47. Vanhatalo S, Voipio J, Kaila K, et al. Use of Full-band EEG for non- 70. James MF, Smith JM, Boniface SJ, et al. Cortical spreading depres-
invasive ictal localization. In: Miller JW, Silbergeld DL, eds. sion and migraine: new insights from imaging? Trends Neurosci.
Epilepsy Surgery, Principles and Controversies. New York: Informa 2001;24:266–271.
Healthcare, Taylor & Francis Group; 2006:745–751. 71. Rogawski MA. Common pathophysiologic mechanisms in
48. Lagerlund TD, Gross RA. DC-EEG recording: a paradigm shift in migraine and epilepsy. Arch Neurol. 2008;65:709–714.
seizure localization? Neurology. 2003;60:1062–1063.
CHAPTER

High-Frequency EEG Activity


JEAN GOTMAN, NATHAN E. CRONE 37
INTRODUCTION nous delta activity, and smaller regions can generate synchro-
nous higher frequencies. As very high frequencies are generated
The electroencephalogram (EEG) is traditionally recorded and in a very small volume, they are unlikely to be “seen” from the
clinically interpreted with a low-pass filter set at 60 or 70 Hz. The scalp, simply because of the small angle under which they would
most common EEG activities are divided into the delta, theta, be seen (5). For the same reason (small generator volume), high
alpha, and beta bands, the latter term usually used for activity frequencies usually have low amplitude and the skull attenua-
extending up to ~30 Hz. There has been much interest recently in tion reduces this amplitude further. So it is indeed unlikely that
the gamma band, a term commonly used for activity faster than high frequencies can be seen from the scalp, but it is not because
beta ( 30 Hz). Because of considerable variability in the fre- they are filtered by the skull; it is because of the characteristics of
quency bounds of gamma band activity reported across different their generator. For these reasons (distance from generator to
studies, it may be premature, or even altogether inappropriate to sensor and small amplitude of signal), it may be just as difficult
select a specific frequency range to define it. Nevertheless, there is to record high frequencies with MEG as it is with scalp EEG.
a practical need for terminological consistency in order to facili- In the context of a small generator volume, the size of
tate communication in both scientific discourse and translation recording electrodes becomes a potentially important factor,
to clinical practice. Based on accumulating evidence from studies although the question of electrode size has rarely been
of both physiologic and pathologic responses in gamma frequen- addressed in the field of intracranial EEG (iEEG). If a generator
cies (see below), it may be useful to arbitrarily, but provisionally is on the order of a few hundred microns, two issues can be
define the gamma band as 30 to 70 Hz. It has recently been dis- raised. The first is that of spatial sampling: if such small gener-
covered that frequencies higher than 70 Hz can also be recorded ators are scattered and present in a small number of locations,
and can have both physiologic and pathologic significance. A the likelihood that an electrode would be in their vicinity is low.
variety of terms have been used to describe physiologic EEG This is one extreme of the general problem of poor spatial sam-
activity above 70 Hz, for example, “high-gamma” (1,2) and the pling of intracranial investigations. The second one is that an
chi-band (3). In the context of epilepsy, pathologic patterns at electrode larger than the field generator and in contact with it
high frequencies have also been described, for example: ripples could significantly alter that field, and an electrode much larger
between 100 and 200 Hz (sometimes between 80 and 160 Hz, than the field generator could attenuate it, possibly to the point
sometimes between 100 and 250 Hz), fast ripples between 160, of making it undetectable. It is very difficult to know the size of
200, or 250 Hz and 500 or 600 Hz. These events have other prop- a generator, unless one performs a systematic exploration with
erties besides their frequency, and in general, we think that fre- microelectrodes (6). It is therefore not easy to determine the
quency bands should be defined independently from specific optimal electrode size. Electrodes have usually been designed to
EEG phenomena at a time when much exploration is taking allow electrical stimulation without an excessive current density
place. Rather than using a name, we will therefore refer to bands (7,8); for this purpose, a relatively large contact surface is
by their boundaries (e.g., the 100- to 250-Hz band). important but this requirement may be in conflict with that of
optimizing the recording of high-frequency activity.
The Skull, the Generator Volume, It may also be useful to discuss some terminology issues. In
and the Electrodes the animal experimentation domain, the notion of “local field
It is often said and sometimes written that “the skull filters out potential” is often used. For instance filtering an extracellular
high frequencies.” This would imply that high frequencies, for microelectrode recording such that only activity below 100 Hz,
instance above 60 Hz, would be more attenuated by the skull or below 500 Hz, remains (i.e., removing action potentials), is
than lower frequencies, making it essentially impossible to said to produce the local field potential (LFP). This extracellular
record high frequencies except with intracranial electrodes or potential fluctuation is not easily differentiated from the EEG,
with magnetoencephalography (MEG) that is not sensitive to as recorded locally with a small electrode. Although the term
skull conductivity. In fact the skull does not attenuate any fre- “EEG” is most often associated with relatively large electrodes,
quency until approximately 10,000 Hz. This has been clearly there is nothing in the definition of the EEG that implies elec-
demonstrated in the excellent study of Oostendorp et al. (4). trode size. It is rather defined as potential fluctuations within a
There seems to be, however, an inverse relationship between the certain frequency range. From our point of view, there is no for-
frequency of an EEG rhythm and the volume of brain that gen- mal difference between LFP and EEG, but the terms tend to be
erates it. For instance, very large regions can generate synchro- used in different contexts.
749
750 Part V ■ Complementary and Special Techniques

We will first discuss the high frequencies that are recorded in ing interact with excitatory inputs to pyramidal cells such that
the context of normal function. These have been documented more excited cells fire earlier in the gamma cycle. This recording
in experimental animals as well as in humans in response to of the amplitude of excitatory drive into phase values relative to
specific sensory, motor, or cognitive activities. In humans, some the gamma cycle would provide a more efficient coding mecha-
of these high frequencies can be recorded from the scalp but nism by enabling the readout of amplitude information within
most were investigated in patients with intracerebral electrodes a single gamma cycle without requiring the temporal integration
in the context of the evaluation of the surgical treatment of of firing rates (22).
their medically refractory epilepsy. We will then discuss the
high-frequency activities that have been related to focal Band-Limited Gamma Oscillations
epilepsy, here again investigated in experimental animals and in A common feature of the aforementioned theories relating high-
patients with intracerebral electrodes. frequency EEG (gamma) activity to neural coding and cortical
function is that they all more or less require rhythmic oscilla-
tions of membrane potentials and/or action potentials at rela-
HIGH-FREQUENCY ACTIVITY tively stable, that is, band-limited, gamma frequencies. Although
AND NORMAL CORTICAL FUNCTION synchronous neural activity is not necessarily oscillatory and
temporal coding could conceivably exist without oscillations,
Experimental Background and Significance the explanatory potential of band-limited gamma oscillations
The functional relevance of high-frequency EEG activity in nor- has been compelling (23). Perhaps inspired by this, many inves-
mal brain physiology has been the subject of many investigations tigators have attempted to correlate band-limited gamma oscil-
over the past several decades in both animals and humans and is lations in human EEG with cognitive and perceptual operations
still hotly debated. Some of the earliest studies using EEG recog- in cortex.
nized that cortical activation was associated with the disappear- Early attempts to relate band-limited gamma oscillations to
ance of low-frequency activity (alpha) and the appearance of fast human cortical function were designed to demonstrate task-
activity, though the extent of this fast activity was difficult to specific lateralization of 40-Hz activity in scalp-recorded EEG
ascertain given the technical limitations of existing equipment during long blocks of task performance. Although compensa-
(9–11). Subsequent microelectrode recordings in rabbit olfactory tions had to be made for 40-Hz EMG artifact, these studies did
cortex (12) showed that different odors elicited different spatial show lateralization of 40 Hz to the left hemisphere during ver-
patterns in the amplitude of sniff-triggered gamma oscillations bal tasks (24), and to contralateral central scalp region during
(38 to 80 Hz), suggesting a mechanistic role in stimulus discrim- both simple and complex choice reaction time tasks (25,26).
ination. Similar studies in cat and rat revealed oscillations with a However, like block designs in fMRI experiments, estimates of
similar frequency range (13). A decade later, local field recordings gamma activity in long temporal segments during both resting
in cat visual cortex found that oscillatory neuronal firing in a fre- and activated conditions can be contaminated with extraneous
quency range of 40 to 60 Hz can become synchronized during behavioral fluctuations. Furthermore, this block-design
visual stimulation between spatially separate columns (14), approach does not exploit the fine temporal scale at which
between areas 17 and 18 (15), and even between areas 17 of the gamma activity can change during task performance. For these
two hemispheres (16), and that this synchronization depends on reasons, event-related designs have been used much more
global stimulus properties. Similar activity, ranging up to 35 and extensively when studying gamma responses.
50 Hz was recorded in monkey sensorimotor cortex during vol-
untary hand movements (17,18). In these studies bursts of single Phase-Locked Gamma Responses
unit activity were commonly synchronized with LFP oscillations, Using event-related analyses of MEG, Pantev et al. (27)
suggesting that these oscillations facilitated and/or were facili- observed band-limited gamma responses to tone bursts at 35 to
tated by the synchronization of neuronal firing between spatially 40 Hz. This was consistent with previous reports of 40-Hz clicks
segregated but functionally related neurons. driving large steady-state evoked responses in auditory cortex
Synchronization of neural firing has been hypothesized to (28). Notably, the gamma-band responses (GBRs) observed by
form the basis for temporal coding by which temporary assem- Pantev et al. (27) were obtained by filtering event-related poten-
blies of neurons represent higher-order, or global, stimulus tials (ERPs) produced by averaging raw signal from many indi-
properties (19). This mechanism of temporal coding (20) was vidual trials in the time domain. This procedure reveals
recognized as an exciting potential solution to the binding prob- phase-locked evoked GBR, consisting of gamma components
lem associated with psychophysiologic phenomena such as sen- in the ERP. It is important to distinguish these evoked GBRs
sory segmentation and invariant object recognition. More recent from induced gamma band activity that is time-locked, but not
theoretical refinements have further proposed that subthreshold phase-locked (29) (Fig. 37.1; see the section “Non–Phase-Locked
gamma oscillations, driven by synchronous inhibitory neuron Gamma Responses”).
firing, produce temporal windows for cortical–cortical commu- Phase-locked gamma responses have been investigated in a
nication, whereby coherently oscillating ensembles of neurons variety of experimental contexts. In many cases narrow band-
interact more effectively because their communication windows pass filters have been used to focus on ERP components in and
are open at the same time (21). Another theory proposes that around 40 Hz. This practice is potentially susceptible to arti-
rhythmic gamma oscillations from inhibitory interneuronal fir- facts from filtering ERP impulses containing a wide range of
Chapter 37 ■ High-Frequency EEG Activity 751

Figure 37.1 Signal analysis of phase-locked and non–phase-locked responses using band-pass filtering. Schematic of sig-
nal analyses for one channel of intracranial EEG recorded over dominant (left) superior temporal gyrus (filled circle on
brain) during an auditory speech discrimination task. To minimize the contribution of phase-locked responses to subse-
quent analyses of non–phase-locked responses, the ERP may be subtracted from the raw EEG signal in each individual trial
(depicted in steps A, B, and C; for alternative approaches, see Kalcher et al. (42) and Trautner et al. (69)). In this approach,
the raw signal (A) is averaged across N trials to obtain the event-related potential (ERP, B), and the ERP is then subtracted
from each individual trial (C) prior to band-pass filtering (D, 80 to 100 Hz in this illustration). Band-pass filtered signals
from each individual trial are then squared to obtain power values (E). These band-specific power estimates, which are all
positive, may then be averaged across trials to obtain the power average (F), or submitted to statistical analyses to calcu-
late the % change in poststimulus power from prestimulus baseline power (G). Event-related power increases (F,G) are
also known as induced responses and are dominated by non–phase-locked response components. Note the variability in
the latency and magnitude of these responses across individual trials (E). (From Crone NE, Boatman D, Gordon B, et al.
Induced electrocorticographic gamma activity during auditory perception. Clin Neurophysiol. 2001;112(4):565–582.)

frequencies. Nevertheless, evoked GBRs have been observed in phase-locked to surface EEG wavelets elicited by electrical
both animals and humans during cortical activation in a vari- median nerve stimuli (Fig. 37.3). Because many central
ety of functional–anatomic domains (30–32), and in humans synapses transmit bursts but filter out single spikes, coincident
this class of responses has been used to explore mind–brain spike bursts from thalamocortical relay neurons may drive
relationships (33,34). postsynaptic cortical cells more efficiently than single spikes.
In addition to ERP components in the gamma band, ERP Furthermore, they may also be capable of richer coding because
components have also been observed in even higher frequen- intraburst spike frequency can convey graded information
cies, for example, ultrafast frequencies (400 to 1000 Hz), in ani- about sensory stimuli while bursts of different durations can
mals and humans. For example, both EEG and MEG recordings code different stimulus features.
(Fig. 37.2) have shown that somatosensory evoked potentials
from electrical stimulation of the median nerve contain a brief Non–Phase-Locked Gamma Responses
(10 to 15 msec) burst of ~600-Hz spikelike wavelets, called Averaging electrophysiologic (EEG, MEG, LFP, etc.) responses
sigma bursts (35–37). Sigma bursts temporally overlap the thal- in the time domain extracts phase-locked signal components as
amic P15 component and the N20 primary cortical response ERPs and discards non–phase-locked components as noise. In
and, based on converging evidence from animals and humans, contrast, averaging these responses in the frequency domain
are thought to represent both far-field and near-field potentials focuses on event-related changes in the spectral energy of
generated by highly synchronized population spikes in responses that may have both phase-locked and non–phase-
cuneothalamic and thalamocortical relay cells and a variety of locked components (41). The latter approach is still temporally
cortical neurons. These sigma bursts are diminished during linked to an event across multiple trials, but the result is not
sleep, particularly slow-wave sleep, though the N20 is not (38). limited to phase-locked components, as in ERPs. Different
EEG wavelet bursts at similar frequencies have been observed in methods have been used to recognize and minimize the contri-
animals (39). In awake monkeys (40), for example, a subset of bution of phase-locked components (ERPs) to analyses of
SI units exhibits bursts of spikes and single spikes that are non–phase-locked electrophysiologic responses (42,43).
752 Part V ■ Complementary and Special Techniques

Figure 37.3 Recordings from single units and epidural EEG in S-I of an
awake monkey. Peristimulus time histograms (PSTHs) summed over 15
single units (thick lines, upper right panel) coincide with the middle
part of epidural EEG burst averages (thin lines); middle right panel
depicts the results of high-pass filtering ( 428 Hz) the upper panel
traces. (Modified from Baker SN, Curio G, Lemon RN. EEG oscillations
at 600 Hz are macroscopic markers for cortical spike bursts. J Physiol.
Figure 37.2 Simultaneous magnetoencephalogram/ electroencephalo-
2003;550(pt 2):529–534.)
gram (MEG/ EEG) recordings of somatosensory responses (8000 aver-
ages) after electrical left median nerve stimulation. Somatosensory
evoked potentials (SEPs) (traces A/ B) recorded with a C4 –C3 derivation usually more jitter of these responses (and their corresponding
(negativity at C4 upward). MEG somatically evoked fields (SEF) (traces cognitive processes) at longer latencies. This probably explains
C/ D) were recorded at the N20 field maximum over the lower part of the why ERPs with high-frequency components (e.g., the sigma
right central sulcus (upward deflections: magnetic field lines entering the bursts described above) are usually confined to early latencies.
head). A/ D: Original records (0.5 to 1500 Hz). B/ C: High-pass-filtered On the other hand, ERPs at longer latencies (e.g., P300) usually
versions ( 430 Hz). The thin line at 21 msec demonstrates the peak consist of low-frequency components that are more resistant
latency preservation before versus after digital filtering. The solid line to to jitter (44,45). Because averaging in the frequency domain
the left indicates median nerve electrical stimulation (artifacts clipped). does not require phase-locking, it may be better suited to inves-
The MEG sigma burst (C) is shorter than the simultaneously recorded tigate cortical processing at longer or more variable latencies and
EEG burst (B), which starts earlier and lasts longer. While MEG detects to investigate high-frequency electrophysiologic responses at
activity mainly from tangential generators (e.g., area 3b), early EEG burst longer latencies. It may therefore be useful to conceive of electro-
wavelets correspond to radially oriented thalamocortical afferences, and physiologic responses as having different combinations of
later EEG peaks reflect radially oriented sources in area 1 at the crown of frequencies, latencies, and phase-locking. Nevertheless, the dis-
the postcentral gyrus. (Modified from Curio G. Ultrafast EEG activities. In: tinction between phase-locked and non–phase-locked responses
Niedermeyer E, Lopes da Silva F, eds. Electroencephalography: Basic is often still a practical one. Furthermore, many studies suggest
Principles, Clinical Applications, and Related Fields. 5th ed. Philadelphia, that these different classes of EEG responses may have distinct
PA: Lippincott, Williams & Wilkins; 2005:495–504.) functional response properties (29,46,47).
Although early studies of gamma activity using block
designs, strictly speaking, did relate non–phase-locked gamma
However, the distinction between phase-locked responses activity to human cortical function, one of the first event-
(ERPs) and non–phase-locked increases in signal energy (often related analyses of this activity, that is, on a temporal scale com-
termed “induced” responses) may depend more on the meth- mensurate with task performance, was made by Pfurtscheller et
ods by which they are extracted, than on fundamental differ- al. (48), who demonstrated a lateralized increase in 40-Hz activ-
ences between their generators. ity during self-paced finger movements in three subjects. One
Averaging in the time domain necessarily yields phase-locked of these subjects was later shown to have a somatotopic pattern
responses. However, significant variability (jitter) in the latency of this activity during movements of the tongue, fingers, and
(or phase) of ERPs can distort their appearance in time-averaged toes (49). Similar non–phase-locked gamma activity has also
responses. High-frequency components of electrophysiologic been observed in a variety of experimental contexts, for exam-
responses are more susceptible to latency jitter, and there is ple, during the perception of illusory triangles (50).
Chapter 37 ■ High-Frequency EEG Activity 753

Broadband “High Gamma” Activity and somatotopically specific than the more widespread power
As mentioned above, most noninvasive studies of gamma activity changes in mu (alpha) and beta bands, and its temporal pattern
vis-à-vis normal cortical function, until recently, have focused on was also briefer, corresponding to movement onset.
relatively narrow band-limited gamma frequencies in and around In the largest series to date (22 subjects) of iEEG recordings
40 Hz. These studies were motivated in part by previous animal in sensorimotor cortex, Miller et al. (56) compared the topo-
studies of band-limited gamma oscillations and by the aforemen- graphic patterns of power suppression in low frequencies (8 to
tioned theories of cortical coding that necessitate rhythmic oscil- 32 Hz) versus power augmentation in high gamma frequencies
latory activity at consistent, narrowband gamma frequencies. (76 to 100 Hz) during a variety of motor tasks. As in the previ-
iEEG recordings in patients undergoing epilepsy surgery, however, ously mentioned studies, high gamma responses had a more
have allowed the investigation of physiologic gamma responses focused spatial distribution than did power suppression in low
across a broader range of frequencies than those studied in frequencies. In addition, these responses had a somatotopic
humans by noninvasive methods. These studies have revealed organization corresponding to the movement of different body
event-related, non–phase-locked responses in gamma frequencies parts and corresponded well to the results of electrocortical
higher than those previously studied noninvasively (2). In addi- stimulation mapping. This and other reports (57,82,83) suggest
tion, these “high gamma” responses have had a characteristically that high-frequency responses may be a useful tool for mapping
broadband spectral profile that spans a large range of frequencies. motor function in patients undergoing surgical resections near
The lower and upper frequency boundaries of these responses or within motor cortex.
have been quite variable, but they have most commonly ranged
Language and Auditory Cortices
from ~60 to ~200 Hz, with the majority of event-related energy
changes occurring between 80 and 150 Hz (51,52). Broadband high gamma responses have also been used to map
Across a variety of iEEG studies broadband high gamma language cortex (74,76–79). In one of these studies (Fig. 37.4),
responses have exhibited functional response properties that dis- the spatiotemporal patterns of high gamma responses were com-
tinguish them from both phase-locked responses (ERPs) and pared during tasks with different modalities of input (visual vs.
non–phase-locked responses previously observed with scalp EEG auditory stimuli) and output (spoken vs. signed responses). The
in other frequency bands (2). For example, the temporal and spa- location and timing of high gamma responses during these tasks
tial distributions of these responses are typically more discrete were consistent with the functional neuroanatomy of human lan-
and/or functionally specific for task-related cortical activation guage and with the latencies of verbal and signed responses. In
than other electrophysiologic responses. Furthermore, broad- addition, high gamma responses often, but not always, corre-
band high gamma responses have been demonstrated in a great sponded to the results of electrocortical stimulation mapping.
variety of functional–anatomic domains, including motor cortex The potential clinical utility of high gamma responses for map-
(1,53–61), frontal eye fields (62), somatosensory cortex (63–66), ping language cortex was subsequently studied in 13 patients.
auditory cortex (43,67–69), visual cortex (70–74), olfactory Maps of language cortex based on high gamma responses during
cortex (75), and language cortex (74,76–79). The functional– picture naming were compared with maps of naming and mouth
anatomic ubiquity of these responses suggests that they may movements (responsible for verbal output) from electrocortical
serve as a general electrophysiologic index of cortical processing. stimulation (77). With respect to electrocortical stimulation (the
“gold standard”), the calculated specificity of maps based on high
gamma responses was 84% but their sensitivity was only 43%.
Motor Cortex The relatively good specificity suggested that high gamma
Investigations of broadband high gamma responses in motor responses were potentially useful for constructing a preliminary
cortex have provided many insights into their functional functional map in order to identify cortical sites of lower priority
response properties. In a study using a visually cued motor task for electrocortical stimulation mapping. However, its low sensi-
(1,80) power augmentation was observed from 75 to 100 Hz tivity indicated that it could not yet replace electrocortical stim-
within somatotopically defined regions of sensorimotor cortex ulation even though stimulation carries the added risk of eliciting
contralateral to the cued movement (1) and corresponded well to seizures. These results also raise the question as to whether some
electrocortical stimulation maps of motor function. The tempo- high gamma responses are falling between the cracks of the stan-
ral patterns of these responses were also quite specific, occurring dard 1-cm spaced electrode arrays. Would smaller, more closely
in brief bursts limited to the onset and offset (81) of movement, spaced electrodes capture more of these responses? A previous
with latencies that covaried with the latencies of movement study of intracranial gamma responses suggested an optimal
onset/offset. In another study using self-paced finger and wrist electrode spacing of less than 5 mm (84).
movements, Ohara et al. (53) observed non–phase-locked Intracranial EEG recordings of auditory cortex have also
gamma activity in S1 and M1 extending up to 90 Hz. High demonstrated high gamma responses during tone and speech
gamma activity (60 to 90 Hz) in particular was time-locked to discrimination (43; Fig. 37.5), during an auditory oddball par-
movement onset and was short-lived after it. Pfurtscheller et al. adigm (67), and during an auditory sensory gating (P50) para-
(54) also observed broadband high gamma activity (60 to 90 Hz) digm (69). Broadband high gamma responses induced by
over sensorimotor cortex while subjects performed self-paced acoustic stimuli are concentrated over posterior superior tem-
tongue and finger movements. As in the previous studies, the poral gyrus in a spatial distribution similar to but not identical
topographic pattern of high gamma activity was more discrete with the N100 of the auditory evoked response associated with
754 Part V ■ Complementary and Special Techniques

Figure 37.4 Plots of induced high gamma


(80 to 100 Hz) activity in one electrode over
posterior superior temporal gyrus, during
three language tasks using either spoken or
signed responses. In each plot the x-axis is
time in milliseconds after stimulus onset,
and data points occur every 100 msec.
Event-related high gamma responses
(HGRs, geometric mean percentage change
in poststimulus 80 to 100 Hz power from
baseline) are plotted as thick lines; thin
lines above and below represent 95% con-
fidence intervals. HGRs begin 100 to 200
msec after onset of the auditory stimulus
during word repetition (peaks marked “S”).
In all three language tasks, HGRs occurred
later during spoken responses (peaks
marked “R”), but not during signed
responses. The onset of the late HGRs
covaried with the latencies of the patient’s
verbal responses. (From Crone NE, Hao L,
Hart J Jr, et al. Electrocorticographic gamma
activity during word production in spoken
and sign language. Neurology. 2001;57(11):
2045–2053.)

the onset of acoustic stimuli. In addition, the magnitudes of related power suppression. If this power suppression extends
these responses appear to be correlated with the degree of func- into low gamma frequencies, it may mask power augmentation
tional activation, for example, greater magnitude in dominant in this frequency range. This is a potential pitfall for analyses of
superior temporal gyrus during discrimination of speech stim- responses with narrow band-pass filters at low gamma frequen-
uli than during discrimination of tone stimuli. Responses in a cies, for example, at 40 Hz. Although the lower boundary of
similar frequency range have been observed in microelectrode broadband gamma power increases may extend down into
recordings of auditory cortex in monkeys (85,86). 40-Hz frequencies, the most consistent responses occur
above 60 Hz. The greater reliability of power changes in high
Band-Limited versus Broadband gamma frequencies was recently demonstrated in a study
Gamma Responses using iEEG to classify movements in different body parts (3).
Studies in humans and animals have shown that non–phase- The limited classification accuracy of lower gamma frequencies
locked responses in the traditional 40-Hz gamma band are (30 to 70 Hz) was interpreted to result from an overlap in the
more variable and less sensitive to functional activation of cor- power spectrum of band-limited power suppression at lower
tex than responses in high gamma frequencies (43,86). This frequencies (ranging up to ~50 Hz) and broadband power
may be due to variability in the frequency range of event- increases that included gamma frequencies greater than 70 Hz.
Figure 37.5 Power spectral analyses (PSA) of ECoG recorded during an auditory speech discrimination task. For selected
electrodes (stars) average power spectra during a baseline, or reference, interval ( 750 to 250 msec before stimulus onset)
are compared with average power spectra during auditory stimulation (0 to 500 msec after stimulus onset). For each elec-
trode, the upper plot is produced by subtracting the lower plot of the prestimulus (reference) power spectrum (gray line) from
the lower plot of the poststimulus (active) power spectrum (black line). Negative values in the upper plots indicate power
suppression; positive values indicate power augmentation. At the second site in Subject #1 (lower plots) there is a relative
power increase at 40 Hz during active versus reference intervals. However, at the first site in Subject #1 (upper plots) and the
second site in Subject #2 (lower plots), there is no net increase in ECoG power at 40 Hz. Furthermore, at the first site in Subject
#2 (upper plots) ECoG power is suppressed at 40 Hz. In all four sites, ECoG power is augmented in high gamma frequencies
greater than 80 Hz. This power augmentation is observed over a broad range of frequencies although it is most prominent
between ~ 80 and ~ 150 Hz. Dips and peaks in power spectrum at 60 Hz are from a 60-Hz notch filter (Subject #1) or from
electronic noise and its harmonics (Subject #2), respectively. (From Crone NE, Boatman D, Gordon B, et al. Induced electro-
corticographic gamma activity during auditory perception. Clin Neurophysiol. 2001;112(4):565–582.)

755
756 Part V ■ Complementary and Special Techniques

Broadband high gamma responses recorded with iEEG in PATHOLOGIC HIGH-FREQUENCY ACTIVITY
humans are difficult to reconcile with earlier conceptualiza-
tions of band-limited gamma oscillations and their putative Ripple activity, in the range of 100 to 200 Hz, had been recorded
role in neural computation. Although more recent observa- in the LFPs of hippocampus in normal experimental animals
tions, of higher frequency and broader band oscillations, in and, like the gamma responses discussed above, were considered
animals are extending the frequency range that is quoted for to be normal physiologic activity. It was then found that the hip-
“gamma,” gamma responses are still largely conceptualized as pocampal formation of experimental animal models of epilepsy
band-limited network oscillations. However, for the broad- presented a ripple type of activity, but at an even higher fre-
band responses observed with iEEG to arise from band-limited quency (93,94). These were termed fast ripples (FRs). Ripples
network oscillations, power changes would have to arise from and FRs were also found in the mesial temporal structures of
summation over multiple spatially overlapping neural popula- epileptic patients in whom microelectrodes were inserted (93).
tions or assemblies, each oscillating at different, perhaps over- This discovery started an extensive series of investigations in
lapping or broadly tuned frequencies (1,87). experimental animals and in humans using microelectrodes. It
was then found that low-amplitude activity in the same fre-
Generators of Broadband Gamma Responses quency range (100 to 500 Hz) could also be recorded from EEG
An alternative explanation for broadband high gamma macroelectrodes (95). This was followed by a series of studies
responses is that they are the time–frequency representations of with clinical EEG recordings. We will review separately the
transient responses with a broad range of frequency compo- microelectrode and the macroelectrode studies and then discuss
nents. Intertrial jitter in the latency of these transients presum- the issues in interpreting their similarities and differences. A first
ably renders them invisible when averaged across trials in the review of this topic was written by Rampp and Stefan (96).
time domain. Recent investigations of microelectrode record-
ings from macaque SII cortex during tactile stimulation have High-Frequency Activity from Microelectrodes
observed broadband high gamma responses with spectral pro- Microelectrodes are normally used to record the activity of one
files identical to those recorded in human iEEG (88,89). or a small number of cells (we only discuss here extracellular
Detailed time–frequency analyses of these responses using recordings) in the form of action potentials. This requires a
matching pursuits revealed that they are temporally tightly small electrode tip (usually 40 µ in diameter or smaller) and a
linked to neuronal spikes though their precise generating mech- sampling rate sufficient to identify action potentials (typically
anisms could not be determined (88). In addition, LFP power 20 kHz). Equipment required for such recordings is common in
in the high gamma range was strongly correlated, both in its the experimental laboratory but is not normally used when
temporal profile and in its trial-by-trial variation, with the fir- investigating epileptic patients. Standard EEG equipment can-
ing rate of the recorded neural population (89). not record properly from microelectrodes because of their high
Whether the underlying signals giving rise to broadband impedance compared to EEG electrodes and because it cannot
gamma responses are oscillations or transients, activity in such a deal with such high sampling rates. Microelectrode investiga-
high frequency range is much more likely to be recorded at the tions in humans therefore require a complex setup where the
mesoscale of subdural electrocorticography if there is some EEG is recorded with a standard clinical EEG system at the
degree of synchronization across a population of neural genera- same time that microelectrode activity is recorded with a paral-
tors. In a recent simulation of iEEG responses using different fir- lel system dedicated to this task.
ing patterns in the underlying cortical population, both an In addition to providing action potentials, microelectrodes
increase in firing rate and an increase in neuronal synchrony also record the field potential, or the local EEG. This can be
resulted in broadband high gamma power increases. However, revealed by filtering out action potentials (removing frequencies
these responses were much more sensitive to increases in neu- above approximately 600 Hz). Figure 37.6 shows how the differ-
ronal synchrony than to increases in firing rate (89). Thus, broad- ent components of a microelectrode recording are obtained. The
band gamma response may index neuronal synchronization to knowledge of when action potentials fire is of course important
some extent even if the underlying firing pattern is not a band- in interpreting the high frequencies of the LFP.
limited oscillation. Synchronization across subpopulations of As indicated above, Bragin et al. (93) recorded ripples in the
neurons, as mentioned above, has been hypothesized to consti- frequency range of 80 to 200 Hz and FRs between 200 and
tute a temporal coding strategy for cortical computation that 500 Hz in the hippocampal region of rats in which chronic
complements rate coding and plays a role in higher cortical func- epilepsy was induced with intrahippocampal injection of kianic
tions such as attention (90,91). Several human iEEG studies have acid, and in human hippocampal regions of epileptic patients
found an augmentation of broadband gamma activity in associ- investigated with intracerebral electrodes, through which micro-
ation with attention (52,61,71,73,79,92). Whether these gamma electrodes were inserted (Fig. 37.7). They noted that FRs were
responses reflect an increase in firing rate or an increase in neu- not present in the recordings from kindled animals. The same
ral synchronization, there is ample evidence from the aforemen- group then demonstrated that ripples were the more frequent
tioned studies that they likely reflect patterns of neural activity pattern in the least abnormal temporal lobe (in patients with
that are relevant to cortical computation and may serve as useful electrodes implanted on both sides because of possible bitempo-
markers for cortical function mapping. ral epilepsy), whereas FRs were more frequent on the more
Chapter 37 ■ High-Frequency EEG Activity 757

Figure 37.6 Example of band-pass filter settings used to detect specific


activities recorded from microelectrodes: (A) unfiltered EEG, (B) ripple
frequency (80 to 160 Hz), (C) fast ripple frequency (250 to 500 Hz), and
(D) unit activity (600 Hz to 5 kHz). Marker pulses shown in B and C
indicate trigger points used for waveform averaging. (From Bragin A,
Mody I, Wilson CL, et al. Local generation of fast ripples in epileptic
brain. J Neurosci. 2002;22(5):2012–2021.)

Figure 37.7 A: MRI scan of an electrode track remaining after removal


abnormal side (97). They also showed that FRs are generated of a clinical electrode implanted into the right posterior entorhinal cor-
over a smaller volume than ripples in patients; in animals the FR tex (EC), with a proportionally sized Photoshop image of the electrode
generator appeared to be less than 1 mm 3 (6). In rats, it was superimposed over the track to illustrate the position of a recording
found that the regions generating FRs were stable over time microelectrode within the EC. B: Enlarged Nissl-stained autopsy section
(days and weeks); it was also found that the broader the region of a entorhinal cortex with an orientation similar to the MRI in A, and
in which FRs were found, the more frequent were the seizures, with the same proportionally superimposed electrode image. Arrows
implying that the FRs represented the epileptogenic region and indicate locations of the recording sizes of the clinical electrode. (From
seizures were more frequent if the epileptogenic region was Bragin A, Mody I, Wilson CL, et al. Local generation of fast ripples in
larger. FRs appeared to be generated in multiple discrete clusters epileptic brain. J Neurosci. 2002;22(5):2012–2021.)
of hyperexcitable neurons. Engel et al. (98) concluded that FRs
may be a more efficient marker of epileptogenic tissue than syn-
chronized cellular activity because the field potential of FRs can
be recorded more easily than cellular activity, given the small in which status epilepticus is generated, followed by a latent
fraction of neurons that participate in the synchronous activity period and then spontaneous recurrent seizures. They found that
of epileptic discharges. In a different experimental context if no HFO developed, no seizure developed. They also found that
(ketamine-induced anesthesia in the cat), Grenier et al. (99) the earlier HFOs appeared following the status, the earlier seizures
argued that fast oscillations result from a vicious cycle in which developed, indicating that HFOs appear to reflect epileptogenicity
bursts of population spikes generate a field potential which itself in this model. In a subsequent study of the same model, they
facilitates neuronal firing. attempted to link seizure onset and HFOs, but found that seizures
In humans, it was observed that ripples and FRs were more fre- started with an EEG wave before the first ictal HFO was observed,
quent in non-REM sleep than during wakefulness and REM sleep. concluding that HFOs do not seem to be the immediate trigger of
In addition, REM sleep suppressed ripples but not FRs, compared seizure discharges (103).
to wakefulness (100). In patients with temporal lobe epilepsy,
High-Frequency Activity from Macroelectrodes
Staba et al. (101) found that the ratio of FRs to ripples increased
with increased hippocampal atrophy, thus establishing a direct EEG Activity between 40 and 120 Hz in Epilepsy
link between the generation of FRs and the pathology underlying Activity in the range of 80 to 120 Hz was reported in subdural
mesial temporal lobe epilepsy. In an attempt to try to link directly recordings at the time of electrodecremental seizure onset, and
the generation of high-frequency oscillations (HFOs, including this activity appeared limited to the region of seizure onset (104).
ripples and FRs) and the generation of seizures, Bragin et al. (102) The same year, Allen et al. (105) reported that frequencies around
studied the evolution of HFOs after kianic acid injection, a model 100 Hz might be useful in localizing the seizure onset in frontal
758 Part V ■ Complementary and Special Techniques

Figure 37.8 Left: Ictal EEG of spasms in series in patient with symptomatic West syndrome. Top: A conventional trace.
Bottom: A temporally expanded trace. The time points of A and B (arrowheads) in the conventional trace correspond to
those of a and b (arrowheads) in the expanded trace. Rhythmic gamma activity at around 60 Hz is dominant over the pos-
terior head area and is associated with each spasm in the expanded trace. The power spectra in the right part of the fig-
ure correspond to the 2-second ictal EEG segments, with each segment beginning from each arrow in the conventional
trace. Delt., deltoid muscle. Right: Time evolution of spasm-associated gamma activity in averaged power. Spectral peaks
of the ictal gamma activity are indicated by arrows at around 60 or 70 Hz in the EEG. Activity from the deltoid muscles
has much broader and noisier spectra than does gamma activity. The peaks of gamma activity on the scalp precede those
of muscle activity by about 200 msec. (From Kobayashi K, Oka M, Akiyama T, et al. Very fast rhythmic activity on scalp
EEG associated with epileptic spasms. Epilepsia. 2004;45(5):488–496.) (See color insert)

lobe epilepsy. It took quite a few years until significant studies differentiating it from muscle activity with energy in the same
evaluated further the importance of activity faster than the beta frequency range (Fig. 37.8). Akiyama et al. (110) also showed
band in epileptic patients. activity between 60 and 150 Hz at the onset of spasms, and sug-
There have been several studies relating activity in the 40- to gested that high-frequency activity triggers the spasm.
150-Hz band and different types of epileptic syndromes. Worrell Moving into frequencies above 120 Hz, but in relation to
et al. (106) found that bursts of interictal activity in the 60 to 100 infantile spasms, Ramachandranair et al. (111) demonstrated
Hz range were often found in the intracerebral EEG of patients activity between 150 and 250 Hz at spasm onset, localized to
with neocortical epilepsy, were indicative of the seizure onset frontal or rolandic areas in intracranial recordings. Meanwhile,
region, and became more frequent as seizures were approaching. Akiyama et al. (112) presented a method to facilitate the visual-
Studying the scalp EEG of a large group of children, Wu et al. ization of activity recorded from subdural grid electrodes
(107) found that interictal paroxysmal beta and gamma activity between 60 and 120 Hz at seizure onset. Gardner et al. (113)
was an accurate indicator of epilepsy and of the seizure onset described a method of automatic detection of short bursts of
region, particularly in children younger than 3 years. In young activity between 40 and 100 Hz.
children with hemimegalencephaly, Yamazaki et al. (108) fre-
quently saw activity around 40 to 50 Hz. Kobayashi et al. (109) EEG Activity Higher Than 100 Hz in Epilepsy
showed that gamma activity (50 to 100 Hz) was present at the Given the size of the presumed generators of HFOs, it would
time of infantile spasms in the vast majority of spasms. The seem unlikely that such activity can be recorded from the stan-
power spectrum of this activity showed a clear peak, thus dard macroelectrodes used in the presurgical evaluation of
Chapter 37 ■ High-Frequency EEG Activity 759

Figure 37.9 Discrete high-frequency oscillations identified in the EEG through visual inspection of the digitally filtered
signal. A: Unfiltered ictal EEG at standard timescale. Highlighted sample for further visual analysis in B and C. Ictal EEG
is shown at an expanded time-scale and increased vertical gain with high-pass filtering at 50 and 100 Hz in B and C. A
brief 160- to 210-Hz segmental oscillation is well visualized in the RAH1-2 channel at both filter settings. D: Unfiltered ictal
EEG at standard timescale. A highlighted sample is shown at increased vertical and horizontal gain in parts E and F with
filtering. A discrete 285- to 375-Hz segmental oscillation is clearly visualized in RMH2-3 and to a lesser extent RMH3-4 at
both filter settings. (From Jirsch JD, Urrestarazu E, LeVan P, et al. High-frequency oscillations during human focal seizures.
Brain. 2006;129(pt 6):1593–1608.)

some patients with intractable epilepsy. Electrodes used for The first study demonstrated HFOs during epileptic seizures
subdural recordings are usually disks of 4 to 5 mm 2 with an (95). High-frequency activity, either in the form of brief oscil-
exposed diameter of 2.3 mm, and depth electrodes used for lations or as prolonged discharges, was found most often in the
intracerebral implantation are made of cylinders of approxi- electrodes in which the seizure had started and rarely in regions
mately the same surface area. This size is approximately 4000 to which the seizure had propagated (Fig. 37.9). It was also
times larger than a microelectrode. A group at the Montreal found that patients for whom it was felt that the seizure onset
Neurological Institute uses electrodes made in house with a sur- had been missed (for instance if clinical signs preceded the first
face contact of 0.8 mm 2, somewhat smaller than commercial EEG change), did not have high-frequency activity at seizure
electrodes. Using a recording system that allowed sampling at onset. It was then demonstrated that interictal HFOs were also
2000 Hz after 500 Hz low-pass filtering, they embarked on a relatively frequent in the ripple frequency band, that is, not just
series of studies that demonstrated that high-frequency activity in the FR frequency band (114). These HFOs occurred in three
in the range of 100 to 500 Hz could be recorded with these situations: (i) on top of traditional EEG spikes (as was most
macroelectrodes and appeared to have a strong relationship often the case for FRs recorded with microelectrodes); (ii)
with the seizure-generating region. totally independently of spikes; (iii) as a result of filtering a
760 Part V ■ Complementary and Special Techniques

A FR-Vis-S B FR-NVis-S C FR-Vis


(Fast Ripples visible in spike) (Fast Ripples not visible in spike) (Fast Ripples visible)

Unfiltered

HP 80 Hz

HP 250 Hz

300 µV 300 µV 50 µV

100 msec 100 msec 100 msec

Figure 37.10 Different types of HFOs, with and without spikes. A: Fast ripple visible in spike. B: Fast ripple not visible
in spike. C: Fast ripple visible, independently of a spike. Top: Nonfiltered EEG. Middle: EEG filtered with high-pass filter
of 80 Hz. Bottom: EEG filtered with high-pass filter of 250 Hz. (From Urrestarazu E, Chander R, Dubeau F, et al. Interictal
high-frequency oscillations (100–500 Hz) in the intracerebral EEG of epileptic patients. Brain. 2007;130(pt 9):2354–2366.)

sharp spike (Fig. 37.10). In both the above studies, HFOs were The importance of HFOs was further underlined when it
found in neocortical regions as well as in mesial temporal lobe was found that, in patients with lesions, HFOs were more
structures. In one recent study, Kobayashi et al. (115), recording closely coupled with the region of seizure onset than with the
with a 3000-Hz filter, demonstrated activity up to 900 Hz from lesion, which is sometimes but not always the source of seizures
subdural electrodes at seizure onset, in the form of short oscil- (120). The above studies were performed by measuring HFO
lations superimposed on spikes. rates during non-REM sleep, since it was shown that, as in
It is known that the zone of interictal spiking and the seizure microelectrode recordings, HFOs are most frequent during
onset zone are related to each other, but the relationship is not non-REM sleep but their relative distribution across brain
always tight. By examining the spatial relationship between the regions is not state-dependent (121).
seizure onset zone, the region in which interictal spiking took If there is a good spatial correspondence between the region of
place and the region in which HFOs were found, it could be deter- seizure onset and HFOs, one can naturally wonder if there is also
mined that HFOs had a tighter correspondence with the seizure a temporal coupling. The most natural question is whether HFOs
onset zone than did interictal spikes (116; Fig. 37.11). HFOs there- become more frequent as a seizure approaches. Khosravani et al.
fore appeared to be a better candidate for a biomarker of ictogen- (122) found that HFOs often increased in the few seconds imme-
esis than spikes. These results were largely confirmed for mesial diately preceding a seizure. This study also demonstrated that
and polar structures of the temporal lobe in the study of Crepon HFOs could be recorded from commercially available subdural
et al. (117). In a study that was not directly related to HFOs as dis- electrodes, with a surface contact of 4 mm 2. In a study that exam-
crete entities but rather to activity in the ripple and FR frequency ined fluctuations in HFO rate and energy in the 15-, 5-, and
bands as calculated by spectral analysis, Urrestarazu et al. (118) 1-minute intervals preceding seizure occurrence, Jacobs et al.
showed that high-frequency activity was suppressed during the (123) did not find any systematic change. These two studies
slow wave following spikes, compared to the background. The appear to indicate that if there is a change in HFOs prior to
suppression was most pronounced in the region closest to the seizures, it is only immediately prior to their occurrence and thus
maximum of the spike and was most prominent in the hippocam- it may be difficult to distinguish them from the seizure onset itself.
pus compared to the amygdala or neocortical region. The sup- Looking at HFO fluctuations at the time scale of days in rela-
pression of high-frequency activity was interpreted as reflecting tion to seizure occurrence and to changing antiepileptic medica-
the strength of the inhibition that follows a spike. A statistical tion, Zijlmans et al. (124) demonstrated that medication
approach to the demonstration of this phenomenon and of other reduction results directly in an increase in the rate of HFO occur-
high-frequency changes was presented by Kobayashi et al. (119). rence. Seizures, on the other hand, when they occurred in the
Chapter 37 ■ High-Frequency EEG Activity 761

Figure 37.11 HFOs are most prominent in the seizure onset region. This patient presented with interictal and
ictal signs of bitemporal epilepsy on scalp EEG. He was implanted bitemporally aiming at the amygdala (LA, RA)
and hippocampus (LH, RH). The MRI revealed a malrotation of the right hippocampus. All seizures originated
from the right mesial temporal structures, where the highest rates of HFOs were found. The figure shows three
sections selected from the same few seconds of EEG. In each panel the original EEG is on the left and the thin sec-
tion marked in grey is expanded at the right, after filtering and changing the gain as indicated. The unfiltered EEG
shows frequent spikes in both mesial TL structures. Top panel: Co-occurrence of ripples and fast ripples outside
spikes in channel RH1. Additionally a ripple is seen in channel RH2. Middle panel: The selection includes two
spikes over LA1 and LH1. Both spikes lied outside the seizure onset zone and in the healthier mesial TL. The
expanded sections show no high-frequency oscillation during this time period. Bottom panel: The gray selection
shows two spikes simultaneously at RH1 and RH2 in the seizure onset zone. No oscillation is visible in the spike
at RH1 in the extended unfiltered EEG, while the spike in RH2 shows a very short and small oscillation. The fil-
tered EEG segments clearly reveal a ripple and fast ripple oscillation during these spikes. Spikes looking similar in
the unfiltered EEG might differ in whether they carry high-frequency oscillations. (From Jacobs J, LeVan P,
Chander R, et al. Interictal high-frequency oscillations (80–500 Hz) are an indicator of seizure onset areas inde-
pendent of spikes in the human epileptic brain. Epilepsia. 2008;49(11):1893–1907.)
762 Part V ■ Complementary and Special Techniques

context of stable medications, did not cause any change in HFO patients to contract their jaws and observe the changes directly
occurrence rates. This is in contrast to interictal spikes (125–127), related to the contractions.
for which medication reduction does not result directly in an
increased occurrence rate; seizures, however, are followed by Discussion
increased spiking (this explains the often seen increase in spiking Activity between 40 and 100 Hz is often seen in relation to the
after medication withdrawal, as seizures occur more often in this seizure onset zone in intracranial recordings. Activity in this fre-
context and mediate the increase in spiking). Comparing HFOs quency range can also be seen in the scalp EEG, particularly in
and spikes, one therefore concludes that HFOs behave more like young children and particularly in the context of infantile
seizures than spikes do in the context of changing medication spasms. Frequencies above 100 Hz have not been reported in the
levels. They may therefore be a better marker of disease activity scalp EEG, most likely as a result of a small generator region. In a
than spikes. In another study relating HFOs to seizure occur- recent review, Engel et al. (137) discussed the origin and possible
rence, Zijlmans et al. (128) were not able to replicate in humans meaning of normal and pathologic microelectrode-recorded
the results of Bragin et al. (129) obtained in rats, to the effect that HFOs in the context of mesial temporal lobe epilepsy. They con-
the frequency of seizure occurrence was related to the size of the cluded that ripples appear to reflect synchronized inhibitory
region generating HFOs. postsynaptic potentials and that FRs probably reflect bursts of
Given these results, the question naturally arises regarding the population spikes. It was possible to demonstrate in human
possible use of HFOs in the surgical evaluation of epileptic recordings that interneurons and pyramidal cells fire in sequence,
patients. In a small study, Ochi et al. (130) noted some correla- thus supporting the generation of ripples by inhibitory activity
tion between the removal of regions generating HFOs and suc- (138). Engel et al. (137) also concluded that not all ripples repre-
cessful surgery. In a larger patient group and studying sent normal phenomena since, for instance, ripples are not nor-
systematically the correlation between regions generating HFOs, mally seen in the dentate gyrus but appear there prior to seizures
spikes and the seizure onset, Jacobs et al. (131) showed that the in kianic acid–treated rats (102). Abnormal ripples could be gen-
outcome of surgery was more closely related to whether the erated by slow bursts of population spikes. It is also possible that
HFO-generating region was removed than to whether spike- or not all FRs represent abnormal phenomena.
seizure-generating regions were removed. This suggests that From the studies of HFOs recorded with intracerebral EEG
regions generating HFOs are potential seizure-generating regions. macroelectrodes, one can conclude that HFOs are an important
The question of what is the optimum electrode size to record marker of the epileptogenic region not only in mesial temporal
HFOs remains open. We have seen above that they were well structures, where they are particularly abundant, but also in
recorded with “small” EEG electrodes (0.8 mm 2) but that they neocortical regions. Although activity in the FR band was
could also be recorded with standard electrodes (4 mm 2; see Ref. sometimes more specific than activity in the ripple band, the
122). Worrell et al. (132) performed combined recordings with distinction that was possible with microelectrodes has not fully
standard depth electrodes (contact size 9.4 mm 2) and with carried over with macroelectrodes. There is clearly a relation-
microelectrodes placed a few millimeters away. They concluded ship between the events recorded with microelectrodes and
that both electrode types could record HFOs but that high fre- those recorded with macroelectrodes, but the nature of this
quencies were better recorded with the microelectrodes. Clemens relationship is uncertain. From microelectrode recordings, it
et al. (133) recorded activity up to 150 Hz with foramen ovale would seem that the field of HFOs should be too small to be
electrodes. In preliminary findings by Chatillon et al. (134), it was clearly visible with macroelectrodes. Microelectrode recordings
found, surprisingly, that electrodes having a surface of 0.2 mm 2 are made with a local reference however, and they may therefore
did not record HFOs better than electrodes having a 0.8-mm 2 be blind to broader field changes that take place at the same
surface. The optimum electrode size therefore remains to be time as the very local event that they are best suited to detect.
determined. The properties of these broader events may not be identical to
When looking for HFOs in intracerebral EEG, recorded by those of the very local events. There is a clear need for a system-
depth or subdural recordings, an important word of caution is atic study of the distribution of these HFO fields and of the
necessary: artifacts from contracting scalp muscles can appear electrode sizes most appropriate for recording the most dis-
surprisingly like HFOs (135). Just as EEG activity is transmitted criminating events. Whereas microelectrodes record activity
through the skull and can be recorded on the scalp, activity gen- that is easiest to interpret scientifically because it relates directly
erated outside the skull is measurable in intracerebral record- to the basic building block of the nervous system, that is, the
ings. This has been shown for eye blinks (136). Similarly, EMG neuron, macroelectrodes record potentials that are more diffi-
activity from scalp muscles is transmitted through the skull and cult to interpret in terms of the activity of individual cells.
can be recorded on subdural electrodes and on the most super- However, activity recorded with macroelectrodes may be more
ficial contacts of depth electrodes. Because of skull attenuation significant from the clinical point of view because it represents
and because of the small surface of synchronized activity in what happens to neuronal populations on a larger scale.
muscles, EMG recorded with intracerebral EEG electrodes is of There is a frequent overlap between the spike-generating
small amplitude, just like HFOs. This is a particular concern region and the seizure-generating region, justifying the interest in
during seizures, when scalp muscles can have strong contrac- spikes. The relationship is, however, uncertain, and it is not clear
tions. One way to become aware of this possibility and to how information about spikes should be integrated in the defini-
observe the intracerebral distribution of EMG activity is to ask tion of the epileptogenic region. The investigations of HFOs
Chapter 37 ■ High-Frequency EEG Activity 763

recorded with macroelectrodes suggest that they are more tightly EEG (140), this requires averaging in the frequency domain
coupled to seizures than are spikes, in both time and in space. across many trials of the same time-aligned activations. The tim-
They may therefore be of greater clinical utility. One study (131) ing of epileptogenic HFOs however, like epileptic seizures in gen-
even suggests that HFOs may be a better indicator of epilepto- eral, is inherently unpredictable, and cannot be exploited as
genicity than the seizure onset itself. More studies are required easily. Nevertheless, future investigations will undoubtedly con-
before these results can become clinically applicable. tinue to test the limits of both invasive and noninvasive record-
It is becoming more common now that clinical EEG record- ings of high-frequency EEG activities and to explore their
ing systems allow recordings of activity up to 500 or 600 Hz, but relationships to both normal and pathologic neurophysiologic
these systems are not equipped for microelectrode recordings. mechanisms.
If it is shown conclusively that microelectrode recordings pro-
vide useful clinical information that cannot be obtained from REFERENCES
EEG electrodes, it is possible that future clinical equipment will
allow microelectrode recordings in addition to the EEG. 1. Crone NE, Miglioretti DL, Gordon B, et al. Functional mapping of
An important issue that has not yet been addressed in human sensorimotor cortex with electrocorticographic spectral
intracerebral EEG studies is the differentiation between normal analysis. II. Event-related synchronization in the gamma band.
high-frequency responses related to cognitive activity and Brain. 1998;121(pt 12):2301–2315.
pathologic HFOs related to epilepsy, which may occur in over- 2. Crone NE, Sinai AS, Korzeniewska A. High-frequency gamma
oscillations and human brain mapping with electrocorticography.
lapping frequency ranges. It is unlikely that the epileptogenic
Prog Brain Res. 2006;159:279–302.
HFOs recorded during slow wave sleep, as done in most of the 3. Miller KJ, Shenoy P, den Nijs M, et al. Beyond the gamma band: the
studies reported above, are influenced by cognitive activity. On role of high-frequency features in movement classification. IEEE
the other hand, even though epileptogenic HFOs are more Trans Biomed Eng. 2008;55(5):1634–1637.
common during sleep, they can also occur during wakefulness, 4. Oostendorp TF, Delbeke J, Stegeman DF. The conductivity of the
and thus can potentially contaminate recordings of normal human skull: results of in vivo and in vitro measurements. IEEE
high-frequency responses. A few differences between these elec- Trans Biomed Eng. 2000;47(11):1487–1492.
trophysiologic phenomena, however, may mitigate this con- 5. Gloor P. Neuronal generators and the problem of localization in
cern. First, event-related high gamma responses are usually electroencephalography: application of volume conductor theory
time-locked to a functional task, and although these responses to electroencephalography. J Clin Neurophysiol. 1985;2(4):327–354.
are robust enough to be differentiated in single trials, their sta- 6. Bragin A, Mody I, Wilson CL, et al. Local generation of fast ripples
in epileptic brain. J Neurosci. 2002;22(5):2012–2021.
tistical significance is usually established over a series of trials.
7. McCreery DB, Agnew WF, Yuen TG, et al. Charge density and
It is thus unlikely that epileptogenic HFOs will consistently charge per phase as cofactors in neural injury induced by electri-
occur at the same times. Second, the epileptogenic HFOs cal stimulation. IEEE Trans Biomed Eng. 1990;37(10):996–1001.
observed to date have typically consisted of brief oscillatory, 8. Wei XF, Grill WM. Current density distributions, field distribu-
that is, sinusoidal, wave packets, whereas the most consistent tions and impedance analysis of segmented deep brain stimulation
high-frequency responses during functional activation are electrodes. J Neural Eng. 2005;2(4):139–147.
broadband and thus not really oscillations per se. The utility of 9. Adrian ED, Matthews BHC. The Berger rhythm: potential changes
this distinction, however, will require further testing as investi- from the occipital lobes in man. Brain. 1934;57:355–385.
gations of both phenomena continue. 10. Jasper HH, Andrews HL. Electro-encephalography. III. Normal
Another major issue remains, of course, the limitation of HFO differentiation of occipital and precentral regions in man. Arch
recordings to intracerebral electrodes. Is there a possibility that Neurol Psychiatry. 1938;39:96–115.
11. Ectors L. Étude oscillographique des activités sensitives et motrice
they might be visible noninvasively, from scalp EEG or from
du cortex cérébral chez l’animal éveillé. Comptes rendus des séances
MEG? Given what we know on the one hand about the size of the de la Société de biologie et de ses filiales. 1935;120:1339–1343.
region over which HFOs are synchronous (possibly up to 1 cm) 12. Freeman WJ. Spatial properties of an EEG event in the olfactory bulb
and the amplitude of this activity (10 to 20 V) and, on the other and cortex. Electroencephalogr Clin Neurophysiol. 1978;44:586–605.
hand, about the region of cortex that needs to be involved for a 13. Bressler SL, Freeman WJ. Frequency analysis of olfactory system
spike of several hundred microvolts to be visible on the scalp (a EEG in cat, rabbit, and rat. Electroencephalogr Clin Neurophysiol.
patch of 2.5 to 3 cm 2), it appears unlikely that either EEG or MEG 1980;50:19–24.
can record HFOs. If such recordings were to be possible, they 14. Gray CM, König P, Engel AK, et al. Oscillatory responses in cat
would probably be limited to the most superficial cortical visual cortex exhibit inter-columnar synchronization which
regions. An indication of this difficulty is given by a recent com- reflects global stimulus properties. Nature. 1989;338:334–337.
bined MEG and intracerebral EEG study (139) evaluating 15. Eckhorn R, Bauer R, Jordan W, et al. Coherent oscillations: a mech-
anism of feature linking in the visual cortex? Multiple electrode and
gamma activity evoked by cognitive tasks. This activity was
correlation analyses in the cat. Biol Cybern. 1988;60:121–130.
clearly visible in the intracerebral EEG but was barely measurable 16. Engel AK, Konig P, Kreiter AK, et al. Interhemispheric synchro-
in the MEG. Epileptogenic HFOs may have smaller generators nization of oscillatory neuronal responses in cat visual cortex.
that the gamma activity generated during functional activation Science. 1991;252:1177–1179.
and may thus have even smaller amplitudes at the scalp surface. 17. Murthy VN, Fetz EE. Coherent 25- to 35-Hz oscillations in the
Although high gamma responses during functional activation sensorimotor cortex of awake behaving monkeys. Proc Natl Acad
have been successfully captured with MEG and even with scalp Sci U S A. 1992;89:5670–5674.
764 Part V ■ Complementary and Special Techniques

18. Sanes JN, Donoghue JP. Oscillations in local field potentials of the 40. Baker SN, Curio G, Lemon RN. EEG oscillations at 600 Hz are
primate motor cortex during voluntary movement. Proc Natl Acad macroscopic markers for cortical spike bursts. J Physiol. 2003;
Sci U S A. 1993;90:4470–4474. 550(pt 2):529–534.
19. Singer W, Gray CM. Visual feature integration and the temporal 41. Pfurtscheller G. Quantification of ERD and ERS in the time
correlation hypothesis. Annu Rev Neurosci. 1995;18:555–586. domain. In: Pfurtscheller G, Lopes da Silva FH, eds. Event-
20. Von der Malsburg C. Binding in models of perception and brain Related Desynchronization. 1st ed. Amsterdam: Elsevier; 1999:
function. Curr Opin Neurobiol. 1995;5:520–526. 89–105.
21. Fries P. A mechanism for cognitive dynamics: neuronal communi- 42. Kalcher J, Pfurtscheller G. Discrimination between phase-locked and
cation through neuronal coherence. Trends Cogn Sci. 2005;9(10): non-phase-locked event-related EEG activity. Electroencephalogr Clin
474–480. Neurophysiol. 1995;94:381–384.
22. Fries P, Nikolic D, Singer W. The gamma cycle. Trends Neurosci. 43. Crone NE, Boatman D, Gordon B, et al. Induced electrocortico-
2007;30(7):309–316. graphic gamma activity during auditory perception. Clin
23. Bressler SL. The gamma wave: a cortical information carrier? Neurophysiol. 2001;112(4):565–582.
[news]. Trends Neurosci. 1990;13:161–162. 44. Spencer KM, Polich J. Poststimulus EEG spectral analysis and
24. Spydell JD, Ford MR, Sheer DE. Task dependent cerebral lateraliza- P300: attention, task, and probability. Psychophysiology. 1999;36(2):
tion of the 40 Hertz EEG rhythm. Psychophysiology. 1979;16: 220–232.
347–350. 45. Demiralp T, Ademoglu A, Comerchero M, et al. Wavelet analysis of
25. Sheer DE, Grandstaff NW, Benignus VA. Behavior and 40-c-sec. P3a and P3b. Brain Topogr. 2001;13(4):251–267.
electrical activity in the brain. Psychol Rep. 1966;19:1333–1334. 46. Yordanova J, Kolev V, Polich J. P300 and alpha event-related desyn-
26. DeFrance J, Sheer DE. Focused arousal, 40-Hz EEG, and motor chronization (ERD). Psychophysiology. 2001;38(1):143–152.
programming. In: Giannitrapani D, Murri L, eds. The EEG of 47. Herrmann CS, Knight RT. Mechanisms of human attention:
Mental Activities. New York, NY: Karger; 1988:153–168. event-related potentials and oscillations. Neurosci Biobehav Rev.
27. Pantev C, Makeig S, Hoke M, et al. Human auditory evoked 2001;25(6):465–476.
gamma-band magnetic fields. Proc Natl Acad Sci U S A. 1991;88: 48. Pfurtscheller G, Neuper C, Kalcher J. 40-Hz oscillations during
8996–9000. motor behavior in man. Neurosci Lett. 1993;164:179–182.
28. Galambos R, Makeig S, Talmachoff PJ. A 40-Hz auditory potential 49. Pfurtscheller G, Flotzinger D, Neuper C. Differentiation between
recorded from the human scalp. Proc Natl Acad Sci U S A. 1981;78: finger, toe and tongue movement in man based on 40 Hz EEG.
2643–2647. Electroencephalogr Clin Neurophysiol. 1994;90:456–460.
29. Pantev C. Evoked and induced gamma-band activity of the human 50. Tallon-Baudry C, Bertrand O, Delpuech C, et al. Stimulus specificity
cortex. Brain Topogr. 1995;7(4):321–330. of phase-locked and non-phase-locked 40 Hz visual responses in
30. Franowicz MN, Barth DS. Comparison of evoked potentials and human. J Neurosci. 1996;16(13):4240–4249.
high-frequency (gamma-band) oscillating potentials in rat audi- 51. Ray S, Jouny CC, Crone NE, et al. Human ECoG analysis during
tory cortex. J Neurophysiol. 1995;74(1):96–112. speech perception using matching pursuit: a comparison between
31. Barth DS, MacDonald KD. Thalamic modulation of high-fre- stochastic and dyadic dictionaries. IEEE Trans Biomed Eng. 2003;
quency oscillating potentials in auditory cortex [see comments]. 50(12):1371–1373.
Nature. 1996;383(6595):78–81. 52. Ray S, Niebur E, Hsiao SS, et al. High-frequency gamma activity
32. Basar-Eroglu C, Struber D, Schurmann M, et al. Gamma-band (80–150 Hz) is increased in human cortex during selective atten-
responses in the brain: a short review of psychophysiological tion. Clin Neurophysiol. 2008;119(1):116–133.
correlates and functional significance. Int J Psychophysiol. 1996; 53. Ohara S, Ikeda A, Kunieda T, et al. Movement-related change of
24(1–2):101–112. electrocorticographic activity in human supplementary motor
33. Sheer DE. Focused arousal and the cognitive 40-Hz event-related area proper. Brain. 2000;123(pt 6):1203–1215.
potentials: differential diagnosis of Alzheimer’s disease. Prog Clin 54. Pfurtscheller G, Graimann B, Huggins JE, et al. Spatiotemporal pat-
Biol Res. 1989;317:79–94. terns of beta desynchronization and gamma synchronization in cor-
34. Tiitinen H, Sinkkonen J, Reinikainen K, et al. Selective attention ticographic data during self-paced movement. Clin Neurophysiol.
enhances the auditory 40-Hz transient response in humans. 2003;114(7):1226–1236.
Nature. 1993;364(6432):59–60. 55. Miller KJ, denNijs M, Shenoy P, et al. Real-time functional brain
35. Curio G, Mackert B, Burghoff M, et al. Localization of evoked neu- mapping using electrocorticography. Neuroimage. 2007;37(2):
romagnetic 600 Hz activity in the cerebral somatosensory system. 504–507.
Electroencephalogr Clin Neurophysiol. 1994;91:483–487. 56. Miller KJ, Leuthardt EC, Schalk G, et al. Spectral changes in corti-
36. Curio G. Linking 600-Hz “spikelike” EEG/MEG wavelets (“sigma- cal surface potentials during motor movement. J Neurosci. 2007;
bursts”) to cellular substrates: concepts and caveats. J Clin 27(9):2424–2432.
Neurophysiol. 2000;17(4):377–396. 57. Szurhaj W, Bourriez JL, Kahane P, et al. Intracerebral study of
37. Curio G. Ultrafast EEG activities. In: Niedermeyer E, Lopes da gamma rhythm reactivity in the sensorimotor cortex. Eur J Neurosci.
Silva F, eds. Electroencephalography: Basic Principles, Clinical 2005;21(5):1223–1235.
Applications, and Related Fields. 5th ed. Philadelphia, PA: Lippincott, 58. Leuthardt EC, Schalk G, Wolpow JR, et al. A brain-computer inter-
Williams & Wilkins; 2005:495–504. face using electrocorticographic signals in humans. J Neural Eng.
38. Emerson RG, Sgro JA, Pedley TA, et al. State-dependent changes in 2004;1:63–71.
the N20 component of the median nerve somatosensory evoked 59. Gonzalez SL, de Peralta RG, Thut G, et al. Very high frequency oscil-
potential. Neurology. 1988;38(1):64–68. lations (VHFO) as a predictor of movement intentions. Neuroimage.
39. Jones MS, Barth DS. Spatiotemporal organization of fast ( 200 Hz) 2006;32(1):170–179.
electrical oscillations in rat Vibrissa/Barrel cortex. J Neurophysiol. 60. Gunji A, Ishii R, Chau W, et al. Rhythmic brain activities related to
1999;82(3):1599–1609. singing in humans. Neuroimage. 2007;34(1):426–434.
Chapter 37 ■ High-Frequency EEG Activity 765

61. Brovelli A, Lachaux JP, Kahane P, et al. High gamma frequency the time–frequency plane. J Neurosci Methods. 2005;145(1–2):
oscillatory activity dissociates attention from intention in the 267–276.
human premotor cortex. Neuroimage. 2005;28(1):154–164. 82. Aoki F, Fetz EE, Shupe L, et al. Increased gamma-range activity in
62. Lachaux JP, Hoffinann D, Minotti L, et al. Intracerebral dynamics human sensorimotor cortex during performance of visuomotor
of saccade generation in the human frontal eye field and supple- tasks. Clin Neurophysiol. 1999;110(3):524–537.
mentary eye field. Neuroimage. 2006;30(4):1302–1312. 83. Brunner P, Ritaccio AL, Lynch TM, et al. A practical procedure for
63. Gross J, Schnitzler A, Timmermann L, et al. Gamma oscillations in real-time functional mapping of eloquent cortex using electrocor-
human primary somatosensory cortex reflect pain perception. ticographic signals in humans. Epilepsy Behav. 2009;15(3):278–286.
PLoS Biol. 2007;5(5):1168–1173. 84. Menon V, Freeman WJ, Cutillo BA, et al. Spatio-temporal correlations
64. Ihara A, Hirata M, Yanagihara K, et al. Neuromagnetic gamma- in human gamma band electrocorticograms. Electroencephalogr Clin
band activity in the primary and secondary somatosensory areas. Neurophysiol. 1996;98:89–102.
Neuroreport. 2003;14(2):273–277. 85. Brosch M, Budinger E, Scheich H. Stimulus-related gamma oscil-
65. Hauck M, Lorenz J, Engel AK. Attention to painful stimulation lations in primate auditory cortex. J Neurophysiol. 2002;87(6):
enhances gamma-band activity and synchronization in human 2715–2725.
sensorimotor cortex. J Neurosci. 2007;27(35):9270–9277. 86. Steinschneider M, Fishman YI, Arezzo JC. Spectrotemporal
66. Bauer M, Oostenveld R, Peeters M, et al. Tactile spatial attention analysis of evoked and induced electroencephalographic
enhances gamma-band activity in somatosensory cortex and responses in primary auditory cortex (A1) of the awake monkey.
reduces low-frequency activity in parieto-occipital areas. J Neurosci. Cereb Cortex. 2008;18(3):610–625.
2006;26(2):490–501. 87. Crone NE, Hao L. The functional significance of event-related spec-
67. Edwards E, Soltani M, Deouell LY, et al. High gamma activity in tral changes (ERD/ERS) from the perspective of electrocorticogra-
response to deviant auditory stimuli recorded directly from phy. In: Reisin RC, Nuwer MR, Hallett M, et al., eds. Advances in
human cortex. J Neurophysiol. 2005;94(6):4269–4280. Clinical Neurophysiology (Suppl to Clin Neurophysiol). Amsterdam:
68. Kaiser J, Lutzenberger W. Human gamma-band activity: a window Elsevier Science; 2002:435–442.
to cognitive processing. Neuroreport. 2005;16(3):207–211. 88. Ray S, Hsiao SS, Crone NE, et al. Effect of stimulus intensity on
69. Trautner P, Rosburg T, Dietl T, et al. Sensory gating of auditory the spike-local field potential relationship in the secondary
evoked and induced gamma band activity in intracranial record- somatosensory cortex. J Neurosci. 2008;28(29):7334–7343.
ings. Neuroimage. 2006;32(2):790–798. 89. Ray S, Crone NE, Niebur E, et al. Neural correlates of high-
70. Hoogenboom N, Schoffelen JM, Oostenveld R, et al. Localizing gamma oscillations (60–200 Hz) in macaque local field poten-
human visual gamma-band activity in frequency, time and space. tials and their potential implications in electrocorticography. J
Neuroimage. 2006;29(3):764–773. Neurosci. 2008;28(45):11526–11536.
71. Vidal JR, Chaumon M, O’Regan JK, et al. Visual grouping and the 90. Steinmetz PN, Roy A, Fitzgerald PJ, et al. Attention modulates
focusing of attention induce gamma-band oscillations at different synchronized neuronal firing in primate somatosensory cortex.
frequencies in human magnetoencephalogram signals. J Cogn Nature. 2000;404(6774):187–190.
Neurosci. 2006;18(11):1850–1862. 91. Fries P, Reynolds JH, Rorie AE, et al. Modulation of oscillatory
72. Lachaux JP, George N, Tallon-Baudry C, et al. The many faces of neuronal synchronization by selective visual attention. Science.
the gamma band response to complex visual stimuli. Neuroimage. 2001;291(5508):1560–1563.
2005;25(2):491–501. 92. Jensen O, Kaiser J, Lachaux JP. Human gamma-frequency oscilla-
73. Tallon-Baudry C, Bertrand O, Henaff MA, et al. Attention modulates tions associated with attention and memory. Trends Neurosci.
gamma-band oscillations differently in the human lateral occipital 2007;30(7):317–324.
cortex and fusiform gyrus. Cereb Cortex. 2005;15(5):654–662. 93. Bragin A, Engel J Jr, Wilson CL, et al. Hippocampal and entorhinal
74. Tanji K, Suzuki K, Delorme A, et al. High-frequency gamma-band cortex high-frequency oscillations (100–500 Hz) in human epilep-
activity in the basal temporal cortex during picture-naming and tic brain and in kainic acid-treated rats with chronic seizures.
lexical-decision tasks. J Neurosci. 2005;25(13):3287–3293. Epilepsia. 1999;40(2):127–137.
75. Miyanari A, Kaneoke Y, Ihara A, et al. Neuromagnetic changes of 94. Bragin A, Engel J Jr, Wilson CL, et al. High-frequency oscillations
brain rhythm evoked by intravenous olfactory stimulation in in human brain. Hippocampus. 1999;9(2):137–142.
humans. Brain Topogr. 2006;18(3):189–199. 95. Jirsch JD, Urrestarazu E, LeVan P, et al. High-frequency oscillations
76. Crone NE, Hao L, Hart J Jr, et al. Electrocorticographic gamma during human focal seizures. Brain. 2006;129(pt 6):1593–1608.
activity during word production in spoken and sign language. 96. Rampp S, Stefan H. Fast activity as a surrogate marker of epileptic
Neurology. 2001;57(11):2045–2053. network function? Clin Neurophysiol. 2006;117(10):2111–2117.
77. Sinai A, Bowers CW, Crainiceanu CM, et al. Electrocorticographic 97. Staba RJ, Wilson CL, Bragin A, et al. Quantitative analysis of
high gamma activity versus electrical cortical stimulation mapping high-frequency oscillations (80–500 Hz) recorded in human
of naming. Brain. 2005;128(pt 7):1556–1570. epileptic hippocampus and entorhinal cortex. J Neurophysiol.
78. Mainy N, Jung J, Baciu M, et al. Cortical dynamics of word recog- 2002;88(4):1743–1752.
nition. Hum Brain Mapp. 2008;29(11):1215–1230. 98. Engel J Jr, Wilson C, Bragin A. Advances in understanding the
79. Jung J, Mainy N, Kahane P, et al. The neural bases of attentive read- process of epileptogenesis based on patient material: what can
ing. Hum Brain Mapp. 2008;29(10):1193–1206. the patient tell us? Epilepsia. 2003;44(suppl 12):60–71.
80. Crone NE, Miglioretti DL, Gordon B, et al. Functional mapping of 99. Grenier F, Timofeev I, Steriade M. Neocortical very fast oscilla-
human sensorimotor cortex with electrocorticographic spectral tions (ripples, 80–200 Hz) during seizures: intracellular corre-
analysis. I. Alpha and beta event-related desynchronization. Brain. lates. J Neurophysiol. 2003;89(2):841–852.
1998;121(pt 12):2271–2299. 100. Staba RJ, Wilson CL, Bragin A, et al. High-frequency oscillations
81. Zygierewicz J, Durka PJ, Klekowicz H, et al. Computationally effi- recorded in human medial temporal lobe during sleep. Ann Neurol.
cient approaches to calculating significant ERD/ERS changes in 2004;56(1):108–115.
766 Part V ■ Complementary and Special Techniques

101. Staba RJ, Frighetto L, Behnke EJ, et al. Increased fast ripple to rip- 121. Bagshaw AP, Jacobs J, LeVan P, et al. Effect of sleep stage on interic-
ple ratios correlate with reduced hippocampal volumes and neu- tal high-frequency oscillations recorded from depth macroelec-
ron loss in temporal lobe epilepsy patients. Epilepsia. 2007;48(11): trodes in patients with focal epilepsy. Epilepsia. 2009;50(4):617–628.
2130–2138. 122. Khosravani H, Mehrotra N, Rigby M, et al. Spatial localization and
102. Bragin A, Wilson CL, Almajano J, et al. High-frequency oscilla- time-dependant changes of electrographic high frequency oscilla-
tions after status epilepticus: epileptogenesis and seizure genesis. tions in human temporal lobe epilepsy. Epilepsia. 2009;50(4):
Epilepsia. 2004;45(9):1017–1023. 605–616.
103. Bragin A, Wilson CL, Fields T, et al. Analysis of seizure onset on 123. Jacobs J, Zelmann R, Jirsch J, et al. High frequency oscillations
the basis of wideband EEG recordings. Epilepsia. 2005;46(suppl (80–500 Hz) in the preictal period in patients with focal seizures.
5):59–63. Epilepsia. 2009;50(7):1780–1792.
104. Fisher RS, Webber WR, Lesser RP, et al. High-frequency EEG activ- 124. Zijlmans M, Jacobs J, Zelmann R, et al. High-frequency oscilla-
ity at the start of seizures. J Clin Neurophysiol. 1992;9(3):441–448. tions mirror disease activity in patients with epilepsy. Neurology.
105. Allen PJ, Fish DR, Smith SJ. Very high frequency rhythmic 2009;72(11):979–986.
activity during SEEG suppression in frontal lobe epilepsy. 125. Gotman J, Marciani MG. Electroencephalographic spiking activ-
Electroencephalogr Clin Neurophysiol. 1992;82(2):155–159. ity, drug levels, and seizure occurrence in epileptic patients. Ann
106. Worrell GA, Parish L, Cranstoun SD, et al. High-frequency oscil- Neurol. 1985;17(6):597–603.
lations and seizure generation in neocortical epilepsy. Brain. 126. Gotman J, Koffler DJ. Interictal spiking increases after seizures
2004;127(pt 7):1496–1506. but does not after decrease in medication. Electroencephalogr Clin
107. Wu JY, Koh S, Sankar R, et al. Paroxysmal fast activity: an inter- Neurophysiol. 1989;72(1):7–15.
ictal scalp EEG marker of epileptogenesis in children. Epilepsy 127. Spencer SS, Goncharova, II, Duckrow RB, et al. Interictal spikes
Res. 2008;82(1):99–106. on intracranial recording: behavior, physiology, and implica-
108. Yamazaki M, Chan D, Tovar-Spinoza Z, et al. Interictal epilepto- tions. Epilepsia. 2008;49(11):1881–1892.
genic fast oscillations on neonatal and infantile EEGs in 128. Zijlmans M, Jacobs J, Zelmann R, et al. High frequency oscillations
hemimegalencephaly. Epilepsy Res. 2009;83(2–3):198–206. and seizure frequency in patients with focal epilepsy. Epilepsy Res.
109. Kobayashi K, Oka M, Akiyama T, et al. Very fast rhythmic activity 2009;85(2–3):287–292.
on scalp EEG associated with epileptic spasms. Epilepsia. 2004; 129. Bragin A, Wilson CL, Engel J. Spatial stability over time of brain
45(5):488–496. areas generating fast ripples in the epileptic rat. Epilepsia. 2003;
110. Akiyama T, Otsubo H, Ochi A, et al. Focal cortical high-frequency 44(9):1233–1237.
oscillations trigger epileptic spasms: confirmation by digital video 130. Ochi A, Otsubo H, Donner EJ, et al. Dynamic changes of ictal
subdural EEG. Clin Neurophysiol. 2005;116(12):2819–2825. high-frequency oscillations in neocortical epilepsy: using multi-
111. Ramachandrannair R, Ochi A, Imai K, et al. Epileptic spasms in ple band frequency analysis. Epilepsia. 2007;48(2):286–296.
older pediatric patients: MEG and ictal high-frequency oscilla- 131. Jacobs J, Zijlmans M, Zelmann R, et al. High-frequency elec-
tions suggest focal-onset seizures in a subset of epileptic spasms. troencephalographic oscillations correlate with outcome of
Epilepsy Res. 2008;78(2–3):216–224. epilepsy surgery. Ann Neurol. 2010;67(2):209–220.
112. Akiyama T, Otsubo H, Ochi A, et al. Topographic movie of ictal 132. Worrell GA, Gardner AB, Stead SM, et al. High-frequency
high-frequency oscillations on the brain surface using subdural oscillations in human temporal lobe: simultaneous microwire
EEG in neocortical epilepsy. Epilepsia. 2006;47(11):1953–1957. and clinical macroelectrode recordings. Brain. 2008;131(pt 4):
113. Gardner AB, Worrell GA, Marsh E, et al. Human and automated 928–937.
detection of high-frequency oscillations in clinical intracranial 133. Clemens Z, Molle M, Eross L, et al. Temporal coupling of
EEG recordings. Clin Neurophysiol. 2007;118(5):1134–1143. parahippocampal ripples, sleep spindles and slow oscillations in
114. Urrestarazu E, Chander R, Dubeau F, et al. Interictal high-fre- humans. Brain. 2007;130(pt 11):2868–2878.
quency oscillations (100–500 Hz) in the intracerebral EEG of 134. Chatillon C-E. The Effect of Contact Size on High Frequency
epileptic patients. Brain. 2007;130(pt 9):2354–2366. Oscillation Detection in Human and Rat Intracerebral EEG
115. Kobayashi K, Inoue T, Watanabe Y, et al. Spectral analysis of EEG Recording. Montreal: McGill; 2009.
gamma rhythms associated with tonic seizures in Lennox-Gastaut 135. Otsubo H, Ochi A, Imai K, et al. High-frequency oscillations of
syndrome. Epilepsy Res. 2009;86(1):15–22. ictal muscle activity and epileptogenic discharges on intracranial
116. Jacobs J, LeVan P, Chander R, et al. Interictal high-frequency EEG in a temporal lobe epilepsy patient. Clin Neurophysiol. 2008;
oscillations (80–500 Hz) are an indicator of seizure onset areas 119(4):862–868.
independent of spikes in the human epileptic brain. Epilepsia. 136. Ball T, Kern M, Mutschler I, et al. Signal quality of simultaneously
2008;49(11):1893–1907. recorded invasive and non-invasive EEG. Neuroimage. 2009;
117. Crepon B, Navarro V, Hasboun D, et al. Mapping interictal oscil- 46(3):708–716.
lations greater than 200 Hz recorded with intracranial macro- 137. Engel J Jr, Bragin A, Staba R, et al. High-frequency oscillations:
electrodes in human epilepsy. Brain. 2010;133(pt 1):33–45. what is normal and what is not? Epilepsia. 2009;50(4):598–604.
118. Urrestarazu E, Jirsch JD, LeVan P, et al. High-frequency intracere- 138. Le Van Quyen M, Bragin A, Staba R, et al. Cell type-specific fir-
bral EEG activity (100–500 Hz) following interictal spikes. ing during ripple oscillations in the hippocampal formation of
Epilepsia. 2006;47(9):1465–1476. humans. J Neurosci. 2008;28(24):6104–6110.
119. Kobayashi K, Jacobs J, Gotman J. Detection of changes of high- 139. Dalal SS, Baillet S, Adam C, et al. Simultaneous MEG and intracra-
frequency activity by statistical time-frequency analysis in epilep- nial EEG recordings during attentive reading. Neuroimage.
tic spikes. Clin Neurophysiol. 2009;120(6):1070–1077. 2009;45(4):1289–1304.
120. Jacobs J, Levan P, Chatillon CE, et al. High frequency oscillations 140. Ball T, Demandt E, Mutschler I, et al. Movement related activity
in intracranial EEGs mark epileptogenicity rather than lesion in the high gamma range of the human EEG. Neuroimage. 2008;
type. Brain. 2009;132(pt 4):1022–1037. 41(2):302–310.
CHAPTER
Intraoperative Evoked
Potential Monitoring
ALAN D. LEGATT
38
S
ensory evoked potentials (EPs) are the electrical signals frequently and thus facilitating more rapid notification to the
produced by the nervous system in response to a sensory surgeons should adverse EP changes occur.
stimulus. Auditory, visual, and somatosensory EPs are EPs should only be recorded to unilateral stimulation; if
used as diagnostic tests to noninvasively assess sensory pathways simultaneous bilateral stimulation were employed, the presence
within the central and peripheral nervous system, as well as to of a normal EP to stimulation of one side might prevent recog-
assess patients for diseases of the ear and eye. Somatosensory nition of an abnormal or absent EP to stimulation of the other
evoked potentials (SEPs) and auditory evoked potentials (AEPs) side. Modern EP recording systems permit interleaving of sen-
have also found wide utility in intraoperative monitoring (IOM) sory stimuli (e.g., auditory stimuli are delivered alternately to the
to detect neural compromise during surgery, when the patient is left and to the right ears). The data sweeps are then sorted (after
anesthetized and a conventional neurologic examination cannot artifact rejection) into separate left- and right-sided averages.
be performed. The anesthetic sensitivity of visual EPs limits their Thus, even though left- and right-sided averages are acquired
utility for IOM. simultaneously, simultaneous bilateral stimulation is never actu-
The central motor tracts can also be monitored by stimulating ally delivered.
them rostrally and recording from either muscles or neural struc-
tures caudal to the part of the nervous system that is at risk. By Assessment of IOM Data
analogy to sensory EPs, the signals that are recorded are typically Sensory EP amplitudes vary substantially across different sub-
called motor evoked potentials (MEPs). The brain can be stimu- jects, whereas component latencies are much more consistent.
lated either electrically or magnetically in order to elicit MEPs. As Therefore, interpretation of extraoperative EP studies per-
discussed in Chapter 52, MEPs to magnetic stimulation are diffi- formed for diagnostic purposes is predominantly based on
cult to record consistently in anesthetized patients during sur- latencies (2,3). However, EP component amplitudes on
gery; intraoperative MEP monitoring is therefore typically repeated recordings in the same subject are usually quite consis-
performed using transcranial electrical stimulation (TES) of the tent if the recording techniques are not altered. Moreover, if
brain. parts of the nervous system are compromised, amplitude
The EP modalities commonly used for IOM are described in changes may occur earlier than, or in the absence of, latency
this chapter. For the sensory EPs, the description includes the changes. Therefore, both the amplitudes and latencies should be
anatomical generators of the components that are useful for assessed during IOM of SEPs and AEPs.
IOM. Recording techniques, assessment of the IOM data, For extraoperative sensory EP studies performed for diagnos-
causes of adverse EP changes, and typical intraoperative appli- tic purposes, the patient’s signals are compared to those
cations are described. recorded in a control population of normal subjects. In order for
this comparison to be valid, the techniques used and the state of
the subjects must be identical in the patient’s study and in the
FACTORS COMMON TO MULTIPLE control studies. During IOM, the anesthetic regimen, the body
MODALITIES temperature, the delivered stimulus intensity, and other factors
will differ from patient to patient. Therefore, during IOM each
Recording Techniques patient serves as his or her own control; EPs recorded during the
Sensory EPs are in general too small to be seen in the raw data time when parts of the nervous system are at risk are compared
following a single stimulus. Signal averaging is therefore to those recorded earlier during the same operation (4). If the
employed—multiple stimuli are delivered and the data epochs or anesthetic regimen or other factors that will affect the EP data
sweeps recorded following the individual stimuli are averaged are changed, the baseline amplitude and latency values should be
together, after rejecting those that contain large artifacts. Signal reassessed.
averaging (Fig. 38.1) improves the signal-to-noise ratio of the
averaged EP waveforms by a factor equal to the square root of the Causes of Adverse EP Changes
number of sweeps included in the average (1). If the EP data are Changes in the signals recorded during IOM can be classified
noisy and/or the EP amplitudes are small, the number of sweeps into three categories: “true positive” changes, which reflect
per average may have to be increased. Conversely, if the signal-to- compromise of the structures that the monitoring is intended
noise ratio of the raw data is favorable, the number of sweeps per to safeguard (e.g., Figs. 38.2 and 38.3); changes produced by
average can be decreased, allowing averages to be acquired more other physiologic mechanisms such as anesthetic effects (e.g.,
767
768 Part V ■ Complementary and Special Techniques

Figure 38.1 Averaged brainstem auditory evoked potential


(BAEP) waveforms in an awake human subject, showing the
improvement in the signal-to-noise ratio as the number of
epochs in the average is increased. The BAEPs were elicited
by right ear stimulation with rarefaction clicks delivered at a
rate of 11.3 per second; the number of epochs included in
the average is shown to the left of each waveform. Amplifier
bandpass 150 to 3000 Hz, vertex (Cz) to right mastoid
recording. In this and subsequent BAEP figures, Cz positivity
is plotted as an upward deflection.

Figs. 38.4 and 38.5) or hypothermia; and changes due to tech- changes in neural function but are different from the neural
nical problems (e.g., Fig. 38.6) rather than to actual changes in dysfunction due to surgical manipulations that the IOM was
neuronal function (5). True positive changes can reflect tissue specifically intended to detect. Irrigation of the surgical field
damage due to direct mechanical or thermal injury. They can with cold fluids can produce localized tissue hypothermia,
also reflect ischemia caused by compromise of the blood supply causing EP changes that resemble those that would be caused by
to the neural tissue (Figs. 38.2 and 38.3), by vasospasm, by gen- surgical compromise of the neural tissue there (Fig. 38.8).
eralized hypotension, or by a combination of these acting syn-
ergistically. AUDITORY EVOKED POTENTIALS
EPs can be lost due to technical problems such as equipment
malfunction, dislodged electrodes or stimulators (Fig. 38.6), dis- The earliest electrical signals recorded following a transient
connected or broken wires, and operator error (the use of incor- auditory stimulus are generated within the cochlea and are
rect protocols or settings). A variety of artifacts can obscure the recorded as part of the electrocochleogram. The subsequent
EPs (Fig. 38.7). The artifact from the cavitational ultrasonic suc- neurally generated signals have been divided into latency
tion aspirator (CUSA) device (Fig. 38.7C) deserves special men- classes—short- latency AEPs, with latencies of under 10 msec;
tion, as it may manifest as a low-voltage, high-frequency artifact long- latency AEPs, with latencies over 50 msec; and middle-
that obscures the EP but does not trigger automatic artifact latency AEPs, with intermediate latencies. The long- latency
rejection (4). Monopolar cautery use can saturate the EP record- AEPs are generated within cerebral cortex, including cortical
ing system’s input amplifiers, preventing recording of valid EP association areas, and are profoundly affected by the degree to
data for some time after the cautery is stopped. which the subject is attending to the auditory stimulus and
Anesthesia (Figs. 38.4 and 38.5), systemic hypothermia, and extracting information from it. The long- latency AEPs are sup-
hypotension can produce EP changes that do in fact reflect pressed by surgical anesthesia, and thus are not useful for IOM.
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 769

Figure 38.2 Consecutive SEP runs recorded to left median nerve stimulation during a right carotid endarterectomy in
a 78-year-old man. There was a delayed attenuation of the cortical SEPs (left) following carotid cross-clamping. The
surgeons were notified and placed a shunt; the SEPs then returned to baseline. The patient was unchanged neurolog-
ically after the surgery. The simultaneously recorded cervicomedullary SEPs (right) did not change, demonstrating that
the left median nerve was still being stimulated and that afferent somatosensory activity was still reaching the level of
the medulla. Vertical calibration: 3 µV/ division. Horizontal calibration: 5 msec/ division. In this and subsequent SEP
figures, negativity at input 1 is plotted as an upward deflection.

Middle- latency AEPs are also generated within cerebral cortex, Components, Generators, and Recording
including primary auditory cortex and surrounding areas (6). Electrode Locations
They are also markedly affected by surgical anesthesia, which BAEP components are typically recorded between the vertex
limits their utility for IOM to detect surgery- related focal (position “Cz” of the International 10-20 System) and both ear-
neurologic dysfunction. However, this anesthetic sensitivity has lobes or both mastoids (labeled “Ai” or “Mi” for the ipsilateral
led to attempts to use middle- latency AEPs as an indicator of ear and mastoid, respectively, and “Ac” or “Mc” for those con-
the depth of anesthesia (7). tralateral to the stimulated ear); recording the two channels pro-
The short-latency AEPs are the most useful AEPs for neuro- vides a measure of redundancy in case one of the ear/mastoid
logic and otologic diagnosis because they are relatively easy to electrodes becomes unusable during surgery and may also help in
record and their waveforms and latencies are highly consistent the identification of wave V (see next paragraph). Positive BAEP
across normal subjects (8). They are also useful for IOM because peaks are most often displayed as upward deflections (the oppo-
surgical anesthesia produces only minor changes in them site polarity convention from SEPs and from electroencephalog-
(5,9,10); some of the intraoperative changes that are observed in raphy and electromyography, in which negativity is displayed as
them may be due more to changes in body temperature than to an upward deflection), and the positive peaks in the Cz-Ai record-
anesthetic effects (11–13). Although the earliest components are ing channel are typically labeled with Roman numerals according
generated, at least in part, in the eighth nerve, most of the com- to the convention of Jewett and Williston (14) (Fig. 38.9). Most of
ponents are generated in the parts of the brainstem auditory the BAEP components are recorded from the scalp as far-field
pathways, and the short-latency AEPs are most commonly potentials, which means that small displacements of the record-
referred to as brainstem auditory evoked potentials (BAEPs). ing electrodes do not significantly alter the BAEP waveform.
770 Part V ■ Complementary and Special Techniques

Figure 38.3 BAEP and SEP changes due to brainstem ischemia and infarction during surgery for a basilar artery
aneurysm in a 50-year-old woman. Cortical SEPs to stimulation of both median nerves and BAEPs to left ear stimulation
are shown. The aneurysm ruptured during an attempt to place a clip on it and the cortical SEP to left median nerve
stimulation disappeared. A clip was placed on the basilar artery to control the bleeding and the cortical SEP to right
median nerve then disappeared as well. A BAEP run was not obtained between the aneurysm rupture and clipping of
the basilar artery, but following the clipping BAEP wave V (arrow) became delayed and attenuated, then only question-
ably present (arrow with question mark), and eventually disappeared, while BAEP wave IV (triangle) persisted. The
cortical SEP to right median nerve stimulation reappeared and returned to baseline; the cortical SEP to left median nerve
stimulation recovered only partially. The patient suffered multiple brainstem infarcts. In the SEP waveforms, cortical
negativity is shown as an upward deflection. The first 8 msec of each SEP waveform was cropped off to remove the
large stimulus artifacts. (Reprinted from Legatt AD. Mechanisms of intraoperative brainstem auditory evoked potential
changes. J Clin Neurophysiol. 2002;19:396–408.)

Thus, if a CPz electrode is being placed to record lower limb wave V is typically a little longer (see Fig. 38.9). The increased
SEPs, it can be used in place of the Cz electrode to record BAEPs, separation of waves IV and V in the Cz- Ac channel may make
with minimal effect on the waveforms obtained. Wave I, how- wave V easier to identify in that channel.
ever, is recorded as a near-field potential on the surface of the Wave I originates in the first volley of action potentials in the
head in the vicinity of the stimulated ear. auditory nerve as it begins in the most distal portion of the nerve
Waves I, III, and V are the most consistent BAEP peaks, and (16). It represents the same electrical phenomenon as the N1
those upon which interpretation of both extraoperative BAEP component of the eighth nerve compound action potential in
studies and intraoperative BAEP monitoring is based. Wave I is the electrocohleogram (17), and at the skin surface, it appears as
recorded as a negativity by the ipsilateral ear/mastoid electrode a negativity in a circumscribed area around the stimulated ear
and is largely absent in the Cz-Ac recording (see Fig. 38.9). (18,19). The negativity picked up by the Ai electrode gives rise to
Wave III is typically seen in both channels, though it is smaller the upgoing (positive) peak in the Cz-Ai recording. Since wave I
in Cz- Ac than in the Cz- Ai recording. Waves IV and V are seen is recorded as a near-field potential around the stimulated ear,
in both channels at similar amplitudes and typically overlap, repositioning of the Ai/Mi recording electrode can substantially
forming a IV–V complex with variable morphology (15) alter it, and alternate electrode positions can be used to obtain a
(Fig. 38.10). In the Cz- Ac channel, the latency of wave IV is typ- clearer wave I. An electrode within the external auditory canal
ically a little shorter than that in the Cz-Ai channel, and that of may pick up an even larger wave I. Because wave I arises from the
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 771

Figure 38.4 Cortical and cervicomedullary SEPs to left median nerve stimulation recorded (simultaneously) during a
left carotid endarterectomy in a 65-year-old man. There were no SEP changes with carotid clamping, but the cortical
SEPs became markedly attenuated after nitrous oxide was turned on during closing. Volume-conducted cortical SEPs
visible in the cervicomedullary SEP waveforms (asterisk) also became attenuated, but the cervicomedullary SEP com-
ponents themselves (arrow) were unchanged. Vertical calibration: 1.5 µV/ division for the cortical SEP waveforms and
0.7 µV/ division for the cervicomedullary SEP waveforms. Horizontal calibration: 5 msec/ division.

most distal portion of the auditory nerve, it may persist after the nerve and to the cochlear nucleus. However, this occurs simulta-
nerve is sectioned at a more proximal location, such as during neously with the onset of the second auditory nerve volley (the N2
surgery for eighth nerve tumors (20,21) (Fig. 38.11). component of the eighth nerve compound action potential) in the
Subsequent BAEP components are the composites of contri- distal nerve (17). The latter contributes to the scalp-recorded
butions of multiple generators. The complexity of the generators BAEP in the same manner as the N1 component did when it was
of human BAEPs derives in part from the anatomy of the brain- at the same location. This can cause persistence of a wave II in
stem auditory pathways, with ascending fibers both synapsing in cases where the proximal eighth nerve has been destroyed (8).
and bypassing various relay nuclei (22–24). It also reflects the Wave III predominantly originates in the caudal pontine
presence of at least two bursts of activity in the auditory nerve tegmentum, including the region of the superior olivary com-
(corresponding to the N1 and N2 components of the eighth plex (8). Since ascending projections from the cochlear nucleus
nerve compound action potentials in the electrocochleogram), are bilateral, wave III may receive contributions from brainstem
which can drive the more rostral pathways. Because of both of auditory structures both ipsilateral and contralateral to the
these factors, several different structures within the infratentor- stimulated ear. In patients with asymmetrical lesions of the
ial auditory pathways may be active simultaneously, and thus brainstem, wave III abnormalities are usually most pronounced
contribute to the same BAEP component (16). following stimulation of the ear ipsilateral to the lesion (25–27),
Wave II originates, in part, in the first auditory nerve volley though occasionally they are more pronounced following con-
(the N1 component of the eighth nerve compound action poten- tralateral stimulation (28).
tial, which gave rise to wave I when it began in the distal eighth The anatomical generators of waves IV and V are most likely
nerve) that has propagated to the proximal end of the auditory in close anatomical proximity, since they are usually either both
772 Part V ■ Complementary and Special Techniques

Figure 38.5 Consecutive MEP runs recorded from right leg muscles to transcranial electrical stimulation with the
anode on the left in a 19-year-old man having spinal instrumentation and fusion surgery for Scheuermann’s kyphosis.
During closing, the upper two pairs of MEP waveforms were recorded under total intravenous anesthesia using propofol
and sufentanil. The anesthetic regimen was then changed to 2% sevoflurane. The MEPs became attenuated and then
disappeared.

affected or both unaffected by brainstem lesions (28–30). They studies typically employ a single click polarity. Stimulation with
may, however, be differentially affected (16,28,31), including by alternating click polarities and averaging the responses to rar-
intraoperative brainstem damage (Fig. 38.12). Wave IV appears efaction and compression clicks together is useful to cancel a
to reflect activity predominantly in ascending auditory fibers large stimulus artifact and/or the cochlear microphonic and is
within the dorsal and rostral pons, just caudal to the inferior often employed during intraoperative BAEP monitoring.
colliculus, whereas wave V predominantly reflects activity at the Since headphones would be impractical for IOM, the
level of the inferior colliculus, perhaps including activity in the acoustic stimuli are typically delivered using ear inserts, incor-
rostral portion of the lateral lemniscus as it terminates in the porating foam cylinders that can be compressed and then grad-
inferior colliculus (8). As is the case with wave III, wave V ually expand to achieve a tight fit with the ear canal. The foam
abnormalities due to unilateral brainstem lesions are usually can be covered with a thin layer of metal foil, to serve as a near-
most pronounced following stimulation of the ear ipsilateral to field electrode for recording of wave I from the external audi-
the lesion (25–27,32,33), though there are exceptions (34,35). tory canal. The ear insert can be held in place with bone wax or
Waves VI and VII are absent in some normal subjects. While cotton padding, and secured with nonporous tape or an adhe-
they may in part reflect activity in more rostral structures such sive waterproof dressing pad (4,38). This is to prevent fluids
as the medial geniculate nucleus, they also receive contributions from entering the ear canal, where they might interfere with
from activity in the inferior colliculus (8); the latter generator transmission of sound to the inner ear. The ear insert is con-
may cause persistence of these waves in patients with auditory nected to the acoustic transducer using a segment of plastic
pathway damage rostral to the inferior colliculus. Therefore, tubing. Care should be taken to ensure that this tubing remains
BAEPs cannot be used to assess or monitor the auditory path- in place during positioning of the patient and is not kinked. The
ways rostral to the mesencephalon. time required for the acoustic signal to propagate through the
tubing typically prolongs the latencies of all BAEP components
Recording Techniques by approximately 0.9 to 1.0 msec as compared to those recorded
Acoustic clicks, produced by delivering trains of 100- µsec- using headphones. This causes no problems in the evaluation of
duration electrical square pulses to the acoustic transducer, are the BAEPs, since each patient serves as his or her own control
most often used to elicit BAEPs for IOM; brief tone pips can and the acoustic propagation delay is constant. Moreover, the
also be used. Since the responses to rarefaction and compres- delay helps to prevent obscuration of wave I by the electrical
sion clicks may differ (36,37), extraoperative diagnostic BAEP stimulus artifact, because (i) it prolongs the latency of wave I,
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 773

Figure 38.6 Loss of BAEPs due to a technical problem in a patient


undergoing surgery for a left-sided vestibular schwannoma. Consecutive
BAEP recordings (Cz-Ai recording) to right ear stimulation are shown,
Figure 38.7 Single (unaveraged) data epochs recorded during intraop-
with the earliest waveform at the top. During positioning of the patient’s
erative BAEP monitoring. A: An electrical stimulus artifact is present at
head, these BAEPs disappeared. Investigation revealed that the tube car-
the beginning of the epoch. The BAEP, which is less than 1 µV in ampli-
rying the acoustic stimulus had been pulled out of the patient’s ear. The
tude, is not visible in this single epoch of raw data. B: Bipolar cautery
tube was replaced and the BAEPs reappeared. The arrow indicates a pos-
produces a large artifact that triggers automatic artifact rejection. C: The
sible delayed BAEP peak when the tube was dislodged, reflecting
cavitational ultrasonic surgical aspirator (CUSA) device produces a very
acoustic stimulus reaching the ear at a reduced intensity. (Reprinted from
high-frequency but low-voltage artifact that completely obliterates the
Legatt AD. Mechanisms of intraoperative brainstem auditory evoked
neurophysiologic data but does not trigger automatic artifact rejection.
potential changes. J Clin Neurophysiol. 2002;19:396–408.)
D: The light source of the operating microscope produces a repetitive,
sharply contoured artifact that recurs at a harmonic of the line frequency
helping to separate it in time from the electrical stimulus arti- but is composed of higher frequencies that would not be removed by a
fact (which remains simultaneous with the activation of the line-frequency notch filter. In A, B, and C, the horizontal dotted lines
acoustic transducer), and (ii) it permits increasing the distance show the input window of the analog-to-digital converter and the thres -
between the acoustic transducer and the recording electrodes, hold for automatic artifact rejection. In D, the amplifier gain was
reducing the amplitude of the electrical stimulus artifact. reduced to show the light source artifacts in their entirety. Voltage cali-
The stimulus intensity delivered during intraoperative BAEP bration bar: 10 µV in A, B, and C, and 40 µV in D. (Reprinted from Legatt
monitoring cannot be precisely controlled, due to variability in AD. Mechanisms of intraoperative brainstem auditory evoked potential
the positioning of the ear insert, but this does not cause a prob- changes. J Clin Neurophysiol. 2002;19:396–408.)
lem since each patient serves as his or her own control. The
intensity setting chosen should be loud enough to produce a problem because acoustic crosstalk is much smaller with ear-
clear BAEP but not loud enough to cause ear damage. A stimu- insert transducers than with headphones (39). Also, the major
lus rate of approximately 10 per second is typical, but a rate of reason for white noise masking is to prevent acoustic crosstalk
exactly 10 Hz or another submultiple of the power line fre- from producing a BAEP when a deaf ear is stimulated during a
quency should be avoided; if such a submultiple were chosen, diagnostic BAEP study; absence of white noise masking will not
signal averaging would not eliminate line- frequency artifact prevent recognition of new auditory pathway compromise
that is present in the data. Contralateral white noise masking, when a functioning ear is stimulated.
such as is typically used during extraoperative diagnostic BAEP Typical filter settings for BAEP recordings are 100 to 150 Hz
studies to prevent acoustic crosstalk, cannot be used during for the low-frequency (high- pass) filter and 3000 Hz for the
IOM if left and right ear stimuli are interleaved. This is not a high- frequency (low-pass) filter. Line- frequency (e.g., 60 Hz)
774 Part V ■ Complementary and Special Techniques

Figure 38.8 BAEPs to right ear


stimulation recorded during surgery
for a right cerebellopontine angle
meningioma. The BAEPs were stable
during cerebellar retraction and
tumor resection. After the tumor had
been removed, copious irrigation of
the surgical field with cold fluids
produced a transient prolongation of
the I-to-III interpeak interval, reflect-
ing slowed neural conduction within
the eighth nerve resulting from
local cooling. The peak latencies of
waves I, III, and V are marked by
the small diamonds, and the clock
times, esophageal temperatures,
and surgical procedures correspon-
ding to each of the BAEP waveforms
are noted at the right. (Reprinted
from Legatt AD. Mechanisms of
intraoperative brainstem auditory
evoked potential changes. J Clin
Neurophysiol. 2002;19:396–408.)

“notch” filters can be used for BAEP recordings. The analog epoch duration, typically 15 msec, should be used during intra-
gain depends on the input window of the analog- to- digital con- operative BAEP monitoring because component latencies can be
verter; a value of approximately 100,000 is typical. prolonged by preexisting pathology, hypothermia, or intraoper-
While an averaging epoch duration of 10 msec is often used ative compromise of the auditory system. The choice of the
for extraoperative diagnostic BAEP recordings in adults, a longer number of sweeps per average will depend on the signal-to-
noise ratio of the raw data. A value of 1000 to 2000 sweeps per
average is typical.
If the risk is to the eighth nerve (e.g., during surgery for an
intracanalicular eighth nerve tumor) and the surgical exposure
permits placement of a recording electrode on the proximal
eighth nerve, a near- field eighth nerve compound action poten-
tial that is typically much larger than the far- field BAEP can be
recorded (4,40) (Fig. 38.13). This permits averaging using fewer
sweeps, facilitating more frequent assessment and more rapid
notification to the surgeons should adverse changes occur.

Assessment of IOM Data


The peak latencies of waves I, III, and V are measured, and the
amplitude of each of these components is measured with
respect to the trough that follows it. The central transmission
time, the interval between the peaks of waves I and V, can also
be calculated. Typical alarm criteria are a 50% drop in compo-
Figure 38.9 Brainstem auditory evoked potentials recorded simultane- nent amplitudes and a 1 msec increase in component latencies
ously from three different recording electrode linkages following monaural or in the central transmission time.
stimulation in a normal subject. The vertical dashed lines indicate the Loss or delay of wave V with preservation of waves I and III
peak latencies of waves IV and V in the Cz-Ai waveforms; these peaks are (Fig. 38.12) reflects compromise of the auditory pathways within
more widely separated in the Cz-Ac waveforms. (Reprinted from Legatt the brainstem, between the lower pons and the mesencephalon.
AD. Brainstem auditory evoked potentials: methodology, interpretation, If the BAEP changes are unilateral or asymmetrical, the predom-
and clinical application. In: Aminoff MJ, ed. Electrodiagnosis in Clinical inant pathology is most often, though not always, ipsilateral to
Neurology. 5th ed. New York, NY: Churchill Livingstone; 2005:489–523.) the ear whose stimulation gives the most abnormal BAEPs.
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 775

Figure 38.11 Intraoperative BAEPs to left ear stimulation recorded dur-


ing surgery for left vestibular schwannomas in two different patients,
showing persistence of wave I (arrows) after transection of the intracra-
nial eighth nerve. The nerves were intentionally sacrificed to permit
total resection of the tumors. (Reprinted from Legatt AD. Mechanisms
of intraoperative brainstem auditory evoked potential changes. J Clin
Neurophysiol. 2002;19:396–408.)

Internal auditory artery compromise probably accounts for


most of the cases where all BAEP components, including wave I,
Figure 38.10 Various IV–V complex morphologies in Cz-Ai wave-
are suddenly lost (Fig. 38.14), during surgery for cerebellopon-
forms recorded in normal subjects. (Reprinted from Chiappa K,
tine angle tumors (42).
Gladstone KJ, Young RR. Brain stem auditory evoked responses. Studies
When the cerebellum is retracted to gain access to the cere-
of waveform variations in 50 normal human subjects. Arch Neurol.
bellopontine angle (e.g., for a cerebellopontine angle tumor or
1979;36:81–87. Copyright © 1979 American Medical Association. All
during microvascular decompression surgery), this also moves
rights reserved.)
the brainstem away from the internal auditory meatus and
Loss or delay of waves III and V with preservation of wave I
(Fig. 38.11) reflects compromise of the eighth nerve (though
not of its most distal part, within the modiolus) or of the lower
pons. If the BAEP changes are unilateral or asymmetrical, the
predominant pathology is again most often ipsilateral to the ear
whose stimulation gives the most abnormal BAEPs.
Loss or delay of wave I and subsequent waves (Fig. 38.14), if
not due to technical problems, reflects cochlear dysfunction.
This may include cochlear ischemia or infarction due to com-
promise of the blood supply to the cochlea.

Causes of Adverse EP Changes


As previously noted, true positive BAEP changes may reflect
direct mechanical or thermal tissue injury as well as
ischemia/infarction. Two situations specifically pertaining to
BAEPs deserve special mention: internal auditory artery com- Figure 38.12 Intraoperative BAEPs to left ear stimulation recorded
promise and eighth nerve stretch. during the surgery of Figure 38.2. A: Clear waves I through VI were
The cochlea receives its blood supply from the intracranial present in these BAEPs, recorded just before the aneurysm ruptured.
circulation via the internal auditory artery. This vessel, which is B: BAEPs recorded after the aneurysm had been clipped and the basilar
usually a branch of the anterior inferior cerebellar artery, passes artery had been reopened show a loss of waves V and VI; waves I
through the internal auditory canal alongside the eighth nerve through IV were unaffected. (Modified from Legatt AD, Arezzo JC,
(41). Damage to this artery will cause cochlea ischemia or infarc- Vaughan HG Jr. The anatomic and physiologic bases of brain stem audi-
tion, affecting wave I and all subsequent BAEP components. tory evoked potentials. Neurol Clin. 1988;6:681–704.)
776 Part V ■ Complementary and Special Techniques

Figure 38.13 BAEPs recorded from a platinum pad electrode placed


on the intracranial eighth nerve (top) compared to surface-recorded
BAEPs (bottom) during surgery in a 52-year-old woman with a left-
sided intracanalicular acoustic neuroma. Note the voltage calibrations;
the near-field response is considerably larger. The patient had transient
BAEP changes during the resection but BAEPs were at baseline at its
end, and postoperative hearing was normal. (Courtesy of Dr. Timothy
A. Pedley.)
Figure 38.14 Consecutive BAEPs to left ear stimulation (earliest wave-
form at the top) recorded in a patient undergoing surgery for a left
acoustic neuroma. A clear wave I and a poorly formed wave V were ini-
stretches the eighth nerve, which can cause hearing loss. BAEP tially present and were stable during the initial dissection, but all BAEP
monitoring can be used to detect excessive eighth nerve stretch components disappeared simultaneously during dissection within the
(Fig. 38.15) and to notify the surgeons that the retraction needs internal auditory canal and remained absent through the end of the
to be reduced or readjusted. Uncorrected excessive traction on operation. This was most likely due to interruption of the blood supply
the eighth nerve can lead to avulsion of the distal nerve from to the cochlea via the internal auditory artery. (Reprinted from Legatt
the cochlea (43), causing hearing loss. AD. Mechanisms of intraoperative brainstem auditory evoked potential
Drilling of bone will produce high noise levels in both ears changes. J Clin Neurophysiol. 2002;19:396–408.)
via bone conduction and can alter the BAEPs due to acoustic
masking (44), simulating cochlear dysfunction. This would rep-
resent a change due to physiologic factors; it is not a “true pos- SOMATOSENSORY EVOKED POTENTIALS
itive” change reflecting compromise of the auditory system. It is
best to pause BAEP recording during drilling. While SEPs can be elicited by mechanical stimulation, for extra-
operative diagnostic studies and for IOM SEPs are elicited by
Typical Intraoperative Applications electrical stimulation of mixed peripheral nerves, most often the
BAEPs can be used for IOM of the ears, the eighth nerves, and median and ulnar nerves in the upper extremities and the
the infratentorial auditory pathways up through the level of the peroneal and posterior tibial nerves in the lower extremities
mesencephalon. They are not useful for monitoring the thala- (Fig. 38.16). Components generated in the peripheral nerves, the
mus (medial geniculate) or the auditory pathways within the spinal cord, the brainstem, and in cerebral cortex can be identi-
cerebrum. They are most often used to monitor surgery for fied. SEP components are typically named by their scalp polarity
eighth nerve tumors such as vestibular schwannomas (formerly and their average latency in unanesthetized normal subjects; for
called acoustic neuromas) and for tumors or vascular abnor- example, N20 is a negativity with a typical latency of 20 msec.
malities within the posterior fossa, both extra- axial and within Upper extremity SEPs are most often elicited by stimulation of
the brainstem. BAEPs are also useful to detect excessive eighth the median and ulnar nerves just proximal to the wrist. Median
nerve stretch, which can lead to hearing loss, from cerebellar nerve SEPs are most often used for extraoperative diagnostic SEP
retraction during surgery in the cerebellopontine angle, such as studies, because of the common occurrence of a degree of ulnar
microvascular decompression procedures (45). neuropathy at the elbow; carpal tunnel syndrome will not affect
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 777

Figure 38.15 Intraoperative BAEPs to


right ear stimulation recorded during sur-
gery for a right acoustic neuroma, showing
two runs recorded prior to (top) and after
(bottom) retraction of the cerebellum. The
most prominent change in the BAEPs was
an increase in the I–III interpeak interval of
more than 1 msec, reflecting stretching of
the eighth nerve. The smaller change in the
III–V interpeak interval may reflect effects
of the retraction on the brainstem.
(Reprinted from Legatt AD. Mechanisms of
intraoperative brainstem auditory evoked
potential changes. J Clin Neurophysiol.
2002;19:396–408.)

Figure 38.16 Examples of the four most common SEPs used for IOM, recorded in four different patients. Data from three
consecutive runs are superimposed. The arrowheads indicate the primary cortical SEPs (upper waveforms in each pair)
and the cervicomedullary SEP components (lower waveforms in each pair). Vertical calibration bar = 1 µV for the cortical
SEPs to peroneal nerve stimulation, 2 µV for the cortical SEPs to median nerve and to posterior tibial nerve stimulation,
and 1.5 µV for all of the other waveforms.
778 Part V ■ Complementary and Special Techniques

them because the stimulation site is proximal to the wrist. eral or medial aspect of the limb, at the same level, maximizes
Sensory fibers of the median nerve gain access to the spinal cord in- phase cancellation of the electrocardiogram (4). The periph-
through the C6 and C7 spinal roots (46). If the spinal cord path- eral nerve volley to posterior tibial nerve stimulation is
ways were compromised at a level below C6, the afferent activity recorded from a midline electrode behind the knee, referred to
traversing the C6 nerve root could generate SEPs and mask the either the lateral or medial aspect of that knee or to the midline
presence of more caudal spinal cord dysfunction. Thus, median electrode on the other knee.
nerve SEPs by themselves are not adequate for monitoring the Components originating in the gray matter of the spinal
spinal cord below the C6 level. Similarly, ulnar nerve SEPs are not cord at the level of root entry, N13 following median nerve
adequate for monitoring the spinal cord below the C8 level, stimulation and N22 following posterior tibial nerve stimula-
because sensory fiber of the ulnar nerve gains access to the spinal tion, are used during clinical SEP studies (3) but are of limited
cord through the C8 and T1 nerve roots (46). utility during IOM. They are often small and difficult to record,
In the lower extremity, the common peroneal nerve is typi- their recording sites may be too close to the surgical field, and
cally stimulated at the level of the knee, behind the head of the they are generated caudal to the central nervous system struc-
fibula. The posterior tibial nerve is stimulated at the ankle, tures that the IOM is intended to safeguard.
behind the medial malleolus; this permits recording of the Activity in the afferent somatosensory pathways at the level
peripheral nerve compound action potential behind the knee, of the dorsal column nucleus/cervicomedullary junction is typ-
which can help in the determination of the cause of a change in ically recorded as a far- field potential, P14 for upper limb stim-
the more rostrally recorded SEPs. Other advantages of posterior ulation and P31 for posterior tibial nerve stimulation (Fig.
tibial nerve SEPs as compared to peroneal nerve SEPs are that 38.16), with the positivity of the equivalent dipole field directed
posterior tibial nerve SEPs are typically larger (47), that stimu- upwards and anteriorly and the negativity directed downwards
lating electrodes at the ankle are usually easier to reach and and posteriorly. These cervicomedullary far- field potentials are
replace than those at the knee should they become dislodged best recorded between an electrode in the area where the dipole
during surgery, when the patient is draped, and that if the field is positive and one in which the dipole field is negative. In
patient is not pharmacologically paralyzed, posterior tibial extraoperative diagnostic SEP studies, these components are
nerve stimulation causes less patient movement than does per- typically recorded between the dorsal scalp and a noncephalic
oneal nerve stimulation. If posterior tibial nerve SEPs cannot be electrode (3). During IOM, recording electrode pairs with such
used due to peripheral neuropathy, the anatomy at the ankle, or large interelectrode distances tend to pick up more noise. A
a distal amputation, then peroneal nerve SEPs offer another way forehead- to- inion recording linkage can also pick up the SEP
to monitor the afferent somatosensory pathways from the legs. components generated at the level of the cervicomedullary
Similar to auditory EPs, a complex series of somatosensory junction; if the inion is within the surgical field, a forehead- to-
EPs with latencies ranging from a few milliseconds to hundreds earlobe recording linkage can also be used (48).
of milliseconds can be recorded following a single peripheral The primary cortical SEP to upper limb stimulation, N20, is
nerve shock. The shorter-latency SEP components, generated in recorded from the scalp over the parietal region contralateral to
the peripheral nerves, the spinal cord, and at the level of the cer- the stimulated arm, for example, at positions CP3 and CP4 of the
vicomedullary junction, are relatively insensitive to anesthetic International 10-10 System (49) for right arm and left arm stim-
effects. The primary cortical SEP components—N20 to stimula- ulation, respectively. The reference can be either the electrode
tion of the median or ulnar nerve at the wrist, P37 to stimulation over the other parietal region (CPi, ipsilateral to the stimulus) or
of the posterior tibial nerve at the ankle, and P27 to stimulation an electrode on the forehead. Recordings to a forehead reference
of the peroneal nerve at the knee—are affected more by anesthe- may also contain the N18 component, which is generated in sub-
sia (Fig. 38.4), but can be recorded and used for IOM if the anes- cortical white matter structures. If SEPs are being used to moni-
thetic regimen is not excessive. Longer-latency SEP components tor the cerebrum, a CPc-Cpi recording linkage can be used to
are even more attenuated by anesthesia and may be completely cancel the N18 component, which has a symmetrical distribution
suppressed, which limits their utility for IOM. Thus, SEPs can be over the top of the head (50), to ensure that the SEPs being mon-
used to monitor the central somatosensory pathways only up itored are entirely of cerebral origin. However, if SEPs are being
through the level of primary somatosensory cortex. used to monitor the spinal cord, a forehead reference may give a
somewhat larger SEP waveform, and is acceptable because N18
Components, Generators, and Recording and N20 would both be affected by spinal cord dysfunction.
Electrode Locations The scalp topographies of the primary cortical SEPs to lower
For extraoperative diagnostic studies of upper limb SEPs, the limb stimulation—P37 following posterior tibial nerve stimula-
signal generated by afferent peripheral nerve volley as it trav- tion at the ankle and P27 following peroneal nerve stimulation at
erses the brachial plexus is typically recorded by an electrode at the knee—vary from one subject to another because of anatom-
Erbs’ point as the N9 component. This can also be done during ical variability in the location and orientation of the foot area of
IOM, but in some cases Erbs’ point is too close to the surgical primary somatosensory cortex (51). These components are typi-
field, or the positioning of the patient may preclude secure cally largest in the midline or over the parietal area ipsilateral to
placement of an electrode there. The afferent peripheral volley the stimulated limb—a so-called “paradoxical” localization
can also be recorded by an electrode near the nerve at, or just (Fig. 38.17). Preoperative SEPs studies can be performed to
proximal to, the elbow; use of a reference electrode on the lat- determine the optimal location for recording electrodes for IOM
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 779

Figure 38.17 Extraoperative recordings of cortical SEPs to posterior tibial nerve stimulation during diagnostic SEP testing
in two different patients, and P37 generator locations and equivalent dipole orientations that would explain them. A P37
component that is largest in the midline (A) is explained by an almost vertically oriented P37 dipole (B). A P37 component
that is “paradoxical,” largest over the hemisphere ipsilateral to the stimulated nerve and inverted over the contralateral hemi-
sphere (C), is explained by a generator in the mesial wall of the hemisphere that produces a horizontally oriented P37 dipole
(D). Note that an electrode at CPz would not pick up a P37 component that is suitable for IOM in the example shown
in C. The larger peak that follows P37 has a wider distribution; suppression by anesthesia makes it not useful for IOM.

of lower limb SEPs in each individual patient, or electrodes can lesions or during surgery for aneurysms or vascular malfor-
be placed at multiple recording positions (e.g., CP3, CPz, and mations when the craniotomy opening permits placement of
CP4) to permit adequate recording of the cortical SEPs in all cortical surface electrodes.
patients no matter what the scalp topographies of their cortical
SEPs are. Recording Techniques
The primary cortical SEP to median or ulnar nerve stimula- SEPs are elicited using a pair of stimulating electrodes over each
tion (N20) is generated in the posterior bank of the central sul- tested peripheral nerve, with the cathode proximal to prevent
cus (Fig. 38.18) and thus inverts in polarity across the central the possibility of anodal block of the afferent signal. Either sur-
sulcus in recordings from the cortical surface (52–54). This face electrodes or subdermal needle stimulating electrodes can
inversion can be used to identify the central sulcus during resec- be used. The spacing between the surface electrodes, or between
tive surgery (55–57). The SEPs are typically recorded from metal the skin entry points of needle electrodes, should be about 3
disk electrodes embedded in a sheet of silastic plastic that is cm. If subdermal needles are used, they should be directed away
placed on the exposed cortical surface after the dura is opened. from each other; otherwise, the tips could be too close to each
Electrode strips containing several electrodes arranged in a lin- other and the stimulating current could travel between them
ear array can be used, but use of an electrode grid containing a without effectively stimulating the underlying nerve. If surface
rectangular grid of electrodes may make identification of the cup electrodes with electrode gel are used, care should be taken
central sulcus easier and faster (58). SEPs recorded from the to ensure that the gel does not leak out and form a salt bridge
cortical surface may also be used to monitor for compromise of between the stimulating electrodes, since this would shunt the
cortical somatosensory tracts during resection of cerebral stimulus current between them. Were this to happen, the nerve
780 Part V ■ Complementary and Special Techniques

In some patients with spinal cord lesions, the volleys along


the afferent pathways may be sufficiently desynchronized that
the cortical and cervicomedullary SEP components are not
identifiable. In such patients, recording electrodes placed
directly on the spinal cord rostral to the lesion may pick up sig-
nals that are sufficiently reproducible to be used for IOM (4).

Assessment of IOM Data


The peak latencies of the peripheral nerve potentials (if appli-
cable), the cervicomedullary far- field potentials, and the pri-
mary cortical SEPs are measured, and the amplitude of each of
these components is measured with respect to the following
peak of the opposite polarity. Depending on the configuration
of the SEP and the degree to which the SEP waveform is con-
taminated by lower- frequency noise, it may also be useful to
measure the amplitude of the SEP peak with respect to the pre-
Figure 38.18 Diagram showing generation of the N20/ P20 component ceding peak of the opposite polarity or with respect to the base-
of the median nerve SEP in primary somatosensory cortex located in the line level of the waveform preceding the SEP. Typical alarm
posterior bank of the central sulcus (shaded area). The arrow represents criteria are a 50% drop in component amplitudes and a 10%
the equivalent voltage dipole produced by activation of this area of cortex. increase in component latencies.
(Reprinted from Legatt AD, Kader A. Topography of the initial cortical If the rostrally recorded SEPs are lost, recording of the
component of the median nerve somatosensory evoked potential: relation- peripheral nerve compound action potential permits immedi-
ship to central sulcus anatomy. J Clin Neurophysiol. 2000;17:321–325.) ate categorization of the cause of the SEP change. If the periph-
eral nerve compound action potential also disappears, the cause
of the SEP change is most likely due to technical factors leading
would not be stimulated but the stimulating equipment would to inadequate stimulation of the peripheral nerve, though the
not indicate an open- circuit condition (4). possibility of mechanical or ischemic compromise of the
During extraoperative SEP studies, the stimulus intensity is peripheral nerve in the limb must also be considered. If the
typically adjusted to just above the threshold level necessary to peripheral nerve compound action potential persists but the
obtain a consistent muscle twitch, so as to not cause undue rostral SEPs disappear, the cause is most likely compromise of
patient discomfort. During IOM, higher stimulus intensities are the afferent somatosensory pathways due to surgical manipula-
used, to give a safety margin should the efficacy of peripheral tions, and the surgeons should be notified accordingly.
nerve stimulation decrease during the surgery due to technical If the region of the neuraxis that is at risk is caudal to the cer-
factors, such as the development of edema that would increase vicomedullary junction (e.g., the spinal cord), then both the
the distance between the nerve and the stimulating electrodes. primary cortical SEPs and the cervicomedullary far- field SEP
Peripheral nerve stimulation is typically accomplished using components are generated rostral to the region that is at risk,
trains of 200- µsec- duration constant-current stimulus pulses. and either may serve to monitor the condition of the spinal
Stimulus rates of 2 to 8 per second have been used for SEP cord. If, however, the part of the nervous system that is at risk
monitoring, but rates that are exact submultiples of the line fre- during the surgery is above the cervicomedullary junction, then
quency (e.g., 5 Hz or 6 Hz in North America) should be the subcortical far- field potentials can be used to verify that the
avoided; if such a submultiple were chosen, signal averaging peripheral nerves are being adequately stimulated and that the
would not eliminate line- frequency artifact that is present in afferent activity has reached the level of the cervicomedullary
the data. Typical filter settings are 5 to 30 Hz for the low- junction, but the cortical potentials must be examined to assess
frequency (high- pass) filter and 1000 to 3000 Hz for the high- the region of the brain or brainstem that is at risk during the
frequency (low- pass) filter. Line- frequency (e.g., 60 Hz) operation (Fig. 38.2).
“notch” filters should not be used for SEP recordings because Cortical SEPs are affected by surgical anesthesia, and
they can cause a “ringing” oscillatory artifact (59). increases in the anesthetic regimen can cause cortical SEP
The epoch or sweep duration used for averaging should be at changes resembling those due to surgical compromise of neural
least twice the latency of the longest EP component that is of structures (compare Figs. 38.2 and 38.4). If the anesthetic regi-
interest. A typical epoch duration for recording of upper limb men is changed substantially, the baseline amplitude and latency
SEPs is 50 msec; epoch durations in the 80- to 100-msec range levels for the patient’s SEPs may have to be reassessed. High
are appropriate for recording of lower limb SEPs. The choice of doses of inhalational agents, especially high doses of multiple
the number of sweeps per average will depend on the signal- to- agents, are undesirable. Even more important than avoiding
noise ratio of the raw data and may range from 200 to 1000. high anesthetic doses is avoiding large changes in the anesthetic
SEPs recorded from the cortical surface are typically much regimen (i.e., a sudden increase in the concentration of inhala-
larger than those recorded from the scalp, and the number of tional anesthetic or a bolus dose of an intravenous anesthetic
sweeps per average can be smaller. agent), particularly around the time of critical maneuvers such
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 781

as positioning of the head and neck in a patient with cervical If the constant- current SEP stimulator is unable to deliver
spinal stenosis or instability, clamping of cerebral vessels, or dis- the preset current level, most contemporary IOM equipment
traction during spinal deformity surgery (4). will indicate a high- impedance or open- circuit stimulus condi-
tion, which can help to identify technical problems with stimu-
Causes of Adverse EP Changes lation such as broken wires or dislodged electrodes. If, however,
As with all IOM, true positive SEP changes may reflect direct the paired stimulating electrodes are short- circuited together, as
mechanical or thermal tissue injury as well as ischemia/ by a salt bridge, then the nerve will not be stimulated effectively
infarction of the tissue due to compromise of its blood supply and the SEPs will deteriorate, but the equipment will not indi-
(Figs. 38.2 and 38.3). cate the error condition.
During aortic surgery with retrograde bypass via the femoral As previously mentioned, an increase in the anesthetic con-
artery, anterograde blood flow in the femoral artery is blocked, centration can cause attenuation and latency prolongation of
and the peripheral nerve in that leg will become ischemic. This the cortical SEPs that resembles SEP changes caused by
may lead to disappearance of all SEP components, including the somatosensory pathway compromise. If the region of the neu-
peripheral nerve components, to stimulation of that leg follow- raxis at risk is caudal to the cervicomedullary junction and cer-
ing the placement of the femoral artery catheter. The absence of vicomedullary far- field SEPs adequate for IOM are obtained,
the peripheral nerve volley would preclude IOM of the central they can be used to determine whether the changes in the cor-
somatosensory pathways from that leg. tical SEPs are due to anesthesia or to surgical manipulations.
Compression of the peripheral nerve in a limb or limb
ischemia due to positioning of the limb may also lead to atten- Typical Intraoperative Applications
uation or disappearance of the SEPs to stimulation of a nerve in SEPs can be used to monitor the condition of the central
that limb. Recording of the peripheral nerve SEP in that limb somatosensory pathways during surgery on the brain, brainstem,
may help to recognize that situation. If peripheral nerve SEPs or spinal cord that places those structures at risk or during ortho-
were not recorded, investigations to determine the cause of an pedic surgery that poses a risk to the spinal cord (Fig. 38.19).
adverse change in the rostral SEPs should include assessment of Neural structures are at risk of ischemic injury during vascular
the stimulated limb. surgery on the aorta, the carotid arteries, and during surgery for

Figure 38.19 Consecutive SEP runs recorded during spinal instrumentation and fusion surgery for congenital kyphosco-
liosis with multiple hemivertebrae in a 15-year-old boy; only the cortical SEPs are shown. The lower limb SEPs and MEPs
disappeared bilaterally during placement of pedicle screws; upper limb SEPs and MEPs remained present. The surgeons
were notified and the most recently placed screws were removed. A wake-up test was performed and the patient had left
leg weakness. The patient was reanesthetized and the remaining hardware was removed. Postoperatively the patient had
motor and sensory deficits in the left leg, with subsequent improvement. Shown here are the cortical SEPs from the last
four runs recorded before the adverse changes (top) and the first four runs recorded after them (bottom). The arrows indi-
cate the primary cortical SEP components (P37 for posterior tibial nerve stimulation and N20 for ulnar nerve stimulation).
Calibrations: 10 msec/ division and 1.5 µV/ division for posterior tibial nerve SEPs, 5 msec/ division and 2 µV/ division for
ulnar nerve SEPs.
782 Part V ■ Complementary and Special Techniques

aneurysms or arteriovenous malformations of vessels that supply and SEPs may show changes in the absence of MEP changes
the brain and spinal cord. during IOM (69).
During surgery on the thoracic and lumbar spine, the
brachial plexus or nerves in the arms may be stretched due to MOTOR EVOKED POTENTIALS
the positioning of the patient’s arms. Monitoring of upper limb
SEPs can be used to detect nerve stretch during surgery (60,61), MEP monitoring is used to directly assess the motor pathways
permitting intraoperative correction of the arm positioning to within the brain and spinal cord during surgery. If the dura is
reduce postoperative morbidity. opened and motor cortex is accessible, MEPs can be elicited by
Afferent somatosensory activity is carried rostrally, within direct cortical stimulation, but most often the brain is stimu-
the spinal cord, in both the dorsal columns and the spinothala- lated through the intact skull using TES. As discussed in
mic tracts. However, due to the slower conduction velocities Chapter 52, transcranial magnetic stimulation is of limited use
and temporal dispersion within the spinothalamic tracts, the in anesthetized patients.
activity in them does not contribute significantly to the earliest Stimulation of the spinal cord rostral to the region that is at
SEP components generated in the brain. The rostral SEPs used risk coupled with recording of responses from peripheral
for IOM are mediated almost entirely by the dorsal columns nerves in the legs has been proposed as another way of moni-
(51,62), and therefore directly assess only that portion of the toring the spinal cord motor tracts (70), but the responses
spinal cord, which is supplied by the posterior spinal arteries. obtained in this way predominantly reflect retrograde conduc-
Since intraoperative spinal cord damage during spinal surgery tion within somatosensory fibers in the dorsal columns (71)
may be on a vascular basis (63), SEP monitoring may fail to and therefore, like SEPs, they may fail to detect intraoperative
detect ischemic spinal cord compromise that involves the ante- damage to the corticospinal tracts (72). Responses recorded
rior spinal artery territory but spares the dorsal columns; SEPs from muscles may be mediated both by descending motor
may also remain unchanged following mechanical trauma lim- tracts and by retrograde conduction in somatosensory tracts
ited to the anterior spinal cord. SEP monitoring may therefore with reflex connections to the alpha motor neurons (73–75);
fail to detect damage that is limited to the motor tracts in the thus, they do not convey the desired specificity. The only way to
anterolateral funiculus of the spinal cord (64–66). Fortunately be sure that the signals being monitored are mediated solely by
such “false- negative” SEP studies are rare; surgical situations the motor tracts within the spinal cord is to stimulate rostral to
that compromise the anterior spinal cord usually compromise the somatosensory synapses in the dorsal column nuclei (76).
the posterior spinal cord as well. MEP monitoring was devel-
oped to directly monitor the central motor pathways. Components, Generators, and Recording Electrodes
Even if MEPs are employed during surgery on the spinal MEPs to TES can be recorded from the spinal cord or from mus-
cord, SEPs should still be monitored. Because of the anatomical cles. Myogenic MEPs (“M-waves”) (Fig. 38.20, left) are more sen-
separation between the somatosensory and motor pathways sitive to anesthetic effects than are the other EPs commonly used
and their different blood supplies, the somatosensory pathways for IOM (Fig. 38.5) due to anesthetic effects at the anterior horn
can be damaged without effects on the motor pathways (67,68), cell synapse, and total intravenous anesthesia (e.g., a combination

Figure 38.20 M-waves and D-waves


recorded during resection of a recurrent
glioma of the cervical spinal cord in a
22-year-old woman. Left: Consecutive
recordings of M-waves in right hand mus-
cles to multiple-pulse TES with the anode
on the left. Right: D-waves recorded from
an epidural electrode placed over the
spinal cord caudal to the tumor to single-
pulse TES with the anode on the left.
D-waves were recorded more frequently
than M-waves; the D-wave runs included
in this figure are those that were recorded
closest in time to the M-waves that are
shown. M-waves are single sweeps;
D-waves are averages of the responses to
25 stimuli. Tumor resection was com-
pleted at 10:34.
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 783

of propofol and narcotic infusions) with avoidance of any inhala- densities capable of stimulating corticospinal tract axons are
tional agents may be required to record MEPs in some patients. produced further and further away from the surface of the head
M-waves will also be attenuated or eliminated by neuromuscular and may be able to stimulate the motor pathways as far caudally
blockade (NMB). They may be recordable under partial NMB, as the medulla (81). During IOM of the brainstem and of the
but that NMB should be maintained by a continuous infusion of corticospinal tracts within the cerebrum, this could prevent
paralytic drug rather than by intermittent bolus doses (77). recognition of surgery-related motor tract compromise if the
MEPs recorded from the spinal cord ( “D-waves”) (Fig. motor tracts were stimulated caudal to that dysfunction.
38.20, right) are relatively insensitive to anesthetic effects, since Therefore, when monitoring these surgeries, the TES stimulus
there are no intervening synapses between the site of corti- intensity should be kept relatively low, for example, to a level that
cospinal tract stimulation in the brain and the spinal cord produces MEPs in one limb but not in multiple limbs.
recording site. They are also more consistent from run to run
and can be recorded in some patients in whom M- waves are not Assessment of IOM Data
identifiable due to preexisting pathology or anesthetic effects. D- waves are highly consistent from run to run (Fig. 38.20,
However, invasive electrodes must be placed close to the spinal right) and are evaluated by criteria similar to those used for sen-
cord to record them, and they may miss unilateral deficits if the sory EPs, for example, a 50% decrease in amplitude or a 10%
motor tracts in the brain are stimulated bilaterally (78). Also, increase in latency would constitute a significant change.
M- waves can be used to monitor the lower spinal cord where M-waves typically show substantial run-to-run variability
D- waves are not usable, may show adverse changes earlier than (Fig. 38.20, left), and uniform use of a 50% amplitude criterion
D- waves when the motor tracts are compromised, and may be would lead to an unacceptably high level of false alarms. In many
monitorable in some patients in whom preexisting spinal cord centers, the alarm criterion is complete disappearance of the
pathology has desynchronized the descending corticospinal MEP, although a higher percentage amplitude criterion (e.g., a
tract volley so that there are no identifiable D- waves (79). 75% or a 90% drop in amplitude) may also be used. When eval-
M- waves can be recorded from surface or needle electrodes uating M-waves, the anesthetic regimen should be taken into
placed in or over the muscles of interest, and reflect the com- account. Increases in the anesthetic doses, especially the addition
pound motor action potentials elicited within the muscles. of an inhalational agent to an intravenous regimen, may cause
D- waves can be recorded using an epidural recording electrode substantial amplitude attenuation or eliminate the MEPs (Fig.
placed percutaneously via a Touhey needle, or by electrodes 38.5). In addition, during surgeries lasting several hours MEPs
placed within the surgical field close to the spinal cord. They are amplitudes tend to decrease, and thresholds to elicit MEPs tend
generated by propagating action potentials within corticospinal to rise, a phenomenon known as “anesthetic fade” (82,83). This
tract axons. may require reassessment of the MEP baseline.
Since TES preferentially stimulates the brain under the anode,
Recording Techniques it predominantly elicits M-waves in muscles contralateral to the
To record M- waves to TES, a high- intensity multipulse stimula- anode (see Chapter 52). M-waves may also be present in muscles
tor is used to produce multiple closely spaced descending vol- contralateral to the TES cathode, but these cathodal stimulation
leys in the corticospinal tract, so that temporal summation of elicited MEPs (CSE-MEPs) may disappear due to anesthetic-
the excitatory postsynaptic potentials that they produce can fire related changes in cortical excitability, in the absence of corti-
the anterior horn cells (see Chapter 52). Typical train parame- cospinal tract compromise. Therefore, adverse changes in
ters (69) are 3 to 7 stimulus pulses per train and interpulse CSE-MEPs should not be interpreted as definite evidence of spinal
intervals of 2 to 4 msec within the train. Wide-open filters (e.g., cord compromise. Instead, MEPs elicited by anodal stimulation
5 to 3000 Hz) permit recording of both lower and higher fre- should be used to more accurately assess the motor pathways to
quency components of the MEP waveform. the limb(s) in which the CSE-MEPs had deteriorated (84).
Only a small percentage of the alpha motor neuron pool is
activated with each TES stimulus, and the subset of alpha motor Causes of Adverse EP Changes
neurons that are activated varies from run to run. Therefore, M- The causes of adverse MEP changes are similar to those
waves typically vary substantially in waveshape from trial to trial described for SEPs, including direct mechanical or thermal tis-
(Fig. 38.20, left), and signal averaging should not be employed. sue injury and ischemia/infarction of neural or muscle tissue
Nor is it required, since the single-trial myogenic MEPs are due to compromise of its blood supply. Apparent significant EP
typically quite large, often hundreds of microvolts in amplitude. attenuations due to changes in the anesthetic regimen are even
D-waves are much smaller, and signal averaging of a small more common for M- wave monitoring than for SEP monitor-
number of sweeps (e.g., 5 to 25) is useful to obtain a clearer and ing. If the spinal cord below the neck is at risk (as in scoliosis or
more consistent waveform. Since there is no intervening synapse, aortic surgery), then MEPs recorded from upper limb muscles
pulse train stimulation is not used. This makes it less likely that can be used as controls to identify anesthetic effects.
the stimulation will cause undesirable patient movement.
D-waves can be elicited by stimulation of subcortical white Typical Intraoperative Applications
matter (80); therefore, M-waves to multipulse stimulation can MEPs are used to monitor the condition of the corticospinal
also be elicited by stimulation of subcortical white matter. As the tracts during surgery on the brain, brainstem, or spinal cord
TES stimulus intensity is increased, intraparenchymal current that places those structures at risk, during orthopedic surgery
784 Part V ■ Complementary and Special Techniques

Figure 38.21 Consecutive MEP runs recorded during the same operation as the data shown in Fig. 38.19. Shown here are
the last four MEP runs recorded from the abductor hallucis and tibialis anterior muscles to contralateral anodal TES before
the adverse changes (top) and the first four runs recorded after them (bottom). The MEPs recorded from the hand muscles
did not change.

that poses a risk to the spinal cord (Fig. 38.21), or during sur- 6. Yvert B, Crouzeix A, Bertrand O, et al. Multiple supratemporal
gery on the aorta or on other blood vessels that supply the brain sources of magnetic and electric auditory evoked middle latency
and spinal cord. They can also be used to assess the function of components in humans. Cereb Cortex. 2001;11:411–423.
cranial nerves or spinal nerve roots by recording the MEPs in 7. Schneider G, Hollweck R, Ningler M, et al. Detection of conscious-
ness by electroencephalogram and auditory evoked potentials.
the muscles that they innervate following stimulation of the
Anesthesiology. 2005;103:934–943.
corticospinal and corticobulbar tracts within the brain.
8. Legatt AD. Brainstem auditory evoked potentials: methodology,
As noted above, the somatosensory pathways may be dam- interpretation, and clinical application. In: Aminoff MJ, ed.
aged without MEP changes, so SEP monitoring should also be Electrodiagnosis in Clinical Neurology. 5th ed. New York, NY:
employed when MEPs are used to monitor the spinal cord. Also, Churchill Livingstone; 2005:489–523.
monitoring both SEPs and MEPs provides a measure of redun- 9. Banoub M, Tetzlaff JE, Schubert A. Pharmacologic and physiologic
dancy for monitoring the integrity of the spinal cord; two inde- influences affecting sensory evoked potentials: implications for
pendent monitoring techniques are employed, each capable of perioperative monitoring. Anesthesiology. 2003;99:716–737.
detecting most cases of spinal cord compromise in case the 10. Stockard JJ, Pope-Stockard JE, Sharbrough FW. Brainstem audi-
other technique cannot provide effective IOM due to preexist- tory evoked potentials in neurology: methodology, interpretation,
ing neurologic compromise, anesthetic effects, NMB, exces- and clinical application. In: Aminoff MJ, ed. Electrodiagnosis in
Clinical Neurology. 3rd ed. New York, NY: Churchill Livingstone;
sively noisy data, or other technical problems (85).
1992:503–536.
11. Litscher G. Continuous brainstem auditory evoked potential moni-
REFERENCES toring during nocturnal sleep. Int J Neurosci. 1995;82:135–142.
12. Markand ON, Lee BI, Warren C, et al. Effects of hypothermia on
1. Epstein CM, Boor DR. Principles of signal analysis and averaging. brainstem auditory evoked potentials in humans. Ann Neurol.
Neurol Clin. 1988;6:649–656. 1987;22:507–513.
2. American Clinical Neurophysiology Society. Guideline 9C: guide- 13. Rodriguez RA, Edmonds HL Jr, Auden SM, et al. Auditory brain-
lines on short- latency auditory evoked potentials. J Clin stem evoked responses and temperature monitoring during pedi-
Neurophysiol. 2006;23:157–167. atric cardiopulmonary bypass. Can J Anaesth. 1999;46:832–839.
3. American Clinical Neurophysiology Society. Guideline 9D: guide- 14. Jewett DL, Williston JS. Auditory- evoked far fields averaged from
lines on short-latency somatosensory evoked potentials. J Clin the scalp of humans. Brain. 1971;94:681–696.
Neurophysiol. 2006;23:168–179. 15. Chiappa K, Gladstone KJ, Young RR. Brain stem auditory evoked
4. Legatt AD. Intraoperative neurophysiologic monitoring. In: Frost responses. Studies of waveform variations in 50 normal human
EAM, ed. Clinical Anesthesia in Neurosurgery. 2nd ed. Boston, MA: subjects. Arch Neurol. 1979;36:81–87.
Butterworth-Heinemann; 1991:63–127. 16. Legatt AD, Arezzo JC, Vaughan HG Jr. The anatomic and physio-
5. Legatt AD. Mechanisms of intraoperative brainstem auditory logic bases of brain stem auditory evoked potentials. Neurol Clin.
evoked potential changes. J Clin Neurophysiol. 2002;19:396–408. 1988;6:681–704.
Chapter 38 ■ Intraoperative Evoked Potential Monitoring 785

17. Gersdorff MCH. Simultaneous recordings of human auditory poten- 38. Little JR, Lesser RP, Lueders H, et al. Brain stem auditory evoked
tials: transtympanic electrocochleography (ECoG) and brainstem- potentials in posterior circulation surgery. Neurosurgery. 1983;12:
evoked responses (BER). Arch Otorhinolaryngol. 1982;234:15–20. 496–502.
18. Hughes JR, Fino JJ. A review of generators of the brainstem audi- 39. Roeser RJ, Clark JL. Clinical masking. In: Roeser RJ, Valente M,
tory evoked potential: contribution of an experimental study. Hosford-Dunn H, eds. Audiology: Diagnosis. New York, NY: Thieme;
J Clin Neurophysiol. 1985;2:355–381. 2000:253–279.
19. Grandori F. Field analysis of auditory evoked brainstem potentials. 40. Møller AR, Jannetta PJ. Monitoring auditory functions during cra-
Hear Res. 1986;21:51–58. nial nerve microvascular decompression operations by direct
20. Raudzens PA, Shetter AG. Intraoperative monitoring of brain- recording from the eighth nerve. J Neurosurg. 1983;59:493–499.
stem auditory evoked potentials. J Neurosurg. 1982;57:341–348. 41. Kim HN, Kim YH, Park IY, et al. Variability of the surgical anatomy
21. Legatt AD, Pedley TA, Emerson RG, et al. Electrophysiological moni- of the neurovascular complex of the cerebellopontine angle. Ann
toring of seventh and eighth nerve function during surgery for Otol Rhinol Laryngol. 1990;99:288–296.
acoustic neuromas. Electroencephalogr Clin Neurophysiol. 1986;64:30P. 42. Nadol JB Jr, Levine R, Ojemann RG, et al. Preservation of hearing in
22. Strominger NL. The origins, course and distribution of the dorsal surgical removal of acoustic neuromas of the internal auditory canal
and intermediate acoustic striae in the rhesus monkey. J Comp and cerebellar pontine angle. Laryngoscope. 1987;97:1287–1294.
Neurol. 1973;147:209–234. 43. Sekiya T, Møller AR. Avulsion rupture of the internal auditory
23. Strominger NL, Nelson LR, Dougherty WJ. Second- order auditory artery during operations in the cerebellopontine angle: a study in
pathways in the chimpanzee. J Comp Neurol. 1977;172:349–366. monkeys. Neurosurgery. 1987;21:631–637.
24. Strominger NL, Strominger AI. Ascending brain stem projections 44. Levine RA, Ronner SF, Ojemann RG. Auditory evoked potential
of the anteroventral cochlear nucleus in the rhesus monkey. J and other neurophysiologic monitoring techniques during tumor
Comp Neurol. 1971;143:217–242. surgery in the cerebellopontine angle. In: Loftus CM, Traynelis VC,
25. Brown RH Jr, Chiappa KH, Brooks E. Brain stem auditory evoked eds. Intraoperative Monitoring Techniques in Neurosurgery. New
responses in 22 patients with intrinsic brain stem lesions: implica- York, NY: McGraw-Hill; 1994:175–191.
tions for clinical interpretations. Electroencephalogr Clin Neuro- 45. Radtke RA, Erwin CW, Wilkins RH. Intraoperative brainstem
physiol. 1981;52:38P. auditory evoked potentials: significant decrease in postoperative
26. Faught E, Oh SJ. Brainstem auditory evoked responses in brain- morbidity. Neurology. 1989;39:187–191.
stem infarction. Stroke. 1985;16:701–705. 46. Kimura J, ed. Electrodiagnosis in Diseases of Nerve and Muscle:
27. Oh SJ, Kuba T, Soyer A, et al. Lateralization of brainstem lesions by Principles and Practice. 3rd ed. Oxford: University Press; 2001.
brainstem auditory evoked potentials. Neurology. 1981;31:14–18. 47. Pelosi L, Cracco JB, Cracco RQ, et al. Comparison of scalp distri-
28. Stockard JJ, Rossiter VS. Clinical and pathologic correlates of brain bution of short latency somatosensory evoked potentials (SSEPs)
stem auditory response abnormalities. Neurology. 1977;27:316–325. to stimulation of different nerves in the lower extremity.
29. Starr A, Hamilton AE. Correlation between confirmed sites of neu- Electroencephalogr Clin Neurophysiol. 1988;71:422–428.
rological lesions and abnormalities of far- field auditory brainstem 48. Fried S, Legatt AD. The utility of a forehead- to-inion derivation in
responses. Electroencephalogr Clin Neurophysiol. 1976;41:595–608. recording the subcortical far- field potential (P14) during median
30. Stockard JJ, Stockard JE, Sharbrough FW. Detection and localiza- nerve somatosensory evoked potential (SEP) testing. J Clin
tion of occult lesions with brainstem auditory responses. Mayo Neurophysiol. In press.
Clin Proc. 1977;52:761–769. 49. American Clinical Neurophysiology Society. Guideline five: guide-
31. Hirsch BE, Durrant JD, Yetiser S, et al. Localizing retrocochlear lines for standard electrode position nomenclature. J Clin
hearing loss. Am J Otol. 1996;17:537–546. Neurophysiol. 1996;23:107–110.
32. York DH. Correlation between a unilateral midbrain-pontine 50. Desmedt JE, Cheron G. Non-cephalic reference recording of early
lesion and abnormalities of brain-stem auditory evoked potential. somatosensory potentials to finger stimulation in adult or aging
Electroencephalogr Clin Neurophysiol. 1986;65:282–288. normal man: differentiation of widespread N18 and contralateral
33. Scaioli V, Savoiardo M, Bussone G, et al. Brain-stem auditory N20 from prerolandic P22 and N30 components. Electroencephalogr
evoked potentials (BAEPs) and magnetic resonance imaging (MRI) Clin Neurophysiol. 1981;52:553–570.
in a case of facial myokymia. Electroencephalogr Clin Neurophysiol. 51. Emerson RG. Anatomic and physiologic bases of posterior tibial
1988;71:153–156. nerve somatosensory evoked potentials. Neurol Clin. 1988;6:
34. Zanette G, Carteri A, Cusumano S. Reappearance of brain- stem 735–749.
auditory evoked potentials after surgical treatment of a brain-stem 52. Allison T, Goff WR, Williamson PD, et al. On the neural origin of
hemorrhage: contributions to the question of wave generation. early components of the human somatosensory evoked potential. In:
Electroencephalogr Clin Neurophysiol. 1990;77:140–144. Desmedt JE, ed. Clinical Uses of Cerebral, Brainstem and Spinal
35. Fischer C, Bognar L, Turjman F, et al. Auditory evoked potentials Somatosensory Evoked Potentials. Progress in Clinical Neurophysiology.
in a patient with a unilateral lesion of the inferior colliculus and Vol 7. Basel: S Karger; 1980:51–68.
medial geniculate body. Electroencephalogr Clin Neurophysiol. 53. Broughton R, Rasmussen T, Branch C. Scalp and direct cortical
1995;96:261–267. recordings of somatosensory evoked potentials in man (circa 1967).
36. Emerson RG, Brooks EB, Parker SW, et al. Effects of click polarity on Can J Psychol. 1981;35:136–158.
brainstem auditory evoked potentials in normal subjects and 54. Lueders H, Lesser RP, Hahn J, et al. Cortical somatosensory evoked
patients: unexpected sensitivity of wave V. Ann N Y Acad Sci. 1982; potentials in response to hand stimulation. J Neurosurg. 1983;58:
388:710–721. 885–894.
37. Schwartz DM, Morris MD, Spydell JD, et al. Influence of click 55. Gregorie EM, Goldring S. Localization of function in the excision
polarity on the brain- stem auditory evoked response (BAER) of lesions from the sensorimotor region. J Neurosurg. 1984;61:
revisited. Electroencephalogr Clin Neurophysiol. 1990;77:445–457. 1047–1054.
786 Part V ■ Complementary and Special Techniques

56. King RB, Schell G. Cortical localization and monitoring during 71. Toleikis JR, Skelly JP, Carlvin AO, et al. Spinally elicited peripheral
cerebral operations. J Neurosurg. 1987;67:210–219. nerve responses are sensory rather than motor. Clin Neurophysiol.
57. Wood CC, Spencer DD, Allison T, et al. Localization of human sen- 2000;111:736–742.
sorimotor cortex during surgery by cortical surface recording of 72. Minahan RE, Sepkuty JP, Lesser RP, et al. Anterior spinal cord
somatosensory evoked potentials. J Neurosurg. 1988;68:99–111. injury with preserved neurogenic “motor” evoked potentials. Clin
58. Legatt AD, Kader A. Topography of the initial cortical component Neurophysiol. 2001;112:1442–1450.
of the median nerve somatosensory evoked potential: relationship 73. Machida M, Weinstein SL, Yamada T, et al. Monitoring of motor
to central sulcus anatomy. J Clin Neurophysiol. 2000;17:321–325. action potentials after stimulation of the spinal cord. J Bone Joint
59. Yamada T. The anatomic and physiologic bases of median nerve Surg. 1988;70:911–918.
somatosensory evoked potentials. Neurol Clin. 1988;6:705–733. 74. Mochida K, Shinomiya K, Komori H, et al. A new method of mul-
60. O’Brien MF, Lenke LG, Bridwell KH, et al. Evoked potential mon- tisegment motor pathway monitoring using muscle potentials after
itoring of the upper extremities during thoracic and lumbar spinal train spinal stimulation. Spine. 1995;20:2240–2246.
deformity surgery: a prospective study. J Spinal Disord. 1994;7: 75. Su CF, Haghighi SS, Oro JJ, et al. “Backfiring” in spinal cord moni-
277–284. toring. High thoracic spinal cord stimulation evokes sciatic response
61. Chung I, Glow JA, Dimopoulos V, et al. Upper- limb somatosen- by antidromic sensory pathway conduction, not motor tract conduc-
sory evoked potential monitoring in lumbosacral spine surgery: a tion. Spine. 1992;17:504–508.
prognostic marker for position- related ulnar nerve injury. Spine J. 76. Legatt AD, Ellen R. Grass lecture: motor evoked potential monitor-
2009;9:287–295. ing. Am J Electroneurodiagnostic Technol. 2004;44:223–243.
62. Cusick JF, Myklebust JB, Larson SJ, et al. Spinal cord evaluation by 77. Sloan T, Heyer EJ. Anesthesia for intraoperative neurophysiologic
cortical evoked responses. Arch Neurol. 1979;36:140–143. monitoring of the spinal cord. J Clin Neurophysiol. 2002;19:430–443.
63. Machida M, Weinstein SL, Yamada T, et al. Dissociation of muscle 78. Burke D, Hicks RG. Surgical monitoring of motor pathways. J Clin
action potentials and spinal somatosensory evoked potentials after Neurophysiol. 1998;15:194–205.
ischemic damage of spinal cord. Spine. 1988;13:1119–1124. 79. Deletis V, Kothbauer K. Intraoperative neurophysiology of the cor-
64. Ben-David B, Haller G, Taylor P. Anterior spinal fusion complicated ticospinal tract. In: Stålberg E, Sharma HS, Olsson Y, eds. Spinal
by paraplegia: a case report of a false-negative somatosensory- Cord Monitoring. Wein: Springer; 1998:421–444.
evoked potential. Spine. 1987;12:536–539. 80. Amassian VE, Stewart M, Quirk GJ, et al. Physiological basis of motor
65. Zornow MH, Grafe MR, Tybor C, et al. Preservation of evoked effects of a transient stimulus to cerebral cortex. Neurosurgery. 1987;
potentials in a case of anterior spinal artery syndrome. Electroen- 20:74–93.
cephalogr Clin Neurophysiol. 1990;77:137–139. 81. Rothwell J, Burke D, Hicks R, et al. Transcranial electrical stimula-
66. Jones SJ, Buonamassa S, Crockard HA. Two cases of quadriparesis tion of the motor cortex in man: further evidence for the site of
following anterior cervical discectomy, with normal perioperative activation. J Physiol. 1994;481:243–250.
somatosensory evoked potentials. J Neurol Neurosurg Psychiatry. 82. Lyon R, Feiner J, Lieberman JA. Progressive suppression of motor
2003;74:273–276. evoked potentials during general anesthesia: the phenomenon of
67. Ben- David B, Taylor PD, Haller GS. Posterior spinal fusion com- “anesthetic fade.” J Neurosurg Anesthesiol. 2005;17:13–19.
plicated by posterior column injury. A case report of a false- 83. MacDonald DB, Al Zayed Z, Khoudeir I, et al. Monitoring scoliosis
negative wake-up test. Spine. 1987;12:540–543. surgery with combined multiple pulse transcranial electric motor
68. Chatrian G-E, Berger MS, Wirch AL. Discrepancy between intraop- and cortical somatosensory- evoked potentials from the lower and
erative SSEP’s and postoperative function. J Neurosurg. 1988;69: upper extremities. Spine. 2003;28:194–203.
450–454. 84. Legatt AD. MEPs elicited by cathodal stimulation during transcra-
69. Legatt AD. Current practice of motor evoked potential monitor- nial electrical stimulation- MEP monitoring. Neurology. 2006;
ing: results of a survey. J Clin Neurophysiol. 2002;19:454–460. 66(suppl 2):A68.
70. Owen JH, Laschinger J, Bridwell K, et al. Sensitivity and specificity 85. Nagle KJ, Emerson RG, Adams DC, et al. Intraoperative monitoring
of somatosensory and neurogenic- motor evoked potentials in ani- of motor evoked potentials: a review of 116 cases. Neurology. 1996;47:
mals and humans. Spine. 1988;13:1111–1118. 999–1004.
CHAPTER

Monitoring EEG during Carotid Surgery


THORU YAMADA AND MALCOLM YEH 39
INTRODUCTION Whether to use a shunt has long been argued by many inves-
tigators. Some have proposed the use of shunt in all CEAs (9)
The studies from the North American Symptomatic Carotid while others have stressed the increase of stroke incidence with
Endarterectomy Trial Collaborators (1) and the European the use of shunt and have argued that the shunt should not be
Carotid Surgery Trialists’ Collaboration Group (2) have shown used at all (10,11). Most CEAs, however, have been done with
the benefit of carotid endarterectomy (CEA) to prevent stroke, selective shunting based on EEG or other monitoring methods.
especially for patients with high-grade carotid artery stenosis. A Irrespective of the shunt used routinely, selectively or not at all,
more recent study has reaffirmed the effectiveness of CEA, the incidences of perioperative stroke were overall similar, 3%
especially in symptomatic patients with severe carotid stenosis (1). In three groups of studies, however, the incidence of stroke
( 70%) (3). Patients with asymptomatic stenosis in the 50% to was highest among those using a shunt routinely and lowest
60% range are in general uncertain candidates for CEA, but among those never using a shunt. These data suggest that intra-
may well benefit, especially for patients with additional medical operative or postoperative stroke is more likely due to emboli
conditions having a higher stroke risk. The CEA benefit is sig- rather than hypoperfusion or ischemia and thus raises a ques-
nificantly less for asymptomatic patients (4,5), but CEA may tion of the value to shunting and monitoring (12). Indeed in a
still be applicable for asymptomatic patients with a high degree study using RA (patients were awake during the CEA), only one
of stenosis (2), plaque with ulceration (6), and an occluded con- of 21 neurologic deficits (a total of 345 patients) was attributable
tralateral carotid artery (7). to cerebral anoxia and remaining 20 patients were due to
Because CEA always carries a risk of morbidity and mortal- thrombo-embolic origin (13). Another large scale of review
ity (allowable rates of stroke and death rate of less than 6%), study (3062 CEAs) showed that intraoperative ischemia, throm-
CEA for asymptomatic patients should only be considered if bosis, embolization, and intracranial hemorrhage accounted for
surgery could be done by an expert surgeon with track record 47 of the 63 deficits (14). Of these, thrombo-embolic events were
of low complication rates of less than 3%. CEAs are generally the most common cause of neurologic deficit accounting for
not indicated for patients with less than 50% symptomatic or 45%. Nonetheless a substantial number of patients would suffer
less than 60% asymptomatic stenosis (8). from ischemic stroke unless appropriate protection is provided.
CEA can be performed under general or regional anesthesia Moreover, in some cases, postoperative stroke could be extensive
(RA). During CEA, the internal carotid artery must be cross- resulting in global cerebral dysfunction. This is where various
clamped; therefore, the brain becomes subject to ischemia or monitoring methods can be used to warn of an impending
hypoperfusion. The brain can tolerate more than 1 hour if cere- ischemic event. One study comparing selective shunting based
bral blood flow is greater than 15 mL/100 g/min, but the severe on EEG (311 patients) and a nonshunt group (591 patients) in a
infarct would occur within 4 minutes with zero blood flow. A single institute reported much higher incidence of postoperative
number of monitoring techniques are available for examining the stroke in the nonshunt group (13 patients) as compared to a
cerebral perfusion and detecting the risk of cerebral ischemia. selective shunt group (1 patient) (15).
These include EEG, somatosensory-evoked potential (SSEP)
(median nerve stimulation), transcranial Doppler, and oxygen ANESTHESIA
saturation of hemoglobin using near-infrared spectroscopy,
stump-pressure measurement, and cerebral blood flow by Xenon- Commonly used anesthesia for CEA surgery is gaseous anesthe-
133. In order to protect brain from ischemic damage during cross- sia with isoflurane ranging between 0.4% and 0.6% with 40%
clamping, placement of a catheter to shunt blood around the to 60% N2O and oxygen. This combination may be supple-
surgical site has been used. Because the shunt placement itself car- mented by sufentanil or remifentanil (16,17). An alternative
ries a significant risk of embolization as a result of dislodging ath- anesthetic regimen consists of nitrous oxide in oxygen and
erosclerotic emboli coming from the vessel wall, only patients with propofol after induction with etomidate and fentanyl (17).
a potentially high risk of developing ischemic brain damage are Other commonly used inhalation agents include desflurane,
selected for shunt placement. Determining which patients require enflurane, and sevoflurane. Overall different anesthetic agents
shunts depend on the multiple factors. EEG has been the most show a similar EEG pattern when used at a concentration below
commonly used monitoring method for shunt determination but their minimal alveolar concentrations (MAC) level (the level
other tests are complimentary or may be more sensitive and accu- necessary to prevent movement to painful stimuli in about 50%
rate for predicting ischemic insult in some cases. of subjects).

787
788 Part V ■ Complementary and Special Techniques

EEG Change during Induction EEG Pattern During Recovery from Anesthesia
Most anesthesia inductions are carried out by etomidate, propo- When discontinuing anesthetics at the sub-MAC level, the WAR
fol, or thiopental via intravenous administration. This initially pattern decreases in its amplitude and increases in frequency,
produces an EEG showing beta activity with a decrease in the which subsequently changes to the beta range activity. There is
alpha rhythm. The beta activity rapidly becomes more wide- also a decrease of the WPS pattern before the patient regains
spread, with increases in amplitude. This is followed by a slowing consciousness (18). During the transition from the anesthetic
in the beta frequency that approaches the alpha frequency activ- state to the arousal state, bursts of intermittent delta activity,
ity. During this phase, the beta or alpha activity may be inter- resembling a FIRDA pattern, may appear which are similar to
mixed with a burst of high amplitude, intermittent rhythmic but less prominent than those seen during induction.
delta activity, which is often of frontal dominance, that is, FIRDA
(frontal intermittent delta activity) before a steady-state anes- Other Factors That Affect the EEG
thetic pattern is established (Figs. 39.1A, B and 39.2A,B) (18). The decreasing PaCO 2 level below 40 mm Hg or decreasing
the amount of stimulation that would be painful if given dur-
EEG Patterns at Sub-MAC Concentration ing awake state would produce a pattern suggestive of a deeper
At this light level of steady-state anesthesia, a widespread ante- level of anesthesia, that is, increase of slow waves, especially
riorly dominant rhythm (WAR) pattern, usually in lower beta during light sub-MAC level of anesthesia. Severe blood pres-
or alpha frequency activity, is seen with all of the above sure drops are associated with increases of slow waves and
described agents (18). The frequency of this pattern tends to eventual flattening of the EEG activity. Hypothermia slows the
slow with increasing concentration of these agents. This anes- EEG frequency spectrum with gradual reduction of amplitude
thetic WAR pattern is strikingly similar to the alpha-coma pat- and eventually leads to EEG “flattening” at about 20 C to
tern often seen in a postanoxic cerebral insult (Figs. 39.1D, 25 C. However, electrocerebral inactivity created by severe
39.2D, 39.3A, and 39.4A). However, the alpha frequency WAR hypothermia could act as cerebral protective mechanism.
pattern may be simply due to slowing of the beta frequency Hypothermia also accentuates the EEG effect of anesthetic
activity induced by anesthesia and may approach alpha or even agents (18).
theta frequency activity.
In addition to the WAR pattern, anterior maximum inter- EEG MONITORING
mittent slow (AIS) waves may appear (Figs. 39.1C and 39.2C)
(18). AIS waves are commonly diphasic having a sharply con- EEG has been used for monitoring the brain function during
toured negative–positive configuration with a duration of less CEA for many years and probably has been the most commonly
than a second occurring singly or in brief trains. used method for the detection of cerebral ischemia that would
Another pattern during this level of anesthesia is widespread determine the need for selective shunting. EEG can be recorded
persistent slow (WPS) waves (Figs. 39.1C and 39.2C) (18). The easily in the operating room despite an electrically hostile envi-
WPS waves consist of polymorphic slow waves with a duration ronment where multiple electrical instruments are connected
usually exceeding 1 second. This pattern is least obvious with to or working nearby the patients and surgical personnel. Most
halothane and enflurane and is most prominent with isoflurane EEG monitoring are performed under general anesthesia and
(18). With deepening of the anesthetic level, there is an increase its activity may vary depending on the type and depth of anes-
in the amplitude and wave length of the WPS pattern and a thesia, blood pressure, body temperature, or other physiological
decrease in WAR pattern. factors such as PaCO2 and PaO2 level. These variables should be
thoroughly understood by the monitoring team.
EEG Pattern w ith Supra-MAC Concentration
The EEG patterns of different anesthetic agents are similar when Recording Technique
the concentrations are at sub-MAC level. But the EEG patterns Because EEG changes during the CEA can be diffuse or focal,
are different when the concentration levels are above MAC. For the electrodes across the entire scalp, using 16 EEG data chan-
instance, enflurane elicits spike and wave at 1.5 MAC and a burst nels, are commonly used. It is preferable to use collodion, which
suppression pattern that may include a spike component at ensures the secure attachment of electrodes to the scalp during
MAC levels greater than 2. At these levels even seizure activity the surgery.
may be elicited when activated by hyperventilation (19). The anterior–posterior longitudinal bipolar (so-called
In contrast, isoflurane tends to exert an antiepileptic effect banana) montage is commonly used because this montage
producing an intermittent suppression pattern at 1.5 MAC and allows easy detection of hemispheric asymmetry.
continuous suppression at MAC above 2. Halothane also has an The filter setting is usually the same as of routine EEG with
antiepileptic property but may not produce EEG inactivity even the high (low pass) filter of 70 Hz. Some laboratories may
with concentrations as high as 4 MAC (18). Desflurane and choose the low (high pass) filter setting of 0.3 Hz and high (low
isoflurane tend to rapidly produce EEG patterns of suppression pass) filter setting of 35 Hz. Because of the electrically “noisy”
or burst suppression at supra-MAC levels. Adding nitrous oxide operating room environment, 60 Hz contamination onto the
to isoflurane or desflurane may potentiate the EEG suppression EEG recording may be inevitable. The monitoring team should
at a lower concentration of the primary anesthetic agent (18). try to find the source of 60-Hz artifacts and to eliminate the
Figure 39.1 EEG examples of the anesthetic effect using isoflurane and nitrous oxide. The patient is a 50-year-old woman
having a history of TIA symptoms with intermittent partial visual loss in the left eye with right extremity weakness.
A: Within 5 seconds after IV propofol injection, the EEG starts to show diffuse delta with superimposed beta activity. (This
patient had preexisting beta activity prior to the induction.) B: Subsequent polymorphic delta activity with superimposed
beta activity is seen.
(continued on next page)
Figure 39.1 (continued) This was followed by C that shows a decrease of delta activity along with the widespread ante-
riorly dominant rhythm (WAR) consisting of dominant alpha and beta activities. In addition there were occasional anterior
dominant intermittent slow (AIS) and widespread (irregular) persistent slow (WPS) patterns. D: When the anesthesia was
stabilized at sub-MAC level, AIS and WPS patterns became less frequent and less prominent, and WAR pattern dominated
throughout the procedure.
790
Figure 39.2 Another EEG example of anesthetic effect using desflurane and nitrous oxide. The EEG monitors a right CEA
in a 66-year-old man without a history of TIA or stroke but having a 70% to 80% stenosis on right and 50% to 60% steno-
sis on left carotid artery. A and B: Within 5 seconds after the IV propofol injection, the EEG started to show diffuse theta
activity mixed with beta activity, which was followed by an increase of irregular delta as well as frontal dominant rhythmic
delta activity (FIRDA) within 30 to 40 seconds after the induction. (continued on next page)
Figure 39.2 (continued) C: About a minute after the induction, delta slow waves became more dominant with irregular
delta (WPS) along with a frontal dominant intermittent delta (AIS) pattern. D: Once the anesthesia was stabilized at the
sub-MAC level, the EEG was dominated by diffuse alpha and slow beta activity with minimal theta–delta components,
resembling “alpha-coma.”
792
Figure 39.3 A 69-year-old man without a history of TIA or stroke has a 90% stenosis of left carotid and a patent right
carotid artery. A: Within several seconds after cross-clamping the left carotid artery, there was a slight reduction and slow-
ing of the alpha activity, noted more prominently in the left hemisphere. Note the dominant WAR pattern with alpha activ-
ity resembling an “alpha-coma” prior to the clamp. B: The suppression of EEG activity in the left hemisphere became more
prominent within 20 seconds after carotid clamp. This persists until the shunt was placed. (continued on next page)
794 Part V ■ Complementary and Special Techniques

Figure 39.3 (continued) Within a few seconds after the shunt was placed, the EEG started to show recovery (C,D). It
took more than 3 minutes to recover fully to the preclamp EEG (E).
Chapter 39 ■ Monitoring EEG during Carotid Surgery 795

Figure 39.3 (continued)

artifacts by technical modifications if possible. If 60-Hz Preclamp Focal EEG Abnormalities in


artifacts cannot be eliminated, the use of 60-Hz notch filter Relationship to the Anesthesia
is allowed. The majority of EEG patterns during CEA show a symmetric
The sensitivity setting is usually 7 or 5 V/mm. It is not baseline pattern before the clamping of the carotid artery.
uncommon for the EEG amplitude to be exceedingly low under However, depending on case selection, 30% to 40% of preclamp
anesthesia, requiring a sensitivity of 3 V/mm. EEG may show focal abnormality of varying degree (20). The
The sweep speed is usually a 10-second display on a full focal abnormality may consist of unilateral reduction of the
video screen but may be adjusted to 5 seconds or 20 to 30 sec- WAR amplitude. This may be accompanied by polymorphic
onds screen depending on the interpretation needs and prefer- delta activity on the side of the reduced amplitude. Most focal
ence of electroencephalographer. EEG abnormalities under anesthetic usually correlate with a
A preoperative, awake resting EEG can be recorded in the preoperative waking EEG abnormalities. However, anesthesia
operating room before anesthesia induction. This allows may activate an abnormality that was either inapparent or less
inspection of any preexisting EEG abnormalities before anes- apparent during the waking state (18). Conversely, the anesthe-
thesia. At least 5 to 10 minutes of baseline preclamp EEG is sia may obscure a focal abnormality in some cases that was
essential while the patient is under stable anesthesia to appreci- apparent in EEG recordings during the awake state.
ate any clamp-associated EEG changes. After the clamp is In patients with a lesion in the anterior head region where
applied, the EEG of the preclamp state may be displayed on half the alpha rhythm tends to be unaffected in the awake state, the
of the video screen, so that the postclamp on-going EEG can be anesthesia may bring out a reduction of the WAR pattern
directly compared with preclamp EEG. Although most clamp- along with decreased beta and increased polymorphic slowing
related EEG changes occur within 20 to 30 seconds after the in the anterior head region. Preexisting intermittent rhythmic
clamp, the EEG should be monitored throughout the procedure delta in the temporal region in the awake state may become
because EEG changes may occur in the midst of procedure, long more prominent and persistent, along with a reduction of the
after the clamp has been applied. Possible causes for the late WAR pattern (18). A more posterior lesion having decreased
EEG changes include hypoperfusion secondary to a lowered alpha activity on the side of lesion may leave the anesthetic
blood pressure or embolism, or shunt malformation if a shunt WAR pattern symmetrical without focal slowing (18).
is used. Preexisting nonlateralizing FIRDA or persistent generalized
Figure 39.4 A 77-year-old man without a history of TIA or stroke has occlusion of right carotid and an 80% stenosis of
left carotid artery. A: The EEG started to show an increase of theta and delta range activity bilaterally, which is more promi-
nently noted on the left hemisphere within 5 seconds after the clamp. B: Within 20 seconds after the clamp, the EEG
showed diffuse suppression and loss of fast activity bilaterally which was to a greater degree over the left hemisphere (B).
796
Figure 39.4 (continued) C: Within a few seconds after the shunt was placed the EEG started to recover. D: Full recovery
was achieved at about 1 minute after the shunt was placed.

797
798 Part V ■ Complementary and Special Techniques

slowing during awake state may be observed and cannot be level. Raising the blood pressure usually corrects the adverse
distinguished from the anesthesia-induced slowing that com- EEG change. Another possibility of late and persistent EEG
monly occurs during induction or during the recovery phase change may be due to embolization especially during dissection
of anesthesia (18). or at the time of clamp release which will likely result in neuro-
logic deficit.
EEG Changes after Cross-Clamping A late EEG change may also occur in shunted patient. If this
the Carotid Artery is not due to blood pressure, anesthesia, or embolization, the
Most preclamp EEG patterns have a symmetrical pattern with- shunt malfunction should be considered
out focal features and the development of an asymmetric pat-
tern after the clamp can be easily recognized. The Predictability of EEG Change
clamp-associated EEG change occurs within the first minute in The overall incidence of clamp-related EEG changes has been
the majority of patients ( 80%) with most (69%) appearing estimated to be about 10% to 30% (20–23). Most studies have
within 20 seconds after the clamp (21). The most common and agreed that the incidence of clamp-related EEG changes was
sensitive EEG change indicating cerebral ischemia is the decrease higher in patients when contralateral carotid artery was
of alpha and beta frequency activity (WAR pattern) on the occluded. When comparing patients with a patent and occluded
clamped side (Figs. 39.3A,B and 39.4A,B). Fortunately the most contralateral carotid artery in a total of 564 patients from a sin-
common EEG pattern under stable sub-MAC level of anesthesia gle institution, a significantly greater incidence of ipsilateral
consists of abundant alpha and beta activity (WAR pattern). EEG change was found in patients with an occluded artery
Further progress of ischemia is associated with an increase of (39%, 22 patients out of 57 patients) than in patients with
delta amplitude or slower frequencies . The most severe degree patent artery (16%, 80 patients out of 479 patients) showing
of EEG change is “flattening” of the EEG pattern with depres- statistically significant difference (P 0.001) (24). The greater
sion of all activity including delta and faster frequency compo- incidence of clamp-associated EEG changes was found with
nents (Figs. 39.3C and 39.4B). These changes occur primarily 90% than 50% stenosis of contralateral carotid artery (25).
on the hemisphere ipsilateral to the side of clamp but a bilateral Other studies also agreed that 30% to 40% of the patients with
change could occur if the blood flow of contralateral hemi- contralateral occlusion required shunt based on EEG findings
sphere depends on the collateral circulation from the ipsilateral (26,27).
hemisphere. This may occur when occlusion or severe stenosis Another study also reported a 48% incidence of clamp-
of the contralateral carotid artery exists. When bilateral changes related EEG change with contralateral carotid occlusion com-
occur, it may be difficult to differentiate if changes are due to pared to 18% for stenosis and 21% for normal carotid artery
global ischemia or systemic factors such as a change in anes- (28). Clamp-related EEG changes are also more common in
thetic level, blood pressure, temperature, or PaCO2/O2 level. patients when contralateral abnormalities on the preoperative
However, the bilateral change secondary to hypoperfusion usu- awake EEG exist than in patients with only ipsilateral or diffuse
ally shows a greater EEG change where more “flattening” of abnormalities (21) or patients with previous history of stroke
EEG activity occurs in the presence of slower delta and/or (29). It should be noted that there are still substantial numbers
greater depression of fast activity over the ipsilateral hemi- of patients who have a complete occlusion of the contralateral
sphere (Fig. 39.4B,C). carotid artery that do not show EEG changes when clamping of
After the shunt is placed, the EEG recovers gradually to the the ipsilateral carotid artery occurs. This is likely due to ade-
baseline state (preclamp EEG) and may take several minutes to quate collateral circulation. In such cases, the surgeon may pre-
full recovery. It appears that the longer the clamp time before fer to shunt even if there is no EEG change.
shunt placement occurs, the slower the recovery time. In a small It is intuitive to expect that the greater degree of ipsilateral
number of patients, the EEG improves but never recovers to the stenosis is associated with a lower chance of EEG change,
preclamp state. Most of these patients wake up with no neuro- because the collateral flow from the contralateral hemisphere
logic deficit. If any, the deficit is minor or transient. In the would be well established in patients with severe stenosis than
majority of shunted or nonshunted cases, the EEG remains in patients with a patent ipsilateral carotid artery. However, one
symmetric throughout the procedure. In a small percentage of study did not support this expectation (15).
patients, transient and focal EEG changes may appear during
the middle of the procedure, long after the cross-clamping has CEA RESULTS WITH AN EEG MONITORING
occurred. These transient and focal EEG changes have been FOR THE SHUNT NEED
thought due to the asymmetrical effects of changing level of
anesthesia or resurgence of preexisting focal abnormality. These There are three types of surgical preferences for CEA: one is a
transient changes have no neurologic consequence (20). This surgical protocol using shunts routinely (9,30), another proto-
finding may be due to decreased blood pressure. Because the col will not use a shunt at all (21,31), still another protocol
threshold of blood pressure to maintain the appropriate cere- places a shunt selectively by one or more monitoring techniques
bral perfusion is not known and varies individually, slight (20,32). The reported incidences of perioperative stroke were
reductions of systemic blood pressure may result in focal areas not much different among these three protocols but the overall
of ischemia bringing out EEG changes especially when the incidence tends to be highest among those using a shunt rou-
blood pressure at the time of clamp is close to the threshold tinely and the lowest among those who do not use a shunt at all.
Chapter 39 ■ Monitoring EEG during Carotid Surgery 799

This suggests that perioperative stroke are more likely due to With EEG monitoring and selective shunting, the incidence of
embolism rather than cerebral ischemia, and thus the necessity stroke appeared to be lower than those without monitoring.
of any type of monitoring can be questioned. Only a large-scale, One study reported that the incidence of stroke was 2.19% in
prospective study in a single institution can make accurate nonmonitored/nonshunted group versus 0.32% in selective
comparisons of these three methods of surgery. Nonetheless, shunting based on EEG (P 0.05) (15). Other studies using
many studies agree that CEAs without shunt appear to be asso- EEG monitoring reported 0.4% (35) and 0.03% (23) stroke
ciated with a substantially higher risk of stroke than CEAs with- rate. Another recent study also reported that selective shunting
out selective shunting (15,23,33). In a retrospective review of based on EEG and SSEP findings resulted in lower stroke rate
902 consecutive CEAs from a single institute, 591 CEAs were of 1% (2 out of 194 patients) in selective shunted group com-
performed without shunting and also without EEG monitor- pared with 4% (47 out of 1217 patients) of routinely shunted
ing. This was compared to 311 CEAs that were done with selec- group (36).
tive shunting based on the EEG findings (15). This study The incidences of perioperative stroke with and without the
reported that clamp-related EEG changes occurred in 40 presence of contralateral carotid occlusion or stenosis have
patients (12.8%) and a postoperative stroke occurring in only been in dispute. In a large-scale study involving 1661 CEAs, the
one patient (0.32%) in the selective shunted group, whereas in statistically significant increases of stroke rate were associated
the nonshunted group (no EEG monitoring), 13 patients with contralateral carotid occlusion (1.8%) while the overall
(2.19%) had postoperative stroke (P 0.05). incidence of stroke rate was 0.03% (23). In this study, the
In another large-scale study involving 1661 CEAs, EEG stroke rate was also higher in patients who had EEG change
changes were noted in 366 procedures (22%) (23). A perioper- (1.1%) and greatest in patients who had both EEG as well as
ative stroke occurred in five patients (0.03%). Of the five contralateral occlusion (3.3%). Another study also reported
patients, four had positive EEG finding and only one had false- the increase of stroke rate from 1.8% to 7.8% with contralat-
negative EEG finding. The authors concluded that EEG moni- eral occlusion and 1.9% to 4.0% with history of stroke (37). A
toring accurately identified the patients who did not need third study also reported a greater incidence of perioperative
shunting. One study reports less satisfactory results of EEG stroke in patients with than without history of stroke (3% vs.
monitoring, in which 70 patients (15%) of 458 CEAs showed 0.3%) (27). However, there were studies reporting no differ-
EEG changes and received shunts (29). In this study, 15 patients ence in stroke rate irrespective of presence or absence of
were found to have immediate stroke or transient neurologic carotid occlusion (24,38).
deficits. Ten of these 15 patients showed no EEG changes after
the clamp. In the remaining five patients, EEG changes
occurred but resolved after the shunt placement or raising LONG-TERM EEG CHANGE AFTER CEA
blood pressure. All 15 EEG studies were re-reviewed and veri-
Thus far little attention has been paid to the long-term outcome
fied by an experienced electroencephalographer. of EEG after the successful CEA. Besides preventing stroke, CEA
One of the reasons for a false-negative EEG finding is due has been reported to improve cerebral circulation (39–41) and
to inability to detect subcortical/capsular or small cortical cognitive brain function (42). In 28 patients with symptomatic
ischemia by visual inspection of the EEG. In this sense, high-grade carotid stenosis, quantitative EEG (q-EEG) showed
comonitoring SSEP may be useful because SSEP can detect an increased mean frequency 1 Hz (P 0.01) postopera-
capsular ischemia. Another reason may be due to embolic tively. The Mini mental test and Set Test for verbal frequency
stroke, which may occur in the recovery room after EEG mon- had a positive correlation with the q-EEG changes (43).
itoring has ended. The EEG monitoring also showed a consid- Another study also showed an increased mean frequency of the
erably high incidence of false-positive findings. Of 176 CEA alpha band and peak frequency of the alpha after CEA in 166
patients with EEG monitoring but without shunt, postopera- patients with 70% stenosis of the internal carotid artery (44).
tive stroke occurred in only 2 (9%) of the 22 patients with In general, patients with occluded contralateral carotid artery
clamp-related major EEG changes and none of the 33 patients improved more than other patients.
with moderate EEG changes (21). Another similar study (with
EEG monitoring but without shunt use) also reported a low
incidence of perioperative stroke associated with positive EEG REGIONAL ANESTHESIA FOR AWAKE CEA
changes; of 72 CEAs with moderate or major EEG changes AND EEG CORRELATES
from a total of 293 CEAs, only 6 (8%) of them resulted in
postoperative stroke (34). The advantage of regional anesthesia (RA) instead of general
anesthesia is that the patient’s neurologic status can be assessed
THE INCIDENCE OF STROKE AFTER CEA during the cross-clamping period. RA can be performed in
most patients, if so elected, unless a significant contraindication
An intraoperative stroke rate was reported to be 2.1% in the exist such as patient’s refusal, cognitive limitation, anxiety, or
North American Symptomatic Carotid Endarterectomy Trial language barrier. RA is accomplished by superficial cervical
(1) study, in which no uniform method of intraoperative cere- block by local anesthetics. Occasionally small doses of intra-
bral monitoring or protection was used. An overall incidence venous narcotic or midazolam may be used, but the patient
of perioperative stroke rate was reported to be 2% to 6% (8). must remain awake and lucid during the procedure.
800 Part V ■ Complementary and Special Techniques

A relatively simple neurologic examination involving speech, ANESTHESIA-INDUCED EEG BURST


sensory and motor and general cognitive functions can be SUPPRESSION PATTERN FOR CEAS
tested when the patient is awake during the procedure. Perhaps
this is the most reliable method to determine the necessity of The barbiturate-induced burst suppression has been known to
shunt placement. protect against cerebral ischemia by decreasing cerebral metab-
Based on EEG findings, the rate of shunt requirement under olism during carotid clamping (52–54). One of the earliest stud-
general anesthesia ranges from about 10% to 30% (15,45–47). ies using barbiturate for cerebral protection during CEAs
This rate is substantially higher than that of RA with the involved seven patients whose EEG changes after cross-clamping
neurologic examination in an awake patient, which is about 5% could not be corrected by raising systolic blood pressure (55).
to 15% (13,46–49). Does this mean that the EEG under general They were placed in a barbiturate-induced burst-suppression
anesthesia is too sensitive to warn the ischemic change, result- pattern with suppression intervals of 15 to 30 seconds. Despite
ing in excessive false-positive finding? significant clamp-related EEG changes including the one with
However, studies comparing EEG findings with cases using occluded contralateral carotid artery showing severe depression
neurologic examination in an awake patient during CEA of EEG activity, none of these seven patients developed neuro-
showed results contrary to this notion. Of 89 patients who logic deficit.
underwent awake CEA with EEG monitoring (45), the con- Disadvantages of barbiturate-induced burst suppression
cordance rate of both normal EEG and neurologic examina- include hemodynamic instability such as intraoperative
tion was 76.4% (68 patients) and the concordance rate of both hypotension and prolonged anesthesia effect after surgery,
abnormal EEG and neurologic examination was 12.4% which makes early postoperative neurologic evaluation diffi-
(11 patients). The total concordance rate was thus 88.8%. The cult. Instead of barbiturate, the more recent studies used
remaining 12.2% (10 patients) showed a discrepancy between Propofol to induce an EEG burst suppression pattern. Unlike
the EEG and the neurologic examination. Abnormal EEG with barbiturate, Propofol does not impair the autoregulation of the
normal examination, that is, false-positive finding, was seen in hemodynamic state. Also recovery from anesthesia is quicker
6.7% (6 patients) and normal EEG with abnormal neurologic than for barbiturates. Using a propofol-induced burst suppres-
examination, that is, false-negative finding, was seen in 4.5% sion pattern, one study reported one major stroke, one postop-
(4 patients) (50). Another study showed a much higher false- erative death due to myocardial infarction, and two patients
negative rate of 40.6% where only 19 (59.4%) of 32 patients with TIA (transient ischemic attack) out of 100 consecutive
showed concordant abnormalities in both EEG and neuro- CEA patients (56). In another study from 102 consecutive
logic examination (51). The false-positive findings were found CEAs, etomidate-induced burst suppression was used in 20
in three patients (1.0%) in this study. Another study also patients who had EEG changes but without shunt. Of these 20
reported a high incidence of false-negative EEG changes; of patients, only one patient developed perioperative stroke (57).
134 awake CEAs, 13 patients required shunt based on neuro- A larger population study is needed to validate the effects of
logic changes but 4 (30.5%) of these did not show EEG deep anesthesia to the point of burst suppression pattern on
changes (48). Increased false-negative results for EEG in EEG as a protective measure against brain ischemia.
awake CEAs may be due to some difficulty in assessing awake
patients, because the EEG recording may have the extra noise
of increased muscle or other artifacts, and at times low- OTHER MONITORING METHODS FOR CEAS
amplitude alpha rhythm or exceedingly low-amplitude back- IN RELATIONSHIP WITH EEG
ground activity in awake patients who are anxious. The most
sensitive EEG parameter is beta activity, which is commonly Cerebral Oxygen Saturation Measurement
seen in general anesthesia. This may not be present in the Cerebral oximetry using near-infrared spectroscopy measures
awake patient (Figs. 39.5A through D). Also some false- SaO2 saturation. Two sensors are placed on the right and left
negative EEG monitors may be due to the inability to detect forehead. The decrease of cerebral oxygen saturation is reported
abnormalities when ischemia or hypoperfusion is limited to to be accompanied by a significant decrease in the mean middle
capsular or small cortical/subcortical region. One study raised cerebral artery blood velocity, even with minimal change of the
a possibility that the awake state may make brain less vulner- SaO2 (58). A fall of 5% or more in cerebral oxygen saturation
able to hypoperfusion and cerebral ischemia than anesthetic following cross-clamping application was accompanied by a
state (50). This appears to contradict the general assumption decrease in mean middle cerebral artery blood velocity of at
that anesthesia, especially deep anesthesia with burst suppres- least 60% (58).
sion, is protective from cerebral ischemic by decreased metab- An earlier study showed fairly good correlation between
olism. However, under general anesthesia, the relationship change in SaO2 saturation and SSEP (59) and/or EEG changes
between EEG and postoperative neurologic deficit appeared (60). The decrease of cerebral oxygen saturation was also
to be opposite since more false-positive ( 90%) than false- correlated with neurologic symptoms in awake CEA
negative ( 10%) EEG findings are noted (21,34) (see also patients (61). Overall cerebral oxygen saturation less than 60%
“Incidence of Stroke”). These two studies suggested that the and/or a greater than 15% to 20% decrease in SaO2 from the
EEG under general anesthesia is overly sensitive for predicting preclamp baseline appeared to correlate with a prediction of
cerebral ischemia. ischemic compromise using EEG as a gold standard (61,62).
Chapter 39 ■ Monitoring EEG during Carotid Surgery 801

Conversely, cerebral oxygen saturations greater than 61% with Transcranial Doppler (TCD)
desaturations less than 6.9% to 7.3% of the preclamp baseline TCD monitoring of middle cerebral artery flow velocity can
was considered to be in a safe range (62). More recent studies, also assess the dependence of the ipsilateral hemisphere on the
however, showed poorer concordance between SaO 2 and middle cerebral artery blood supply (25,73,74). Stenosis or
EEG/SSEP findings. Of 323 CEAs under general anesthesia, 24 occlusion of the contralateral carotid artery was associated with
patients (7.4%) showed significant discrepancies between SaO2 a large middle cerebral artery velocity decrease upon clamping
and EEG/SSEP measurement (63); 16 patients showed no sig- (25). TCD can also detect signals representing embolic flow
nificant EEG/SSEP change but profound changes occurred in after clamp release (73). Increased numbers of emboli on
SaO2, and seven patients showed no change in SaO2 but pro- closure may warn of impending luminal thrombosis (75). TCD
found change in EEG/SSEP. The sensitivity of SaO2 compared may also be useful to identify postoperative risk of hyperperfu-
with EEG/SSEP was 68% and the specificity was 94%. This gave sion syndrome (76). Using a 75% reduction of mean blood flow
a positive-prediction value of 47% and negative-prediction of middle cerebral artery as the threshold for shunt placement,
value of 98% (63). This study concluded that relying on SaO2 one study showed one true-positive and four false-positive and
alone for selection of shunting is potentially dangerous. no false-negative results from 91 patients (77). In this study
Another recent study also showed positive prediction of 33% EEG yielded one false-negative and no false-positive results,
and negative-prediction value of 100% (64). Other studies also and SEP had no false-positive or false-negative results. Another
discourage the use of SaO2 as a sole guide for determining shunt study also showed higher false-positive results in TCD (78). Of
placement (65,66). The overall threshold for the identification 85 patients, 13 showed a drop of 70% and only one of them
of ischemia has been difficult to define (59). required shunt based on EEG and SSEP. The authors concluded
that using a drop of 70% or more of the systolic blood flow
Carotid Stump Pressure (CSP)
velocity in the middle cerebral artery during internal carotid
Measurement of stump pressure was one of the earliest tech- artery cross-clamping would lead to excessive unnecessary
niques used in an attempt to measure the adequacy of collateral shunting. On the other hand, another study reported a dra-
flow after carotid clamping. The residual pressure in the distal matic reduction of TCD in association with normal EEG results
portion of the internal carotid system following temporary in three patients with a resulting transient postoperative neuro-
occlusion of the common and external carotid arteries has been logic deficit (79).
thought to reflect arterial pressure through the circle of Willis and Overall TCD appears to show more false-positive finding for
thus to indicate the efficacy of collateral blood supply to the the prediction of ischemia. Another problem of TCD is the rela-
hemisphere of the occluded vessel (67). Although earlier reports tively high rate of unsuccessful recordings ranging from 20% to
doubted its usefulness (20,68), later studies obtained close corre- 40% (72,77). Perhaps TCD is more useful in detecting embolic
lation of CSP with EEG changes to indicate the patency of the and cerebral hyperperfusion after endarterectomies (80,81).
contralateral carotid artery. For example, one study reported EEG
changes in 58% of cases where CSP was 25 mm Hg, 32% of Xenon-133 Washout Study
cases with CSP between 25 and 50 mm Hg, and only 4% of cases The xenon washout study is an accurate method of measuring
when CSP exceeded 50 mm Hg (69). Based on neurologic exam- cerebral blood flow. Xenon-133 is injected into either internal
ination in 474 patients with awake CEA, another study showed carotid artery above the plaque or the common carotid artery
false-negative rate requiring shunt placement was only 1.0% below the plaque with external carotid artery occlusion. EEG
using 40 mm Hg systolic as a threshold for the need of shunting changes have been found to occur with blood flow at or below
(70). If these 474 CEAs had been performed using general anes- 18 to 20 mL/100 g brain tissue/min (20). The method, however,
thesia the shunt determination rate based on CSP would have is not widely available and is less commonly used for CEAs.
been 29% of patients for CSP 50 mm Hg and 15% of patients
for CSP 40 mm Hg. A more recent and larger scale study (1135 Somatosensory-Evoked Potential (SSEP)
patients) concluded that 45 mm Hg CSP was a reliable predic- SSEP is dependent upon the integrity of sensory pathways start-
tion of adequate cerebral perfusion (71). In another study of 48 ing from the peripheral nerve to the sensory cortex via the
patients undergoing awake CEA, CSP provided similar accuracy internal capsule and thalamus. Unlike EEG that is not sensitive
with transcranial Doppler sonography and near-infrared spec- to detect small subcortical ischemia, SSEP can reflect ischemic
troscopy for the detection of cerebral ischemia (72). However, in changes occurring in small areas of the capsular region, which
another study of 100 patients under general anesthesia, 34 is important for neurologic function. However, one should real-
patients had CSP 50 mm Hg and only 4 of these needed shunt- ize that the area covered by SSEPs is relatively small as com-
ing based on EEG change (64). Conversely 2 of 66 patients who pared to EEG.
had SP 50 mm Hg, 2 patients needed shunting. If a shunting One of the disadvantages of SSEP is the technical challenge
policy has been used on a CSP of 50 mm Hg, 30 patients would of recording such small potentials in an electrically hostile
have been shunted unnecessarily (positive predictive value of environment common to an operating room. One encounters
12%), whereas CSP 50 mm Hg would determine that a shunt technical difficulties more often with SSEP recordings than
was not required in 64 of 66 patients (negative predictive value of with EEG recording. Electrical noise or artifacts are relatively
97%). The authors concluded that CSP should not be used as the easy to recognize in the EEG recording because of its continu-
sole predictor for shunting for CEA (64). ous nature. Artifacts or electrical noise that are often difficult
Figure 39.5 An 83-year-old woman with history of stroke and TIAs affecting the left hemisphere has a 90% stenosis of
the left carotid artery. CEA was performed during the awake state with regional anesthesia. A: The EEG showed preexisting
delta activity over the left hemisphere. B: Within 30 seconds after the clamp, there were increases of delta slow waves and
decreases of alpha-theta background activity over the left hemisphere.
802
Chapter 39 ■ Monitoring EEG during Carotid Surgery 803

Figure 39.5 (continued) C: This persisted until a shunt was placed. Despite obvious EEG changes, the patient remained
asymptomatic during the clamp period. D: Final recovery was achieved about 3 minutes after the shunt was placed.
804 Part V ■ Complementary and Special Techniques

to identify can contaminate SSEP responses. These artifacts in express the power spectrum amplitude of a range of frequen-
turn can change the response making it appear as if real cies along a time axis. The X-axis is designated as the time
pathology is present. Unlike EEG that shows changes instanta- scale and the Y-axis expresses both the power scale by color-
neously, SSEP recordings require averaging the responses. coded differential or gray scale differential and frequency by
Thus, there is a delay of a few minutes to yield one response height along the Y-axis. The SEF is expressed as a percentage
and an additional few minutes delay to verify the response of frequency power at 90% to 97% of total power and is
changes. Another disadvantage of SSEP is that the recording defined to be the highest frequency component of the EEG
often cannot be performed in the patients who have peripheral visible in the current spectrum. Using DSA and SEF on a sin-
neuropathy or impaired peripheral nerve conduction of the gle channel of EEG (without conventional EEG recording and
upper extremities, which is not uncommon in the elderly without shunt placement irrespective of q-EEG findings), one
patient’s population. Nonetheless, SSEP monitoring has shown earlier study showed all 7 out of 70 neurologically intact
favorable results for the purpose of monitoring to detect cere- patients preoperatively who had the changes in SEF developed
bral ischemia. postoperative neurologic deficit (84). The criteria used
Comparing the SSEP, EEG, and TCD in 156 patients, EEG included a decrease in high frequencies by SEF lasting longer
yielded one false-negative and no false-positive results, and than 10 minutes where the amplitude was less than 50% of its
TCD showed four false-positive and no false-negative results, average value over the preceding moments. In the same study,
whereas SEP had no false-positive or false-negative results (77). however, 31 patients who had preoperative neurologic deficit
The criteria used for determining the need for shunting was showed one false-positive and one false-negative result using
greater than 50% amplitude reduction of the N20/P25 potential the same criteria.
using median nerve SSEP. Using the same criteria, another One study compared visually inspected EEG with q-EEG
study also supported the SEP to be most reliable in warning of analyses, which included total power, SEF, spectral variance, and
ischemia, as compared to TCD or spectral edge frequency log spectral variance. Overall q-EEG showed significant changes
analysis of EEG (82). The same criteria could be used in only in patients who had severe degree of slow waves (continu-
patients who had history of previous stroke (83). ous delta or persistent amplitude attenuation, or both) that was
SSEP monitoring in addition to EEG may indeed help to evident visually as an EEG abnormality (85). Of the various
identify the subcortical ischemia not detected by EEG, which in q-EEG parameters, total power was most sensitive and SEF was
turn would promote the accuracy of warning for cerebral least sensitive in correlating with EEG findings. Another study
ischemia. also reported that the total power spectrum was more sensitive
than SEF as an indicator of cerebral ischemia (86). In another
Quantitative EEG Analysis (q-EEG) study, two electroencephalographers independently interpreted
The visually assessed EEG changes indicative of cerebral DSA and conventional EEG from 103 CEA patients under gen-
ischemia are reflected by a reduction or amplitude decrease of eral anesthesia (87). One interpreted the significant DSA
fast activity and an increase of delta/theta bands activity. The change in 21 of 29 patients (sensitivity of 72% and specificity of
reliable assessment and the accuracy are dependent on the skill 96%). The other identified significant DSA change in 16 of
and experience of electroencephalographers. The type, magni- 27 patients (sensitivity of 59% and specificity of 96%). Failure
tude, and duration of EEG changes needed to accurately predict of DSA to detect potential ischemia was greater in patients who
cerebral ischemia have not been clearly established. Thus the had mild degrees of EEG change. The authors concluded that
precise EEG criteria for shunt requirement are still a matter of DSA did not reliably detect mild EEG pattern changes indica-
debate. Also the reasons for false-positive or false-negative EEG tive of cerebral ischemia and is not a reliable substitute for
results have not been clarified. 16-channel conventional EEG (87).
Since most EEGs are now performed by digital recording Separating fast (factor 1: beta and alpha) and slow (factor 2:
techniques, various quantitive EEG (q-EEG) analyses using theta and delta) band frequencies obtained slightly better
appropriate algorithms are possible, either on- or off-line. results of power spectrum analysis (88). Decrease of factor 1
These include power spectrum frequency analysis, spectral edge and increase of factor 2 was considered indicative of cerebral
frequency parameter, brain symmetric index (BSI), bispectral ischemia. Minor ischemia was distinguished by decrease of fac-
index profile (BIS), and brain mapping. It is hoped that q-EEG tor 1. Of 16 patients who had significant decrease of factor 1,
will provide a more accurate and objective measure to reflect 15 patients correlated with EEG that showed a visual change
the subtle EEG changes indicative of cerebral ischemia with the and were subsequently shunted. The factor 2 was, however, not
goal of identifying when to use a shunt with more accuracy. revealing. Another study measured the 8 to 15 Hz band power
This, in turn, is expected to reduce the false-positive and false- before and after the carotid clamp and calculated the reduction
negative EEG finding for the prediction of perioperative neuro- ratio of this band power after the clamp that reflected the
logic deficit. degree of power change and designated it as “the desynchro-
A fast Fourier transform (FFT) is used to generate spectral nization index” (D-index) (89). In 47 consecutive patients with
power of specified or pre-selected frequency bands. The den- CEAs, the major EEG changes (11 patients) found were severe
sity spectral array (DSA) and/or spectral edge frequency (SEF) depression or loss of 8- to 15-Hz activity and/or at least
derived from FFT data have been used as monitors during twofolds increase of slow delta ( 1 Hz) noted on visual inspec-
CEA. The DSA is a three-dimensional data display designed to tion. This was consistently associated with a D-index of greater
Chapter 39 ■ Monitoring EEG during Carotid Surgery 805

than 65% (mean of 76.85%). The authors concluded that the of EEG were reflected by changes on the temporal symmetry
D-index of greater than 65% correctly identified the patient measure with tBSI greater than 0.02. A large-scale clinical study
with cerebral hypoxic risk. The moderate EEG changes is needed to find out if the above BSI parameters accurately pre-
(4 patients) with decreased but persisting 8- to 15-Hz activity dict cerebral ischemia or provide more sensitive information
and/or persistent increase of delta activity ( 1 Hz) were asso- with respect to the criteria for shunt requirement.
ciated with mean D-index of 40.23%. Because two patients of
this group awoke without any neurologic deficit despite no Computed Topographic Brain Mapping (CTBM)
shunting used, the potential risk of D-index reduction of about CTBM is based on power/amplitude spectrum derived from
65% to 50% could not be determined. Fourier transform measured at multiple scalp electrodes. This is
Comparing the difference of power spectrum before and displayed on two-dimensional scalp figure. There have been a
after cross-clamping appears to show a better result in distin- few studies claiming that CTBM is superior to 16-channel stan-
guishing between shunt and nonshunt group (90). However, dard EEG in detecting cerebral ischemia.
there were some differences in sensitivity between two anesthet- One study involving 46 consecutive CEA patients showed
ics; the difference of absolute power spectrum was better when that CTBM changes secondary to ischemia were noted in
propofol anesthesia was used and SEF 90% was a better moni- 23 patients, whereas visually inspected conventional EEG
tor when isoflurane anesthesia was used. showed changes only in 13 patients (100). The ischemic
change detected by CTBM was significantly greater in
Bispectral Analysis (BSA) patients with previous stroke (6 out of 7 patients) than the
The power spectral analysis quantifies only power distribution as patients without stroke (17 out of 39 patients). CTBM
a function of frequency, ignoring phase information. It also changes were also correlated with average stump pressure of
assumes that the signal arises from a linear process and ignores less than 38 mm Hg, compared to 57 mm Hg in patients
any nonlinear process of interaction between the components of without CTBM change (P 0.05). Overall, 51 of 138 CTMB
a signal that are manifested as phase coupling. BSA is a signal- studies (37%) showed “abnormalities,” whereas standard
processing technique that determines both EEG linear (frequency EEG showed abnormality in only 23 studies (17%). The
and power) and nonlinear components (harmonics) which authors concluded that CTBM was much more sensitive
quantitates the interfrequency phase coupling of EEG signals. than the 16-channel standard EEG in showing electrophysi-
BSA has primarily been used by anesthesiologists to monitor ologic changes, particularly during times of carotid cross-
the depth of anesthesia by placing a few electrodes over the fore- clamping.
head (Fp1, Fp2, F3, F4) usually referenced to the Cz electrode Another CTBM study for CEA showed that 10 patients
(88,91,92). Earlier studies suggested potential usefulness of BSA (15%) of 65 CEAs had ischemic change after cross-clamping
as an application for CEA to determine the adequacy of cerebral (101). The changes were resolved after shunt placement in all
perfusion (93,94). However, the more recent studies have patients. This study also showed a higher incidence of ischemic
doubted the reliability of BSA in detecting cerebral ischemia. change in patients who had abnormal preoperative CTBM as
Of the 52 patients monitored with BSA during awake CEA, compared to those with normal CTBM (102).
5 patients showed clinical evidence of cortical ischemia during Although both studies claimed that CTBM was more sensi-
cross-clamping of the carotid but there was no change in BSA tive for selective shunting than EEG and predicted more accu-
value in these five patients (95). In another recent study, BSA rately postoperative neurologic deficits, no specific and
increased in 47%, decreased in 25%, and unchanged in 28% of objective criteria for selective shunting was provided. Also the
36 patients tested during first 3 minutes after cross-clamping of number of patients involved in these studies were small. A
the carotid (96). Because of this paradoxical increase of BSA in larger scale study is needed for further validation of CTBM as a
some patients, the authors suggested that caution should be used monitor for CEA.
in the interpretation of BSA value during CEA.
Summary of q-EEG Assessment
Brain Symmetric Index (BSI) It is hoped that q-EEG will reveal the EEG change not detected
The BSI is determined by the relative difference of the average by visual inspection and more accurately predicts cerebral
spectral density of the right and left hemispheres in the fre- ischemia. However, none of the q-EEG parameters studied so
quency range from 1 to 25 Hz. The perfect symmetry of all far have shown greater accuracy than conventional visually
change is 0, whereas BSI = 1 indicates maximum asymmetry. inspected EEG in predicting cerebral ischemia, that is, no false-
The authors who introduced BSI found that a change in the BSI positive or negative finding for perioperative neurologic
smaller than 0.03 during cross-clamping corresponded with no deficits. Another shortcoming of q-EEG is the artifact contam-
change in visual EEG analysis and a change of BSI equal to or ination, which cannot be readily identified and may lead to
greater than 0.06 showed EEG change that warranted shunting erroneous conclusions if based solely on q-EEG data. Although
(97). Because BSI depends only on the asymmetry of the EEG the above discussed q-EEG parameters may assist in the visual
spectrum spatial brain symmetric index (sBSI) and is insensi- assessment of raw EEG data for selective shunting in CEA, at
tive for temporary changes in the EEG, the same author intro- present none of the q-EEG parameters can replace the visual
duced temporal brain symmetry index (tBSI) (98,99). The inspection of 16-channel EEG interpreted by a qualified elec-
authors found that diffuse changes noticed by visual inspection troencephalographer.
806 Part V ■ Complementary and Special Techniques

APPENDIX 6. Norris JW, Zhu CZ, Borenstein NM, et al. Vascular risks of asymp-
tomatic carotid stenosis. Stroke. 1992;22:1485–1490.
CEA and Carotid Angioplasty and Stenting (CAS) 7. Rutgers DR, Klijn CJM, Kappelle, et al. Sustained bilateral hemo-
More than 10 years has passed since the introduction of CAS dynamic benefit of contralateral carotid endarterectomy in patient
with symptomatic internal carotid artery occlusion. Stroke. 2001;
and it continues to grow. The surge of CAS has dramatically
32:728–734.
reduced the request of EEG monitoring for carotid surgery in 8. Findlay JM, Marchak BE, Pelz DM, et al. Carotid endarterectomy:
many EEG laboratories. The advantages of CAS are less costly a review. Can J Neurol Sci. 2004;31:22–36.
than CEA and can be applied to the patients having high cervi- 9. Giannotta SL, Dicks RE, Kindt GW. Carotid endarterectomy:
cal and surgically inaccessible stenosis, or patients who have technical improvements. Neurosurgery. 1980;7:309–312.
significant medical conditions that would contraindicate CEA 10. Ott DA, Cooley DA, Chapa L, et al. Carotid endarterectomy with-
performed under general anesthesia such as recent myocardia out temporary intraluminal shunt. Ann Sug. 1980;191:708–714.
infarction, unstable angina, or uncontrolled congestive heart 11. Baker WH, LiHooy FN, Hayes AC, et al. Carotid endarterectomy
failure. without a shunt: the control series. J Vasc Surg. 1984;1:50–56.
In a recent large-scale multicenter randomized study 12. Ferguson GG. Intra-operative monitoring and internal shunt: are
involving 3182 patients, CAS seemed to carry a slightly higher they necessary in carotid endarterectomy? Stroke. 1982;13:287–289.
13. Steed DL, Peitzman AB, Grundy BL, et al. Causes of stroke in
risk for stroke within 30 days after the procedure as compared
carotid endarterectomy. Surgery. 1982;92:634–641.
with CEA (8.2% vs. 6.2%, P = 0.04), whereas the rates of non- 14. Riles TS, Imparato AM, Jacobowitz GR, et al. The cause of periop-
fatal myocardial infarction or death within 30 days did not erative stroke after carotid endarterectomy. J Vasc Surg. 1994;19:
differ significantly (P = 0.47) (103). Another large-scale study 206–216.
of randomized controlled trials from 3182 patients showed 15. Pletis KA, Loubser P, Mizrahi EM. Continuous electroencephalo-
that CAS had no significant increase of any stroke or major graphic monitoring and selective shunting reduce neurologic mor-
disabling stroked compared to CAE (104). The risks of death bidity rate in carotid endarterectomy. J Vasc Surg. 1997;25:620–628.
and nonfatal myocardial infarction were the same between 16. Dogle PW, Cole JP, Leary TM, et al. A comparison of remifentanil
CAE and CAS. The large scale of asymptomatic patients is not and fentanyl in patients undergoing carotid endarterectomy. Eur
available, but one study of 326 patients showed no statistical J Anesthesiol. 2001;18(1):13–19.
difference between CAS and CEA: stroke 2 patients (1.7%) 17. Laman DM, vander Reijden CS, Wieneke GH, et al. EEG evidence
for shunt requirement during carotid endarterectomy: optimal
versus 2 patients (1%), 2 myocardial infarction (1.7%) versus
EEG derivation with respect to frequency bands and anesthetic
3 (1.5%) and 1 death (0.8%) versus 0 death, respectively regimen. J Clin Neurophysiol. 2001;18(4):353–363.
(104). Recent long-term follow-up study comparing CEA and 18. Blume WT, Sharbrough FW. EEG monitoring during carotid
CAS revealed that significantly higher rate of re-stenosis endarterectomy and open heart surgery. In: Niedermeyer E, Lopes
( 70%) (6 of 32 vs. 0 of 29) occurred after CAS compared de Silva F, eds. Electroencephalogrphy, Basic Principles, Clinical
with CEA (105). Large-scale multicenter trials are needed for Application and Related Fields. 5th ed. Philadelphia: Lippincott
more accurate assessment of asymptomatic patients and long- Williams & Wilkins; 2005:815–828.
term follow-up after CAS. In recent years, there has been an 19. Stockard JJ, Bickford RG, Myers RR. The neurophysiology of anes-
increasing trend for the use of CEA, perhaps due to more thesia. In: Gordon E, ed. A Basis and Practice of Neuroanesthesia.
careful selection of CAS candidates. Amsterdam: Excepta Medica; 1975:3–46.
20. Sundt TM, Sharbough FW, Piepgras DG, et al. Correlation of cere-
bral blood flow and electroencephalographic changes during
REFERENCES carotid endarterectomy. Mayo Clin Proc. 1981;56:533–543.
21. Blume WT, Ferguson GG, McNeill DK. Significance of EEG
1. North American Symptomatic Carotid Endarterectomy Trial changes at carotid endarterectomy. Stroke. 1986;17:891–897.
Collaborators. Beneficial effect of carotid endarterectomy in 22. Chiappa KH, Burke SR, Young RR. Results of electroencephalo-
symptomatic patients with high-grade carotid stenosis. N Eng graphic monitoring during 367 carotid endarterectomies. Stroke.
J Med. 1991;325:445–453. 1979;10:381–388.
2. Europian Carotid Surgery Trialists’ Colloboration Group. MRC 23. Pinkerton JA Jr. EEG as a criteria for shunt in carotid endarterec-
Europe Carotid Surgery Trial: interim results for symptomatic tomy. Ann Vasc Surg. 2002;16(6):756–761.
patients with severe (70%–99%) or with mid (0%–29%) carotid 24. Schneider JR, Droste JS, Schendler N, et al. Carotid endarterec-
stenosis. Lancet. 1991;337:1235–1243. tomy with routine electroencephalography and selective shunting:
3. Barnett HJM, Meldrum H, Eliaziw M, et al. Treatment of sympto- influence of contralateral internal carotid artery occlusion and
matic arteriosclerotic carotid artery disease. In: Barnett HJM, utility in prevention of perioperative strokes. J Vas Surg.
Bogousslavsky J, Meldrum H, eds. Advances in Neurology Ischemic 2002;35(6):1114–1122.
Stroke, Vol. 92. Philadelphia: Lippincott William & Wilkins; 25. Arnold M, Sturzenegger M, Schaffler L, et al. Continuous intraop-
2003:307–317. erative monitoring of middle cerebral artery blood flow velocities
4. Hobson RW, Weiss DG, Fields, WS, et al., for the Veterans Affairs and electroencephalography during carotid endarterectomy. A
Cooperative Study Group. Efficiency of carotid endarterectomy of comparison of the two methods to detect cerebral ischemia.
asymptomatic carotid stenosis. N Eng J Med. 1993;328:221–227. Stroke. 1997;28(7):1345–1350.
5. Executive Committee for the Asymptomatic Carotid Athero- 26. Whittemore AD, Kauffman JL, Kohler TR, et al. Routine electroen-
sclerosis Study. Endarterectomy for asymptomatic carotid artery cephalographic (EEG) monitoring during carotid endarterectomy.
stenosis. JAMA. 1995;273:1421–1428. Ann Surg. 1983;197:707–713.
Chapter 39 ■ Monitoring EEG during Carotid Surgery 807

27. Green RM, Messick WJ, Ricotta JJ, et al. Benefits, short-comings, 47. Lawrence PF, Alves JC, Jicha D, et al. Incidence, timing and causes
and costs of EEG monitoring. Ann Surg. 1985;201:785–792. of cerebral ischemia during carotid endarterectomy with regional
28. Cherry KJ, Roland CF, Hallett JW Jr, et al. Stump pressure, the con- anesthesia. J Vasc Surg. 1998;27:329–337.
tralateral carotid artery, and electroencephalographic changes. Am 48. Evans WE, Hayes JP, Waltke EA, et al. Optimal cerebral monitoring
J Surg. 1991;162(2):185–188. during carotid endarterectomy: neurologic response under local
29. Kresowik TF, Worsey MJ, Khonry MD, et al. Limitations of elec- anesthesia. J Vasc Surg. 1985;2:775–777.
troencephalographic monitoring in the detection of cerebral 49. Santamaria G, Birtti RD, Tescione M, et al. Comparison between
ischemia accompanying carotid endarterectomy. J Vasc Surg. local and general anesthesia for carotid endarterectomy: a retro-
1991;13:439–443. spective analysis. Minerva Anesthesiol. 2004;72(12):1013–1014.
30. Thompson JE, Austin DJ, Patman RD. Carotid endarterectomy for 50. Illig KA, Sternbach Y, Zhang R, et al. EEG changes during awake
cerebrovascular insufficiency: long-term results in 592 patients carotid endarterectomy. Ann Vasc Surg. 2002;16:6–11.
followed up to thirteen years. Ann Surg. 1970;172:663–679. 51. Hans SS, Jareupoon O. Prospective evaluation of electroen-
31. Whitney DG, Kahn EM, Estes JW, et al. Carotid artery surgery cephalography, cerebral artery stump pressure and neurologic
without temporary indwelling shunt: 1917 consecutive proce- change during 314consequitive carotid endarterectomies per-
dures. Arch Surg. 1980;115:1393–1399. formed in awake patients. J Vasc Surg. 2007;45(3):511–515.
32. Ojemann RG, Crowell RM, Robertson GH, et al. Surgical treat- 52. Kassell NF, Hitchon PW, Gerk MK, et al. Alteration in cerebral
ment of extracranial carotid occlusive disease. Clin Neurosurg. blood flow, oxygen metabolism and electrical activity produced by
1975;22:214–263. high dose sodium thiopental. Neurosurgery. 1980;7:598–603.
33. Lesage R, Paris E, Koskas F, et al. Surgical reconstruction of the 53. Spetzler RF, Martin N, Hadley MN, et al. Microsurgical
internal carotid artery with contralateral occlusion without use of endarterectomy under barbiturate protection: a prospective study.
a shunt. Ann Vasc Surg. 1991;52:55–60. J Neurosurg. 1986;65(1):63–73.
34. Redekop G, Ferguson G. Correlation of contralateral stenosis and 54. Frawley JE, Hicks RG, Gray U, et al. Carotid endarterectomy with-
intraoperative electroencephalographic changes with risk of stroke out shunt for symptomatic lesions associated with contralateral
during carotid endarterectomy. Neurosurgery. 1992;30:191–194. severe stenosis or occlusion. J Vasc Surg. 1996;23(3):421–427.
35. Archie JP Jr. A fifteen-year experience with carotid endarterectomy 55. Gross CE, Adams HP, Sokoll MD, et al. Use of anticoagulants, elec-
after a formal operative protocol requiring highly frequent patch troencephalographic monitoring and barbiturate cerebral protec-
angioplasty. J Vasc Surg. 2000;31:724–735. tion in carotid endarterectomy, Neurosurgery, 1981; 9(1): 1-5.
36. Woodworth GF, McGirt MJ, Than KD, et al. Selective versus rou- 56. D’Angelo V, Catapano G, Bozzini V, et al. Propofol EEG burst
tine intraoperative shunting during carotid endarterectomy, a suppression in carotid endarterectomy. J Neurosurg Sci. 2001;
multi-variant outcome analysis. Neurosurgery. 2007;61(6): 45:159–162.
1176–1177. 57. Melgar MA, Mariwalla N, Madhusudan H. Carotid endarterec-
37. Imparato AM, Ramirez A, Riles T, et al. Cerebral protection in tomy without shunt: the role of cerebral metabolic protection.
carotid surgery. Arch Surg. 1982;117:1073–1078. Neurol Res. 2005;27(8):850–856.
38. Grego F, Antonello M, Lepidi S, et al. Is contralateral carotid artery 58. Williams IM, Picton A, Farrell A, et al. Light-reflective cerebral
occlusion a risk factor for carotid endarterectomy? Ann Vasc Surg. oximetry and jugular bulb venous oxygen saturation during
2005;19(16):882–889. carotid endarterectomy. Br J Surg. 1994;81:1291–1295.
39. Cikrit DG, Burt RW, Dalring EG, et al. Acetazolaminde enhanced 59. Beese U, Langeer H, Lang W, et al. Comparison of near-infrared
single photo emission computed tomography (SPECT) evaluation spectroscopy and somatosensory evoked potentials for the detec-
of cerebral perfusion before and after carotid endarterectomy. tion of cerebral ischemia during carotid endarterectomy. Stroke.
J Vas Surg. 1992;15:747–754. 1998;29:2032–2037.
40. Araki CT, Babikian VL, Cantelmo NL, et al. Cerebrovascular 60. Ferrari M, Zanette E, Sideri G, et al. Effects of carotid compression, as
hemodynamic changes associated with carotid endarterectomy. assessed by near infrared spectroscopy, upon cerebral blood volume
J Vasc Surg. 1991;13:854–860. and hemoglobin oxygen saturation. J R Soc Med. 1987;80:83–87.
41. Blankensteijn JD, Van der Grand J, Mali WPthM, et al. Flow vol- 61. Samara SK, Dy EA, Welch K, et al. Evaluation of a cerebral oxime-
ume changes in major cerebral arteries before and after carotid ter as a monitor of cerebral ischemia during carotid endarterec-
endarterectomy: an MR angiography study. Eur J Vas Endovasc tomy. Anesthesiology. 2000;93:964–970.
Surg. 1997;14:446–450. 62. Oyama H, Endoh O, Iizuka H, et al. The effectiveness of regional
42. Irvine CD, Gardner FV, Davies AH, et al. Cognitive testing in cerebral oxygen saturation monitoring using near-infrared spec-
patients undergoing carotid endarterectomy, Eur J Vas Endosvasc troscopy in carotid endarterectomy. J Clin Neurosci.
Surg. 1998;15:195–204. 2003;10(1):79–83.
43. Ucle’s P, Almarcegui S, Lorente S, et al. Evaluation of cerebral func- 63. Friedell ML, Clark JM, Graham DA, et al. Cerebral oximetry does
tion after carotid endarterectomy. J Clin Neruophysiol. not correlate with electroencephalography and somatosensory
1997;14(3):242–249. evoked potential in determining the need for shunting during
44. Vriens EM, Wieneke GA, Van Huffelen AC, et al. Increase in alpha carotid endarterectomy. J Vasc Surg. 2008;48:601–606.
rhythm frequency after carotid endarterectomy. Clin Neurophysiol. 64. Botes K, Le Roux DA, Van Marle J. Cerebral monitoring during
2000;111(8):1505–1513. carotid endarterectomy – a comparison between electroen-
45. Stoughton J, Nath RL, Abbott WM. Comparison of simultaneous cephalography, transcranial cerebral oximetry and carotid stump
electroencephalographic and mental status monitoring during pressure. S Afr J Surg. 2007;45(2):43–46.
carotid endarterectomy with regional anesthesia. J Vasc Surg. 65. Rigamonti A, Scandroglio M, Minicucci F, et al. A clinical evalua-
1998;28:1014–1023. tion of near-infrared cerebral oximetry in the awake patients to
46. Cheng MA, Theard MD, Templehoff R. Anesthesia for carotid monitor cerebral perfusion during carotid endarterectomy. J Clin
endarterectomy: a survey. J Neurosurg Anesthesiol. 1997;9:211–216. Anesth. 2005;17(6):426–430.
808 Part V ■ Complementary and Special Techniques

66. Vets P, ten Broeke P, Adriaensen H, et al. Cerebral oximetry in 86. Hanowell LH, Soriano S, Bennett HL. EEG power changes are
patients undergoing carotid endarterectomy: preliminary results. more sensitive than spectral edge frequency variation for detec-
Acta Anesthesiol Belg. 2004;55(3):215–220. tion of cerebral ischemia during carotid artery surgery: a
67. Wilkinson HA, Wright RL, Sweet WH. Correlation of reduction in prospective assessment of processed EEG monitoring.
pressure and angiographic cross-filling with tolerance of carotid J Cardiothorac Vasc Anesth. 1992;6:292–294.
occlusion. J Neurosurg. 1965;22:241–245. 87. Kearse LA, Martin D, McPeck K, et al. Computer-derived density
68. Beebe HG, Starr C, Slack D. Carotid artery stump pressure: its vari- spectral array in detection of mild analog electroencephalo-
ability when measured serially. J Cardiovasc Surg. 1989;30:419–423. graphic ischemic pattern changes during carotid enterectomy.
69. Harada RN, Comerota AJ, Good GM, et al. Stump pressure, elec- J Neurosurg. 1993;78:884–890.
troencephalographic changes, and the contralateral carotid artery: 88. Visser GH, Wieneke GH, van Huffelen AC. Carotid endarterec-
another look at selective shunting. Am J Surg. 1995;170:148–153. tomy monitoring: patterns of spectral EEG changes due to
70. Calligaro KD, Dougherty MJ. Correlation of carotid artery stump carotid artery clamping. Clinical Neurophysiol. 1999;110:286–
pressure an neurological changes during 474 carotid endarterec- 294.
tomies performed in awake patients. J Vas Surg. 2005;42(4):684–689. 89. Cursi M, Meraviglia MV, Fanelli GF, et al. Electroencephalographic
71. Jacob T, Hingorani A, Ascher F. Carotid artery Stump Pressure background desynchronization during cerebral blood flow reduc-
(CASP) in 1135 consecutive endarterectomies under general anes- tion. Clin Neurophysiol. 2005;116(11):2577–2785.
thesia: an old method that survived the test of time. J Cardiovasc 90. Larman DM, Wieneke GH, van Duijin H, et al. Q-EEG changes
Surg. 2007;48(6):677–681. during carotid clamping in carotid endarterectomy: spectral edge
72. Moritz S, Kasprzak P, Arlt M, et al. Accuracy of cerebral monitor- frequency parameters and relative band power parameters. J Clin
ing in detecting cerebral ischemia during carotid endarterec- Neurophysiol. 2005;22(4):244–252.
tomies: a comparison of transcranial Doppler sonography, 91. Liu J, Singh H, White PF. Electroencephalographic bispectral
near-infrared spectroscopy, stump pressure, and somatosensory analysis predicts the depth of midazolam induced sedation.
evoked potentials. Anesthesiology. 2007;107(4):563–569. Anesthesiology. 1996;84(1):64–69.
73. Fiori L, Parenti G, Marconi, F. Combined transcranial Doppler and 92. Hoffman WE, Zsigmond E, Albrecht RF. The bispectral index
electrophysiologic monitoring for carotid endarterectomy. during induction of anesthesia with midazolam and propofol.
J Neurosurg Anesthesiol. 1977;9(1):11–16. J Neurosurg Anesthesiol. 1996;8(1):15–20.
74. McCarthy RJ, Nasr MK, McAteer P, et al. Physiological advantages 93. el-Dawlatly AA. EEG bispectral index during carotid endarterec-
of cerebral blood flow during carotid endarterectomy under local tomy. Middle East J Anesthesiol. 2003;17(2):287–293.
anesthesia. A randomized clinical trial. Eur J Vas Endvasc Surg. 94. M’erat S, L’evecque JP, Le Gulluche, et al. BIS monitoring may
2002;24(3):215–221. allow the detection of serve cerebral ischemia. Can J Anesth.
75. Ackerstaff RG, Moons KG, van de Vlasakker CJ, et al. Association 2001;48(11):1055–1060.
of intraoperative transcranial Doppler monitoring variables with 95. Deogaonkar A, Vivar R, Bullock RE, et al. Bispectral index mon-
stroke from carotid endarterectomy. Stroke. 2000;31:1817–1823. itoring may not reliably indicate cerebral ischemia during awake
76. Dalman JE, Beenokkers ICM, Moll FL, et al. Transcranial Doppler carotid endarterectomy. Br J Anesth. 2005;94(4):559–560.
monitoring during carotid endarterectomy helps to identify 96. Bonhomme V, Desiron Q, Lemineur T, et al. Bispectral index pro-
patients at risk of postoperative hyperperfusion. Eur J Vas file during carotid cross clamping. J Neurosurg Anesthesiol. 2007;
Endovasc Surg. 1999;18:222–227. 19(1):49–55.
77. Rowed DW, Houlden DA, Burkholder LM, et al. Comparison of 97. Van Putten MJAM, Peters JM, Mulder SM, et al. A brain symme-
monitoring techniques for intraoperative cerebral ischemia. Can J try index (BSI) for online EEG monitoring in carotid endarterec-
Neurol Sci. 2005;32(1):131–132. tomy. Clin Neurophysiol. 2004;115:1189–1194.
78. Lacroix H, Beyens G, Van Hemelrijck J, et al. Is transcranial 98. Van Putten MJAM. Extended BSI for continuous EEG monitoring
Doppler useful in the detection of internal carotid artery cross- in carotid endarterectomy. Clin Neurophysiol. 2006;117:2661–2666.
clamp intolerance? Cardiovasc Surg. 1999;7(2):203–207. 99. Van Putten MJAM. The revised brain symmetry index. Clin
79. Costin M, Rampersad A, Soloman RA, et al. Cerebral injury pre- Neurophysiol. 2007;118:2362–2367.
dicted by transcranial Doppler ultrasonography but not electroen- 100. Ahn SS, Jordan SE, Nuwer MR, et al. Computed electroen-
cephalography during carotid endarterectomy. J Neurosurg cephalographic topographic brain mapping. A new and accurate
Anesthesiol. 2002;14(4):287–292. monitor of cerebral circulation and function for patients having
80. Dunne VG, Besser M, Ma WJ. Transcranial Doppler in carotid carotid endarterectomy. J Vasc Surg. 1988;8:247–254.
endarterectomy. J Clin Neurosci. 2001;8(2):140–145. 101. Larkin M. Should endarterectomy decision be based on non-
81. Totaro R, Varroni A, Gizzi E, et al. Transcranial Doppler sonogra- invasive imaging? Lancet. 2001;357:1343.
phy in the pre-intra- and post-operative evaluation of 85 patients 102. Elmore JR, Eldrup-Jorgensen J, Leschey WH, et al. Computerized
undergoing carotid endarterectomy. Clin Ter. 1998;149:267–270. topographic brain mapping during carotid endarterectomy. Arch
82. Stejskal L, Kramar F, Ostry S, et al. Experience of 500 case of neu- Surg. 1990;125:734–738.
rophysiological monitoring in carotid endarterectomy. Acta 103. Murad MH, Flynn DN, Elamin MB, et al. Endarterectomy vs
Neuroclir (Wein). 2007;149(7): 681–688. stenting for carotid artery stenosis: a systematic review and meta-
83. Manninen PH, Tan TK, Sarjeant RM. Somatosensory evoked analysis. J Vasc Surg. 2008;48(2):487–493.
potential monitoring during carotid endarterectomy in patients 104. Tang GL, Matsumura JS, Morasch MO, et al. Carotid angioplasty
with a stroke. Anesth Analg. 2001;93(1):39–44. and stenting vs. carotid endarterectomy for treatment of asymp-
84. Rampil IJ, Holzer JA, Quest DQ, et al. Prognostic value of comput- tomatic disease: single-center experience. Arch Surg. 2008;143(7):
erized EEG analysis during carotid endarterectomy. Anesth Analg. 653–658.
1983;62:186–192. 105. Steinbauer MG, Pfister K, Greindl M, et al. Alert for increased
85. Young WL, Moberg RS, Ornstein E, et al. Electroencephalographic long-term follow-up after carotid artery stenting: results of a
monitoring for ischemia during carotid endarterectomy: visual prospective, randomized, single-center trial of carotid artery
versus computer analysis. J Clin Mont. 1988;4:78–85. stenting vs. carotid endarterectomy. J Vasc Surg. 2008;48(1):93–98.
CHAPTER

ANTON KAMP, GERT PFURTSCHELLER, GÜNTER EDLINGER, AND FERNANDO H. LOPES DA SILVA
Polygraphy
40
P
olygraphy denotes the simultaneous recording of several behavioral variables simultaneously, employing separate
physiologic and/or behavioral variables. The main rea- recording systems, the time relations between the various types
sons for the simultaneous recording of several variables of signals must be preserved by using a form of time indexing
are to obtain information on behavioral aspects and to differen- or time marking on both recording media.
tiate artifacts in the electroencephalographic (EEG) data. These
objectives usually do not require precise representation; in many
instances, the relevant information concerns only the occur- CARDIOVASCULAR VARIABLES
rence of a certain phenomenon or is easily obtained from clearly
discernible characteristics of a variable. Therefore, most poly- Electrocardiogram and Heart Rate
graphic data of interest in EEG studies can be obtained using There are several reasons for recording the electrocardiogram
simple recording methods that allow appreciable distortion of (ECG) simultaneously with the EEG. It may be desirable in spe-
the original data. If, however, the polygraphic variables are of cific cardio- or cerebrovascular studies. In most cases, however,
primary concern, then sophisticated and precise recording the ECG recording is not intended to carry out a vascular study
methods are necessary. In view of the techniques used and the but only serves as an indicator of ECG artifacts in EEG records
interpretation of the recorded data, these methods go far beyond or as a general parameter of vegetative functions; in these latter
the simple polygraphic methodology commonly applied to EEG circumstances, one is mainly interested in the heart rate (HR).
studies. This chapter discusses these simple methods and pres- An ECG can be recorded perfectly using an EEG system because
ents a number of examples of variables that are of interest in the electrical characteristics of its input circuit and the provi-
certain EEG studies. A classic survey of variables of interest in sions commonly available for adjustment of frequency response
polygraphic studies and of corresponding recording methods and gain are adequate. The bandwidth required for appropriate
can be found in Manual of Psychophysiologic Methods, edited by ECG recording goes from 0.8 to 60 Hz; the recording sensitivity
Venables and Martin (1). required is approximately 1 mV/cm using conventional ECG
In practice, polygraphic recordings are made with an EEG electrode placements. When the ECG electrodes are placed on
apparatus; this may be of primary interest for determining the the chest wall, a higher sensitivity may be necessary. The sub-
temporal relations between the EEG and the other signals, ject’s behavioral activities might lead to artifacts in the ECG
which reflect different physiologic functions and/or behavioral record, owing to muscular activity or electrode motion. The lat-
states. Unfortunately, the frequency characteristics and the ter can be reduced significantly by using an appropriate type of
magnitude ranges of many signals of interest for polygraphic electrode, such as cup electrodes with a jelly bridge between
studies fall outside those provided by a conventional EEG skin and electrode surface. Interference caused by electromyo-
recording system; moreover, they might not be recordable due graphic potentials can be minimized by choosing electrode
to the electrical characteristics of the input circuit of the EEG positions carefully and lowering the high-frequency response
recorder. Such signals, therefore, require special provisions, of the recording system (20 Hz; 3 dB) in order to attenuate the
such as the use of specialized preamplifiers or input couplers to high-frequency electromyogram (EMG) potentials. High-
obtain an adaptation or a conversion; in this way, the recording frequency filtering can be obtained by means of the EEG appa-
of such signals can be carried out with the EEG apparatus. ratus’s adjustable high-frequency filters.
Many variables of interest in polygraphic studies, such as blood HR recording is best carried out by using a series of pulses
pressure, respiratory parameters, temperature, and electroder- generated at the top of clearly distinguishable R waves of the
mal signals, vary slowly as a function of time; therefore, their ECG. If, however, owing to less favorable electrode positions,
recording requires highly sensitive universal direct current the R wave cannot be easily distinguished, extra signal process-
(DC) amplifiers that are equipped with means of sensitivity ing must be applied. This processing may consist of high-pass
control and adjustable high- and low-pass filters for selection of filtering and/or the introduction of a refractory period, during
the appropriate frequency response. Modern EEG recorders which the instrument is insensitive. Commercially available
have low sensitivity auxiliary input terminals that also permit heart frequency meters or HR counters usually have such pro-
DC recording; these inputs can be used to record other signals visions. HR measurements may be given in terms of the num-
of sufficiently large amplitude (e.g., in Fig. 40.8, the traces indi- ber of beats over a certain period of time (HR) or in terms of
cated by EDG, STIM, BUTTON PRESS, and TIME CODE). heart period (HP), the average interval between a number of
When the EEG is used to record different physiologic and/or successive heartbeats. Because HR and HP are reciprocal, the
809
810 Part V ■ Complementary and Special Techniques

Figure 40.2 Principle of finger photophethysmography for transmis-


sion and reflexion of light.

in polygraphy. Figure 40.2 shows the principle of finger photo-


plethysmography. Two photosensors measure the transmission
and reflection of the light emitted from a light-emitting diode.
The fraction of transmitted light through the tissue and the frac-
Figure 40.1 Method for recording instantaneous heart rate (HR); the tion of reflected light from the tissue depend on the amount of
momentary heart period (HP) value, transformed into HR, is plotted in blood in the tissue. Extensive discussions of the measuring prin-
relation to an adjustable preset mean HR value. ciples, amplifier recorder requirements, and recorded waveforms
have been provided by Lader (3) (pneumatic plethysmography)
and Weiman (4) (photoplethysmography).
instrument, although calibrated in terms of HR, may have a Impedance plethysmography of the thorax for impedance
meter deflection or another output signal proportional to T int, cardiography is the basis for noninvasive beat-to-beat monitor-
the interval between successive heartbeats. In the available ing of the stroke volume (5). An electric current is introduced
instruments, HR is more commonly presented than HP. In into the thorax and the corresponding voltage is measured. The
some applications of polygraphy, the main interest is not in the ratio of voltage to current yields the impedance (Z) that varies
nominal HR but rather in HR changes and the relation to other (in a very simplified model) with the amount and distribution
physiologic, psychological, or behavioral variables. So that rela- of blood in the thorax. Based on the ECG, the phonocardiogram
tively small HR changes can be distinguished, the recording is (PCG), and impedance cardiogram (ICG), the stroke volume
best carried out with a preadjusted preset HR value. The model can be determined noninvasively (Fig. 40.3).
given in Figure 40.1 demonstrates a simple method for sub-
tracting a preset value from the HR meter’s electrical output.

Plethysmography
Plethysmography is the measurement of the variations in organ
or limb volume due to changes in the quantity of blood it con-
tains. Because such volume changes are related to increased or
decreased blood flow, plethysmographic methods can be used to
obtain estimates of the mean blood flow rate and of pulsatile and
transient flow changes. Plethysmography may be of interest in
psychophysiologic studies because mental processes and behav-
ioral responses are often accompanied by changes in such cardio-
vascular parameters as blood flow, accompanied by measurable
changes in limb volume. Continuous measurement of the latter
is known as pulse volume plethysmography; under certain
restricted conditions, an index of the blood flow rate can also be
obtained in this way (2). In most psychophysiologic studies, how-
ever, the variable of interest relates to changes in blood volume
and in blood volume pulses. The most common methods for Figure 40.3 Principle of impedance cardiography. (Adapted from
measuring limb volume changes are pneumatic and photoelec- Gratze G, Fortin J, Holler A, et al. A software package for noninvasive,
tric. Pneumatic methods are more complicated and are not real-time beat-to-beat monitoring of stroke volume, blood pressure,
suited to psychophysiologic studies. Therefore, although provid- total peripheral resistance and for assessment of autonomic function.
ing more precise information, they are much less frequently used Comp Biol Med. 1998;28:121–142.)
Chapter 40 ■ Polygraphy 811

Penaz (7) developed an important improvement in the non-


invasive determination of blood pressure, using continuous
measurement of the blood pressure in the finger. This method
uses a finger cuff. By means of a servosystem the cuff pressure is
maintained equal to the arterial pressure. This is achieved by
minimizing arterial diameter changes using a photoelectric
plethysmographic feedback method. The working principle is
shown in the block diagram of Figure 40.4. The further develop-
ment and evaluation of methods based on this approach (8,9)
are of great interest to those interested in the continuous meas-
urement and recording of beat-to-beat diastolic, systolic, and
mean arterial pressure. This is indicated in Figure 40.5 by the
Figure 40.4 Block diagram of a system for noninvasive continuous
similarity between continuous blood pressure and curves
recording of blood pressure based on the Penas principle. (From
recorded simultaneously by way of noninvasive and invasive
Wesseling KH, van Bemmel RA, van Dieren A, et al. Two methods for
methods.
the assessment of hemodynamic parameters for epidemiology. Acta
The noninvasive blood pressure measurement was first
Cardiol. 1978;33:84–87.)
applied during anesthesia (10) and was also used for long-term
sleep monitoring in patients with systemic hypertension and
Blood Pressure sleep-related breathing disorders (11). The new system gives
The catheter-manometer system is, at present, the fundamental valuable results if the position of the finger cuff is carefully con-
method for continuous accurate measurement of the full arte- trolled. An example of blood pressure recordings combined
rial pressure waveform. It is, however, an invasive procedure with respiratory measurements is displayed in Figure 40.6. A
and should be avoided unless the introduction of a catheter review of noninvasive continuous blood pressure measurements
into an artery is absolutely necessary. The Riva–Rocci– is found in Ruddel and Curio (12) and more recently in Parati
Korotkoff method (using an upper arm cuff and a stethoscope) et al. (13).
is noninvasive and commonly used, but it does not provide The capacity for measuring acute and immediate changes in
continuous blood pressure information. However, most auto- autonomic, EEG, and hemodynamic physiologic variables dur-
matic methods developed for determining blood pressure have ing different sleep stages on a continuous basis has played an
been based primarily on the Riva–Rocci–Korotkoff method. important role in enabling us to understand the interplay
For instance, Roy and Weiss (6) described a technique for pro- between changes in EEG and changes in circulatory variables
viding intermittent determinations of the systolic and diastolic and in autonomic neural functions. In this way the possibility
blood pressure obtained over several heartbeats. Such systems of recording simultaneously with the EEG, HR variability, and
have also become commercially available. The method indi- blood pressure (BP), among other cardiovascular physiologic
cated above, however, is sensitive to motion artifacts and not variables, has advanced our understanding of mechanisms link-
continuous. ing sleep and cardiovascular physiology (14).

Figure 40.5 Example of a comparison of


continuous recordings of blood pressure,
simultaneously obtained by an invasive
(intraarterial) and a noninvasive method. The
latter was performed according to the method
introduced by Penaz.
812 Part V ■ Complementary and Special Techniques

Figure 40.6 Recording of a patient with obstructive apneas and systemic hypertension. The trace of nasal airflow (NAF) shows
complete cessation of respiratory flow, whereas rib cage (RC) and abdominal (Abd) movement show obstructive efforts. Noninvasive
(FINA-PRES) and invasive BP were recorded in parallel. SaO2, oxygen saturation; micro, snoring noise. (From Penzel T, Ducke E,
Peter JJ, et al. Noninvasive monitoring of blood pressure in a sleep laboratory. In: Ruddel H, Curio I, eds. Non-invasive Continuous
Blood Pressure Measurement. Frankfurt am Maim: Peter Lang; 1991.)

Respiration obtained on frequency and on depth of chest movements. Other


The polygraphic recording of respiration patterns is usually car- methods should be used to obtain exact information concerning
ried out to obtain information on frequency or changes in respiratory volume.
inhalation depth. Changes in thoracic volume due to respira- For information concerning respiratory volume and thoracic
tory movements are usually estimated in terms of changes in and abdominal diameter, variations can be measured by using
the perimeter of the chest; this can be measured easily using elastic strips provided with strain gauges that encircle the thorax
transducers working on the principle of a strain gauge. This and abdomen at the level of the nipple and umbilicus, respec-
type of respiration transducer, which is commercially available, tively. Using both measurements, the respiratory volume can be
consists of a tube with an inner diameter of a few millimeters; calculated. This method of spirometry determines the contribu-
the tube is elastic and is filled with an electrically conductive tions of the chest and abdomen separately, then adds them
liquid substance with measurable electrical resistance. The tube together to mimic the total spirometric volume. As the chest and
forms part of a belt strapped around the chest; changes in abdominal volumes change during breathing, changes in electri-
resistance resulting from changes in chest perimeter are meas- cal impedance of the bands are related to changes in the spiro-
ured by means of the Wheatstone bridge circuit. The rather metric volume contributions using a calibration and gain
smooth slow respiration rhythm can be recorded with most adjustment procedure. A combination of techniques is com-
EEGs without further measures; in case of insufficient fre- monly used, namely the measurement of oral and nasal airflow
quency response of the apparatus, an alternating current (AC) and, in parallel, of abdominal and thoracic movements
voltage can be applied to the measuring bridge instead of a DC (Fig. 40.4). The best method to analyze the respiratory effort is
voltage. If respiration must be recorded from moving subjects, the use of an esophageal pressure probe (15).
chest size changes not related to respiration can easily occur; in Pulse oximetry is a noninvasive method to measure the arte-
these cases other methods should be chosen. rial hemoglobin oxygen saturation (SaO2). Continuous pulse
A simple and easily implemented solution is to use a thermis- and oxygen saturation measurements are obtained by ear,
tor or equivalent temperature-sensitive device placed in the finger, or soft probes (Fig. 40.4 contains an example). The
mouth and/or nostrils. Such a device works as a respiration trans- widely used pulse oximeter quantifies the SaO2 as a percentage
ducer by signaling the characteristic differences in temperature of based on spectrophometric and photoelectric plethysmography
the inspired and expired air. With such methods, information is (16). The SaO2 measurement yields information about the
Chapter 40 ■ Polygraphy 813

effectiveness of respiration and is recommended for all types of Classic extensive basic and practical information about electro-
sleep monitoring (17). dermal response recording can be found in the publications by
Venables and Martin (18) and Montagu (19) on skin resistance
Electrodermal Activities potential. Interesting applications of the GSR in psychophysio-
The variations of skin electrical properties in relation to psycho- logic studies have been published by Deschaumes-Molinaro et
logical variables, commonly known as the galvanic skin response al. (20) and Vernet-Maury et al. (21).
(GSR) or psychogalvanic response (PGR), consist of changes of
the electrical conductivity of the skin or of the electrical skin Eye Movements
potential, which can be measured by means of electrodes placed In various behavioral studies, particularly in sleep research for
on the palms or the soles in reference to an electrode placed the recognition of sleep stages, eye movement recording (i.e., by
elsewhere, such as on the back of the hand. means of the electro-oculogram, EOG) is necessary. Eye move-
For such measurements, the skin should be intact; when plac- ment recording is also useful in EEG recording for identifying
ing the electrodes, the skin must not be abraded. The skin poten- eye movement artifacts and studying lambda waves. An example
tial can be recorded easily, but it is unstable and of little use; of the former is the recording of eye movement in relation to
therefore, skin electrical conductance is much more useful in that of the CNV; in this case, it is possible either to reject the EEG
this respect. It can be estimated by applying a constant voltage epochs, where the amount of eye movement exceeds a certain
across two electrodes and by measuring the resulting electrical predetermined threshold in order to carry out selective averag-
current. This is, of course, equivalent to estimating the skin elec- ing (22) or to average the EOG along with the CNV to obtain an
trical resistance by applying a constant current through the elec- indication of the reliability of the latter (see also Chapter 36).
trodes and measuring the voltage across the electrodes. Skin Preferably, the method chosen for identifying eye movement
resistance can vary considerably among subjects, assuming val- artifacts will make use of the electrical field generated by the
ues from kilo- to megaohms. Transient skin resistance responses eyes. The EOG is, in general, easy to measure. The usual princi-
related to sudden changes in psychological state are in the order ple of EOG measurement is demonstrated by the model in
of 100 ohms. In practice, it is preferable to measure skin electri-
cal conductance by applying a constant voltage, instead of meas-
uring resistance by applying a constant electrical current. The
circuit of Figure 40.7A illustrates a constant voltage measuring
procedure. An example of electrodermal responses recorded
during the performance of a contingent negative variation
(CNV) paradigm from a normal subject is shown in Figure 40.8.

Figure 40.8 Polygraphic recording obtained during a contingent nega-


tive variation (CNV) investigation carried out in a normal subject. EEG
activities were recorded from three direct-coupled CNV derivations (FP2,
C4, and C3 against linked mastoids) and six anteroposterior symmetrical
Figure 40.7 A: Schematic diagram of a basic electronic circuit for the derivations. Other variables recorded were electrodermogram (EDG),
recording of electrodermal conductance using a constant voltage electrocardiogram (ECG), instantaneous heart rate, electro-oculogram
source. B: Bridge circuit for the recording of relatively small changes in horizontal eye movements [EOG (HOR)], CNV stimulus presentation
electrodermal conductance. [STIM (CNV)], the button press, and a time code (1-second intervals).
814 Part V ■ Complementary and Special Techniques

Saccades are rapid eye movements that move the line of sight
between successive points of fixation; they are among the best
understood of movements, possessing dynamic properties that
are easily measured (28). Saccades have become a popular
means to study motor control and cognitive functions, particu-
larly in conjunction with other techniques such as EEG, evoked
potentials, functional imaging, and transcranial magnetic
stimulation.

MUSCLE ACTIVITY AND BODY MOVEMENTS


Figure 40.9 Positions of EOG electrodes for recording horizontal and EMG Activity
vertical eye movements.
Part of the electrical activity of muscles (EMG) can be recorded
easily, by means of either surface electrodes placed on the sur-
Figure 40.9. As a result of the corneoretinal standing potential rounding skin or needle electrodes inserted into muscle. The
(the cornea is positive relative to the fundus), a DC potential dif- frequency range of EMG potentials, particularly those recorded
ference can be measured either between the pair of electrodes by means of intramuscular needles, goes far beyond the fre-
(EH) placed in a horizontal plane near the canthi of the eyes or quency response of most EEG recording systems. Even an EEG
between the two electrodes (EV) placed in a vertical plane, apparatus having an ink-jet writing system with a frequency
depending on the position of the eyeballs. Any change in eyeball response of up to 1000 Hz does not provide a faithful represen-
position results in a corresponding change of these two potential tation of EMG potentials. However, using a filter giving the
differences. DC recording is necessary to measure exact eye posi- highest possible EEG frequency response (usually 70 to 100 Hz),
tions, whereas AC recording suffices for determining changes in the recording of EMG activity in most instances adequately indi-
eye position. cates the presence of muscular activity and may even provide a
To carry out a DC recording of the EOG, nonpolarizable rough quantitative measure of the amount of such activity. In
electrodes must be used, and the drift of the electrodes’ offset certain applications, the latter can be better obtained by passing
potential should be taken into account. To obtain sufficiently the EMG potentials through a two-way rectifier, the output of
high recording sensitivity, the electrodes must be placed as close which is integrated with a specified time window. In this way, a
to the eye as possible. To record horizontal movements, the rectified smoothed EMG record is obtained. It is important to
electrodes should be placed near the external canthus of each recognize that the recording of EMG along with EEG signals
eye; for vertical movements, the electrodes should be placed may be necessary for the interpretation of some specific features
closely above and below one or both eyes. The EOG measured of the latter, particularly in those applications such as EEG-
in this way has an amplitude of about 20 V per degree of eye- based brain–computer interfaces that rely on automated meas-
ball rotation. A frequency response up to about 30 Hz is ade- urements of EEG features (29).
quate to record the most rapid eye movements, and thus an In behavioral studies, EMG activity is often used to monitor
EEG recording channel can be used without further provision the onset of certain motor activities such as limb movements.
(see, e.g., trace EOG (HOR) in Fig. 40.8). Other principles for Another way of determining limb movements and tremor (30)
monitoring eye movement in clinical EEG are the measurement makes use of displacement transducers or accelerometers. A
of changes in impedance related to eye movements (23) and use special form of tremor is the microtremor (microvibration),
of a pressure transducer system (24). In the latter system, a thin which is a phenomenon of the sensorimotor system that is
membrane covering one end of a tube held in a spectacle frame modulated by central mechanisms (31). This microtremor is in
is adjusted to touch the closed eyelid lightly. A pressure trans- the frequency range of 8 to 12 Hz (amplitudes of 1 to 10 m)
ducer connected at the other end of the tube detects pressure and can be recorded by an accelerometer fixed, for example, at
changes from eye movements. Robinson (25) describes eye the wrist. The measurement range of such an accelerometer
movement recording systems that also provide information on should be between 5 and 5g., such that the microtremor in
exact eye position; eye position is determined from the voltage the range of 10 mV/g can be recorded. Using accelerometry and
generated by an alternating magnetic field in a coil embedded EMG recordings of the forearm, the characteristics of physio-
in a scleral contact lens worn by the subject. A similar photo- logical tremor have been studied (32). In addition to the
electric device (26) consists of four small infrared-detecting microtremor, ballistic movements also be measured with this
cells mounted on a light spectacle frame together with a minia- technique.
ture infrared (9000Å)-emitting diode. This system, which pre-
serves maximum vision field size, has a resolution less than 1 Body Movements
minute of arc, a bandwidth of 1000 Hz, and 5% linearity of the The detection of limb or whole-body movements can be of
maximum range. Currently, systems are available for recording interest in sleep studies. With a simple wrist actigraph, a dis-
and analysis of a wide range of eye movements, both saccadic crimination between sleep and wakefulness is possible (33).
and smooth pursuit movements, where these are measured Actigraphy has been found to be reliable for evaluating sleep
using a scleral reflection technique (IRIS instrument) (27). patterns in patients with insomnia, for studying the effect of
Chapter 40 ■ Polygraphy 815

treatments designed to improve sleep, and in the diagnosis of 6. Roy R, Weiss M. Automatic blood pressure indicator. IRE Trans
circadian rhythm disorders, including shift work (34). Such a Biomed Electron 1962;9:244–246.
wrist actigraph can be used not only in the sleep laboratory but 7. Penaz J. Photoelectric measurement of blood pressure volume and
also with outpatients to obtain a picture of the degree of sleep flow in the finger. In: Alben R, Vogt W, Helbig W, eds. Digest of the
10th International Conference on Medicine and Biological
disturbances within, for example, a 24-hour period (17). The
Engineering. Dresden; 1973:104.
recording of movements along with the EEG can also be impor-
8. Reeben V, Epler M. Detection and many-side use of signals from
tant in differentiating authentic EEG activity from movement controlled counter pressure finger cuffs. Proc Biocapt Paris.
artifacts (35), which, on the basis of their waveforms and ampli- 1975;1:265–270.
tude, cannot easily be identified as noncerebral. When monitor- 9. Wesseling KH, van Bemmel RA, van Dieren A, et al. Two methods
ing movements of epileptic patients to determine the for the assessment of hemodynamic parameters for epidemiology.
occurrence of seizures during the night, transducers indicating Acta Cardiol. 1978;33:84–87.
global body movements may be used. With this objective, con- 10. Wesseling KH, Settels JJ, de Wit B. The measurement of continu-
ventional pressure or displacement transducers or other special ous finger arterial pressure noninvasively in stationary subjects. In:
methods developed for the detection of whole-body (36) or Schmidt TH, Dembroski TM, Blumchen G, eds. Biological and
limb movements (37) have been used. Psychological Factors in Cardiovascular Disease. Berlin: Springer-
Verlag; 1986:355–375.
Another method used for recording body movements is the
11. Penzel T, Ducke E, Peter JJ, et al. Non-invasive monitoring of blood
static charge-sensitive bed, which consists of a mattress with
pressure in a sleep laboratory. In: Ruddel H, Curio I, eds. Non-
two electrically active layers (38) with the use of filters; the bal- invasive Continuous Blood Pressure Measurement. Frankfurt am
listocardiogram, respiratory signals, and movement signals can Main: Peter Lang; 1991.
be differentiated. 12. Ruddel H, Curio I. Non-invasive Continuous Blood Pressure
For movement quantification in epileptic seizures advanced Measurement. Frankfurt am Main: Peter Lang; 1991.
video analysis methods can be applied (39). Markers at land- 13. Parati G, Ongaro G, Bilo G, et al. Non-invasive beat-to-beat blood
mark points are attached to the patient. Then EEG is acquired pressure monitoring: new developments. Blood Press Monit.
and the movement of the body parts is monitored using special 2003;8(1):31–36.
cameras. Quantified motion trajectories of body parts can be 14. Murali NS, Svatikova A, Somers VK. Cardiovascular physiology
extracted based on the fiducial markers. The trajectories reflect and sleep. Front Biosci. 2003;8:s636–s652.
15. Roberts S, Davies WL. Comparison of simple (slope based) and
the motion pattern of patients during seizures yielding addi-
more complex (simple syntactic) algorithms for central apnoea
tional movement information that cannot be obtained from
detection. In: Chase MH, Lydic R, O’Connor C, eds. Sleep Research.
standard video-EEG analysis. Los Angeles: Brain Information Service/Brain Research Inst;
1989:398.
Temperature 16. West P, George CF, Kryger MH. Dynamic in vivo response charac-
Temperature can be measured with sensors placed on the skin teristics of three oximeters: Hewlett-Packard 47201A, Biox III, and
or with a rectal probe. Temperature measurements are espe- Nellcor N-100. Sleep. 1987;10:263–271.
cially important in sleep studies. Detailed studies of rectal tem- 17. Penzel T, Stephan K, Kubicki S, et al. Integrated sleep analysis, with
perature recordings over 24 hours are reported by Stephan and emphasis on automatic methods. In: Degen R, Roding EA, eds.
Epilepsy, Sleep and Sleep Deprivation. 2nd ed. (Epilepsy Res. Suppl.
Dorow (40). Experiments with long-term isolation of subjects
2). Amsterdam: Elsevier; 1991:177–204.
have revealed a relationship between the body temperature and
18. Venables PH, Martin I. Skin resistance and skin potential. In:
the duration and stage of sleep (41). To assess core temperature, Venables PH, Manin I, eds. A Manual of Psychophysiological
for instance during monitoring under anesthesia, the recording Methods. Amsterdam: North Holland; 1967:53–102.
of tympanic temperature may be carried out. 19. Montagu JD. The psycho-galvanic reflex: a comparison of d.c. and
a.c. methods of measurement. J Psychosom Res. 1964;8:49–65.
20. Deschaumes-Molinaro C, Dittmar A, Vernet-Maury E. Autonomic
REFERENCES
nervous system response patterns correlate with mental imagery.
1. Venables PH, Martin I, eds. A Manual of Psychophysiological Physiol Behav. 1992;51(5):1021–1027.
Methods. Amsterdam: North Holland; 1967. 21. Vernet-Maury E, Alaoui-Ismaili O, Dittmar A, et al. Basic emo-
2. Melrose DG, Lynn RB, Rainbow RLG, et al. A sensitive digital tions induced by odorants: a new approach based on autonomic
plethysmograph. Lancet. 1954;1:810–812. pattern results. J Auton Nerv Syst. 1999;75(2–3):176–183.
3. Lader MH. Pneumatic plethysmography. In: Venables PH, Martin 22. Papakostopoulos D, Winter A, Newton P. New techniques for the
I, eds. A Manual of Psychophysiological Methods. Amsterdam: control of eye potential artifacts in multichannel CNV recordings.
North Holland; 1967:159–183. Electroencephalogr Clin Neurophysiol. 1973;34:651–653.
4. Weiman J. Photoplethysmography. In: Venables PH, Martin I, eds. 23. Sullivan GH, Weltman G. Impedance oculograph—a new tech-
A Manual of Psychophysiological Methods. Amsterdam: North nique. J Appl Physiol. 1963;18:215.
Holland; 1967:185–217. 24. Winter AL, Kellenyi L. Eye movement monitoring. Proc
5. Gratze G, Fortin J, Holler A, et al. A software package for noninva- Electrophysiol Technol Assoc. 1971;18:121.
sive, real-time beat-to-beat monitoring of stroke volume, blood 25. Robinson DA. A method of measuring eye movement using a scle-
pressure, total peripheral resistance and for assessment of auto- ral search coil in a magnetic field. IEEE Trans Biomed Eng BME.
nomic function. Comp Biol Med. 1998;28:121–142. 1963;10:137–145.
816 Part V ■ Complementary and Special Techniques

26. Gauthier GM, Volle M. Two-dimensional eye movement monitor 35. Buchthal F, Dahl E, Projaborg W. Simultaneous recording of accel-
for clinical and laboratory recordings. Electroencephalogr Clin eration and brain waves. Electroencephalogr. Clin Neurophysiol.
Neurophysiol. 1975;39:285–291. 1973;34:550–552.
27. Muir SR, MacAskill MR, Herron D, et al. EMMA—an eye move- 36. van Nimwegen C, Boter J, van Eijnsbergen B. A method of detect-
ment measurement and analysis system. Australas Phys Eng Sci ing epileptic seizures. Epilepsia. 1975;16:689–692.
Med. 2003;26(1):18–24. 37. Kripke DR, Mullaney DJ, Messin S, et al. Wrist actigraphic meas-
28. Leigh RJ, Kennard C. Using saccades as a research tool in the clin- ures of sleep and rhythms. Electroencephalogr Clin Neurophysiol.
ical neurosciences. Brain. 2003;127(3):460–477. 1978;44:674–676.
29. Goncharova II, McFarland DJ, Vaughan TM, et al. EMG contami- 38. Alihanka J, Vaahtoranta K, Saarikivi J. A new method of long-term
nation of EEG: spectral and topographical characteristics. Clin monitoring of the ballistocardiogram, heart rate, and respiration.
Neurophysiol. 2003;114(9):1580–1593. Am J Physiol. 1981;240:348–392.
30. Oppel F, Umbach WW. A quantitative measurement of tremor. 39. Li Z, da Silva AM, Cunha JPS. Movement quantification in epilep-
Electroencephalogr Clin Neurophysiol. 1977;43:885–888. tic seizures: a new approach to video-EEG analysis. IEEE Trans
31. Burne JA, Lippold OC, Pryor M. Proprioceptors and normal Biomed Eng. 2002;49:565–573.
tremor. J Physiol. 1984;348:559–572. 40. Stephan K, Dorow R. Circadian variations of core body tempera-
32. Elble RJ. Characteristics of physiologic tremor in young and eld- ture, performance and subjective ratings of fatigue in “morning”
erly adults. Clin Neurophysiol. 2003;114(4):624–635. and “evening” types. In: Redfern PA, Campall IC, Davies JA, eds.
33. Sadeh AJ, Alster J, Urbach D, et al. Actigraphically based automatic Circadian Rhythms in the Central Nervous System. Weinheim:
bedtime sleep-wake scoring: validity and clinical applications. J Verlagsgesellschaft, 1985:233–236.
Ambul Monit.1989;2:209–216. 41. Zulley J, Wever RA, Aschoff J. The dependence of onset and dura-
34. Ancoli-Israel S, Cole R, Alessi C, et al. The role of actigraphy in the tion of sleep on the circadian rhythm of rectum temperature.
study of sleep and circadian rhythms. Sleep. 2003;26(3):342–392. Pflugers Arch. 1981;391:314–318.
CHAPTER
Polysomnography:
Technical and Clinical Aspects
SUDHANSU CHOKROVERTY, RODNEY RADTKE, AND JANET MULLINGTON
41
P
olysomnography (PSG) is the single most important lab- during the deepening stages of sleep (5). It is interesting to note
oratory technique for assessment of sleep and its disor- that Kohlschutter (6,7), a 19th century German physiologist,
ders. PSG consists of recordings of multiple physiologic thought that sleep was deepest in the first few hours and became
characteristics during sleep, whereas polygraphy indicates the lighter as time went on, and he also described the varying
recording of similar characteristics at any time during the day. arousal thresholds throughout the night. The REMs of sleep
An understanding of the importance of the laboratory evalua- were not known in those days. In 1953, Aserinsky and Kleitman
tion of sleep and its disorders has been evolving slowly, but in (2) obtained characteristic REMs during sleep using surface
the last century great advances have been made in this direc- electrodes over the eye lids. Over the 4-year period, Loomis and
tion. The discoveries of the human electroencephalogram (EEG) his colleagues (5) published 12 papers, describing the oscillating
by Berger (1) in 1929 and rapid eye movements (REMs) during nature of EEG along with five distinct stages of sleep as well as
sleep by Aserinsky and Kleitman (2) in 1953 are the real driving arousals manifested by increased body movements and respira-
forces behind this understanding. In 1974, Holland and his col- tory effort. In 1957, Dement and Kleitman (8) described sleep
leagues (3) during a presentation at the 14th annual meeting of evolving through non-rapid eye movement (NREM) and REM
the Association of the Psychophysiological Study of Sleep (later sleep states in a cyclic manner throughout the night. All the
named American Sleep Disorders Association, which was components of REM sleep, however, have not been described by
finally changed to the American Academy of Sleep Medicine them until Jouvet and Michel (9) in 1959 observed markedly
[AASM]) coined the term PSG. This chapter is divided into sev- decreased muscle tone during REM sleep in cats. In 1961, Berger
eral parts: The initial part includes a brief review of the histori- (10) recorded markedly decreased muscle tone from extrinsic
cal milestones, functional neuroanatomy of sleep, physiologic laryngeal muscles during human REM sleep. In order to stan-
changes emphasizing those pertinent to overnight PSG inter- dardize the scoring of different stages of sleep, an ad hoc com-
pretation and clinical relevance as well as homeostatic and mittee led by Rechtschaffen and Kales (R–K) (11) in 1968
circadian factors, and functions of sleep. Next we deal with produced the now-famous sleep scoring technical manual (The
laboratory procedures including PSG recording and scoring R–K Scoring Technique). This remained the “gold standard” until
techniques, indications for PSG, video PSG, ambulatory and the AASM published the AASM Manual for the Scoring of Sleep
computerized PSG, artifacts during PSG recording, and pitfalls and Associated Events (12), which modified the R and K tech-
of PSG. We then discuss clinical considerations, briefly describ- nique and extended the scoring rules. The R and K sleep scoring
ing clinical presentation, diagnosis, and treatment but mainly manual (11) was devised only for normal sleeping adults, but
focusing on PSG findings in common sleep disorders as well as later infant sleep scoring manual was developed. R and K sleep
sleep-related movement disorders and neurologic disorders, scoring technique is based on three physiologic characteristics:
and sleep-related epilepsies. The final part addresses related EEG, electro-oculography (EOG), and electromyography
laboratory procedures for assessment of sleep including multi- (EMG). PSG, however, includes more than just sleep staging,
ple sleep latency test (MSLT), maintenance of wakefulness test and other physiologic characteristics such as respiration, limb
(MWT), and actigraphy. muscle activity, blood oxygen saturation, electrocardiogram
(ECG), body position, snoring, and other special recordings
HISTORICAL MILESTONES have been incorporated, which are described later in this chap-
ter. Since the discovery of REM sleep, dream was thought to be
In 1875, Richard Caton (4), an English neurophysiologist, first associated with REM sleep during 80% of the time. In 1868
recorded electrical activities from rabbit and monkey brains. Griesinger (13) suggested that dreaming was associated with eye
It was, however, Hans Berger (1), an Austrian psychiatrist, who movements and in 1895, Freud (14) indicated that dreaming was
in 1924 first obtained electrical activities from the scalp surface associated with relaxation of the major muscles of the body.
of a 17-year-old young man with a skull defect and published his Amazingly, centuries ago (ca. 1000 BC), Upanishads (15), the
findings in 1929. Although sleep was known since antiquity, ancient Indian text of Hindu religion, sought to divide human
brain activities in sleep could not have been recorded before the existence into four states: the waking, the dreaming, the deep
discovery of EEG. It was in 1937 that Loomis and his colleagues dreamless sleep, and the superconscious (“the very self”). This is
classified sleep into five stages (A–E) based on the EEG activities reminiscent of modern classification of three states of existence.

817
818 Part V ■ Complementary and Special Techniques

FUNCTIONAL NEUROANATOMY OF SLEEP


The control of sleep is quite complex. Whereas for the most part
during the whole sleep period we are in an unconscious state,
changes occur that are physiologically quite distinct. During slow
wave sleep, the cortical neurons fire in relative synchrony, pro-
ducing what is described as slow wave sleep, so named after
the polygraphic pattern of this brain activity. During REM sleep,
the eyes show phasic, rapid conjugate movements, whereas at the
same time skeletal muscle tone is at its lowest levels. Brain activ-
ity during REM sleep is rapid and desynchronous, almost like
waking, which is why it is also referred to as “paradoxical sleep.”

ASCENDING AROUSAL SYSTEM


There are two major branches to the ascending arousal or retic-
ular activating system (ARAS) (16). One is an ascending path-
way from pedunculopontine (PPT) and laterodorsal tegmental
(LDT) nuclei to the reticular nucleus and thalamic relay nuclei
in the thalamus. These thalamic relay neurons transmit signals
to the cortex. Neurons in the PPT and LDT fire most rapidly
during REM and wakefulness, and are at their most inactive Figure 41.1 The ascending arousal system. Two major pathways are
state during NREM. shown. One (shown in light gray) providing upper brainstem input to
The second branch of the ARAS comes from locus coeruleus the thalamic-relay nuclei and to the reticular nucleus of the thalamus
(LC), dorsal and median raphe nuclei, periaqueductal gray mat- coming from the pedunculopontine and laterodorsal tegmental
ter, and tuberomammillary neurons. Cortical input goes through (PPT/ LDT) nuclei, which are acetylcholine (ACh)-producing neuronal
lateral hypothalamic neurons and basal forebrain neurons. The groups. The second major group of neurons (shown in dark gray) come
neuron groups in the two branches of the ARAS are shown in from the noradrenergic (NA) locus coeruleus (LC), serotoninergic (5-HT)
Figure 41.1 along with their respective neurotransmitters (17). dorsal and median raphe nuclei, dopaminergic (DA) periaquiductal gray
The Hypocretin (Orexin) peptidergic system (18) located in matter (vPAG), and histaminergic (His) tuberomamillary neurons (TMN).
the lateral hypothalamic and perifornical regions with its wide- Additional cortical input merges from the basal forebrain (BF) neurons
spread ascending and descending projections is thought to play containing GABA or ACh, and by lateral hypothalamic (LH) peptidergic
an important role in the control of arousal and wakefulness. A neurons that contain melanin-concentrating hormone (MCH) or orexin
reduction of activities of the hypocretin projections to the LC, (hypocretin) (ORX). (Reproduced from Saper CB, Scammell TE, Lu J.
midline raphe, mesopontine, and posterior hypothalamic and Hypothalamic regulation of sleep and circadian rhythms. Nature. 2005;
tuberomedullary regions will cause sleepiness. 437:1257–1263, with permission.)
REM-Generating Neurons
The existence of REM sleep-generating neurons in the pons has neurons in the deep mesencephalon, ventral periaqueductal
been proved by transection experiments in cats through differ- gray, and lateral pontine tegmentum, and GABAergic REM-on
ent regions of the midbrain, pons, and medulla (19) (Fig. 41.2). neurons in the sublaterodorsal (SLD) nucleus (equivalent to
A transection of the pontomesencephalic (A) and pon- perilocus coeruleus alpha in the cat) and the dorsal extension of
tomedullary (B) junctions produced an isolated pons that shows the SLD named precoeruleus to explain REM sleep mechanism.
all the signs of REM sleep (see Fig. 41.2). There are mainly two Ascending glutamatergic projections from precoeruleus neurons
animal models available to explain the mechanism of REM to the medial septum are responsible for the hippocampal EEG
sleep. McCarley–Hobson (20) reciprocal interaction model theta rhythm during REM sleep. Muscle atonia during REM
based on reciprocal interactions of REM-on and REM-off neu- sleep is related to descending glutamatergic projections from the
rons is the earliest and most well-known model. A reciprocal ventral SLD directly to the spinal interneurons inhibiting spinal
interaction in the brainstem between REM-on neurons (the ventral horn cells by both glycinergic and GABAergic mecha-
cholinergic pedunculopontine tegmental and LDT nuclei in the nisms. Cholinergic and aminergic neurons play a modulatory
pontomesencephalic region) and REM-off neurons (the amin- role in this model and are not part of the “flip-flop” switch.
ergic LC and dorsal raphe [DR] nuclei) is responsible for REM Brooks and Peever (22) recently challenged the glycinergic and
generation and maintenance. The role of gamma-aminobutyric GABAergic neurochemical mechanisms of REM motor atonia
acid (GABA) in the REM sleep generation has been emphasized based on experimental evidence in rats that REM atonia per-
in the latest modification of the reciprocal interaction model by sisted even when glycine and GABA receptors were blocked.
McCarley (20). Lu and co-workers (21) described a “flip-flop” These authors suggested that multiple biochemical pathways are
switch interaction model in rats between GABAergic REM-off responsible for controlling muscle tone in REM sleep.
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 819

Schematic: Sagittal Section of Brain Stem pathophysiologic mechanism and the therapeutic intervention for
this disorder. These physiologic changes are most commonly
C B A noted in the respiratory, cardiovascular, gastrointestinal, and
endocrine systems. Respiration is controlled by the automatic or
metabolic and behavioral systems complemented by the third sys-
tem known as the arousal system or the system for wakefulness
stimulus. These respiratory systems receive inputs from various
peripheral and central neural structures to maintain acid–base
regulation and respiratory homeostasis. The location of the respi-
ratory neurons makes them easily vulnerable to a variety of neu-
rologic disorders, particularly those involving the brainstem.
Some conditions may affect control of breathing only during
sleep, causing undesirable, often catastrophic, results including
Spinal Cord Medulla Pons Midbrain Forebrain cardiorespiratory failure or even sudden death. The two control
systems (metabolic and voluntary) are active during wakefulness,
but during NREM sleep, the voluntary system is inactive and res-
Figure 41.2 Schematic sagittal section of the brainstem of the cat. piration is entirely dependent upon the metabolic controller. The
A: Junction of midbrain and pons. B: Junction of pons and medulla. behavioral mechanism is probably responsible for controlling
C: Junction of medulla and spinal cord. (Reproduced from Chokroverty breathing, at least in part during REM sleep. Tidal volume and
S. Sleep Disorders Medicine: Basic Science, Technical Considerations and alveolar ventilation decrease during sleep. Arterial oxygen tension
Clinical Aspects. 3rd ed. Philadelphia: Saunders/ Elsevier; 2009, with is slightly decreased and arterial carbon dioxide tension is
permission.) increased during both NREM and REM sleep. Hypoxic ventila-
tory response is impaired in NREM sleep in adult men but not in
women. Hypoxic ventilatory response during REM sleep is signif-
Anatomical Substrates for NREM Sleep icantly decreased in both sexes. Hypercapnic ventilatory response
NREM sleep-generating neurons are located primarily in the is also decreased during NREM and further decreased during
ventrolateral preoptic area (VLPO) of the hypothalamus, the REM sleep. Thus, ventilation is unstable during sleep and few
basal forebrain area as well as the solitary nucleus region of periods of apneas may occur particularly at sleep onset and dur-
the medulla. The reticular nucleus of the thalamus is thought to ing REM sleep in normal individuals. Respiratory homeostasis is
be responsible for sleep spindle generation. Both passive and thus relatively unprotected during sleep, making those individuals
active mechanisms play a role in the generation of sleep. The con- with intrinsic respiratory disease, such as chronic obstructive pul-
temporary theory (23) for the mechanism of NREM sleep sug- monary disease (COPD) or bronchial asthma, highly vulnerable
gests a reciprocal interaction between two antagonistic neurons for respiratory failure during sleep.
in the VLPO region of the anterior hypothalamus and wake-pro- As a result of increased parasympathetic and decreased sym-
moting neurons in the tuberomammillary nuclei of the posterior pathetic activity during sleep, heart rate, blood pressure, cardiac
hypothalamus and the hypocretinergic neurons in the lateral output, and peripheral vascular resistance decrease during
hypothalamus, as well as the LC, DR nuclei, basal forebrain, and NREM sleep and decrease still further during REM sleep. In
mesopontine tegmentum. The extended VLPO input to LC and REM sleep, however, there is an intermittent activation of the
DR is thought to be involved in regulating REM sleep. Reciprocal sympathetic nervous system accounting for rapid fluctuations
interaction between sleep-promoting neurons in the region of in blood pressure and heart rate. Cardiac output falls progres-
the solitary nucleus and the wake-promoting neurons within the sively during sleep and the greatest decrement occurs during
ARAS of the brainstem independent of the reciprocal interaction the last sleep cycle, particularly during the last REM sleep cycle
of the neurons of the forebrain also plays a minor role in the gen- early in the morning. This may explain why normal individuals
eration of NREM sleep. It has been suggested that adenosine, a and patients with cardiopulmonary disease are most likely to
neuromodulator, may act as a physiologic sleep factor modulat- die during the early morning hours. Cerebral blood flow and
ing the somnogenic effects of prolonged wakefulness (24). cerebral metabolic rates for glucose and oxygen decrease during
NREM sleep but increase to that of the waking values during
PHYSIOLOGIC CHANGES DURING SLEEP REM sleep. Because of these hemodynamic and sympathetic
changes during REM sleep during the last third of the sleep
A vast number of physiologic changes take place during sleep in cycle in the early hours of the morning, there could be increased
humans, affecting almost every system in the body (Table 41.1) platelet aggregability, plaque rupture, and coronary arterial
(25). It is important to have a basic knowledge about these spasms possibly triggering thrombotic events causing myocar-
changes during sleep and how they affect various sleep disorders. dial infarction, ventricular arrhythmias, or even sudden cardiac
These changes may be documented in the Polysomnography. A death. These circadian variations in cardiovascular and cere-
case in point is obstructive sleep apnea syndrome (OSAS) causing brovascular events with the highest rates of events occurring
dramatic changes in the respiratory control of the upper airway during the early morning hours have been documented by
muscles during sleep, directing our attention to a very important meta-analysis of epidemiologic studies.
820 Part V ■ Complementary and Special Techniques

Tabl e 41 . 1

Physiologic Changes during Wakefulness, NREM Sleep, and REM Sleep

Physiology Wakefulness NREM Sleep REM Sleep


Parasympathetic activity
Sympathetic activity Decreases or variable ( )
Heart rate Normal sinus rhythm Bradycardia Bradytachyarrhythmia
Blood pressure Normal Decreases Variable
Cardiac output Normal Decreases Decreases further
Peripheral vascular resistance Normal Normal or decreases slightly Decreases further
Respiratory rate Normal Decreases Variable; apneas may occur
Alveolar ventilation Normal Decreases Decreases further
Upper airway muscle tone Decreases or absent
Upper airway resistance
Hypoxic and hypercapnic Normal Decreases Decreases further
ventilatory responses
Cerebral blood flowa
Thermoregulation –
Gastric acid secretion Normal Variable Variable
Gastric motility Normal Decreases Decreases
Swallowing Normal Decreases Decreases
Salivary flow Normal Decreases Decreases
Migrating motor complex Normal Slow velocity Slow velocity
(a special type of intestinal
motor activity)
Penile or clitoral tumescence Normal Normal Markedly increased

aThere is, in general, global decrease in cerebral blood flow with regional variation during NREM sleep, but this is not homogeneous. It may not decrease in some
areas and may even show phasic increase in certain areas.
NREM, non-rapid eye movement; REM, rapid eye movement; , mild; , moderate; , marked; , very marked; , absent.
Reproduced from Chokroverty S. Sleep and its disorders. In: Bradley WG, Daroff RB, Fenichel GM, et al., eds. Neurology in Clinical Practice. Philadelphia: Elsevier;
2008:1960, with permission.

There are profound changes in endocrine secretions during Homeostatic and Circadian Factors
sleep. There is a pulsatile increase of growth hormone during and Functions of Sleep
NREM sleep in the first third of the normal sleep period. Prolactin The normal sleep–wake rhythm is controlled by both the circa-
secretion also rises 30 to 90 minutes after sleep onset. Cortisol dian and the homeostatic systems. The circadian system is
secretion is inhibited by sleep, whereas thyroid-stimulating entrained by the normal light–dark cycle and the hormone con-
hormone reaches a peak in the evening and then decreases trolling the circadian timing of physiologic systems is the pineal
throughout the night. Testosterone levels in men increase during hormone, melatonin. Bright light suppresses melatonin and it is
sleep, rising from low levels at 8 PM to peak levels at 8 AM, but there released during the dark phase. Human circadian rhythm has a
is no clear relationship noted between levels of gonadotrophic cycle close to 24 hours (24.2). The paired suprachiasmatic
hormones and sleep–wake cycle in children or adults. Melatonin, nuclei (SCNs) of the hypothalamus above the optic chiasm
the hormone of darkness released by the pineal gland, reaches its serve as the master biologic clock. SCN receives afferent infor-
highest secretion level between 3 AM and 5 AM and then decreases mation from the retinohypothalamic tract sending signals to
to low levels during the day. Thermoregulation is maintained multiple synaptic pathways including other parts of the hypo-
during NREM sleep, but it is nonexistent in REM sleep. Body tem- thalamus and the pineal gland where melatonin is released.
perature begins to fall at sleep onset and reaches its lowest point Remarkable progress including identification of many genes
during the third sleep cycle. and their protein products within the circadian clock in both
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 821

fruit flies (Drosophila) and mammals has been made (26). TECHNICAL PERSONNEL
Dysfunction of the circadian rhythm results in several impor-
tant sleep disorders including delayed sleep phase syndrome The hiring of technologists who are mature and competent and
and advanced sleep phase syndrome. highly motivated is a critical step in the operations of a sleep
During wakefulness, it is thought that a build up of some laboratory. A feeling of trust between the patient and technolo-
sleep factor, or process “S,” occurs and is burnt off or dissipated gist is very important for sleeping in an unusual environment
at night through sleep (27). In the morning after waking, cog- because electrodes and other devices attached to the body can
nitive and physiologic functioning takes a while to get to a full put the patient in a somewhat uneasy state. Coupled with the
waking state due to an inertia from the sleep system (28). The fact that they are in the laboratory for reasons of disturbed sleep
wake system takes a while to fully prime and synchronize all and/or wakefulness, and that continuous positive pressure may
components for optimal output. This mechanistic framework be tested, even on their first night in the laboratory, it is very
has explanatory utility and does a reasonable job of providing a important that they have a trusting professional interaction
framework for much of the physiologic and cognitive data. with the technologist.
Notably, the concept of homeostasis is exemplified by the find- In addition to professionalism, maturity, and excellent bed-
ing of recovery sleep following a period of sleep deprivation side manner, the technologist needs to be able to function well
that is intensified with respect to slow wave activity. This sug- at night, and, as much as possible, maintain a consistent shift.
gests that physiologic systems need slow wave sleep and there- Frequent rotations from night to day lead to stress and result in
fore attempt to catch up for what was lost by intensifying and unnecessary sleep loss and misalignment of biologic rhythms.
lengthening the sleep bout as well (29). This can in turn lead to impaired performance and mood dys-
The function of sleep remains the greatest biologic mystery regulation.
of all times. Sleep is essential and sleep deprivation leads to Technologists chosen for performing diagnostic and treat-
impaired attention and decreased performance in addition to ment initiation sleep recordings frequently have full EEG techni-
sleepiness. Sleep is thought to have restorative, conservative, cian training and/or training as a respiratory therapist. In
adaptive, thermoregulatory, and consolidative functions, as well addition, they require well-supervised training in specialized
as maintenance of synaptic and neuronal network integrity. polygraphic recording methods and in visual sleep staging,
Recent scientific data have strengthened the theory that mem- including identification and classification of arousals, leg move-
ory reinforcement and consolidation take place during sleep. ments, and respiratory disturbance. Technologists are responsible
for the maintenance of equipment and for the identification and
fixing of technical problems at night during a recording if they
SLEEP ARCHITECTURE should arise. Although most recordings are now performed with
The term “sleep architecture” refers to the pattern of sleep commercially available digital systems, the technologists still
stages that cycle through the night in what is referred to as the need to quickly and correctly identify problems and swap out
REM–NREM cycle. During development, there are changes parts as necessary. Therefore, they need to be able to make sure
that occur in the general pattern of sleep stages, but the adult that adequate supplies and back up equipment are available. The
shows a pattern of stages N1, N2, and N3 followed by REM “American Association of Sleep Technologists” has developed
sleep, and the periodicity of this REM–NREM cycle throughout standards of training, practice, and certification for registration
the night is generally 90 to 120 minutes in duration. The first of technologists in North America (http://www.aastweb.org/).
REM period of the night is very often brief and may even be
missed in young healthy sleepers. SETTING: EQUIPMENT AND
During infancy, the sleep–wake cycle is approximately 50% RECORDING ROOM
dominated by REM sleep, and the sleep pattern involves fre-
quent napping through the day. As the infant matures, there is A report for the technologist including the patient’s presenting
a consolidation of sleep into the night period. During infancy, symptoms and reason for the referral needs to be provided. The
the mother’s melatonin rhythm is conveyed to the infant technologist should also have access to list of medications and
through breast milk, helping to consolidate the sleep period other information pertinent to the care of the patient through
into the nocturnal dark phase. The last sleep to be given up dur- the night in the laboratory. It is most common for patients to be
ing the day period is generally the afternoon nap. studied outside of a hospital setting, but even within the hospi-
tal setting, it is important for the technologist to have explicit
POLYSOMNOGAPHIC TECHNIQUE instructions for when and whom to call with respect to critical
levels of oximetry, abnormal ECG, or other medical needs of
In 2007, the AASM (12) published a new manual for the scor- the patient. As in any hospital ward setting, it is not desirable for
ing of sleep and associated events, almost 40 years after the first a technologist to work alone, without any back up personnel
consensus on the technical standards for the field was published immediately available, for safety reasons.
by Rechtschaffen and Kales in 1968 (11). The AASM has a Web The equipment and technologist are usually housed in a room
site (http://www.aasmnet.org/) with links to resources for sleep separate from that of the sleeping subject. Rather than an institu-
medicine clinicians, scientists, technical personnel, and labora- tional-like bedroom, it is preferable for the patient to have an
tory managers, and to several position papers. environment that is as home-like as possible, with a comfortable
822 Part V ■ Complementary and Special Techniques

mattress, a closet, and night stand. The sleeping room should be the diameter of the electrode and about 5 to 8 cm along the wire.
quiet and sound attenuated so that noises from outside the sleep A length of tape added at right angles over the end of the first
laboratory and technologist’s area do not disturb the sleeping tape helps anchor the latter. A high-frequency filter of 35 Hz is
patient. An intercommunication system for contact with the used to reduce EMG artifact on the tracing. The low-frequency
patient is essential. Toilet facilities should preferably be attached filter is generally selected at 0.3 to 0.5 second (time constant) and
to the bedroom and a shower should also be readily available. permits recording of both the slow rolling eye movements of
Patients are generally asked to report to the laboratory facil- drowsiness and the REMs of REM sleep.
ity 1 to 3 hours before their normal bedtimes. They are shown
their room and the facility around to let them know they are EMG
appropriate. The technician asks them to change into their bed Mylohyoid muscle EMG is recorded with electrodes placed on
clothes and then they begin to attach the electrodes and record- the skin, secured with flexible sticky tape. It is recommended
ing devices. The technologist turns lights out after he/she con- that three electrodes be attached, one electrode positioned mid-
ducts a test of the equipment and performs biologic calibrations. line just above the inferior rim of the mandible and two other
Biologic calibrations enable the technologist to demonstrate to electrodes each 2 cm below the inferior edge of the mandible, to
the individual, who subsequently will review the recording, the the right and left. The technologist can in this way select the
normal waking eye movement pattern, and pattern of gritting bipolar combination of electrodes giving the best tonic EMG.
teeth, breathing deeply, coughing, and pointing and flexing the Pairs of electrodes have also been used on the jaw edge or over
toes and feet. All of these need to be documented by the technol- the masseter muscles to record axial muscle tone (31,32). There
ogist at the outset of the recording, and when lights are turned is a great deal of variability in the resting chin tone EMG level
off for the night, the technologist must document the time, in that is related to adipose tissue and age, and therefore it is
the recording. important that the technologist adjusts the EMG amplification
such that the background level during drowsiness shows some
THE ELECTRODE SETUP baseline elevation so that transition into sleep and particularly
REM is easily discernable.
EEG In addition to submental EMG, muscle activity is routinely
Sleep EEG has always been based on the standard “international recorded from the legs to document leg muscle activation.
10 to 20 EEG system” derivations originally defined by Jasper in These extra long electrodes are placed on the anterior tibialis
1958 (30). According to the most recent AASM scoring guide- muscles, again fixed with soft flexible skin tape such as microp-
lines (12), the recommended EEG derivations for sleep include ore, and need to be well fixed with tape so that they don’t come
scalp EEG derivations (from the frontal, central, and occipital off with normal leg movements during sleep.
regions) linked to the left mastoid (M1) or right mastoid (M2).
Recommendations for sleep stage discrimination are F4–M1, ECG
C4–M1, and O2–M1 with back up electrodes placed at F3, C3, A single lead that is modified from lead II used in clinical cardi-
O1, and M2 to allow for re-referencing to F3–M2, C3–M2, and ology is used to monitor the heartbeat during sleep, with an elec-
O1–M2 if there are problems that develop over the course of trode placed on the right subclavicle area and left torso over the
the recording with the right hemisphere selections. An alterna- lower ribcage beneath the heart. Standard EEG electrodes may be
tive montage for stage scoring is Fz–Cz, Cz–Oz, and C4–M1 with used, but ECG electrode applications are preferable in that they
back up electrodes Fpz, C3, O1, and M2. The EEG is the most are more resistant to artifact. Heart rate, extrasystoles, and other
important physiologic information for the determination of arrhythmias can be monitored using this lead, but due to differ-
sleep stages. Of course, for studies where epileptic discharge ences in sampling rate and amplifier characteristics, this tracing
during sleep is under investigation, additional electrodes would is not suitable for detecting PQRST complex abnormalities.
be included depending on the brain regions of interest.

EOG RESPIRATORY MONITORING


For routine EOG, the AASM scoring manual (12) recommends The measurement of respiration during sleep is critical to the
referential recordings from each outer canthus (1 cm below the diagnosis of sleep disorders and includes chest and abdominal
left and 1 cm above the right) to the ipsilateral mastoid. These excursion, most often using inductance plethysmography or
derivations show horizontal out-of-phase potentials very well, sometimes strain gauges, and also a means of assessing nasal air-
which are used in the identification of REMs associated with flow. Syndrome definitions and measurement techniques are
REM sleep. EOG electrodes should be attached with soft tape described for sleep-disordered breathing in an AASM task force
made for use on skin, and they should be never attached with col- report (33), and a more recent guidelines to the classification and
lodion-soaked gauze because of the potential for corneal damage scoring of respiratory events was published in the AASM Manual
and also because collodion becomes inflexible when dry, so not for the Scoring of Sleep and Associated Events in 2007 (12).
only itchy and uncomfortable but prone to lifting and coming off Careful monitoring of both upper airway airflow and thora-
during sleep. A micropore surgical tape maintains good contact coabdominal movement in the respiratory tracings is used to
for more than 48 hours of continuous recording and can be eas- categorize central, mixed, and obstructive sleep apnea (OSA),
ily trimmed to needed dimensions. Tape width is usually twice which is further discussed later. In addition, the monitoring and
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 823

quantification of blood oxygen levels are necessary and accom- PNEUMOTACHOGRAPHY


plished using pulse oximetry. Endoesophageal (intrathoracic)
pressure recording is also sometimes used in the detection of This technique is rarely used in diagnostic testing because it
partial obstructions or upper airway resistance that can exist involves the placement of an airtight mask on the face and uses
without actual apneas or hypopneas. The nasal pressure trans- a pressure transducer system to measure flow rate, tidal volume,
ducer (NPT) is recommended for the measurement of subtle and other respiratory variables. This method is adopted fre-
reductions in airflow and can also be used in the identification quently in the sleep testing laboratory during CPAP pressure
of absent airflow, characteristic of apnea. titration in order to monitor adequacy of pressure and flow (see
Nasal Pressure heading above).
Airflow
Upper Airway Exchange: Thermistors, RESPIRATORY EFFORT
Thermocouples, and Pressure Piezoelectric Strain Gauges
Upper airway exchange can be monitored by thermistor resist-
Thoracoabdominal movement is frequently monitored with
ance fluctuations or by thermocouples, both sensitive to temper-
piezoelectric crystal sensors incorporated into bands that are
ature changes of air passing in and out through the mouth and
fitted around the torso at the level of the chest and abdomen.
nostrils. Both of these sensors are attached under the nostrils
Generally these sensors can be plugged directly into the head-
and can also be designed or applied with an additional sensor
box and connected to the AC amplifier. Respiratory excursion is
pointing to the mouth, they are useful for documenting mouth
monitored and the traces are used in detecting reductions in
breathing. Thermistors generally use a ceramic or polymer
breathing effort, but it does not produce a measured signal,
material and detect temperature changes based on a Wheatstone
rather only a relative one.
bridge using DC voltage. Thermocouples are constructed of dis-
similar metals (e.g., constantin and copper) that generate a Inductance Plethysmography
potential in response to temperature change and are therefore Inductive plethysmography uses wire coils placed around the
thermoelectric generators. They can be connected directly to the chest and abdomen, sometimes woven into a vest, to monitor sep-
plug-in box and used with AC voltage for recording. arate movements and then add them together, mimicking total
Nasal Flow Using Nasal Airway Pressure spirometric volume. In contrast to the piezoelectric belts, this
measuring technique is not only adjusted for visual characteristics
Nasal flow can be monitored using a simple cannula, typically of the wave but also calibrated before the sleep period begins. It
used to deliver oxygen, attached to a pressure transducer and produces a measured output for chest movement, abdominal
may be recorded using very low-frequency filters with AC or movement, and total volume. This system has an advantage in
DC amplifiers. This technique is more informative and reliable being able to clearly distinguish paradoxical breathing associated
than thermistors and thermocouples and is a recommended with obstruction in contrast with central cessation of breathing
method for the detection of hypopneas (12). With appropriate that is reflected in a loss of signal in all three tracings.
filtering of the slow component to reveal the vibration signal
using an AC amplifier, nasal pressure can also be used to record INTERCOSTAL EMG
snoring. The primary concern in using nasal pressure exclu-
sively to measure upper airway flow is that the signal may drop Long electrode leads are sometimes applied on the lower back
due to mouth breathing. However, additionally a simple oral intercostal muscles to aid in the detection of effort during
thermistor/thermocouple can be used to resolve this problem. breathing and the placement is sensitive to muscle activity dur-
The pressure transducer can also be used during continuous ing coughing.
positive airway pressure (CPAP) titration to monitor respira-
tory waveform and mask leak. SNORING MONITOR
In addition to the NPT method mentioned earlier in this chap-
CAPNOGRAPHY
ter, both piezo sensors (encased in a rubber disk attached to the
Capnography refers to the monitoring of the concentration of throat and plugged into the headbox) and microphones are
carbon dioxide (CO2) in the respiratory gases and is measured used to monitor snoring. Snoring is associated with reduced
in the breath at the end of complete expiration (end tidal). A upper airway diameter, and bursts of loud guttural inspiratory
CO2 analyzer is sometimes used to document CO2 retention in snorting are signs of OSA.
sleep-related respiratory disorders, especially COPD and
hypoventilation syndromes. INDIRECT MEASUREMENT
OF BLOOD GASES
END TIDAL CO2 Transcutaneous (TcCO2) Measurement
End tidal CO2 refers to the measurement of the CO2 in the air Transcutaneous (TcCO2) measurement is the measurement of
exhaled from the lungs, and the normal range is 4% to 6% CO2 gas tension of tissue underlying a specialized electrode that
(equivalent to 35 to 45 mm Hg). is usually placed on the chest or forearm. The electrode method
824 Part V ■ Complementary and Special Techniques

uses a Stow–Severinghaus glass/ceramic electrochemical sensor, A sleep study is scored in 30-second epochs with each stage
with a small heater unit to facilitate blood flow. Transcutaneous labeled as stage W (or wakefulness), stage N1, stage N2, stage
monitoring of CO2 tension is more reliable than transcutaneous N3, or stage R. If two or more stages coexist in an epoch, the
measurement of O2 because CO2 diffuses better through the skin. epoch is labeled as the stage that represents the greatest portion
of the epoch. Stage W represents the waking state ranging from
Finger Oximetry for Oxygen Saturation full alertness through early drowsiness. Stage W is scored when
Standard polygraphic diagnostic recording must include mon- more than 50% of the epoch demonstrates alpha frequency
itoring of an index of blood oxygen saturation or the percent- activity over the occipital region (Fig. 41.3). Stage W is scored
age of available circulating hemoglobin that is saturated with in the absence of alpha activity (which occurs in 10% to 20% of
oxygen (SaO2). Probes are easily attached to the finger with a individuals) if eye blinks, reading eye movements, or irregular
specially designed clip or with tape. Measurement is achieved conjugate REMs are identified with accompanying normal or
using an optical device that uses a DC channel involving com- elevated chin muscle tone. The chin EMG during stage W is
putation of absorption of certain wavelengths of light. variable but is usually higher than that seen during sleep stages.
Stage N1 represents late drowsiness and light sleep. It is
BODY POSITION scored when greater than 50% of an epoch shows alpha attenu-
ation and is replaced by low amplitude, mixed frequency EEG
Body position is the useful information to have when interpret- activity (Fig. 41.4). K-complexes and sleep spindles are absent
ing a pattern of respiratory disturbance and can be obtained by definition. If an individual does not generate alpha activity,
visually through infrared camera monitoring and/or with the stage N1 is scored when over 50% of the epoch demonstrates
aid of a position sensor. Such sensors are available and can be theta range slowing (4 to 7 Hz), vertex sharp waves, or slow eye
attached to respiratory bands with accurate output of informa- movements. In individuals who do generate alpha activity, slow
tion including prone, supine, right side, and left side. eye movements are frequently seen before the disappearance of
Table 41.2 lists appropriate filter settings for recording vari- alpha activity, so individuals who do not generate alpha activity
ous physiologic characteristics in the PSG. may have stage N1 scored slightly earlier than those who do
generate alpha activity. During stage N1, chin EMG is variable
SLEEP STAGING CRITERIA but usually lower than that seen in stage W.
Stage N2 represents the predominant sleep stage during an
In 1968, the publication of the sleep scoring atlas by overnight recording and is scored when there is the appearance
Rechtschaffen and Kales (11) represented the consensus agree- of a K complex or sleep spindle (Fig. 41.5). In sleep medicine, a
ment between sleep researchers of the time to the scoring of sleep K complex is defined as a biphasic negative sharp wave maxi-
stages. Remarkably, this document remains the dominant force in mum at the vertex or frontal regions that lasts greater than 0.5
sleep scoring and analysis. With the recent publication of the second. Sleep spindles are 11 to 16 Hz sinusoidal activity lasting
AASM Manual for the Scoring of Sleep and Associated Events, the at least 0.5 second seen maximally in the central head region.
standardization of sleep and event scoring has reached a new Less than 20% of an epoch of stage N2 can be delta activity
level of consensus (12). The scoring of sleep in this recent docu- ( 2 Hz, 75 V in amplitude). Stage N2 is scored from the
ment varies only limitedly from that published in 1968 (11). first epoch of stage N2 until a clear epoch of stage W or another
The nomenclature of sleep stages has changed with the recent stage of sleep is identified. Although eye movements are usually
AASM scoring manual with NREM stage 1 now called stage N1, absent in stage N2, slow eye movements can sometimes be seen.
and NREM stage 2 is now stage N2. What had previously been Chin or axial EMG is variable but usually lower than stage W.
NREM stages 3 and 4 (or slow wave sleep) is now unified into a Stage N3 (which encompasses both former stages 3 and 4
single stage N3. Stage REM is now labeled as stage R. of R and K criteria (11)) is the deepest stage of sleep and is

Tabl e 41 . 2

Filter Settings for PSG Recordings

High-Frequency Time Constant Low-Frequency


Characteristics Filter (Hz) (second) Filter (Hz) Sensitivity
Electroencephalogram 70 or 35 0.4 0.3 5–7 V/mm
Electro-oculogram 70 or 35 0.4 0.3 5–7 V/mm
Electromyogram 90 0.04 5.0 2–3 V/mm
Electrocardiogram 15 0.12 1.0 1 V/cm to start; adjust
Airflow and effort 15 1 0.1 5–7 V/mm; adjust
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 825

Figure 41.3 The limited PSG montage illustrates


alpha activity (well seen after eye closure) consis-
tent with wakefulness or stage W (30-second
epoch, chin = submental EMG).

Figure 41.4 The limited PSG montage illustrates


the absence of alpha activity and the appearance
of slow eye movements (out-of-phase activity in
channels E1 and E2) consistent with stage N1
sleep (30-second epoch, chin = submental EMG).

Figure 41.5 The limited PSG montage illustrates


sleep spindles (sinusoidal activity in top two
channels) and broad vertex waves consistent
with stage N2 (30-second epoch, chin = submen-
tal EMG).
826 Part V ■ Complementary and Special Techniques

Figure 41.6 The limited PSG montage illus-


trates high-amplitude delta activity ( 2 Hz)
occupying greater than 20% of the epoch and
represents an example of stage N3. Waveforms
that span the two dotted lines in the top two
channels identify activity that is at least 75 V in
amplitude (30-second epoch, chin = submental
EMG).

associated with increasing delta activity. Stage N3 is scored first sleep cycle, a pattern that repeats itself three to five times
when 20% or greater of an epoch is delta activity (Fig. 41.6). during the typical night’s sleep. With each ensuing 90 minute
For the purposes of sleep scoring, delta activity is defined as sleep cycle, there is decreasing amounts of stage N3 sleep and an
activity of 0.5 to 2 Hz that is at least 75 V in amplitude when increasing amount of stage R sleep. Predictable changes are
measured over the frontal or central regions. Sleep spindles may noted in sleep architecture with aging and are illustrated by the
persist into stage N3, but eye movements are usually absent. histograms in Figure 41.8. Beginning in middle age, stage 3 sleep
Axial EMG is usually lower than stage N2 and may approach lessens and more wakefulness after sleep onset (WASO) is noted.
that seen in stage R. The number of arousals and awakening continues to increase
Stage R (formerly stage REM) requires three components for with aging, becoming particularly notable in the elderly (35,36).
it to be scored: low-amplitude mixed frequency EEG, low chin
EMG, and the presence of REMs (Fig. 41.7A,B). Phasic EMG AROUSAL SCORING
activity may occur, but tonic EMG activity must be at a level
that is as low as, or lower, than that occurring at any other time The original scoring guidelines of Rechtschaffen and Kales (11)
during the study. Sleep spindles and K-complexes are absent. focused primarily on the scoring of the stages of sleep (as dis-
Series of 2 to 5 Hz vertex negative “sawtooth” waves occur, par- cussed above). The R and K criteria defined that the appearance of
ticularly just before phasic REM activity. 30 seconds or more of waking background (which resulted in the
Movement time (or stage M as described by Rechtschaffen epoch being labeled as wake) would be labeled an awakening. The
and Kales (11)) is no longer scored. During major body move- R and K manual made reference to movement arousals, but no
ments that obscure greater than 50% of an epoch, the epoch is other mention of brief EEG frequency changes was made. There
scored as stage W if alpha activity is present at any time in the was little standardization to the scoring of arousals before the
epoch. If no alpha activity is identified, but an epoch of stage W publication of the position paper by the American Sleep Disorders
precedes or follows the epoch with a major body movement, Association (ASDA) task force in 1992 (37). This consensus paper
then the epoch is scored as stage W. If neither of these require- carefully defined rules for scoring an arousal. In the recent AASM
ments can be met, the movement is scored the same as the scoring manual (12), these rules were distilled to a single rule.
epoch that follows it. Score arousal during sleep stages N1, N2, N3, or R if there
is an abrupt shift of EEG frequency to alpha, theta, or frequen-
HYPNOGRAMS cies greater than 16 Hz (but not spindles) that lasts at least
3 seconds, with at least 10 seconds of stable sleep preceding
Normal sleep has a clearly defined architecture that occurs each the change in EEG frequency. Scoring arousals during REM
night. Hypnograms are a graphic display of the ultradian cycle requires a concurrent increase in submental EMG lasting at
within a night’s sleep (34). Sleep stage is depicted in the vertical least 1 second (Figs. 41.9 and 41.10).
axis with the hours of sleep on the horizontal axis (Fig. 41.8). The use of the 3-second duration of EEG change is not based
Sleep onset begins with a transition to stage N1 sleep followed on any judgment of physiologic impact, rather it is an arbitrary
quickly by stage N2. Stage N3 (formerly NREM stage 3 or slow- value chosen by the task force partly due to increased interob-
wave sleep) comes next and is particularly sustained in this first sever reproducibility of arousal scoring (as compared to shorter
sleep cycle in children and young adults. Sleep then briefly light- duration events). The requirement for an accompanying EMG
ens to stage N2 and transitions into stage R for the first time, change in stage R is due to the routine appearance of faster EEG
usually about 90 minutes after sleep onset. This completes the frequencies during normal sustained stage R sleep.
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 827

Figure 41.7 A: An example of stage R sleep with


low-amplitude mixed frequency EEG activity,
absent axial EMG activity (chin channel repre-
sents submental EMG), and REMs (out-of-phase
activity in channels E1 and E2). (30-second
epoch). B: Another example of stage R sleep. Note
2 to 3 Hz low-amplitude sharp activity (seen in
C4–M1 channel) just prior to REMs. This activity
represents sawtooth waves, a pattern frequently
seen in stage R.

Periodic Limb Movements in Sleep Scoring During PSG, the PLMS are quantified as to how many
Periodic limb movements in sleep (PLMS) (also called periodic events occurred and also whether an EEG arousal was associ-
leg movements of sleep) are repetitive, stereotyped movements ated with them (Fig. 41.11A,B). A significant leg movement is
of the legs characterized by tonic extension of the great toe defined as an increase in EMG lasting between 0.5 and 10 sec-
(with occasional superimposed clonic activity) variably accom- onds. The EMG increase has to be at least 8 V above the rest-
panied by ankle dorsiflexion and knee flexion (38). These peri- ing EMG. The leg movement is labeled as part of a PLM series
odic movements can rarely preferentially involve the arm and if it is one of the four consecutive leg movement events occur-
thus the use of the term PLMS. These periodic movements may ring with an interval between 5 and 90 seconds. If each leg
sometimes be associated with an accompanying EEG arousal. movement event is separated by less than 5 seconds, they are
PLMS are commonly seen on PSGs particularly in the setting of considered a single leg movement. Arousals are scored as asso-
disrupted sleep or in the elderly. The clinical significance of ciated with the leg movement if there is less than 0.5 second
PLMS is often unclear, but may sometimes contribute to interval between the end of one event and the onset of the
insomnia or daytime sleepiness. In 1980, Coleman and co- other regardless of which is first. Limb movements should not
workers (38) made a major contribution in understanding be scored if they occur within 0.5 second of the onset or end
PLMS and various rules of scoring the movements have been of an apnea or hypopnea.
proposed, including that from the ASDA task force in 1993 (39) Similar periodic leg movements can be seen in wakefulness,
and the World Association of Sleep Medicine—International particularly in patients suffering from restless legs syndrome
Restless Legs Syndrome Study Group (IRLSSG) in 2006 (40). (RLS). RLS is a disorder where the patient suffers from an
From here on we will focus on the recent guidelines given in the uncomfortable sensation in their legs, which is worse at night,
AASM scoring manual. worse while at rest, and relieved with movement. RLS is a
828 Part V ■ Complementary and Special Techniques

clinical diagnosis made in the clinic based primarily on the his-


tory and does not require PSG evaluation. PLMS is a diagnosis
made on the PSG as most patients do not have daytime symp-
toms and observer history is not usually helpful. Most patients
with RLS will also have PLMS, but PLMS most commonly
occur in the absence of restless legs symptoms. Periodic limb
movements in wakefulness (PLMW) are not part of the recently
published AASM scoring guidelines (12). However, several
investigators have suggested the utility of PLMW in assessing
patients with RLS. The scoring criteria are exactly the same as
those outlined above for the scoring of PLMS. These investiga-
tors have also proposed a SIT test (suggested immobilization
test) with associated quantification of PLMW as a diagnostic
test for patients with RLS (41).

Scoring of Respiratory Events


With the advent of the recognition of apneic events during
sleep (42), the cessation of identified airflow for 10 seconds has
been the standard definition of an apneic event. With subse-
quent recognition of the significance of partial airflow inter-
ruption (hypopneas), no consistent standard definition was
accepted and technical and scoring standards varied widely. The
recent AASM scoring manual defines both technical recording
requirements as well as scoring rules (12) which hopefully will
lead to an improvement in the standardization of quantifying
respiratory abnormalities during sleep.
The sensor used to score apnea is the oronasal thermal sensor
(thermistor or thermocouple) that has long been used to qualita-
tively assess airflow during PSG recording. The sensor for detec-
tion of a hypopnea is the NPT. The NPT is much more sensitive
to the identification of a decrease in airflow and is appropriate to
Figure 41.8 Histograms representing normal sleep cycles for age. REM use for the identification of a hypopnea. Frequently during a
sleep represented by darkened area. Note NREM stages 3 and 4 would study, a clear decrease in airflow is identified in the NPT, with no
now be combined into stage N3. Horizontal axis is hours of PSG record- evidence of change in the thermal sensor. Previously, it was com-
ing. (From Kales A, Kales J. Recent findings in the diagnosis and treat- mon to recognize cyclical arousals associated with loud snoring
ment of disturbed sleep. N Engl J Med. 1974;487–499, with permission.) and oxygen desaturation that were not scorable using the less
sensitive thermal sensors. This led to the recognition of the upper
airway resistance syndrome in which the patient had a clinical

Figure 41.9 Example of arousal from NREM


sleep (stage N2) with appearance of fast activity
lasting at least 3 seconds. Note increase in EMG
activity, which commonly accompanies an EEG
arousal pattern but is not required for scoring an
arousal from NREM sleep (30-second epoch).
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 829

Figure 41.10 An arousal from stage R with


appearance of 3 seconds of faster frequencies
accompanied by the appearance of EMG in the
axial (chin) EMG channel (30-second epoch).

Figure 41.11 A: An illustration of periodic limb


movements of sleep. This is a 2-minute epoch
with the appearance of EMG activity in the right
anterior tibialis (RAT) channel occurring approx-
imately every 30 seconds. The patient is in NREM
sleep (which is not able to be confidently deter-
mined in this 2-minute epoch). B: A 30-second
epoch taken from the time period illustrated in A.
Note the appearance of faster frequencies on the
EEG demonstrating an arousal associated with
this periodic movement.
830 Part V ■ Complementary and Special Techniques

syndrome consistent with OSA but without scorable apneic lasting at least 10 seconds characterized by increasing respira-
events. With the use of NPT and the associated identification of tory effort or flattening of the nasal pressure waveform leading
scorable hypopneas, it is much rarer to have a clinical suspicion to an arousal from sleep when the sequence of breaths does not
of unrecognized respiratory events contributing to a patient’s meet criteria for an apnea or hypopnea.” Flattening of NPT
symptoms. However, the NPT is frequently overly sensitive in waveform is thought to identify increased airway pressure
that it demonstrates no airflow (an apnea) while the thermal sen- reflecting partial airway occlusion. The flattening of the nasal
sor continues to demonstrate obvious airflow. As such, the NPT waveform is illustrated in Figure 41.13.
is not to be used for determination of apneic events, which are to The number of scored respiratory events is then divided by
be scored off of the thermal sensor recording. the hours of sleep to yield an AHI (apnea–hypopnea index)
An apnea is scored using the thermal sensor when there is a which represents the number of respiratory events per hour of
90% decrease in identified airflow lasting at least 10 seconds. sleep. This value is most commonly used to categorize the
The apnea is classified based on the accompanying inspiratory severity of respiratory abnormality. The consensus statement
effort identified using the inductive plethysmography belts from from the AASM classifies apnea severity as per the following
the chest and abdomen. An apnea is labeled as obstructive if there criteria: AHI 5: normal; AHI 5 but 15: mild apnea; AHI
is continued or increased respiratory effort throughout the entire 15 but 30: moderate apnea; AHI 30: severe apnea (33).
period of absent airflow (Fig. 41.12B). The apnea is classified as Many observers suggest this categorization of apnea severity
central if the event has no associated inspiratory effort through- may be too severe, particularly given the lack of normative data.
out the entire apneic period (Fig. 41.12A). The event is scored as Several large studies have demonstrated that nonobese healthy
a mixed apnea if there is initially an absence of inspiratory effort middle-aged adults have an AHI 5 in 20% of individuals, and
followed by resumption of inspiratory effort in the later portion an AHI 15 in approximately 6% of individuals. The AHI is
of the event (Fig. 41.12C). The duration of the absent respiratory also recognized to increase with age, even in individuals with no
effort needed to score a mixed apnea is not defined, but usually sleep complaint (43–45).
would be at least one complete breath cycle (4 to 6 seconds). It
should be noted that there is no requirement for any accompany- Scoring of Cardiac Events
ing oxygen desaturation or EEG arousal to score an apnea. Before the AASM scoring manual (12), there had been no
Two separate rules for scoring hypopneas were presented in attempt to define or standardize the documentation or scoring
the AASM scoring manual (12). The “recommended” rule is of cardiac events during a PSG. The AASM scoring manual
that used by Medicare and was chosen in an attempt to be in defined the use of a modified ECG lead II (recording electrodes
concert with Medicare definitions and decision regarding cov- placed on torso at approximately second rib to right of sternum
erage of CPAP and other therapies. However, the “alternative” and the sixth rib near the apex of the heart on the left chest).
set of rules for scoring hypopneas is actually more widely used The following ECG scoring rules for adults were defined:
and captures a larger number of hypopneas, particularly in
1. Score sinus tachycardia for sustained sinus heart rate 90
individuals with healthy lungs where oxygen desaturations
beats per minute.
occur much less frequently.
2. Score sinus bradycardia for sustained sinus heart rate 40
The recommended rule for scoring a hypopnea requires the
beats per minute.
following:
3. Score asystole for cardiac pauses 3 seconds.
1. NPT signal decreases by 30% for at least 10 seconds. 4. Score wide complex tachycardia for a rhythm of at least three
2. There is an accompanying oxygen desaturation of 4% or consecutive beats at a rate greater than 100 beats per minute
more. and a QRS duration of 120 msec.
5. Score narrow complex tachycardia for a rhythm of at least
The alternative rule for scoring a hypopnea requires the
three consecutive beats at a rate 100 beats per minute and
following:
a QRS duration of 120 msec.
1. NPT signal decreases by 50% for at least 10 seconds. 6. Score atrial fibrillation if there is an irregularly irregular ven-
2. There is an accompanying oxygen desaturation of 3% or tricular rhythm associated with replacement of consistent P
more or the event is accompanied by an EEG arousal within waves with variable rapid oscillations.
3 seconds of the event. 7. Other significant arrhythmias (such as heart block) should
be reported if quality of the single lead is sufficient for accu-
Either scoring method is acceptable but the rules used
rate identification.
should be clearly defined in the PSG report. Most labs do not
attempt to classify hypopneic events as obstructive or central
due to the inaccuracy of assessing respiratory effort in the SCORING OF CYCLIC
absence of more invasive monitoring techniques such as ALTERNATING PATTERN
esophageal manometry.
An optional scoring rule outlines the scoring of respiratory The cyclic alternating pattern or CAP was first described by
effort–related arousals (RERAs). This may be particularly help- Terzano and colleagues in 1985 (46). CAP has been proposed as
ful in identifying potential clinically significant respiratory a tool for the comprehensive analysis of sleep microstructure in
events if only the recommended rule for hypopnea identifica- both normal and pathologic conditions. Traditional sleep scoring
tion is used. A RERA is scored if “there is a sequence of events using the criteria of Rechtschaffen and Kales (11) has been the
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 831

Figure 41.12 A: Limited PSG montage demon-


strating no evidence of airflow as measured by
nasal pressure transducer (PTAF channel),
oronasal thermocouple (flow channel) accompa-
nied by the absence of ventilatory effort in the
thorax, and abdominal channels (measured by
respiratory inductive plethysmography belts).
This is an example of a central apnea lasting
approximately 16 seconds. B: Limited PSG mon-
tage demonstrating no evidence of airflow as
measured by nasal pressure transducer (PTAF
channel), oronasal thermocouple (flow channel)
accompanied by the evidence of continued venti-
latory effort in the thorax channel (measured by
respiratory inductive plethysmography belts).
This is an example of an obstructive apnea lasting
approximately 18 seconds. Note the appearance
of snore artifact (in snore and PTAF channel) with
resumption of airflow. C: Limited PSG montage
demonstrating an apneic event lasting approxi-
mately 40 seconds (in this 60-second epoch). No
evidence of airflow is noted by the nasal pressure
transducer (PTAF channel) or the oronasal ther-
mocouple (flow channel) throughout the event
identifying it as an apnea. During the first half of
the apneic period, there is no evidence of ventila-
tory effort, but then increasing evidence of effort
in the thorax channel is seen leading up to the ter-
mination of the apneic event. This is an illustra-
tion of a mixed apnea, having central features at
onset but then demonstrating obstructive features
as the event progresses.
832 Part V ■ Complementary and Special Techniques

Figure 41.13 Polysomnographic recording show-


ing an example of upper airway resistance syn-
drome. Note that peak increase in effort (indicated
by the solid arrowhead) is associated with a small
drop in peak flow and tidal volume, triggering a
transient electroencephalographic arousal. ECG,
electrocardiogram; EMGFACIAL, facial muscle elec-
tromyogram; EOG, electro-oculogram (right and
left); FLOWPNEUMOTACH, pnuemotachometer to
quantify airflow; Pes, esophageal manometry to
record esophageal pressure; RESPSUM, respiratory
effort; SaO2, saturation with oxygen. (Reproduced
from Chokroverty S. Sleep Disorders Medicine:
Basic Science, Technical Considerations and Clinical
Aspects. 3rd ed. Philadelphia: Saunders/ Elsevier;
2009, with permission.)

gold standard for looking at sleep macrostructure. CAP presents A CAP sequence is composed of repetitive CAP cycles, with
another way of looking at NREM sleep within those sleep stages. each cycle composed of a phase A and a phase B. Each phase of
NREM sleep has been recognized to have high amplitude EEG the CAP is 2 to 60 seconds in duration. If there is no phase A
bursts such as K-complexes or delta bursts. These are often seen activity for 60 seconds, then that portion of NREM sleep is
as an arousal response to external stimuli but also occur when scored an non-CAP. Figure 41.14 illustrates an example of the
sleep disturbance was not evident. An alternative view to this CAP in stage 2 sleep. Figure 41.15 illustrates a brief period of
arousal process is that these phenomena are associated with sleep CAP surrounded by non-CAP sleep on either side. Phase A can
instability (due to an internal or external challenge to the sleep take on three patterns: A1 shows primarily slow ( 1 Hz) delta
process) and that this type of slow wave activity marks the brain’s activity with a small degree of autonomic activation; A3
attempt to preserve or sustain the sleep state. If sleep becomes too demonstrates an increase in fast rhythms with strong auto-
unstable, the preservation attempt fails and the high-amplitude nomic activation (an EEG arousal as per ASDA rules) (37); A2
activity is accompanied by a more complete EEG arousal. It is represents a middle ground between the A1 and A3 subtypes
proposed that the addition of a periodicity dimension to the con- (Fig. 41.16). A summary paper outlining the rules for scoring
cept of sleep stability and arousal will provide a valuable perspec- CAP and including an atlas to illustrate those rules was pub-
tive on sleep and its relationship to underlying physiologic and lished by Terzano and colleagues in 2001 (47).
pathophysiologic mechanisms. CAP represents a slow oscillation In several studies, the CAP A-phase is identified as the “gate”
of EEG activity that manifests in the appearance of EEG arousal or “amplifier” that allows the appearance of pathologic events
patterns with a periodicity of 20 to 40 seconds. such as PLMS, bruxism, sleepwalking, and sleep-disordered
CAP is represented by three characteristics: breathing to occur. The majority of nocturnal partial seizures
presenting during NREM sleep also occur predominantly in
1. The recurring high-amplitude EEG activity (phase A of the
CAP in association with phase A (in particular in association
period)
with K-complexes and delta bursts) (48). At this point, the clin-
2. The intervening background EEG activity (phase B of the
ical significance of CAP remains unclear but further research is
period)
warranted.
3. The period or cycle that is the sum of phase A and phase B
Figure 41.14 An example of CAP in stage N2.
The box outlines a CAP cycle (C) composed of
phase A and following phase B. Bipolar EEG
channels for first six channel top to bottom
Fp2–F4, F4–C4, C4–P4, P4–O2, F8–T4, and
T4–T6; bottom eight channels are top to bottom
Fp1–F3, F3–C3, C3–P3, P3–O2, F7–T3, T3–T5,
EOG, and ECG. (From Terzano et al. Atlas, rules,
and recording techniques for the scoring of cyclic
alternating pattern (CAP) in human sleep. Sleep
Med. 2001;2:537–553, with permission.)

Figure 41.15 Consecutive stretches of non-CAP


(top), CAP (middle), and non-CAP (bottom).
The middle section illustrates the minimal
requirements for definition of a CAP sequence (at
least three phase A’s in succession). The CAP
sequence occurs between two black arrows and
the transition between phases is delineated by
the dotted line. EEG derivation is Fp2–F4, F4–C4,
C4–P4, P4–O2, and C4–A1. (From Terzano et al.
Atlas, rules, and recording techniques for the
scoring of cyclic alternating pattern (CAP) in
human sleep. Sleep Med. 2001;2:537–553, with
permission.)

Figure 41.16 The three phase A subtypes. The


dotted line indicates the fast low-amplitude por-
tion of phase A. EEG derivation as in Figure 41.15.
(From Terzano et al. Atlas, rules, and recording
techniques for the scoring of cyclic alternating
pattern (CAP) in human sleep. Sleep Med. 2001;
2:537–553, with permission.)

833
834 Part V ■ Complementary and Special Techniques

INDICATIONS FOR PSG AMBULATORY PSG RECORDING


The indications for PSG have been summarized by the pub- Ambulatory recording of sleep in the evaluation of sleep disor-
lished practice parameters of the AASM (49,50). The domi- ders has been rarely utilized due to the limitations of the tech-
nant indication for PSG is for the evaluation of sleep-related nique and the lack of third-party reimbursement (51). With the
breathing disorders. The increasing awareness of sleep apnea recent CMS (Centers for Medicare and Medicaid Services)
and its possible impact on long-term cardiovascular health approval for coverage and the push to control health care costs,
has lead to a marked increase in evaluation of sleep- the role of ambulatory or home sleep tests (HSTs) is being re-
disordered breathing. While the obese individual who pres- evaluated (52). As outlined earlier in this chapter, in-laboratory
ents with snoring, witnessed apneas, and excessive daytime PSG is the gold standard for the evaluation of sleep disorders and
sleepiness (EDS) represents the obvious candidate for PSG is classified as a type 1 study. A full but unattended PSG (seven or
evaluation, there are many others who likely need PSG evalu- more channels including the recording of EEG) is classified as a
ation. Symptoms of concern for possible sleep apnea include type 2 study. The focus on the HSTs has been with the use of type
daytime sleepiness, loud snoring, witnessed apneas, and unre- 3 studies which have four to seven channels and focus on the
freshing sleep. Additional medical conditions that warrant evaluation of respiratory parameters, but without recording of
exploring a possible role of OSA include intractable hyperten- EEG and therefore have no evaluation of sleep stages or arousals.
sion, new-onset atrial fibrillation (particularly with onset in A type 4 study is a one- or two-channel overnight recording, with
sleep), intractable congestive heart failure, and stroke. Once at least one of them being oximetry (53).
the diagnosis of sleep apnea has been documented, patients In the typical HSTs, the patient presents to the laboratory
usually return for repeat study for CPAP titration. The goal is during the day and is educated regarding use of the equipment
to document the minimum CPAP pressure that eliminates and placement of electrodes and recording devices. After prepar-
apnea, hypopneas, and arousals with particular attention to ing for bed, the patient places the recording sensors and pushes
stage R sleep and supine sleep. It is recommended that a button indicating that they are retiring for the night. The but-
patients undergoing upper airway surgery to treat snoring or ton is pressed again upon awakening in the morning and a total
apnea have a study to document apnea severity and ensure recording time is obtained, which is supplemented by a brief
appropriate choice of therapy and perioperative management. sleep log from the patient outlining any sustained periods of
Similarly, after upper airway surgery to treat apnea, a follow- wakefulness or other important information. Usual recorded
up PSG is warranted to document the residual degree of sleep parameters include nasal/oral airflow, respiratory effort, oxygen
apnea and whether additional therapeutic efforts are saturation, body position, snoring, and heart rate. The studies
warranted. are scored by automated scoring systems, but the data should be
In addition to the evaluation of sleep-disordered breathing reviewed and edited by a technologist to ensure that artifactual
(SDB), PSGs are also utilized to evaluate other causes of EDS, information is discarded and that the results represent a reason-
sleep disruption, and unusual nocturnal behavior. In an indi- able interpretation of the data. From this recording, an estimate
vidual with EDS where sleep apnea is not a concern, a PSG can of an AHI and oxygen desaturation index is obtained.
be done to evaluate possible PLMS as a contributor to the The recent guidelines from the AASM for unattended sleep
patient complaint. PSG is routinely done the night before an studies outline minimal technical expectations for portable
MSLT (discussed later in this chapter) to document adequate monitoring (53). HSTs should at a minimum record airflow,
sleep quantity and quality to allow interpretation of the multi- respiratory effort, and blood oxygenation and should use the
ple nap study. In patients with RLS, PSG is not routinely done same sensors that are used in the laboratory studies. Accurate
as this is a clinical diagnosis and the restrained environment of data collection is paramount to successful use of the techniques,
the sleep laboratory can be very uncomfortable for the sympto- so the devices should not be used by technologists or physicians
matic patient with RLS. PSG may be appropriate if a patient who are inexperienced in sleep disorders and their evaluation.
with RLS does not respond to therapy or has other potential The devices must also allow review of the raw data so that man-
sleep pathology. Similarly, PSG is not routinely indicated in ual scoring and/or adjustment of the automated scoring algo-
patients presenting with insomnia. Both behavioral and phar- rithms can be made.
macologic therapies are pursued before considering PSG evalu- The advantages of HSTs are obvious and include lower cost
ation in a patient with insomnia. and greater patient accessibility. Patients who cannot access in-
Most PSG recordings are presently done with video accom- laboratory studies due to laboratory backlog or physical immo-
panying the polygraph recording. Video recording is absolutely bility can be evaluated with HST. Other patients with irregular
essential if the goal is to evaluate unusual nocturnal behavior. or unusual sleep–wake cycles can also have an evaluation that
The diagnosis of a parasomnia can usually be made on a clini- does not have to fit into the usual laboratory schedule. The pri-
cal basis and a video PSG is not required in many patients. mary disadvantages include unreliable or lost data and the lack
However, if there are unusual features or a concern about coex- of any assessment of sleep stage or quality.
istent sleep problems, PSG may be warranted. If the differential Because of their obvious limitations, portable or ambulatory
diagnosis include nocturnal seizures, additional EEG channels sleep studies are not appropriate in many clinical settings and
are often helpful in more confidently differentiating parasom- this is recognized by the CMS guidelines as well as those of
nias from epileptic seizures. the AASM. Both bodies agree that the HST may be used as an
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 835

alternative to in-laboratory PSG for the diagnosis of OSA in to document large negative intrathoracic pressure swings that
patients with a high pretest probability of moderate to severe correlate with the EEG arousals. Esophageal manometry is not
OSA. If positive, the caregiver can move forward with therapy commonly used in the sleep laboratory due to its effect on sleep
for the significant respiratory abnormality. Additionally, HSTs and patient comfort. In the last several years, the consistent use
can be helpful after CPAP therapy has been initiated to assess of the NPTs to monitor airflow and airway pressure has
adequacy of treatment if there is a concern regarding response markedly increased our sensitivity to this phenomena, with
to therapy (52–55).- many of these events now being recognized as hypopneas due
However, HST is not indicated for use in patients with sig- to the superior identification of airflow resistance by NPT
nificant comorbidity (COPD, congestive heart failure, neuro- (60–62).- However, the caveat remains that the sleep physician
muscular disease) as the coexistence of these disorders may lead needs to be sensitive to the possibility of the under recognition
to a greater degree of inaccuracy. HST has not been evaluated in of respiratory events in the sleep laboratory.
asymptomatic but high-risk individuals such as bariatric
surgery patients or commercial truck drivers. ARTIFACTS DURING PSG RECORDING
Computerized PSG and Computer-Assisted Scoring PSG recording frequently contains extraneous electrical activi-
Visual sleep scoring has been the standard for analyzing sleep ties not originating from the brain, eyes, heart, or other regions
from the very beginning and subsequent standardizations of of interest interfering with the biologic signals of interest (63).
sleep scoring has emphasized that (11,12). Any attempt to uti- These include physiologic, environmental and instrumental
lize computerized sleep scoring in clinical sleep studies has artifacts.
attempted to model itself after the visual sleep analysis.
Although it is extremely useful for quantifying easily identified Physiologic Artifacts
events (oxygen desaturations), its inability to adjust to hostile It is important to recognize physiologic artifacts resulting from
“pathologic states” such as frequent apneas, disrupted sleep, movements of the head, eyes, and other body parts (Figs. 41.17
and movement artifacts has limited its clinical application. through 41.19), rhythmic leg movements causing tremor-like
Although many of the commercially available PSG equipment artifacts (Fig. 41.20), movements originating from muscles
do come with computerized analysis of sleep stage, the clinical causing myogenic artifacts (Fig. 41.21), and movements of the
standard remains visual scoring by a trained technologist or, at heart giving rise to electrocardiographic artifacts (Fig. 41.22),
least, the review and over scoring of the computerized sleep which may interfere with recordings from the regions of inter-
staging by a technologist. Similarly, computerized identification est and may sometimes be mistaken for slow waves in the EEG.
of respiratory or movement events also requires review and Sweating by causing excessive baseline swaying produces very
editing by a technologist. slow oscillations lasting for 1 to 3 seconds mainly during non-
As part of the efforts to create the new scoring manual, there REM sleep as a result of action potentials generated by salt con-
was a critical evidence-based look at computerized analysis of tent of the sweat glands (Fig. 41.23).
PSG. The individuals in charge of this section suggested that
“computer scoring and quantitative analysis are still in the Environmental Artifacts
formative stage of development” and have not been proved to The most important environmental artifact is the 60-Hz artifact
be useful in clinical practice (56). (Fig. 41.24) resulting from electromagnetic radiation from AC
The real strength of computerized analysis of sleep is in its current in power lines (in many other countries the main fre-
ability to look at sleep as a continuum rather than the arbitrary quency is 50 Hz). Most often this results from high impedance
definitions provided for identifying stages N1, N2, N3, and of the electrodes. The impedance should be kept below 5 K.
stage R sleep. Similarly, the microstructure of sleep is likely lost
in the 30-second epoch. Research is still needed to determine Instrumental Artifacts
whether this technology will contribute to new methods for These artifacts result from faulty electrodes (Fig. 41.25) resem-
understanding sleep and its disorders (57). bling transient sharp or slow waves limited to one electrode,
wires, switches, and the PSG machine itself due to random
fluctuations of charges causing instrumental noise artifacts.
PITFALLS OF PSG
One of the biggest pitfalls of PSG is the recognition of respira- CLINICAL CONSIDERATIONS
tory events that may be clinically significant but may not be
consistently recognized by the sensors used for the recording. Narcolepsy and Other Hypersomnias
The clearest example of this is the upper airway resistance syn- Narcolepsy is a disorder of EDS that is usually accompanied by
drome (UARS) (58,59). UARS is a syndrome in which patients other “auxiliary” symptoms such as cataplexy, hypnagogic hal-
experience repetitive increases in upper airway resistance that lucinations, and sleep paralysis. Additional features commonly
result in brief arousals with associated EDS. Not all of these include disrupted night-time sleep and excessive motor activity
individuals snore, and the repetitive arousals can sometimes be in sleep. A more complete review of the clinical features of nar-
attributed to other causes such as PLMS. The “gold standard” of colepsy can be found in the literature (64,65). During the initial
documenting this disorder is the use of esophageal manometer diagnostic evaluation of a patient with possible narcolepsy, a
836 Part V ■ Complementary and Special Techniques

Figure 41.17 A 30-second excerpt from


overnight PSG recording shows body movement
artifact following an obstructive apnea as demon-
strated by variable, high-voltage, asymmetrical
and asynchronous slow activity on EEG channels
and increased electromyography activity on chin
and tibialis anterior electrode channels. Top 10
channels: EEG; LT and RT EOG, left and right
electro-oculograms; chin EMG, electromyogra-
phy of chin muscles; Lt. and Rt. Tib EMG, left
and right tibialis anterior electromyography;
P Flow, peak flow; oronasal thermistor; chest
and abdomen effort channels; snore monitor;
EKG, electrocardiography. (Reproduced from
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of
Sleep Medicine. Philadelphia: Elsevier; 2005, with
permission.)

PSG is usually done in tandem with an MSLT. The PSG excludes identification of sleep-onset REM (SOREM) in the brief nap
possible pathologic entities such as sleep apnea or PLMS that trials. In addition, it has long been recognized that patients with
can contribute to EDS. The PSG is also used to document ade- narcolepsy seem to have an increased incidence of sleep apnea,
quate sleep duration (usually 6 hours) and sleep quality to PLMS, RBD, and sleep-related eating disorders.
allow confident interpretation of the MSLT performed the fol- Idiopathic hypersomnia is another disorder of EDS that also
lowing day. The focus of the MSLT is to document the patho- presents in adolescence or young adulthood. However, it does
logic hypersomnolence (as a shortened sleep latency) not have the accompanying auxiliary symptoms or SOREM
and evaluate any abnormal REM sleep pressure through the sleep noted in narcolepsy. The EDS of idiopathic hypersomnia

Figure 41.18 A 30-second epoch from overnight


PSG showing repetitive blink artifact best noted
in the electro-oculogram channels and on the
frontal and anterior temporal EEG electrode
recordings. This can be misinterpreted for
abnormal cerebral activity. Top 10 channels:
EEGs; LT and RT EOG, left and right electro-ocu-
lograms; chin EMG, electromyography of chin
muscles; Lt. and Rt. Tib EMG, left and right tib-
ialis anterior electromyography; P. Flow, peak
flow; chest and abdomen effort channels; snore
monitor; EKG, electrocardiography. (Reproduced
from Chokroverty S, Thomas R, Bhatt M, eds.
Atlas of Sleep Medicine. Philadelphia: Elsevier;
2005, with permission.)
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 837

Figure 41.19 A 20-second epoch from overnight


PSG showing rapid eye flutter artifact best
recorded in F3 and F4 electrode recordings dur-
ing the latter half of the epoch. A blink artifact is
noted in the initial part of the recording and is
best seen on the electro-oculogram channels.
Well-formed alpha rhythm is noted in the back-
ground on all EEG channels when eyes are
closed. (Reproduced from Chokroverty S,
Thomas R, Bhatt M, eds. Atlas of Sleep Medicine.
Philadelphia: Elsevier; 2005, with permission.)

is usually less severe than that seen in narcolepsy, but it has a noted. More recent studies (68,69) have expanded on the obser-
more pervasive all-day character that is not impacted by day- vations of PSG in narcolepsy patients. The night-time sleep of
time naps. Night-time sleep in idiopathic hypersomnia is usu- narcoleptics is characterized by increased awakenings,
ally described as long and uninterrupted (66). decreased stage N1, decreased stage N3, and shortened REM
An early study of night-time sleep in narcolepsy and idio- latency. Total sleep time and total stage R (REM sleep) are not
pathic hypersomnia (67) confirmed most of these clinical effected. Evidence of abnormal motor activity during sleep is
impressions. Patients with idiopathic hypersomnia demon- prominent in narcolepsy (particularly in patients with accom-
strated better sleep efficiency, less stage N1, more stage N3, less panying cataplexy). PLMS index was greater than 15 in approx-
WASO, and longer REM latency. In addition, the coexistence of imately 75% of middle-aged narcolepsy patients, and somewhat
sleep-disordered breathing and PLMS was less frequently uniquely they also had PLMS during REM sleep, something

Figure 41.20 Overnight PSG from a 50-year-old


man. Nocturnal PSG shows the presence of mild
sleep apnea with an AHI of 12.3. A 30-second
excerpt from nocturnal polysomnography shows
bursts of rhythmic foot and leg movements on
the left/ right tibialis anterior muscle recording
channel during stage 2 NREM sleep (A). The
movement is not associated with respiratory
events, oxygen desaturation, or arousal from
sleep. Top 10 channels: EEGs; LEFT and RT EOG,
left and right electro-oculograms; chin EMG, elec-
tromyography of chin muscles; Lt/ Rt Tib EMG,
left/ right tibialis anterior EMG; oronasal thermis-
tor; chest and abdomen effort channels; snore
monitor; EKG, electrocardiography; SaO2, oxygen
saturation by finger oxymetry. (Reproduced from
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of
Sleep Medicine. Philadelphia: Elsevier; 2005, with
permission.)
838 Part V ■ Complementary and Special Techniques

Figure 41.21 A 30-second excerpt of PSG


recording shows spikelike potentials of varying
amplitude and frequency denoting muscle activ-
ity not only on the chin electromyography chan-
nel but also in all EEG channels, particularly
prominent in T3, T4, A2, and A1 electrode deri-
vations. Top 10 channels: EEGs; LT and RT EOG,
left and right electro-oculograms; chin EMG, elec-
tromyography of chin muscles; Lt. and Rt. Tib
EMG, left and right tibialis anterior electromyog-
raphy; P Flow, peak flow; oronasal thermistor;
chest and abdomen effort channels; snore moni-
tor; EKG, electrocardiography. (Reproduced from
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of
Sleep Medicine. Philadelphia: Elsevier; 2005, with
permission.)

which is distinctly unusual in non-narcolepsy patients. Even somnia. KLS is a rare disorder with onset during teenage years
more remarkable is the high incidence of RBD. In a group of and is predominantly seen in men. Diagnosis is based on the
narcolepsy patients off medication (68), 60% provided a his- clinical presentation of recurrent episodes of hypersomnia along
tory of RBD and 40% demonstrated RBD during PSG. In addi- with at least one of the following symptoms during the hyper-
tion, another 14% had evidence of REM sleep without atonia. somnolent episodes: binge eating, hypersexuality, irritability,
It is clear that excessive and pathologic motor activity during aggression, other odd behaviors, or cognitive abnormalities. The
sleep is common in individuals with narcolepsy. pathogenesis is unknown with hypotheses centering on neuro-
Episodic hypersomnias are also described and include transmitter imbalance or autoimmune mechanisms. There is no
Kleine–Levin syndrome (KLS) and menstrual-associated hyper- consensus on accompanying PSG findings in KLS, likely related

Figure 41.22 This is a 30-second excerpt from


an overnight PSG selected to show electrocardio-
graphy artifact, which is characterized by its
morphology, rhythm, and synchrony with elec-
trocardiographic recording. It is seen throughout
the epoch on electroencephalography, electro-
oculography, and tibialis electromyography chan-
nels. Incidentally, an obstructive apnea is seen in
the airflow and effort channels. Top 10 channels:
EEGs; LEFT and RT EOG, left and right electro-
oculograms; CHIN, electromyography of chin;
Lt/ Rt Tib EMG, left and right tibialis anterior
electromyography; oronasal thermistor; chest
and abdomen effort channels; snore monitor;
EKG, electrocardiography. (Reproduced from
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of
Sleep Medicine. Philadelphia: Elsevier; 2005, with
permission.)
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 839

Figure 41.23 Sweat artifact at 30 seconds.


(Reproduced from Siddiqui F, Osuna E, Walters
AS, et al. Sweat artifact and respiratory artifact
occurring simultaneously in polysomnogram.
Sleep Med. 2006;7(2):197–199, with permission.)

to variability in the timing of the studies with reference to the reported but are not consistently noted (71). Menstrual-associ-
hypersomnolent period. One recent study (70) documented a ated hypersomnia is a disorder in which a young woman (usu-
reduction in slow wake sleep early in the hypersomnolent phase, ally near the time of menarche) experiences excessive sleep need
returning to normal during second half, at which time REM and daytime sleepiness in association with her menses. It is usu-
sleep was decreased. Other studies suggested increased total ally improved with oral contraceptive medication and has not
sleep time, reduced sleep efficiency, increased WASO, decreased been evaluated extensively in the sleep laboratory.
stage N3, and sometimes shortened REM latency. MSLTs have
consistently demonstrated shortened MSL during the episodic Parasomnias
hypersomnolence (MSL in range of 3 to 5 minutes) and normal NREM parasomnias include confusional arousals, sleep walk-
MSL during asymptomatic intervals. SOREMs have been ing, and sleep terrors (72,73). Typically, arousal parasomnias

Figure 41.24 A 30-second excerpt from an


overnight PSG recording shows a uniform
monorhythmic artifact at 60 Hz on left/ right tib-
ialis electromyography channel, which is further
superimposed by an electrocardiography artifact.
The 60-Hz artifact is due to electrical interference
from power lines and equipment occurring at a
frequency of 60 Hz in North America (but at
50 Hz in many other countries). Maximum inter-
ference is seen in the presence of poor electrode
contact. Incidentally, an obstructive apnea is
noted in the airflow and respiratory effort chan-
nels. Top 10 channels: EEGs; LEFT and RT EOG,
left and right electro-oculograms; chin EMG, elec-
tromyography of chin muscles; Lt/ Rt Tib EMG,
left and right tibialis anterior electromyography;
oronasal thermistor; chest and abdomen effort
channels; snore monitor; EKG, electrocardiogra-
phy; SaO2, oxygen saturation. (Reproduced from
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of
Sleep Medicine. Philadelphia: Elsevier; 2005, with
permission.)
840 Part V ■ Complementary and Special Techniques

Figure 41.25 A 30-second excerpt from an


overnight PSG recording shows the presence of a
C3 electrode pop artifact. Near the middle of the
epoch, it is accentuated by a movement artifact
simultaneously recorded on the chin electromyo-
graphy, left and right tibialis anterior electromyo-
graphy, and snoring channel. Top 10 channels:
EEGs; LEFT and RT EOG, left and right electro-
oculograms; chin EMG, electromyography of chin
muscles; Lt/ Rt Tib EMG, left and right tibialis
anterior electromyography; oronasal thermistor;
chest and abdomen effort channels; snore moni-
tor; EKG, electrocardiography. (Reproduced from
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of
Sleep Medicine. Philadelphia: Elsevier; 2005, with
permission.)

occur in the first third of the night during a transition from routinely indicated for the evaluation of nightmares, but when
slow wave sleep (stage N3) into lighter stages of sleep. All of such an event is recorded, the patient is usually observed to
these parasomnias are more common in children (aged 3 to 10 abruptly awaken out of REM sleep without pathologic motor
years) but can occur in adults. During confusional arousals the activity or any other PSG-identified abnormality.
individual whimpers, moans, moves about slowly, may speak in RBD is a disorder in which the individual usually demon-
short phrases, and resists consoling or attention from others. strates aggressive motor activity during dream enactment.
These events may last up to 30 minutes. Sleep terrors tend to be The patient can be awakened and then usually is able to recall
briefer but are associated with screams and autonomic activa- dream content that explains his behavior. The disorder is
tion (sweating, tachycardia, flushed face). Again the child resists more common in men, particularly in the elderly. It usually
the consoling efforts of the parents and returns to sleep without comes to medical attention when the aggressive dream enact-
memory of the event. Sleep walking events are similar to confu- ment has resulted in injury to the patient or bed partner (75).
sional arousals in terms of time of night, duration, and charac- In up to 50% of patients, it may be a heralding symptom of a
ter of the behavior, but the individual moves about the house in degenerative neurologic condition such as the synucle-
a somewhat slow clumsy fashion and without emotional con- inopathies (Parkinson disease [PD], Lewy body dementia)
tent. In each of these parasomnias, PSG recordings usually doc- (76). It is also seen more frequently in other neurologic disor-
ument generalized, high-amplitude rhythmic delta or theta ders with brainstem dysfunction including multiple sclerosis
activity during the event. No other abnormalities are usually and narcolepsy (77).
identified on PSG. The CAP discussed earlier in this chapter has The behavioral events of RBD occur during REM sleep
been described to be increased in children with sleep terrors. and as such tend to be more frequent in the early morning
This makes sense as CAP represents a pattern of “unstable hours. Frequency is variable and as such a single-night PSG
sleep” and may trigger a variety of events in sleep that can range recording will often not record any of the dream enactment
from epileptic seizures to periodic movements to bruxism to behaviors. Greater attention has been paid to objectively evalu-
confusional arousals. Other sources of sleep disruption, such as ating the degree of muscle atonia present in REM sleep as a way
sleep-disordered breathing and PLMS, may also play a role in to identifying patients with increased risk of dream enactment
precipitating the arousals from slow-wave sleep. (78) (Fig. 41.26). The AASM scoring manual has defined crite-
REM sleep–related parasomnias include nightmare disorder ria for scoring pathologic muscle activity during REM sleep
and REM behavior disorder (RBD). The International (12). Sustained muscle activity (tonic activity) in REM sleep is
Classification of Sleep Disorder, 2nd ed. (ICSD-2) (74) defines defined as an epoch of REM in which at least 50% of the epoch
nightmares as “recurrent episodes of awakening from sleep with duration has a chin EMG amplitude greater than the minimum
recall of intensely disturbing dream mentation, usually involv- amplitude in NREM sleep. Excessive transient muscle activity
ing fear or anxiety, but also anger, sadness, disgust, or other dys- (phasic activity) in REM sleep is defined as when at least 5 of
phoric emotions.” There is generally full alertness upon the 10 three-second “mini-epochs” (in a 30-second epoch) have
awakening and intact recall of dream experience. PSG is not bursts of transient muscle activity. In RBD, the excessive transient
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 841

Figure 41.26 A 60-second epoch of PSG tracing from a 66-year-old man with idiopathic RBD
showing stage R without muscle atonia and with excessive phasic (transient) EMG bursts in cra-
nially innervated (Masseter, Chin, and Sternomastoid), upper limbs (RT Biceps, and LT Biceps),
lower limbs (RT Quadriceps, LT Quadriceps, RT and LT TIBialis, RT and LT Gastrocnemius),
and trunk (RT upper Rectus and RT Lumbar Paraspinal) muscles. Top four channels: EEGs
(international electrode nomenclature); left and right EOGs (E1–M1; E2–M1); EKG, electrocar-
diogram; respiratory air flow (OroNasal and PFlow), and effort (Chest and Abd); HR, heart rate;
Intercostal 1, intercostal muscle EMG; snore; SAO2, oxygen saturation by finger oximetry; BODY,
body position.

muscle activity bursts are 0.1 to 5.0 seconds in duration and at interictal epileptiform discharges. In addition, review of the
least four times the amplitude of background EMG activity. recording utilizing 30 or 60 seconds per page further limits the
The PSG characteristics of RBD can include either or both the identification of epileptiform activity. If PSG is used to evaluate
tonic and phasic EMG observations. REM sleep without atonia interictal epileptiform activity, a full 16- or 18-channel EEG
can occur without accompanying clinical behaviors and as such recording with review at the rate of 10 seconds per page is rec-
RBD remains a clinical diagnosis that can be complemented by ommended (79,80).
the abnormal observations of muscle activity during REM sleep PSG recordings may be utilized to assist in the diagnosis of
as defined by the AASM scoring guidelines (12). Selective sero- unusual nocturnal behavior. In most settings, prolonged video-
tonin reuptake inhibitors and selective norepinephrine reup- EEG recording is used as the primary diagnostic tool if the pri-
take inhibitors commonly provoke REM sleep without atonia, mary diagnostic concern is seizures. Overnight PSG may be
with or without associated clinical RBD episodes. Most investi- used to evaluate potential epileptic seizures, particularly in the
gations have also reported an increased incidence of PLMS in absence of the availability of prolonged video-EEG monitoring.
individuals with RBD. Other abnormalities are not usually PSG does more confidently identify sleep stage (and any
described on PSG. accompanying sleep abnormality such as sleep apnea) than
video-EEG recordings. However, the limited EEG montage used
Sleep-Related Epilepsies on routine PSG represents a significant disadvantage. Foldvary
An extensive overview of sleep and epilepsy is beyond the scope and colleagues (79) published a study in 2000 illustrating that a
of this chapter. However, sleep-related epilepsies are briefly dis- four-channel EEG recording was inadequate for electrographic
cussed in this chapter, with a focus on their differentiation from seizure detection during PSG recording. That same group
other sleep disorders, namely confusional arousals and RBD. recently extended their observations to the evaluation of 8- and
We also briefly review the role that PSG may have in the evalu- 18-channel montages (80). In that study at least 25% of seizures
ation of a patient with epilepsy. with an EEG correlate were missed on the eight-channel
An EEG recording during sleep is more sensitive to the iden- recording. The authors conclude that abbreviated montages are
tification of interictal epileptiform abnormality as compared to inadequate in differentiating seizures from nonepileptic noc-
an EEG recording only during wakefulness. Given the difficul- turnal events during PSG (79,80).
ties in recording a sleep EEG during the daytime (usually Nocturnal frontal lobe epilepsy (NFLE) is a remarkable dis-
requiring sleep deprivation or sedation), the use of an all-night order where the seizures are manifest by bizarre motor behav-
sleep recording to identify interictal epileptiform activity may ior that occurs almost exclusively during sleep. The events are
be useful. No objective data is available to identify how often usually stereotyped, but vary in their duration and complexity.
this may be helpful. Clinical experience has demonstrated that The issue of stereotyped behavior is important as most para-
the limited three- or four-channel EEG montage routinely used somnias will not have the same degree of stereotypy common
during nocturnal PSG is of very limited use in identifying to epileptic seizures. Recording multiple events may help in the
842 Part V ■ Complementary and Special Techniques

identification of the stereotyped nature of these events. problem lies in the ventilatory control mechanism in the central
Interictal EEG recordings are commonly normal and structural nervous system.
brain abnormalities are unusual in patients with NFLE. So the
recording of the events is frequently required for confident Upper Airway Obstructive Sleep Apnea Syndrome
diagnosis. Even with the recording of the events, unambiguous Upper airway OSAS is characterized by repetitive episodes of
ictal EEG correlate may be absent in up to 50% of frontal lobe apnea (see Fig. 41.12A–C) or hypopnea (Fig. 41.27) during
seizures, further complicating the diagnosis (81,82). sleep resulting in arterial oxygen desaturation and repeated
As previously discussed, the usual EEG pattern seen during arousals. OSAS causes significant morbidity and mortality and
a confusional arousal is diffuse hypersynchronous slowing in is often associated with a variety of comorbid conditions. This
the theta or delta range. During RBD, the EEG is that of REM often remains undiagnosed or underdiagnosed because of
sleep. In both cases, superimposed EMG and movement artifact insufficient knowledge and awareness of serious consequences
complicate the interpretation. A wide range of electrographic resulting from this disorder. Apnea or cessation of breathing
patterns may accompany nocturnal frontal lobe seizures, but during sleep can be of three types: central, obstructive, and
bilateral rhythmic sharp waves are most commonly seen. In the mixed. In obstructive apnea, the airflow stops while the effort
absence of a definite ictal electrographic correlate, final diagno- continues (see Fig. 41.12B), whereas in central apnea, both the
sis is often reached clinically. As opposed to parasomnias, the airflow and the upper airways (e.g., pharynx and nose) and the
seizures of NFLE can begin at any age, occur more frequently effort of the diaphragm and other respiratory muscles cease
(often many times) per night, are briefer (several seconds up to (see Fig. 41.12A). An initial period of central apnea followed by
3 minutes), and have a stereotyped pattern that is absent from a period of obstructive apnea constitutes mixed apnea (see Fig.
parasomnias. Several reviews in the literature attempt to review 41.12C). Upper airway OSAS is the most common cause of
how clinical information may help in differentiating the char- apnea in the adult. The AHI (number of episodes of
acter of these nocturnal events (82). apnea–hypopnea per hour of sleep) must be at least 5 to be con-
sidered pathologic. Respiratory disturbance index (RDI) should
Sleep Apnea–Hypopnea Syndrome not be confused with AHI, as RDI includes, in addition to the
Sleep apnea–hypopnea syndrome is divided broadly into two number of apneas and hypopneas, the number of flow limita-
types: upper airway OSAS and central sleep apnea syndrome tions accompanied by arousals per hour of sleep (RERAs).
(CSAS) depending upon pathophysiologic mechanisms. The These rules are meant for the adult sleep scoring of breathing
most common indication for referral of patients to sleep labo- events, but for the pediatric population the rules are slightly dif-
ratories for PSG recording is the suspicion of OSAS. Two ferent (12).
groups of neurologists, Gastaut et al. (42) from France and Jung OSAS is common in middle aged and elderly men. The inci-
and Kuhlo (83) from Germany independently discovered in dence of OSAS in a previously healthy population is not known.
1965 the site of obstruction in the upper airway. In CSAS, the However, based on a definition of AHI of 5 per hour of sleep

Figure 41.27 A 120-second excerpt from stage


N2 showing an initial brief period of obstructive
apnea followed by two sequential hypopneas.
Top 10 channels: EEGs (international 10 to 20
nomenclature); LT and RT EOG: left and right
electro-oculograms; chin EMG, electromyogra-
phy of chin muscles; Lt. and Rt. Tib EMG, elec-
tromyography of left and right tibialis anterior
muscles; P FLOW, peak flow; nasal pressure
recording; oronasal, thermistor for air flow; chest
and abdomen, respiratory effort channels; EKG,
electrocardiogram; SAO2, oxygen saturation by
finger oximetry.
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 843

accompanied by EDS, the prevalence of OSAS is thought to be arousals on resumption of breathing with increased arousal
4% in men and 2% in women between the ages 30 and 60 years index (the number of arousals per hour of sleep). Percentage of
(43). In women, the prevalence of OSAS rises after menopause. oxygen desaturation associated with abnormal breathing events
The pathogenesis of OSAS includes both local anatomical fac- may be classified into mild with desaturation of 80% to 89%,
tors (e.g., narrowing of the upper airway and excessive relax- moderate to moderately severe with desaturation of 70% to
ation of the upper airway muscles during sleep, increased upper 79%, and severe with 69% or below. An arousal index may be
airway resistance) and neurologic factors causing dysfunction arbitrarily considered to be normal when the index is 10 or
of the respiratory neurons in the brain stem. The symptoms of below, whereas an index of 10 to 15 may be borderline and an
OSAS can be divided into nocturnal and daytime symptoms. index above 15 is abnormal. In terms of sleep architectural
Nocturnal symptoms consist of loud snoring (often with a long changes, patients with OSAS characteristically show reduction
history), cessation of breathing as witnessed by bed partners, of slow wave and REM sleep and spend their sleep mostly in
choking, abnormal motor activities, and thrashing about in stage N2 (stage 2 NREM sleep). Short sleep latency and
bed, as well as hyperactivity and nocturnal enuresis, particu- increased time spent awake after sleep onset are the other sleep
larly in children, and occasionally excessive sweating (84). architecture findings. In a high percentage of OSAS patients
Major daytime symptoms of OSAS include EDS, with patients (may be up to 25%), sleep maintenance insomnia has also been
falling asleep during the day at inappropriate times and in inap- observed. In addition, patients with OSAS have abnormal sleep
propriate places often causing accidents on the road and at microstructure as evidenced by increased arousal index as well
work. The daytime function is impaired complaining some- as increased CAP rate. The best treatment for moderate to
times about morning headache and forgetfulness, and men may severe OSAS is the application of CPAP titration causing upper
report impotence. This pattern of sleepiness of prolonged dura- airway pressurization opening up the airway passages acting as
tion and nonrefreshing nature is quite different from short- a pneumatic splint, thereby eliminating obstructive apnea, hypox-
duration refreshing narcoleptic sleep attacks. On physical emias, snoring, and sleep fragmentation. Figure 41.28 shows
examination, obesity was noted in approximately 70% of the respiratory and sleep hypnogram before and after CPAP titra-
patients and in many cases local anatomical abnormalities of tion. The optimal CPAP pressure must first be determined by
the upper airway were found. Repeated hypoxemias associated an overnight PSG study and the patient can then purchase a
with cessation of breathing and arousals during sleep may lead home unit for use during the night’s sleep. There are also auto-
to hypertension, cardiac arrhythmias, heart failure, and stroke CPAP machines available, automatically titrating the pressure
(84,85). The diagnosis is confirmed by laboratory techniques according to detected abnormal breathing events. In terms of
(see PSG). PSG findings of OSAS include recurrent episodes of patient adherence and clinical outcomes, the roles and advan-
apnea and hypopneas (see Figs. 41.12A–C and 41.27), which are tages of these devices over the standard CPAP machine remain
mostly obstructive or mixed with a few episodes of central to be determined (86,87). The role of portable unattended
apneas accompanied by oxygen desaturation followed by monitor or home sleep testing for diagnosis and treatment of

Figure 41.28 Sleep and respiratory hypnogram


before and after positive airway pressurization
(Bilevel) in a 56-year-old man with severe upper
airway obstructive sleep apnea syndrome
(OSAS). Note the initial part (left-hand side) of
the hypnogram before pressurization treatment
(None) shows repeated episodes of obstructive
apneas and hypopneas ( Respira tory Event)
accompanied by oxygen desaturation (Desatu -
ration) recorded by finger oximetry and followed
by repeated arousals (Arousal) and snoring
(Snore) during stages N1 and N2. Following
pressurization (right-hand column) treatment
(Treatment: Start), respiratory events and oxygen
desaturation are gradually eliminated. W, wake-
fulness; R: stage REM; N1, sleep stage N1; N2,
stage N2; N3, stage N3; Position: body position;
HR, heart rate; SAO2, oxygen saturation (note
artifacts).
844 Part V ■ Complementary and Special Techniques

OSAS patients is currently evolving (see above under “Ambulatory Tabl e 41 . 3


PSG Recording”) and has not been firmly established yet based on
validated scientific studies. In some mild to moderate cases of Circadian Rhythm Sleep Disorders
OSAS, dental appliances or uvulopalatopharyngoplasty (UPPP)
may be tried. Jet lag
Shift work
Central Sleep Apnea Syndrome
Delayed sleep phase type
CSAS includes primary or idiopathic type and CSAS due to
Cheyne–Stokes breathing (CSB) pattern (Fig. 41.29), CSAS due Advanced sleep phase type
to high altitude and due to central neurologic disorders, partic- Irregular sleep–wake type
ularly brain stem dysfunction, as well as central sleep apnea due
Free-running type (non–24 hour or nonentrained type)
to drug or alcohol abuse (e.g., use of opiates). Idiopathic or pri-
mary CSAS is rare and the patient may present with years of Circadian rhythm sleep disorder comorbid with medical
frequent awakenings due to repeated episodes of central apneas conditions
followed by arousals and sometimes may present with insom- Circadian rhythm sleep disorder due to drug or substance
nia–hypersomnia syndrome. CSB is noted in patients with con- abuse
gestive cardiac failure, neurologic disorders, and sometimes
with renal failure. The presence of CSB in cardiac failure
increases mortality. In primary CSAS, patients are usually
normocapnic or hypocapnic with PaCO2 of 35 mm Hg or who work nonstandard shifts) are the two most common
lower. In addition, the cycle length (apnea plus hyperpnea dura- circadian rhythm sleep disorders. Other circadian rhythm sleep
tion) is more than 45 seconds in CSB and less than 45 seconds disorders are listed in Table 41.3.
in primary CSAS (74). Repeated arousals causing sleep frag-
mentation are important factors causing EDS. CSB is a type of Jet Lag Disorder
periodic breathing characterized by cyclic changes in breathing Jet lag is experienced as a result of eastward or westward jet
with a crescendo-decrescendo sequence separated by central travel after crossing several time zones, disrupting synchroniza-
apneas or hypopneas (Fig. 41.29). tion between the body’s inner clock and external cues.
Symptoms do not occur after North/South travel. This desy-
Circadian Rhythm Sleep Disorders chronization causes sleepiness, tiredness, general malaise,
A mismatch between the body’s internal clock (in the SCN) and impairment of judgment and concentration, and sometimes
geophysical environment either as a result of malfunction of the disorientation. Sleep problems include difficulty maintaining
biologic clock or as a shift in the environment causing this to be sleep, frequent arousals, and EDS. These symptoms may also be
out-of-phase results in circadian rhythm sleep disorders. These associated with gastrointestinal problems and the body’s other
patients present with sleep difficulties as a result of desynchro- rhythms including those that control endocrine secretions, and
nization between their internal circadian rhythm and the exter- body temperature. These symptoms may last for several days
nal time. Jet lag (associated with high-speed air travel across depending on the direction of travel and the number of time
several time zones) and shift work sleep disorder (in patients zones traversed. They also depend on age; older individuals take

Figure 41.29 A 500-second excerpt showing the


classic Cheyne–Stokes breathing (CSB)
crescendo–decrescendo pattern from an
overnight polysomnographic (PSG) recording of a
69-year-old man. The presence of CSB through-
out most of the NREM sleep with marked decre-
ment or absence during REM sleep in this patient
with a history of hypertension and EDS suggests
incipient left ventricular failure. (Reproduced
from Chokroverty S. Sleep and its disorders. In:
Bradley WG, Daroff RB, Fenichel GM, et al., eds.
Neurology in Clinical Practice. 4th ed. 2004:
1993–2054, with permission.)
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 845

a longer time to readjust than younger individuals. It takes and wake time, and sleep onset occurs at irregular hours. There
longer to resynchronize the internal and external clocks when is no entrainment or synchronization with the usual time cues
traveling East than when traveling West. It takes approximately such as sunlight or social activities. This disorder is most often
1½ hours per day to readjust the internal body clock when trav- seen in blind people. In each 24-hour sleep–wake cycle, the
eling East as compared with 1 hour per day when traveling patient displays increasing delay of sleep onset by approxi-
West. It is easier to lengthen a day (for example, when traveling mately 1 hour, causing an eventual progression of sleep onset to
in a westward direction) than to shorten a day (for example, the daytime hours into the evening. This can be documented
when traveling East). Other factors that may contribute to jet clearly in an actigraphic recording (see later).
lag disorder include limited mobility when flying, dryness of
the eyes on the plane, headache, fatigue, gastrointestinal distur- Irregular Sleep–Wake Circadian Rhythm Disorder
bances, and nasal congestion (jet symptoms). Circadian rhythm sleep disorder, irregular sleep–wake type, is
characterized by lack of clearly defined circadian rhythm of
Shift Work Sleep Disorder sleep and wake. Patients are seen to be napping throughout the
In the United States, up to 2%–5% workers may suffer from 24-hour period, but total sleep time is normal. This condition
shift work sleep disorder as a result of working irregular shifts, may be seen in patients with degenerative neurologic disorders
particularly night shifts. Symptoms include sleep disruption, such as dementia, and children with mental retardation.
fatigue, and gastrointestinal symptoms, increasing chances of Actigraphic recordings are very useful in diagnosing this condi-
being involved in traffic accidents and making errors on the job. tion or any of the circadian rhythm sleep disorders (discussed
Adjustment of the work time schedule rarely improves the later).
symptoms of shift work sleep disorder.
Sleep-Related Movement Disorders
Delayed Sleep Phase State This new category of sleep-related movement disorder is
In delayed sleep phase state (DSPS), the patient’s major sleep included in the ICSD-2 (74). These movements consist of rela-
episode is delayed in relation to desired clock time causing tively simple stereotyped movements disturbing sleep. This cat-
sleep-onset insomnia or difficulty awakening at the desired egory includes RLS, PLMS, sleep-related rhythmic movement
time. A typical schedule consists of going to sleep late between disorder, bruxism, and nocturnal leg cramps.
2 AM and 6 AM and waking up late between 10 AM and 2 PM (74).
These patients have difficulty functioning adequately during Restless Legs Syndrome
daytime hours if they must wake up early in the morning to go RLS is a sensory-motor neurologic movement disorder with
to school or work. They cannot function normally in society lifelong symptoms causing considerable morbidity. RLS is the
due to the disturbed sleep schedule; their sleep architecture, most common movement disorder, but it is uncommonly
however, is generally normal if these individuals are allowed to recognized and treated despite a lucid description of the entity
follow their own uninterrupted sleep schedule. The onset of in the middle of the last century. There is not a single diagnos-
DSPS generally occurs during childhood or adolescence. tic laboratory test for RLS, therefore the diagnosis depends
Sometimes a history of DSPS in other family members exists entirely on physician’s critical evaluation of symptoms. The
and some patients may complain of depression. An unusually diagnosis is based on the IRLSSG criteria established first in
long intrinsic circadian period due to an abnormality in the 1995 (88) and modified slightly in 2003 (89). These criteria
biologic clock in the SCN is the fundamental problem. This include essential, supportive, and associated features and are
type of sleep problem can be clearly documented by keeping a listed in Table 41.4.
sleep log for one to two weeks and by recording sleep–wake RLS is a lifelong disorder that often begins at a very young
activities with an actigraph (discussed further under ‘Related age but is often diagnosed in the middle to later years. The
Laboratory Procedures’). prevalence of RLS increases with age and the overall prevalence
has been estimated at about 10% of the adult population in the
Advanced Sleep Phase State North American continent and Western Europe, but the preva-
Advanced sleep phase state (ASPS) is the converse of DSPS (74). lence of most severe cases (symptoms occurring at least twice a
These patients go to sleep early in the evening and wake up early week) is about 2.7% (90). There is an increased incidence (50%
in the morning. They experience sleep disruption and daytime to 60%) in first degree relatives of idiopathic cases of RLS.
sleepiness when not going to sleep at an early hour. The condi- Recent genome-wide association and linkage analysis docu-
tion is often seen in patients with depression and in normal eld- mented common variations in certain genomic regions con-
erly individuals. Familial ASPS has been ascribed to mutation in firming more than 50% increase in risks for RLS and PLMS
the hPer 2 gene. This circadian rhythm disorder can be easily (91,92). Most of the abnormal leg movements are noted in
documented by monitoring with an actigraph for 1 to 2 weeks. the evening while the patients are resting in bed. In severe
cases, however, movements are noted in the daytime while
Free-Running Circadian Rhythm Disorder patients are sitting or lying down, hence it is aptly termed as
A circadian rhythm sleep disorder, free-running type (nonen- quiescegenic disease. At least 80% of RLS patients have PLMS
trained type or non–24-hour sleep–wake syndrome), is charac- and may also have PLMW. The greatest advance made in
terized by a patient’s inability to maintain a regular bedtime recent years is in the treatment of RLS. Several double-blind
846 Part V ■ Complementary and Special Techniques

Tabl e 41 . 4

Clinical Diagnostic Criteria for Idiopathic Restless Legs Syndrome

Essential criteria
An urge to move the legs usually accompanied or caused by uncomfortable sensations in the legs
The urge to move or unpleasant sensations beginning or worsening during periods of rest or inactivity such as lying or sitting
The urge to move or unpleasant sensations are partially or totally relieved by movements, such as walking or stretching, at
least as long as the activity continues
The urge to move or unpleasant sensations are worse in the evening or night than during the day and only occur in the
evening or night
Supportive features
Dopaminergic responsiveness
Presence of periodic limb movements in sleep or in wakefulness
Positive family history
Associated features
Usually progressive clinical course
Normal neurologic examination in the idiopathic form
Sleep disturbance

placebo-controlled clinical trials have established the value of They noted no difference in sleep stage percentages between
dopaminergic treatment in RLS. The condition may have a pro- those with and without RLS. All the studies therefore docu-
found impact on sleep, although sleep disturbance is not listed mented significant sleep disruption in RLS patients, but there
as an essential or supportive feature of RLS. In the RLS epi- are no studies adequately addressing the long-term conse-
demiology, symptoms, and treatment (REST) Primary study quences of such sleep disruption. A significant number of RLS
(93), the most troublesome symptom rated by almost half of patients are left with chronic insomnia despite improvement or
the patients included sleep-related symptoms. The same study complete resolution of the RLS symptoms and the mechanism
noted that about 69% of those with moderately severe to severe of such chronic sleep disturbance remains undetermined. In
idiopathic RLS subjects took more than 30 minutes to fall summary, the typical PSG findings in an RLS patient are pro-
asleep and 60% woke up three or more times at night, thus longed sleep-onset latency, increased WASO, increased PLMS,
causing sleep initiation and sleep maintenance difficulties with evidence of sleep-related breathing disorders in some patients,
sleep fragmentation. It is notable that despite disturbed sleep and transient rise of blood pressure and heart rate associated
being a prominent complaint in RLS patients, the topic of sleep with PLMS and arousal.
disturbance in RLS and its potential consequences (both short
term and long term) have not been adequately addressed. Periodic Limb Movements in Sleep and Periodic
There are only limited polysomnographic (PSG) studies to Limb Movement Disorder
characterize sleep in RLS patients. Hornyak et al. (94) noted PLMS is a PSG finding (see Fig. 41.11) characterized by recur-
prolonged sleep-onset latency, shorter total sleep time, and ring stereotyped limb movements, particularly dorsiflexion of
longer REM sleep latency in moderately severe RLS patients (29 the ankles and sometimes flexion of the knees and hips (see
women and 16 men) with a mean IRLSSG score of 24 as com- Section 1). A PLMS index (number of PLMS per hour of sleep)
pared to equal number of age- and sex-matched healthy con- exceeding 5 in children and 15 in most adult cases is clinically
trols. These patients also had markedly increased PLMS index significant. PLMS appears most commonly in RLS occurring in
and sleep fragmentation. Saletu et al. (95) also found increased at least 80% of cases but may also occur in a large number of
REM latency and decreased REM density but normal sleep other medical, neurologic, and sleep disorders, and with inges-
onset latency and total sleep time in 12 RLS patients compared tion of medications (e.g., selective serotonin reuptake
with healthy controls. Winkelman et al. (96) in a recent study inhibitors, tricyclic antidepressants) and even in normal indi-
analyzed sleep data obtained from unattended, in-home PSG viduals, particularly in subjects older than 65. In the current
recordings from 2971 subjects included in the sleep heart health ICSD-2 (74), periodic limb movement disorder (PLMD) is
study (SHHS) which is a population and cross-sectional obser- characterized by PSG findings of increased PLMS without asso-
vational study, and found increased mean sleep latency and ciated RLS, causing repeated awakenings and sleep fragmenta-
arousal index in RLS patients compared to those without RLS. tion giving rise to insomnia or daytime excessive sleepiness, and
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 847

is listed as a separate entity. There is, however, some controversy Many normal individuals have nocturnal leg cramps and the
regarding the diagnosis of PLMD causing sleep fragmentation, cause remains unknown. PSG recordings during the cramps
arousals, and EDS. There is a growing body of evidence that show nonperiodic bursts of gastrocnemius EMG activity. The
PLMS may simply be a PSG phenomenon and may not have sleep-related leg cramps are not explained by another sleep dis-
any specific clinical significance except the presence in the order, medical or neurologic disorder, medications, or sub-
majority of patients with RLS (97). Many sleep specialists think stance use disorder.
PLMD is a separate entity but occurring in a very small number
of patients. Further investigations including outcome studies Sleep-Related Bruxism
are needed to document that PLMS or PLMD may cause sleep (Nocturnal Tooth Grinding)
disturbance or EDS. Sleep-related bruxism or nocturnal tooth grinding often pres-
ents between the ages of 10 and 20 years, but it may persist
Sleep-Related Rhythmic Movement Disorder throughout life often leading to secondary problems such as
Sleep-related rhythmic movement disorder is characterized by temporomandibular joint dysfunction. It is also commonly
repetitive rhythmic stereotyped behavior occurring predomi- noted in children with mental retardation or cerebral palsy.
nantly during transition from relaxed wakefulness to sleep Nocturnal bruxism is noted most prominently during stage N1
involving large muscle groups (74). The condition is mostly or N2 and REM sleep. The episode is characterized by stereo-
seen in infants and children. The three most common types of typical tooth grinding and is precipitated by anxiety, stress, and
movements seen in this condition consist of head banging, head dental disease. Occasionally, familial cases have been described.
rolling, and body rocking. Other types of sleep-related move- The scoring rules in the PSG for sleep bruxism include the fol-
ments in this disorder include body rolling, leg rolling, and leg lowing (12): They must consist of brief phasic or tonic eleva-
banging. The behavior results in significant complaint as man- tion of EMG activity of twice the amplitude of background
ifested by one of the following: interfering with normal sleep, EMG; the chin EMG elevations are scored as bruxism if they are
significant impairment in daytime function, or self-inflicted 0.25 to 2 seconds in duration and must occur in a regular
bodily injury. PSG studies show characteristic rhythmic move- sequence of three consecutive elevations; sustained elevations
ments occurring in the transition stage or stage N1 or N2 sleep are scored as bruxism if the elevation is more than 2 seconds
(Fig. 41.30). Rarely, these movements are noted in stage N3 or (Fig. 41.31). There must be a period of at least 3 seconds of sta-
REM sleep. The recent AASM scoring manual (12) established ble background chin EMG before a new episode of bruxism can
the rules defining PSG characteristics of rhythmic movement be scored. Sleep-related bruxism can also be scored reliably by
disorders as follows: the frequency varies from 0.5 to 2.0 Hz audio in combination with EMG with minimum of two audi-
with four consecutive movements forming a cluster of move- ble tooth grinding episodes per night of PSG in the absence of
ments showing an EMG activity that is twice the baseline back- epilepsy.
ground activity.
Sleep-Related Eating Disorders
Nocturnal Leg Cramps Sleep-related eating disorders classified under “Other
These are intensely painful contractions accompanied by mus- Parasomnias” in ICSD-2 (74) are commonly seen in women
cle tightness occurring during sleep. These spasms usually last between the ages of 20 and 30. This disorder consists of recur-
for a few seconds but sometimes persist for several minutes. rent episodes of involuntary eating and drinking during
Cramps during sleep are generally associated with awakening. partial arousals from sleep (74,98). Sometimes the patient

Figure 41.30 A 120-second excerpt from a 5½ -


year-old boy showing four episodes of head
banging (a manifestation of rhythmic movement
disorder) at a rate of about 1.5 Hz during transi-
tioning from wakefulness to sleep stage N2. Note
rhythmic movement artifacts in all channels of
PSG. Top 8 channels: EEGs (international 10 to 20
electrode nomenclature); E1–M1 and E2–M1, left
and right EOG; Chin 1–Chin 2, chin EMG; EKG:
electrocardiogram; LTIB and RTIB, EMGs of left
and right tibialis anterior; LGAST and RGAST,
gastrocnemius muscles; OroNs 1–OroNs 2 and
Pflw1–Pflw2, respiratory air flows; Chest and
ABD, effort.
848 Part V ■ Complementary and Special Techniques

Figure 41.31 A 20-year-old woman referred for


sudden nocturnal awakenings with vocalization
and sleepwalking. PSG shows an arousal from
NREM stage 4 sleep with subsequent rhythmic
masticatory muscle activation and teeth gnashing
(EMG artifacts on EEG channels) typical of sleep
bruxism. (Reproduced from Chokroverty S,
Thomas R, Bhatt M, eds. Atlas of Sleep Medicine.
Philadelphia: Elsevier; 2005, with permission.)

displays strange eating behavior (e.g., consumption of inedi- it may also occur during NREM sleep (100–102). PSG findings
ble or toxic substances such as frozen pizza, raw bacon, and cat resemble central apnea with protracted expiratory bradypnea
food). The episodes cause sleep disruption and weight gain, without oxygen desaturation (Fig. 41.33). Simultaneous audio
and occasionally injury has been reported. The condition can recordings will bring out the characteristic groaning sounds.
be idiopathic or can be comorbid with other sleep disorders The clinical relevance and pathophysiology of this condition
(e.g., sleep walking, RLS, PLMS, OSAS, narcolepsy, and irreg- remain unknown.
ular sleep–wake circadian rhythm sleep disorder) and with the
use of medications such as zolpidem, triazolam, and other Neuromuscular Disorders
psychotropic agents (98,99). The most common PSG findings In neuromuscular disorders, SDB is commonly associated with
are multiple, confusional arousals with or without eating, insidiously developing chronic respiratory failure, especially in
arising predominantly from stage N3 sleep, but also from the advanced stages, but is often unrecognized and untreated
other stages of NREM sleep and occasionally from REM sleep (103–106).- The most common SDB in neuromuscular disor-
(see Fig. 41.32) (74). ders is sleep related, especially REM-related hypoventilation.
Both central and upper airway obstructive apneas also occur.
Catathrenia (Expiratory Groaning) Nocturnal hypoventilation giving rise to hypoxemia and
This parasomnia is characterized by recurrent episodes of expi- hypercapnea during sleep in the initial stage of neuromuscular
ratory groaning (high-pitched, loud humming or roaring disorders causes chronic respiratory failure. In later stages of
sounds) occurring in clusters predominantly during REM, but the illness, abnormal blood gases may persist even during the

Figure 41.32 A 240-second excerpt of PSG showing awakening (right half of the recording)
out of sleep-stage REM (left half of the tracing) and an episode of nocturnal eating in a
51-year-old man. He woke up with a snore from an episode of REM-related hypopnea. Top four
channels: EEGs; left and right electro-oculographic (E1–M1; E2–M1); chin muscle
(CHIN1–CHIN2) show chewing artifacts. EKG: electrocardiogram; HR: heart rate; LtTib and
RtTib, left and right tibialis anterior EMGs; PFlow1–PFlow2, airflow by thermistor (oronasal)
and by nasal pressure recording; CHEST; ABD: respiratory effort; SaO2, oxygen saturation by
finger oximetry.
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 849

Figure 41.33 Expiratory groans. A polysomno-


graphic segment of 120-second epoch showing
EEGs (C3–A2, O1–A2, C4–A1, O2–A1), left and
right eye movements (LOC–A2; ROC–A1), chin,
left tibialis (LT. TIBI), and right tibialis (RT. TIBI)
EMGs, snoring (SNORE), EKG, oronasal airflow
(ORONAS), thoracic (THORAX) and abdominal
(ABDM) effort channels, oxygen saturation
(SaO2). Note prolonged expiration in the flow
and effort channels followed by arousals without
oxygen desaturation in stage 2 NREM sleep.
(Reproduced from Siddiqui F, Walters AS,
Chokroverty S. Catathrenia: a rare parasomnia
which may mimic central sleep apnea on
polysomnogram. Sleep Med. 2008;9(4):460–461,
with permission.)

daytime. As a result of alveolar hypoventilation and ventila- manifesting these symptoms deserve to be investigated to
tion–perfusion mismatching, hypoxemia and hypercapnea uncover nocturnal hypoventilation to prevent serious conse-
occur, giving rise to chronic respiratory failure even during the quences of chronic respiratory failure such as pulmonary
daytime at a later stage of neuromuscular disorders. Nocturnal hypertension and congestive cardiac failure. Neuromuscular
hypoventilation and chronic respiratory failure, however, in disorders causing chronic respiratory failure include primary
neuromuscular disorders may present insidiously and may ini- muscle disease, neuromuscular junction disorders and motor
tially remain asymptomatic (103–106). For the diagnosis, a neuron diseases or motor neuronopathies, acute and chronic
high index of clinical suspicion is needed. Historical clues inflammatory demyelinating polyneuropathies, hereditary
include the presence of EDS, daytime fatigue, morning sensory-motor neuropathy, and phrenic neuropathy (107).
headache, restless and disturbed nocturnal sleep, and unex- The characteristic PSG findings in neuromuscular disorders
plained leg edema. Patients with neuromuscular disorders consist of central, mixed, and upper airway obstructive apneas

Figure 41.34 Ten- and 30-second excerpts from


a nocturnal PSG showing alpha–delta sleep in a
30-year-old man with history of snoring for many
years. He denied any history of joint or muscle
aches and pains. The alpha frequency is inter-
mixed with and superimposed on underlying
delta activity. Alpha–delta sleep denotes a non-
specific sleep architectural change noted in many
patients with complaints of muscle aches and
fibromyalgia. It is also seen in other conditions
and many normal individuals. Top 10 channels:
EEGs; LEFT EOG and RT EOG, left and right
electro-oculograms; chin EMG, EMG of chin;
Lt/ Rt Tib EMG, left/ right tibialis anterior EMG;
oronasal thermistor; chest and abdomen effort
channels; snore monitor; EKG, electrocardiogra-
phy. (Reproduced from Chokroverty S, Thomas
R, Bhatt M, eds. Atla s of Sleep Medicin e.
Philadelphia: Elsevier; 2005, with permission.)
850 Part V ■ Complementary and Special Techniques

and hypopneas associated with oxygen desaturation, sleep Tabl e 41 . 5


hypoventilation showing nonapneic oxygen desaturation that
is worse during REM sleep (108). The painful neuromuscular Molecular Neurobiologic Classification of
disorders may also show sleep-onset insomnia and reduced Neurodegenerative Diseases
sleep efficiency.
Tauopathies
Neurodegenerative Disorders
Alzheimer disease (AD)
Neurodegenerative diseases of the central nervous system
include a group of heterogeneous disorders with insidious Progressive supranuclear palsy (PSP)
onset and relentlessly progressive course without an identifi- Corticobasal degeneration (CBD)
able cause except for genetic predisposition in some cases Frontotemporal dementia (FTD)
(109). The other characteristics include bilateral symmetry of
the clinical manifestations and increasing incidents with Synucleinopathies
advancing age and pathologic involvement of system of neu- Parkinson disease (PD) with or without dementia
rons that are anatomically and physiologically related with Diffuse Lewy body disease (DLBD) with dementia
common pathologic features of simple atrophy or apoptosis
without intense cellular reactions but accompanied by mild Multiple system atrophy (MSA)
gliosis. The concept of neurodegenerative disease has changed Polyglutamine triplet repeat disorders
rapidly with the modern scientific understanding of the Huntington disease (HD)
molecular neurobiologic mechanisms responsible for nerve
cell degeneration. In contemporary neuroscience, neurodegen- Spinocerebellar ataxia (SCA)
erative diseases are believed to be proteinopathies due to exces- Hypoprogranulinopathies
sive protein misfoldings and intracellular protein aggregation. FTD with parkinsonism linked to progranulin gene on
They are mainly classified into tauopathies and synucle- chromosome 17 (FTDR-17P)
inopathies. The main tauopathies (Table 41.5) include
Alzheimer disease (AD), progressive supranuclear palsy (PSP), Miscellaneous diseases
corticobasal degeneration (CBD), and frontotemporal demen- Torsion dystonia
tia (FTD). Synucleinopathies are a group of disorders with Chorea-acanthocytosis
abnormal deposition of alpha-synuclein in the cytoplasm of
neurons or glial cells as well as extracellular deposits of amy- Amyotrophic lateral sclerosis (ALS)
loid, and these include PD, diffuse Lewy body disease (DLBD)
Reproduced from Chokroverty S. Sleep and neurodegenerative diseases. Semin
with dementia, and multiple system atrophy (MSA), which is
Neurol. 2009;29:446–468.
inclusive of Shy–Drager syndrome, striatonigral degeneration,
and sporadic olivopontocerebellar atrophy (see Table 41.5).
The major sleep complaints in neurodegenerative diseases
include insomnia, hypersomnia, parasomnia, excessive noctur- Polysomnographic Findings in
nal motor activity, “sundowning,” circadian sleep–wake distur- Neurodegenerative Disorders
bances, and respiratory dysrhythmias. PSG is the single most important laboratory test for the diagno-
Sleep fragmentation characterized by increased awakenings, sis of sleep disorders particularly hypersomnia. Simultaneous
stage transitions, and arousals is an important sleep dysfunc- video monitoring (Video PSG) additionally helps in the diag-
tion in neurodegenerative diseases. Sleep fragmentation results nosis of parasomnia (109).
in reduced slow wave and REM sleep. Hypersomnia includes PSG findings in AD show reduced total sleep time, increased
EDS and irresistible sleep attacks. The parasomnia most com- nocturnal awakenings, decreased REM and slow-wave sleep,
monly associated with neurodegenerative diseases is RBD, loss of phasic components (spindles and K-complexes) of
which is characterized by REM sleep associated with intense NREM sleep, and sleep–wake rhythm disturbances. Other
motor activity related to dream-enacting behavior and absence abnormalities in PSG of AD patients are sleep apnea and PLMS.
of muscle atonia of REM sleep. It has been increasingly obvious In PD, PSG findings include reduction of total sleep time,
recently that in the setting of degenerative dementia or frequent awakenings, decreased slow-wave sleep, decreased sleep
Parkinsonism, RBD is not only a manifestation of an evolving spindles, and K-complexes in stage N2. REM sleep may intrude
synucleinopathies but also seen in tauopathies occasionally. into NREM sleep and normal REM/NREM cycling may be
RBD carries the potential for injury to self and others and abolished. Repetitive blinking may be noted at sleep onset and
therefore its early recognition is very important. Circadian blepharospasm is seen during slow-wave sleep before REM
rhythm sleep–wake disturbances are noted in some conditions episodes. Recently, there has been PSG documentation of
most prominent in AD. It may also present as a cyclic agitation abrupt daytime sleep attacks in PD patients characterized by
syndrome popularly known as the “sundowning syndrome.” A abrupt occurrence of stage N2 sleep. Absence of muscle atonia
commonly encountered excessive nocturnal motor activity that in REM sleep may be noted in patients with RBD (see
may cause sleep disturbance is PLMS. Fig. 41.26) or as a preclinical sign of RBD.
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 851

In DLBD, PSG findings include decreased total sleep time, patients with nonrestorative sleep such as alpha–delta sleep
frequent awakenings, and absence of muscle atonia in REM (Uncommon and atypical PSG patterns). Fatal familial insomnia
sleep in those with RBD, which may be a preclinical sign of (FFI) is a rare prion disease originally described by Lugaresi and
DLBD. Because of sleep fragmentation and repeated awaken- associates (116). Clinical features of FFI include impaired control
ings, the patient may have EDS as reflected in abnormal MSLT. of sleep–wake cycle, including circadian rhythms, autonomic and
PSG findings in PSP patients have shown consistent abnor- endocrine dysfunction, as well as somatic, neurologic, cognitive,
malities in sleep architecture with the decreased total sleep and behavioral manifestations (117). Profound sleep distur-
time, decreased sleep efficiency, increased wake time after sleep bances and in particular severe insomnia are noted from the very
onset, increased percentage of N1 sleep, decreased percentage of beginning of the illness. In the PSG study, there is almost total
REM sleep, and decreased REM duration. Another characteris- absence of deep sleep patterns and only short episodes of REM
tic is the reduction in abundance and amplitude of sleep spin- sleep lasting for a few seconds or minutes without muscle atonia
dles. RBD has also been described in PSP patients (110,111). associated with dream-enacting behavior in the form of complex
PSG findings in Huntington disease show decreased sleep effi- gestures and myoclonus. There is progressive reduction of the
ciency, increase in stage N1, frequent arousals, and reduced total sleep time and reduced sleep cycle with the progression of
slow-wave sleep. The involuntary movements decrease progres- the disease. Sleep spindles and K-complexes are totally absent in
sively from wake to stages N1 and N2 to REM sleep. The dysk- the late stage of illness and slow-wave sleep is never recorded. The
inetic movements in sleep are also of shorter duration, lower neurophysiology hallmark of FFI is severe atrophy of the thala-
amplitude, and more fragmented as compared to wakefulness. mus, particularly anteroventral and dorsomedial thalamic nuclei.
An increased density of sleep spindles in Huntington disease FFI has thus rekindled the role of the thalamus in the sleep–wake
patients has also been reported. regulatory mechanisms by demonstrating that the thalamus is
PSG findings in torsion dystonia show frequent awakenings, essential for generation of slow-wave sleep and for suggesting a
reduced sleep efficiency, and increased sleep latency. High- role for prions in sleep regulation.
amplitude sleep spindles have been described in some cases but
have been an inconsistent finding in other studies. Segawa and Uncommon and Atypical PSG Patterns
colleagues (112) recorded movement counts in PSG studies in In certain normal individuals, particularly following sleep dep-
patients with hereditary progressive dystonia. They reported a rivation, withdrawal of medications, and alteration of sleep
decrease in gross body movements in stage N1, an increase in habits, certain atypical and uncommon sleep patterns may be
stage N2, and a decrease in REM sleep. Localized twitch move- observed in the PSG (118). Similar patterns, however, have also
ments were depressed in all-sleep stages but followed the nor- been described in a variety of sleep disorders. Some of these pat-
mal relative distribution between stages. terns include alpha–delta or alpha–NREM sleep, REM–spindle
In olivopontocerebellar degeneration (OPCD), PSG study has sleep, REM intrusion into NREM sleep, REM sleep without
been reported to show increased awakenings, reduced slow-wave atonia in conditions other than RBD, abnormal REM–NREM
sleep, and reduced or absent REM sleep (113,114). In several cycling, rhythmic leg movements, bruxism with arousals after
cases of OPCD, REM sleep without muscle atonia accompanied termination of apnea, alternating leg muscle activation (ALMA),
by features of RBD has been described (115). Central, obstruc- propriospinal myoclonus at sleep onset, and ataxic (biot’s)
tive, and mixed apneas have been reported by several authors. breathing in patients on opiates for pain management.
In MSA, PSG findings include reduction of total sleep time,
decreased sleep efficiency, decreased sleep efficiency, increased Alpha–Delta Sleep
sleep latency, increased night-time awakenings, decreased slow- Hauri and Hawkins (119) originally described the so-called
wave sleep and REM sleep, absence of muscle atonia in patients alpha–delta sleep pattern in patients with depression.
with RBD, and a variety of respiratory dysrhythmias (109). However, since then it has been found in many subjects with
nonrestorative sleep as a result of muscle and joint aches and
Insomnia Including Fatal Familial Insomnia pains, and rheumatic disorders including fibromyalgia. A sim-
Insomnia is the most common sleep disorder affecting the pop- ilar pattern has also been noted in many normal individuals.
ulation and the most common disease encountered in the prac- Therefore, it is not specific for fibromyalgia but often noted in
tice of sleep medicine. Patients present with difficulty initiating large number of such patients. In this PSG pattern, bursts of
and maintaining sleep and nonrestorative sleep occurring three alpha activities are seen during slow-wave sleep or during stage
to four times per week persisting for more than a month and N2 sleep overriding the slow waves or spindles causing alpha
associated with impairment of daytime function (74). Acute intrusions into different NREM sleep stages (see Fig. 41.34).
insomnia is generally associated with an identifiable stressful
situation. Most cases of insomnia are chronic and comorbid REM–Spindle Sleep
with other conditions including psychiatric, medical, and neu- On many occasions bursts of sleep spindles are noted during
rologic disorders and drug and alcohol abuse. If no cause is REM sleep, giving rise to a pattern called REM–spindle sleep
found, the condition is labeled idiopathic, primary, or psy- pattern (Fig. 41.35). This is noted in many normal individuals,
chophysiological insomnia. There is no indication for PSG in particularly in the beginning of the first REM period. Similar
uncomplicated insomnia unless insomnia is associated with REM–spindle sleep pattern is noted also in many patients with
sleep apnea. Some nonspecific findings have been described in hypersomnia and narcolepsy–cataplexy syndrome.
852 Part V ■ Complementary and Special Techniques

Figure 41.35 A 67-year-old man with difficulty


sleeping, loud snoring, and EDS for 5 years.
Medical history is significant for high blood pres-
sure and coronary artery disease. Nocturnal PSG
showed the presence of moderate sleep apnea
with an AHI of 29.5. A 30-second excerpt from
nocturnal PSG showing the presence of frequent
sleep spindles throughout the epoch of REM
sleep. Prominent sawtooth waves of REM sleep
in C3- and C4-derived EEG channels, prominent
phasic eye movements of REM sleep on electro-
oculogram channels, and decreased chin muscle
tone characteristic of REM atonia are seen.
(Reproduced from Chokroverty S, Thomas R, Bhatt
M, eds. Atlas of Sleep Medicine. Philadelphia:
Elsevier; 2005, with permission.)

REM Sleep Without Muscle Atonia the head, trunk, or limbs during wake–sleep transition (74). As
This pattern is of course characteristically seen in patients with an unusual manifestation, this also has been noted in patients
RBD (see Fig. 41.26). However, sometimes similar REM sleep with upper airway OSAS on arousals after termination of
without atonia may be noted in patients who have been treated apneic–hypopneic events (120–122) (Fig. 41.36).
with REM suppressant medications such as tricyclic antidepres-
Rhythmic Leg Movements
sant, monoamine oxidase (MAO) inhibitors, selective serotonin
reuptake inhibitors, and phenothiazines, as well as in patients Rhythmic leg movements in wakefulness (Fig. 41.37) and
with PD or narcolepsy in absence of the typical behavioral NREM (Fig. 41.38) and REM (Fig. 41.37) sleep are sometimes
manifestation of RBD. Whether this pattern indicates a subclin- noted as unusual PSG findings in normal individuals as well as
ical or preclinical RBD remains undetermined. in patients with sleep pathologies (121). The significance of
such findings remains undetermined.
A Mixture of REM Bursts in NREM Sleep
Occasionally, REM-like bursts are noted during NREM sleep in Sleep Bruxism or Tooth Grinding
normal persons. Such pattern, however, is seen particularly in As an unusual manifestation, bruxism may rarely be noted on
narcoleptics and patients with depression who have been arousal after termination of apneic events in patients with
treated with REM suppressant medications. This pattern may OSAS (121) (Fig. 41.39).
also be seen in PD.
Alternating Leg Muscle Activation
Abnormal REM–NREM Cycling ALMA is a recently described unusual polysomnographic
REM–NREM goes through a normal cycling of four to five observation characterized by ALMA preceding or following
cycles throughout the night. The initial cycle is of brief duration an arousal or awakening (122,123) (Fig. 41.40). The usual
and the longest period of REM cycle is noted toward the end of frequency is 1 to 2 Hz (range of 0.5 to 3Hz), with a duration
the night when it may last up to 1 hour. Such cycling may be of 0.1 to 0.5 second for individual activations, and showing
disrupted in patients with narcolepsy–cataplexy and those with at least four discrete and alternating muscle activations (12).
narcolepsy who have been on REM suppressant medications. The specific significance of ALMA remains undetermined at
Such abnormal cycling may also be seen in patients with head present, but it may be a variant of hypnagogic foot tremor.
injury and PD. In addition, some normal individuals may dis-
play an abnormal REM–NREM cycling. Propriospinal Myoclonus in Predormitum
Propriospinal myoclonus has recently been described (124,125)
Rhythmic Limb and Body Movements on Arousal as axial jerks occurring during relaxed wakefulness immediately
After Termination of Apneic–Hypopneic Events before falling asleep (Fig. 41.41), the period termed predormitum
Rhythmic movement disorder is an abnormal sleep-related by McDonald Critchley (126). The etiology remains undeter-
movement disorder characterized by rhythmic oscillations of mined but may prevent from falling asleep, causing sleep-onset
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 853

Figure 41.36 A 90-second excerpt from a portion of an overnight polysomnography during


stage 2 (N2) NREM sleep in a 51-year-old man with upper airway OSA, RLS, and rhythmic
movement disorder showing two episodes of rhythmic EMG bursts (about 0.75 Hz) beginning
in the right biceps (RtBiceps) and right triceps (RtTriceps) and spreading to other muscles.
These bursts were noted following termination of the apneic events and accompanied by
arousals and resumption of normal breathing as well as rhythmic body and head-rolling move-
ments. Top six channels (international nomenclature): EEGs; E1–M1, left electro-oculogram;
E2–M1, right electro-oculogram; EKG, electrocardiogram; OralNasal, oronasal thermistor (air
flow); PFlow, nasal pressure recording for airflow; Chest, thoracic breathing effort; Abd, abdom-
inal breathing effort; SaO2, oxygen saturation by finger oximetry; Snore, snoring recording;
Masseter, right masseter EMG; Chin: submental EMG; RtSterno: right sternocleidomastoideus
EMG; LtBiceps, left biceps EMG; LtTriceps, left triceps EMG; RtBiceps, right biceps EMG;
RtTriceps, right triceps EMG; RtQuad, right quadriceps (biceps femoris) EMG; LtQuad, left
quadriceps EMG; RtTib, right tibialis EMG; RtGast, right gastrocnemius EMG; LtTib, left tibialis
EMG; LtGast, left gastrocnemius EMG. (Reproduced from Gharagozlou P, Seyffert M, Santos R,
et al. Rhythmic movement disorder associated with respiratory arousals and improved by CPAP
titration in a patient with restless legs syndrome and sleep apnea. Sleep Med.
2009;10(4):501–503, with permission.)

Figure 41.37 A 50-year-old man with history of


loud snoring, choking in sleep, and intermittent
leg jerking at night. Nocturnal PSG shows the
presence of mild sleep apnea with an AHI of
12.3. This 30-second excerpt from nocturnal
polysomnography shows bursts of rhythmic foot
and leg movements on the left/ right tibialis ante-
rior muscle recording channel during wakeful-
ness. The movement is not associated with
respiratory events, oxygen desaturation, or
arousal from sleep. EEG, Top 10 channels: EEGs;
LEFT and RT EOG, left and right electro-oculo-
grams; CHIN, EMG of chin; Lt/ Rt Tib EMG,
left/ right tibialis anterior EMG; oronasal thermis-
tor; chest and abdomen effort channels; snore
monitor; EKG, electrocardiography; SaO2, oxygen
saturation by finger oxymetry.
854 Part V ■ Complementary and Special Techniques

Figure 41.38 A 50-year-old man with history of


loud snoring, choking in sleep, and intermittent
leg jerking at night. Nocturnal PSG shows the
presence of mild sleep apnea with an AHI of
12.3. This 30-second excerpt from nocturnal
polysomnography shows bursts of rhythmic foot
and leg movements on the left/ right tibialis ante-
rior muscle recording channel during REM sleep.
The movement is not associated with respiratory
events, oxygen desaturation, or arousal from
sleep. Top 10 channels: EEGs; LEFT and RT EOG,
left and right electro-oculograms; CHIN, EMG of
chin; Lt/ Rt Tib EMG, left/ right tibialis anterior
EMG; oronasal thermistor; chest and abdomen
effort channels; snore monitor; EKG, electrocar-
diography; SaO2, oxygen saturation by finger
oxymetry.

insomnia. The generator is thought to be in the mid-thoracic an unstable or destabilized central controller for breathing
spinal cord with propagation of muscle bursts rostrally and cau- (medullary respiratory neurons). This is well known to occur in
dally along slowly conducting propriospinal pathways. medullary lesions, but this has recently been observed in
patients taking large doses of opiates for pain management
Ataxic or Biot’s Breathing in Patients on Opiates (127,128) (Fig. 41.42). Biot’s breathing is a type of ataxic
Ataxic breathing is a type of periodic breathing with central breathing with two to three normal breaths interspersed with
apnea without crescendo–decrescendo pattern resulting from periodically recurring central apnea.

Figure 41.39 A 120-second excerpt from PSG


recording reveals sleep bruxism in a 70-year-old
woman with a history of insomnia, early morn-
ing awakenings, and EDS. Overnight PSG
revealed the presence of mild to moderate OSA
with an AHI of 21.5. Episodes of bruxism are
recorded repeatedly as part of the arousal
response following respiratory events accompa-
nied by tooth grinding on simultaneous
audio–video recording. Respiratory-related limb
movements are recorded on tibialis anterior
channel in association with the arousal response.
PSG sleep bruxism is characterized by a series of
teeth grinding associated with rhythmic EMG
artifacts with a frequency of approximately 0.5 to
1.0 hertz in stage 1 NREM sleep. Top 10 chan-
nels: EEGs; LEFT and RT EOG, left and right elec-
tro-oculograms; CHIN, EMG of chin; Lt/ Rt Tib
EMG, left/ right tibialis anterior EMG; oronasal
thermistor; chest and abdomen effort channels;
snore monitor EKG, electrocardiography.
(Reproduced from Chokroverty S, Thomas R,
Bhatt M, eds. Atla s of Sleep Medicin e.
Philadelphia: Elsevier; 2005, with permission.)
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 855

Figure 41.40 A 20-second except of PSG show-


ing alternating leg muscle activation (ALMA) in
the left and right tibialis anterior muscles (LTIB
and RTIB) at a state of approximately 1.5 Hz in
an adult man during stage N2 sleep. Top 8 chan-
nels: EEGs; E1–M1 and E2–M1, left and right
electro-oculograms; Chin 2–Chin 3, chin muscle
EMG; EKG, electrocardiogram; HR, heat rate;
OroNs1–OroNs2 and Pflw1-Pflw2, respiratory
airflow; Chest and ABD, respiratory Effort; SaO2,
oxygen saturation by finger oximetry; EtCO2,
expiratory carbon dioxide.

RELATED LABORATORY PROCEDURES well as other history and physical examination. The first step
FOR DIAGNOSIS OF SLEEP DISORDERS before ordering the laboratory tests is an assessment of EDS.
There are certain clinical clues that may suggest EDS (131):
PSG and video-PSG recordings remain the most important lab- falling asleep in inappropriate places and inappropriate circum-
oratory tests for diagnosis of sleep disorders. Three other tech- stances, for example, sleepiness while sitting and relaxing on a
nical procedures, however, have found an important place in couch, watching television, reading, watching movies, sitting in
the assessment of sleep disorders: MSLT, MWT, and actigraphy. classes or conferences, listening to a lecture and in very severe
Additionally, there are two other procedures (129,130) that also cases even talking on the telephone and giving the history to the
have some place in assessment of sleep disorders and these physician; dosing off while driving; poor attention span and
include peripheral arterial tonometry (PAT) and pulse transit difficulty coping with work and school work; driving accidents
time (PTT), but these are not described in this chapter. or accidents at work; frequent naps during the day and nonre-
freshing sleep; and feeling sleepy and tired upon waking up first
Multiple Sleep Latency Test thing in the morning. For assessment of persistent sleepiness,
The most common indication for MSLT is EDS. The initial step Epworth Sleepiness Scale (ESS) (132) is often used to assess a
in assessment of a patient with EDS is a detailed sleep history general level of sleepiness. This is a subjective propensity to
covering not only sleep at night but also daytime function as sleepiness assessed by the patient under eight situations on a

Figure 41.41 Propriospinal myoclonus at the


wake–sleep transition. A 40-year-old man pre-
sented with a 4-year history of axial jerks during
relaxed wakefulness impending falling asleep.
PSG recording shows repetitive myoclonic axial
jerks. The EMG activity originates in the left rec-
tus abdominis muscle, thereafter propagating to
rostal (sternocleidomastoid, masseter, mylohy-
oid) and caudal (biceps femoris) muscles. The
left panel shows jerks at low speed; the right
panel shows one of the jerks at a high speed.
R, right; L, left; Mylohyoid, mylohyoideus;
S.C.M., sternocleidomastoideus; I.T.C., inter-
costalis; Rectus abd, abdominis; Rectus fem., rec-
tus femoris; Biceps fem, biceps femoris.
(Reproduced from Chokroverty S, Thomas R,
Bhatt M, eds. Atla s of Sleep Medicin e.
Philadelphia: Elsevier; 2005, with permission.)
856 Part V ■ Complementary and Special Techniques

Figure 41.42 A 240-second excerpt of PSG dur-


ing sleep stage N2 showing periodic breathing
(ataxic or Biot’s breathing with two to three nor-
mal breaths interspersed with periodically recur-
ring central apneas) in a 68-year-old woman on
large doses of OxyContin (opiate) for pain man-
agement. Her abnormal breathing improved
somewhat on bilevel positive upper airway pres-
surization but could not eliminate the events
most of the time. Top four channels, EEGs;
E1–M1 and E2–M1, left and right EOGs;
CHIN1–CHIN2, chin EMG; EKG, electrocardio-
gram; HR, heart rate; LtTib and RtTib, left and
right tibialis anterior EMG (); CFLOW, air flow
during positive pressure titration; CHEST and
ABD, respiratory effort; CPRES; positive pressure
in centimeter; SaO2, oxygen saturation.

scale of 0 to 3 with three indicating the chances of dosing off are out. The test must be conducted in a quiet, dark room. The
at the highest. The maximum score is 24 and a score of 10 or recordings may include three to six channels of EEG, submen-
above suggests the presence of EDS. The test has been weakly tal EMG, and EOG recordings.
correlated with the MSLT score. The ESS and MSLT, however, The measurements include average sleep onset latency and
test different types of sleepiness. MSLT tests the propensity to the presence of SOREMs, defined as the occurrence of REMs
sleepiness objectively and ESS tests the general feeling of sleepi- within the first 15 minutes of sleep onset. Sleep onset is defined
ness or subjective propensity to sleepiness. The Stanford as the time from lights out to the first epoch of any stage of
Sleepiness Scale (SSS) (133) is a 7-point scale to measure sub- sleep including stage N1 sleep. The first epoch of 15 seconds of
jective sleepiness at the time of testing, but it does not measure sleep or greater in a 30-second epoch is considered sleep onset.
persistent sleepiness. There is a somewhat similar scale called If no sleep occurs, the test is concluded 20 minutes after lights
Karolinska Sleepiness Scale (KSS) (134), which is used mostly out. The test is terminated 15 minutes after the first 30-second
in Europe using 9-point scale but again does not measure per- epoch in any stage of sleep. If the finding is indefinite, it is bet-
sistent sleepiness. Visual Analog Scale is the other scale used to ter to continue the test than to end it prematurely. Mean sleep
assess alertness and well-being in which subjects indicate their latency is calculated as the average of the latencies to sleep onset
feelings of alertness at an arbitrary point on a line of 0 to 100 for each of the four to five naps. A sleep latency of less than 8
mm scale with 100 being the maximum sleepiness and 0 being minutes is consistent with pathologic sleepiness. A mean sleep
the most alert. latency of 8 to 10 minutes is consistent with mild sleepiness and
a mean sleep latency of 10 to 15 minutes is considered normal.
Technique of MSLT Repeat MSLT is indicated if the patient is strongly suspected to
The MSLT has been standardized and includes several general have narcolepsy but did not show the characteristic findings of
and specific procedures (135,136). The general procedures pathologic sleepiness, with sleep-onset latency of less than 8
before the actual recording include keeping a sleep diary for 1 minutes and the presence of two or more SOREMs during four
to 2 weeks before the test, which records the information about to five tests as may be seen in certain percentage of narcolepsy
bedtime, time of rising, napping, and any drug use. The patient patients after the first MSLT. MSLT may not be diagnostic in the
should discontinue central nervous system active drugs before initial test and the yield increases after the second test. The
the test. The test is preceded by an overnight PSG study and is other situation for repeating MSLT is when the finding is
scheduled about 2 to 3 hours after the conclusion of the PSG ambiguous and the sleep onset or REM sleep cannot be ade-
study. The actual test consists of four to five nap opportunities quately interpreted. Finally, if the MSLT guidelines have not
at 2-hour intervals and each recording session lasts for 20 min- been followed, the test results may be technically invalid.
utes. Between sessions, test subjects must remain awake. The
subject must not smoke for 30 minutes before lights are turned Indications for MSLT
out. Physiologic calibrations (e.g., grit teeth, blink your eyes, The AASM developed indications and practice parameters for
look up, look down, look to the right, look to the left, open your clinical use of the MSLT (136). The single most important indi-
eyes, and close your eyes) are then performed and the patients cation for performing MSLT is for the evaluation of patients
are instructed to relax and fall asleep, and the lights are turned with suspected narcolepsy. The MSLT may also be useful in the
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 857

evaluation of patients with suspected idiopathic hypersomnia and MWT do have separate functions. The MSLT unmasks
in which condition the patient develops pathologic sleepiness physiologic sleepiness, which depends on both the circadian
or sleep onset of less than 8 minutes with one or no SOREM and homeostatic factors. In contrast, the MWT is a reflection of
during 4 to 5 nap tests. The MSLT is not routinely indicated in the individual’s capability to resist sleep and is influenced by
the initial evaluation and diagnosis of OSAS nor for the moni- physiologic sleepiness. The validity, reliability, and specificity of
toring of the treatment effects after nasal CPAP therapy. The MWT need to be studied using large samples and different pop-
MSLT is also not routinely indicated for patients complaining ulations of sleep disorders. No universally accepted normative
of EDS due to medical or neurologic disorders except nar- data for the MWT are available. Using standard deviation crite-
colepsy, insomnia, or circadian rhythm sleep disorders. ria, lower limit of normative data for sleep onset latency (mean
2 SD) has been defined by Mitler et al. (140) as 19.4 minutes.
Reliability, Validity, and Limitations of the MSLT In contrast, the AASM practice parameters (136) advocate the
The sensitivity and specificity of the MSLT in detecting sleepi- use of a percentile cut-off score. Accordingly a mean sleep
ness have not been clearly determined (137). The test–retest latency of less than 8 minutes on the 40-minute protocol is con-
reliability of the MSLT, however, has been documented in both sidered abnormal and values greater than this but less than 40
normal subjects and patients with narcolepsy. In subjects with minutes are of uncertain significance. It should be noted that
sleepiness caused by circadian rhythm sleep disorders, sleep there is little evidence linking mean sleep latency on the MWT
deprivation, and ingestion of alcohol, pathologic sleepiness has with the risk of accidents in real-world circumstances, therefore
been validated by MSLT. There is, however, poor correlation the sleep clinician should not rely exclusively on mean sleep
between the MSLT and ESS. The patient’s psychological and latency for determining the risk of accidents but should take
behavioral states also interfere with the MSLT results. The into consideration clinical history, physical findings, and com-
MSLT objectively measures tendency to sleep rather than the pliance with treatment. An important study by Philip et al.
likelihood of falling asleep. If the patient suffers from severe (141) correlated subjective and objective measures of sleepiness
anxiety or psychological disturbances causing behavioral stim- and driving performance in patients suffering from EDS. The
ulation, the MSLT may not show sleepiness even in a patient study included 38 untreated sleep apnea patients and 14 healthy
complaining of EDS. controls. Based on the number of inappropriate line-crossings
during a 90-minute real-life driving session, they determined
The Maintenance of Wakefulness Test the MWT sleep-onset score as follows: very sleepy, 0 to 19 min-
The MWT is a variant of the MSLT to measure a patient’s abil- utes; sleepy, 20 to 33 minutes; and alert, 34 to 40 minutes.
ity to stay awake (138,139). The MWT is performed similar to
MSLT at 2-hour intervals in a quiet dark room beginning about Actigraphy
1
12 to 3 hours after waking up. Four to five such tests are per- Colburn et al. (142) first documented use of accelerometers to
formed and each one lasts for 40 minutes. A polysomnogram record body movements. Tyron (143) in 1991 gave a compre-
and sleep logs are not required before the MWT. The patient hensive account of activity monitoring in psychology and med-
should be seated in bed with the back and head supported by a icine. Actigraphy is defined as the technique of recording and
bed rest, ensuring that the neck is comfortable. The patient quantifying movements. Monitoring of body movements and
should not smoke at least 30 minutes before each test and other activities can be performed continuously for days, weeks,
should not have caffeinated beverages on the day of the test. or even months by using an actigraph, also known as actometer
The conventional recording montage for the MWT is similar to or actimeter (144,145). This can be worn on the wrist or alter-
that used for the MSLT. At the start of the test, after lights out, natively on the ankle for recording arm, leg, and body move-
the patient is instructed to sit still and remain awake for as long ments. Actigraph uses piezoelectric sensors which generate
as possible. The patient is not allowed to use extraordinary electric currents in direct proportion to the amount of acceler-
measures to stay awake. Unequivocal sleep is defined as three ation. The activity usually covers frequency range of 0.5 to 15
consecutive epochs of stage N1 sleep or one epoch of any other Hz, which is generally the dominant range for human move-
stage of sleep. Trials are ended after 40 minutes if no sleep ments. The mechanical movements are converted into electrical
occurs and also after sleep onset as defined above. signals that are then sampled every tenth second over a prede-
termined time or epoch, which is generally one minute. These
Clinical Indications, Normative Data, samples are stored in the memory of the actigraph and then
Validity, and Reliability retrieved and analyzed in a computer. The principle of analysis
The MWT 40-minute protocol may be indicated in assessment is based on the fact that increased movements (as indicated by
of individual’s ability to remain awake when his/her inability to black bars in the actigraphy) are seen during wakefulness in
remain awake constitutes a public or personal safety issue contrast to markedly decreased movements or no movements
(136). The MWT is also indicated in patients with excessive (as indicated by the white bars interrupting the black bars dur-
sleepiness to assess response to treatment (e.g., response to ing sleep), although normal physiologic body and limb move-
stimulants in narcolepsy and idiopathic hypersomnia and ments and postural shifts during sleep will cause interruptions
response to CPAP titration in OSA patients). The MWT is less (small black bars) of the white background (Fig. 41.43). Several
useful and less sensitive than the MSLT as a diagnostic test for actigraph models are in the development stage to carefully reg-
narcolepsy. It should, however, be noted that the MSLT ulate the sampling frequencies, durations, filters, sensitivities,
858 Part V ■ Complementary and Special Techniques

Figure 41.43 Normal sleep–wake schedule.


This shows a wrist actigraph recording from a
healthy 60-year-old man with a regular
sleep–wake schedule. The subject goes to bed
around 10:00 PM and wakes up at 6:00 AM.
Physiologic body shifts and movements are indi-
cated by a few black bars in the white (sleep
period) areas. The waking period is indicated by
black bars reflecting activities.

and the dynamic range in order to detect and quantify PLMS, Recommendations for Actigraph
but no generally accepted standardized technique of quantify- The AASM’s Standards of Practice Committee suggests the fol-
ing and identifying PLMS discriminating from other move- lowing recommendations for actigraphy. Actigraphy may be a
ments (e.g., those resulting from parasomnia, nocturnal useful adjunct to history and physical examination and sleep
seizures, and other dyskinesias) is currently available. The role logs in patients with insomnia including sleep state mispercep-
of actigraphy in detecting, quantifying, and differentiating tion (paradoxical insomnia) and inadequate sleep hygiene
abnormal motor activities currently remains controversial, but (Figs. 41.45 and 41.46), circadian rhythm sleep disorders (151)
there is immense potential for such applications with the devel- (see Figs. 41.47 and 41.48), and excessive daytime somnolence.
opment of sophisticated models and techniques. More recently, Actigraphy may be a useful adjunct to detect the rest time and
in some models, sophisticated systems for counting movements activity patterns during modified portable sleep apnea testing.
have been developed and validated (146,147). This system Actigraphy may be useful in assessing EDS in situations where
therefore may be useful for therapeutic monitoring and for MSLT is not practical.
diagnostic ability in patients with PLMS and RLS (148,149). Actigraphy is useful to detect rest time and activity patterns
One of the available monitors (PAM-RL, Respironics, over days and weeks when sleep log or other methods cannot
Pittsburgh, PA) through a fine-grained analysis with 40-Hz provide such data.
sampling and storage at 10 Hz provides a description closely Actigraphy may be useful in monitoring circadian rhythm
matching the EMG recordings for these movements. The data disturbances in special populations (e.g., the elderly and nurs-
provide an excellent agreement with the EMG recordings of ing home patients); newborns, infants, children, and adoles-
the legs (Fig. 41.44) and with the nocturnal PSG for number cents; hypertensive individuals (to monitor circadian patterns
of leg movements (150). Studies have been conducted using of blood pressure and circadian therapeutic effectiveness of
actigraphy and PSG recordings simultaneously to validate the medications); depressed or schizophrenic patients; and individ-
ability of the actigraph to distinguish sleep from wakefulness. uals during space flight.
Assessment of sleep–wakefulness for over a prolonged period Actigraphy may be useful to measure outcomes in clinical
of time (days to weeks) has been the main utility of actigraph. trials. Actigraphs are not indicated for the diagnosis, assess-
Total sleep time and sleep-onset time, however, have shown ment, or severity of any sleep disorder including insomnia
variable results with discrepancy from PSG results. Although and OSAS.
computer algorithms are available for automatic sleep–wake
scoring, visual inspection of the raw data is necessary. Advantages of Actigraph over PSG
Rreliable and valid data are obtained using a specific actigraph These include the following: easy accessibility; inexpensive
model, but no universally validated data are available. recording over extended periods (e.g., days, weeks, or even
Actigraphs can differentiate sleep from wakefulness but can- months); recording of 24-hour activities at all sites: home, work,
not differentiate REM from NREM sleep and cannot identify or laboratories; usefulness in uncooperative and demented
different NREM sleep stages. Actigraphs and sleep logs are patients when laboratory PSG study is not possible; ability to
complementary. conduct longitudinal studies to monitor disease progression,
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 859

Figure 41.44 Example of the real-time output of


a high-precision activity monitor worn on the
ankle (bottom line) compared to anterior tibialis
EMG activity (top line). The middle line shows
the real-time automatic detection of a significant
leg movement made by the activity meter. The
decision rules for the real-time leg movement
detector create a 7-second delay in the detection.
(Reproduced from Chokroverty S. Sleep Disorders
Medicine: Basic Science, Technical Considerations
a nd Clinica l Aspects. 3rd ed. Philadelphia:
Saunders/ Elsevier; 2009, with permission.)

Figure 41.45 Actigraphy in insomnia (sleep state misperception): a 59-year-old man complain-
ing of insomnia since the age of 12 years. He was diagnosed to have DSM IV Axis personality
disorder (dependent personality) and panic attacks in the past, treated with benzodiazepines
(clordemetildiazepam 3 mg, flurazepam 30 mg) and zolpidem 10 mg. He denies any symptoms
of RLS, EDS, or daytime sleep attacks. Subjective sleep duration is 3 to 4 hours per night. In the
past he had numerous drugs for sleep amelioration but no clear and stable subjective improve-
ment was noted. An actigraphic monitoring (during drug reduction: clordemetildiazepam 2 mg,
flurazepam 15 mg, and no zolpidem) shows a clear misperception of sleep duration and qual-
ity. The recording shows normal nocturnal motor activity and sleep efficiency and duration.
Note sleep period during the afternoon. He complained of sleeping not more than 3 hours each
night. PSG, polysomnography; TST, total sleep time; SE, sleep efficiency; WASO, wakefulness
after sleep onset; SWS, slow wake sleep; PLMS, periodic limb movements in sleep. (Reproduced
from Chokroverty S, Thomas R, Bhatt M, eds. Atlas of Sleep Medicine. Philadelphia: Elsevier;
2005, with permission.)
860 Part V ■ Complementary and Special Techniques

Figure 41.46 This wrist actigraphic recording is


taken from a 50-year-old woman with chronic
insomnia due to inadequate sleep hygiene. The
actigraph worn for 11 days shows highly irregular
bedtime and wake-up time. The waking period is
indicated by black bars and the sleep period by
gray bars. Note excessive body movements (indi-
cated by black bars) during sleep period and brief
periods of sleepiness (white areas) during wake-
fulness (black bars). Note the subject did not
wear the actigraph on Monday, 5/ 16/ 05 from
7:00 PM to 6:30 AM (fourth from the top showing
a continuous, thick, black line).

Figure 41.47 Primary delayed sleep phase syn-


drome. This wrist actigraphic recording is taken
from a 29-year-old man with a lifelong history of
delayed sleep onset and delayed wake-up time.
The actigram shows his typical sleep period from
3:00 AM to 4:00 AM to 9:00 AM to noon (white
areas). If he has to wake up early in the morning,
he feels exhausted and sleepy all day. He feels
fine if he is allowed to follow his own schedule.
Melatonin at night did not help him. Morning
bright light therapy was suggested but the patient
declined. (Reproduced from Chokroverty S,
Thomas R, Bhatt M, eds. Atlas of Sleep Medicine.
Philadelphia: Elsevier; 2005, with permission.)

Figure 41.48 Sleep–awake schedule disorder in


a patient with acquired immunodeficiency syn-
drome (AIDS). This wrist actigram is taken from
a 46-year-old man with AIDS. The patient pre-
sented with sleep difficulty due to inability to fall
asleep and wake at desired bedtime and wake-up
time. This 10-day recording shows disorganiza-
tion of the sleep–wake schedule. There is a sug-
gestion of non–24-hour (hypernychthemeral)
syndrome with progressive delay of sleep onset
(arrows) from day 1 to day 6, and again delayed
sleep onset (arrows) from day 7 to day 10.
(Reproduced from Chokroverty S, Thomas R,
Bhatt M, eds. Atla s of Sleep Medicin e.
Philadelphia: Elsevier; 2005, with permission.)
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 861

remission, and therapeutic responses; ability to overcome the 10. Berger RJ. Tonus of extrinsic laryngeal muscles during sleep and
problem of night-time variability noted in many sleep disorders dreaming. Science. 1961;134:840.
and some nocturnal movement disorders; ability to discriminate 11. Rechtschaffen K, Kales A. A Manual of Standardized Terminology,
real movements from clinically insignificant EMG potentials; Techniques and Scoring System for Sleep Stages of Human Subjects.
Washington DC: US Government Printing Office; 1968.
usefulness in sleep state misperception; and ability to document
12. American Academy of Sleep Medicine. The AASM Manual 2007 for
delayed or advanced sleep phase syndrome or non–24-hour cir-
the Scoring of Sleep and Associated Events: Rules, Terminology and
cadian rhythm disorders although sleep logs may suggest such Technical Specifications. Westchester, IL: American Academy of Sleep
diagnosis. Finally, actigraphs are small, lightweight, easily main- Medicine; 2007.
tained, and inexpensive in contrast to bulky, labor intensive, and 13. Griesinger W. Berliner medizinisch-psychologische Gesclleschaft.
expensive, as well as inconvenient PSG recordings. Arch Psychiatr Nervenkr. 1868;1:200–204.
14. Freud S. Project from a scientific psychology. In: Bonaparte M,
Disadvantages and Limitations of Actigraph Recordings Freud A, Kris, E eds. The Origins of Psychoanalysis: Letters to
These include the inability to diagnose sleep apnea and to clarify Wilhelm Fliess, Drafts and Notes, 1895 to 1902. New York: Basic
the etiology of insomnia, overestimation of sleep when some Books; 1954:400.
insomniacs may lie down in bed for prolonged periods without 15. Wolpert S. A New History of India. New York: Oxford University
Press; 1982:48.
moving; inability to identify subjects with feigning sleep prob-
16. Jones B. Neurology of NREM sleep. In: Montagna P, Chokroverty S,
lems and to discriminate types of movements such as PLMS from
eds. Sleep Disorders, Handbook of Clinical Neurology. Amsterdam:
other body movements and provide any information about other Elsevier 2011 (In Press).
physiologic characteristics (e.g., EEG, EOG, and respiration); and 17. Saper CB, Scammell TE, Lu J. Hypothalamic regulation of sleep
inability to record specific data as the actigraph records all kinds and circadian rhythms. Nature. 2005;437:1257–1263.
of movements. Lack of standardization of placement of the acti- 18. Peyron C, Tighe DK, Van den Pol AN, et al. Neurons containing
graph may also be seen as a pitfall. Most commonly, the actigraph hypocretin (orexin) project to multiple neuronal systems. J Neurosci.
is placed on the nondominant side. Activity is somewhat differ- 1998;18:9996.
ent between the two sides with more activities being recorded 19. Siegel JM. Mechanisms of sleep control. J Clin Neurphysiol.
from the dominant than from the nondominant limbs. The over- 1990;7:49.
all agreement, however, for sleep period between the two sites is 20. McCarley RW. Neurobiology of REM and NREM sleep. Sleep Med.
2007;8:302–330.
not frequently different when compared with the PSG data.
21. Lu J, Sherman D, Devor M, et al. A putative flip-flop switch for
In conclusion, the actigraph is an inexpensive, useful
control of REM sleep. Nature. 2006;441:589.
method for longitudinal assessment of sleep–wake pattern, can 22. Brooks PL, Peever JH. Glycinergic and GABAA-mediated inhibi-
differentiate normal sleep patterns from those of disturbed tion of somatic motoneurons does not mediate rapid eye move-
sleep due to insomnia and SDB, and can differentiate normal ment sleep motor tonia. J Neurosci. 2008;28:3535.
sleep from sleep–wake schedule disturbances by recording over 23. Saper CB, Chou T, Scammell TE. The sleep switch: hypothalamic
long periods and longitudinal monitoring. control of sleep and wakefulness. Trends Neurosci. 2001;24:726.
24. Porkka-Hieskanen T, Strecker RE, Thakkar M, et al. Adenosine: a
REFERENCES mediator of the sleep-inducing effects of prolonged wakefulness.
Science. 1997;276:1265.
1. Berger H. Uber Das elektroencephalogramm Des Menschen. Arch 25. Chokroverty S. Physiological changes in sleep. In: Chokroverty S,
Psychiatr Nervenkr. 1929;87:527–570. ed. Sleep Disorders Medicine: Basic Science, Technical Considerations
2. Aserinsky E, Kleitman N. Regularly occurring periods of eye motil- and Clinical Aspects. 3rd ed. Philadelphia: Elsevier; 2009:80–104.
ity and concomitant phenomenon during sleep. Science. 1953; 26. Moore RY. Circadian timing and sleep-wake regulation. In:
119:273. Chokroverty S, ed. Sleep Disorders Medicine: Basic Science,
3. Holland JV, Dement WC, Raynal DM. Polysomnography: a response Technical Considerations and Clinical Aspects. 3rd ed. Philadelphia:
for a need for improved communication. Paper presented at the Elsevier; 2009:105–111.
14th annual meeting of the Association of the Psychophysiological 27. Daan S, Beersma DGM, Borbely AA. The timing of human sleep:
Study of Sleep, Association of the Psychophysiological Study of A recovery process gated by a circadian pacemaker. Am J Physiol.
Sleep, Jackson Hole, WY, 1974:121. 1984;246:R161–R178.
4. Caton R. The electric currents of the brain. BMJ. 1975;2:278. 28. Dinges DF, Broughton RJ. Sleep and Alertness: Chronobiological,
5. Loomis AL, Harvey EN, Hobart GA. Cerebral states during sleep, Behavioral and Medical Aspects of Napping. Raven Press; 1989.
as studied by human brain potentials. J Exp Physiol. 1937;21:127. 29. Dijk DJ, Czeisler CA. Contribution of the circadian pacemaker and
6. Kohlschutter E. Messeungen Dar Festigkei Des Schlafes. Zeitschrift the sleep homeostat to sleep propensity, sleep structure, electroen-
Fur Rationelle Medicin. 1863;17(3):209–253. cephalographic slow waves and sleep spindle activity in humans.
7. Basner M. Arousal threshold determination in 1862: Kohlschutter’s J Neurosci. 1995;15:3525–3538.
measurements on the firmness of sleep. Sleep Med. 2010;11:417–422. 30. Jasper HH. The ten-twenty electrode system of the International
8. Dement WC, Kleitman N. Cyclic variations of EEG during sleep Federation. Electroencephalogr Clin Neurophysiol. 1958;10:371.
and their relation to eye movements, body motility and dreaming. 31. Mouret J, Delorme F, Jouvet M. Activité des muscles de la face au
Electroencephalogr Clin Neurophysiol. 1957;9:673. cours du sommeil paradoxal chez l’homme. Compt Rend Soc Biol
9. Jouvet M, Michel F. Correlations electromyographiques Du Sommeil Paris. 1965;159:391–394.
Chez Le Chat Decortique et Mesencephalique Chroniue. Comp Rend 32. Bliwise D, Coleman R, Bergmann B, et al. Facial muscle tonus dur-
Soc Biol (Paris). 1959;153:422–425. ing REM and NREM sleep. Psychophysiology. 1974;11:497–508.
862 Part V ■ Complementary and Special Techniques

33. Report of AASM Task Force. Sleep-related breathing disorders in Task Force of the American Academy of Sleep Medicine. J Clin
adults: recommendations for syndrome definition and measure- Sleep Med. 2007;3:737–747.
ment techniques in clinical research. Sleep. 1999;22(5):667–689. 54. White DP, Gibb TJ, Wall JM, et al. Assessment of accuracy and
34. Kales A, Kales J. Recent findings in the diagnosis and treatment of analysis time of a novel device to monitor sleep and breathing in
disturbed sleep. N Engl J Med. 1974;209:487–499. the home. Sleep. 1995;18:115.
35. Williams RI, Karacan I, Hursch CJ. EEG of Human Sleep: Clinical 55. Tonelli de Oliveira AC, Martinez D, Vasconcelos LF, et al. Diagnosis
Applications. New York: Wiley; 1974. of obstructive sleep apnea syndrome and its outcomes with home
36. Bonnet MH, Arand DL. EEG arousal norms by age. J Clin Sleep portable monitoring. Chest. 2009;135:330.
Med. 2007;3(3):271–274. 56. Penzel T, Hirshkowitz M, Harsh J, et al. Digital analysis and tech-
37. Bonnet M, Carley D, Carskadon M, et al. EEG arousals: scoring nical specifications. J Clin Sleep Med. 2007;3(2):109–120.
rules and examples: a preliminary report of the Sleep Disorders 57. Schulz H. Rethinking sleep analysis: comment on the AASM
Atlas Task Force of the ASDA. Sleep. 1992;15:173–184. Manual for Scoring Sleep and Associated Events. J Clin Sleep Med.
38. Coleman RM, Pollak C, Weitzman ED. Periodic movements in 2008;4:99–103.
sleep (nocturnal myoclonus): relation to sleep-wake disorders. Ann 58. Guilleminault C, Stoohs R, Clerk A, et al. A cause of daytime
Neurol. 1980;8:416–421. sleepiness: the upper airway resistance syndrome. Chest. 1993;104:
39. Bonnet M, Carley D, Carskadon M, et al. Recording and scoring 781–787.
leg movements. Sleep. 1993;16:748–759. 59. Exar E, Collop N. The upper airway resistance syndrome. Chest.
40. Zucconi M, Ferri R, Allen R, et al. The official World Association 1999;115:1127–1139.
of Sleep Medicine standards for recording and scoring PLMS and 60. Norman RG, Ahmed MM, Walsleben JA, et al. Detection of respi-
PLMW developed in collaboration with a task force from the ratory events during NPSG: nasal cannula/pressure sensor versus
International RLS Study Group. Sleep Med. 2006;7:175–183. thermistor. Sleep. 1997;20:1175–1184.
41. Montplaisir J, Boucher S, Lesperance P, et al. Immobilization tests 61. Epstein MD, Chicoine SA, Hanumara RC. Detection of UARS using
and periodic leg movements in sleep for the diagnosis of restless a nasal cannula/pressure transducer. Chest. 2000;117:1073–1077.
legs syndrome. Mov Disord. 1998;13(2):324–329. 62. Budhiraja R, Goodwin JL, Parthasarathy S, et al. Comparisons of
42. Gastaut H, Tassinari C, Duron B. Etude polygraphique des manifes- NPT and thermistor for detection of respiratory events during
tations episodiques (hypniques et respiratoires) du syndrome de PSG in children. Sleep. 2005;28(9):1117–1121.
Pickwick. Rev Neurol. 1965;112:568–579. 63. Chokroverty S, Bhatt M, Goldhammer T. Polysomnographic record-
43. Young T, Palta M, Dempsey J, et al. The occurrence of sleep disor- ing technique. In: Chokroverty S, Thomas RJ, Bhatt M, eds. Atlas
dered breathing among middle-ages adults. N Engl J Med. 1993; of Sleep Medicine. Philadelphia: Elsevier/Butterworth-Heinemann;
328:1230–1235. 2005:1–28.
44. Bixler EO, Vgontas AN, Ten Have T, et al. Effects of age on sleep 64. Dauvilliers Y, Arnulf I, Mignot E. Narcolepsy with cataplexy. Lancet.
apnea in men: prevalence and severity. Am J Resp Cricial Care Med. 2007;369:499–511.
1998;157:144–148. 65. Nishino S. Clinical and neurobiological aspects of narcolepsy. Sleep
45. Pavlova MK, Duffy JF, Shea SA. PSG respiratory abnormalities in Med. 2007;8:373–399.
aymptomatic individuals. Sleep. 2008;31:241–248. 66. Frenette E, Kushida CA. Primary hypersomnia of central origin.
46. Terzano MG, Mancia D, Salati MR, et al. The cyclic alternating Semin Neurol. 2009;29:354–367.
pattern as a physiologic component of normal NREM sleep. Sleep. 67. Baker TL, Guilleminault C, Nino-Murcia G, et al. Comparative
1985;8:137–145. PSG study of narcolepsy and idiopathic CNS hypersomnia. Sleep.
47. Terzano MG, Parrino L, Sherieri A, et al. Atlas, rules, and record- 1986;9:232–242.
ing techniques for the scoring of cyclic alternating pattern (CAP) 68. Mattarozzi K, Bellucci C, Campi C, et al. Clinical, behavioral and
in human sleep. Sleep Med. 2001;2:537–553. PSG correlates of cataplexy in patients with narcolepsy/cataplexy.
48. Parrino L, Halasz P, Tassinari CA, et al. CAP, epilepsy, and motor Sleep Med. 2008;9:425–433.
events during sleep: the unifying role of arousal. Sleep Med Rev. 69. Harsh J, Peszka J, Hartwig G, et al. Night-time sleep and EDS in
2006;10:267–285. narcolepsy. J Sleep Res. 2000;9:309–316.
49. Indications for PSG Task Force. Practice parameters for the 70. Huang YS, Lin YH, Guilleminault C. PSG in Kleine–Levine syn-
indications for PSG and related procedures. Sleep. 1997;20: drome. Neurology. 2008;70(10):795–801.
406–422. 71. Rosenow F, Kotagel P, Cohen BH, et al. MSLT and PSG in diagnos-
50. Kushida CA, Littner MR, Morgenthaler T, et al. Practice parame- ing Kleine–Levin syndrome and periodic hypersomnia. J Clin
ters for the indications for PSG and related procedures: an update Neurophysiol. 2000;17(4):519–522.
for 2005. Sleep. 2005;28:499–521. 72. Kales A, Soldatos CR, Kales JD. Sleep disorders: insomnia, sleep-
51. Chesson AL Jr, Berry RB, Pack A. Practice parameters for the use walking, night terrors, nightmares, and enuresis. Ann Intern Med.
of portable monitoring devices in the investigation of suspected 1987;106:582–592.
obstructive sleep apnea in adults. Sleep. 2003;26:907. 73. Ohayon M, Guilleminault C, Priest R. Night terrors, sleepwalking,
52. Department of Health and Human Services, Center for Medicare and confusional arousals in the general population: their frequency
and Medicaid Services. Decision Memo for Continuous Positive and relationship toother sleep and mental disorders. J Clin
Airway Pressure (CPAP) Therapy for Obstructive Sleep Apnea (OSA). Psychiatry. 1999;60:268–276.
CAG#0093R. March 13, 2008. Available at: http://www.cms.hhs.gov/ 74. The International Classification of Sleep Disorders. Diagnostic and
mcd/viewdecisionmemo.asp?from2 =viewdecisionmemo.asp&id= Coding Manual. 2nd ed. Westchester, IL: American Academy of
204&. Accessed August 7, 2010. Sleep Medicine; 2005.
53. Collop NA, Anderson WM, Boehlecke B, et al. Clinical guidelines 75. Schenck CH, Bundie SR, Ettinger MG, et al. Chronic behavioral
for the use of unattended portable monitors in the diagnosis of disorders of human REM sleep. A new category of parasomnia.
obstructive sleep apnea in adult patients. Portable Monitoring Sleep. 1986;9:293–308.
Chapter 41 ■ Polysomnography: Technical and Clinical Aspects 863

76. Boeve B, Silber M, Parisi J, et al. Synucleinopathy pathology and 97. Mahowald M. Hope for the PLMS quagmire. Sleep Med. 2002;3:
REM sleep behavior disorder plus dementia or parkinsonism. 463–464.
Neurology. 2003;61:40–45. 98. Howell MJ, Schenck CH, Crow SJ. A review of nighttime eating
77. Schenck C, Mahowald M. REM sleep behavior disorder: clinical disorders. Sleep Med Rev. 2009;13(1):23–34.
developmental, and neuroscience perspectives 16 years after for- 99. Siddiqui F, Osuna E, Chokroverty S. Writing emails as part of sleep-
mal identification. Sleep. 2002;25:120–138. walking after increase in Zolpidem. Sleep Med. 2009;10(2):262–264.
78. Lapierre O, Montplaisir J. PSG features of REM sleep behavior 100. Pevernagie D, Boon P, Mariman A, et al. Vocalization during
disorder: development of a scoring method. Neurology. 1992;42: episodes of prolonged expiration: a parasomnia related to REM
1371–1374. sleep. Sleep Med. 2001;2:19–30.
79. Foldvary N, Caruso AC, Maschaa E, et al. Identifying montages 101. Vetrugno R, Provini F, Plazzi G, et al. Catathrenia (nocturnal
that best detect EEG seizure activity during polysomnography. groaning): a new type of parasomnia. Neurology. 2001;56:681–683.
Sleep. 2000;23:221–229. 102. Siddiqui F, Walters AS, Chokroverty S. Catathrenia: A rare para-
80. Foldvary-Schaefer N, DeOcamp J, Mascha E, et al. Accuracy of somnia which may mimic central sleep apnea on polysomno-
seizure detection using abbreviated EEG during polysomnogra- gram. Sleep Med. 2008;9:460–461.
phy. J Clin Neurophysiol. 2006;23:68–71. 103. Chokroverty S. Sleep disordered breathing in neuromuscular dis-
81. Walczak TS, Radtke RA, Lewis DV. Accuracy and interobserver orders: a condition in search of recognition. Muscle and Nerve.
reliability of scalp ictal EEG. Neurology. 1992;42:2279–2285. 2001;24:451–455.
82. Derry CP, Davey M, Johns M, et al. Distinguishing sleep disorders 104. Chokroverty S. Neuromuscular disorders and sleep dysfunction.
from seizures: diagnosing bumps in night. Arch Neurol. 2006;63: In: Montagna P, Chokroverty S, eds. Sleep Disorders, Handbook of
705–709. Clinical Neurology. Amsterdam: Elsevier 2011 (In Press).
83. Jung R, Kuhlo W. Neurophysiological studies of abnormal night 105. Labanowski M, Schmidt-Nowara W, Gulleminault C. Sleep and
sleep and the Pickwickian syndrome. Prog Brain Res. 1965;18:140. neuromuscular disease: frequency of sleep disordered breathing
84. Banno K, Kryger MH. Sleep apnea: clinical investigations in humans. in a neuromuscular disease clinic population. Neurology. 1996;17:
Sleep Med. 2007;8:400. 1173–1180.
85. Yaggi HK, Concato J, Kernan WN, et al. Obstructive sleep apnea as 106. Langevan B, Petijean T, Philip F, et al. Nocturnal hypoventilation in
a risk factor for stroke and death. N Engl J Med. 2005; 353:2034. chronic respiratory failure (CRF) due to neuromuscular disease.
86. Sanders MH, Strollo PD, Jr, Atwood CW, Jr., et al. Positive airway Sleep. 2000;23(suppl 4):S204–S208.
pressure in the treatment of sleep related breathing disorders. In: 107. Martin TJ, Sanders MH. Chronic alveolar hypoventilation: a
Chokroverty S, ed. Sleep Disorders Medicine: Basic Science, Technical review for the clinician. Sleep. 1995;18:617–634.
Considerations and Clinical Aspects. 3rd ed. Philadelphia: Saunders- 108. Chokroverty S. Polysomnography in movement disorders. In:
Elsevier; 2009:340. Hallett M, ed. Handbook of Clinical Neurophysiology: Movement
87. Berry RB, Hill G, Thompson L, et al. Portable monitoring and auto Disorders. Amsterdam: Elsevier; 2002:139.
titration vs. polysomnography for the diagnosis and treatment of 109. Chokroverty S. Sleep and neurodegenerative diseases. Semin
sleep apnea. Sleep. 2008;31:1423. Neurol. 2009;29:446–468.
88. Walters AS. Toward a better definition of the restless legs syn- 110. Arnulf I, Merino-Andreu M, Blocs F, et al. REM sleep behavior
drome. The International Restless Legs Syndrome Study Group. disorder and REM sleep without atonia in patients with progres-
Mov Disord. 1995;10:634–642. sive supranuclear palsy. Sleep. 2005;28:349–354.
89. Allen RP, Picchietti D, Hening WA, et al. Restless legs syndrome: 111. Sixel-Doring F, Scweitzer M, Mollenhauer B, et al. Polysomno-
diagnostic criteria, special considerations, and epidemiology. A graphic findings, a video-based sleep analysis and sleep percep-
report from the restless legs syndrome diagnosis and epidemiology tion in progressive supranuclear palsy. Sleep Med. 2009;10:
workshop at the National Institute of Health. Sleep Med. 2003; 407–415.
4:101–119. 112. Segawa M, Hysaka A, Miwakawa F, et al. Hereditary progressive
90. Hening WA, Allen RP, Chokroverty S, et al., eds. Restless Legs dystonia with marked diurnal fluctuation. Adv Neurol. 1976;
Syndrome. Philadelphia: Saunders/Elsevier; 2009. 14:215–233.
91. Winkelmann J, Schormair B, Lichtner P, et al. Genome-wide asso- 113. Salazar-Grueso EF, Rosenberg RS, Roos RP. Sleep apnea in olivo-
ciation study in restless legs syndrome identifies common variants pontocerebellar degeneration: treatment with trazodone. Ann
in three genomic regions. Nat Gen. 2007;39:1000–1006. Neurol. 1988;23:399–401.
92. Stefansson H, Rye DB, Hicks A, et al. A genetic risk factor for peri- 114. Neil JF, Holzer BC, Spiker DG, et al. EEG sleep alterations in
odic limb movements in sleep. N Engl J Med. 2007;357:703–705. olivopontocerebellar degeneration. Neurology. 1980;30:660–662.
93. Hening W, Walter AS, Allen RP, et al. Impact, diagnosis and treat- 115. Salva MA, Gulleminault C. Olivopontocerebellar degeneration,
ment of restless legs syndrome (RLS) in a primary care study. Sleep abnormal sleep and REM sleep without atonia. Neurology. 1986;36:
Med. 2004;5(3):237–246. 576–577.
94. Hornyak M, Feig B, Voderholzer U, et al. Polysomnography find- 116. Lugaresi E, Medori R, Montagna P. Fatal familial insomnia and
ings in patients with restless legs syndrome and in healthy con- dysautonomia with selective degeneration of thalamic nuclei. N
trols: a comparative observational study. Sleep. 2007;30:861–865. Eng J Med. 1986;315:997.
95. Saletu B, Anderer P, Saletu M, et al. EEG mapping, psychometric, 117. Montagna P. Fatal familial insomnia and the role of the thalamus
and polysomnographic studies in restless legs syndrome (RLS) in sleep regulation. In: Montagna P, Chokroverty S, eds. Handbook
and periodic limb movement disorder (PLMD) patients as com- of Clinical Neurology. Amsterdam: Elsevier 2011 (In Press).
pared with normal controls. Sleep Med. 2002;3(suppl):S35–S42. 118. Broughton RJ, Mullington JM. Polysomnography; principles and
96. Winkelman JW, Redline S, Baldwin CM, et al. Polysomnographic applications in sleep and arousal disorders. In: Niedermeyer E,
and health related quality of life correlates of restless legs syn- Lopes Da Silva F, eds. Electroencephalography. 5th ed. Philadelphia:
drome in the sleep heart health study. Sleep. 2009;32:772–778. Lippincott Williams & Wilkins; 2005:899–936.
864 Part V ■ Complementary and Special Techniques

119. Hauri P, Hawkins DR. Alpha-delta sleep. Electroencephalogr Clin 136. Littner MR, Kushaida C, Wise M, et al. Practice parameters for
Nuerophysiol. 1973;34:233. clinical use of the multiple sleep latency test and the maintenance
120. Gharagozlou P, Seyffert M, Santos R, et al. Rhythmic movement of wakefulness test. Sleep. 2005;28:113–121.
disorder associated with respiratory arousals and improved by 137. Cherbin R. Assessment of sleepiness. In: Chokroverty S, Hening
CPAP titration in a patient with restless legs syndrome and sleep WA, Walters AS, eds. Sleep and Movement Disorders. Boston:
apnea. Sleep Med. 2009;10:501–503. Butterworth-Heinemann; 2002.
121. Chokroverty S, Thomas RJ, Bhatt M. Atlas of Sleep Medicine. 138. Doghramji K, Mitler M, Sangal RB, et al. A normative study of
Philadelphia: Butterworth-Heinemann; 2005. the maintenance of wakefulness test (MWT). Electroencephalogr
122. Cosentino FL, Lero I, Tripodi M, et al. the neurophysiology of the Clin Neurophysiol. 1997;103:554.
alternating leg muscle activation (ALMA) during sleep: study of 139. Doghramji K. The maintenance of wakefulness test. In:
one patient before and after treatment with pramipexole. Sleep Chokroverty S, ed. Sleep Disorders Medicine: Basic Science,
Med. 2006;7:63–71. Technical Considerations and Clinical Aspects. Philadelphia:
123. Chervin RD, Consens FB, Kutluay E. Alternating leg muscle acti- Saunders/Elsevier; 2009:224–228.
vation during sleep and arousal: a new sleep-related motor phe- 140. Mitler MM, Miller JC, Lipsitz JJ, et al. The sleep of long-haul
nomenon? Mov Disord. 2003;18:551–559. truck drivers. N Engl J Med. 1997;337:755–761.
124. Montagna P, Provini F, Plazzi G, et al. Propriospinal myoclonus 141. Philip P, Sagaspe P, Taillard G, et al. Maintenance of wakefulness
upon relaxation and drowsiness: a cause of severe insomnia. Mov test, obstructive sleep apnea syndrome, and driving risk. Ann
Disord. 1997;12:66–72. Neurol. 2008;64:410–416.
125. Vetrugno R, Provini F, Meletti S, et al. Propriospinal myoclonus 142. Colburn TR, Smith BM, Guarini JJ, et al. An ambulatory activity
at the sleep-wake transition: a new type of parasomnia. Sleep. monitor with solid state memory. ISA Transaction. 1976;15:
2001;24:835–843. 114–154.
126. Critchley M. The Predormitum. Rev Neurol (Paris). 1955;93:101. 143. Tyron WW. Activity Measurement in Psychology and Medicine.
127. Alattar MA, Scharf SM. Opioid-associated central sleep apnea: a New York: Plenum Press; 1991.
case series. Sleep Breath. 2009;13:201–206. 144. Sadeh A, Hauri PJ, Kripke DF, et al. The role of actigraphy in the
128. Farney RJ, Walker JM, Boyle KM, et al. Adaptive servoventilation evaluation of sleep disorders. Sleep. 1995;18:288–302.
(ASV) in patients with sleep disordered breathing associated with 145. Littner M, Kushida CA, Anderson WM, et al. Practice parameters
chronic opioid medication for non-malignant pain. J Clin Sleep for the role of actigraphy in the study of sleep and circadian
Med. 2008;4:311–319. rhythms: An update for 2002. Sleep. 2003;26:337–341.
129. Pepin JL, Dale D, Argod J, et al. Recommendations for practical use 146. Sforza E, Johannes M, Claudio B. The PAM-RL ambulatory device
of pulse transit time as a tool for respiratory effort measurements for detection of periodic leg movements: a validation study. Sleep
and microarousal recognition. In: Chokroverty S, Thomas R, Bhatt Med. 2005;6:407.
M, eds. Atlas of Sleep Medicine. Philadelphia: Elsevier/Butterworth- 147. Kemlink D, Pretal M, Sounka K, et al. A comparison of
Heinemann; 2005:262–271. polysomnographic and actigraphic evaluation of periodic limb
130. Pittman SD, Thomas RJ. Peripheral arterial tonometry. In: movements in sleep. Neurol Res. 2008;30:234.
Chokroverty S, Thomas R, Bhatt M, eds. Atlas of Sleep Medicine. 148. Allen RP. Improving RLS diagnosis and severity assessment:
Philadelphia: Elsevier/Butterworth-Heinemann; 2005:285–288. polysomnography, actigraphy and sleep log. Sleep Med. 2007;8:S13.
131. Chokroverty S. Sleep disorders. In: Bradley W, Daroff R, Fenichel 149. Kohnen R, Allen RP, Benes H, et al. Assessment of restless legs
G, et al., eds. Neurology in Clinical Practice. Philadelphia: Elsevier/ syndrome—methodological approaches for use in practice and
Butterworth-Hinemann; 2008:1947. clinical trials. Move Disord. 2007;22:S485.
132. Johns MW. A new method for measuring daytime sleepiness: the 150. Allen RP. Activity monitoring to diagnose and evaluate motor
Epworth Sleepiness Scale. Sleep. 1991;14:540. abnormalities of sleep. In: Henning W, Chokroverty S, eds. Topics
133. Keenan SA. Polysomnography technique: an overview. In: in Movement Disorders in Sleep, Course Syllabus: ASDA Annual
Chokroverty S, ed. Sleep Disorder Medicine: Basic Science, Clinical Meeting. San Francisco: American Sleep Disorders Association;
Consideration and Technical Aspects. Philadelphia: Saunders/ 1997.
Elsevier; 2009:137–156. 151. Morganthaler TI, Lee-Chiung T, Friedman AG, et al. Practice
134. Ackersted T, Gilberg M. Subjective and objective sleepiness in the parameters for the clinical evaluation of circadian rhythm sleep
active individual. Int J Neurosci. 1990;52:29–37. disorders; an American Academy of Sleep Medicine report. Sleep.
135. Carskadon MA, Dement WC, Mitler M, et al. Guidelines for the 2007;30:1445–1459.
multiple sleep latency test (MSLT: a standard measure of sleepi-
ness). Sleep. 1986;9:519.
CHAPTER
Magnetoencephalography:
Methods and Applications
RIITTA HARI
42
R COMPARISON OF EEG AND MEG
ecording of weak magnetic fields outside the head by
means of magnetoencephalography (MEG) emerged
in the late 1960s, 40 years after the invention of MEG is closely related to EEG. Despite different sensitivities of
the human EEG. The first instrument was an induction coil EEG and MEG to sources of different orientations and locations,
magnetometer with 2 million turns of wire. It was used to the primary currents causing the signals are the same. Similarities
detect the magnetic alpha rhythm by means of signal averaging, between the MEG and EEG waveforms are therefore to be
with the electric alpha as the time reference (1). The MEG expected. The advantage of MEG over EEG in source identifica-
method became more practical with the introduction tion results mainly from the transparency of the skull and other
of SQUID (superconducting quantum interference device) extracerebral tissues to the magnetic field, in contrast to the sub-
magnetometers in the early 1970s; since then it has been possi- stantial distortion and smearing of the electric potentials. Thus
ble to record both spontaneous and evoked magnetic signals of the MEG pattern outside the head is less distorted than the EEG
the human brain without any EEG reference. Rapid develop- distribution on the scalp. The magnetic recording is also refer-
ment of the technology has taken place during the last few ence-free, whereas the electric brain maps depend on the location
years: sensor arrays with whole-scalp coverage are now com- of the reference electrode. As a result it is often difficult to make
mercially available, and signal analysis methods have progressed a reasonable guess of the source locations by visual inspection of
quickly. At present, tens of laboratories worldwide utilize MEG the EEG data. Reference-free EEG presentations can be obtained
for exploration of normal and abnormal functions of the by calculating the “surface Laplacians” (19,20). However, even
human brain, and MEG results continue to influence interpre- then one has to proceed to neural sources for proper interpreta-
tation of EEG data. tion, and problems arise for several simultaneous source. The
In this chapter, the basic principles of MEG are reviewed Laplacians cannot be calculated for the outermost electrodes,
briefly, with examples mainly from our own laboratory. Both which reduces the brain areas that can be characterized.
spontaneous and evoked activities in normal subjects and in Figure 42.1 shows a current dipole in a spherical volume
some neurologic patients are discussed to the extent which is conductor consisting of four layers of different conductivities
supposed to be of immediate interest to a clinical neurophysiol- that simulate the brain, the cerebrospinal fluid, the skull, and
ogist. Several review articles are available for consulting MEG the scalp. The resulting distributions of electric potential and
findings directly related to basic brain research and relevant magnetic field are “dipolar,” that is, display two extrema of
methodology (2–18). The main advantages of MEG are its good opposite polarities, but rotated by 90 with respect to each
spatial resolution in locating cortical events and its selectivity to other. The isocontour lines are relatively more tight in the mag-
activity of the fissural cortex. The sites of active brain areas can netic than in the electric pattern because concentric electric
be located with respect to external landmarks on the head, inhomogeneities smear only the electric potential, while the
brain structures, or functional brain regions (identified by magnetic pattern is not influenced. In an ideal sphere, a single
means of sources of evoked responses). superficial tangential dipole can be found one third more accu-
One goal of MEG recordings is to obtain information about rately on the basis of magnetic than electric recordings (21). For
the neural generators of various signals. The EEG research has interpretation of MEG signals recorded from the real brain, it is
traditionally focused on temporal waveforms, and a whole sufficient to use a realistically shaped model consisting of the
branch of electrophenomenology has arisen around EEG brain only, while accurate EEG calculations require a full mul-
“graphoelements,” which have been correlated with different ticompartment model with known conductivities and shapes
tasks and stimulation parameters, clinical states, and even with for the brain, skull, cerebrospinal fluid, and scalp.
personality factors. It is clear that a better understanding of the MEG’s selectivity to tangential currents in the presence of
generators underlying the EEG signals would permit a more several simultaneous sources is an important advantage in
precise and physiologic interpretation of abnormalities. MEG practical work, as is illustrated, for example, by the early differ-
has considerably clarified these issues during the last decade, entiation between multiple cortical areas activated by
both at conceptual and practical level, and may have much to somatosensory stimuli (12,22–26). Moreover, it is often more
offer in the future. straightforward to interpret MEG than EEG data.

865
866 Part V ■ Complementary and Special Techniques

“The contribution of neuronal models: in vitro and computa-


tional models” in Chapter 5).

MEG INSTRUMENTATION
The magnetic field generated by cerebral currents is two orders
of magnitude weaker than that produced by the heart and only
a tiny part of the steady magnetic field of the earth (Table 42.1).
To avoid external magnetic artifacts caused by moving vehicles,
power lines, radio transmission, and so on, it is common to
carry out the recordings within a magnetically shielded room,
usually made of several layers of aluminum and mu-metal.
Biomagnetic measurements can also be performed without
magnetic shielding when special compensation techniques are
available. In general, it is however better to prevent than to
compensate for artifacts. Adequate magnetically shielded rooms
Figure 42.1 Magnetoencephalographic (MEG) and electroencephalo-
are commercially available.
graphic (EEG) field patterns over a concentric four-layer sphere when a
Figure 42.2A gives a schematic illustration of, now already
tangential current dipole (shown by the arrow) is active in an area
old-fashioned, arrangement during the MEG recording. The
approximating the second somatosensory cortex in the upper lip of the
subject lies in a magnetically shielded room and the neuromag-
Sylvian fissure. The shadowed areas indicate the magnetic flux out of
netometer, containing SQUID sensors immersed in liquid
the head (MEG) and positive potential (EEG). The lower part of the
helium (at –269 C), is positioned close to the head. To replace
figure illustrates schematically, with shadowed ellipsoids, the inaccu-
the evaporating helium, the Dewar container has to be refilled
racy region of the dipole location when computed “backwards” from
regularly, in modern devices about once a week. During record-
the signals on the surface. The dipole is assumed to be in a wall of a
ings with the present-day devices that cover the whole scalp in
cortical fissure.
a helmet-shaped array, such as the 306-channel neuromagne-
tometer in Figure 42.3, the subject is typically sitting during the
measurement.
Since both electric and magnetic signals are generated by The cerebral magnetic field is coupled into the SQUID
the same primary currents that flow in the brain and the sensors through superconducting flux transformers. The
nearby tissues, one should pay attention to both MEG and transformer configuration is important for the device’s sensi-
EEG data when drawing conclusions on brain function from tivity to different source current configurations and to arti-
either type of recordings. For example, a dipolar potential dis- facts. A magnetometer, containing a single pick-up loop in
tribution could be explained equally well with two radial cur- the flux transformer, is most sensitive to signals but also to
rent dipoles or with one tangential dipole. However, the artifacts (Fig. 42.2C). A more elaborate axial first-order gra-
existence of a clear magnetic pattern, rotated 90 with respect diometer contains a compensation coil, wound in the direc-
to the electric one, implies that the latter explanation should tion opposite to the pickup coil (Fig. 42.2B,C); this
be favored (27). The minimum requirement for sound data
interpretation is that the conclusions based on EEG and MEG
do not contradict. Tabl e 4 2 . 1
Some current sources (very deep and radial) are more reli-
ably picked up by EEG than MEG, but MEG counterparts of Orders of Magnitude of Magnetic Fields
auditory brainstem responses can be detected with MEG as (in femtotesla, fT = 10–15 T)
well and they can give important information about the
source configuration (28). However, the contribution of even Magnetic resonance 1,000,000,000,000,000 (=1 T)
deep activity to the MEG signals can be probed by inserting imaging
sources to interesting brain structures (such as thalamus or Steady magnetic field 100,000,000,000
hippocampus) and then calculating the best fit of these of the earth
sources, as a function of time, to the measured signals (29,30).
In theory, current loops are electrically silent but magnetically Magnetocardiogram 100,000
visible. In practice, however, ideal current loops have not been Cerebral -rhythm 1000
observed, and all cerebral currents that give rise to MEG sig- Cerebral evoked response 100
nals are also expected to generate electric potentials on the
scalp. For maximum information, the MEG and EEG tech- Sensitivity of magnetometers 10
niques should be combined, and the simultaneously recorded Noise within a shielded room 1
data should be interpreted with methods that take advantage
of the complementarity of the records (see also the section Note that the sensitivity of the present-day magnetometers is below 5 fT/ 1 Hz.
Chapter 42 ■ Magnetoencephalography: Methods and Applications 867

Figure 42.2 (A) Magnetoencephalogram


recording. The subject is lying in a magnet-
ically shielded room with his head sup-
ported by a vacuum cast. The Dewar
containing the SQUID sensors is brought as
close to the head as possible, without direct
contact, and the magnetic field (or its gradi-
ent) is picked up outside the head at several
locations simultaneously. (B) Two flux
transformer configurations. The first-order
axial gradiometer (left) measures essentially
Br and detects the maximum signals at both
sides of the dipole. The planar gradiometer
(right) measures the tangential derivative
e Br/ x or Br/ y; the maximum signal is
e e e
detected just above the dipole. (C)
Dependence of signal strength (in arbitrary
units) on the depth of a current dipole when
measured by (i) a magnetometer, (ii) a first-
order axial gradiometer, (iii) a second-order
axial gradiometer, and (iv) a planar figure-
of-eight gradiometer. Simulations in a
sphere of 10-cm radius.

configuration decreases the influence of distant disturbances tion. Methods have been developed to present data measured
that link the same magnetic flux into both coils. Therefore, with different coil configurations in a standard format (31).
the output of the axial first-order gradiometer is essentially On the basis of axial gradiometers, some users compute vir-
determined by the signal of the nearby neuronal source itself. tual planar gradiometer signals that work well as indicators of
Furthermore, since the distance between the pickup and the most likely source areas but have ( 2 times) more noise
compensation coils of a first-order axial gradiometer is sev- than the original signals.
eral centimeters, the measured signal approximates the An essential part of the measurement system is the head-posi-
amplitude of the radial field component Br rather than its tion indicator, which gives the exact measurement sites and the
axial derivative. Higher-order gradiometers are necessary for orientations of the sensors with respect to the head. We obtain
measurements performed without a magnetic shield; in these this information by placing three to four small wire loops on
systems the sensitivity is reduced to the distant artifacts but known sites on the scalp. The field pattern produced by currents
to some extent also to the nearby brain currents. led through the loops is then measured with the multichannel
The contour plots in Figure 42.2B illustrate the pattern of magnetometer. In another commonly used head-position indica-
Br , the magnetic field radial to the surface, generated by a tan- tor system a transmitter is connected to magnetic sensors, which
gential current dipole in a sphere. The signal strengths meas- are fixed on the dewar, and three receivers are placed on the sub-
ured with an axial gradiometer form a spatial pattern similar ject’s head. The head-position indicator devices allow the posi-
to the field itself, with extrema of opposite polarities at the tion of the magnetometer to be determined with respect to
two sides of the dipole. In contrast, the planar gradiometer external landmarks on the head with 2- to 3-mm accuracy. The
yields the maximum signal when centered just over the dipole coordinate system is preferably global, typically fixed on the basis
at the location of the steepest field gradient. The planar gra- of the nasion, inion, and the preauricular points so that it can be
diometer is able to detect the location of the source even at the easily transported to, for example, Talairach space commonly
edge of the sensor array and the essential information from used in functional magnetic resonance imaging (MRI). The
the field pattern can thus be obtained from a rather small important landmarks on the head and the shape of the skull can
measurement area. On the other hand, information about the be determined with a three-dimensional digitizer.
depth of the source is more accurate with axial than planar To determine the current distribution within the head, the
sensors since the gradient decreases relatively more rapidly as magnetic field must be sampled at several locations, preferably
the function of source depth than does the field itself (Fig. simultaneously. When only single-channel magnetometers were
42.2C). However, whole-scalp sensor coverage largely coun- in use, it sometimes took several days to complete a field map.
terbalances this drawback. Although the patterns measured by For example, the MEG recordings of the first epileptic patients
both axial and planar gradiometers are easily interpreted, in lasted for 16 hours (32)! Changes in the subject’s attentive state
practice it is often useful that the maximum signal detected by and vigilance were thus unavoidable. With the present-day
the planar gradiometers suggests the approximate source loca- helmet-shaped neuromagnetometers that cover the whole scalp
868 Part V ■ Complementary and Special Techniques

Figure 42.3 A modern 306-SQUID neuromagnetometer used in our laboratory. Each of the 102 three-channel sensor
elements comprises two orthogonal planar gradiometers and one magnetometer; the helmet-shaped arrangement of the
elements is shown on the right (courtesy of Mika Seppä). The planar flux transformers measure the tangential derivatives
Br/ x and Br/ y of the radial field component Br.
e e e e

(Fig. 42.3), the whole field pattern can be measured without However, the different devices provide practically very similar
moving the instrument. This progress of instruments has finally information about the brain’s neuronal currents.
made MEG recordings feasible for comprehensive studies of With the present technology, the intrinsic noise of the
integrative brain functions in health and disease. SQUID is no longer a problem, and the main “noise” arises
The optimal spacing of the sensors is determined by the spa- from the brain itself. Taking into account the distance of the
tial frequency of the signal distribution and, therefore, only coils from the source, pickup coils with diameters around 2 cm
marginal benefit can be obtained by reducing the sensor spac- are reasonably sensitive and do not lose significant information.
ing below the distance between the source and the sensor, that Decreasing the distance to the source improves the resolution of
is, about 3 cm (33,34). Thus about 150 sensors would suffice to the system. Thus it would be important to develop flux trans-
cover the whole cortex, and the newest instruments with 200 to former arrays that can be put close to the scalp of each subject,
300 sensors provide dense-enough spatial sampling for MEG independent of the head shape. Such systems could be made
recordings. The diameter of the sensor coil is always a matter of with high-Tc superconductor arrays that require less insulation.
compromise: the larger the coil, the more sensitive it is, but also However, despite the promising recordings of MEG signals with
the more it averages the magnetic field, thereby leading to loss a single-channel high-Tc neuromagnetometer (35), the tech-
of information. nique has not developed during the last years. The main reasons
Currenlty (in 2010) about 140 whole-scalp neuromagnetome- are the higher (thermal) noise of the sensors and the difficulty
ters are in use in clinical and research settings all over the world. to easily manufacture multichannel high-Tc sensor arrays.
The devices of different manufacturers vary in the number of Interestingly, totally new sensor principles have been
channels, in the pickup coil configuration (e.g., first-, second-, or recently introduced: atomic magnetometer (36) and magne-
third-order axial gradiometer, planar gradiometer, or magne- toresistive sensors operating up to 77 K (37), in contrast to the
tometer), as well as in the area covered by the sensor array. SQUIDs that are operated at 4 K.
Chapter 42 ■ Magnetoencephalography: Methods and Applications 869

SOURCE IMAGING
Since the aim of MEG studies is to obtain information about
brain function, the field pattern should be interpreted in terms
of cerebral currents. Yet, due to the nonuniqueness of the
inverse problem—namely that several current distributions
can, in principle, produce identical magnetic field patterns out-
side the head—the interpretation usually requires the use of
specific source and volume conductor models. Thus the situa-
tion is more complicated than in MRI, functional magnetic res-
onance imaging (fMRI), or in positron emission tomography
(PET), where the inverse problem can be solved uniquely.
However, physiologically meaningful solutions of MEG pat-
terns can be found by utilizing constraints based on the known
anatomy and physiology of the brain.

CURRENT DIPOLE
The most commonly used source model in MEG studies is a
current dipole within a sphere. The dipole can be characterized
by means of five parameters, three for its three-dimensional
location, one for its orientation (only the plane parallel to the
surface of the sphere is relevant), and one for its strength. In a
sphere, only primary currents tangential to the surface will
produce a magnetic field. In reality, signals may also be
detected from the convexial cortex where the currents are con-
Figure 42.4 The dependence of the radial component of the magnetic
siderably closer to the detector than are the currents within the
field on the distance between the source and the detector. Two current
wall of a fissure. Deviation of the convexial source by only 10
dipoles are situated in a homogeneous sphere (radius 10 cm), 1 cm
to 20 degrees from the radial orientation is enough to give a
beneath the surface, 3 cm from each other, and symmetrically with
signal as large as that produced by a tangential dipole of the
respect to the origin. The field was calculated on an arc perpendicular
same size but 2 cm deeper in the fissural cortex. It therefore
to the orientation of the dipoles (x-axis). The pattern is complicated on
seems probable that under realistic conditions nearly radial
the surface, whereas 2 cm outside the sphere the higher spatial frequen-
sources may contribute significantly to the MEG signals. This
cies have faded away and the dipolar term dominates. The amplitudes
notion is supported by simulations that took into account the
have been normalized according to the maximum value; the maximum
real geometry of the cerebral cortex (38). A practical example
field would be about seven times stronger on the surface than 2 cm
of the possibility to detect tilted currents is the identification of
above it. (Adapted from Hari R. Interpretation of cerebral magnetic
magnetic counterparts of the P22 and P25 somatosensory
fields elicited by somatosensory stimuli. In: Basar E, ed. Springer Series
responses, which are believed to arise in the convexial cortex
of Brain Dynamics. Berlin/ Heidelberg: Springer-Verlag; 1988:305–310.)
(39–41).
In dipole modeling, the location of the “equivalent current
dipole” (ECD) is found by a least-squares fit to the data, typically
at a time point of a clearly dipolar field pattern. In progressing poor. It is also often useful to study the residual field, that is, the
toward a multidipole model, one may extract the field patterns difference between the measured field and that predicted by the
produced by the already identified sources to facilitate further model, or to compare the waveforms predicted by the model with
analysis (42–44). The success of dipole models in MEG interpre- the measured waveforms. If the residual field shows systematic
tation derives, in part, from the difficulty to discern the details of features that cannot be explained by noise, a different source con-
the brain activation pattern from the typical measurement dis- figuration must be considered.
tance of at least 3 cm from the source (Fig. 42.4). Consequently, The inaccuracy of MEG localization is smallest in the direc-
a single current dipole is a reasonably accurate description of a tion transverse to dipole orientation (cf. Fig. 42.1) and largest
local active cortical area of less than 2 to 3 cm in diameter. (about double) in the direction of depth. The locating accuracy
The adequacy of the model can be evaluated by calculating the depends drastically on the signal-to-noise ratio, which is
goodness-of-fit (g) value (5,45), that is the squared correlation improved by signal averaging for evoked responses but cannot
coefficient between the measured signals and those predicted by be affected much—except by filtering—in the case of sponta-
the model. The g-value, however, depends on several factors such neous activity. For comparison, note that the best localization
as the number and distribution of the measurement locations accuracy for EEG is in the direction along the dipole (Fig. 42.1).
(46). A low g-value means that either the brain source signifi- Since the dipoles mainly reflect synchronous activation of the
cantly deviates from the model or that the signal-to-noise ratio is cortical pyramidal cells and thus are perpendicular to the
870 Part V ■ Complementary and Special Techniques

the driving force for both intra- and extracellular current flow.
ECD calculated from the measured MEG signals reflects the
direction of the net intracellular current flow. A PSP at the end
of one dendrite produces an “elementary” dipole moment Q =
I (~10–14 nAm), where I is the intracellular current driven by
the synapse and is the length constant of the cell membrane
(48). The strength of the observed dipole moment can be
strongly affected by the simultaneous calcium currents.
Since it is not known how many PSPs occur synchronously
in each pyramidal cell and how much cancellation takes place
within the cortex, it is not possible to derive accurate estimates
for the source area of a typical evoked response on the basis of
cell/synapse density and the size of the elementary dipole.
Okada et al. (49) note that many animal species across a wide
phylogenetic scale (turtle, guinea pig, and swine) show an
apparent invariance in the maximum current dipole moment
density of 1 to 2 nAm/mm 2. Considerably smaller dipole
moment densities have been estimated on the basis of available
information on intracortical current densities and values of
cortical (50). However, intracortical current densities depend
strictly on the degree of neuronal synchrony, and thus on the
specimen studied, and may vary according to background
activity (51). Since is proportional to the square root of the
fiber diameter, the dipole moments weigh the larger neurons.
Glial cells may affect the MEG and EEG signals: they change
Figure 42.5 Schematic illustration of equivalent current dipole (ECD)
the return paths of volume currents and have activation latencies
locations when the source is (A) a layer of less than 2 cm in diameter,
that fit with the occurrence of long-latency-evoked responses.
(B) a layer with angular extension, and (C) a layer extended along the
Several important electrical events probably remain beyond the
radius of the sphere. The activity is assumed to occur in the wall of a
reach of both MEG and EEG measurements. For example, intra-
cortical fissure. This behavior depends slightly on the coil configuration
cortical short-distance interactions are—when viewed from the
used. (Adapted from Hari R. On brain’s magnetic responses to sensory
top of the cortex—radially symmetric; only their net effect along
stimuli. J Clin Neurophysiol. 1991;8:157–169.)
the dendrites of the pyramidal cells gives rise to an MEG signal.

MINIMUM-NORM ESTIMATES
cortical surface, changes in the focus of activation along the cor-
tex are more easily seen with MEG than EEG. On the other An example of a different approach to the neuromagnetic
hand, EEG may be more accurate in indicating which wall of a inverse problem is the minimum-norm estimate (MNE) pres-
fissure has been activated, since here the distinction must be entation of the source currents (52). MNE gives the most prob-
made along the direction of the dipole. able current distribution, in the sense of the minimum norm,
When a single current dipole is used to model an extended giving rise to the recorded magnetic field. Calculation of MNE
cortical area consisting of a layer of dipoles, the estimates of does not require specific assumptions about the source config-
dipole depth and, consequently, of dipole strengths may be uration (one dipole, multiple dipoles, quadrupoles, etc.), which
erroneous (Fig. 42.5). Better source models are thus needed for is an advantage when there is no basis for explicit models. On
extended areas. When multiple brain regions are simultane- the other hand, the unconstrained MNE solution favors super-
ously active, the relative contribution of each area to the meas- ficial currents and does not give a reliable estimate of the source
ured signal depends on its site, strength, and synchrony. depth. Therefore MNE solutions have been weighted with con-
Multidipole models with time-varying source strengths are use- straints based on the known brain anatomy and physiology,
ful in interpreting the resulting complex field patterns (47). especially source depth (53,54). Minimum-current estimates
(MCEs) (55), a subclass of MNE solutions that give more local
NEURAL CURRENTS UNDERLYING solutions, have been applied for visualization and analysis of
THE CURRENT DIPOLE single-subject and group-level MEG data. One example of MCE
visualization is given in Figure 42.6.
To understand the cellular events underlying the ECD, it is rea- The distributed MNE-based source activations often look
sonable to divide the currents associated with postsynaptic “more physiologic” than the point-like current dipoles, and
potentials (PSPs) to transmembrane currents at the active therefore current dipoles have been considered old-fashioned
synaptic area, intracellular currents within the neuron, and and less appropriate than the distributed models to describe the
extracellular “volume” currents. Transmembrane currents are complex brain activity. However, a word of caution has its place
Chapter 42 ■ Magnetoencephalography: Methods and Applications 871

Figure 42.6 Minimum current estimates of the 20-Hz band activity projected to the brain surface of a single subject before
(left) and after (middle) oral benzodiazepine administration; the brain is viewed from right. Coregistration of the sources
on the subject’s MRI on the right indicates activation of the primary motor cortex, with virtually identical source areas
before and after application of benzodiazepines. (Adapted from Jensen O, Goel P, Kopell N, et al. On the human motor-
cortex beta rhythm: sources and modelling. Neuroimage. 2005;26:347–355.)

here because the appearance of the result depends strongly on fMRI results may significantly differ in certain conditions (60),
the method used: the MNE approach will give a distributed and the MEG/EEG and fMRI/PET methods remain comple-
solution and the dipole approach a local solution, whatever the mentary in studies of human brain function. New approaches to
real current distribution is (see Ref. (16))! In general, both combine fMRI and MEG data have been introduced, and the
dipole and MCE solutions give rather similar results, although theoretical basis of the fusion is improving (see, e.g., Ref. (61)).
the temporal accuracy may be better with dipole modeling and
the group data may be easier to visualize with MCE; however, VOLUME CONDUCTOR MODELS
even well-informed users tend to report more false positives
with MCE than with current dipole modeling (56). The sphere model works well in most areas of the head when
the radius of the sphere is fitted to the local radius of curvature
COMBINATION OF MEG WITH of the measurement area (62), likely because the realistic noise
MRI/ fMRI/ PET DATA largely masks the errors caused by the different conductor mod-
els (63). Realistic head models that take more computing time
Functional information from MEG is at present routinely com- may be needed for proper modeling of the temporobasal and
bined with structural information from MRI. Moreover, all frontobasal brain areas (64). Fortunately, it is sufficient for
source estimates could be improved by constraints based on the MEG to model the intracranial space since only a relatively
known anatomy of the brain, derived from MRI data, and forc- small proportion of the currents flow in the poorly conducting
ing the dipoles/currents to the cortical tissue. In preoperative skull. In contrast, proper modeling of the EEG signals necessi-
evaluation, reconstruction of the three-dimensional outer tates a multicompartment model consisting of 3 to 4 concentric
surface of the brain from MRI scans helps the surgeon to recog- layers with known resistivities.
nize the landscape after opening the skull when the brain tissue Some nonspherical electric inhomogeneities may also affect
retracts and the relationship between the brain and the land- MEG distributions (65). For example, the falx cerebri may
marks on the skull changes (57). Adding surface vessels to the change volume current paths and lead to slight mislocalizations
reconstruction further facilitates the orientation during surgery of current dipoles in the mesial wall of the hemisphere. There
(58) (Fig. 42.30). has been discussion about the significance of holes in the skull
Compared with PET and fMRI (59), the advantage of MEG is to the distribution of volume currents. In the intact human
its good temporal resolution that allows monitoring of cortical skull, the main holes are in the base of the skull and in the
dynamics in millisecond scale. In combined use of multiple orbits. The absence of electro-oculographic artifacts in
methods, active brain regions have been first determined with intracranial recordings from the frontal lobe (66) suggest a
PET/fMRI and then used as source constrains in the inverse rather good isolation between the outer and the inner sides of
solution of the MEG data to reveal the corresponding temporal the skull. Therefore the effects of normal holes of the skull seem
behavior. However, not all changes in the synchronization of a negligible for the interpretation of MEG distributions, as is also
neuronal population, reflected in the MEG/EEG signals, induce supported by calculations showing that radial anisotropy added
significant changes in the mean neuronal firing rates or the to a spherically symmetric conductor does not affect the exter-
blood flow and metabolism, and vice versa. Therefore MEG and nal magnetic field (67).
872 Part V ■ Complementary and Special Techniques

PRACTICAL ISSUES least monitor, contaminated signals. Muscular artifacts cause


problems less frequently. Magnetic lung contamination or
Successful clinical MEG recordings need sophisticated instru- magnetic material in clothing can cause respiration-related slow
mentation and close interdisciplinary collaboration. The exper- shifts. Cardiac artifacts can be either magnetocardiographic sig-
imental situation introduces some restrictions. Since the head nals (69,70), picked up at distance, or ballistocardiographic fluc-
has to be immobile, recordings cannot be performed during tuations, caused by the movement of magnetized material at the
major motor seizures, and problems are encountered in studies rhythm of the cardiac cycle. For recognition of both artifacts,
of uncooperative subjects who either cannot keep still during simultaneously recorded ECG is essential: the magnetocardio-
the recording or who are not willing or able to perform the gram coincides with the ECG, whereas the main peak of the
tasks. However, continuous monitoring of the head position is magnetic ballistocardiogram is broader and lags the QRS com-
currently possible with some neuromagnetometers. Moreover, plex by several hundred milliseconds.
multichannel MEG mappings are quick to perform and the rel- All magnetic materials must be avoided in the clothing of the
ative locations of the sensors, required for accurate source subject. Although intraoral metallic devices for orthodontics or
analysis, are exactly known without extra effort. magnetized material used in staples and sutures during brain
Figure 42.7 illustrates some common MEG artifacts. Since surgery may contaminate the MEG measurement, recent devel-
significant contamination can be caused by eyeblinks and move- opment of signal-space separation (SSS) method and its tem-
ments (68), the electro-oculogram should be used to reject, or at poral extension (tSSS) help to remove artifacts in the
postprocessing phase (71–73). It is even possible to clean the
MEG data from artifacts elicited by deep brain stimulator (74),
as is illustrated in Figure 42.8.

Raw data
VERTEX FRONTAL
EOG OCCIPITAL

After tSSS-cleaning
VERTEX FRONTAL
EOG OCCIPITAL

Figure 42.7 Different artifacts in magnetoencephalogram (MEG)


recordings of spontaneous brain activity over the temporal area; the
measurements were made with a planar gradiometer. Blinking and ver-
tical eye movements produce signals of about equal size; for horizontal
eye movements the signals are similar in waveform but have different Figure 42.8 The effect of tSSS artifact suppression on spontaneous
spatial distributions. Muscular tension refers to biting the teeth magnetoencephalogram (MEG) of a person with deep brain stimulator.
together. Respiration artifacts were due to a metallic piece over the Top: Spontaneous MEG signals from the left hemisphere when the stim-
chest of the subject. The magnetic ballistocardiogram was due to move- ulator was on. The approximate locations of the planar gradiometers are
ment of magnetized metal on the bed, on which the subject was lying indicated; total duration 30 seconds. Bottom: After tSSS when even the
on his left side. Note the timing differences between the maximum sig- normal posterior alpha activity is visible. The amplitude scales are the
nals in the electrocardiogram (ECG) and the ballistocardiogram. The same in both panels. Courtesy of Jyrki Mäkelä, Helsinki University
lowest artifact was due to a digital watch 70 cm from the Dewar. Central Hospital. (See color insert)
Chapter 42 ■ Magnetoencephalography: Methods and Applications 873

52
Bandwidth 0.1–145 Hz
50 fT/cm right hemisphere
31
Left Hand
41
28
46

25
Left Lip
optic fiber switch
bundle
receiver 32
transmitter
reflectance emittance 18

skin trigger
–100 0 100 200 300 ms

Figure 42.9 Tactile stimulator made of a bundle of optical fibers and the somatosensory responses obtained by stimulating
the left hand and the left lip. The corresponding source areas in the right-hemisphere primary somatosensory cortex
and the current directions are indicated on the right (hand up, lip below). (Adapted from Jousmäki V, Nishitani N,
Hari R. Brush stimulator for functional brain imaging. Clin Neurophysiol. 2007;118:2620–2624.) (See color insert)

Special attention must also be paid to the proper design of Figure 42.10 draws attention to dangers of filtering. A too
stimulators. High-quality sounds can be produced with elec- high high-pass filter setting will cause well-known distortions
troacoustic transformers placed outside the shielded room and in the signal waveforms but—what may be less obvious—also
connected through plastic tubes to the subject; the system may transfer the dipolar field patterns, with opposite polarities, to
need sophisticated equalizing to guarantee flat frequency trans- latencies distant from the real signal and thereby lead to erro-
fer. Excessive artifacts from electric somatosensory stimuli can neous interpretations of brain activation.
be avoided by twisting the stimulator wires tightly together.
Multichannel devices with balloon diaphragms driven by com- ANALYSIS OF SPONTANEOUS ACTIVITY
pressed air (75) provide a nice and artifact-free method for tac-
tile stimulation. A hand-held bundle of optical fibers, half Very useful information about spontaneous activity (for a
emitting red light and the other half detecting the reflected light review, see Ref. (11)) can be obtained by calculating frequency
from the skin can be used to evoke reliable somatosensory spectra of the signals (Fig. 42.11) and by mapping the abun-
responses to taps to the skin (76); see Figure 42.9. dance of different frequencies at various sensor sites. The
Noxious laser heat stimuli for pain studies can be brought to sources can then be identified either in time or in frequency
the shielded room via optical cables. Visual stimuli can be trans- domain (77–79). In multichannel recordings, cross-spectra
mitted through mirrors, a data projector, a bundle of optic between channels suggest which signals arise from the same
fibers, or the subject can view a monitor through a hole in the source, and time lags may tell about the sequence of activation.
wall. EEG can be measured with nonmagnetic electrodes and One may quantify the level of different brain rhythms by
wires without causing problems to the MEG recordings. Eye using time–frequency representations, or more simply using the
tracking can be performed during the MEG recordings with an temporal spectral evolution (TSE) method (80) by focusing on
infrared camera. one frequency band at a time. The TSE method resembles the

Figure 42.10 Dangers of filtering,


illustrated with simulated data. A cur-
rent dipole with a monophasic activa-
tion curve (gray line on the left, “no
filter”) and with 30 nAm dipole
moment was inserted to a site corre-
sponding to the right auditory cortex.
The field patterns above show a dipolar
field pattern at peak activation at 100
msec. After high-pass filtering at 6 Hz
the waveform changes (black curve on
the left, “HP 6 Hz”) and ghost sources
appear at 60 and 150 msec.
874 Part V ■ Complementary and Special Techniques

activity, and the method works only if well-discernible spikes or


sharp waves are present. With multichannel low-noise instru-
ments, averaging is necessary only for the detection of prespikes,
which might be accurate indicators of the onset area of the
paroxysmal activity (83). With template matching, signals can
be automatically classified and the source locations determined
separately for each class.

SPONTANEOUS ACTIVITY OF
AWAKE NORMAL SUBJECTS
Alpha Rhythm from Parieto-Occipital Areas
The typical parieto-occipital alpha rhythm, with a peak fre-
quency around 10 Hz, is damped by opening the eyes
(Fig. 42.11). In the first study of the human magnetic alpha
rhythm (1), a phase reversal was observed between signals meas-
ured from the right and left hemispheres, and the currents were
therefore suggested to be parallel to the longitudinal fissure.
Later, the sources of the alpha rhythm have been suggested
to cluster mainly in the region of the calcarine sulcus (84) and
around the parieto-occipital sulcus (POS) (85–88). Figure
42.12, based on whole-scalp MEG recordings, shows sources in
both of these locations, typically the POS region is the far most
dominant source of the MEG alpha rhythm.
Vvedensky et al. (85) suggested that alpha spindles often
Figure 42.11 Amplitude spectra from a 1-minute period of spontaneous have the same generators for about 1 second, whereas the
activity when the subject was resting with eyes closed. Two orthogonal sources differ for successive spindles. Later, a multitude of MEG
derivatives of the radial magnetic field were measured at each location, alpha sources were found with the help of “signal-space analy-
along the longitude and latitude. The insets show reactivity of the - and sis” (89). Different spindles seemed to have different sources
-rhythms to opening of the eyes and to movements of the left and right but the source configuration of one spindle was rather stable.
hand. (Adapted from Salmelin R, Hari R. Characterization of sponta- Later recordings, analyzed with time-varying fixed-location
neous magnetoencephalogram (MEG) rhythms in healthy adults. dipole models, however, suggest that a typical alpha spindle
Electroenceph Clin Neurophysiol. 1994;91:237–248.) cannot be explained by a single source (88).
The parieto-occipital alpha rhythm is strongly suppressed
during visual stimuli, visual memory tasks, and visual imagery
(90–93). When the subject has to differentiate between visually
“event-related desynchronization” technique (81) used to study
presented images of objects vs. nonobjects, the poststimulus
task-dependent changes in the human scalp EEG. However, TSE
alpha level is consistently higher after nonobjects (94). This
preserves the original units and the signal levels can thus be
directly compared with the sizes of evoked responses. In TSE,
the brain signals are first bandpass filtered, then rectified (tak-
ing absolute values of the signals), and thereafter averaged with
respect to the triggering event. Thus, all signals that are time-
locked but not exactly phase-locked to the event will be
detected.
Sometimes one may be interested in just seeing the signal
waveform and the temporal changes in the field pattern. This
type of analysis, which resembles the classical use of EEG
recordings, can be helpful in screening candidates for surgical
treatment: clear changes of the field pattern from one irritative
phenomenon to the next discourage the assumption of a local
onset of the discharge, although they do not rule out a deeper
common trigger.
Due to noise problems and the use of single-channel instru- Figure 42.12 Sources of alpha oscillations superimposed on two sagit-
ments, the first studies of epileptic MEG activity employed tal MRI slices demonstrating a calcarine (left) and a parieto-occipital
averaging, with the simultaneously recorded EEG signal as a (right) source cluster. Source current orientations are also indicated.
trigger (32,82). The drawback of such a procedure is that the (Adapted from Hari R, Salmelin R, Mäkelä JP, et al. Magnetoence -
choice of the trigger channel largely determines the observed phalographic cortical rhythms. Int J Psychophysiol. 1997;26:51–62.)
Chapter 42 ■ Magnetoencephalography: Methods and Applications 875

modulation may reflect interaction between the dorsal and


ventral visual pathways during an attention-demanding task.
The level of parietal alpha activity has been related to memory
load (95).
Early visual deprivation may lead to the abolishment of
parieto-occipital spontaneous rhythms: from the five early
blind subjects in Figure 42.13, four do not display alpha activ-
ity and opening/closing the eyes did not have any effect on
their brain rhythms. Only one subject (B5) has rhythmic
alpha-like activity; she had lost her sight earlier than the
other four subjects. The reason for the observed alpha
absence in blinds, also known from EEG recordings (96),
is unknown, but it certainly speaks against a purely idling
nature of the normal alpha rhythm; otherwise one might
expect strong alpha during the continuous lack of
visual input.

Mu Rhythm from Sensorimotor Areas


The electroencephalographic -rhythm has a magnetic coun-
terpart that starts to dampen 1 to 2 seconds before a movement
(Fig. 42.14). In association with unilateral movements, the sup-
pression is bilateral, although contralaterally dominant (97).
The -rhythm consists of two main frequency components,
one around 10 Hz and the other around 20 Hz (see inset in
Fig. 42.11); however, these two frequencies are usually not exact
harmonics and show independent temporal behaviors (98).
Additional evidence for functionally separate frequency compo-
nents of the -rhythm derives from the source locations, which
center on average 5 mm more anterior for the 20 Hz than the
10 Hz frequencies (80), suggesting the existence of separate pre-
central (20 Hz) and a postcentral (10 Hz) rhythms (Fig. 42.15).
In addition to movements (99), the magnetic -rhythm is
modified by electric stimulation of peripheral nerves, with a
Figure 42.13 Traces of parieto-occipital alpha activity from five early
more clear increase, “rebound” of the 20-Hz than the 10-Hz
blind subjects and seven control subjects. The blind subjects were 25 to
component around 400 msec after the stimulus (80,97). The
32 years in age. Subjects B2 and B3 were able to see some light; subject
poststimulus rebound is abolished when the subject moves the
B5 is blind due to retinal cancer and has seen light only during her first
fingers of the same hand. Interestingly, a similar, but weaker,
year. M. Huotilainen and T. Kujala participated in data collection.

Figure 42.14 Spontaneous magnetic activity over the rolandic area, shown on six gradiometer channels, when the subject
has his eyes open and clenches his contralateral fist (see the electromyogram [EMG] channel). (Unpublished data from
F. Lado and R. Hari.)
876 Part V ■ Complementary and Special Techniques

The 20-Hz rebounds do—whereas the 10-Hz rebounds do


not—follow the moved body part in a somatotopic manner:
they appear at lateral rolandic areas after mouth movements,
more medially after finger movements, and close to midline
after foot movements (102).
Recent MEG recordings after oral administration of benzo-
diazepine demonstrated a strong increase in the power of
approximately 20-Hz oscillations in both hemispheres, with
sources in the primary motor cortex close to the hand area
(103) (Fig. 42.6). These data suggest that the motor cortex is
an important effector site of benzodiazepine, and they also
agree with the proposed generation of the rolandic 20-Hz
oscillations in the motor cortex. The frontal-lobe dominance
of benzodiazepine-related scalp-EEG beta activity can be
explained by tangential current dipoles in the wall of the
central sulcus.
Oscillatory Cortex–Muscle Coupling
Figure 42.15 Top: A 5-second trace of magnetic -rhythm from the The human rolandic MEG activity has been observed to have
rolandic region. Bottom: Isodensity clusters of sources of 10- and 20-Hz a close temporal relationship to peripheral muscular activity
oscillations over the rolandic region of one subject, based on thousands of (54,104–106). For example, during isometric contraction of
equivalent current dipole (ECD) locations. The black ovals show the area different muscles, the muscular and cortical signals are coher-
activated by median nerve stimulation at the wrist, and the line shows the ent at frequencies varying between 15 and 33 Hz in individ-
estimated course of the rolandic fissure. (Adapted from Salmelin R, Hari ual subjects (106). The sites of maximum coherence in the
R. Spatiotemporal characteristics of rhythmic neuromagnetic activity motor cortex show gross somatotopic organization, with
related to thumb movement. Neuroscience. 1994;60:537–550.) activations during foot muscle contraction closer to the head
midline than during hand muscle contractions (Fig. 42.17).
suppression is seen when the subject just imagines making the
movements, indicating involvement of the primary motor cortex
in motor imagery (100). Suppression of the rebound, as a sign of
motor–cortex activation, also occurs when the subject just views
another person to make certain movements (Fig. 42.16) (101).

Figure 42.16 Level of the 20-Hz rolandic activity after stimulation of Figure 42.17 Top: Surface electromyogram (EMG) from isometrically
the contralateral median nerve when the subject either rested (solid contracted left hallucis brevis muscle and simultaneously recorded
lines), viewed another person to manipulate a small object with the magnetoencephalogram (MEG) signal over the parietal midline (3 to
right hand fingers (dashed line; “viewing”), or manipulated the same 100 Hz). Middle: Coherence spectra between MEG and EMG during iso-
object herself (“acting”). The sources of these 20-Hz signals were in the metric contraction of small hand and foot muscles (left- and right-sided
precentral primary motor cortex. (Adapted from Hari R, Forss N, contractions superimposed). Bottom: Spatial distributions of the
Avikainen S, et al. Activation of human primary motor cortex during strongest peaks of the coherence spectra. (Adapted from Salenius S,
action observation: a neuromagnetic study. Proc Natl Acad Sci USA. Portin K, Kajola M, et al. Cortical control of human motoneuron firing
1998;95:15061–15065.) during isometric contraction. J Neurophysiol. 1997;77:3401–3405.)
Chapter 42 ■ Magnetoencephalography: Methods and Applications 877

The MEG signals lead the EMG signals in time, with increas-
ing time lags with increasing brain–muscle distance. Such
data strongly suggest that the 20-Hz rolandic rhythm reflects,
at the population level, the common central drive to spinal
motoneurons (for reviews, see Refs. (107,108)). The
corresponding cortex–muscle coherence can also be shown
with EEG recordings (109).
Studies with different gripping tasks have led to the proposal
that the cortical oscillations have a role in recalibrating the control
system after a change in the cortex–muscle relationship (110,111).
At strong contractions, the frequency of the coherence jumps
from 20 to 40 Hz (“Piper rhythm”; (112,113)). The cortex–muscle
coherence provides a complementary tool to identify the primary
motor cortex as a part of preoperative functional mapping (58).
Gross et al. (114) demonstrated, using a sophisticated
dynamic imaging of coherent sources (DICS) method, developed
Figure 42.18 Locations of equivalent current dipoles for the tau oscil-
to characterize synchronously firing neural networks in different
lations (6.5 to 9.5 Hz range) in two subjects. The sources (clusters of
parts of the brain, that 6- to 9-Hz velocity changes of slow finger
white dots) are projected to the surface of the brain and shown on the
movements are directly correlated to oscillatory activity of the
subject’s own MRI. The black dot indicates the source of the auditory
primary motor cortex. Moreover, the coherence patterns sug-
N100m response. In the coronal sections in the middle, the dipole dis-
gested that the pulsatile velocity changes were sustained by a cere-
tributions are presented as contour plots, with highest dipole densities
bellar drive through thalamus and premotor cortex.
in white and lowest in black. (Adapted from Lehtelä L, Salmelin R, Hari
Tau Rhythm from Auditory Areas R. Evidence for reactive magnetic 10-Hz rhythm in the human auditory
cortex. Neurosci Lett. 1997;222:111–114.)
A third MEG rhythm, which I have started to call -rhythm
( referring to temporal lobe), was described by Tiihonen et al.
(115). This 8- to 10-Hz oscillatory activity is best seen in the
planar gradiometer recordings just over the auditory cortex. the occipital alpha and from the temporal tau generators.
Occasionally the rhythm is reduced by sound stimuli, such as Epidural electrode recordings (120,121) show that also the tem-
bursts of noise, but it is not dampened by opening the eyes—a poral-lobe 6- to 11-Hz activity arising from the convexial cortex
feature clearly distinct from the occipital alpha. The sources of is strikingly resilient to decreased vigilance.
single-tau oscillations cluster to the supratemporal auditory
cortex close to the generations site of auditory-evoked fields, A Note on Nomenclature
with right-hemisphere dominance (Fig. 42.18) (116). The existence of local cortical rhythms explains in part the
On the basis of the observed current orientations, the corre- confusion in the literature concerning effects of various tasks
sponding electric -rhythm should be seen mostly in the fron- on the spontaneous EEG. Some people consider alpha to refer
tocentral midline where the tau sources, with dipole moments to the occipital “Berger rhythm,” independently of its shape
of 40 nAm, would lead to potentials of about 10 to 20 V. In (i.e., frequency composition), whereas others refer to alpha as
fact, such an EEG rhythm may appear during drowsiness: all signals falling in the 8- to 13-Hz frequency range,
Decreased vigilance is often considered to be associated with a independently of the brain area where they are generated.
“spread” of occipital EEG alpha toward more anterior regions, To avoid confusion, it would be necessary to characterize
with simultaneously slightly decreasing frequency (117,118). A each rhythm both by its site of origin and by its frequency
real spread of the alpha would be in clear contrast to the fixed, range.
although distributed, generators of alpha activity, and a more
plausible explanation is that the anterior “alpha” in fact reflects Other Brain Rhythms
“tau” generated in the supratemporal auditory cortex. A 7- to 9-Hz “sigma” rhythm has been demonstrated in MEG
The combined MEG and EEG data of Lu et al. (119) would recordings in the second somatosensory cortex (122). Single
agree with such a hypothesis: During the awake state (the first median-nerve stimuli elicited some sigma oscillations, and the
panel of Fig. 42.19) the occipital EEG displays rhythmic 10 to level of sigma was enhanced during rhythmic stimulation at
11 Hz alpha activity, and the simultaneous MEG in the tempo- the rhythm’s dominant frequency.
ral region is of low amplitude, with little rhythmic activity. Theta-range MEG activity has been scarcely studied. Sasaki
However, when the occipital electric alpha becomes discontin- et al. (123) recorded 5- to 7-Hz MEG signals from the frontal
uous during light drowsiness (Stage 1a) and seems to spread cortex during mental calculation and intensive thinking. Tesche
more anteriorly, with slightly slowed down frequency, rhythmic (30) estimated the temporal waveforms of sources (computa-
activity of the same frequency appears in the MEG sensors over tionally) inserted to the hippocampus and found in some sub-
the temporal lobe. jects around 5-Hz rhythmic activity during mental calculation.
Therefore, the apparent spread of the EEG “alpha” during Since 1980s, much attention has been paid to the 40-Hz fre-
drowsiness might reflect changed relative contributions from quency band, often supposed to have a role in perceptual binding
878 Part V ■ Complementary and Special Techniques

Figure 42.19 Different vigilance stages in one subject. The recording locations (24-channel planar gradiometer) are indi-
cated on the schematic head; the x-gradients at 12 locations are plotted above and the y-gradients below. In the electric
channels EOGv and EOGh refer to vertical and horizontal oculograms, respectively. The electric amplitude scale refers to
all other electric recordings except the electromyogram (EMG) for which only relative amplitudes are of importance. Note
the increase of rhythmic magnetoencephalographic activity during Stage 1a, compared with the awake stage. A typical V-
wave occurs during Stage 1b. During Stage 2, light sleep, K-complexes occur both in MEG and in electroencephalogram
(EEG). In deep sleep, Stages 3 and 4, slow activity is seen in the whole measurement area. Note the eye movements and
the decreased EMG activity during the REM stage. (Adapted from Lu ST, Kajola M, Joutsiniemi SL, et al. Generator sites of
spontaneous MEG activity during sleep. Electroenceph Clin Neurophysiol. 1992;82:182–196.)

(124,125). Both stimulus-related and later “induced” 40-Hz how strongly the subcortical structures contribute to the MEG
activity has been reported, with differences between visual EEG signals that are biased toward cortical currents.
and MEG 40-Hz signals (53,126). Even higher MEG frequencies
(from 30 to 150 Hz) have been recently used to demonstrate dis-
sociation between visual awareness and spatial attention (127). Spontaneous Activity during Sleep
Although the 40-Hz activity, according to animal experiments, In the first MEG studies of sleep, performed with single-chan-
results from thalamocortical interaction, it is at present unknown nel MEG devices, it was difficult to draw conclusions about the
Chapter 42 ■ Magnetoencephalography: Methods and Applications 879

Figure 42.20 Two K-complexes from


the same subject, recorded over the
same location. In the magnetic sig-
nals, the upper traces of each pair
indicate the vertical gradient and the
lower trace the horizontal gradient of
the magnetic field; the recording
locations are shown on the inset
head. Simultaneous electric signals
from the midline are shown in the
insets; their amplitude calibration is
100 V. (Adapted from Lu ST, Kajola
M, Joutsiniemi SL, et al. Generator
sites of spontaneous MEG activity
during sleep. Electroenceph Clin
Neurophysiol. 1992;82:182–196.)

relationship between the electric and the magnetic spontaneous do not appear to be stereotyped responses of the cortex to exter-
signals (128–130). nal or internal stimuli, comparable to evoked responses, but
Figure 42.19 illustrates the first multichannel MEG record- they rather seem to reflect a diffuse and variable reaction
ing of sleep where it was possible to classify the different stages involving large cortical areas.
of vigilance on the basis of both EEG and MEG (119). The Up to now, no focal activation spots have been detected
activity during awake stage and the appearance of -rhythm during slow-wave sleep. The sources underlying the spin-
during Stage 1a were discussed above. During deep drowsiness dles also seem very complex. In principal component analy-
and light sleep (Stages 1b and 2), vertex waves of 150 to sis, four to six separate components were often needed to
250 msec duration appeared in the frontocentral EEG leads and explain at least 90% of the field variance during 1 second of
on several MEG channels. The electric and magnetic V-waves the spindle activity (119). Complexity of spindle generation,
did not always coincide. with a multitude of sources and frequencies, has also been
Magnetic spindles of 11 to 15 Hz in frequency and 0.3 to suggested in animal experiments (133), in human depth elec-
2 seconds in duration appeared during Stage 2. A later study trode and scalp topography studies (134,135), as well as in
has shown that the sources of the spindles differ from the recent MEG recordings applying synthetic aperture magne-
sources of wake-state oscillatory activity (131). The spindles tometry (136).
were occasionally superimposed on high-amplitude tran- Llinas and Ribary (137) point out that while the spontaneous
sients, thereby resembling the typical electric K-complexes. activity is rather similar during awake and REM sleep stages,
Often the successive magnetic V-waves and K-complexes dif- responses to repetitive 40-Hz stimulation are dampened during
fered in waveform and distribution. For example, the midline REM sleep compared with wakefulness, possibly reflecting the
distributions of two electric K-complexes of Figure 42.20 brain’s isolation from the external world during the REM stage.
resemble each other, whereas the simultaneous magnetic dis-
tributions differ. A typical magnetic K-complex lasted 0.8 to Anesthesia
1.2 seconds. Burst-suppression patterns have been recorded from a dog dur-
During slow-wave sleep (Stages 3 and 4; Fig. 42.19), 0.5 to ing enflurane and propofol anesthesia (138). The signal pattern
2 Hz polymorphic MEG activity was widely spread over the during the bursts was complex and the signal did not show any
measurement area. During the REM stage, defined on the basis organized structure (Fig. 42.21). A practical message from these
of rapid eye movements and decreased submental muscular recordings is that an essentially artifact-free MEG measurement
tone, MEG activity was lower in amplitude and faster in can be obtained during respirator-assisted anesthesia when the
frequency than during the other sleep stages. Sharp magnetic respirator is kept outside the magnetically shielded room and
transients, resembling the V-waves, were frequently seen. when special care is taken in choosing nonmagnetic tubings
In the first whole-scalp MEG study of sleep (132), the and valves.
sources of K-complexes in the two hemispheres were active
independently. Only in one subject out of six were the K- MEG IN EPILEPSY
complex distributions satisfactorily explained by two current
dipoles, one in each inferior parietal lobe, that is, in line with The first MEG recording of an epileptic individual was reported
the model suggested by Lu et al. (119). In five other subjects the by Cohen (139): slow EEG and MEG activity increased during
distributions were more complex. Therefore the K-complexes hyperventilation in a patient with psychomotor epilepsy.
880 Part V ■ Complementary and Special Techniques

Figure 42.21 Four magnetoencephalogram (MEG) channels and one


electroencephalogram (EEG) channel from a dog during burst-suppres-
sion phase induced by enflurane anesthesia. (Adapted from Jäntti V,
Baer G, Yli-Hankala A, et al. MEG burst suppression in an anaesthetized
dog. Acta Anaesth Scand. 1995;39:126–128.)

Starting from the early 1980s, a number of epileptic subjects


have been studied in several laboratories (4,32,82,140–157). In
November 2003, the PubMed database already contained 400
publications with keywords of “MEG/magnetoencephalogra-
phy/neuromagnetic and epilepsy,” and thereafter more than one
study per week has appeared in this field. Therefore, it is impos-
sible to cover here the progress in this rapidly expanding field.
The earliest MEG recordings were used to locate both single
and multiple irritative brain areas (32,82,140). In one study,
both scalp and sphenoidal EEG spikes were used as triggers for Figure 42.22 Top: Spatial distribution of one multispike displayed on
averaging the magnetic spikes. The latter had different latencies the 122-channel sensor array viewed from top. Bottom left: Field pat-
for the two triggers, consistent with the propagation of the tern during the spike (asterisk above the curves); the sensor array is
epileptic activity (142). The sources of the MEG spikes were viewed from left, top, and right. The shadowed areas indicate magnetic
deeper for sphenoidal than for scalp triggers in all five subjects flux emerging from the head; the isocontours are separated by 400
studied, suggesting propagation of the discharge between dif- fT/ cm. The arrows indicate the sites and orientations of the two ECDs
ferent parts of the temporal lobe. required to account for the field pattern. Bottom right: Dipole moments
Differentiation between ictal and interictal spikes is as diffi- as a function of time in both hemispheres. Each trace corresponds to
cult in MEG as it is in EEG. Since MEG recordings can only one unaveraged spike, whose distribution was explained by the two-
infrequently be performed during motor seizures, the relevance dipole model. (Adapted from Hari R, Ahonen A, Forss N, et al. Parietal
of the interictal MEG focus has been questioned. Sometimes epileptic mirror focus detected with a whole-head neuromagnetometer.
the activity just preceding the seizure, assumed to have the best Neuroreport. 1993;5:45–48.)
localizing value, can be detected. One possibility to reveal the
significance of the observed focus is to tailor an electrode array
on the basis of the interictal MEG distribution, by calculating speech areas, and whether the source configuration is stable over
the forward potential solution from the identified sources, and time. MEG seems especially suitable for identifying epileptic foci
then to monitor the ictal activity during telemetric EEG record- in the convexial neocortex. MEG results have been also used to
ings to ascertain whether the interictal focus is responsible for guide placement of intracranial electrodes and to suggest re-eval-
triggering the seizure. uation of structural MRI data.
In preoperative evaluation of epileptic patients it is important Two early examples of epileptic MEG discharges are presented
to know whether the epileptic discharges are focal, whether there below. Figure 42.22 shows spikes, field patterns, and dipole
are multiple foci, and whether these foci show any systematic moments of an 18-year-old female who suffered from complex
time lags. One should also find how close the foci are to function- partial epilepsy (158). During night her 20- to 30-second
ally irretrievable brain regions, such as the sensorimotor and seizures included awakening and bilateral increase of muscular
Chapter 42 ■ Magnetoencephalography: Methods and Applications 881

Figure 42.23 Epileptic discharges after


voluntary triggering. The top trace illustrates
magnetoencephalographic activity from the
right hemisphere during the whole 14-second
seizure. In the middle and lower parts,
selected periods (A–C) are expanded and sig-
nals from the corresponding area in the left
hemisphere are shown for comparison.
(Adapted from Forss N, Mäkelä JP, Keränen T,
et al. Trigeminally triggered epileptic facial
convulsions. Neuroreport. 1995;6:918–920.)

tone, as well as tonic jerks in left extremities. During daytime she SPONTANEOUS ACTIVITY
had epochs of paresthesia and loosening of the grip in the left IN OTHER PATIENTS
hand. The abundant spikes, recorded with a 122-channel
whole-scalp magnetometer, could be adequately modeled with Ischemic Lesions
two current dipoles, one in each posterior parietal cortex. The Ischemic hemispheric lesions are associated with slow magnetic
peak activity occurred consistently 20 msec later in the left than shifts and with considerable slowing of cortical activity (167).
the right hemisphere, suggesting callosal conduction of the sig- Gallen et al. (168) showed that 2- to 3-Hz MEG activity was
nals from right to left. Such time lags are useful in differentiating mainly concentrated in the damaged but viable tissue at the
the primary and the secondary (mirror) foci. periphery of the infarcted area. It is not known, though, to
Figure 42.23 illustrates an ictal MEG recording of a man which extent the changed tissue conductivities at the lesion area
who suffered from left-sided hemifacial convulsions that were contribute to the measured signals.
often triggered by touching the left mouth region (159). Local unilateral lesions in the left anteromedial thalamus
Computerized tomography and PET revealed no abnormali- caused extensive and surprisingly symmetric changes in the
ties. The MEG recording displays abundant epileptic spikes parieto-occipital spontaneous activity: the frequency spectra
that become increasingly polyphasic during the 14-second were broadened in the parieto-occipital area, but the reactivity
seizure. In the beginning, spikes appear only in the right hemi- to eye opening was preserved and the generation sites of the
sphere, with a source in the motor face area (defined on the activity were similar as in normal subjects (169,170).
basis of evoked responses and anatomical structures), whereas
later during the seizure spikes also appear in the homolog area Infantile Neuronal Ceroid Lipofuscinosis
in the left hemisphere, again (as in Fig. 42.24) with about 20-
msec time lag compared with the other side. As expected, patients with infantile neuronal ceroid lipofusci-
In patients with Landau–Kleffner syndrome (LKS), stereo- nosis (INCL) have flat MEG, pointing toward a total absence of
typic 3 c/s spike-and-wave complexes have been shown to be cortical activity (171). Brains of INCL patients show almost total
generated in the auditory cortex (160–163), and the continuous neuronal loss and strong gliosis. Therefore, the flat MEG from
epileptic discharges in the auditory cortices of LKS children these patients also indicates that glial cells have negligible contri-
prevent the proper analysis of sounds, including speech. bution to the MEG signals in the absence of neuronal activity.
Many groups are currently using MEG in preoperative
evaluation of epileptic patients, but the discussion about the Migraine
benefits of the procedure continues in quite a heated manner Spreading depression (SD) has been suggested as the neuro-
(164–166). physiologic basis of migraine aura. In turtle cerebellum,
882 Part V ■ Complementary and Special Techniques

Figure 42.24 Effects of electroconvulsive ther-


apy on rolandic magnetoencephalographic sig-
nals and their frequency spectra. (A) Before
treatment, (B) after four treatments, (C) after
eight treatments, and (D) 1 month after the last
treatment. On the right, the before-treatment (A)
spectrum is shown for comparison in all other
conditions ( dashed lin es). (Adapted from
Salmelin R, Mäkelä JP, Heikman P, et al. Human
brain rhythms and electroconvulsive therapy. Soc
Neurosci Abstr. 1996;22(pt 1):342.)

induced SD is associated with strong magnetic fields (172). who were withdrawn from medication from the preceding
Consequently, it has been proposed that SD propagating in the evening; the coherence was, to a large part, restored with the
human brain during the prodromal phase of migraine could patients’ normal levodopa treatment. Abolishment of the nor-
be detected by MEG and such shifts have been reported during mal 15- to 30-Hz synchronized oscillatory activity has been
the migraine aura (173). The propagation of SD, assumed to be proposed to lead in bradykinesia (179).
about 3 mm/min on the basis of changes in the scotoma in the In a more extensive analysis with the DICS method,
visual field, has been elegantly demonstrated in fMRI record- Timmermann et al. (180) demonstrated coherence between 4
ings (174). and 6-Hz Parkinsonian tremor and activity of several cerebral
areas. Coherence was seen both at the tremor frequency and,
Effects of Electroconvulsive Therapy even more strongly, at its double. The analysis of partial coher-
Electroconvulsive therapy (ECT), efficient in treatment of ence and phase shifts was especially interesting: The interaction
severe depression, is known to increase slow EEG rhythms. between M1 and EMG agreed with previous studies, indicating
Heikman et al. (175) tracked the temporal and spatial modula- that cortex leads the muscle, whereas the interaction between
tion of spontaneous oscillations in five ECT-treated depressive EMG and diencephalic areas was bidirectional. Moreover, a
patients. ECT resulted in a prominent increase of frequencies direct afferent coupling was evident from EMG to second
below 8 Hz, which largely disappeared within 1 month after the somatosensory cortex and to posterior parietal cortex.
last treatment. In three patients, relief of depression was accom- Cerebellum and premotor areas seemed to be connected with
panied by an increase of the 4- to 8-Hz oscillations by at least the periphery via other cerebral areas. Similar widespread cor-
50% after the first four treatments and up to 500% after eight tical circuitries seem to support postural tremor accentuated in
treatments, particularly in the temporal and rolandic areas. hepatic encephalopathy (181).

Encephalitis Auditory-Evoked Fields


In herpes simplex encephalitis, sources of abnormal MEG The first demonstrations of the generation of the main
waveforms have to be identified in the temporal lobes (176). auditory-evoked field (AEF) deflections in the superior surface
Further similar studies might help in the evaluation of the sites of the temporal lobe (182,183), deduced from the measured
and extent of the functionally disturbed brain areas in posten- field patterns, considerably enlightened the origin of the coin-
cephalitic patients. ciding EEG evoked potentials that had been previously believed
to be “nonspecific” due to their maximum amplitude at the ver-
Parkinsonian Tremor tex. Consequently, various long-latency EEG and MEG
Volkmann et al. (54) found strong coherence, at the frequency responses are used nowadays widely as tools to study functions
of the 3- to 6-Hz resting parkinsonian tremor, between oscilla- of the supratemporal auditory cortex.
tory MEG activity in premotor and somatomotor cortices and
muscle activity (54). Previously, Mäkelä et al. (177) had noted Transient Responses
that in patients with hemiparkinsonism the onset of tremor Figure 42.25 shows examples of middle-latency and long-
induces a similar dampening of the 10- and 20-Hz components latency auditory-evoked magnetic fields, obtained using differ-
of the -rhythm as do normal movements. It was thus sug- ent stimulus repetition rates, different filter settings, different
gested that Parkinsonian tremor activates cortical mechanisms analysis periods, and different number of averaged responses.
that normally produce rapid alternating movements. After the earliest cortical auditory responses peaking around 19
Salenius et al. (178) observed suppression of the normal to 20 msec, or even around 11 msec according to some reports
15- to 30-Hz cortex–muscle coherence in Parkinsonian patients (184), the interindividually variable but otherwise reliable
Chapter 42 ■ Magnetoencephalography: Methods and Applications 883

Figure 42.25 Example of middle-latency (left) and long-latency (right) auditory-evoked magnetic fields recorded from
one subject with a 122-channel device. (Adapted from McEvoy L, Mäkelä JP, Hämäläinen M, et al. Effect of interaural time
differences on middle-latency and late auditory evoked magnetic fields. Hear Res. 1994;78:249–257.)

middle-latency responses peak around 30 msec (185–188) and tested for example by changes in the interaural time difference
are followed by longer-latency responses around 50, 100, and of binaural click trains (205,206).
200 msec (P50m, N100m, and P200m, respectively). A sus- The reactivity of the auditory cortex to physical changes in the
tained field is seen during long sounds. All these responses have stimuli in an otherwise monotonous sequence has been studied
cortical origin and their generation sites differ slightly, indicat- extensively (for reviews, see Refs. (6,50,207–210)). Infrequent
ing contribution from several cytoarchitectonic areas in the deviations in a monotonous sound sequence evoke “mismatch
supratemporal auditory cortex. Also hippocampal activity may fields” (MMFs) in the close vicinity of the generation site of the
be seen during an auditory detection task (189). The clear auditory N100m, often with right-hemisphere dominance. MMF,
dependence of the auditory N100m response on the interstim- related to automatic sound processing, can be used in testing of
ulus interval (ISI), with saturation at ISIs of 4 to 8 seconds auditory cortical functions, including the duration of echoic
(190,191), has been related to the duration of sensory memory memory (194). A total omission of a tone from a monotonous
(192–194). sequence activates several brain areas, including the supratempo-
Different ECD locations for N100m depending on the fre- ral auditory cortex, the superior temporal sulcus and the frontal
quency and amplitude of the sound have been suggested to lobe; all with right-hemisphere dominance (211).
reflect tonotopic and amplitopic organization of the auditory The AEFs with known sources are good tools to study brain
cortex ((195,196); see, however, the critical evaluation by function. For example, Sams et al. (212) showed, applying the
Lütkenhöner et al. (197)). The auditory cortex is very sensitive so-called McGurk illusion related to speech-sound perception,
to amplitude and frequency modulations (198) as well as to var- that articulation movements seen from a videotape have an
ious changes within the stimuli (199,200). Direction of atten- effect on AEFs evoked by acoustical syllables. It was thus
tion can modulate activity of at least two areas in the demonstrated that visual information from lip movements can
supratemporal cortex (201–204). The auditory cortex also modify activity of the auditory cortex, a finding supported by
reacts to small changes in the direction of binaural stimuli, later fMRI recordings (213).
884 Part V ■ Complementary and Special Techniques

Steady-State Responses In deaf patients with cochlear prosthesis, AEFs have been used
Tonotopic organization of the human auditory cortex was first to show that the auditory cortex reacts to very artificial input
shown by steady-state recordings (214). The amplitude of the (227–230). AEFs also provide a tool to study the reorganization
auditory steady-state response reaches its maximum at repeti- of central auditory pathways after various peripheral lesions or
tion rates around 40 Hz. This so-called 40-Hz response (215) congenital disorders (231–233).
has a cortical origin (216,217). (To avoid confusion, it is impor- MMFs are very sensitive indicators of different sensory and
tant to emphasize that the notion “cortical origin” refers to the brain disorders at group level (for a review, see Ref. (209)) but,
major contributor to the measured signal. It does not, by any unfortunately, the specificity of the responses has not yet been
means, deny the possibility of a subcortical trigger or simulta- rigorously addressed and thus the clinical usefulness of MMFs
neous activity in some subcortical areas. However, in many at single-subject level remains to be shown. For example, MMFs
cases the adequate explanation of the signal patterns by cortical are altered in patients with thalamic infarction, but without
sources indicates negligible contribution from subcortical areas clear relationship to the degree of memory deficit (170).
to the measured MEG/EEG signals.) Patients with Alzheimer disease have dampened cortical
The amplitude enhancement of the steady-state response of 40-Hz responses and delayed ipsilateral N100m responses, prob-
around 40 Hz has been adequately explained by the summation ably as signs of deteriorated cortical processing (53,234). The
of subsequent single responses, each with a 40-Hz content, N100m responses were delayed and dampened during auditory
thereby suggesting linearity of the responses, at least at stimula- hallucinations in schizophrenic patients, most likely indicating
tion rates from 10 to 40 Hz (218,219). In other words, the ampli- top-down activation of the auditory projection cortices (235).
tude enhancement does not indicate stronger reactivity or Patients with tinnitus may have a significantly decreased
“resonance” of the auditory cortex at stimulation rates of 40 Hz. amplitude ratio between P200m and N100m (236). In tinnitus
However, PET data, based on quantifying regional cerebral due to acute noise trauma, the P200m/N100m amplitude ratio
blood flow during repetitive auditory stimulation, suggest that can normalize during the period of recovery from tinnitus
the 40-Hz response enhancement reflects increased cortical (237). However, many contradictory findings have been pre-
synaptic activity (220); further studies are required because the sented as well (238,239), most likely due to different types of
behavior of response strengths as a function of stimulus repeti- patients studied and due to variations in the experimental
tion rate differed between EEG and PET recordings. setups and filter settings. In support of the early tinnitus stud-
Stimulus-related 40-Hz oscillations, elicited by paired clicks, ies, Shiomi et al. (240) showed that lidocaine-injection-induced
have been related to temporal binding (221). tinnitus release is associated with narrowing of the N100m
During normal binaural hearing, sounds from each ear reach deflection; such a change could reflect tinnitus-related modula-
the auditory cortices of both hemispheres. Therefore the binau- tion of neuronal mechanisms underlying the P200m response.
ral cortical responses, even when recorded from one hemisphere Tinnitus, similarly as exposure to external sounds, can be
only, are unknown mixtures of inputs from both ears. One solu- related to plastic reorganization of the auditory cortex (241).
tion to this problem is to “frequency-tag” the inputs from both AEFs have been frequently studied in dyslexic individuals,
ears (222): Continuous tones, presented either monaurally to both as indicators of problems of phonologic processing and as
left or right ear, or binaurally, are amplitude modulated with dif- signs of a more general disorder in processing of rapidly pre-
ferent frequencies in the two ears, and the MEG signals of each sented auditory stimuli. For example, Nagarajan et al. (242)
hemisphere are analyzed either in time or in frequency domain. observed that N100m to the second sound of a pair is, at short
Such analysis demonstrates significantly stronger suppression of stimulus onset asynchronies, smaller in dyslexic than normal-
the ipsilateral than contralateral input during binaural hearing. reading adults. The result was interpreted to indicate abnormal
The effect is robust despite interaural intensity differences (223). neural representation of brief and rapidly successive sensory
input, and thereby be a probable correlate of poor reading abil-
ity. Renvall and Hari (243) noted decreased responses to acoustic
AEFs in Patients frequency changes in the left auditory cortex of dyslexic adults.
Ischemic lesions of the temporal lobe often cause marked defects Gootjes et al. (244) developed a simple paradigm to predict
in speech perception and production. Details of symptomatol- language lateralization in right-handed subjects by presenting
ogy cannot, however, be determined from the anatomical lesion vowels, tones, and piano notes in pairs of two stimuli.
alone and additional methods are welcome. In patients with dif- Responses recorded when the subjects had to indicate when the
ferent types of ischemic brain lesions AEFs disappeared only two stimuli were the same showed, in all the 11 subjects studied,
when the lesion extended to the deep region of the temporal left-hemisphere dominance for responses to vowels.
lobe (224,225). Figure 42.26 illustrates the extent of the lesion in
one such patient (226). The responses are normal in the healthy
left hemisphere but totally abolished in the lesioned left hemi- SOMATOSENSORY-EVOKED FIELDS
sphere. The finding is of interest also from the source analysis
point of view: although MEG source modeling is hampered by Responses from SI
the nonunique inverse problem, this kind of result strongly sup- The spatial distribution of sources of somatosensory-evoked
ports the correctness of dipole modeling of normal AEFs, that is, fields (SEFs) represents well the known somatotopic organiza-
sources in the auditory cortices of both hemispheres. tion of the primary somatosensory cortex SI (22,23,41,245–
Chapter 42 ■ Magnetoencephalography: Methods and Applications 885

the median-nerve 30-msec response at the contralateral SI but


enhances it in the ipsilateral SI (253), implying that tactile input
from one hand has access to the SI cortices of both hemi-
spheres, probably through callosal connections. Some long-
latency MEG responses from the ipsilateral SI have been
reported (254). However, transfer of stimulus- or movement-
related vibrations to the other hand has been demonstrated as
one possible cause of ipsilateral responses (255).
Interesting changes occur during the development in the
latencies, relative amplitudes, and polarities of the main SEF
deflections (Fig. 42.28) (256).
Brief bursts of about 600-Hz oscillations are superimposed
on the earliest cortical deflections, the 20-msec N20m response
(257,258). These high-frequency responses will be discussed in
detail in Chapter 37.
Quantification of source strengths may be very useful in
some applications. For example, Elbert et al. (259), determining
source strengths for tactile finger stimulation at SI cortex, found
increased cortical representations for the left hands of string
players, implying practice-related cortical reorganization (see
also Ref. (260)).
Figure 42.29 illustrates the use of SEFs in preoperative iden-
tification of the sensorimotor strip. Superposition of sources
Figure 42.26 Auditory responses from a patient with an extended for median and tibial nerve stimulation on the patient’s MRI
lesion of the right temporal lobe; the shadowed areas in the brain slices illustrates that her cavernous angioma is about 1.5 cm away
show the extent of the lesion. Responses are shown for a representative from the somatomotor hand area. The angioma was later suc-
left- and right-hemisphere channels. The stimuli were noise/ square-wave cessfully operated, without causing any sensory or motor
sequences (shown on the time scale) that typically elicite one onset impairments.
response (N100m) and one transition-related response N100m. (Adapted In Figure 42.30, the preoperative evaluation includes, in
from Mäkelä JP, Hari R. Neuromagnetic auditory evoked responses after addition to SEF recordings, identification of the primary motor
a stroke in the right temporal lobe. Neuroreport. 1992;3:94–96.) cortex, anterior to the central sulcus, by means of cortex–mus-
cle coherence (see Fig. 42.17 and the related text). Visualization
of the somatosensory and motor landmarks on a brain render-
247). Since MEG mainly picks signals from tangential currents, ing with veins superimposed (right) helps the neurosurgeon to
the convexial cortex is either silent or poorly represented in the navigate during the operation (58).
SEF distributions. At SI, the hand-area signals should therefore
arise mainly from cytoarchitectonic area 3b, whereas in the foot Responses from SII and Other Nonprimary
SI all cytoarchitectonic areas should produce currents tangen- Cortical Areas
tial to the skull and thus their changed relative contributions MEG recordings have indicated that peripheral stimuli also
should be seen as rotation of the field patterns. Evoked field activate bilaterally areas close to the upper lip of the Sylvian fis-
data to lower-limb stimulation suggest that such rotations really sure, that is, most probably at the second somatosensory area
occur as signs of successive activation of several cytoarchitec- SII (23,45,261,262). Although the SI/SII differentiation was
tonic areas (248–252). possible already with small MEG sensor arrays, the more recent
Figure 42.27 shows one subject’s responses to left tibial- whole-scalp recordings have given more detailed information
nerve stimuli, with largest signals at the top of the head (251). about the cortical somatosensory network, with spatially and
The field patterns rotate as a function of time, as predicted temporally distinct activations also in the posterior parietal cor-
above. A two-dipole model, with one dipole in area 3b and the tex, in the mesial cortex, and in the frontal lobes (12,24–26).
other in area 5, both in the mesial wall of the hemisphere, The field patterns of Figure 42.31 illustrate activation of the
explained the data during the first 100 msec, with a consistent right (contralateral) SI, left and right SII, and the posterior pari-
3-msec time difference in their initial peak latencies. Other etal cortex after electric stimulation of the left thumb. Tactile
source models were rejected on the basis of their inconsisten- stimulation activates essentially the same brain regions as does
cies with experimental data and/or anatomical information. A electric stimulation, but with slightly different latencies and rel-
slightly different two-source model for tibial-nerve SEFs was ative amplitudes (263). The MEG source locations in SI and SII
suggested by Hashimoto et al. (252) on the basis of beam- cortices agree with fMRI data collected from the same subjects
former analysis. (264,265).
SEFs from the SI cortex are typically strictly contralateral. The representations of fingers of both hands overlap
However, tactile interference applied on the palm suppresses strongly in the bilaterally activated SII cortices, whereas the SI
886 Part V ■ Complementary and Special Techniques

SEFs in Multiple Sclerosis


In patients with clinically diagnosed multiple sclerosis (271),
the N20m deflection was dampened and delayed by 2.5 to
3 msec. The 50- to 80-msec responses were often of larger
amplitude than in control subjects, especially in patients who
had periventricular plaques in MRI. The source locations of
N20m and P60m in the somatosensory hand area did not differ
between the groups.
It is probable that enhancement of the corresponding elec-
tric SEPs has remained undetected with the typical high-pass
filter settings at 10 to 20 Hz and the analysis periods of 30 to
50 msec applied in many laboratories.

SEFs in Myoclonus Epilepsy


Karhu et al. (272) suggested that progressive myoclonus
epilepsy (PME; Unverricht-Lundborg type) is associated with
thalamocortical hyperreactivity in the sensorimotor but not in
the auditory system. The patients were adequately medicated
and their MEG did not show spikes. The amplitudes of the
30-msec SEFs were up to sixfold compared with the response of
normal subjects, but the source locations agreed with the acti-
vation of the somatosensory hand area. Patients of juvenile
neuronal ceroid lipofuscinosis (JNCL) and late INCL (CLN5)
also show enhanced SEFs (273,274).
“Giant” SEFs from SI have been recorded also from a genet-
ically homogeneous group of PME (Unverricht-Lundborg)
patients (275). Figure 42.32 shows, as a sign on hyperreactivity,
strongly enhanced responses at the contralateral SI of a patient.
Responses are also seen in the ipsilateral SI, in contrast to the
healthy control subject; the latency difference of about 20 msec
suggests facilitated callosal transfer to the ipsilateral side.
Moreover, despite the enhanced SI responses, no clear SII
responses are seen in the patient. Findings like this encourage
Figure 42.27 Top: Responses of one subject to electric stimulation of the the clinical neurophysiologist to assess functions of the
left tibial nerve at the ankle. The traces are averages of 800 single somatosensory circuitry beyond the primary somatosensory
responses. The inset shows two enlarged sets of 400 responses from suc- cortex, as the dysfunctions may have direct consequences on the
cessive experiments superimposed. The passband is from 0.03 to 275 Hz. symptoms, prognosis, and/or rehabilitation of the patient.
Bottom: Magnetic field patterns and equivalent current dipoles of the same
subject as a function of time (indicated in msec). The isocontours are sep-
Noxious Stimulation
arated by 20 fT. (Adapted from Hari R, Nagamine T, Nishitani N, et al.
Time-varying activation of different cytoarchitectonic areas of the human MEG responses to painful stimuli imply activation around SII
SI cortex after tibial nerve stimulation. NeuroImage. 1996;4:111–118.) and/or the frontal operculum to electric stimulation of the
tooth pulp, carbon dioxide stimulation of the nasal mucosa,
and noxious stimulation (by means of electric current or CO2
or thulium laser) of the skin (276–283). Whole-scalp record-
cortex (area 3b) has representations of fingers of one hand only ings indicated a clear right- hemisphere predominance to
and with much weaker overlap (266). Bilateral median-nerve painful CO 2 stimulation of the nasal mucosa (284).
stimulation elicits left-hemisphere-dominant SII activation, With laser heat it is possible to stimulate rather selectively
suggesting handedness-independent functional specialization A - and C-fibers by applying different stimulus intensities to
of the human SII cortices (267). stimulus areas of different sizes (280,283,284). Figure 42.33
Differentiation of various source areas of SEFs has started to shows that the evoked responses from the contralateral SII cor-
enlighten functions of the cortical somatosensory network tex peak to A -fiber stimuli at about 160 msec and to C-fiber
(12,23,159,268) and recordings of SEFs in patients with various stimuli around 800 msec, in agreement with the conduction
lesions (259), and analysis of, for example, phase locking velocities of these two fiber types (283). Interestingly, monitor-
between areas (270) may bring new information about the ing of the level of the 20-Hz motor–cortex activity strongly sug-
debate on parallel versus serial processing in different gests that the painful laser stimuli automatically activate the
somatosensory cortical areas. primary motor cortex and therefore might be informative
Chapter 42 ■ Magnetoencephalography: Methods and Applications 887

–50 0 50 200 ms –50 0 50 200 ms

50 fT/cm

N20m P30m
M30 M60

Newborn Adult

Figure 42.28 Somatosensory-evoked fields and field patterns to median nerve stimulation in a newborn and an adult.
The signals are from one planar gradiometer channel over the right somatosensory cortex. (Adapted from Lauronen L,
Nevalainen P, Wikström H, et al. Immaturity of somatosensory cortical processing in human newborns. Neuroimage.
2006;33:195–203.) The photograph on the left illustrates how with an adult-shape neuromagnetometer, just one hemi-
sphere at the time can be recorded from the small baby brain. (Courtesy of Elina Pihko.) (See color insert)

Figure 42.29 Left: Sources for responses to left


median and tibial nerve stimulation superposed
on the MRI reconstruction of the subject. The
section passes through both sources to illustrate
the central sulcus just anterior to them. Right: A
parallel section slightly deeper, with the cav-
ernous angioma indicated. The patient, a 50-
year-old female, was later successfully operated.

Figure 42.30 Preoperative functional localiza-


tion in a patient with a parietal-lobe tumor. The
postcentral somatosensory cortex has been iden-
tified by measuring SEFs to median-nerve (hand)
and tibial-nerve (foot) stimulation. In addition,
cortex–muscle coherence for hand and foot small
muscles has been used to identify the precentral
primary motor cortex. On the right, veins are
superimposed on the surface rendering.
(Courtesy of CliniMEG, Brain Research Unit, Low
Temperature Laboratory, Aalto University School
of Science and Technology.)
888 Part V ■ Complementary and Special Techniques

patterns. The retinotopic organization of the occipital visual


cortex has been studied with MEG by presenting stimuli to dif-
ferent parts of the visual field (293–296).
Outside the occipital cortex the visual field patterns are very
complex and the utilization of VEFs as tools of clinical neuro-
physiology is still scarce. Figure 42.34 shows responses to
half-field pattern and luminance stimuli. The first responses are
generated in the occipital cortex but a later response occurs,
especially for luminance stimuli, in the region of the parieto-
occipital sulcus (POS).
The very same POS area close to the midline can be activated
by various visual stimuli (295) as well as by voluntary eyeblinks
(297,298) and saccades (299); the finding has been confirmed
in functional magnetic resonance recordings. Anatomically, the
posterior parietal midline region, which also produces the most
prominent MEG alpha activity, could well be the human homo-
logue of the monkey V6/V6A region, influenced by, for exam-
ple, saccades and eye position and proposed to be related to
spatial coding of extrapersonal visual space and to visuomotor
integration (300). The V6/POS region is a part of the dorsal
visual stream and thus of interest for both basic neuroscience
and for some neurological disorders affecting visuomotor
coordination.
The human homologue of the monkey MT/V5 cortex, a
brain area selective to visual motion, has also been identified by
MEG recordings (301–304). Better understanding of neural ori-
gin of different VEF components brings invaluable information
about temporal relationships in the activation of different
visual areas. Interestingly, the activation order of human visual
cortices can deviate from the simple serial activation pattern
(305), in agreement with both monkey recordings (306) and
Figure 42.31 Field patterns of SEFs to electric stimulation of the simulation studies (307), thereby indicating the importance of
subject’s left thumb (ISI 4 seconds). Illustrative responses are shown reciprocal interareal connections in a hierarchically connected
from four areas (contralateral SI, both SII regions, and the PPC; panels network.
A, B, C, and D, respectively) and the corresponding field patterns at the
time of maximum response from each region. (Adapted from Forss N, Responses to Faces and Language Stimuli
Jousmäki V, Hari R. Interaction in the human somatosensory cortex In agreement with both clinical and other imaging data, MEG
between afferent input from different fingers. Brain Res. 1995;685:68–76.) responses to pictures of faces indicate activation of extrastriate
visual areas, predominantly in the right posterior fusiform
about the mechanisms underlying pathological muscle contrac- gyrus (308–311). Face-categorization-related activity has been
tion in tension pain (286–288). In patients with chronic pain, reported as early as 100 msec after the stimulus (312), whereas
enhanced activation of the motor cortex occurs after innocuous the typical face-specific response peaks at 140 to 170 msec (60),
tactile stimuli (289). MEG recordings have also demonstrated and the activation shows a systematic amplitude dependence on
the spread of a pain-related disorder of somatosensory process- the degree of “faceness” of the stimulus (311).
ing from one hemisphere to another in patients suffering from MEG’s excellent temporal resolution is of high value in
complex regional pain syndrome (290). studies of language perception and production. Salmelin et al.
(313) were able to follow the cortical activation sequence
Visual-Evoked Fields from the posterior brain areas toward more anterior regions
Visual-evoked fields (VEFs) were reported for the first time in when the subjects named pictures of objects. Signs of consid-
1975 (291,292). With flash stimuli, VEFs and VEPs did not peak erable parallel processing were evident and both hemispheres
simultaneously and the responses depended on luminance in were activated by the task. Interestingly, MEG studies of
different manner, thereby suggesting separate sources for VEFs incongruities in musical syntax suggest that Broca’s region
and VEPs (292). Steady-state recordings with gratings reversing and its right-hemisphere homologue area process syntactic
at 13 Hz (291) allowed for the determination of the sources: information that is less language-specific than usually
The field patterns evoked by half-field stimuli agreed with acti- believed (314).
vation of the contralateral occipital cortex and the patterns to More recently, MEG has been increasingly applied for stud-
full-field stimulation were the sums of the two half-field ies of reading and naming, both in healthy subjects (315) and in
Chapter 42 ■ Magnetoencephalography: Methods and Applications 889

Figure 42.32 Source locations (left) and source waveforms as a function of time (right) to electric stimulation of the left
median nerve in a healthy control subject and in a patient suffering from Unverricht-Lundborg-type progressive myoclonus
epilepsy. The black circles indicate source locations and the white circles indicate possible source locations that did not
show any activation; c and I refer to contralateral and ipsilateral hemispheres. (Adapted from Forss N, Silén T, Karjalainen
T. Lack of activation of human secondary somatosensory cortex in Unverricht-Lundborg type of progressive myoclonus
epilepsy. Ann Neurol. 2001;49:90–97.)

Figure 42.33 Top left: Averaged responses (N = 100)


of a single subject from right SII region to selective A -
(upper trace; weak pricking pain) and C-fiber (lower
trace; weak burning pain) thulium-laser stimuli pre-
sented to the dorsum of the left hand once every 4.5 to
5.5 seconds. Top right and bottom: The mean source
locations of responses in the SII (top) and PPC (bot-
tom) region to A -fiber stimuli. The ovals indicate the
mean SEM locations across eight subjects superim-
posed on slices of a mean brain of the subjects, calcu-
lated by means of elastic transformations of their
magnetic resonance images. (Modified from Forss N,
Raij TT, Seppä M, et al. Common cortical network for
first and second pain. Neuroimage. 2005;24:132-142.)
890 Part V ■ Complementary and Special Techniques

Figure 42.34 Left: Responses to stimulation of the right visual hemifield by luminance stimuli. Right: Responses of the
same subject to left and right hemifield (LVF, RVF) luminance and pattern stimuli (radius 8 deg) from the parietal region
(Par) and from left and right occipital areas (Locc, Rocc). Note the predominance of the parietal response to luminance
stimuli and the very clear LVF versus RVF difference in the occipital channels for pattern, but not for luminance, stimuli.
(Adapted from Portin K, Salenius S, Salmelin R, et al. Activation of the human occipital and parietal cortex by pattern and
luminance stimuli: neuromagnetic measurements. Cereb Cortex. 1998;18:253–260.)

aphasic patients (316), stuttering subjects (317), as well as in maxima were observed, one in the parietal midline and another
dyslexic individuals (318–320). New interesting approaches in the lowest posterior channels. Source modeling suggested
include the analysis of long-range connectivity (321) and the that the former response was generated in the POS region and
study of language development (322). the latter signal in cerebellar vermis. Both source activations
peaked about 170 msec after the saccade onset, and the cerebel-
Saccade-Related Activity lar signal started already 30 msec before the saccade. The POS
Figure 42.35 shows saccade-related MEG signals, averaged on signal was abolished when the lights of the experimental room
the basis of the saccade onsets (299). The MEG channels close were turned off whereas the cerebellar response was only
to the orbits record eyeball-related signals. Two main signal slightly delayed. The possibility to noninvasively follow the

Figure 42.35 Magnetoencephalographic signals


from three locations (shown on the schematic
head) during a visually-guided saccade task. The
signals were averaged with respect to the onset of
horizontal saccades (leftwards or rightwards);
the horizontal EOG used as the trigger is depicted
above. Signals recorded during darkness and
with lights on are illustrated with dashed and
solid lines, respectively. (Adapted from Jousmäki
V, Hämäläinen M, Hari R. Magnetic source imag-
ing during a visually guided task. Neuroreport.
1996;7:2961–2964.)
Chapter 42 ■ Magnetoencephalography: Methods and Applications 891

A B
12 Hz

Spectral density
12 Hz 30

(fT/cm/ Hz)
15 Hz 15 Hz
20

10

0 10 20 30
Frequency (Hz)
Time (s)
–2 –1 0 1 2
C

Frequency (Hz)
15 Hz (face-tag) 20

10
12 Hz (vase-tag)

PERCEPT “vase” “faces”

Figure 42.36 An example of frequency tagging. (A) The Rubins vase/ faces figure used as the stimulus. Dynamical noise
was superimposed to the image and updated at 12 Hz in the vase area and at 15 Hz in the face areas. (B) Frequency
spectrum from one occipital magnetoencephalogram (MEG) channel, based on 10-minute recording. (C) Time–frequency
representation of MEG activity computed time-locked to the change in percept, indicated by button press. (Adapted
from Parkkonen L, Andersson J, Hämäläinen M, et al. Early visual areas reflect the percept of an ambiguous scene.
Proc Nat Acad Sci USA. 2008;105:20500–20504.)(See color insert)

temporal behavior of human cerebellar activation may open monkey or by the experimenter. The mirror neurons have
new windows to the monitoring of neurologic patients. Early been assumed to match action observation and execution
cerebellar signals related to somatosensory stimulation have (for a review, see Ref. (327)). In humans, the MNS comprises
been observed (323), as have oscillations anticipating omissions at least the primary motor cortex (see Fig. 42.16), inferior
of somatosensory stimuli (324). frontal cortex (Broca’s area in the left hemisphere), and infe-
rior parietal lobe, with contribution from the superior
Cortical Correlates of Perceptual Sw itches temporal sulcus that does not contain proper mirror neu-
Some percepts change so unexpectedly that it is not possible to rons. These areas are activated in a specific order within 250
time-lock the analysis of MEG signals to the event. Figure 42.36 to 400 msec during viewing of other person’s motor acts
shows how frequency tagging can help to follow cortical activa- (328,329). Moreover, the primary and secondary somatosen-
tion changes related to the spontaneous switching of percepts of sory cortices react to observed hand movements. The pri-
the Rubin’s vase (325). Dynamic noise was superimposed to the mary motor cortex is activated before and during observed
picture and updated at 12 Hz in the vase region and at 15 Hz in actions (that the viewer already more or less knows) and is
the face region. The subjects were not able to tell the difference stabilized afterwards, very similarly as happens during the
between the update frequencies, but their occipital MEG signals person’s own actions; these conclusions are based on the
displayed distinct frequency peaks both at 12 and 15 Hz. reactivity of the motor-cortex 20-Hz rhythm (330). The human
On the basis of the changes in the relative strengths of the MNS may play an important role both in action imitation
12- and 15-Hz frequencies during the percepts it was possible to and in understanding the meaning of actions made by other
conclude that the activation of the early visual cortices covaried subjects, thereby also having relevance for social interaction
with the percept, most likely because of top-down influences. (for reviews, see Refs. (332,333)).
Eye tracking ruled out the possibility that the changes in the
MEG signals could have been related to gaze deviations (325). CONCLUSIONS
Human Mirror-Neuron System MEG has established a position as a tool of basic neuroscience,
MEG recordings have been recently used for studies of the and clinical applications have started to rapidly develop when
human mirror-neuron system (MNS). Mirror neurons were neuromagnetometers with whole-scalp coverage have been
first described in monkey premotor cortical area F5 (326): installed in hospital environments. During the last few years,
the neurons discharged both when a monkey executed a hand combination of functional information, deduced from MEG,
action and when it observed a similar action made by another and structural information, obtained from MRI, has become a
892 Part V ■ Complementary and Special Techniques

routine, and coregistration of MEG and fMRI data have started 6. Näätänen R, Ilmoniemi R, Alho K. Magnetoencephalography in
to become popular as well. Instead of instrumental develop- studies of human cognitive brain function. Trends Neurosci.
ment, which already has reached a very user-friendly stage, 1994;17:389–395.
the efforts of the neuromagnetism community are now 7. Baumgartner C, Deecke L, Stroink G, et al., eds. Biomagnetism:
Fundamental Research and Clinical Applications. Amsterdam:
increasingly focusing on development of new signal analysis
Elsevier; 1995.
approaches (14).
8. Hashimoto I, Okada YC, Ogawa S, eds. Visualization of Information
MEG’s clinical applications in neurology, neurosurgery, and Processing in the Human Brain. Amsterdam: Elsevier; 1996.
audiology are expanding rapidly and the method will probably 9. Lounasmaa OV, Hämäläinen M, Hari R, et al. Information pro-
be useful also in developmental neuroscience and in neuropsy- cessing in the human brain – magnetoencephalographic approach.
chiatry. In addition to conventional studies of the auditory, Proc Natl Acad Sci USA. 1996;93:8809–8815.
visual, and somatomotor systems, stimulation techniques are 10. Aine C, OkadaYC, Stroink G, et al., eds. Advances in Biomagnetism:
available to obtain selective MEG responses also from olfactory Biomag96. Springer Verlag; 1997.
and gustatory systems, and reliable data have been obtained 11. Hari R, Salmelin R. Human cortical rhythms: a neuromagnetic
with real-life-like stimulation. With the whole-scalp neuromag- view through the skull. Trends Neurosci. 1997;20:44–49.
netometers and with improved head-position monitoring tools 12. Hari R, Forss N. Magnetoencephalalography in the study of
human somatosensory cortical processing. Proc Royal Soc Lond B.
as well as sophisticated artifact suppression methods, MEG can
1999;354:1145–1154.
be applied in a variety of behavioral conditions, designed for
13. Forss N, Nakasato N, Ebersole J, et al. Clinical use of magnetoen-
assessment of neural basis of cognitive functions. cephalography. Suppl Clin Neurophysiol. 2000;53:287–297.
MEG allows studies of the temporal activation order of 14. Baillet S, Mosher JC, Leahy RM. Electromagnetic brain mapping.
several cortical areas, thereby offering new noninvasive infor- IEEE Signal Proc Magaz. 2001;18:14–30.
mation of brain dynamics and temporal hierarchies (334). A 15. Del Gratta C, Pizzella V, Tecchio F, et al. Magnetoencephalography –
clinical neurophysiologist naturally looks for practical applica- a noninvasive brain imaging method with 1 ms time resolution.
tions, appropriate to the functional assessment of sensory path- Rep Prog Phys. 2001;64:1759–1814.
ways and the cortex. In this respect, MEG’s ability to locate 16. Hämäläinen M, Hari R. Magnetoencephalographic characteriza-
sources of various electric signals that are frequently measured tion of dynamic brain activation: basic principles, and methods
with scalp EEG is very helpful. Better knowledge of cortical- of data collection and source analysis. In: Toga AW, Mazziotta JC,
eds. Brain Mapping. The Methods, 2nd ed. Amsterdam: Academic
evoked responses and spontaneous rhythms will further widen
Press; 2002:227–253.
the clinical scope of both EEG and MEG recordings. Necessity
17. Lu ZL, Kaufman L, eds. Magnetic Source Imaging of the Human
to map and interpret complex dynamical activation sequences Brain. Mahwah, New Jersey: Lawrence Erlbaum Associates; 2003.
of cortical circuitries requires the future clinical neurophysiol- 18. Hansen P, Kringelbach M, Salmelin R, eds. MEG. An Introduction
ogists to obtain good understanding about modern electro- to Methods. New York: Oxford University Press; 2010.
physiologic and imaging methods, but also about systems-level 19. Pernier J, Perrin F, Bertrand O. Scalp current density fields: con-
basic and cognitive neuroscience. cept and properties. Electroencephalogr Clin Neurophysiol. 1988;69:
385–389.
20. Nunez PL. Estimation of large scale neocortical source activity
ACKNOWLEDGMENTS with EEG surface Laplacians. Brain Topogr. 1989;2:141–154.
21. Cohen D, Cuffin B. Demonstration of useful differences between
This study has been supported by the Academy of Finland and
MEG and EEG. Electroencephalogr Clin Neurophysiol. 1983;56:38–51.
the Sigrid Jusélius Foundation. The skill, support, and enthusi-
22. Hari R, Reinikainen K, Kaukoranta E, et al. Somatosensory evoked
asm of the members of the Brain Research Unit of the Low cerebral magnetic fields from SI and SII in man. Electroencephlogr
Temperature Laboratory, Helsinki University of Technology, Clin Neurophysiol. 1984;57:254–263.
have been decisive for collecting the MEG data presented in this 23. Hari R, Karhu J, Hämäläinen M, et al. Functional organization of
chapter. the human first and second somatosensory cortices: a neuromag-
netic study. Eur J Neurosci. 1993;5:724–734.
24. Forss N, Hari R, Salmelin R, et al. A novel source area in the human
REFERENCES
parietal cortex activated by median nerve stimulation. Exp Brain
1. Cohen D. Magnetoencephalography: evidence of magnetic field Res. 1994;99:309–315.
produced by alpha-rhythm currents. Science. 1968;161:784–786. 25. Forss N, Merlet I, Vanni S, et al. Activation of human mesial cortex
2. Hari R, Lounasmaa OV. Recording and interpretation of cerebral during somatosensory attention task. Brain Res. 1996;734:229–235.
magnetic fields. Science. 1989;244:432–436. 26. Mauguière F, Merlet I, Forss N, et al. Activation of a distributed
3. Grandori F, Hoke M, Romani G-L, eds. Auditory Evoked Magnetic somatosensory cortical network in the human brain. A dipole
Fields and Potentials. Advances in Audiology, Vol. 6. Basel: modelling study of magnetic fields evoked by median nerve stim-
Karger; 1990. ulation. Part I: location and activation timing of SEF sources.
4. Sato S, eds. Magnetoencephalography. Advances in Neurology. Vol. Electroencephalogr Clin Neurophysiol. 1997;104:281–289.
54. New York: Raven Press; 1990. 27. Hari R. On brain’s magnetic responses to sensory stimuli. J Clin
5. Hämäläinen M, Hari R, Ilmoniemi R, et al. Magnetoence- Neurophysiol. 1991;8:157–169.
phalography—theory, instrumentation, and applications to non- 28. Parkkonen L, Fujiki N, Mäkelä J. Sources of auditory brainstem
invasive studies of the working human brain. Rev Mod Phys. responses revisited: contribution by magnetoencephalography.
1993;65:413–497. Hum Brain Mapp. 2009;30:1772–1782.
Chapter 42 ■ Magnetoencephalography: Methods and Applications 893

29. Tesche CD. Non-invasive imaging of neuronal population dynam- inverse solutions. Tenth International Conference on Biomagnetism.
ics in human thalamus. Brain Res. 1996;729:253–258. Santa Fe, NM, Volume of Abstracts; 1996:149.
30. Tesche CD. Non-invasive detection of ongoing neuronal population 50. Hari R. The neuromagnetic method in the study of the human
activity in normal human hippocampus. Brain Res. 1997;749:53–60. auditory cortex. In: Grandori F, Hoke M, Romani G, eds. Auditory
31. Numminen J, Ahlfors S, Ilmoniemi R, et al. Transformation of Evoked Magnetic Fields and Potentials. Advances in Audiology.
multichannel magnetoencephalographic signals to standard grid Vol. 6. Basel: Karger; 1990:222–282.
form. IEE Trans Biomed Eng. 1995;42:72–77. 51. Bernander O, Douglas R, Martin K, et al. Synaptic background
32. Barth D, Sutherling W, Engel JJ, et al. Neuromagnetic localization activity determined spatial-temporal integration in pyramidal
of epileptiform spike activity in the human brain. Science. cells. Proc Natl Acad Sci USA. 1991;88:11569–11573.
1982;218:891–894. 52. Hämäläinen M, Ilmoniemi R. Interpreting magnetic fields of the
33. Duret D, Karp P. Figure of merit and spatial resolution of super- brain: minimum norm estimates. Med Biol Eng Comput.
conducting flux transformers. J Appl Phys. 1984;56:1762–1768. 1994;32:35–42.
34. Romani G, Leoni R. Localization of cerebral sources by neuromag- 53. Ribary U, Ioannides A, Singh K, et al. Magnetic field tomography
netic measurements. In: Weinberg H, Stroink G, Katila T, eds. of coherent thalamocortical 40-Hz oscillation in humans. Proc
Biomagnetism: Applications and Theory. Oxford: Pergamon; Natl Acad Sci USA. 1991;88:11037–11041.
1985:205–220. 54. Volkmann J, Joliot M, Mogilner A, et al. Central motor loop oscil-
35. Curio G, Drung D, Koch H, et al. Magnetometry of evoked fields lations in parkinsonian resting tremor revealed by magnetoen-
from human peripheral nerve, brachial plexus and primary cephalography. Neurology. 1996;46:1359–1370.
somatosensory cortex using a liquid nitrogen cooled superconduct- 55. Uutela K, Hämäläinen M, Somersalo E. Visualization of magne-
ing quantum interference device. Neurosci Lett. 1996;206:204–206. toencephalographic data using minimum current estimates.
36. Kominis IK, Kornack TW, Allred JC, et al. A subfemtotesla multi- Neuroimage. 1999;10:173–180.
channel atomic magnetometer. Nature. 2003;422:596–599. 56. Stenbacka L, Vanni S, Uutela K, et al. Comparison of minimum cur-
37. Pannetier M, Fermon C, Le Goff G, et al. Femtotesla magnetic field rent estimate and dipole modeling in the analysis of simulated acti-
measurement with magnetoresistive sensors. Science. 2004;304: vity in the human visual cortices. Neuroimage. 2002;16:936–943.
1648–1650. 57. Jack C, Marsh W, Hirschorn K, et al. EEG scalp electrode projec-
38. Hillebrand A, Barnes GR. A quantitative assessment of the sensi- tions onto three-dimensional surface rendered images of the
tivity of whole-head MEG to activity in the adult human cortex. brain. Radiology. 1990;176:413–418.
Neuroimage. 2002;16:638–650. 58. Mäkelä J, Kirveskari E, Seppä M, et al. Three-dimensional integration
39. Sutherling W, Crandall P, Darcey T, et al. The magnetic and electric of brain anatomy and function to facilitate intraoperative navigation
fields agree with intracranial localizations of somatosensory cor- around the sensorimotor strip. Hum Brain Mapp. 2001;12:181–192.
tex. Neurology. 1988;38:1705–1714. 59. Aine CJ. A conceptual overview and critique of functional neu-
40. Tiihonen J, Hari R, Hämäläinen M. Early deflections of cerebral roimaging techniques in humans: I. MRI/fMRI and PET. Crit Rev
magnetic responses to median nerve stimulation. Neurobiol. 1995;9:229–309.
Electroencephalogr Clin Neurophysiol. 1989;74:290–296. 60. Furey ML, Tanskanen T, Beauchamp MS, et al. Dissociation of face
41. Baumgartner C. Clinical Neurophysiology of the Somatosensory selective cortical responses by attention. Proc Natl Acad Sci USA.
Cortex. Wien: Springer Verlag; 1993 2006;103:1065–1070.
42. Lounasmaa OV, Williamson SJ, Kaufman L, et al. Visually evoked 61. Nangini C, Hlushchuk , Hari R. Predicting stimulus-rate sensitivity
responses from non-occipital areas of human cortex. In: Weinberg of human somatosensory fMRI signals by MEG. Human Brain
H, Stroing G, Katila T, eds. Biomagnetism: Applications & Theory. Mapp. 2009;30:1824–1832.
New York: Pergamon Press; 1985:348–353. 62. Hari R, Ilmoniemi RJ. Cerebral magnetic fields. CRC Crit Rev
43. Tesche CD, Uusitalo MA, Ilmoniemi RJ, et al. Signal-space projec- Biomed Eng. 1986;14:93–126.
tions of MEG data characterize both distributed and well-localized 63. Tarkiainen A, Liljeström M, Seppä M, et al. The 3D topography of
neuronal sources. Electroencephalogr Clin Neurophysiol. 1995;95: MEG source localization accuracy: effects of conductor model and
189–200. noise. Clin Neurophysiol. 2003;114:1977–1992.
44. Uusitalo M, Ilmoniemi R. Signal-space projection method for sep- 64. Hämäläinen MS, Sarvas J. Realistic conductivity geometry model
arating MEG and EEG into components. Med Biol Eng Comput. of the human head for interpretation of neuromagnetic data. IEEE
1997;35:135–140. Trans Biomed Eng. 1989;36:165–171.
45. Kaukoranta E, Hämäläinen M, Sarvas J, et al. Mixed and sensory 65. Cuffin B. Effects of fissures in the brain on electroencephalograms
nerve stimulations activate different cytoarchitectonic areas in the and magnetoencephalograms. J Appl Phys. 1985;57:146–153.
human primary somatosensory cortex SI. Exp Brain Res. 66. Cooper R, Winter A, Crow H, et al. Comparison of subcortical,
1986;63:60–66. cortical and scalp activity using chronically indwelling electrodes
46. Hari R, Joutsiniemi SL, Sarvas J. Spatial resolution of neuromag- in man. Electroencephalogr Clin Neurophysiol. 1965;18:217–228.
netic records: theoretical calculations in a spherical model. 67. Ilmoniemi R. Radial anisotropy added to a spherically symmetric
Electroencephalogr Clin Neurophysiol. 1988;71:64–72. conductor does not affect the external magnetic field due to inter-
47. Scherg M. Fundamentals of dipole source potential analysis. In: nal sources. Europhys Lett. 1995;30:313–316.
Grandori F, Hoke M, Romani GL, eds. Auditory Evoked Magnetic 68. Antervo A, Hari R, Katila T, et al. Magnetic fields produced by eye
Fields and Potentials. Advances in Audiology. Vol. 6. Basel: Karger; blinking. Electroencephalogr Clin Neurophysiol. 1985;61:247–254.
1990:40–69. 69. Jousmäki V, Hari R. Cardiac artifacts in magnetoencephalogram.
48. Vvedensky V, Hari R, Ilmoniemi R, et al. Physical basis of neuro- J Clin Neurophysiol. 1996;13:172–176.
magnetic fields. Biophysica. 1985;30:154–158. 70. Samonas M, Petrou M, Ioannides A. Identification and elimina-
49. Okada YC, Papuashvili N, Xu C. Maximum current dipole tion of cardiac contribution in single-trial magnetoencephalo-
moment density as an important physiological constraint in MEG graphic signals. IEEE Trans Biomed Eng. 1997;44:386–393.
894 Part V ■ Complementary and Special Techniques

71. Taulu S, Simola J, Kajola M. Applications of the signal space sepa- 92. Salenius S, Kajola M, Thompson WL, et al. Reactivity of magnetic
ration method. IEEE Trans Sign Proc. 2005;53:3359–3372. parieto-occipital alpha rhythm during visual imagery.
72. Taulu S, Simola J. Spatiotemporal signal space separation method Electroencephalogr Clin Neurophysiol. 1995;95:453–462.
for rejecting nearby interference in MEG measurements. Phys Med 93. Portin K, Salenius S, Salmelin R, et al. Activation of the human
Biol. 2006;51:1759–1768. occipital and parietal cortex by pattern and luminance stimuli:
73. Taulu S, Hari R. Removal of MEG artifacts with temporal signal- neuromagnetic measurements. Cereb Cortex. 1998;8:253–260.
space separation: demonstration with single-trial auditory evoked 94. Vanni S, Revonsuo A, Hari R. Modulation of the parieto-occipital
responses. Hum Brain Mapp. 2008;30:1524–1534. alpha rhythm during object detection. J Neurosci. 1997;17:7141–
74. Mäkelä JP, Taulu S, Pohjola J, et al. Effects of subthalamic nucleus 7147.
stimulation on spontaneous sensorimotor MEG activity in a 95. Tuladhar AM, ter Huurne N, Schoffelen JM, et al. Parieto-
Parkinsonian patient. Int Congr Ser. 2007;1300:345–348. occipital sources account for the increase in alpha activity with
75. Mertens M, Lütkenhöner B. Effective neuromagnetic determina- working memory load. Hum Brain Mapp. 2007;28:785–792.
tion of landmarks in the somatosensory cortex. Clin Neurophysiol. 96. Jeavons P. The electro-encephalogram in blind children. Br
2000;111:1478–1487. J Opthalmol. 1964;48:83–101.
76. Jousmäki V, Nishitani N, Hari R. Brush stimulator for functional 97. Salenius S, Schnitzler A, Salmelin R, et al. Modulation of human
brain imaging. Clin Neurophysiol. 2007;118:2620–2624. rolandic rhythms during natural sensorimotor tasks.
77. Lütkenhöner B. Frequency-domain localization of intracerebral dipo- Neuroimage. 1997;5:221–228.
lar sources. Electroencephalogr Clin Neurophysiol. 1992;82:112–118. 98. Tiihonen J, Kajola M, Hari R. Magnetic mu rhythm in man.
78. Tesche C, Kajola M. A comparison of the localization of sponta- Neuroscience. 1989;32:793–800.
neous neuromagnetic activity in the frequency and time domains. 99. Nagamine T, Kajola M, Salmelin R, et al. Movement-related slow
Electroencephalogr Clin Neurophysiol. 1993;87:408–416. cortical magnetic fields and changes of spontaneous brain
79. Salmelin R, Hämäläinen M. Dipole modelling of MEG rhythms in rhythms. Electroencephalogr Clin Neurophysiol. 1996;99:274–296.
time and frequency domains. Brain Topogr. 1995;7:251–257. 100. Schnitzler A, Salenius S, Salmelin R, et al. Involvement of primary
80. Salmelin R, Hari R. Spatiotemporal characteristics of rhythmic motor cortex in motor imagery: a neuromagnetic study.
neuromagnetic activity related to thumb movement. Neuroscience. Neuroimage. 1997;6:201–208.
1994;60:537–550. 101. Hari R, Forss N, Avikainen S, et al. Activation of human primary
81. Pfurtscheller G, Aranibar A. Event-related desynchronization motor cortex during action observation: a neuromagnetic study.
detected by power measurements of scalp EEG. Electroenceph Clin Proc Natl Acad Sci USA. 1998;95:15061–15065.
Neurophysiol. 1977;42:138–146. 102. Salmelin R, Hämäläinen M, Kajola M, et al. Functional segrega-
82. Barth DS, Sutherling W, Engel J, et al. Neuromagnetic evidence of tion of movement-related rhythmic activity in the human brain.
spatially distributed sources underlying epileptiform spikes in the Neuroimage. 1995;2:237–243.
human brain. Science. 1984;223:293–296. 103. Jensen O, Goel P, Kopell N, et al. On the human motor-cortex beta
83. Stefan H, Schneider S, Abraham-Fuchs K, et al. The neocortico to rhythm: Sources and modelling. Neuroimage. 2005;26:347–355.
mesio-basal limbic propagation of focal epileptic activity during 104. Conway B, Halliday D, Farmer S, et al. Synchronization between
the spike-wave complex. Electroencephalogr Clin Neurophysiol. motor cortex and spinal motoneuronal pool during the perform-
1991;79:1–10. ance of a maintained motor task in man. J Physiol. 1995;489:
84. Chapman RM, Ilmoniemi RJ, Barbanera S, et al. Selective localiza- 917–924.
tion of alpha brain activity with neuromagnetic measurements. 105. Salenius S, Salmelin R, Neuper C, et al. Human cortical 40-Hz
Electroencephalogr Clin Neurophysiol. 1984;58:569–572. rhythm is closely related to EMG rhythmicity. Neurosci Lett.
85. Vvedensky V, Ilmoniemi R, Kajola M. Study of the alpha rhythm 1996;21:75–78.
with a 4-channel SQUID magnetometer. Med Biol Eng Comput. 106. Salenius S, Portin K, Kajola M, et al. Cortical control of human
1986;23:11–12. motoneuron firing during isometric contraction. J Neurophysiol.
86. Williamson S, Wang J-Z, Ilmoniemi R. Method for locating 1997;77:3401–3405.
sources of human alpha activity. In: Williamson S, Hoke M, 107. Hari R, Salenius S. Rhythmical corticomuscular communication.
Stroink G, et al., eds. Advances in Biomagnetism. New York: Plenum Neuroreport. 1999;10:R1–R10.
Press; 1989:257–260. 108. Salenius S, Hari R. Synchronous oscillatory cortical activity
87. Lu ZL, Wang JZ, Williamson S. Neuronal sources of human during motor action. Curr Opin Neurobiol. 2003;13:678–684.
parieto-occipital alpha rhythm. In: Hoke M, Erne S, Okada Y, et al., 109. Mima T, Hallett M. Corticomuscular coherence: a review. J Clin
eds. Biomagnetism. Clinical Aspects. Amsterdam: Excerpta Medica; Neurophysiol. 1999;16:501–511.
1992:33–37. 110. Kilner J, Baker S, Salenius S, et al. Human cortical muscle coher-
88. Salmelin R, Hari R. Characterization of spontaneous MEG ence is directly related to specific motor parameters. J Neurosci.
rhythms in healthy adults. Electroencephalogr Clin Neurophysiol. 2000;20:8838–8845.
1994;91:237–248. 111. Kilner JM, Salenius S, Baker SN, et al. Task-dependent modula-
89. Ilmoniemi R, Williamson S, Hostetler W. Method for the study of tions of cortical oscillatory activity in human subjects during a
spontaneous brain activity. In: Atsumi K, Kotani M, Ueno M, et al., bimanual precision grip task. Neuroimage. 2003;18:67–73.
eds. Biomagnetism ’87. Tokyo: Tokyo Denki University Press; 112. Brown P, Salenius S, Rothwell JC, et al. The cortical correlate of
1987:182–185. the Piper rhythm in man. J Neurophysiol. 1998;80:2911–2917.
90. Kaufman L, Schwarz B, Salustri C, et al. Modulation of sponta- 113. Brown P. Cortical drives to human muscle: the Piper and related
neous brain activity during mental imagery. J Cogn Neurosci. rhythms. Prog Neurobiol. 2000;60:97–108.
1990;2:124–132. 114. Gross J, Timmermann L, Kujala J, et al. The neural basis of inter-
91. Michel CM, Kaufman L, Williamson SJ. Duration of EEG and mittent motor control in humans. Proc Natl Acad Sci USA.
MEG suppression increases with angle in a mental rotation 2002;99:2299–2302.
task. J Cogn Neurosci. 1994;6:139–150.
Chapter 42 ■ Magnetoencephalography: Methods and Applications 895

115. Tiihonen J, Hari R, Kajola M, et al. Magnetoencephalographic 137. Llinas R, Ribary U. Coherent 40-Hz oscillation characterizes dream
10-Hz rhythm from the human auditory cortex. Neurosci Lett. state in humans. Proc Natl Acad Sci USA. 1993;90:2078–2081.
1991;129:303–305. 138. Jäntti V, Baer G, Yli-Hankala A, et al. MEG burst suppression in
116. Lehtelä L, Salmelin R, Hari R. Evidence for reactive magnetic an anaesthesized dog. Acta Anaesth Scand. 1995;39:126–128.
10-Hz rhythm in the human auditory cortex. Neurosci Lett. 139. Cohen D. Magnetoencephalography: detection of the brain’s
1997;222:111–114. electrical activity with a superconducting magnetometer. Science.
117. Bente D. Die Insuffizienz des Vigilitätstonus. Habilitationsschrift. 1972;1175:664–666.
University of Erlangen-Nürnberg; 1964. 140. Modena I, Ricci GB, Barbanera S, et al. Biomagnetic measure-
118. Santamaria J, Chiappa K. The EEG of drowsiness in normal ments of spontaneous brain activity in epileptic patients.
adults. J Clin Neurophysiol. 1987;4:327–382. Electroencephalogr Clin Neurophysiol. 1982;54:622–628.
119. Lu ST, Kajola M, Joutsiniemi SL, et al. Generator sites of sponta- 141. Rose DF, Smith PD, Sato S. Magnetoencephalography and
neous MEG activity during sleep. Electroencephalogr Clin. epilepsy research. Science. 1987;238:329–335.
Neurophysiol. 1992;82:182–196. 142. Sutherling WW, Barth DS. Neocortical propagation in temporal
120. Niedermeyer E. Alpha-like rhythmical activity of the temporal lobe spike foci on magnetoencephalography and electroen-
lobe. Clin Electroencephalogr. 1990;21:210–224. cephalography. Ann Neurol. 1989;25:373–381.
121. Niedermeyer E. The ‘third rhythm’: further observations. Clin 143. Paetau R, Kajola M, Hari R. Magnetoencephalography in the
Electroencephalogr. 1991;22:83–96. study of epilepsy. Neurophysiol Clinique. 1990;20:169–187.
122. Narici L, Forss N, Jousmäki V, et al. Evidence for a 7- to 9-Hz ‘sigma’ 144. Paetau R, Kajola M, Karhu J, et al. Magnetoencephalographic
rhythm in the human SII cortex. Neuroimage. 2001;13:662–668. localization of epileptic cortex – impact on surgical treatment.
123. Sasaki K, Tsujimoto T, Nambu A, et al. Dynamic activities of the Ann Neurol. 1992;32:106–109.
frontal association cortex in calculating and thinking. Neurosci. 145. Paetau R, Hämäläinen M, Hari R, et al. Presurgical MEG evaluation
Res. 1994;19:229–233. of children with intractable epilepsy. Epilepsia. 1994;35:275–284.
124. Gray M, Singer W. Stimulus-specific neuronal oscillations in ori- 146. Tiihonen J, Hari R, Kajola M, et al. Localization of epileptic foci
entation columns of cat visual cortex. Proc Natl Acad Sci USA. using a large-area magnetometer and functional brain anatomy.
1989;86:1698–1702. Ann Neurol. 1990;27:283–290.
125. Llinas R. Intrinsic electrical properties of mammalian neurons 147. Ebersole JS. New applications of EEG/MEG in epilepsy evalua-
and CNS function. In: Fidia Research Foundation Neuroscience tion. Epilepsy Res Suppl. 1996;11:227–237.
Award Lectures. New York: Raven Press; 1990;4:175–194. 148. Nakasato N, Seki K, Kawamura T, et al. Cortical mapping using an
126. Tallon-Baudry C, Bertrand O, Wienbruch C, et al. Combined MRI-linked whole head MEG system and presurgical decision
EEG and MEG recordings of visual 40 Hz responses to illusory making. In: Hashimoto I, Okada Y, Ogawa S, eds. Visualization of
triangles in human. Prog Neurobiol. 2000;60:97–108. Information Processing in the Human Brain: Recent Advances in
127. Wyart V, Tallon-Baudry C. Neural dissociation between visual MEG and Functional MRI (EEG Suppl. 47). Elsevier; 1996:333–341.
awareness and spatial attention. J Neurosci. 2008;28:2667–2679. 149. Watanabe Y, Fukao K, Watanabe M, et al. Epileptic events
128. Hughes JR, Hendrix DE, Cohen J, et al. Relationship of the mag- observed by multichannel MEG. In: Hashimoto I, Okada Y,
netoencephalogram to the electroencephalogram. Normal wake Ogawa S, eds. Visualization of Information Processing in the
and sleep activity. Electroencephalogr Clin Neurophysiol. 1976;40: Human Brain: Recent Advances in MEG and Functional MRI (EEG
261–278. Suppl. 47). Elsevier; 1996:283–391.
129. Nakasato N, Kado H, Nakanishi M, et al. Magnetic detection of 150. Merlet I, Paetau R, Garcia-Larrea L, et al. Apparent asynchrony
sleep spindles in normal subjects. Electroencephalogr Clin between interictal electric and magnetic spikes. Neuroreport.
Neurophysiol. 1990;76:123–130. 1997;8:1971–1076.
130. Ueno S, Iramina K. Modeling and source localization of MEG 151. Lamusuo S, Forss N, Ruottinen H-M, et al. [ 18F]FDG-PET and
activities. Brain Topogr. 1990;3:151–165. whole-scalp MEG localization of epileptogenic cortex. Epilepsia.
131. Manshanden I, De Munck JC, Simon NR, et al. Source localiza- 1999;40:921–930.
tion of MEG sleep spindles and the relation to sources of alpha 152. Baumgartner C, Pataraia E, Lindinger G, et al. Magneto-encephalog-
band rhythms. Clin Neurophysiol. 2002;113:1937–1947. raphy in focal epilepsy. Epilepsia. 2001;41 (suppl 3): S39–S47.
132. Numminen J, Mäkelä J, Hari R. Distribution and sources of mag- 153. Iwasaki M, Nakasato N, Shamoto H, et al. Surgical implications
netoencephalographic K-complexes. Electroencephalogr Clin of neuromagnetic spike localization in temporal lobe epilepsy.
Neurophysiol. 1996;99:544–555. Epilepsia. 2002;43:415–424.
133. Buser P. Thalamocortical mechanisms underlying synchronized 154. Barkley GL, Baumgartner C. MEG and EEG in Epilepsy. J Clin
EEG activity. In: Halliday A, Butler S, Paul R, eds. A Textbook of Neurophysiol. 2003;20:163–178.
Clinical Neurophysiology. Chichester: Wiley; 1987:595–621. 155. Knowlton RC. Magnetoencephalography: clinical application in
134. Niedermeyer E. Depth electroencephalography. In: Niedermeyer epilepsy. Curr Neurol Neurosci Rep. 2003;3:341–348.
E, Lopes da Silva F, eds. Electroencephalography. Basic Principles, 156. LinY-Y, Chang K-P, Hsieh J-C, et al. Magnetoencephalographic
Clinical Applications and Related Fields. Baltimore-Munich: analysis of bilaterally synchronous discharges in benign rolandic
Urban & Schwarzenberg; 1982:519–536. epilepsy of childhood. Seizure. 2003;12:448–455.
135. Scheuler W, Kubicki S, Scholz G, et al. Two different activities in 157. Van’t Ent D, Manshanden I, Ossenblok P, et al. Spike cluster analysis
the sleep spindle frequency band-discrimination based on the in neocortical localization related epilepsy yields clinically signifi-
topographical distribution of spectral power and coherence. In: cant equivalent source localization results in magnetoencephalo-
Horne J, ed. Sleep ’90. Bochum: Pontenagel Press; 1990:13–16. gram (MEG). Clin Neurophysiol. 2003;114:1948–1962.
136. Ishii R, Dziewas R, Chau W, et al. Current source density distri- 158. Hari R, Ahonen A, Forss N, et al. Parietal epileptic mirror focus
bution of sleep spindles in humans as found by synthetic aper- detected with a whole-head neuromagnetometer. Neuroreport.
ture magnetometry. Neurosci Lett. 2003;340:25–28. 1993;5:45–48.
896 Part V ■ Complementary and Special Techniques

159. Forss N, Jousmäki V, Hari R. Interaction in the human 180. Timmermann L, Gross J, Dirks M, et al. The cerebral oscillatory
somatosensory cortex between afferent input from different network of parkinsonian resting tremor. Brain. 2003;126:199–212.
fingers. Brain Res. 1995;685:68–76. 181. Schnitzler A, Timmermann L, Gross J. Physiological and patho-
160. Paetau R, Kajola M, Korkman M, et al. Landau-Kleffner syn- logical oscillatory networks in the human motor system. J Physiol
drome: epileptic activity in the auditory cortex. Neuroreport. Paris. 2005;99:3–7.
1991;2:201–204. 182. Elberling C, Bak C, Kofoed B, et al. Magnetic auditory responses
161. Paetau R, Granström ML, Blomstedt G, et al. Magnetoence- from the human brain. A preliminary report. Scand Audiol.
phalography in presurgical evaluation of children with the 1980;9:185–190.
Landau-Kleffner syndrome. Epilepsia. 1999;40:326–335. 183. Hari R, Aittoniemi K, Järvinen ML, et al. Auditory evoked tran-
162. Paetau R. Sounds trigger spikes in the Landau-Kleffner sient and sustained magnetic fields of the human brain.
syndrome. J Clin Neurophysiol. 1994;11:231–241. Localization of neural generators. Exp Brain Res. 1980;40:237–240.
163. Morrell F, Whisler WW, Smith MC, et al. Landau-Kleffner 184. Kuriki S, Nogai T, Hirata Y. Cortical sources of middle latency
syndrome. Treatment with subpial intracortical transection. responses of auditory evoked magnetic field. Hering Res.
Brain. 1995;118:1529–1546. 1995;92:47–51.
164. Lau M, Yam D, Burneo JG. A systematic review on MEG and its 185. Pelizzone M, Hari R, Mäkelä JP, et al. Cortical origin of middle-
use in the presurgical evaluation of localization-related epilepsy. latency auditory evoked responses in man. Neurosci Lett. 1987;82:
Epilepsy Res. 2008;79:97–104. 303–307.
165. Lewine JD. Commentary on Lau et al. A systematic review on 186. Mäkelä JP, Hämäläinen M, Hari R, et al. Whole-head mapping of
MEG and its use in the presurgical evaluation of localization middle-latency auditory evoked fields. Electroencephalogr Clin
related epilepsy. Epilepsy Res. 2008;82:235–236. Neurophysiol. 1994;92:414–421.
166. Schwartz ES, Dlugos DJ, Storm PB, et al. Magnetoence- 187. McEvoy L, Mäkelä JP, Hämäläinen M, et al. Effect of interaural
phalography for pediatric epilepsy: how we do it. Am time differences on middle-latency and late auditory evoked
J Neuroradiol. 2008;29:832–837. magnetic fields. Hear Res. 1994;78:249–257.
167. Vieth J, Sack G, Schueler P, et al. Ischemic and epileptic lesions 188. Yoshiura Y, Ueno S, Iramina K, et al. Effects of stimulation site on
measured by AC- and DC-MEG. In: Williamson S, Hoke M, human middle latency auditory evoked magnetic fields. Neurosci
Stroink G, et al., eds. Advances in Biomagnetism. New York: Lett. 1994;172:159–162.
Plenum Press; 1989:307–310. 189. Nishitani N, Nagamine T, Fujiwara N, et al. Cortical-hippocampal
168. Gallen C, Schwartz B, Pantev C, et al. Detection and localization auditory processing identified by magnetoencephalography.
of delta frequency activity in human stroke. In: Hoke M, Erné J Cogn Neurosci. 1998;10:231–247.
SN, Okada YC, et al., eds. Biomagnetism. Clinical Aspects. 190. Hari R, Kaila K, Katila T, et al. Interstimulus-interval dependence
Amsterdam: Excerpta Medica; 1992;301–305. of the auditory vertex response and its magnetic counterpart:
169. Mäkelä JP. Neurological application of MEG. In: Hashimoto I, Implications for their neural generation. Electroencephalogr Clin
Okada Y, Ogawa S, eds. Visualization of Information Processing in Neurophysiol. 1982;54:561–569.
the Human Brain: Recent Advances in MEG and Functional MRI 191. Hari R, Pelizzone M, Mäkelä JP, et al. Neuromagnetic responses
(EEG Suppl. 47). Elsevier; 1996;343–355. of the human auditory cortex to on- and offsets of noise bursts.
170. Mäkelä JP, Salmelin R, Kotila M, et al. Modification of neuromag- Audiology. 1987;25:31–43.
netic cortical signals by thalamic infarctions. Electroencephalogr 192. Lu ZL, Williamson S, Kaufman L. Behavioral lifetime of human
Clin Neurophysiol. 1998;106:433–443. auditory sensory memory predicted by physiological measures.
171. Paetau R, Santavuori P, Vanhanen S-L, et al. unpublished obser- Science. 1992;258:1668–1670.
vations, 1991 193. Mäkelä JP, Ahonen A, Hämäläinen M, et al. Functional differ-
172. Okada Y. Magnetoencephalography as a noninvasive tool for ences between auditory cortices of the two hemispheres revealed
electrophysiological characterization of auras in classic migraine. by whole-head neuromagnetic recordings. Hum Brain Mapp.
In: Sato S, ed. Advances in Neurology, Vol 54: Magnetoence- 1993;1:48–56.
phalography. New York: Raven Press; 1990:133–140. 194. Sams M, Hari R, Rif J, et al. The human auditory sensory mem-
173. Barkley G, Moran J, Takanashi Y, et al. Techniques for DC ory trace persists about 10 s: neuromagnetic evidence. J Cogn
magnetoencephalography. J Clin Neurophysiol. 1991;8:189–199. Neurosci. 1993;5:363–370.
174. Hadjikhani N, Sanchez del Rio M, Wu O, et al. Mechanisms of 195. Pantev C, Hoke M, Lehnertz K, et al. Tonotopic organization of
migraine aura revealed by functional MRI in human visual the human auditory cortex revealed by transient auditory evoked
cortex. Proc Natl Acad Sci USA. 2001;98:4687–4692. magnetic fields. Electroencephalogr Clin Neurophysiol. 1988;69:
175. Heikman P, Salmelin R, Mäkelä J, et al. Relation between the 160–170.
frontal 3–7 Hz MEG activity and the efficacy of ECT in major 196. Pantev C, Hoke K, Lehnertz K, et al. Neuromagnetic evidence of
depression. J ECT. 2001;17:136–140. an amplitopic organization of the human auditory cortex.
176. Mäkelä JP, Salmelin R, Hokkanen L, et al. Neuromagnetic seque- Electroencephalogr Clin Neurophysiol. 1989;72:225–231.
lae of herpes simplex encephalitis. Electroencephalogr Clin 197. Lütkenhöner B, Krumbholtz K, Seither-Preisler A. Studies of
Neurophysiol. 1998;106:251–258. tonotopy based on wave N100 of the auditory evoked field are
177. Mäkelä JP, Hari R, Karhu J, et al. Suppression of magnetic mu problematic. NeuroImage. 2003;9:935–949.
rhythm during Parkinsonian tremor. Brain Res. 1993;617:189–193. 198. Mäkelä JP, Hari R, Linnankivi A. Different analysis of frequency
178. Salenius S, Avikainen S, Kaakkola S, et al. Defective cortical drive and amplitude modulations of a continuous tone in the human
to muscle in Parkinson’s disease and its improvement with auditory cortex: A neuromagnetic study. Hear Res. 1987;27:257–
levodopa. Brain. 2002;125:491–500. 264.
179. Brown P, Marsden J, Defebvre L, et al. Intermuscular coherence 199. Kaukoranta E, Hari R, Lounasmaa OV. Responses of the human
in Parkinson’s disease: relationship to bradykinesia. Neuroreport. auditory cortex to vowel onset after fricative consonants. Exp
2001;12:2577–2581. Brain Res. 1987;69:19–23.
Chapter 42 ■ Magnetoencephalography: Methods and Applications 897

200. Mäkelä JP, Hari R, Leinonen L. Magnetic responses of the human ear to the human auditory cortex during binaural hearing.
auditory cortex to noise/tone-transitions. Electroencephalogr Clin J Neurosci. 2002;22:RC205.
Neurophysiol. 1988;69:423–430. 223. Kaneko K, Fujiki N, Hari R. Binaural interaction in the human
201. Hari R, Hämäläinen M, Kaukoranta E, et al. Selective listening auditory cortex revealed by neuromagnetic frequency-tagging:
modifies activity of the human auditory cortex. Exp Brain Res. no effect of stimulus intensity. Hearing Res. 2003;183:1–6.
1989;74:463–470. 224. Leinonen L, Joutsiniemi SL. Auditory evoked potentials and
202. Arthur D, Lewis P, Medwick P, et al. A neuromagnetic study of magnetic fields in patients with lesions of the auditory cortex.
selective auditory attention. Electroenceph. Clin Neurophysiol. Acta Neurol Scand. 224;79:316–325.
1991;78:348–360. 225. Mäkelä JP, Hari R, Valanne L, et al. Auditory evoked magnetic
203. Kuriki S, Takeuchi F. Neuromagnetic responses elicited by fields after ischemic brain lesions. Ann Neurol. 1991;30:76–82.
auditory stimuli in dichotic listening. Electroencephalogr Clin 226. Mäkelä JP, Hari R. Neuromagnetic auditory evoked responses after
Neurophysiol. 1991;80:406–411. a stroke in the right temporal lobe. Neuroreport. 1992;3:94–96.
204. Rif J, Hari R, Hämäläinen M, et al. Auditory attention affects two 227. Pelizzone M, Hari R, Mäkelä J, et al. Cortical activity evoked by a
different areas in the human auditory cortex. Electroencephalogr multichannel cochlear prosthesis. Acta Oto-laryngol. 1987;103:
Clin Neurophysiol. 1991;79:464–472. 632–636.
205. McEvoy L, Hari R, Imada T, et al. Human auditory cortical mech- 228. Hari R, Pelizzone M, Mäkelä J, et al. Neuromagnetic responses
anisms of sound lateralization: II. Interaural time differences at from a deaf subject to stimuli presented through a multichannel
sound onset. Hear Res. 1993;67:98–109. cochlear prosthesis. Ear Hear. 1988;9:148–152.
206. Sams M, Hämäläinen M, Hari R, et al. Human auditory cortical 229. Hoke M, Pantev C, Lütkenhöner B, et al. Magnetic fields from the
mechanisms of sound lateralization: I. Interaural time differences auditory cortex of a deaf human individual occurring sponta-
within sound. Hear Res. 1993;67:89–97. neously or evoked by stimulation through a cochlear prosthesis.
207. Näätänen R, Picton T. The N1 wave of the human electric and Audiology. 1989;28:152–170.
magnetic response to sound: a review and analysis of the compo- 230. Hari R. Neuromagnetic approach to human auditory cortical
nent structure. Psychophysiology. 1987;24:375–425. functions, with emphasis on subjects with cochlear implants. In:
208. Näätänen R. Attention and Brain Function. Hillsdale, New Jersey: Honjo I, Takahashi H, eds. Cochlear Implant and Related Sciences
Erlbaum; 1992. Update Adv. Otorhinolaryngol. Vol. 52. Basel: Karger; 1997:15–18.
209. Näätänen R. Mismatch negativity: clinical research and possible 231. Vasama JP, Mäkelä JP, Parkkonen L, et al. Auditory cortical
applications. Int J Psychophysiol. 2003;48:179–188. responses in humans with congential unilateral conductive hear-
210. Alho K. Cerebral generators of mismatch negativity (MMN) and ing loss. Hear Res. 1994;78:91–97.
its magnetic counterpart (MMNm) elicited by sound changes. 232. Vasama JP, Mäkelä JP, Pyykkö I, et al. Modification of central
Ear Hear. 1995;16:38–51. auditory pathways after unilateral permanent deafness due to
211. Raij T, McEvoy L, Mäkelä J, et al. Human auditory cortex is activated removal of acoustic neuroma. Neuroreport. 1994;6:961–964.
by omissions of auditory stimuli. Brain Res. 1997;745:134–143. 233. Vasama JP, Mäkelä JP. Auditory pathway plasticity in adult
212. Sams M, Aulanko R, Hämäläinen M, et al. Seeing speech: visual humans after idiopathic sudden unilateral sensorineural hearing
information from lip movements modifies activity in the human loss. Hear Res. 1995;87:132–140.
auditory cortex. Neurosci Lett. 1991;127:141–145. 234. Pekkonen E, Huotilainen M, Virtanen J, et al. Alzheimer’s disease
213. Calvert GA, Bullmore ET, Brammer MJ, et al. Activation of audi- affects parallel processing between the auditory cortices.
tory cortex during silent lipreading. Science. 1997;276:593–596. Neuroreport. 1996;7:1365–1368.
214. Romani GL, Williamson SJ, Kaufman L. Tonotopic organization 235. Tiihonen J, Hari R, Naukkarinen H, et al. Auditory hallucinations
of the human auditory cortex. Science. 1982;216:1339–1340. may modify activity of the human auditory cortex. Am
215. Galambos R, Makeig S, Talmachoff PJ. A 40-Hz auditory poten- J Psychiatry. 1992;149:255–257.
tial recorded from the human scalp. Proc NY Acad Sci. 1981;78: 236. Hoke M, Feldman H, Pantev C, et al. Objective evidence of tinni-
2643–2647. tus in auditory evoke magnetic fields. Hear Res. 1989;37:281–286.
216. Mäkelä J, Hari R. Evidence for cortical origin of the 40-Hz audi- 237. Pantev C, Hoke M, Lütkenhöner B, et al. Tinnitus remission
tory evoked response in man. Electroencephalogr Clin objectified by neuromagnetic measurements. Hear Res. 1989;40:
Neurophysiol. 1987;66:539–546. 261–264.
217. Ross B, Picton TW, Pantev C. Temporal integration in the human 238. Jacobson G, Ahmad B, Moran J, et al. Auditory evoked cortical
auditory cortex as represented by the development of the steady- magnetic field (M100/M200) measurements in tinnitus and nor-
state magnetic field. Hear Res. 2002;165:68–84. mal groups. Hear Res. 1991;56:44–52.
218. Hari R, Hämäläinen M, Joutsiniemi SL. Neuromagnetic steady- 239. Colding-Jørgensen E, Lauritzen M, Johnsen NJ, et al. On the evi-
state responses to auditory stimuli. J Acoust Soc Am. dence of auditory evoked magnetic fields as an objective measure
1989;86:1033–1039. of tinnitus. Electroencephalogr Clin Neurophysiol. 1992;83:
219. Gutschalk A, Mase R, Roth R, et al. Deconvolution of 40 Hz 322–327.
steady-state fields reveals two overlapping source activities of the 240. Shiomi Y, Nagamine T, Fujiki N, et al. Tinnitus remission by lido-
human auditory cortex. Clin Neurophysiol. 1999;110:856–868. caine demonstrated by auditory-evoked magnetoencephalo-
220. Pastor MA, Artieda J, Arbizu J, et al. Activation of human cerebral gram. Acta Otolaryngol (Stockh.) 1997;117:31–34.
and cerebellar cortex by auditory stimulation at 40 Hz. 241. Muhlnickel W, Elbert T, Taub E, et al. Reorganization of auditory
J Neurosci. 2002;22:10501–10506. cortex in tinnitus. Proc Natl Acad Sci USA. 1998;95:10340–10343.
221. Joliot M, Ribary M, Llinas R. Human oscillatory brain activity 242. Nagarajan S, Mahncke H, Salz T, et al. Cortical auditory signal
near 40 Hz coexists with cognitive temporal binding. Proc Natl processing in poor readers. Proc Natl Acad Sci USA. 1999;96:
Acad Sci USA. 1994;91:11748–11751. 6483–6488.
222. Fujiki N, Jousmäki V, Hari R. Neuromagnetic responses to fre- 243. Renvall H, Hari R. Diminished auditory mismatch fields in
quency-tagged sounds: a new method to follow inputs from each dyslexic adults. Ann Neurol. 2003;53:551–557.
898 Part V ■ Complementary and Special Techniques

244. Gootjes L, Raij T, Salmelin R, et al. Left-hemisphere dominance 263. Forss N, Salmelin R, Hari R. Comparison of somatosensory
for processing of vowels: a whole-scalp neuromagnetic study. evoked fields to airpuff and electric stimuli. Electroencephalogr
Neuroreport. 1999;10:2987–2991. Clin Neurophysiol. 1994;92:510–517.
245. Brenner D, Lipton J, Kaufman L, et al. Somatically evoked mag- 264. Del Gratta C, Della Penna S, Ferretti A, et al. Topographic organ-
netic fields of the human brain. Science. 1978;199:81–83. ization of the human primary and secondary somatosensory cor-
246. Suk J, Ribary U, Cappell J, et al. Anatomical localization revealed tices: comparison of fMRI and MEG findings. NeuroImage.
by MEG recordings of the human somatosensory system. 2002;17:1373–1383.
Electroencephalogr Clin Neurophysiol. 1991;78:185–196. 265. Tuunanen PI, Kavec M, Jousmäki V, et al. Comparison of BOLD
247. Yang TT, Gallen CC, Schwartz BJ, et al. Noninvasive fMRI and MEG characteristics to vibrotactile stimulation.
somatosensory homunculus mapping in humans by using a Neuroimage. 2003;19:1778–1786.
large-array biomagnetometer. Proc Natl Acad Sci USA. 1993;90: 266. Simoes C, Mertens M, Forss N, et al. Functional overlap of finger
3098–3102. representations in human SI and SII cortices. J Neurophysiol.
248. Huttunen J, Kaukoranta E, Hari R. Cerebral magnetic responses 2001;86:1661–1665.
to stimulation of tibial and sural nerves. J Neurol Sci. 1987;79: 267. Simões C, Alary F, Forss N, et al. Left-hemisphere-dominant SII
43–54. activation after bilateral median nerve stimulation. Neuroimage.
249. Fujita S, Nakasato N, Matani A, et al. Short latency somatosen- 2002;15:686–690.
sory evoked field for tibial nerve stimulation: rotation of dipole 268. Huttunen J, Wikström H, Korvenoja A, et al. Significance of the
pattern over the whole head. In: Baumgartner C, Deecke L, second somatosensory cortex in sensorimotor integration:
Stroink G, et al., eds. Biomagnetism: Fundamental Research and enhancement of sensory responses during finger movements.
Clinical Applications. Amsterdam: Elsevier; 1995:95–98. Neuroreport. 1996;7:1009–1012.
250. Kakigi R, Koyama S, Hoshiyama M, et al. Topography of 269. Forss N, Hietanen M, Hari R. Modified activation of somatosen-
somatosensory evoked magnetic fields following posterior tibial sory cortical network in patients with right hemisphere stroke.
nerve stimulation. Electroencephalogr Clin. Neurophysiol 1995;95: Brain. 1999;122:1989–1899.
127–134. 270. Simões C, Jensen O, Parkkonen L, et al. Phase locking between
251. Hari R, Nagamine T, Nishitani N, et al. Time-varying activation human primary and secondary somatosensory cortices. Proc Natl
of different cytoarchitectonic areas of the human SI cortex after Acad Sci USA. 2003;100:2691–2694.
tibial nerve stimulation. Neuroimage. 1996;4:111–118. 271. Karhu J, Hari R, Mäkelä J, et al. Somatosensory evoked magnetic
252. Hashimoto I, Sakuma K, Kimura T, et al. Serial activation of dis- fields in multiple sclerosis. Electroenceph Clin Neurophysiol.
tinct cytoarchitectonic areas of the human SI cortex after poste- 1992;83:192–200.
rior tibial nerve stimulation. Neuroreport. 2001;12:1857–1862. 272. Karhu J, Hari R, Paetau R, et al. Cortical reactivity in progressive
253. Schnitzler A, Salmelin R, Salenius S, et al. Tactile information myoclonus epilepsy. Electroenceph Clin Neurophysiol.
from the human hand reaches the ipsilateral primary somatosen- 1994;90:93–102.
sory cortex. Neurosci Lett. 1995;200:25–28. 273. Lauronen L, Heikkilä E, Autti T, et al. Somatosensory evoked
254. Korvenoja A, Wikström H, Huttunen J, et al. Activation of ipsilat- magnetic fields from primary sensorimotor cortex in juvenile
eral primary sensorimotor cortex by median nerve stimulation. neuronal ceroid lipofuscinosis. J Child Neurol. 1997;12:
Neuroreport. 1995;6:2589–2593. 355–360.
255. Hari R, Imada T. Ipsilateral movement-evoked fields (MEFs) 274. Lauronen L, Huttunen J, Kirveskari E, et al. Enlarged SI and SII
reconsidered. Neuroimage. 1999;10:582–588. somatosensory evoked responses in the CLN5 form of neuronal
256. Lauronen L, Nevalainen P, Wikström H, et al. Immaturity of ceroid lipofuscinosis. Clin Neurophysiol. 2002;113:1491–1500.
somatosensory cortical processing in human newborns. 275. Forss N, Silén T, Karjalainen T. Lack of activation of human sec-
Neuroimage. 2006;33:195–203. ondary somatosensory cortex in Unverricht-Lundborg type of
257. Curio G, Mackert B, Burghoff M, et al. Localization of evoked progressive myoclonus epilepsy. Ann Neurol. 2001;49:90–97.
neuromagnetic 600 Hz activity in the cerebral somato - 276. Hari R, Kaukoranta E, Reinikainen K, et al. Neuromagnetic local-
sensory system. Electroencephalogr Clin Neurophysiol. 1994;91: ization of cortical activity evoked by painful dental stimulation in
483–487. man. Neurosci Lett. 1983;42:77–82.
258.Hashimoto I, Mashiko T, Imada T. Somatic evoked high-frequency 277. Huttunen J, Kobal G, Kaukoranta E, et al. Cortical responses to
magnetic oscillations reflect activity of inhibitory interneurons in painful CO2 stimulation of the nasal mucosa. Electroencephalogr
the human somatosensory cortex. Electroencephalogr Clin Clin Neurophysiol. 1986;64:347–349.
Neurophysiol. 1996;100:189–203. 278. Joseph J, Howland E, Wakai R, et al. Late pain-related magnetic
259. Elbert T, Pantev C, Wienbruch C, et al. Increased cortical repre- fields and electric potentials evoked by intracutaneous electric
sentation of the fingers of the left hand in string players. Science. finger stimulation. Electroencephalogr Clin Neurophysiol.
1995;270:305–307. 1991;80:46–52.
260. Pantev C, Engelien A, Candia V, et al. Representational cortex in 279. Kakigi R, Koyama S, Hoshiyama M, et al. Pain-related magnetic
musicians. Plastic alterations in response to musical practice. Ann fields following painful CO2 laser stimulation in man. Neurosci
NY Acad Sci. 2001;930:300–314. Lett. 1995;192:45–48.
261. Hari R, Hämäläinen M, Kaukoranta E, et al. Neuromagnetic 280. Kakigi R, Tran TD, Qiu Y, et al. Cerebral responses following
responses from the second somatosensory cortex in man. Acta stimulation of unmyelinated C-fibers in humans: electro- and
Neurol Scand. 1983;68:207–212. magnetoencephalographic study. Neurosci Res. 2003;45:255–275.
262. Hari R, Hämäläinen H, Tiihonen J, et al. Separate finger repre- 281. Ploner M, Schmitz F, Freund HJ, et al. Parallel activation of
sentations at the human second somatosensory cortex. primary and secondary somatosensory cortices in human pain
Neuroscience. 1990;37:245–249. processing. J Neurophysiol. 1999;81:3100–3104.
Chapter 42 ■ Magnetoencephalography: Methods and Applications 899

282. Kanda M, Nagamine T, Ikeda A, et al. Primary somatosensory 304. Uusitalo M, Virsu V, Salenius S, et al. Human cortical activation
cortex is actively involved in pain processing in human. Brain related to perception of visual motion and movement after-
Res. 2000;853:282–289. effect. Neuroimage. 1997;5:241–250.
283. Forss N, Raij T, Seppä M, et al. Common cortical network for first 305. Vanni S, Tanskanen T, Seppä M, et al. Coinciding early activation
and second pain. Neuroimage. 2005;24:132–142. of human primary visual cortex and anteromedial cuneus. Proc
284. Hari R, Portin K, Kettenmann B, et al. Right-hemisphere prepon- Natl Acad Sci USA. 2001;98:2776–2780.
derance of responses to painful CO2 stimulation of the human 306. Schmolesky MT, Wang Y, Hanes DP, et al. Signal timing across the
nasal mucosa. Pain. 1997;72:145–151. macaque visual system. J Neurophysiol. 1998;79:3272–3278.
285. Bragard D, Chen AC, Plaghki L. Direct isolation of ultra-late (C- 307. Petroni F, Panzeri S, Hilgetag CC, et al. Simultaneity of responses
fibre) evoked brain potentials by CO2 laser stimulation of tiny in a hierarchical visual network. Neuroreport. 2001;12:2753–2759.
cutaneous surface areas in man. Neurosci Lett. 1996;209:81–84. 308. Lu S, Hämäläinen M, Hari R, et al. Seeing faces activates three
286. Hari R, Forss N, Raij TT. Neuromagnetic exploration of the brain areas outside the occipital visual cortex in man.
connection between pain and the motor cortex. In: Kalso E, Neuroscience. 1991;43:287–290.
Estlander A-M, Klockars M, eds. Psyche, Soma and Pain. Acta 309. Sams M, Hietanen J, Hari R, et al. Face-specific responses from
Gyllenbergiana IV. Helsinki: The Signe and Ane Gyllenberg the human inferior occipitotemporal cortex. Neuroscience.
Foundation; 2003:145–153. 1997;77:49–55.
287. Raij TT, Forss N, Stancak A, et al. Noxious input activates the 310. Linkenkaer-Hansen K, Palva JM, Sams M, et al. Face-selective
human motor cortex: implications for tension-type pain? Soc processing in human extrastriate cortex around 120 ms after
Neurosci Ann Meet. 2003;238:3. stimulus onset revealed by magneto- and electroencephalogra-
288. Raij TT, Forss N, Stancak A, et al. Modulation of motor-cortex phy. Neurosci Lett. 1998;253:147–50.
oscillatory activity by painful A - and C-fiber stimuli. 311. Halgren E, Raij T, Marinkovic K, et al. Cognitive response profile
Neuroimage. 2004;23:569–573. of the human fusiform area as determined by MEG. Cereb
289. Juottonen K, Gockel M, Silén T, et al. Altered central sensorimo- Cortex. 2000;10:69–81.
tor processing in patients with complex regional pain syndrome. 312. Liu J, Harris A, Kanwisher N. Stages of processing in face percep-
Pain. 2002;98:315–323. tion: an MEG study. Nat Neurosci. 2002;5:910–916.
290. Forss N, Kirveskari E, Gockel M. Mirror-like spread of chronic 313. Salmelin R, Hari R, Lounasmaa OV, Sams M. Dynamics of brain
pain. Neurology. 2005;65:748–750. activation during picture naming. Nature. 1994;368:463–465.
291. Brenner D, Williamson SJ, Kaufman L. Visually evoked magnetic 314. Maess B, Koelsch S, Gunter TC, et al. Musical syntax is processed
fields of the human brain. Science. 1975;190:480–481. in Broca’s area: an MEG study. Nat Neurosci. 2001;4:540–545.
292. Teyler TJ, Cuffin BN, Cohen D. The visual magnetoencephalo- 315. Cornelissen P, Tarkiainen A, Helenius P, et al. Cortical effects of
gram. Life Sci. 1975;17:683–692. shifting letter position in letter strings of varying length. J Cogn
293. Maclin E, Okada Y, Kaufman L, et al. Retinotopic map on the Neurosci. 2003;15:731–746.
visual cortex for eccentrically placed patterns: first noninvasive 316. Cornelissen K, Laine M, Tarkiainen A, et al. Adult brain plasticity
measurement. Nuovo Cimento C. 1983;2D:410–419. elicited by anomia treatment. J Cogn Neurosci. 2003;15:444–461.
294. Ahlfors S, Ilmoniemi R, Hämäläinen M. Estimates of visually 317.Salmelin R, Schnitzler A, Schmitz F, et al. Single word reading in devel-
evoked cortical currents. Electroencephalogr Clin Neurophysiol. opmental stutterers and fluent speakers. Brain. 2000;123:1184–202.
1992;82:225–236. 318. Salmelin R, Service E, Kiesilä P, et al. Impaired visual word pro-
295. Aine C, Supek S, George J, et al. Retinotopic organization of cessing in dyslexia revealed with magnetoencephalography. Ann
human visual cortex: Departures from the classical model. Cereb Neurol. 1996;40:157–162.
Cortex. 1996;6:354–361. 319. Salmelin R, Helenius P, Service E. Neurophysiology of fluent and
296. Tabuchi H, Yokoyama T, Shimogawara M, et al. Study of the impaired reading: a magnetoencephalographic approach. J Clin
visual evoked magnetic field with the m-sequence technique. Neurophysiol. 2000;17:163–174.
Invest Ophthalmol Vis Sci. 2002;43:2045–2054. 320. Helenius P, Salmelin R, Service E, et al. Semantic cortical activa-
297. Hari R, Salmelin R, Tissari S, et al. Visual stability during eye- tion in dyslexic readers. J Cogn Neurosci. 1999;11:535–550.
blinks. Nature. 1994;367:121–122. 321.Salmelin R, Kujala J. Neural representation of language: activation
298. Bristow D, Frith C, Rees G. Two distinct neural effects of blinking versus long-range connectivity. Trends Cogn Sci. 2006;10:519–525.
on human visual processing. Neuroimage. 2005;27:136–145. 322. Imada T, Zhang Y, Cheour M, et al. Infant speech perception acti-
299. Jousmäki V, Hämäläinen M Hari R. Magnetic source imaging vates Broca’s area: a developmental magnetoencephalography
during a visually guided task. Neuroreport. 1996;7:2961–2964. study. Neuroreport. 2006;17:957–962.
300. Galletti C, Battaglini P, Fattori P. Eye position influence on the 323. Tesche CD, Karhu J. Somatosensory evoked magnetic fields aris-
parieto-occipital area PO (V6) of the macaque monkey. Eur ing from sources in the human cerebellum. Brain Res.
J Neurosci. 1995;7:2486–2501. 1997;744:23–31.
301. Ffytche DH, Guy CN, Zeki S. The parallel visual motion inputs 324. Tesche CD, Karhu JY. Anticipatory cerebellar response during
into areas V1 and V5 of human cerebral cortex. Brain. somatosensory omission in man. Hum Brain Mapp. 2000;9:
1995;118:1375–1394. 119–142.
302. Anderson SJ, Holliday IE, Singh KD, Harding GFA. Localization 325. Parkkonen L, Andersson J, Hämäläinen M, et al. Early visual areas
and functional analysis of human cortical area V5 using magneto reflect the percept of an ambiguous scene. Proc Nat Acad Sci USA.
encephalography. Proc R Soc Lond B Biol Sci. 1996;263: 423–431. 2008;105:20500–20504.
303. Uusitalo M, JousmäkiV, Hari R. Activation trace lifetime of 326. Rizzolatti G, Fadiga L, Gallese V, et al. Premotor cortex and the
human cortical responses evoked by apparent visual motion. recognition of motor actions. Brain Res Cogn Brain Res.
Neurosci Lett. 1997;224:45–48. 1996;3:131–141.
900 Part V ■ Complementary and Special Techniques

327. Rizzolatti G, Fogassi L, Gallese V. Neurophysiological mecha- 331. Hari R, Nishitani N. From viewing of movement to imitation and
nisms underlying the understanding and imitation of action. Nat understanding of other persons’ acts: MEG studies of the human
Rev Neurosci. 2001;2:661–670. mirror-neuron system. In: Kanwisher N, Duncan J, eds.
328. Nishitani N, Hari R. Temporal dynamics of cortical representa- Functional Neuroimaging of Visual Cognition. Attention and
tion for action. Proc Natl Acad Sci USA. 2000;97:913–918. Performance XX. Oxford: Oxford University Press; 2004:463–479.
329. Nishitani N, Hari R. Viewing lip forms: cortical dynamics. 332. Hari R, Kujala MV. Brain basis of human social interaction. From
Neuron. 2002;36:1211–1220. concepts to brain imaging. Physiol Reviews. 2009;89:453–479.
330. Caetano G, Jousmäki V, Hari R. Actor’s and viewer’s primary 333. Hari R, Parkkonen L, Nangini C. The brain in time: Insights from
motor cortices stabilize similarly after seen or heard motor neuromagnetic recordings. Ann NY Acad Sci. 2010;1191:89–109.
actions. Proc Natl Acad Sci USA. 2007;104:9058–9062.
Part VI EEG: Neuropharmacology and Anesthesia
CHAPTER
EEG, Drug Effects, and
Central Nervous System Poisoning
GERHARD BAUER AND RICHARD BAUER
43
A
large number of patients examined in an electroen- observed. At the deepest level, EEG activity ceases. The burst-
cephalographic (EEG) laboratory are under treatment suppression pattern exhibits no differences to those seen with
with drugs that alter EEG activity. Therefore, it is deep comatose states under pathologic circumstances (see
important to know changes due to the particular drug(s) used, Chapter 23) or intoxication with sedative drugs (see below).
chronic overdosage, the patterns of overt intoxication, and
withdrawal effects. The abundant literature of the topic makes
a selection inevitable. This chapter discusses EEG findings of HYPNOTICS AND SEDATIVES
drugs most frequently used in modern medicine, provides an
Barbiturates
overview of toxic encephalopathies of clinical importance, and
concentrates on visual EEG reading. Pharmacogenomics of Barbiturates remained on the market only as anesthetics and
drug actions and toxicities are not addressed in detail (for sum- antiepileptic drugs, especially in developing countries (8). They
mary see Ref. 1). apparently act at all levels of the neuraxis by binding to a specific
Superimposed fast frequencies (Fig. 43.1) are suggestive for site on the -aminobutyric acid (GABAA) receptor, different
sedating drugs. Otherwise, EEG abnormalities due to substance from the binding site of benzodiazepines (9). Low blood concen-
intake are unspecific. Diffuse slow activities and coma patterns trations produce inhibition of higher cortical functions and dis-
in cases of severe intoxication cannot be distinguished from inhibition of more primitive behavior. With increasing blood
those seen with other etiologies. This is also true for epilepti- levels, clinical signs are determined by generalized inhibition.
form activities and triphasic waves regardless whether they are
encountered in intoxicated patients with or without acute EEG Changes with Therapeutic Doses
symptomatic seizures. With localized EEG abnormalities an When administered in small doses, the barbiturates produce an
additional focal lesion should be excluded (2). increase in fast activities in the range of 25 to 35 cycles/sec (Hz),
soon shifting to 15 to 25 Hz. This activity is predominant over
DRUGS ACTING ON THE
CENTRAL NERVOUS SYSTEM
General Anesthetics
Molecular and cellular mechanisms of general anesthesia have
been reviewed by Franks and Lieb (3) and Campagna et al. (4).
Anesthetics exert their effects primarily at the neuronal ion chan-
nels via neurotransmitter receptors and by binding directly to the
protein site. Initially, anesthetics induce amnesia, euphoria, anal-
gesia, excitation, and hyperreflexia. Surgical anesthesia consists of
deep sedation, muscle relaxation, and diminished or abolished
motor and autonomous responses to noxious stimuli. Specific
effects of different substances (nitrous oxide, halothane, enflu-
rane, isoflurane, sevoflurane, desflurane, propofol, etc.) are due to
specific binding sites and genetic peculiarities of the patient.
Correlation studies of EEG and the stage of anesthesia have
been summarized by Winters (5) and Sloan (6). The initial
phase is dominated by the appearance of frontal fast activity,
gradually becoming more generalized and associated with dis-
solution of the alpha rhythm. During excitation, epileptiform
activities become discernible in nonepileptic and epileptic
patients with different anesthetic drugs (for summary see Ref. Figure 43.1 St.St., a 14-year-old female teenager, who took an
7). As anesthesia deepens, activity becomes slower and the unknown amount of diazepam. Clinically slight drowsiness. Twelve- to
voltage increases. Eventually, a burst-suppression pattern can be 13-Hz rhythmical activities, maximal over the anterior regions.

901
902 Part VI ■ EEG: Neuropharmacology and Anesthesia

Figure 43.2 W.M., a 33-year-old female and chronic abuser of alcohol and barbiturates (Optalidon). EEG at the sixth day
after admission for withdrawal. No seizures. Paroxysmal 2- to 3-Hz waves with intermingled small spikes.

the frontal cortex and spreads to parietal and occipital areas. EEG Changes after Withdrawal
Increasing doses are accompanied by intermixed slow activities After long-term ingestion of barbiturates, abrupt withdrawal
and dissolution of the alpha rhythm, indicating drowsiness and leads to paroxysmal abnormalities. Myoclonic jerks, even as
sleep. In children, the elderly, and patients with organic brain myoclonic status, generalized tonic–clonic seizures, and delir-
lesions, barbiturates even in low doses may induce irritability, ium are major complications. The EEG frequently shows gener-
hyperactivity, or delirium (10). Lack of barbiturate-induced fast alized paroxysmal activities and spikes, especially with photic
activity indicates a diffuse organic brain lesion; asymmetries are stimulation (14). These changes are usually transient in nature
suggestive of a focal lesion (11). and most often occur within the first few days after withdrawal,
but they may occasionally persist for 3 to 4 weeks. They also
Chronic Overdosage occur without clinical seizures and even with medically con-
Signs of chronic barbiturate intoxication in seizure patients trolled gradual withdrawal (Fig. 43.2).
might be misinterpreted as personality changes attributed to
the epileptic disorder. Signs include general sluggishness, Acute Intoxication
lethargy, difficulty in thinking, poor memory, learning disabili- Sedatives such as barbiturates, anxiolytic drugs, phenoth-
ties, and cerebellar deficits (10). Chronic neurotoxicity may iazines, and tricyclic antidepressants can all produce coma.
result in insidiously developing disturbances of higher cognitive They directly depress cellular oxidative metabolism and do not
functions without concomitant abnormalities (12). Compared permanently damage neuronal functions. The mere functional
with other antiepileptic drugs, cognitive adverse effects are pro- character of CNS depression after an acute overdose indicates a
nounced with phenobarbital (for review see Ref. 13). EEG less grave prognosis even in the presence of very serious clinical
changes are similar to those seen in the first stage of acute intox- and EEG signs.
ication. In epileptic patients, an increase in slow activities in According to the multilevel action of barbiturates, the clin-
serial recordings without increase or even with reduction in ical syndrome encompasses cortical, reticular, vestibular, and
seizure frequency is indicative of overdosage and should other brainstem dysfunctions. Coma due to overdose with
prompt an evaluation of the serum level. sedatives is fairly characteristic if one considers the depth of
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 903

Figure 43.3 E.A., a 24-year-old female.


Suicidal attempt by ingestion of a combina-
tion of cyclobarbital, hexobarbital, meproba-
mate, and carbromal (= Somnupan, banned
from the Austrian market for several years).
Coma with decerebrate posturing (with fast-
acting barbiturates cerebral functions can be
depressed in a rostro-caudal fashion and
motor signs may initially evolve) (16). EEG
with diffuse slow activity superimposed by
high-voltage 10- to 13-Hz activities. Patient
recovered completely.

unresponsiveness in combination with flaccid muscle tone, Miscellaneous Hypnotic Drugs


absent plantar responses, and preserved pupillary reactions Melatonin
(15). With fast-acting barbiturates cerebral functions can be
Melatonin has been extensively discussed in the popular media.
depressed in a rostro-caudal fashion and flexor and extensor
It is the hormone of darkness (22). The substance may act as a
postures may initially evolve (Fig. 43.3) (16). In severe cases,
phase-setter for sleep–wake cycles in subjects with a delayed
circulatory effects lead to a typical shock syndrome.
sleep phase syndrome (23). However, no evidence was found
Subsequent cerebral hypoxia may turn the functional distur-
that melatonin is effective in treating secondary sleep disorders
bance into structural damage with serious prognostic implica-
or jet lag and shift work disorders (24). For difficulties in sleep
tions.
EEG signs in different stages of acute intoxication closely
resemble those observed with other CNS depressant substances.
Initial dissolution of the alpha rhythm and appearance of inter-
spersed theta frequencies are followed by predominant slow
activities superimposed by 10- to 16-Hz rhythmical activity
maximal over the anterior regions. Such an EEG is suggestive of
intoxication with depressant drugs of any type. However, a so-
called spindle coma after head injury and the alpha coma pat-
tern with cerebral hypoxia may look similar. Superimposed fast
activities after intoxications have also been termed “drug-
induced alpha coma” (17). The separation from alpha coma
pattern with hypoxic states has prognostic significance with a
much more favorable outlook with intoxications (17). With
deepening coma, fast frequencies disappear and diffuse delta
activity becomes prominent. With impending breakdown of
vegetative functions, periods of flattening and, eventually, a
burst-suppression pattern appears (18). Preceding or following
the development of a burst-suppression pattern, the record may
be characterized by bi- or triphasic sharp transients (Fig. 43.4)
(19). Electrocerebral silence signifies the most advanced cases.
The prognostic meaning of this otherwise ominous sign is less
grave with intoxication. Several patients survived without per-
manent neurologic sequels (20).
Attempts have been made to correlate the EEG with clinical
signs and blood levels (14,19,21). In the earliest phases, clinical
signs are superior to the EEG in the assessment of the severity
of the functional disturbance, whereas the EEG permits grading Figure 43.4 B.E., a 33-year-old female. Coma after suicidal attempt by
of stages during the later phases. ingestion of barbiturates. Diffuse slow activity with triphasic waves.
904 Part VI ■ EEG: Neuropharmacology and Anesthesia

onset, ramelteon, a melatonin receptor agonist was recom- Acute Intoxication


mended (25). Effects on the conventional EEG have not been Overdosage with neuroleptics usually leads to sedation and
reported for melatonin or the agonist. coma. In milder cases, patients can be agitated, delirious, or
confused. Involuntary extrapyramidal movements, parkinson-
Bromides ism, and generalized tonic–clonic seizures can be prominent.
Bromide was the drug of choice as anticonvulsant and sedative Fatalities are rare if neuroleptics alone are taken; death is attrib-
during the second half of the 19th century. It is still used in the utable to cardiac effects. The EEG is dominated by slow waves,
treatment of therapy-resistant tonic–clonic seizures (26). With frequently occurring as generalized paroxysmal activities. Fast
acute bromide intoxication, the EEG shows mixed slow and fast frequencies as in barbiturate poisoning are not recorded (14).
activities. Very pronounced EEG slowing is found in chronic
bromide encephalopathies. Drug level determinations have Atypical Antipsychotic Drugs
improved the handling of the drug, so the typical bromism of A long list of newer antipsychotic drugs is on the market (cloza-
the old days has disappeared. pine, amisulpride, olanzapine, quetiapine, risperidone, sertin-
dole, ziprasidone, zotepine, aripiprazole). In a systematic
Antipsychotic Drugs meta-analysis only clozapine exhibited fewer parkinsonian
Antipsychotic drugs are used for the treatment of schizophrenia. symptoms and a higher antipsychotic effect than conventional
They are listed as typical, classic or first-generation antipsychotic drugs (33). Compared with the classic antipsychotics, the newer
agents (chlorpromazine, chlorprothixen, flupentixol, melperone, drugs are less extensively studied with quantitative EEG methods.
fluphenazine, perphenazine, trifluoperazine, thiothixene,
haloperidol among 18 drugs developed between the 1950s and the
Clozapine
1970s), and atypical or second-generation drugs (clozapine, Clozapine has affinity for serotonin (5-HT2a, 5-HT6, 5-HT7,
risperidone, olanzapine, quetiapine, ziprasidone, aripiprazole, 5-HT2c, and 5-HT3), alpha-adrenergic, and dopamine D4
amisulpride). Major progress has been made to elucidate their receptors but weak affinity for the D2 receptor (34). It causes no
mechanism of action (for review see Ref. 27). Typical antipsy- extrapyramidal symptoms and tardive dyskinesia (33). The
chotic drugs act by blocking the dopamine D2 receptor and are major adverse effect of clozapine relates to its potential for
connected with parkinsonian side effects. The newer antipsychotic damaging the granulocyte cell line. A cumulative risk figure for
compounds bind at many different receptors including members agranulocytosis of 0.8% at 1 year and 0.9% at 18 months was
of the dopamine receptor family and 5-HT2A receptors. calculated (35).
EEG power spectra show an increase in delta, theta, and above-
Typical Antipsychotic Drugs 21-Hz beta activities. The EEG became abnormal in up to 53% of
patients as a function of clozapine serum levels (36). Increased
EEG changes due to neuroleptics are not particularly different
theta and delta activities were prominent over the frontal, central,
for the numerous substances under this pharmacologic heading
and parietal areas (37). EEG abnormalities included paroxysmal
and therefore are discussed jointly.
slowing and spikes. Clozapine can provoke seizures. Seizure fre-
EEG Changes with Therapeutic Doses quency amounts up to 4% (38), increases with doses over
600 mg/day, and seems to be greater than with other typical and
A great number of quantitative analyses of EEG changes due to atypical antipsychotic drugs (39). Besides generalized tonic–clonic
therapeutic doses of neuroleptics have been summarized by seizures, myoclonic jerks have been observed (40).
Saletu (28). Neuroleptics increase the alpha activity with a slight
shift to the lower frequency range and increase the amount of Risperidone
slow activity and the general voltage output, but decrease per- Risperidone is a benzisoxazole derivative with combined
cent time of beta activity, variability of frequencies, and the dopamine D2 receptor and serotonin 5-HT2 receptor blocking
average frequency. Mild to moderate diffuse abnormalities have properties (41). The substance has an established efficacy in
been observed on visual EEG reading with considerable but acute psychotic states. Risperidone does not induce EEG changes
inconsistent variations among the different substances (29). in the waking state (42). The risk of risperidone-related seizures
The EEG abnormalities in treated patients exceeded the occur- amounts to 0.3%, a low number compared with the 3.5% risk of
rence in psychiatric patients not treated with antipsychotics for clozapine, that ranks highest among all antipsychotic drugs (39).
at least 5 days.
Other Atypical Antipsychotic Drugs
Epileptogenic Potency Little is known about EEG changes of other new antipsychotic
Although the risk of seizures under treatment with typical drugs. After Alper et al. (39) the seizure risk extracted from
antipsychotic drugs does not differ substantially from the gen- approval reports is 0.9% for olanzapine, 0.8% for quetiapine,
eral population (30), with high doses seizures and spikes in 0.4% to 0.5% for ziprasidone, and 0.4% for aripiprazole.
the EEG may occur (31). EEG and clinical seizures can deteri-
orate in chronic epileptic patients. Nonconvulsive status Antidepressant Drugs
epilepticus has been reported under treatment with typical Since the introduction of the first-generation tricyclic anti-
neuroleptics (32). depressants, several new drugs have been marketed. New
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 905

Figure 43.5 P.M., a 22-year-old male.


Comatose due to intoxication with an
unidentified amount with tricyclic antide-
pressants. Unresponsive slowing with super-
imposed fast activities. The patient survived
and was transferred to the psychiatric ward
the next day.

antidepressants might be classified according to their central hyperpyrexia, hypertension, seizures, and coma. The prognosis
mode of action into selective serotonin reuptake inhibitors (flu- depends largely on the effects on the cardiovascular system.
oxetine, fluvoxamine, paroxetine, citalopram, sertraline), dual Even with therapeutic doses, there is an increased tendency
serotoninergic antidepressants (reuptake inhibitors plus recep- toward cardiac arrhythmias, and there have been several reports
tor antagonism; nefazodone), selective serotonin and noradren- of unexpected death. Greater than 10-fold differences in the
alin reuptake inhibitors (venlafaxine), noradrenergic and number of deaths per million prescriptions have been shown
specific serotonergic antidepressants (mirtazapine), selective comparing tricyclic antidepressants with the newer antidepres-
noradrenalin reuptake inhibitors (reboxetine), and reversible sants (51). The EEG during acute intoxication shows wide-
specific monoamine oxidase inhibitors (moclobemide) (43). spread, poorly reactive, irregular 8- to 10-Hz activity and
Antidepressants, especially amitriptyline, and several paroxysmal abnormalities including spikes as well as unspecific
antiepileptic drugs (see below) are also important in the treat- coma patterns (Fig. 43.5) (14).
ment of neuropathic pain (44).
New Antidepressants
Tricyclic Antidepressants Seizures have also been reported with the newer antidepres-
sants. There are differences in seizure propensity among the dif-
EEG Changes with Therapeutic Doses
ferent substances, particularly pronounced with maprotiline
Tricyclic antidepressants such as imipramine, amitriptyline, and bupropion (1.5% seizure risk) and low with trazodone,
doxepin, desipramine, nortriptyline, and protriptyline increase nefazodone, mirtazapine, and the selective serotonin reuptake
the amount of slow and fast activities along with instability of inhibitors (for review see Refs. 39, 43, and 52). However,
frequencies and voltage. Furthermore, they slow down the fre- second-generation antidepressants other than bupropion
quency of the alpha rhythm (for review see Ref. 28). apparently have antiepileptic properties (39).
In general, the new antidepressant drugs have a higher ther-
Epileptogenic Potency apeutic index than the tricyclic compounds. Overdosage with
Paroxysmal slow waves, spikes, and polyspikes occur with ther- several of the new drugs has been reported, but deaths seem to
apeutic doses (45,46). The seizure frequency may increase in be an exception (for review see Ref. 43). Fatal toxicity index for
chronic epileptic patients. Furthermore, single or multiple venlafaxine is higher than that for other serotoninergic antide-
seizures occur in nonepileptic patients, especially with high pressants (51). EEG signs during the intoxicated state are rarely
doses (47,48). Several cases of absence status have been thought demonstrated and show no particular features.
to be due to treatment with tricyclic agents (49,50).
The Serotonin Syndrome
Acute Intoxication The serotonin syndrome is an adverse drug reaction associated
Unlike the phenothiazines, tricyclic antidepressants have much with a number of drug–drug interactions (53). Among the drugs
narrower therapeutic ranges and quickly reach toxic levels. at risk are selective serotonin reuptake inhibitors, other antide-
Overdosage may result in serious life-threatening conditions. pressant drugs, and monoamine oxidase inhibitors. The protean
This is of great concern, because depression is notorious for manifestations of the syndrome include mental status changes,
suicidal attempts. The clinical picture is characterized by autonomic hyperactivity, and neuromuscular abnormalities.
906 Part VI ■ EEG: Neuropharmacology and Anesthesia

Figure 43.6 E.R., a 43-year-old female with intoxication from lithium carbonate. A: Delirious, multiple hyperkinesia, gen-
eral rigidity. Blood serum level of Li 2.95 mval/ L. Diffuse slowing with rhythmical 1- to 3-Hz activity maximal over anterior
regions. Some triphasic-like waves. B: Nine days after A. No abnormal results at neurologic examination. No more lithium
could be found in the blood. The EEG was also normal.

Since the condition is potentially life-threatening, an early diag- improvement, but the EEG abnormalities regularly outlast the
nosis with immediate removal of the responsible drugs is essen- abnormal serum levels (Fig. 43.6A and B).
tial. Case reports of the syndrome have noted EEG abnormalities
with diffuse slow activities, spikes, and triphasic waves (54).
ANXIOLYTIC DRUGS
Lithium Benzodiazepines
Lithium is still used in the prophylactic treatment of bipolar The main actions of benzodiazepines can be described as hyp-
mood disorders and has not been entirely replaced by valproate notic, anxiolytic, anticonvulsant, myorelaxant, and amnesic.
or lamotrigine. EEG changes during the treatment are frequent They exert their actions by binding at specific sites at the GABA-
and marked (55,56). In general, they parallel the blood serum A receptor chloride ionophore (65). The distribution of GABA-
levels, but there are also remarkable discrepancies. A receptors within brain regions and changing compositions of
Acute intoxication with lithium salts can be a life-threaten- receptor subunits might contribute to differences in the efficacy
ing condition. Early symptoms include fatigue, muscular weak- of the numerous benzodiazepines (9,66). Furthermore, pharma-
ness, and tremor. When plasma concentrations rise above cokinetic properties like rapidity of absorption, half-life time,
2 mEq/L, more serious toxic effects occur. Disturbances in renal binding to fat deposits, and activities of metabolites are impor-
elimination or fever with liberation of tissue-bound lithium are tant differences among the substances. In general, however, all
the most frequent causes of intoxication. Obtundation, stupor, benzodiazepines exert actions mentioned above and share the
or delirium is always present. Neurologic signs show a bewilder- same side effects. Therefore, those on the list of benzodiazepines
ing variety of cortical and subcortical dysfunctions, optomotor (clobazam, clonazepam, diazepam, estazolam, flunitrazepam,
disturbances, and peripheral neuropathies (57,58). Movement flurazepam, lorazepam, nitrazepam, quazepam, temazepam,
disorders are especially dramatic. Myoclonic jerks (59), convul- triazolam among others) are considered together.
sions, choreiform hyperkinesis, other complex extrapyramidal
movements, and several other signs may be observed. If accom- EEG Changes with Therapeutic Doses
panied by triphasic waves in the EEG, such conditions have The benzodiazepine derivatives are potent activators of beta
been called lithium-induced Creutzfeldt–Jakob syndrome (60) activity, which persist in the EEG as long as 2 weeks after the last
or nonconvulsive status epilepticus (61). However, the epileptic ingestion. As with barbiturates, benzodiazepine-induced fast
or encephalopathic nature of these confusional states remains activities are reduced over the site of a cerebral lesion (11).
undecided (62). Permanent neurologic sequels due to lithium Furthermore, benzodiazepines produce a decrease in alpha
toxicity have also been reported (63). activity and general voltage, and a slight increase of 4- to 7-Hz
The EEG is always markedly abnormal with lithium intoxi- activity (28).
cation and shows diffuse slowing, paroxysmal abnormalities
including spikes and triphasic waves. Focal slowing also occurs Effect on Seizures
and does not have to be taken as a sign of focal brain lesion Benzodiazepines reduce generalized spikes but have no effect on
(64). Improvement of electrical activity parallels clinical interictal focal spikes (67) except for discharges in rolandic
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 907

Figure 43.7 M.C., a 17-year-old male. Had


a severe brain trauma 1 year ago. Marked
spasticity. Now comatose due to an over-
dose of baclofen (failure of the intrathecal
drug delivery system). Had left-sided focal
motor seizures the first day after intoxica-
tion. The EEG taken the second day exhibits
periodic paroxysmal slow and spiky waves
on a severely abnormal slow background
activity. Recovered up to the habitual condi-
tion after the brain trauma. No further
seizures.

epilepsy (68). They are drugs of first choice for status epilepticus Sleep architecture is claimed to resemble normal patterns more
and repetitive acute symptomatic seizures (69). Despite their closely. Zopiclone belongs to the cyclopyrrolones. The final
antiepileptic action, they may provoke tonic status epilepticus if pathway constitutes an opening of the chloride ionophore. The
intravenously administered to children with absence status in most frequent adverse effects include bitter taste, dry mouth,
Lennox–Gastaut syndrome (70,71). Lorazepam is superior in the and complaints comparable to the benzodiazepines (78).
treatment of status epilepticus because it is less extensively bound Microstructural analysis of sleep architecture and decrease
to fat (72). Withdrawal of benzodiazepines has been considered of EEG arousals allow discrimination of benzodiazepines,
an etiologic factor of “de novo” absence status of late onset (50). zolpidem, and zopiclone (79). There was no consistent pattern
of superiority in therapeutic efficacy among the Z-drugs.
Side Effects of Benzodiazepines
Baclofen
All benzodiazepines share a long list of side effects consisting of
tiredness during the day following ingestion (hangover), Baclofen is a selective GABA-B receptor agonist and is used for
rebound anxiety, anterograde amnesia, rebound insomnia, low- treatment of spasticity (80). Oral and intrathecal baclofen have
dose dependency, and withdrawal syndromes. The profile of been associated with epileptic seizures (Fig. 43.7) (81,82). After
side effects depends on the specific binding site, the dose, the intrathecal baclofen overdose the EEG can exhibit coma pat-
half-life time, and other pharmacokinetic variables. With short- terns characterized by periodic generalized epileptiform dis-
acting substances, the amnesic effects are comparatively marked charges without overt seizures (83).
(73,74). In general, benzodiazepines have significantly fewer
side effects than barbiturates, and it is rational to prefer them as Psychotogenic Drugs
hypnotics. The substances under the heading of psychotogenic drugs are
intimately connected with addiction. Exciting progress has been
Acute Intoxication made elucidating the neurobiology of addiction and its relation
The clinical picture resembles that seen with other CNS depres- to various intrinsic neurotransmitter systems (84). This scien-
sant drugs and is not truly specific for benzodiazepines (75). tifically as well as clinically important issue can only be touched
The prognosis is generally good, although in patients with on a few references.
decreased respiratory reserve and in the very young even thera-
peutic doses may dangerously depress cardiorespiratory func- Lysergic Acid Diethylamide (LSD) and Mescaline
tion (76). The EEG shows prominent fast activity with no These agents cause decreased amplitude and depression of slow
response to stimuli. With larger doses, coma patterns, as in waves as well as acceleration of the dominant frequencies dur-
other intoxications, are recorded (14). ing the drug-induced psychotic state (45,85).

So-called Z-drugs Marijuana


Zaleplon, zolpidem, and zopiclone are non-benzodiazepine hyp- Cannabis exerts its central effects through the CB1 cannabinoid
notics. Zolpidem belongs to the class of imidazopyridines (77). receptor (for review see Ref. 86). These effects include disrup-
It binds to the alpha 1 unit of the GABA-A receptor complex (9). tion of psychomotor behavior, short-term memory impair-
Similar side effects as with benzodiazepines have been reported. ment, stimulation of appetite, and antinoceptive and antiemetic
908 Part VI ■ EEG: Neuropharmacology and Anesthesia

actions. Neuropsychologic deficits have been noted after years characteristic cocaine “high” (100). The induction of long-term
of heavy frequent cannabis use (87), and the risk of psychosis in synaptic plasticity in neurons of the brain’s reward system
later life seems to be increased (88). Cannabis is used in the seems to represent a common pathway for the addictive poten-
treatment of neuropathic pain and spasticity (89), but the risk tial of cocaine, morphine, nicotine, ethanol, and amphetamine
to benefit ratio has to be clarified. (101). Variants of the DRD2 gene have been associated with
Marijuana smoking produces no visible effects on the con- cocaine, nicotine, and opioid dependency (102).
ventional EEG (64,90). The EEG of chronic marijuana users Berger’s historic experiments on the EEG effects of cocaine
was associated with reduced power in the alpha and beta bands intake have been confirmed. Cocaine increases beta power cor-
at posterior sites (91). These EEG changes continued for the related with the area under the cocaine plasma versus time
month of abstinence. curve (103). Several neurologic complications are associated
with chronic cocaine intake. It induces strokes (104–107),
CNS Stimulants orbital infarction (108), subarachnoid and intracerebral hem-
Central stimulants potentiate central dopamine activity. orrhages (109,110), vasospasms after aneurysmal subarachnoid
Different modes of dopaminergic potentiation have been hemorrhage (111), cerebral vasculitis (112), persistent dyskine-
shown in such drugs as the amphetamines, methylphenidate, sias (113), oculomotor nerve palsies (114), and spinal and
and cocaine (92). medullary vascular syndromes (106), and can provoke seizures
or exacerbate a preexisting seizure disorder (92). Seizure activ-
Low -Dose Effects on EEG ity can present as status epilepticus (115).
CNS stimulants increase the amounts of beta and alpha activi-
ties, reduce the general voltage and the amount of slow waves, Antiepileptic Drugs
and tend to suppress seizure discharges in pertinent cases, espe- Drug therapy of epilepsies represents the long-term therapy par
cially the 3-Hz spike-and-wave complexes (28). Abnormal slow excellence. Dose- or interaction-related CNS toxic effects are
waves during stupor and coma are diminished by the adminis- common. Delayed toxic effects and drug-induced diseases are
tration of methylphenidate. So-called recreational club drugs not dose-related, occasionally life-threatening and not always
illegally offered as “ecstasy” contain CNS stimulants, especially reversible. Antiepileptic drugs act directly on voltage-gated ion
methamphetamines and dioxymethamphetamine (MDMA) channels and by influencing GABA-mediated effects. They can
(93). A number of acute and long-lasting medical complica- bind at the GABA-A receptor at different binding sites, or can
tions are associated with abuse of these drugs. Therefore, most inactivate GABA-metabolizing enzymes and inhibit uptake of
CNS stimulants have been banned from the market. Modafinil, GABA into nerve cells and glia cells. Other antiepileptic drugs
a pharmacologically unique wake-promoting agent, has influence different transporter systems or the glutaminergic
replaced the traditional amphetamines for the treatment of nar- system via NMDA/AMPA receptors. For detailed and compre-
colepsy (94). Psychostimulants are also used for treatment of hensive information, the reader is referred to Levy et al. (116).
attention-deficit hyperactivity disorder (ADHD). Increased Several antiepileptic drugs are also used in the treatment of
slow activities in unmedicated ADHD girls are normalized neuropathic pain, migraine, and alcohol dependency, and as so-
under psychostimulant therapy (95). With atomoxetine a non- called mood stabilizers.
stimulant pharmacotherapy is now available for ADHD (96). EEG changes due to antiepileptic drugs have to be divided
into effects of therapeutic doses on background activity and on
Acute Intoxication frequency and morphology of preexisting spikes as well as into
Intoxications with CNS stimulants have increased due to their the effects of overdoses, overt intoxication, and withdrawal. In
illegal use as “ecstasy.” In a 1996 survey it was reported that general, the standard antiepileptic drugs slow down the fre-
nearly 5 million Americans have used methamphetamine at quency of the occipital basic rhythm even with nontoxic serum
some time in their lives (97). Symptoms of mild intoxications levels and increase the percentage of power in the theta and
are those of sympathetic activity. In more severe poisoning, delta bands with visual (117,118) and with quantitative analysis
hypertension, confusion, cardiac anomalies, and, finally, hyper- (119). These changes correlate with cognitive effects and sub-
thermia, convulsions, circulatory failure, and coma occur. jective complaints. The effect on interictal spikes varies consid-
Death due to amphetamines is related either to direct pharma- erably with a positive correlation between seizure frequency
cologic effects or to secondary complications in drug addicts and the number of spikes in some patients (120).
(98). The EEG with overdose of stimulant drugs shows the
usual coma pattern without any particular features. Hydantoins
Methamphetamines can damage dopaminergic and serotonin- Signs of cerebellovestibular dysfunction signal initial hydan-
ergic neurons (99) and produce corresponding permanent neu- toin overdosage. Cerebellar atrophy occurs with chronic
rologic abnormalities. hydantoin use but also with acute intoxication (121,122).
Overt hydantoin intoxication is further characterized by
Cocaine altered higher cognitive function, pyramidal signs, and several
Cocaine has become a major substance in the field of drug extrapyramidal movement disorders. Epileptic seizures may be
addiction. It binds strongly to the dopamine reuptake trans- exacerbated (123,124). Cardiovascular toxicity is rare unless
porter and blocks such reuptake after normal neuronal activity. the substance has been given parenterally for treatment of
High dopamine concentrations at the synapse produce the status epilepticus.
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 909

Figure 43.8 K.B., a 17-year-old female.


Temporal lobe epilepsy with rare seizures.
Self-poisoning with a huge amount of a
slow release preparation of carbamazepine.
CBZ level the next day 16 g/ mL. Soporos.
Diffuse slowing and 5- to 6-Hz rhythmical
activities over the posterior regions.
Recovered promptly.

In contrast to barbiturates and benzodiazepines, the EEG seizures (123,137). However, several other antiepileptic drugs
shows no increase in fast activities with visual EEG reading can worsen seizures. Overdosage represents only one of the
(125). Phenytoin increases the power in the theta and delta three possible causes (124).
bands with blood levels in the usual range and without clinical Carbamazepine was claimed to have fewer adverse neu-
signs of overt intoxication (117,118). There are conflicting ropsychological effects than phenobarbital (138). However, car-
reports on the influence of phenytoin on interictal epileptiform bamazepine has cognitive side effects without clinically
discharges. No changes (126), an increase (127), or a decrease of significant differences to phenytoin (139). Clinical signs of
spikes (128) have been reported. acute poisoning include ataxia, nystagmus, diplopia, drowsi-
No changes in background activity occurred with reduction ness, and diffuse slowing in EEG (Fig. 43.8). Cardiovascular
of phenytoin dosage (Duncan et al., 1989). Withdrawal of complications are common (140). Coma indicates a severe
antiepileptic drugs routinely is used as a seizure-provoking intoxication, and EEG can deteriorate to burst-suppression pat-
method in intensive epilepsy monitoring. No misleading infor- tern with accompanying myoclonus (137).
mation was gained with this procedure localizing the seizure
onset zone (129). The same was true withdrawing carba- Oxcarbazepine
mazepine and valproate. Oxcarbazepine is an analog of carbamazepine and shares its
At toxic levels, phenytoin can cause marked diffuse delta efficacy against focal seizures (for summary see Ref. 141).
activity and paroxysmal slow-wave abnormalities (125). This is Oxcarbazepine is not metabolized via the epoxide-diol pathway,
also true in cases of chronic encephalopathy with near-normal possibly resulting in fewer adverse events. However, hypona-
serum levels (130). tremia is more frequent and more severe with oxcarbazepine
than with carbamazepine (133). EEG changes were character-
Carbamazepine ized by generalized spikes not seen prior to oxcarbazepine,
Carbamazepine is chemically related to the tricyclic antidepres- occasionally accompanied by worsening of generalized seizure
sants. It may augment diffuse slow activities (118,126). Besser et types (142,143).
al. (131) found no correlation between serum levels of carba-
mazepine or the epoxide and the increase of power in the theta Succinimides
and delta bands. Furthermore, EEG slowing was not associated The use of succinimides directed against absence seizures has
with rise in seizure frequency (120,132). Severe hyponatremia declined in recent years due to antiepileptic drugs with a
occurred in 2.8% of carbamazepine-treated patients (133). This broader efficacy spectrum (valproate, lamotrigine). The succin-
adverse effect might contribute to the development of diffuse imides sometimes cause somnolence, lethargy, and emotional
slow activities in the EEG. Generalized paroxysmal activities instability accompanied by signs of altered vigilance in EEG.
including spikes can be increased (126). Preexisting interictal Acute intoxication has been reported rarely (see Ref. 144).
focal spikes are either increased or unchanged by carba-
mazepine (126). New appearance of generalized epileptiform Valproic Acid
discharges after introduction of carbamazepine treatment was Valproate in the usual doses did not significantly change EEG
observed, and spikes can be accompanied by seizure exacerba- background activity (117). With processed EEG analysis a
tion (134–136). Carbamazepine overdose may also exacerbate decreased EEG synchronization in the delta and theta frequencies
910 Part VI ■ EEG: Neuropharmacology and Anesthesia

was observed (145). The most important change consists of Observations of concentric and irreversible visual field
reduction or even disappearance of generalized spikes along with deficits (174) limited the use of the substance to the West syn-
seizure reduction (146–149; Villareal et al., 1978). Photosensitive drome and to otherwise untreatable focal epilepsies (175). The
spikes likewise disappear with valproate treatment (150). maximum daily dose was the single most important factor for
Intoxication with valproate was occasionally accompanied visual field deficits (176).
by marked diffuse slowing considered to be partly caused by
drug interactions (151). Enormous interindividual differences Lamotrigine
have been observed with acute valproate poisoning. Lamotrigine blocks voltage-gated sodium channels (for review
Drowsiness, stupor, and coma occur with normal doses of val- see Ref. 177). Rash is the most common cause for withdrawal of
proic acid, with or without evident metabolic changes such as lamotrigine treatment. Comedication with valproic acid and
hyperammonemia and low carnitine (for review see Refs. 152 rapid titration are risk factors.
and 153). In the diagnosis of valproate-associated encephalopa- No slowing in background activity was seen in volunteers
thy the EEG plays an important role. It is characterized by a dra- (178) and in epileptic patients (179). Lamotrigine has a marked
matic change in sequential recordings with the occurrence of decreasing effect on the occurrence and frequency of sponta-
bilaterally synchronous high-voltage slow-wave activity. Case neous and photosensitive generalized spikes and waves
reports exist about valproic acid-induced encephalopathy with (180,181) as well as on interictal epileptiform activity in young
triphasic waves (154). patients with drug-resistant epilepsy (182).
Fatal liver failure with valproate therapy has been reported Lamotrigine can lead to exacerbation of myoclonic epilep-
(155–157). There are no indications that repeated EEG records sies (183,184), and triggers convulsive (185) and nonconvulsive
can herald this insidious complication. status epilepticus (186). One report deals with self-poisoning
Although valproate is increasingly used for treatment of sta- with lamotrigine (187). No serious toxicity was observed. There
tus epilepticus, the substance by itself can trigger tonic status was a prolongation of QRS in ECG, but no EEG examination
epilepticus (158). was reported.
Clonazepam
Gabapentin
Clonazepam is a benzodiazepine and produces the correspon-
Gabapentin is a chemical derivative of GABA, which penetrates
ding EEG changes (159). It has little influence on focal interictal
the blood brain barrier. The substance has proven antiepileptic
epileptic activity (160). Given intravenously, the substance is a
properties. There are several lines of evidence for its mecha-
powerful blocker of status epilepticus, but the effect remains
nisms of action in a variety of animal models, which suggest
temporary in absence status occurring in patients suffering
that gabapentin differs pharmacologically from other
from Lennox–Gastaut syndrome. Like diazepam, clonazepam
antiepileptic drugs (for review see Ref. 188). Gabapentin is
can produce tonic seizures if given intravenously in this setting
extensively used in other indications than epilepsy like migraine
(161).
(189), chronic daily headache (190), painful neuropathy (191),
Vigabatrin and psychiatric disorders (for summary see Ref. 192).
Gabapentin toxicity is low (for review see Ref. 193). The
Vigabatrin binds irreversibly to GABA transaminase and
most common adverse events are somnolence, dizziness, and
inhibits the catabolism of the neurotransmitter. GABA is
ataxia. Overdoses showed no serious toxicity (194).
increased in the brain after vigabatrin exposure (for review see
Prolonged therapy with gabapentin induced EEG slowing
Ref. 162).
that correlates with cognitive complaints (195). Gabapentin-
In humans, one study reports diffuse slowing with vigaba-
treated subjects had an increase in slow-wave sleep compared
trin (163), whereas several other studies found no change in
with baseline (196).
background EEG activity (164–166). No consistent influence
on interictal spikes was reported by Ben-Menachem and
Treiman (167) and Marciani et al. (163). On the other hand, Pregabalin
development of generalized spikes was seen, occasionally Like gabapentin pregabalin is a structural analog of GABA
accompanied by myoclonic jerks or absence seizures, even in (197). Its action is not entirely clarified. Compared with
patients with focal epilepsies (168–171). An excess GABA con- gabapentin, pregabalin seems to be more potent against
tent in the brain was held responsible for the induction of gen- seizures (198) and pain disorders (199). To our knowledge no
eralized spikes (see also the section “Tiagabine”). systematic EEG studies have been published. Myoclonic jerks
Acute encephalopathies characterized by EEG abnormalities have been observed associated with pregabalin treatment
occurred after the start of vigabatrin treatment (171,172). (200,201). The EEG obtained in these patients exhibited no
These anecdotical case reports do not allow a decision whether changes time-locked to the jerks, so their cortical origin was not
these encephalopathies are related to comedication or to a pre- proven. In two patients treated with pregabalin for chronic pain
existing cerebral anomaly. There are few reports of overdoses generalized myoclonic status epilepticus occurred (202).
with vigabatrin (see Ref. 144). The development of psychoses in Pregabalin withdrawal led to a delirious encephalopathy in a
the course of vigabatrin treatment was not uniformly related to postherpetic-neuralgia patient (203). MRI showed a splenial
overdose (173). edema, but no EEG report was added.
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 911

Felbamate reversible encephalopathy with marked delta slowing in EEG


Felbamate is the first drug with proven efficacy against seizures was reported under levetiracetam (227).
occurring in Lennox–Gastaut syndrome (204). Observations of Zonisamide
aplastic anemia and liver failure including fatalities have been
Zonisamide is a 1,2-benzisoxazole with a sulfonamide side
reported after the drug was marketed in the United States
chain. Its antiepileptic profile is similar to hydantoins and car-
(Kaufman et al., 1997). The use of felbamate now is limited to
bamazepine with multiple mechanisms of action (for summary
patients with otherwise intractable epilepsies (French et al.,
see Ref. 228). The development of the substance started in
1999) (205). Some overdoses have been reported with low
Europe and the United States and was stopped because of high
toxicities. Published EEG investigations are limited to animal
incidence of renal calculi (229). Afterwards, zonisamide was
experiments.
further used in Japan (230), and finally reintroduced in the
Western countries (231).
Tiagabine Zonisamide decreases cortical excitability in patients with
Tiagabine has an established mode of anticonvulsant action via idiopathic generalized epilepsy (232). After zonisamide an
inhibition of GABA uptake. The substance has a proven almost complete resolution of generalized spike-and-wave dis-
antiepileptic potency in focal seizures and a low toxicity (for charges and EEG absence seizures have been noted in a patient
review see Ref. 206). Overdose has been reported (207). with juvenile myoclonic epilepsy (233). To our knowledge, no
The effects of tiagabine on electroencephalogram and spike- systematic study deals with EEG changes after zonisamide.
wave discharges have been studied in animals (208,209). In a rat
model of absence epilepsy spike waves and other forms of parox- Lacosamide
ysmal activity were facilitated by tiagabine. As with vigabatrin, an Lacosamide was recently approved as an antiepileptic drug in
increase of GABA content in the brain can result in generalized Europe. It is a member of a series of amino acids and has a dual
EEG abnormalities and seizures in epilepsy patients. Several cases mode of action. It selectively enhances slow inactivation of volt-
of nonconvulsive status epilepticus have been reported during age-gated sodium channels, and has a collapsin response medi-
therapy with tiagabine, in seven patients electroclinically con- ator protein as a binding partner (234). Lacosamide employs
firmed (revisited by Koepp et al. (210)). The FDA safety informa- proven antiepileptic properties in adults with partial-onset
tion and adverse event reporting program warned that seizures seizures (235). EEG effects are not investigated.
and status epilepticus are known to occur with Tiagabin.
Other Drugs
Topiramate A great number of drugs produce EEG changes and CNS intox-
Topiramate is a broad-spectrum antiepileptic drug with multi- ications (for review see Ref. 14). The following part selects some
ple modes of action (for summary see Ref. 211). Cognitive and drugs with particular interest for neurologists.
behavioral effects are slightly worse than with valproic acid Morphine and the Opiates
(212) or with lamotrigine (213). Language deficits are a specific
Morphine and the opiates produce only mild to moderate effects
side effect (214,215). No changes were seen on EEG background
on the EEG; the frequency of the alpha rhythm may be slowed
measures or on daytime vigilance (Salinsky et al., 2007) (216).
down, and paroxysmal changes may occasionally appear. REM
sleep depression has been observed. In neonates, morphine pro-
Levetiracetam
duces profound reversible EEG alterations, which have to be
Levetiracetam is chemically related to piracetam and was taken into consideration in the interpretation of abnormalities in
screened for second-generation nootropic substances. It is these age groups (236). With opioid anesthesia spike discharges
devoid of anticonvulsant activities in traditional seizure screen- resembling benign epileptiform transients of sleep appeared
ing models but exhibits a broad antiepileptic spectrum in within 3 minutes of the first opioid dose in 19 of 20 patients
epileptic patients. So-called antiepileptogenic properties in undergoing coronary artery revascularization (237). Abrupt ces-
experimental investigations have been claimed to be clinically sation of spikes after administration of midazolam was held an
relevant (for review see Ref. 217). Levetiracetam has become a argument for an epileptogenic mechanism of the opioid-induced
major antiepileptic drug for partial epilepsies. Antimyoclonic activity. Morphine and heroin addicts usually show unremark-
efficacy has been reported recently (218–220). able EEG tracings unless they present in a comatose state after
Levetiracetam reduces the corticospinal response to mag- overdose, which produces diffuse slowing. EEG abnormalities
netic stimulation (221). If related to serum levels, there was a observed in an alert addict should prompt the suspicion of a
substantial time lag to these responses (222). This effect might HIV-related brain disease.
correspond to a sustained efficacy against seizures in refractory
epilepsies (223). The amount of interictal spikes in epilepsy Aminophylline
patients was reduced by levetiracetam. (224). It has no major Aminophylline is a bronchodilator commonly used for the
effects on sleep nature (225). treatment of chronic destructive pulmonary disease. Agitation,
In general, levetiracetam is well tolerated. Adverse psychi- tremor, delirium, and even coma occur with therapeutic doses.
atric effects occur in a group of patients generally prone to such Acute convulsions may complicate a comatose state (238). Focal
complications during therapy with antiepileptic drugs (226). A motor status epilepticus (239,240) might be accompanied by
912 Part VI ■ EEG: Neuropharmacology and Anesthesia

Figure 43.9 K.H., a 58-year-old male. Chronic asthmatic and alcoholic. Relapsing episodes of focal motor status with
epilepsia partialis continua-like features with twitchings of left facial and upper extremity muscles. Euphyllin overdose
due to self-medication. No abnormalities with CT and MRI scan. The EEG exhibited constantly repeated episodes of left
parieto-temporal slow and superimposed fast activities accompanied by periodic twitches (see muscle artifacts).

periodic lateralized epileptiform discharges (PLEDs) in the status epilepticus of other etiologies, penicillin should not be
EEG (Fig. 43.9). Aminophylline-associated seizures are difficult administered as an antibiotic drug.
to stop by diazepines. Besides the removal of the responsible
substance, the use of barbiturates as the drug of first choice was Cephalosporins
recommended (241). Cephalosporin antibiotics comprise a varied group of beta-
lactam antibiotics. They produce seizures and nonconvulsive
Isoniazid (INH) status epilepticus with corresponding EEG abnormalities
INH, a highly effective tuberculostatic drug, is known to inter- (246,247). The confusional states after cephalosporin have also
act with several antiepileptic drugs. This interaction may lead to been called encephalopathy with diffuse rhythmic nonreactive
intoxication from previously well-tolerated doses of anticon- triphasic sharp waves in the EEG (248).
vulsants. Overdose of INH can produce coma complicated by
acute convulsions, metabolic acidosis, hyperglycemia, and ace- Immunosuppressant Drugs
tonuria (242). The interictal EEG is characterized by diffuse Cyclosporine is a lipophillic cyclic undecapeptide, which today
slowing and generalized paroxysmal slow waves with intermin- is the major immunosuppressant after organ transplantations.
gled bilateral sharp waves (243). With a large overdose of INH, It is also used in the treatment of rheumatic and other autoim-
status epilepticus has been observed (244). mune diseases. Cyclosporine neurotoxicity is one of the most
significant clinical side effects occurring in up to 60% of
ANTIBIOTICS patients (for summary see Ref. 249). The clinical symptoms
consist of encephalopathies, seizures, and stroke-like episodes
Penicillin among others. In these cases the EEG exhibited diffuse and focal
The parenteral administration of very large doses of penicillin slowing and epileptiform discharges (see Ref. 249). A significant
G (40 to 80 million units/day) may produce jerks, generalized risk for seizure recurrence was associated with persistent EEG
seizures, or even status epilepticus (245). In the treatment of abnormalities (250).
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 913

Antineoplastic Agents activity to PLEDs and, with resolution of the status, to contin-
A rapidly growing number of antineoplastic substances increase uous lateralized slow activities. Seizures have also been seen
the armamentarium fighting cancer. Many, if not all, of these after intravenous application of several different contrast media
drugs cause encephalopathies with personality changes, confu- (265). In a case of gadolinium-related encephalopathy the EEG
sions, hallucinations, and coma (see also Chapter 17). Singh et al. showed generalized atypical triphasic waves (266).
(251) reviewed seizures and epilepsy in people with cancers other Levodopa
than primary brain tumors. Among different other causes anti-
neoplastic agents trigger seizures through varied mechanisms. Levodopa is the main drug used to treat symptoms of
Detailed reports exist for ifosfamide. It causes nonconvulsive sta- Parkinson disease. Reports on EEG changes are sparse. Neufeld
tus epilepticus (252,253), diazepam-sensitive encephalopathy (267) described four patients in subacute confusional states fol-
(254), and encephalopathy characterized by periodic generalized lowing an increase in the dose of levodopa. The EEG revealed
triphasic waves and negative myoclonus (255). periodic generalized triphasic waves.

Salicylate Sildafenil
Salicylate overdose occurs with suicidal attempts or acciden- Sildafenil (Viagra R) is the first drug approved by drug adminis-
tally. Most persons lack prominent neurologic symptoms, but trations to treat erectile dysfunction. In mice, sildafenil induces
severe illness with coma and seizures can occur (15). Two a proconvulsant effect, probably due to the release of nitric oxide
reports mention EEG changes (256,257). The records showed (268). Generalized tonic–clonic seizures are reported after
diffuse slowing accentuated over the anterior regions and occa- sildafenil intake in two patients (269). Seizures also occurred
sionally in rhythmical trains. after vardenafil, a substance related to sildafenil (270). In both
reports the EEG even after sleep deprivation has been normal.
Contrast Media
Toxic Encephalopathies
Metrizamide (Amipaque) is a nonionic water-soluble radio-
logic contrast agent. Metrizamide is used for myelography and A selection of toxic encephalopathies relevant for neurologists
cisternography. Nausea, vomiting, myoclonus, seizures, epilep- is given in the following section. For a comprehensive review
sia partialis continua (258), stroke-like episodes, and aphasia the reader is referred to textbooks of toxicology and to Mellerio
are symptoms of metrizamide-induced encephalopathy. Several and Kubicki (271).
cases of nonconvulsive status epilepticus have also been Toxic encephalopathies present themselves as acute, suba-
reported (Fig. 43.10) (259–262). Ropper et al. (263) reported cute, or chronic disorders. Neurologic and EEG abnormalities
EEG changes after metrizamide for myelography and posterior indicate alterations of neuronal structures, receptor composi-
fossa cisternography in 61 patients. The EEG exhibited abnor- tion and sensitivity, or neuronal death. In general, clinical signs
malities in 34% of patients. A number of changes required 24 to may be classified as coma, organic mental impairment, seizures,
48 hours to develop. movement disorders, involvement of cranial and spinal periph-
After myelography with iopamidol, sold under the trade eral nerves, and neuromuscular dysfunction. EEG changes
name Omnipaque, status epilepticus was reported (264). The occur with coma, acute encephalopathies, and severe chronic
EEG demonstrated the evolution of lateralized rhythmic ictal cerebral deficit syndromes.

Figure 43.10 E.A., a 56-year-old female.


Comatose after myelography with metriza-
mide. Continuous repetitive triphasic
waves. These conditions have also been
called symptomatic nonconvulsive status
(see text).
914 Part VI ■ EEG: Neuropharmacology and Anesthesia

Lead Motor vehicle exhaust gases are the most common source. The
The neurotoxic actions of lead include apoptosis, excitotoxicity, CNS effects are related to its extremely strong affinity to hemo-
and influences on neurotransmitter storage and release processes, globin. If the acute intoxication with different comatose stages
mitochondria, second messengers, cerebrovascular endothelial is survived, vegetative states, diffuse encephalopathies, focal
cells, and glia cells (for review see Ref. 272). Chronic exposure to abnormalities, and extrapyramidal syndromes may persist and
lead is associated with intellectual impairment. Declines in IQ may be accompanied by corresponding MRI findings. A delayed
have been correlated to blood lead concentrations, even those encephalopathy appears after a period free of symptoms
below 10 g/dL (273). Neurodegeneration linked to cumulative (287,288).
lead exposure can be measured by MRI (274). Diffuse or focal epileptiform abnormalities may be seen in
In acute intoxications, the EEG shows the usual signs of dif- acute and protracted encephalopathies (289). Leweke et al.
fuse encephalopathies. Fejerman et al. (275) reported lead (1999) found a good correlation between the clinical and EEG
encephalopathy as a cause of a Lennox–Gastaut syndrome with course. An improvement of the occipital alpha activity was
slow spikes and waves. In chronic forms, the EEG is inconclu- observed by hyperbaric oxygen therapy (290). This effect was
sive (Burchfield et al., 1980) (276). held an indicator for preventing delayed neuropsychiatric
sequels of carbon monoxide poisoning.
Mercury
Methyl Alcohol (Methanol)
Exposure to high doses of methyl mercury causes devastating
damage to the nervous system, resulting in abnormalities in Methyl alcohol is metabolized to formaldehyde and formic
motor function and impairment in the visual, auditory, and acid. Formaldehyde is particularly toxic for retinal cells, and
somatosensory systems (for review see Ref. 277). Chronic toxi- formic acid causes acidosis, generally considered the main cause
city is related to industrial exposure and mainly affects the kid- of CNS involvement (291). Intoxications mostly occur with low
ney. Exposure to mercury from dental amalgams and fish social status and may acquire an epidemic character. Leading
consumption has been a concern for decades. The hypothesis of symptoms are visual disturbances and consecutive permanent
prenatal mercury as a neurodevelopmental risk factor could not blindness. Epileptic seizures, stupor, and coma are signs of
be supported (278). Ayurvedic medicines contain detectable severe acute intoxications. Necrosis of the putamen and cere-
lead, mercury, or arsenic (279). A negative impact to human’s bellar cortex and corresponding parkinsonian syndromes (292)
health, however, is not substantiated. have been reported.
The EEG abnormalities consist of unspecific diffuse slowing The EEG shows marked slowing that correlates mainly with
and reflect the clinical state (280). Quantified EEG may docu- acidosis rather than with blood and cerebrospinal fluid
ment early effects of exposure to mercury vapor (281). methanol level (293).

Manganese Ethylene Glycol


The most characteristic for manganese poisoning is parkinson- Ethylene glycol is used as antifreeze. Intoxications occur in
ism. After prolonged exposure spasticity, epileptic seizures, and developing countries, if the substance is used for elixir prepara-
dementia may be associated (282). Myoclonus and high-inten- tions of drugs, with suicidal attempts, and if the substance is
sity signals in the globus pallidus on T1-weighted MR images illegally added to wine. Glycol causes renal failure and death
have been reported (283). EEG slowing and fast activities (271) (294,295) as well as peripheral nerve palsies mimicking
as well as normal records were observed. polyradiculitis (296–298). In comatose states with acute intoxi-
cation the ubiquitous EEG patterns without any specific signs
Aluminum can be observed.
Toxic effects of aluminum have been recognized particularly
with hemodialysis treatment for chronic renal failure. The EEG Ethyl Alcohol (Ethanol)
in the dialysis encephalopathy syndrome (284) shows diffuse Hallucinosis, delirium tremens, and epileptic seizures are usu-
slow activities and spikes. However, the disease virtually has dis- ally understood as withdrawal signs following dependence and
appeared with improvement of dialysis technique. habituation. EEG activity is desynchronized, voltages are low,
and generalized spikes may be encountered, if recording occurs
Thallium within the first 48 hours after withdrawal (299,300). Seizures
Thallium has been widely used as a raticide. Ingestion of thal- and spikes reflect reduced neurotransmission in GABA and
lium occurs with suicidal or murderous intention. The clinical NMDA pathways (301). A constantly abnormal EEG in an alco-
picture and the EEG signs are essentially of nonspecific charac- holic with seizures suggests epilepsy or symptomatic seizures
ter (285). Figure 43.11A and B demonstrates the EEG course in unrelated to alcohol (302). Chronic alcohol use was not consid-
a case of criminal intoxication. ered a risk factor for a first seizure symptomatic to other etiolo-
gies (303).
Carbon Monoxide Wernicke encephalopathy, Korsakov syndrome, sensorimo-
Carbon monoxide intoxication is a frequent incident in coun- tor polyneuropathy, retrobulbar neuritis, and cerebral pellagra
tries with traditional heating. It is still the leading cause of death are related to nutritional deficiencies, mainly aneurine defi-
by poisoning in the United States (for review see Ref. 286). ciency, which serves as a cofactor in many enzyme systems. In
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 915

Figure 43.11 A: N.R., 41 years. Was


transferred to the neurologic ICU in coma-
tose state with some irregular myoclonic
jerks. Diffuse 1- to 3-Hz activities with
triphasic waves, more on the left side.
Note current artifact due to high imped-
ance T4. Some muscle artifacts due to
involuntary movements. B: Ten days after
A. Investigations by police detected a mur-
derous attempt with thallium. The condi-
tion of the patient changed to a vegetative
state. Diffuse slow activities with continu-
ously superimposed muscle artifacts and
some eye movement artifacts. The patient
died due to intercurrent infections.

the Wernicke encephalopathy the EEG is variable, showing parkinsonism (310). Persistent EEG alterations with severe
mainly diffuse slowing. With other alcohol-related cerebral dis- paroxysmal abnormalities have been reported (311).
turbances severe dysfunctions are reflected in EEG slowing.
A subacute encephalopathy in alcoholics may lead to focal Organophosphate Pesticides
seizures, transient focal cortical deficits, and PLEDs in the EEG Organophosphates exhibit anticholinesterase activity. Sources of
(304). Moreover, alcohol withdrawal has been shown to activate organophosphate pesticides are garden sheds, room sprays, baits
PLEDs in chronic focal cerebral lesions (305). Although still for insects, shampoos against head lice, and pet preparations
controversial, in some instances seizures may be directly related (312). Organophosphates were used as sarin by terrorist attacks
to alcohol intoxication (306). in Japan and found and destroyed in the Gulf war. Three different
syndromes can be recognized (313). The acute cholinergic crisis
Organic Solvents includes muscle weakness, respiratory failure, massive bronchor-
Trichloroethylene and methylbenzene (toluene) are used as rhea, epileptic seizures, coma, and death. An intermediate syn-
industrial solvents. Workers are at an occupational hazard for drome is characterized by pre- and postsynaptic neuromuscular
intoxication. A major source for intoxication represents the sol- junction failure. Late syndromes show polyneuropathy and
vent abuse by children and young adults in developing coun- parkinsonism. A highly significant prolongation of P300 latency
tries (307). Neurotoxicity leads to cranial and peripheral highlighted the possibility of the development of long-lasting
neuropathies, cerebellar, pyramidal, and extrapyramidal signs cognitive deficits following poisoning (314).
as well as cognitive decline. MR images revealed white matter EEG changes in organophosphorus poisoning show nonspe-
lesions, brain atrophy, and thalamic hypointensity (308). A cific enhancement of slow activity and paroxysmal discharges
chronic toxic encephalopathy can be recognized after excessive (14,243). According to Okonek and Rieger (315), there is a char-
occupational exposure to solvents (309). Furthermore, acteristic sequence of EEG patterns in alkyl phosphate poisoning.
trichloroethylene acts as a risk factor for the development of Unlike other acute intoxications, in a first stage of deep coma fast
916 Part VI ■ EEG: Neuropharmacology and Anesthesia

rhythmical activities were recorded. Paroxysmal discharges were 22. Utiger RD. Melatonin—the hormone of darkness. New Engl J Med.
found to be exceptional although massive myoclonic jerks and 1992;327:1377–1379 [editorial].
other convulsive manifestations occurred. 23. Dahlitz M, Alvarez B, Vignau J, et al. Delayed sleep phase syndrome
Further details about environmental toxins may be gleaned response to melatonin. Lancet. 1991;337:1121–1124.
24. Buscemi N, Vandermeer B, Hooton N, et al. Efficacy and safety of
from Lockwood (316).
exogenous melatonin for secondary sleep disorders and sleep dis-
orders accompanying sleep restriction: metaanalysis. Br Med J.
REFERENCES 2006;332:385–393.
25. Simpson D, Curran M. Ramelton: a review of its use in insomnia.
1. Nakamura Y. Pharmacogenomics and drug toxicity. New Engl J Drugs. 2008;68:1901–1919.
Med. 2008;359:856–858. 26. Dreifuss FE, Bertram EH. Bromide therapy for intractable
2. Blume WT, Kaibara M, Young GB. Atlas of Adult seizures. Epilepsia. 1986;27:593.
Electroencephalography. 2nd ed. Philadelphia: Lippincott Williams 27. Freedman R. Schizophrenia. New Engl J Med. 2003;349:1738–1749.
& Wlikins; 2002:266. 28. Saletu B. Psychopharmaka, Gehirntätigkeit und Schlaf. Bibliotheka
3. Franks NP, Lieb WR. Molecular and cellular mechanisms of gen- Psychiatrica. No. 155. Basel: S. Karger; 1976.
eral anesthesia. Nature. 1994;367:607–613. 29. Centorrino F, Price BH, Tuttle M, et al. EEG abnormalities during
4. Campagna JA, Miller KW, Forman SA. Mechanisms of action of treatment with typical and atypical antipsychotics. Am J
inhaled anesthetics. New Engl J Med. 2003;348:2110–2124. Psychiatry. 2002;159:109–115.
5. Winters WD. Effects of drugs on the electrical activity of the brain: 30. Alldredge BK. Seizure risk associated with psychotropic drugs:
anesthetics. Annu Rev Pharmacol Toxicol. 1976;16:413–426. clinical and pharmacokinetic considerations. Neurology.
6. Sloan TB. Anesthetic effects on electrophysiologic recordings 1999;53(suppl 2):S68–S75.
[review articles]. J Clin Neurophysiol. 1998;15:217–226. 31. Pisani F, Oteri G, Costa C, et al. Effects of psychotropic drugs on
7. Chatrian GE. Intraoperative electrocorticography. In: Ebersole JS, seizure threshold. Drug Saf. 2002;25:91–110.
Pedley TA, eds. Current Practice in Clinical Electroencephalography. 32. Van Sweden B. Toxic “ictal” confusion in middle age: treatment with
3rd ed. Lippincott Williams & Wilkins; 2003:681–712. benzodiazepines. J Neurol Neurosurg Psychiatry. 1985;48:472–476.
8. Kwan P, Brodie MJ. Phenobarbital for the treatment of epilepsy in 33. Leucht St, Wahlbeck K, Hamann J, et al. New generation antipsy-
the 21st century: a critical review. Epilepsia. 2004;45:1141–1149. chotics versus low-potency conventional antipsychotics: a system-
9. McKernan RM, Whiting PJ. Which GABA-A receptor subtypes atic review and meta-analysis. Lancet. 2003;361:1581–1589.
really occur in the brain? Trends Neurosci. 1996;19:139–143. 34. Schmidt CJ, Sorensen SM, Kehne JH, et al. The role of 5-HT2a
10. Ban TA, Amin M. Hypnotics, minor tranquilizers and sedatives. In: receptors in antipsychotic activity. Life Sci. 1995;56:2209–2222.
Vinken PJ, Bruyn GW, eds. Handbook of Clinical Neurology. Vol 37. 35. Alvir JMJ, Liebermann JA, Saffermann AZ, et al. Clozapine-
Amsterdam: North Holland; 1979;347–364. induced agranulocytosis: incidence and risk factors in the United
11. Gotman J, Gloor P, Quesney LF, et al. Correlations between EEG States. New Engl J Med. 1993;329:162–167.
changes induced by diazepam and the localization of epileptic 36. Freudenreich O, Weiner RD, McEvoy JP. Clozapine-induced elec-
spikes and seizures. Electroencephalogr Clin Neurophysiol. troencephalogram changes as a function of clozapine serum levels.
1982;54:614–621. Biol Psychiatry. 1997;42:132–137.
12. Trimble MR. Neurobehavioral effects of anticonvulsants. JAMA. 37. Joutsiniemi S-L, Gross A, Appelberg B. Marked clozapine-induced
1991;13:1307–1308. slowing of EEG background over frontal, central, and parietal
13. Cramer JA, Mattson RH. Phenobarbital toxicity. In: Levy RH, scalp areas in schizophrenic patients. J Clin Neurophysiol.
Mattson RH, Meldrum BS, eds. Antiepileptic Drugs. 4th ed. New 2001;18:9–13.
York: Raven Press; 1955:409–420. 38. Baldessarini RJ, Frankenburg FR. Clozapine: a novel antipsychotic
14. Kurtz D. The EEG in acute and chronic drug intoxications. In: agent. N Engl J Med. 1991;324:746–754.
Glaser GH, ed. Metabolic, Endocrine and Toxic Diseases/Handbook 39. Alper K, Schwartz KA, Kolts RL, et al. Seizure incidence in psy-
of Electroencephalography and Clinical Neurophysiology. Vol 15. chopharmacological clinical trials: an analysis of Food and Drug
Amsterdam: Elsevier; 1976:88–104. Administration (FDA) summary basis of approval reports. Biol
15. Plum F, Posner JP. Diagnosis of Stupor and Coma. 2nd ed. Psychiatry. 2007;62:345–354.
Philadelphia: F.A. Davis; 1972. 40. Sajatovic M, Meltzer HY. Clozapine-induced myoclonus and gen-
16. Greenberg DA, Simon RP. Flexor and extensor postures in sedative eralized seizures. Biol Psychiatry. 1996;39:367–370.
drug-induced coma. Neurology. 1982;32:448–451. 41. Mortimer AM. Newer and older antipsychotics. A comparative
17. Kuroiwa Y, Furukawa T, Inaki K. Recovery from drug-induced review of appropriate use. CNS Drugs. 1994;2:381–396.
alpha coma. Neurology. 1981;31:1359–1361. 42. Cunnigham Owens DG. Adverse effects of antipsychotic agents.
18. Ellington AL. Electroencephalographic pattern of burst suppres- Drugs. 1996;51:895–930.
sion in a case of barbiturate coma. Electroencephalogr Clin 43. Frazer A. Pharmacology of antidepressants. J Clin Pharmacol.
Neurophysiol. 1968;25:491–493. 1997;17(suppl 1):S1–S18.
19. Kubicki S, Rieger H, Busse G, et al. Elektroencephalographische 44. Sindrup SH, Jensen TS. Pharmacologic treatment of pain in
Befunde bei schweren Schlafmittelvergiftungen. Z EEG-EMG. polyneuropathy. Neurology. 2000;55:915–920.
1970;1:80–93. 45. Itil TM. Convulsive and anticonvulsive properties of neuro-psy-
20. Bennett DR, Hughes JR, Korein J, et al. Atlas of cho-pharmaca. In: Niedermeyer E, ed. Epilepsy. Modern Problems
Electroencephalography in Coma and Cerebral Death. New York: in Pharmacopsychiatry. Vol 4. Basel: Karger; 1970:270–305.
Raven Press; 1976. 46. Kugler J, Lorenzi E, Spatz R, et al. Drug-induced paroxysmal EEG
21. Haider J, Matthew H, Oswald J. Electroencephalographic changes activities. Pharmacopsychiatry. 1979;12:165–172.
in acute drug poisoning. Electroencephalogr Clin Neurophysiol. 47. Jabbari B, Byran GE, Marsh EE, et al. Incidence of seizures with tri-
1971;30:23–31. cyclic and tetracyclic antidepressants. Arch Neurol. 1985;42:480–481.
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 917

48. Skowron DM, Stimmel GL. Antidepressants and the risk of 74. Greenblatt DJ, Harmatz JS, Shapiro L, et al. Sensitivity to triazolam
seizures. Pharmacotherapy. 1992;12:18–22. in the elderly. N Engl J Med. 1991;324:1961–1698.
49. Rumpl E, Hinterhuber H. Unusual “spike-wave stupor” in a 75. Bruni J, Wilder BJ. The toxicology of antiepileptic drugs. In:
patient with manic-depressive psychosis treated with amitripty- Vinken PJ, Bruyn GW, eds. Handbook of Clinical Neurology. Vol 37.
line. J Neurol. 1981;226:131–135. Amsterdam: North Holland; 1979:199–222.
50. Thomas P, Beaumanoir A, Genton P, et al. “De novo” absence sta- 76. Mattson RH. Other antiepileptic drugs. Benzodiazepines. In:
tus of late onset: report of 11 cases. Neurology. 1992;42:104–110. Woodbury DM, Penry JK, Schmidt RP, eds. Antiepileptic Drugs.
51. Buckley NA, McManus PR. Fatal toxicity of serotoninergic and New York: Raven Press; 1972:497–518.
other antidepressant drugs: analysis of United Kingdom mortality 77. Langtry HD, Benfield P. Zolpidem: a review of its pharmacody-
data. Br Med J. 2002;325:1332–1333. namic and pharmacokinetic properties and therapeutic potential.
52. Schmitz B. Antidepressant drugs: indications and guidelines for Drugs. 1990;40:291–313.
use in epilepsy. Epilepsia. 2002;43(suppl 2):14–18. 78. Allain H, Delahaye CH, LeCoz F, et al. Postmarketing surveillance
53. Boyer EW, Shannon M. The serotonin syndrome. N Engl J Med. of zopiclone in insomnia: analysis of 20513 cases. Sleep.
2005;352:1112–1120. 1991;14:408–413.
54. Dike GL. Triphasic waves in serotonin syndrome. J Neurol 79. Parrino L, Boselli M, Spaggiari MC, et al. Multidrug comparison
Neurosurg Psychiatry. 1997;62:200. (lorazepam, triazolam, zolpidem, and zopiclone) in situational
55. Fetzer J, Kader G, Danahy S. Lithium encephalopathy: a clinical, insomnia: polysomnographic analysis by means of the cyclic alter-
psychiatric and EEG evaluation. Am J Psychiatry. 1981;138: nating pattern. Clin Neuropharmacol. 1997;20:253–263.
1622–1623. 80. Misgeld U, Bijak M, Jarolimek W. A physiological role for GABA-
56. Mayfield D, Brown RG. The clinical, laboratory and electroen- B receptors and the effects of baclofen in the mammalian central
cephalographic effects of lithium. J Psychiatr Res. 1966;4:207–219. nervous system. Prog Neurobiol. 1995;46:423–462.
57. Lee MS, Lessel S. Lithium-induced periodic alternating nystagmus. 81. Kofler M, Kronenberg MF, Rifici C, et al. Epileptic seizures associated
Neurology. 2003;60:344. with intrathecal baclofen application. Neurology. 1994;44:25–27.
58. Sansone MEG, Ziegler DK. Lithium toxicity: a review of neuro- 82. Schuele SU, Kellinghaus C, Shook SJ, et al. Incidence of seizures in
logic complications. Clin Neuropharmacol. 1985;8:242–248. patients with multiple sclerosis treated with intrathecal baclofen.
59. Caviness JN, Evidente JN. Cortical myoclonus during lithium Neurology. 2005;63:1086–1087.
exposure. Arch Neurol. 2003;60:401–404. 83. Fakhoury T, Abou-Khalil B, Blumenkopf B. EEG changes in
60. Casanova B, de Entrambasaguas M, Perla C, et al. Lithium-induced intrathecal baclofen overdose: a case report and review of the liter-
Creutzfeldt–Jakob syndrome. Clin Neuropharmacol. 1996;19:356–359. ature. EEG Clin Neurophysiol. 1998;107:339–342.
61. Lee SI. Nonconvulsive status epilepticus. Ictal confusion in later 84. Chou I, Narasimhan K. Neurobiology of addiction. Nat Neurosci.
life. Arch Neurol. 1985;42:778–781. 2005;8:1427.
62. Kaplan PW, Birbeck G. Lithium-induced confusional states: non- 85. Brown BB. Subjective and EEG responses to LSD in visualizer and
convulsive status epilepticus or triphasic encephalopathy. nonvisualizer subjects. Electroencephalogr Clin Neurophysiol.
Epilepsia. 2006;47:2071–2074. 1968;25:372–379.
63. Kores B, Lader MH. Irreversible lithium neurotoxicity: an 86. Iversen L. Cannabis and the brain. Brain. 2003;126:1252–1270.
overview. Clin Neuropharmacol. 1997;20:283–299. 87. Messinis L, Kyprianidou A, Malefaki S, et al. Neuropsychological
64. Low MD. Evaluation of psychiatric disorders and the effects of deficits in long-term frequent cannabis users. Neurology.
psychotherapeutic and psychomimetic agents. In: Klass DW, Daly 2006:737–739.
DD, eds. Current Practice of Clinical Electroencephalography. 88. Nordentoft M, Hjorthoj C. Cannabis use and risk of psychosis in
1979:395–410. later life. Lancet. 2007;370:293–294.
65. Gardner CR, Tully RW, Hedgecock JR. The rapidly expanding 89. Karst M, Salim K, Burstein S, et al. Analgesic effect of the synthetic
range of neuronal benzodiazepine receptor ligands. Prog cannabinoid CT-3 on chronic neuropathic pain. JAMA.
Neurobiol. 1993;40:1–61. 2003;290:1757–1762.
66. Doble A, Martin IL. Multiple benzodiazepine receptors: no reason 90. Rodin EA, Domino EF, Porzak JP. The marihuana-induced “social
for anxiety. TiPS. 1992;13:76–81. high.” Neurological and electroencephalographic concomitants.
67. Niedermeyer E. Electroencephalographic studies on the anticon- JAMA. 1970;213:1800.
vulsive action of diazepam. Eur Neurol. 1970;3:88–96. 91. Herning RI, Better W, Cadet JL. EEG in chronic marijuana users dur-
68. Mitsudone A, Ohfu M, Yasumoto S, et al. The effectiveness of clon- ing abstinence: relationship to years of marijuana use, cerebral blood
azepam on the rolandic spikes. Brain Dev. 1997;19:274–278. flow and thyroid function. Clin Neurophysiol. 2008;119:321–331.
69. Riss J, Cloyd J, Gates J, et al. Benzodiazepines in epilepsy: pharma- 92. Nausieda PA. Central stimulant toxicity. In: Vinken PJ, Bruyn GW,
cology and pharmacokinetics. Acta Neurol Scand. 2008;118:69–86. eds. Handbook of Clinical Neurology. Vol 37. Amsterdam: North
70. Prior PF, Maclaine GN, Scott DF, et al. Tonic status epilepticus pre- Holland; 1979:223–297.
cipitated by intravenous diazepam in a child with petit mal status. 93. Ricaurte GA, McCann UD. Recognition and management of compli-
Epilepsia. 1972;13:467–472. cations of new recreational drug use. Lancet. 2005;365:2137–2145.
71. Tassinari CA, Dravet C, Roger JA, et al. Tonic status epilepticus by 94. US Modafinil in Narcolepsy Multicenter Study Group.
intravenous benzodiazepine in five patients with Lennox–Gastaut Randomized trial of modafinil as a treatment for the excessive day-
syndrome. Epilepsia. 1972;13:421–435. time somnolence of narcolepsy. Neurology. 2000;54:1166–1175.
72. Treiman DM, Meyers PD, Walton NY, et al., for the Veterans Affairs 95. Clarke AR, Barry RJ, McCarty R, et al. Effects of stimulant medica-
Status Epilepticus Cooperative Study Group. A comparison of four tions on the EEG of girls with attention-deficit/hyperactivity dis-
treatments for generalized convulsive status epilepticus. N Engl J order. Clin Neurophysiol. 2007;118:2700–2708.
Med. 1998;339:792–798. 96. Allen AJ, Kurlan RM, Gilbert DL, et al. Atomoxetine treatment in
73. Bixler EO, Kales A, Manfredi RL, et al. Next-day memory impair- children and adolescents with ADHD and comorbid tic disorders.
ment with triazolam use. Lancet. 1991;337:827–831. Neurology. 2005;65:1941–1949.
918 Part VI ■ EEG: Neuropharmacology and Anesthesia

97. Office of Applied Studies. Preliminary Results from the 1996 120. Duncan JS. Antiepileptic drugs and the electroencephalogram.
National Household Survey on Drug Abuse. Substance Abuse and Epilepsia. 1987;28:259–266.
Mental Health Administration; 1996. 121. Kuruvilla Th, Bharucha NE. Cerebellar atrophy after acute
98. Schifano F, Oyefeso A, Webb L, et al. Review of deaths related to phenytoin intoxication. Epilepsia. 1997;38:500–502.
taking ecstasy, England and Wales, 1997–2000. Br Med J. 122. Luef G, Burtscher J, Kremser Ch, et al. Magnetic resonance vol-
2003;326:80–81. umetry of the cerebellum in epileptic patients after phenytoin
99. Ernst Th, Chang L, Leonido-Yee M, et al. Evidence for long-term overdosage. Eur Neurol. 1996;36:273–277.
neurotoxicity associated with methamphetamine abuse. 123. Bauer J. Seizure-inducing effects of antiepileptic drugs. Acta
Neurology. 2000;54:1344–1349. Neurol Scand. 1996;4:367–377.
100. Leshner AI. Molecular mechanisms of cocaine addiction. N Engl 124. Perucca E, Gram L, Avanzini G, et al. Antiepileptic drugs as a
J Med. 1996;335:128–129. cause of worsening seizures. Epilepsia. 1998;39:5–17.
101. Lüscher Ch, Bellone C. Cocaine-evoked synaptic plasticity: a key 125. Roseman E. Dilantin toxicity: a clinical and electroencephalo-
to addiction? Nat Neurosci. 2008;11:737–738. graphic study. Neurology (Minneapolis). 1961;11:912–921.
102. Noble EP. D2 dopamine receptor gene in psychiatric and neuro- 126. Wilkus RJ, Dodrill CB, Troupin AS. Carbamazepine and the elec-
logic disorders and its phenotypes. Am J Med Genet. troencephalogram of epileptics: a double-blind study in compar-
2003;116B:103–125. ison to phenytoin. Epilepsia. 1978;25:467–475.
103. Herning RI, Jones RT, Hooker WD, et al. Cocaine increases EEG 127. Carrie JRG. Computer-assisted EEG sharp transient detection
beta: a replication and extension of Hans Berger’s historic and quantification during overnight recordings in an epileptic
experiments. Electroencephalogr Clin Neurophysiol. 1985;60: patient. In: Kellaway P, Petersen I, eds. Quantitative Analytic
470–477. Studies in Epilepsy. New York: Raven Press; 1976:225–235.
104. Caplan LR, Heir DB, Banks G. Stroke and drug abuse. Stroke. 128. Buchthal F, Svensmark O, Schiller PJ. Clinical and electroen-
1982;13:869–872. cephalographic correlations with serum levels of diphenylhydan-
105. Globe LI, Merkin M. Cerebral infarction in a user of free-base toin. Arch Neurol. 1960;2:624–630.
cocaine (“crack”). Neurology. 1986;36:1602–1604. 129. Spencer SS, Spencer DD, Williamson PD, et al. Ictal effects of
106. Mody CK, Miller BL, McIntyre HB, et al. Neurologic complica- anticonvulsant medication withdrawal in epileptic patients.
tions of cocaine abuse. Neurology. 1988;38:1189–1193. Epilepsia. 1981;22:297–307.
107. Qureshi AI, Akbar MS, Czander E, et al. Crack cocaine use and 130. Ambrosetto C, Tassinari CA, Baruzzi A, et al. Phenytoin
stroke in young patients. Neurology. 1997;48:341–345. encephalopathy as probable idiosyncratic reaction: case report.
108. Van Stavern GP, Gorman M. Orbital infarction after cocaine use. Epilepsia. 1977;18:405–408.
Neurology. 2002;59:642–643. 131. Besser R, Hornung K, Theison M, et al. EEG changes in patients
109. Aggarwal SK, Williams V, Levine SR, et al. Cocaine-associated during the introduction of carbamazepine. Electroencephalogr
intracranial hemorrhage: absence of vasculitis in 14 cases. Clin Neurophysiol. 1992;83:19–23.
Neurology. 1996;46:1741–1743. 132. Saunders MG, Westmoreland BF. The EEG in the evaluation of
110. Nolte KB, Brass LM, Fletterick CF. Intracranial hemorrhage asso- disorders affecting the brain diffusely. In: Klass DW, Daly DD,
ciated with cocaine abuse: a prospective autopsy study. eds. Current Practice of Clinical Electroencephalography. New
Neurology. 1996;46:1291–1296. York: Raven Press; 1979:343–379.
111. Conway JE, Tamergo RJ. Cocaine use is an independent risk fac- 133. Dong X, Leppik IE, White J, et al. Hyponatremia from oxcar-
tor for cerebral vasospasm after aneurysmal subarachnois hem- bazepine and carbamazepine. Neurology. 2005;65:1976–1978.
orrhage. Stroke. 2001;32:2338–2343. 134. Kochen S, Giagante B, Oddo S. Spike-and-wave complexes and
112. Krendel DA, Ditter SM, Frankel MR, et al. Biopsy-proven cerebral seizure exacerbation caused by carbamazepine. Eur J Neurol.
vasculitis associated with cocaine abuse. Neurology. 2002;9:41–47.
1990;40:1092–1094. 135. Prasad AN, Stefanelli M, Nagarajan L. Seizure exacerbation and
113. Weiner WJ, Rabinstein A, Levin B, et al. Cocaine-induced persist- developmental regression with carbamazepine. Can J Neurol Sci.
ent dyskinesias. Neurology. 2001;56:964–965. 1998;25:287–294.
114. Migita DS, Devereaux MW, Tomsak RL. Cocaine and pupillary- 136. Talwar D, Arora MS, Sher PK. EEG changes and seizure exacerba-
sparing oculomotor nerve palsies. Neurology. 1997;49: tion in young children treated with carbamazepine. Epilepsia.
1466–1467. 1994;35:1154–1159.
115. Ogunyemi AO, Locke GE, Kramer LD, et al. Complex partial sta- 137. De Rubeis AA, Young GB. Continuous EEG monitoring in a
tus epilepticus provoked by “crack” cocaine. Ann Neurol. patient with massive carbamazepine overdose. J Clin
1989;26:785–786. Neurophysiol. 2001;18:166–168.
116. Levy RH, Mattson RH, Meldrum BS, et al., eds. Antiepileptic 138. Meador KJ, Loring DW, Huh K, et al. Comparative cognitive
Drugs. 5th ed. Philadelphia: Lippincott Williams & Wilkins; effects of anticonvulsants. Neurology (Minneapolis).
2002. 1990;40:391–394.
117. Herkes GK, Lagerlund TD, Sharbrough FW, et al. Effects of 139. Dodrill CB, Troupin AS. Neuropsychological effects of carba-
antiepileptic drug treatment on the background frequency of mazepine and phenytoin: a reanalysis. Neurology. 1991;41:
EEGs in epileptic patients. J Clin Neurophysiol. 1993;10:210–216. 141–143.
118. Salinsky MC, Oken BS, Morehead L. Intraindividual analysis of 140. Leslie PH, Heyworth R, Prescott LF. Cardiac complications of
antiepileptic drug effects on EEG background rhythms. carbamazepine intoxication: treatment by hemoperfusion. Br J
Electroencephalogr Clin Neurophysiol. 1994;90:186–193. Med. 1983;286:1018.
119. Salinsky MC, Oken BS, Storzbach D, et al. Assessment of CNS 141. Grant SM, Faulds D. Oxcarbazepine. A review of its pharmacol-
effects of antiepileptic drugs by using quantitative EEG measures. ogy and therapeutic potential in epilepsy, trigeminal neuralgia
Epilepsia. 2003;44:1042–1050. and affective disorders. Drugs. 1992;43:873–888.
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 919

142. Gelisse P, Genton P, Kuate C, et al. Worsening of seizures by 163. Marciani MG, Stantione P, Maschio M, et al. EEG changes
oxcarbazepine in juvenile generalized epilepsies. Epilepsia. induced by vigabatrin monotherapy in focal epilepsy. Acta Neurol
2004;45:1282–1286. Scand. 1997;95:115–120.
143. Venrdrame M, Khurana DS, Cruz M, et al. Aggravation of 164. Hammond EJ, Wilder BJ. Effects of gamma-vinyl-GABA on the
seizures and/or EEG features in children treated with oxcar- human electroencephalogram. Neuropharmacology. 1985;24:
bazepine monotherapy. Epilepsia. 2007;48:2116–2120. 975–984.
144. Jones AL, Proudfoot AT. Features and management of poisoning 165. Kälviäinen R, Keränen T, Riekkinen PJ. Place of newer
with modern drugs used to treat epilepsy. Q J Med. 1998;91: antiepileptic drugs in the treatment of epilepsy. Drugs.
325–332. 1993;46:1009–1024.
145. Clemens B. Valproate decreases EEG synchronization in a use- 166. Mervaala E, Partanen J, Nousiainen U, et al. Electrophysiologic
dependent manner in idiopathic generalized epilepsy. Seizure. effects of gamma-vinyl GABA and carbamazepine. Epilepsia.
2008;17:224–233. 1989;30:189–193.
146. Adams DJ, Lüders H, Pippenger Ch. Sodium valproate in 167. Ben-Menachem E, Treiman DM. Effect of gamma-vinyl GABA
the treatment of intractable seizure disorders: a clinical and on interictal spikes and sharp waves in patients with intractable
electroencephalographic study. Neurology (Minneapolis). complex partial seizures. Epilepsia. 1989;30:79–83.
1978;28:152–157. 168. Luna D, Dulac O, Pajot N, et al. Vigabatrin in the treatment of
147. Braathen G, Theorell K, Persson A, et al. Valproate in the treat- childhood epilepsies: a single-blind placebo-controlled study.
ment of absence epilepsy in children: a study of dose–response Epilepsia. 1989;30:430–437.
relationship. Epilepsia. 1988;29:548–552. 169. Marciani MG, Maschio M, Spanedda F, et al. Development of
148. Bruni J, Wilder BJ, Baumann AW, et al. Clinical efficacy and long- myoclonus in patients with partial epilepsy during treatment
term effects of valproic acid therapy on spike-and-wave dis- with vigabatrin: an electroencephalographic study. Acta Neurol
charges. Neurology. 1980;30:42–46. Scand. 1995;91:1–5.
149. Sato S, White BG, Penry JK, et al. Valproic acid versus ethosux- 170. Michelucci R, Tassinari CA. Response to vigabatrin in relation to
imide in the treatment of absence seizures. Neurology. seizure type. Br J Clin Pharmacol. 1989;27:119–124.
1982;32:157–163. 171. Sälke-Kellermann A, Baier H, Rambeck B, et al. Acute
150. Harding GFA, Herrick CE, Jeavons PM. A controlled study of the encephalopathy with vigabatrin. Lancet. 1993;342:185 [letter].
effect of sodium valproate on photosensitive epilepsy and its 172. Sharief MK, Sander JWA, Shorvon SD. Acute encephalopathy
prognosis. Epilepsia. 1978;19:555–565. with vigabatrin. Lancet. 1993;342:619 [letter].
151. Simon D, Penry JK. Sodium-di-N-propylacetate (DPA) in the 173. Sander JW, Hart YM, Trimble MR, et al. Vigabatrin and psy-
treatment of epilepsy. A review. Epilepsia. 1975;16:549–573. chosis. J Neurol Neurosurg Psychiatry. 1991;54:435–439.
152. Dreifuss FE. Valproic acid. Toxicity. In: Levy RH, Mattson RH, 174. Malmgren K, Ben-Menachem E, Frisén L. Vigabatrin and visual
Meldrum BS, eds. Antiepileptic Drugs. 4th ed. Philadelphia: toxicity: evolution and dose dependence. Epilepsia. 2001;42:
Lippincott-Raven; 1995:641–648. 609–615.
153. Segura-Bruna N, Rodriguez-Campello A, Puente V, et al. 175. Camposano SE, Major P, Halpern E, et al. Vigabatrin in the treat-
Valproate-induced hyperammonemic encephalopathy. Acta ment of childhood epilepsy: a retrospective chart review of effi-
Neurol Scand. 2006;114:1–7. cacy and safety profile. Epilepsia. 2008;49:1186–1191.
154. Kifune A, Kubota F, Shibata N, et al. Valproic acid-induced hyper- 176. Conway M, Cubbidge RP, Hosking SL. Visual field severity
ammonemic encephalopathy with triphasic waves. Epilepsia. indices demonstrate dose-dependent visual loss from vigabatrin
2000;41:909–912. therapy. Epilepsia. 2008;49:108–116.
155. Dreifuss FE, Santili N, Langer DH, et al. Valproic acid fatalities: a 177. Fitton A, Goa KL. Lamotrigine. An update of its pharmacology
retrospective review. Neurology. 1987;37:379–385. and therapeutic use in epilepsy. Drugs. 1995;50:691–713.
156. Koenig SA, Buessing D, Longin E, et al. Valproic acid-induced 178. Mervaala E, Koivisto K, Hänninen T. Electrophysiological and
hepatopathy: nine new fatalities in Germany from 1994 to 2003. neuropsychological profiles of lamotrigine in young and age-
Epilepsia. 2006;47:2027–2031. associated memory impairment (AAMI) subjects. Neurology.
157. Scheffner D, König S, Rauterberg-Ruland I, et al. Fatal liver fail- 1995;46(suppl 4):S259.
ure in 16 children with valproate therapy. Epilepsia. 1988;29: 179. Foletti G, Volanschi D. Influence of lamotrigine addition on
530–542. computerized background EEG parameters in severe epilepto-
158. Capocchi G, Balducci A, Cecconi M, et al. Valproate-induced genic encephalopathies. Eur Neurol. 1994;34(suppl 1):S87–S89.
epileptic tonic status. Seizure. 1998;7:237–241. 180. Besag FM. Use of the “monolog” spike wave monitor to evaluate
159. Pindar RM, Brogden RN, Speight TM, et al. Clonazepam: a lamotrigine for absence seizures. Epilepsia. 1991;32(suppl
review of its pharmacological properties and therapeutic efficacy 1):S89–S90 [abstract].
in epilepsy. Drugs. 1976;12:321–361. 181. Binnie CD, van Emde-Boas W, Kasteleijn-Nolste-Trenite DG.
160. Petsche H. Zum Nachweis des kortikalen Angriffspunktes des Acute effects of lamotrigine (BW 430C) in persons with epilepsy.
antikonvulsiven Benzodiazepinderivats Clonazepam (Ro 5– Epilepsia. 1986;27:248–254.
4023). Z EEG-EMG. 1972;3:145. 182. Erikson AS, Knudson E, Nergardth A. The effect of lamotrigine
161. Martin D, Hirt HR. Clinical experience with clonazepam on epileptiform discharges in young patients with drug-resistant
(Rivotril) in the treatment of epilepsies in infancy and childhood. epilepsy. Epilepsia. 2001;42:230–236.
Neuropediatrie. 1973;4:245–266. 183. Biraben A, Allain H, Scarabin JM, et al. Exacerbation of juvenile
162. Grant SM, Heel RC. Vigabatrin. A review of its pharmacody- myoclonic epilepsy with lamotrigine. Neurology. 2000;55:1758.
namic and pharmacokinetic properties, and therapeutic poten- 184. Guerrini R, Dravet C, Genton P, et al. Lamotrigine and seizure
tial in epilepsy and disorders of motor control. Drugs. aggravation in severe myoclonic epilepsy. Epilepsia. 1998;39:
1991;41:889–926. 508–512.
920 Part VI ■ EEG: Neuropharmacology and Anesthesia

185. Dinnerstein E, Jobst BC, Williamson PD. Lamotrigine intoxica- 207. Laech JP, Stolarek I, Brodie MJ. Deliberated overdose with the
tion provoking status epilepticus in an adult with localization- novel anticonvulsant tiagabine. Seizure. 1995;4:155–157.
related epilepsy. Arch Neurol. 2007;64:1344–1346. 208. Coenen AML, Blezer EHM, van Luijtelaar ELJM. Effects of the
186. Trinka E, Dilitz E, Unterberger I, et al. Non convulsive status GABA-uptake inhibitor tiagabine on electroencephalogram,
epilepticus after replacement of valproate with lamotrigine. J spike-wave discharges and behavior of rats. Epilepsy Res.
Neurol. 2002;249:1417–1422. 1995;21:89–94.
187. Buckley NA, Whyte IM, Dwason AH. Self-poisoning with lamot- 209. Walton NY, Gunawan S, Treiman DM. Treatment of experimen-
rigine. Lancet. 1993;342:1552–1553 [letter]. tal status epilepticus with the GABA uptake inhibitor, tiagabine.
188. Taylor CP. Gabapentin. Mechanisms of action. In: Levy RH, Epilepsy Res. 1994;19:237–244.
Mattson RH, Meldrum BS, et al., eds. Antiepileptic Drugs. 5th ed. 210. Koepp MJ, Edwards M, Collins J, et al. Status epilepticus and
Philadelphia: Lippincott Williams & Wilkins; 2002:321–339. tiagabine revisited. Epilepsia. 2005;46:1625–1632.
189. Mathew NT, Rapoport A, Saper J, et al. Efficacy of gabapentin in 211. Cross JH. Topiramate. In: Shorvon S, Perucca E, Fish D, et al., eds.
migraine prophylaxis. Headache. 2001;41:119–128. The Treatment of Epilepsy. 2nd ed. Oxford: Blackwell; 2003:
190. Spira PJ, Beran RG. Gabapentin in the prophylaxis of chronic 535–547.
daily headache. Neurology. 2003;61:1753–1759. 212. Meador KJ, Loring DW, Hulihan JF, et al. Differential cognitive
191. Backonja M, Beydoun A, Edwards KR, et al. Gabapentin for the and behavioral effects of topiramate and valproate. Neurology.
symptomatic treatment of painful neuropathy in patients with 2003;60:1483–1488.
diabetes mellitus. JAMA. 1998;280:1831–1836. 213. Blum D, Meador K, Biton V, et al. Cognitive effects of lamotrigine
192. Greist JH. Gabapentin. Clinical efficacy and use in psychiatric compared with topiramate in patients with epilepsy. Neurology.
disorders. In: Levy RH, Mattson RH, Meldrum BS, et al., eds. 2006;67:400–406.
Antiepileptic Drugs. 5th ed. Philadelphia: Lippincott Williams & 214. Gross-Tour V, Shalev RS. Reversible language regression as an
Wilkins; 2002:349–353. adverse effect of topiramate treatment in children. Neurology.
193. Ramsey RE, Pryor FM. Gabapentin. Toxicity. In: Levy RH, 2004;62:299–300.
Mattson RH, Meldrum BS, et al., eds. Antiepileptic Drugs. 5th ed. 215. Mula M, Trimble MR, Thompson P, et al. Topiramate and word-
Philadelphia: Lippincott Williams & Wilkins; 2002:354–359. finding difficulties in patients with epilepsy. Neurology.
194. Fischer JH, Barr AN, Rogers SL, et al. Lack of serious toxicity fol- 2003;60:1104–1107.
lowing gabapentin overdose. Neurology. 1994;44:982–983. 216. Bonanni E, Galli R, Maestri M, et al. Daytime sleepiness in
195. Salinsky MC, Binder LM, Oken BS, et al. Effects of gabapentin epilepsy patients receiving topiramate monotherapy. Epilepsia.
and carbamazepine on the EEG and cognition in healthy volun- 2004;45:333–337.
teers. Epilepsia. 2002;43:482–490. 217. Dooley M, Plosker GL. Levetiracetam. A review of its adjunctive
196. Foldvary-Schaefer N, De Leon Sanchez I, Karafa M, et al. use in the management of partial seizures. Drugs. 2000;60:
Gabapentin increases slow-wave sleep in normal adults. 871–893.
Epilepsia. 2002;43:1493–1497. 218. Sharpe DV, Patel AD, Abou-Khalil B, et al. Levetiracetam
197. Taylor CP, Angelotti T, Fauman E. Pharmacology and mechanism monotherapy in juvenile myoclonic epilepsy. Seizure.
of action of pregabalin: the calcium channel 2– (alpha2–delta) 2008;17:64–68.
subunit as a target for antiepileptic drug discovery. Epilepsy Res. 219. Specchio N, Boero G, Michelucci R, et al. Effects of levetirazetam
2007;73:137–150. on EEG abnormalities in juvenile myoclonic epilepsy. Epilepsia.
198. Beydoun A, Uthman BM, Kugler AR, et al. Safety and efficacy of 2008;49:663–669.
two pregabalin regimens for add-on treatment of partial epilepsy. 220. Striano P, Sofia V, Capovilla G, et al. A pilot trial of levetiracetam
Neurology. 2005;64:475–480. in eyelid myoclonia with absences (Jeavons syndrome). Epilepsia.
199. Lesser H, Sharma U, LaMoreaux L, et al. Pregabalin relieves 2008;49:425–430.
symptoms of painful diabetic neuropathy. Neurology. 221. Sohn YH, Kaelin-Lang A, Jung HY, et al. Effect of levetiracetam
2004;63:2104–2110. on human corticospinal excitability. Neurology. 2001;57:858–863.
200. Asconape JJ, Hartman LM, Salanova V. Pregabalin-associated 222. Epstein CM, Girad-Siqueira L, Ehrenberg JA. Prolonged neuro-
myoclonus. Epilepsia. 1999;40(suppl 7):143. physiologic effects of levetiracetam after oral administration in
201. Huppertz HJ, Feuerstein TJ, Schulze-Bonhage A. Myoclonus in humans. Epilepsia. 2008;49:1169–1173.
epilepsy patients with anticonvulsive add-on therapy with prega- 223. French J, di Nicola S, Arrigo C. Fast and sustained efficacy of lev-
balin. Epilepsia. 2001;42:790–792. etiracetam during titration and the first 3 months of treatment in
202. Knake S, Klein KM, Hattemer K, et al. Pregabalin-induced gener- refractory epilepsy. Epilepsia. 2005;46:1304–1307.
alized myoclonic status epilepticus in patients with chronic pain. 224. Stodieck S, Steinhoff BJ, Kolmsee S, et al. Effect of levetiracetam
Epilepsy Behav. 2007;11:471–473. in patients with epilepsy and interictal epileptiform discharges.
203. Oaklander AL, Buchbinder BR. Pregabalin-withdrawal Seizure. 2001;10:583–587.
encephalopathy and splenial edema: a link to high-altitude ill- 225. Bazil CW, Battista J, Basner RC. Effects of levetiracetam on sleep
ness? Ann Neurol. 2005;58:309–312. in normal volunteers. Epilepsy Behav. 2005;7:539–542.
204. Felbamate Study Group in the Lennox–Gastaut Syndrome. 226. Mula M, Trimble MR, Sander JW. Are psychiatric adverse events
Efficacy of felbamate in childhood epileptic encephalopathy of antiepileptic drugs a unique entity? A study on topiramate and
(Lennox–Gastaut syndrome). N Engl J Med. 1993;328:29–33. levetiracetam. Epilepsia. 2007;48:2322–2326.
205. Pellock JM, Faught E, Leppik IE, et al. Felbamate: consensus of 227. Bauer J. Encephalopathy induced by levetiracetam added to val-
current clinical practice. Epilepsy Res. 2006;71:89–101. proate. Acta Neurol Scand. 2008;117:374–376.
206. Adkins JC, Noble St. Tiagabine. A review of its pharmacody- 228. Macdonald RL. Zonisamide. Mechanism of action. In: Levy RH,
namic and pharmacokinetic properties and therapeutic potential Mattson RH, Meldrum BS, et al., eds. Antiepileptic Drugs. 5th ed.
in the management of epilepsy. Drugs. 1998;55:437–460. Philadelphia: Lippincott Williams & Wilkins; 2002:867–872.
Chapter 43 ■ EEG, Drug Effects, and Central Nervous System Poisoning 921

229. Leppik IE, Willmore LJ, Homan RW, et al. Efficacy and safety of 252. Kilickap S, Cakar M, Onal IK, et al. Nonconvulsive status epilep-
zonisamide: results of a multicenter study. Epilepsy Res. ticus due to ifosfamide. Ann Pharmacother. 2006;40:332–335.
1993;14:165–173. 253. Wengs WJ, Talwar D, Bernard J. Ifosfamide-induced nonconvul-
230. Seino M, Fujitani B. Zonisamide. Clinical efficacy and use in sive status epilepticus. Arch Neurol. 1993;50:1104–1105.
epilepsy. In: Levy RH, Mattson RH, Meldrum BS, et al., eds. 254.Simonian NA, Gilliam FG, Chiappa KH. Ifosfamide causes a
Antiepileptic Drugs. 5th ed. Philadelphia: Lippincott Williams & diazepam-sensitive encephalopathy. Neurology. 1993;43:2700–2702.
Wilkins; 2002:885–891. 255. Meyer Th, Ludolph AC, Münch C. Ifosfamide encephalopathy
231. Baulac M, Leppik IE. Efficacy and safety of adjunctive zon- presenting with asterixis. J Neurol Sci. 2002;1999:85–88.
isamide therapy for refractory partial seizures. Epilepsy Res. 256. Brown GL, Wilson WP. Salicylate intoxication and the CNS with
2007;75:75–83. special reference to EEG findings. Dis Nerv Syst. 1971;32:135–140.
232. Joo EY, Kim SH, Seo DW et al. Zonisamide decreases cortical 257. Münthe ThF, Knehans A. EEG-Befunde bei rezidivierender
excitability in patients with idiopathic generalized epilepsy. Clin Salizylat-Intoxication. Z EEG-EMG. 1997;28:52–54.
Neurophysiol. 2008;119:1385–1392. 258. Shiozawa Z, Sasaki H, Ozaki Y, et al. Epilepsia partialis continua fol-
233. Szaflarski JP. Effects of zonisamide on the electroencephalogram lowing metrizamide cisternography. Ann Neurol. 1981;10:400–401.
of a patient with juvenile myoclonic epilepsy. Epilepsy Behav. 259. Elian M, Fenwick P. Metrizamide and the EEG: three case reports
2004;5:1024–1026. and a review. J Neurol. 1985;232:341–345.
234. Beyreuther BK, Freitag J, Heers C, et al. Lacosamide: a review of 260. Obeid T, Yaqub B, Panayiotopoulos Ch, et al. Absence status
preclinical properties. CNS Drug Rev. 2007;13:21–42. epilepticus with computed tomographic brain changes following
235. Ben-Menachem E, Biton V, Jatuzis D, et al. Efficacy and safety of metrizamide myelography. Ann Neurol. 1988;24:582–584.
oral lacosamide as adjunctive therapy in adults with partial-onset 261. Russel D, Anke IM, Nyberg-Hansen R, et al. Complex partial sta-
seizures. Epilepsia. 2007;48:1308–1317. tus epilepticus following myelography with metrizamide. Ann
236. Young GB, Da Silva OP. Effects of morphine on the electroen- Neurol. 1980;8:325–327.
cephalograms of neonates: a prospective, observational study. 262. Vollmer ME, Weiss H, Beanland C, et al. Prolonged confusion
Clin Neurophysiol. 2000;111:1955–1960. due to absence status following metrizamide myelography. Arch
237. Kearse LA, Koski G, Husain MV, et al. Epileptiform activity Neurol. 1985;42:1005–1008.
during opioid anesthesia. EEG Clin Neurophysiol. 1993;87: 263. Ropper AH, Chiappa KH, Young RR. The effect of metrizamide
374–379. on the electroencephalogram: a prospective study in 61 patients.
238. Jakobs MH, Senior RM. Theophylline toxicity due to impaired Ann Neurol. 1979;6:222–226.
theophylline degradation. Am Rev Respir Dis. 1974;110:342–345. 264. Klein KM, Shiratori K, Knake S, et al. Status epilepticus and
239. Schwartz KA, Scott DF. Aminophylline-induced seizures. seizures induced by iopamidol myelography. Seizure.
Epilepsia. 1974;15:501–505. 2004;13:196–199.
240. Yarnell PR, Chu NS. Focal seizures and aminophylline. Neurology 265. Nelson M, Bartlett RJV, Lamb JT. Seizures after intravenous con-
(Minneapolis). 1975;25:819–822. trast media for cranial computed tomography. J Neurol
241. Yoshikawa H. First-line therapy for theophylline-associated Neurosurg Psychiatry. 1989;52:1170–1175.
seizures. Acta Neurol Scand. 2007;115:57–61. 266. Maramatton BV, Manno EM, Wijdicks EFM, et al. Gadolinium
242. Katz BE, Carver MW. Acute poisoning with isoniazid treated with encephalopathy in a patient with renal failure. Neurology.
exchange transfusions. Pediatrics. 1956;18:77. 2005;64:1276–1278.
243. Mellerio F. L’électroencéphalographie Dans Les Intoxications 267. Neufeld MY. Periodic triphasic waves in levodopa-induced
Aigues. Paris: Masson; 1964. encephalopathy. Neurology. 1992;42:444.
244. Terman D, Teitelbaum DT. Isoniazid self-poisoning. Neurology 268. Riazi K, Roshanpour M, Rafiei-Tabatabaei N, et al. The procon-
(Minneapolis). 1970;20:299–304. vulsant effect of sildafenil in mice: role of nitric oxide–cGMO
245. Weinstein L. Penicillins and cephalosporins. In: Goodman LS, pathway. Br J Pharmacol. 2006;147:935–943.
Gilman A, eds. The Pharmacological Basis of Therapeutics. New 269. Gilad R, Lampl Y, Eshel Y, et al. Drug points: tonic–clonic seizures
York: Macmillan; 1975:1130–1166. in patients taking sildafenil. Br Med J. 2002;325:869.
246. Fernández-Torre JL, Martínez-Martínez M, González-Rato J, et 270. Striano P, Zara F, MinettiC, et al. Epileptic seizures can follow
al. Cephalosporin-induced nonconvulsive status epilepticus: high doses of oral vardenafil. Br Med J. 2006;333:785.
clinical and electroencephalographic features. Epilepsia. 271. Mellerio F, Kubicki S. Encephalopathy due to poisoning. In: Remond
2005;46:1550–1552. A, ed.-in-chief. Handbook of Electroencephalography and Clinical
247. Maganti R, Jolin D, Rishi D, et al. Nonconvulsive status epilepti- Neurophysiology. Vol 15A. Amsterdam: Elsevier; 1977:108–135.
cus due to cefepime in a patient with normal renal function. 272. Lidsky TI, Schneider JS. Lead neurotoxicity in children: basic
Epilepsy Behav. 2006;8:312–314. mechanisms and clinical correlates. Brain. 2003;126:5–19.
248. Jallon P, Fankhauser L, Du Pasquier R, et al. Severe but reversible 273. Canfield RL, Henderson CR, Cory-Slechta DA, et al. Intellectual
encephalopathy associated with cefepime. Neurophysiol Clin. impairment in children with blood lead concentrations below
2000;30:383–386. 10 g per deciliter. N Engl J Med. 2003;348:1517–1526.
249. Serkova NJ, Christians U, Benet LZ. Biochemical mechanisms of 274. Stewart WF, Schwartz BS, Davatzikos C, et al. Past adult lead
cyclosporin neurotoxicity. Mol Intervent. 2004;4:97–107. exposure is linked to neurodegeneration measured by brain MRI.
250. Gleeson JG. Cyclosporin A acute encephalopathy and seizure Neurology. 2006;66:1474–1484.
syndrome in childhood: clinical features and risk of seizure 275. Fejerman N, Gimenez ER, Vallejo NE, et al. Lennox–Gastaut syn-
recurrence. J Child Neurol. 1998;13:336–344. drome and lead intoxication. Pediatrics. 1973;52:227–234.
251. Singh G, Ress JH, Sander JW. Seizures and epilepsy in oncological 276. Benignus VA, Otto DA, Müller KE, et al. Effects of age and body
practice: causes, course, mechanisms and treatment. J Neurol lead burden on CNS functions is young children. II. EEG spectra.
Neurosurg Psychiatry. 2007;78:342–349. Electroencephalogr Clin Neurophysiol. 1981;52:240–248.
922 Part VI ■ EEG: Neuropharmacology and Anesthesia

277. Mahaffey K. Methylmercury: a new look at the risk. Public Health 297. Rollins YD, Filley CM, McNutt JT, et al. Fulminant ascending
Rep. 1999;114:397–413. paralysis as a delayed sequela of diethylene glycol (Sterno) inges-
278. Myers GJ, Davidson PW, Cox C, et al. Prenatal methylmercury tion. Neurology. 2002;59:1460–1463.
exposure from ocean fish consumption in the Seychelles child 298. Zhou L, Zabad R, Lewis RA. Ethylene glycol intoxication: electro-
development study. Lancet. 2003;361:1686–1692. physiological studies suggest a polyradiculopathy. Neurology.
279. Saper RB, Phillips RS, Sehgal A, et al. Lead, mercury, an arsenic in 2002;59:1809–1810.
US- and Indian-manufactured ayurvedic medicines sold via 299. Chan AWK. Alcoholism and epilepsy. Epilepsia. 1985;26:323–333.
internet. J Am Med Assoc. 2008;300;915–923. 300. Krauss GL, Niedermeyer E. Electroencephalogram and seizures
280. Brenner RP, Snijder RD. Late EEG-findings and clinical status in chronic alcoholism. Electroencephalogr Clin Neurophysiol.
after organic mercury poisoning. Arch Neurol. 1980;37:282–284. 1991;78:97–104.
281. Pirkivi L, Tolonen U. EEG findings in chlor-alkali workers sub- 301. Davis KM, Wu JY. Role of glutaminergic and GABAergic systems
jected to low long term exposure to mercury vapour. Br J Ind in alcoholism. J Biomed Sci. 2000;8:7–19.
Med. 1989;46:370–375. 302. Sand T, Brathen G, Michler R, et al. Clinical utility of EEG in
282. Lee JW. Manganese intoxication. Arch Neurol. 2000;57:597–599. alcohol-related seizures. Acta Neurol Scand. 2002;105:18–24.
283. Ono K, Komai K, Yamada M. Myoclonic involuntary movement 303. Leone M, Tonini C, Bogliun G, et al., for the ARES (Alcohol
associated with chronic manganese poisoning. J Neurol Sci. Related Seizures) Study Group. Chronic alcohol use and first
2002;199:93–96. symptomatic epileptic seizure. J Neurol Neurosurg Psychiatry.
284. Alfrey AC, Le Gendre GR, Kachny WD. The dialysis encephalopa- 2002;73:495–499.
thy syndrome. Possible aluminium intoxication. N Engl J Med. 304. Niedermeyer E, Freund G, Krumholz A. Subacute encephalopa-
1976;294:184–188. thy with seizures in alcoholics: a clinical electroencephalographic
285. Prick JJG. Thallium poisoning. In: Vinken PJ, Bruyn GW, eds. study. Clin Electroencephalogr. 1981;12:113–129.
Handbook of Neurology. Vol 36. Amsterdam: North Holland; 305. Mani J, Sitajayalakshmi S, Borgohain R, et al. Subacute
1979:239–278. encephalopathy with seizures in alcoholism. Seizure.
286. Prockop LD, Chichkova RI. Carbon monoxide intoxication: an 2003;12:126–129.
updated review. J Neurol Sci. 2007;262:122–130. 306. Ng SKC, Hauser WA, Brust YCM, et al. Alcohol consumption and
287. Kim J, Chang K, Song IC, et al. Delayed encephalopathy of acute withdrawal in new-onset seizures. N Engl J Med. 1988;319:637–666.
carbon monoxide intoxication: diffusivity of cerebral white mat- 307. Ashton CH. Solvent abuse. Br Med J. 1990;20:135–136.
ter lesions. Am J Neuroradiol. 2003;24:1592–1597. 308. Aydin K, Sencer S, Demir T, et al. Cranial MR findings in chronic
288. Zagami AS, Lethlan AK, Mellick R. Delayed neurological deterio- toluene abuse by inhalation. Am J Neuroradiol. 2002;23:1173–1179.
ration following carbon monoxide poisoning: MRI findings. J 309. Baker EL. Chronic toxic encephalopathy caused by occupational
Neurol. 1993;240:113–116. solvent exposure. Ann Neurol. 2008;63:545–547.
289. Bokonjic N. Stagnant Anoxia and Carbon Monoxide Poisoning. 310. Gash DM, Rutland K, Hudson NL, et al. Trichloroethylene:
Amsterdam: Elsevier; 1963. parkinsonism and complex 1 mitochondrial neurotoxicity. Ann
290. Murata M, Suzuki M, Hasegawa Y, et al. Improvement of occipital Neurol. 2007;63:184–192.
alpha activity by repetitive hyperbaric oxygen therapy in patients 311. Stracciari A, Gallassi R, Ciardulli C, et al. Neuropsychological and
with carbon monoxide poisoning: a possible indicator for treat- EEG evaluation in exposure to trichloroethylene. J Neurol.
ment efficacy. J Neurol Sci. 2005;235:69–74. 1985;232:120–122.
291. Schneck SA. Methyl alcohol. In: Vinken PJ, Bruyn GW, eds. 312. Roberts DM, Aoron CK. Clinical review. Management of acute
Handbook of Neurology. Vol 36. Amsterdam: North Holland; organophosphorus pesticide poisoning. Br Med J. 2007;334:
1979:351–360. 629–634.
292. Ley CO, Gali FG. Parkinsonian syndrome after methanol intoxi- 313. Lockwood AL. Editorial comment: organophosphate pesticides
cation. Eur Neurol. 1983;22:405–409. and public policy. Curr Opin Neurol. 2002;15:725–726.
293. Jameson HD, Kane R. EEG records during an epidemic of 314. Dassanyake T, Weerasinghe V, Dangahadeniya U, et al. Long-term
methanol intoxication. Electroencephalogr Clin Neurophysiol. event-related potential changes following organophosphorus
1969;26:112 [abstract]. insecticide poisoning. Clin Neurophysiol. 2008;199:144–150.
294. Hanif M, Mobarak MR, Ronan A, et al. Fatal renal failure caused 315. Okonek S, Rieger H. EEG Veränderungen bei
by diethylene glycol in paracetamol elixir: the Bangladesh epi- Alkylphosphatvergiftungen. Z EEG-EMG. 1975;6:19–27.
demic. Br Med J. 1995;311:88–91. 316. Lockwood AL. Exposure to environmental toxins. Curr Opin
295. O Brien KL, Selanikio JD, Hecdivert Ch, et al., for the Acute Neurol. 1997;10:507–511.
Renal Failure Investigation Team. Epidemic of pediatric deaths 317. Walder B, Tramer MR, Seeck M. Seizure-like phenomena and
from acute renal failure caused by diethylene glycol poisoning. propofol. Neurology. 2002;58:1327–1332.
JAMA. 1998;279:1175–1180.
296. Lewis LD, Smith BW, Mamourian AC. Delayed sequelae after
acute overdoses or poisonings: cranial neuropathy related to
ethylene glycol ingestion. Clin Pharmacol Ther. 1997;61:692–699.
Part VII Evoked Potentials and Event-Related EEG Phenomena
CHAPTER
Event-Related Potentials: General
Aspects of Methodology
and Quantification
44
FERNANDO H. LOPES DA SILVA

E
lectroencephalography as a general method for the the ongoing activity while providing, at the same time, an indi-
investigation of human brain function includes ways of cation of response variability. It is, however, difficult to quantify
determining the reactions of the brain to a variety of the results in this way. Since the introduction of the summation
stimuli. Some of these reactions may be associated with clear- method (13) and the development of digital computer tech-
cut changes in the EEG; some others, however, consist of niques (14–16), ERPs are obtained in special-purpose digital
changes that are difficult to visualize. The research field dedi- computers and expressed as time-varying functions. In general
cated to the detection, quantification, and physiologic analysis terms, there is no fundamental argument to justify the prefer-
of those slight EEG changes that are related to particular events ence for either time or frequency ERP analysis. Both reflect the
has steadily grown as a field of great interest in the last decades. same reality. The choice of analytic domain, therefore, is deter-
These EEG changes may be treated globally under the common mined by practical considerations. Nevertheless, a certain
term event-related potentials (ERPs); a subset of the ERPs is sen- model of the generation of ERPs is always implicit in the choice
sory (visual, auditory, somatosensory) evoked potentials of a method of analysis of these potentials.
(ERPs). This chapter focuses on some general aspects of ERPs.
Detailed descriptions of specific aspects of ERPs to stimuli of
BASIC MODELS OF ERPS
different modalities are presented in separate chapters (visual in
Chapter 46, auditory in Chapter 47, and somatosensory in According to the most widely accepted model, ERPs are signals
Chapter 48), as well as general aspects of event-related generated by neural populations that are time-locked to the
(des)ynchronization in Chapter 45. Specialized aspects of ERPs stimulus; these signals would be summed to the ongoing EEG
of children and infants are presented in Chapter 49 and the use activity. According to another model, however, ERPs are
of ERPs in the operating room is covered in Chapter 38. assumed to result, at least partially, from a reorganization of the
Regan published an authoritative specialized textbook on ongoing activity. This view arose from the seminal work of
this topic in 1972 and a thorough revised form in 1989; other Sayers et al. (17) who analyzed segments of EEG recorded
early general sources include Storm van Leeuwen et al. (1), John before and immediately after the presentation of auditory stim-
and Thatcher (2), Callaway et al. (3), Barber (4), Chiappa (5), uli. They carried out a Fourier analysis computing the real and
and Misulis and Fakhoury (6). Reviews of the applications of imaginary coefficients of the signal’s frequency components. In
computer methods to ERP detection and analysis are those of this way they constructed amplitude and phase spectra, and
Barrett (7), Gevins and Cutillo (8), and Regan (9). Insightful found that the brainstem-evoked potentials (BAEPs) to audi-
accounts of steady-state (10) and transient evoked potentials tory stimuli could be distinguished much better using the dis-
(11) are presented in the book edited by Nunez Neocortical tributions of phase spectral values, rather than the distributions
Dynamics and Human EEG Rhythms (10). of amplitude values. In a later study McClelland and Sayers (18)
showed further that the magnitude of the differences between
EVOKED-RELATED POTENTIAL amplitude spectra of pre- and post-stimulus epochs decreased
ANALYSIS: GENERAL ASPECTS progressively as stimulus intensity was reduced. Examining sep-
arately amplitude and phase spectra of the response ensemble,
ERPs are usually defined in the time domain as the brain elec- these authors showed that the phase standard deviation for
trical activity that is triggered by the occurrence of particular responses at auditory threshold was generally significantly
events or stimuli. A basic problem of analysis is how to detect smaller than for the corresponding harmonics obtained from
ERP activity within the often much larger ongoing EEG or subthreshold responses. In contrast, the amplitude differences
background activity (i.e., the activity not related to the stimu- between populations of threshold and subthreshold responses
lus). Dawson (12), a pioneer in the field, attempted first to solve were generally non-significant. These findings led the authors
this problem by the photographic superposition on the screen to conclude that the use of BAEPs for auditory threshold deter-
of an oscilloscope of a number of time-locked responses. This mination should preferentially be based on ensemble phase
method had the property of enhancing response in contrast to measures rather than amplitude measures.

923
924 Part VII ■ Evoked Potentials and Event-Related EEG Phenomena

These experimental results indicate that the brain response same idea was formulated by Shah et al. (25) who noted that
to a stimulus may consist, at least partially, of a decrease of EEG ongoing activity and ERPs are generated by overlapping
phase variance of certain frequency components, that is, an neuronal elements. According to this model, ERPs may engage
enhanced alignment of the phase components of ongoing EEG a group of neurons, by way of two basic mechanisms: either by
or MEG signals, or phase resetting. enhancing the synchrony of increases, or decreases, of neu-
More recently Makeig et al. (19) demonstrated that ERPs ronal firing rates and/or by synchronized membrane tran-
could be generated by stimulus-induced phase resetting of sients or oscillations. These two mechanisms are not mutually
ongoing EEG components. Mazaheri and Picton (20) showed, exclusive. It follows that the former mechanism will lead to
by subtracting the spectra of the average ERP from the average changes of amplitude, but this will also be the case for the lat-
spectra of single trials, that ERPs can involve phase resetting of ter, since the activity of those neurons that work synchro-
the ongoing EEG rhythms. Nonetheless, Mäkinen et al. (21) nously in phase corresponds to larger amplitude at the level of
argued that a high degree of independence exists between the population than that of neurons that display a random, or
ongoing brain activity and auditory ERPs, and suggested to use weak, alignment of phases. This reinforces the conclusions
amplitude variance to distinguish whether or not phase reset- that amplitude variance is not sufficient to determine whether
ting is present in an ERP. An unambiguous assessment of phase phase resetting occurs in event-related responses. In any case,
resetting, however, implies a measure of variance of phase val- the assessment of phase resetting needs the direct estimation
ues and cannot be based just on amplitude variance, as appro- of phase variance of frequency components. This means that,
priately pointed out by Klimesch et al. (22). in the most general case, the analysis of event-related
This issue was addressed by Mazaheri and Jensen (23) in an EEG/MEG changes of activity should take into consideration
interesting way; they introduced a measure—the phase preser- not only spectral amplitude, but also phase. In this respect, it
vation index—to compare quantitatively the phase of alpha is also interesting to note that recently more attention is being
oscillations before and after a visual stimulus. The main finding put on the use of the imaginary part of coherency, which in
was that the alpha oscillations preserve their phase relationship fact constitutes the phase spectrum, to analyze interactions
with respect to the phase before the stimulus, and their conclu- between different EEG signals (26).
sion was that different neuronal populations appear to generate Notwithstanding the importance of phase in the generation
the ongoing oscillations and the ERPs, but this does not of ERPs in general, the wide use of time averaging of ERP
imply that the neuronal populations are macroscopically amplitude waveforms in practical ERP work is so overwhelm-
distinguishable. ing that this form of analysis is treated first in the following sec-
An underlying problem, however, is the fact that in all these tion. After that we consider analyses in the frequency domain
cases of event-related responses the signal-to-noise ratio plays a and lastly analyses according to integrated time–frequency
fundamental role. The original findings of Sayers et al. (17) that approaches.
the phase spectrum was more powerful than the amplitude
spectrum in distinguishing auditory ERPs were obtained at very TIME AVERAGING OF ERPs
low stimulus intensities, that is, at the threshold for auditory
discrimination. This is not the case in many other experiments. According to the classic view, ERP analysis is based on two basic
In a recent overview of this issue we (24) proposed that it is assumptions: (i) the electrical response evoked from the brain
likely that the relative amount of phase preservation from the is invariably delayed relative to the stimulus and (ii) the ongo-
pre- to the post-stimulus epochs will depend on several factors; ing activity is a stationary noise, the samples of which may or
at least four appear to be particularly relevant: may not be correlated. In this way, ERP can be considered a sig-
nal (s(k)) corrupted by additive noise (n(k)), the ongoing activ-
• the baseline level of the ongoing oscillations,
ity. (Assume that the signals are sampled, with k as the discrete
• the frequency band of the oscillations,
time variable.) ERP detection becomes, in this way, a question
• the stimulus modality and its strength, and
of improving signal-to-noise ratio. In the more general and
• the specific cortical area, since EEG rhythmic activities
simplified case one assumes a simple additive model (stochastic
are not equally distributed over the cortex.
variables are indicated by underlined symbols). The recorded
Moreover, it is important to note that a solution to this ques- stochastic signal x(k) is given as a sum of two terms:
tion may not be resolved using only scalp EEG/MEG records, as
also stated by Mazaheri and Jensen (23) who propose that x(k) s(k) n(k) (44.1)
intracranial recordings will be necessary, combining single cell The expected value of n(k) is zero. The average of x(k) over N
and local field potential recordings with EEG signals. realizations is then defined as:
We may state from a theoretical point of view that it is not
probable that two independent neuronal populations, one 1 N
x(k) a xi (k) (44.2)
generating exclusively ongoing activity and another one Ni 1
responsible for the evoked response, would exist side by side The expected value of the average is given as:
as distinct entities in a given brain area. It is more likely that
the same neuronal elements are involved in the generation of 1 N
E[x(k)] Ec a xii (k) d s(k) (44.3)
both types of signals but working in different modes. This Ni 1
Chapter 44 ■ Event-Related Potentials: General Aspects of Methodology and Quantification 925

[since E(n(k))] = 0. The variance of x(k) is as follows: Nevertheless, the method can be very useful for data reduction
in EP research where one wants to go further than the mean
1 N 2
1 value of the EP.
var[x(k)] Ec a a n i (k) b d var[n(k)] (44.4)
Ni 1 N In principal components analysis, the first component is the
one accounting for the greatest variance; as many components
since s(k) is assumed to be invariant. Therefore, the signal-to-
as one wishes can be determined in order to account for a cer-
noise ratio in terms of amplitude improves proportionally to
tain percentage of the total variance. In EP analysis, it is usually
1 n . In more general terms, however, it should be assumed that
not necessary to go further than i = 4 or 5. In this way, a parsi-
the signal s(k) is a stochastic signal, so that expression 44.1
monious description of EP can be achieved. Principal compo-
should be reformulated as follows:
nents analysis, however, is no more than a transformation of the
x(k) s(k) n(k) (44.5) original data. In this form of analysis, no provision can be made
for variance components attributable only to the unreliability
The importance of this apparently small difference lies in the
of the observations (32). This and other shortcomings of prin-
fact that the model given by expression 44.5 implies that the EP
cipal components analysis can be solved by way of the mathe-
is not fully described by the mean value of x(k), but also by the
matical model of factor analysis.
higher statistical moments, as, for instance, the variance. In
In factor analysis, each observation xi is represented in terms
most practical cases, the latter model (expression 44.5) gives a
of a linear function of common factor variables and of a signal
better account of reality. As pointed out by John et al. (27), the
latent-specific variable that can be represented as follows (pro-
former model (expression 44.1) is valid only for anesthetized
vided that the means of the observations are zero):
preparations and possibly for the short-latency components of
EP that mainly reflect sensory processes; this is, however, not x1 l ijyj p l im ym ej
the case for long-latency components. p (44.11)
xn l njyj l nm ym en
For a comprehensive study of EP, multivariate analysis meth-
ods may be used (28–31). In this case, one considers an EP to be where yj is the jth common factor variable, ij is called the load-
a multidimensional stochastic variable, that is, a vector x where ing of the ith observation on the jth common factor, and ei is
the elements are the values in subsequent time points: the ith specific factor variable. The covariances of the observa-
x (x(1) p x(NS) ) T (44.6) tions xi are reflected only on the specific factors. Factor analysis
is based on a specific statistical model in which the number of
Thus, the expression 44.5 can be rewritten as a vectorial sum: common factors is specified beforehand; an example of factor
x s n (44.7) analysis in EP research is shown in Figure 44.1. These methods
of analysis can be of great usefulness in order to provide a small
assuming that E(n ) 0 as before. Therefore: set of features that can be employed for the classification
and clustering of EP as discussed below (28,34–37). As an
E(x) E( s ) (44.8)

The second-order moment of x is given by the covariance


matrix C; considering any two samples represented by k and 1,
one may write:
Covk1 E[ (x(k) E(x(k) ) ) (x(1) E(x(1) ) )] (44.9)
There are multivariate statistical methods that enable the struc-
ture of such signals as EPs to be represented as functions of a
small number of factors. A technique to achieve this form of
representation is the principal components method. The essence
of the principal components method is to account for the EP as
a linear combination of a small number of basis functions. It
can be proved (e.g., Ref. 32) that, if one chooses as basic func-
tions the eigenfunctions c i, of the covariance matrix, the fol-
lowing expression gives the best p-dimensional approximation
(in a least-squares sense) to the EP (P NS):
p
x E(x) a l 1c i (44.10) Figure 44.1 Average evoked potential (ERP) of one subject (parietal
i 1 leads; N = 512 epochs; epoch length, 765 msec) and the corresponding
In the expression, the coefficients l are uncorrelated and the seven factor loading plots. The reality of the first two factors in the
eigenfunctions c 1 are orthogonal. It should be noted, however, waveform of this example is questionable, because the factors also
that this form of description is only a statistical way of describ- showed loadings in the prestimulus period. (Adapted from Rebert CS.
ing EP; one cannot assume that the eigenfunctions necessarily Neuroelectric measures of lateral specialization in relation to perform-
correspond to independent physiologic generators (33). ance. Electroencephalogr Clin Neurophysiol. 1978;34:231–238.)
926 Part VII ■ Evoked Potentials and Event-Related EEG Phenomena

illustration of the usefulness of principal components and dis-


criminant analysis, the study of Donchin and Cohen (38) found
that the discriminant between EP recorded to task-relevant and
task-irrelevant stimuli was based essentially on time points at
300 msec.
Another form of data reduction of average EP involves
describing EP as a sum of a set of analytic functions, such as
damped sinusoids as thoroughly investigated by Freeman
(39,40). Later, this chapter discusses the possibility of describ-
ing EP as a sum of harmonically related sine waves (i.e., Fourier
components).

SPECIAL PROBLEMS IN ERP


TIME AVERAGING
ERPs in Strong Rhythmic Background EEG
One problem is whether the statistical properties of the back-
ground activity may have influence on the degree of improve-
ment regarding signal-to-noise ratio realized by the averaging
procedure. In detecting ERPs by classic time averaging, the
main objective, of course, is to increase the signal-to-noise ratio
so that the EEG background activity will be attenuated.
Equation 44.4 illustrates that the ratio between the variance of
the averaged signal and the variance of the noise is 1/N. This Figure 44.2 Average ERPs recorded between vertex and mastoid; stim-
holds for the case where the noise samples are uncorrelated. ulus is a sinusoidal tone of 1000 Hz, 40 dB above 2104 bar, 600 msec.
Often, however, this is not the case, as, for instance, in the pres- A: The results of simple averaging are shown for a number of averag-
ence of a strong rhythmic background such as an alpha rhythm. ing steps. B: The results obtained using an adaptive filter (based on a
In such a case, subsequent samples of n(k) are not independent. maximum likelihood detector adapted to broadband colored noise) fol-
The degree of dependence of the samples of n(k) is given by the lowed by synchronization and averaging are shown. Note the improve-
autocorrelation function of the background activity (see for a ment in ERP discrimination. The plots are in relative vertical scale; t st
definition Chapter 54). is the stimulus onset. (Courtesy of G.H. Steeger.)
Assuming that 2 is the var[n(k)], the ERP variance
[var(x(k))] is not equal to [1/N(var(n(k)))], as in Equation 44.4,
but equal to [ 2/N(var(n(k)))] multiplied by a factor that (49). According to one of these methods (44), a matched filter
depends on the properties of the autocorrelation function of the is created that is adapted optimally to the template (the aver-
background activity (41). It is therefore important to emphasize age ERP) as well as to the actual ongoing activity, which is
that rhythmicity of the background can influence the signal-to- estimated by its autocorrelation function (Woody’s adaptive
noise ratio. McGillem and Aunon (42), Steeger (43), and Steeger filter was only adapted optimally to the template and to white
and Reinhardt (44) described methods that can provide better noise). Each single-trial EEG segment is then passed through
estimates of average ERP because they take into account the the matched filter and the maximum output signal is found;
statistical properties of the ongoing EEG activity. in this way an optimal average ERP can be obtained after cor-
A related problem is whether the stimulation should be peri- rection for latency deviations. A method related to those
odic or aperiodic; aperiodic stimulation can lead to reduced described above but also involving a classification process was
ERP variance as shown theoretically by Ten Hoopen (45) and proposed by Pfurtscheller and Cooper (50) under the term
experimentally by Arnal and Gerin (46). More specifically, selective averaging; according to this technique, single-trial
Ruchkin (47) has demonstrated that the variance of the averag- ERPs are cross-correlated with different templates. The max-
ing estimate is closer to zero when the interstimuli intervals are ima of the cross-correlation functions are used to estimate the
exponentially distributed (Fig. 44.2). amplitude and latency of particular components. Thereafter,
those trials that fall within certain amplitude and/or latency
Single-Trial ERPs and Classification limits are averaged. In this way, different types of selected
In the last decades there have been several studies with the aim averages can be obtained that may correspond to different
of identifying single-trial ERPs and/or to discriminate subsystems and/or physiologic conditions. In cases where all
subgroups of ERPs among a series of trials. Initially most of trials are averaged together, there is a considerable informa-
the approaches applied were based on cross-correlation of tion loss. The problem of measuring single-trial ERPs has
EEG signals, including the ERP, and some template a priori been extensively investigated by Coppola et al. (51). In this
determined (42,48), and on maximum likelihood detectors study, equations have been proposed to estimate empirically
Chapter 44 ■ Event-Related Potentials: General Aspects of Methodology and Quantification 927

S/N that may be useful in ERP clinical or psychophysiological Therefore, the averaging reduces the power spectrum of the
studies (for details see Ref. 51). noise term in the unaveraged signal Pnn (fi) by a factor 1/N.
The analysis of single-trial ERP is also important in deter- Moreover, assume that the spectrum of each realization has
mining whether a certain single-trial ERP belongs to a specific been made and that an average of all these spectra has been
ERP class. This problem of classification can be solved using computed: Pxx(fi ) ; thus, it can be written as follows:
multivariate statistical methods (34). In general terms, the
question involves: (i) describing ERP by a set of features (in the Pxx(fi ) Pss(fi ) Pnn (fi ) (44.13)
simplest case, the values of the sample points) and establishing
a feature profile, (ii) making a learning set where ERPs corre- Combining Equations 44.12 and 44.13 one can derive the fol-
sponding to a priori defined classes are pooled, and (iii) finding lowing expression:
a discriminant function (i.e., a decision rule) that must parti-
N 1
tion the space occupied by all objects (the ERP) so that unique Pss(fi ) Pxx(fi ) Pxx(fi ) (44.14)
N 1 N 1
patterns corresponding to different classes can be identified.
(iv) Finally, the class to which any new object (i.e., a single-trial Wiener (54) has shown that for a signal x(k) = s(k) n(k) with
ERP) belongs must be determined; this is accomplished by power spectra Pxx(fi), Pss(fi), and Pnn (fi), a linear filter can be
computing a discriminant score for each object. Thus, each sin- constructed that provides an optimal estimator of the signal
gle-trial ERP is classified in the subspace within which the dis- s(k); this linear filter has a transfer function h(k), or in the fre-
criminant score falls. The ERP features used in the procedure quency domain H(fi), which is given by the following expres-
can be simply the corresponding sample values or a reduced sion (55):
amount of data, such as can be obtained using factor or princi- Pss(fi )
pal component analysis. H(fi ) (44.15)
Pss(fi ) Pnn (fi )
The modern tendency is to use spatiotemporal filtering
methods for single-trial ERP component estimation, such Doyle (56) has shown, however, that one should use an expres-
as methods that make use of the spatial diversity of multi- sion other than 44.15 for defining the filter H(fi) for use on the
channel recordings and thus can extract signals in poor average ERP, based on N realizations:
signal-to-noise ratio conditions (52). Another example of Pss(fi )
such an integrated methodology has been proposed H(fi ) (44.16)
Pss(fi ) (1>N)Pnn (fi )
by Poolman et al. (53); this method consists of multiple pro-
cessing modules that can be applied as a whole, or in part, In this case, notice is taken of the situation after averaging,
including noise mitigation based on directed components because the averaging procedure reduces the power of the noise
analysis (DCA), source–space transformation utilizing a term by a factor 1/N as shown in Equation 44.12. Expression
finite difference model (FDM) of the human head, and 44.16 has been critically reviewed by Albrecht et al. (57), who
discrimination modules that interface with performance pointed out that in the denominator the term (1/N)Pnn(fi)
evaluation modules to generate classification statistics of should be replaced by a more general term (hN(fi) 2)Pnn(fi),
ERPs. This method performed very well in a task consisting where the function hN(fi) 2 reflects the attenuation of the noise
of discriminating targets from non-targets in a rapid serial due to averaging; this function depends not only on N, but also
visual presentation (RSVP). on the length of the realization and on the time interval
Other advanced methods in time–frequency domain are between the end of one realization and the following stimulus
currently used for single-trial ERP estimation combined with (for details see Ref. 57). Using Equation 44.16, the optimal filter
the analysis of the changes in ongoing EEG activities as dis- according to Doyle becomes:
cussed below.
[N>(N 1)] Pxx(fi ) [1>(N 1)] Pxx(fi )
H¿(fi ) (44.17)
Pxx(fi )
Improvement of Time-Averaging Efficiency
It has been suggested that a posteriori or Wiener (54) filtering, The application of filter H(fi) to the averaged signal x(k) can be
introduced by Walter (55) in the EEG field, can improve aver- written formally as follows:
aging efficiency. The following description uses the concepts of
x(k)*F 1 [H¿(fi )] (44.18)
power spectrum and related functions as explained in more
detail in Chapter 54. Assume that the additive model of where H(fi) is the impulse response of the Wiener filter accord-
Equation 44.5, in which the ERP, x(k), is considered the sum of ing to Doyle, the asterisk represents the convolution operation,
a signal s(k) and a noise term n(k), is valid, that s(k) and n(k) and the symbol F–1 indicates the inverse Fourier transforma-
are uncorrelated, and that both have means of zero. The power tion. However, in a theoretical study in which Wiener filtering
spectrum of the average signal x(k) obtained by averaging N was compared to ERP averaging, Albrecht et al. (57) demon-
realizations is given by the following expression: strated that the former yields a better estimate of s(k) than the
latter, although the difference between both decreases with an
1 increased number of realizations. (It can be seen from Equation
Pxx(fi ) Pss(fi ) Pnn (fi ) (44.12)
N 44.16 that as N becomes large, H(fi) tends to unity.) Moreover,
928 Part VII ■ Evoked Potentials and Event-Related EEG Phenomena

they also pointed out, in both cases, the importance of using ones) are of great importance. An interesting addition to the
interstimuli intervals leading to the smallest error of the aver- analytic tools that can be used for assessing the variability of
age (i.e., the intervals should be exponentially distributed), as ERPs was recently proposed by Hu et al. (63) who developed
also shown by Ruchkin (47). Therefore, it appears that Wiener an unsupervised correlation-based clustering method to
filtering would be of practical interest in ERP analysis only in quantify trial-to-trial variability of auditory evoked poten-
those cases where a small number of realizations are available. tials (AEPs).
In fact, Strackee and Cerri (58) have shown in a theoretical
study that a posteriori (Wiener) filtering does not improve ERP ANALYSIS IN THE FREQUENCY DOMAIN
averages, although this technique does improve the signal-to-
noise ratio for individual realizations. Albrecht et al. (57) have In the introductory remarks we discussed the benefit of an
performed a number of simulations to study the influence of analysis in the frequency domain of ERPs. This section consi-
interstimuli intervals and other factors on averaged or Wiener- ders, in particular, (i) those ERPs that are difficult to detect in
filtered ERP; when averaging only five realizations, the mean the time domain because they occur without a fixed phase or
square error of the estimate was, for the Wiener filter, about time relation to the trigger and (ii) ERPs caused by continuous
76% that of the straightforward average ERP obtained using stimuli that are preferably analyzed in the frequency domain.
random interstimuli intervals. If 20 realizations were averaged ERPs consisting of changes in spectral intensity, for example, in
using randomly distributed stimuli, the error for the Wiener fil- the alpha frequency band in relation to voluntary hand move-
ter was about 86% of that of the average ERP. The effectiveness ments (64,65) exemplify the first category. This issue is
of a posteriori (Wiener) filtering in cases using few (5) stimuli, described in detail in Chapter 45 and thus we just mention here
but not larger sets (20), has also been demonstrated by Hartwell a few general aspects. This type of ERP can best be shown by
and Erwin (59). De Weerd et al. (60,61) have described an adap- decomposing the EEG in frequency bands and averaging the
tive time-varying filter capable of optimizing the estimation of power within different bands in relation to the trigger. It is thus
both low-frequency components of relatively long duration and necessary to use statistical methods of ERP evaluation to test
higher frequency components of short duration. whether or not the changes occurred by chance.
Steady-state ERPs are elicited by continuous stimuli as, for
Variability of ERPs example, sine wave–modulated light or amplitude-modulated
It is highly recommended that the averaging procedure tone. These ERPs may be described by measuring their ampli-
should include some form of estimate of ERP variability and tudes and phases as functions of the frequency of the stimulus
the ongoing activity. A very simple solution is to compute as performed in Amsterdam’s school of van der Tweel (66)
partial averages of ERPs (e.g., five sets of N realizations each) and collaborators (41,67) and by Regan (9,68,69). If the phys-
and the corresponding means and standard deviations or, at iologic systems underlying the generation of the ERP were lin-
least, to compare graphically the different realizations. This ear, the information from a study of the system by way of
solution has the disadvantage of requiring long stimulation transient or continuous stimuli would be the same. However,
sessions. Another useful procedure, which may be combined considering that most of these systems have nonlinear proper-
with the former, is to perform a similar computation of par- ties, both types of stimuli can provide different information.
tial averages of EEG segments, but without the application of To construct an amplitude and phase plot of a steady-state
the stimulus while averaging the same number of realiza- visual ERP, for example, it is necessary to stimulate succes-
tions. In this way one can estimate the mean and standard sively with different frequencies. An alternative is to stimulate
deviation of the ongoing activity in the absence of the stimu- with a light of a given mean luminosity, the intensity of which
lus. In any case, it is always best to include in the signal to be is modulated by Gaussian noise. In this way, the stimulus con-
averaged a period of time prior to the delivery of the stimu- tains all frequencies at the same time. The ERP is thus
lus in order to get an estimate of the mean and variability of obtained by calculating the cross-correlation function
the ongoing activity. An interesting method that can be com- (Equation 56.10 in Chapter 56) or the cross-power spectrum
bined with the normal averaging procedure in order to esti- (Equation 56.11) between the stimulus and the corresponding
mate the magnitude and structure of ongoing activity is the EEG (67,70,71); it can be shown that the result of the cross-
plus–minus ( ) average proposed by Schimmel (62). The correlation in such a case is equivalent to the impulse
( ) average is computed by alternate addition and subtrac- response of the linear components of the system (Fig. 44.3).
tion of each realization and division by N (even number); the The system, however, may contain nonlinear elements; in such
deviation of each ERP about the average value and the ( ) a case, its characteristics may be analyzed using noise-
average has the same statistical structure. The latter can be modulated light by computing higher order cross-spectra or
implemented more easily by either manually or automatically cross-correlation functions that can be defined similarly to
setting the averaging computer to “add” or “subtract” mode the bispectrum (see Equation 56.16). This question, however,
or by alternating input polarity. In many instances, the main is too specialized to be considered in detail here (see review in
limitation encountered in practice is the great difficulty of Refs. 9, 41, and 71). Steady-state stimuli particularly of the
recording a sufficiently large number of ERPs under equiva- visual modality (SSVEP) have been applied extensively in a
lent conditions. Therefore, the methods of improving signal- series of investigations by the groups of Nunez and Silberstein
to-noise ratio using a small number of trials (or even single (10,72–74) in both neurocognitive and clinical investigations
Chapter 44 ■ Event-Related Potentials: General Aspects of Methodology and Quantification 929

nervous system, but cortical sources appear to contribute more


than brainstem neuronal sources to responses at lower
modulation frequencies. Several studies showed that these
steady-state ERPs can be used for an objective evaluation of
auditory thresholds, and also to analyze suprathreshold hear-
ing. These stimuli were shown to be useful for the study of not
only specific auditory functions, but also attentional processes
(79) and other neurocognitive processes (80,81).

TIME/ FREQUENCY ANALYSIS:


NEW APPROACHES
The findings that have been obtained in the course of the last
decades, regarding the relationships between ongoing EEG sig-
nals and ERPs, as discussed above, have led to the development
of new tools to analyze these responses. The main purpose is to
Figure 44.3 A cross-correlation function between the noise-modulated integrate time- and frequency-domain analyses. The basic con-
light stimulus field and the EEG. Note that in the cross-correlation cept is that ERPs “do not emerge out a flat EEG baseline” (19),
responses several components can be distinguished; the values of the but interfere in a more or less complex way with the ongoing
cross-correlation responses at negative times give a measure for the EEG activity. In this respect the neuronal sources contributing
variance of the response owing to the finite time length (4 minutes) of to the ERP and to the ongoing EEG may share the same neu-
the EEG record used for analysis. The responses were derived from two ronal networks, although these may work in different modes.
electrodes placed on the midline (occipital region) in relation to a refer- Among other observations the finding of Vijn et al. (82) who
ence placed on the earlobe. Below the Fourier transformation are the showed that visual stimuli reduce the ongoing EEG amplitude
cross-correlation responses (i.e., the cross-power and phase spectra are supports the notion that an ERP is not simply a response added
shown along with the normalized cross-power spectrum or coherence to the background EEG. This notion is also in line with the
function). From the slopes of the phase spectra, estimates of the time experimental results of McClelland and Sayers (18), already
delays of the responses can be obtained. It can be seen that up to about mentioned above, indicating that the brain response to a stim-
20 Hz the phase lag increases more rapidly with frequency (i.e., the ulus may consist, at least partially, of a decrease of phase vari-
time delays are longer) than for frequencies above 20 Hz. In the coher- ance of certain frequency components, that is, an enhanced
ence spectra there are two maxima; one is in the range 14 to 20 Hz and alignment of some phase components of the ongoing
the other is in the range of 30 to 45 Hz. The former corresponds to the EEG/MEG activity. This phenomenon is commonly called
response components in the medium frequency range that have laten- “phase resetting” of ongoing EEG frequency components.
cies of 100 to 120 msec; the latter corresponds to the early components According to this way of reasoning, ERPs can be considered to
of higher frequency content with latency of about 30 msec. (Adapted result from a process incorporating phase resetting of specific
from Spekreijse H, Estevez O, Reits D. Visual evoked potentials and the frequency components. In this manner, the notion of signal-to-
physiological analysis of visual processes in man. In: Desmedt JE, ed. noise ratio in terms of power, or amplitude, does not provide all
Visual Evoked Potentials in Man. Oxford: Clarendon Press; 1976:3–15.) the relevant information. Therefore, a method of detecting
ERPs based on phase spectra evaluation may be more fruitful
than simple time averaging. This issue was more recently inves-
of brain function, namely of memory and aging (75). In these tigated by Makeig et al. (19,83), reinforcing the concept that
studies SSVEPs are used as a way to characterize preferred fre- phase resetting of neuronal oscillations, as originally hypothe-
quencies of neocortical dynamic processes, especially because sized by Sayers et al. (17) and also by Basar (84), can contribute
of the excellent signal-to-noise ratios that can be obtained to the generation of ERPs.
using this form of stimulation, rather than with the aim of A practical approach uses independent component analysis
investigating visual processes as such. (ICA) to decompose the EEG activity into the sum of distinct
Besides these studies where the visual modality was stimu- temporally independent component processes (see also
lated under steady-state conditions, also steady-state ampli- Chapter 54) (85–87). In contrast to principal component analy-
tude- or frequency-modulated sound stimuli were used to sis, ICA does not require the components to be orthogonal. In
investigate functional properties of the auditory system in an summary this novel approach consists of several steps. First, the
important series of studies (76,77). The corresponding ERPs signal is decomposed into a number of independent compo-
can be analyzed using frequency analysis. As described by nents (ICA); second, the spectral power of the components
Picton et al. (78) the auditory steady-state ERP amplitude (ERSP), and, third, the inter-trial phase coherence (ITC) are
increases as a function of modulation depth and/or sound computed. Applying ITC the phase-locked frequency compo-
intensity, and the phase delay of the response increases as the nents of the ERPs can be identified. It should be noted that ITC
intensity or the carrier frequency decreases. Auditory steady- is similar to the “phase-averaging” measure proposed by Tallon-
state responses are generated at various levels of the auditory Baudry et al. (88) to quantify phase-locking of EEG oscillatory
930 Part VII ■ Evoked Potentials and Event-Related EEG Phenomena

Figure 44.4 Maximally independent EEG components (ICA) compatible with a single cortical source.
Scalp maps, event-related spectra perturbation (ERSP) and inter-trial phase coherence (ITC) images,
and ERPs for two typical independent EEG processes during a task consisting of visual letter encoding
(green in images indicates p. 0.001). Below are the best fitting equivalent current dipole (ECD) models
(“r.v.” is the residual scalp map variance across the 69 scalp electrodes). A: Frontal midline theta com-
ponent with radial ECD. Spectral power increases shown in the ERSP accompany weak phase locking
on ITC and slow ERP effects (mV as at Fz). B: Mu-rhythm component with tangential ECD in right
somatomotor cortex. Alpha- and beta-band (ERSP) power changes, accompanied by partial phase lock-
ing (ITC) contributing to the component ERP. (Adapted from Makeig S, Debener S, Onton J, et al.
Mining event-related brain dynamics. Trends Cogn Sci. 2004;8(5):204–210.) (See color insert)

bursts with respect to a certain stimulus. The latter method and time width, but varying amplitude in a study of habitua-
used a decomposition of the EEG signals using a family of tion to auditory stimuli.
wavelets that can provide a time–frequency representation of
the energy of the signal (TF energy) (Fig. 44.4). ERP TOPOLOGY
One aspect that merits being underlined is that these
novel tools may be used in combination with each other to In separate chapters the topology of special ERP types is dis-
solve specific problems. In this context the study of Jung et cussed. This section presents only some of the general aspects
al. (89) demonstrated how ICA applied to single-trial multi- concerning the topologic analysis of ERP. Different forms of
channel EEG data from ERP experiments can be used com- ERP involve activity within more or less well-circumscribed
bined with spatial filters to identify components arising areas of the cortical surface and even subcortical areas, as in the
from distinct or overlapping brain or extra-brain sources. case of the far-field ERP. Therefore, at scalp level, the ensuing
These authors created a new visualization tool, the “ERP electrical activity can be recorded over a relatively large region
image,” in order to display single-trial variations in the due to the presence of volume conduction. In this sense, ERP
amplitudes and latencies of ERPs. This approach can be used recording using two electrodes gives a very limited image of the
to discriminate ERPs associated with specific behavioral or real underlying neural activity. It is in some way an equivalent
physiologic variables. of an ERP in the time domain based on the amplitude value at
Time–frequency analysis of ERPs can be performed using two times samples only. Nevertheless, valuable ERP research
different kinds of basic functions. One kind of interesting func- can be carried out using only one bipolar deviation if it is
tions in this respect is the time–frequency dictionary of Gabor placed on the appropriate brain region. To estimate the localiza-
functions, that is, sine functions modulated by Gaussians as tion of the neural systems responsible for ERP generation, how-
proposed by Sieluzycki et al. (90), who applied a multivariate ever, it is essential to sample the scalp surface appropriately. As
version of the matching pursuit algorithm (MMP) to perform discussed in Chapter 54, sampling of the EEG in space must
an iterative search for auditory MEG ERP waveforms, common take into consideration spatial frequencies present in the ERP.
to all trials with the same time of occurrence, frequency, phase, To determine these frequencies, it is necessary to have a priori
Chapter 44 ■ Event-Related Potentials: General Aspects of Methodology and Quantification 931

knowledge of the spatial characteristics of the ERP in question. studies present the authors’ own methods implemented in
This can be achieved by multichannel recordings with small their “home” environment, which does not allow a comparative
interelectrode distances. In this way, Vaughan (91), Vaughan et and objective evaluation of the performance of different
al. (92), and Vaughan and Ritter (93) mapped the scalp for methodologies applied to the same data. A few studies have
several ERP types. In these studies, derivations against a com- included comparative analysis of different methods, such as the
mon reference were used; the presence of significant activity in demonstration by Iyer and Zouridakis (113) that the applica-
the reference (chin or nose) and artifacts (muscular and eye tion of an iterative ICA procedure to the analysis of single-trial
movements) was carefully assessed. It should be noted that, in auditory N100 component can yield significantly improved
this type of studies, the pitfalls of simple averaging must be results compared to plain averaging and wavelet denoising, and
taken into consideration. In other words, the fact that the EEG that of Wang et al. (114) who demonstrated that wavelet-based
background activity may differ substantially with scalp location approaches yield better results than other methods including
should be considered when processing ERPs recorded at differ- the Wiener filter, least mean square (LMS), and recursive least
ent sites; as a rule, the number of realizations for an identical squares (RLS), especially based on translation-invariant
S/N improvement should not be the same at all recording sites. wavelets. In this field an important step forward was the intro-
Vaughan (91) was able to make isopotential maps for different duction of open source toolboxes that can be downloaded from
time samples of several forms of ERP and to relate the spatial an Internet site. One of these is the “EEGLAB” toolbox for the
distributions with results of a computer model. analysis of single-trial EEG dynamics and ERPs developed by
The early studies of Rémond and collaborators (94,95) and Delorme and Makeig (115) based on a MATLAB environment,
of Lehmann (96,97) also provide estimates of spatial gradients and the “ERPWAVELAB” toolbox developed by Morup et al.
characteristic of some ERP types. The general problem of spa- (2007) for multichannel time–frequency analysis of event-
tial properties of EEG signals is discussed in more detail in related activity of EEG and MEG data and that is an extension
Chapters 54 and 55. In addition to the early studies mentioned of “EEGLAB,” which is required for its performance. “EEGLAB”
above, the new methodologic approaches have contributed to a development is now centered at the Swartz Center for
much finer analysis of processes of sensory information in dif- Computational Neuroscience of the Institute for Neural
ferent modalities, namely in auditory processing (98–100), in Computation at the University of California San Diego (UCSD)
the analysis of the visual parallel pathways that lead to the stri- in collaboration with the CNRS CERCO laboratory in France.
ate (V1) and prestriate cortex in the human brain (101), in the In summary there is a need for critical and open discussions
functional localization of face-evoked MEG and EEG responses among users with experience in the field of the application of
using each modality separately, and by their “fusion” under a these methodologies presented here, and for comprehensive
common generative model (102) and of the somatosensory comparative studies of different methodologies, that may lead
system (103,104) including pain-related fields (105,106). to useful advances in the practical application of ERPs in neu-
Methodologic aspects of MEG and EEG analysis of event- rophysiologic investigations. Furthermore, the combination of
related events were reviewed by Kakigi et al. (107) and by more sophisticated models of the process of generation of ERPs
Michel et al. (108). The relevance of spatial analysis of ERPs with advanced analytic methods is likely to lead to major
based on high-density EEG, or MEG, recordings must be insights in our understanding of brain functional dynamics.
emphasized (109). Significant advances were obtained regard-
ing the integration of EEG and MEG evoked potentials or fields REFERENCES
using dynamic causal modeling (110–112). This modeling
approach combined with advanced time–frequency analysis is 1. Storm van Leeuwen W, Lopes da Silva FH, Kamp A. Evoked
most promising in yielding new insights in the neural processes responses. Part A. In: Buser P, ed., Rémond A, ed.-in-chief.
underlying the generation of ERPs. Handbook of Electroencephalography and Clinical Neurophysiology.
Vol 8. Amsterdam: Elsevier; 1975.
2. John ER, Thatcher RW. Functional Neuroscience/Neurometrics.
CONCLUDING REMARKS Vol II. Hillsdale, NJ: Lawrence Erlbaum; 1977.
3. Callaway E, Tueting P, Koslow SH, eds. Event-related Brain
Since the early phase of ERPs studies successive waves of Potentials in Man. New York: Academic Press; 1978.
methodologic advances have taken place. This field has been 4. Barber C, ed. Evoked Potentials. Lancaster: MTP Press; 1980.
always a fruitful area for collaborative investigations between 5. Chiappa KH, ed. Evoked Potentials in Clinical Medicine. 3rd ed.
biomedical engineers, particularly signal analysts, statisticians, Lippincott-Raven Press; 1997:700pp.
and physicists on the one hand, and neurophysiologists, neurol- 6. Misulis KE, Fakhoury T. Spehlmann’s Evoked Potential Primer. 3rd
ogists, and psychologists on the other. The last decade has seen ed. Butterworth-Heinemann; 2001:184pp.
an impressive increase of the number of analytic tools within 7. Barrett G. Analytic techniques in the estimation of evoked poten-
this field and of theoretical models with the emergence of novel tials. In: Lopes da Silva FH, Storm van Leeuwen W, Rémond A, eds.
concepts about the generation and nature of ERPs. In this way Clinical Applications of Computer Analysis of EEG and Other
Neurophysiological Signals. Handbook of Electroencephalography
a plethora of methods are now available but the practical clini-
and Clinical Neurophysiology. Vol 2. Amsterdam: Elsevier;
cal neurophysiologist may have difficulty in finding his way 1986:311–334 [new series].
within this multitude of challenging possibilities. The relative 8. Gevins AS, Cutillo BA. Signals of cognition. In: Lopes da Silva FH,
value of different methods with respect to performance in ana- Storm van Leeuwen W, Rémond A, eds. Clinical Applications of
lyzing ERPs cannot, in general, be assessed since most published Computer Analysis of EEG and Other Neurophysiological Signals.
932 Part VII ■ Evoked Potentials and Event-Related EEG Phenomena

Handbook of Electroencephalography and Clinical Neurophysiology. Springfield, VA: Defense Documentation Center, Defense Supply
Vol 2. Amsterdam: Elsevier; 1986:335–384 [new series]. Agency, Clearing House for Federal Scientific and Technical
9. Regan D. Human Brain Electrophysiology: Evoked Potentials and Information; 1965.
Evoked Magnetic Fields in Science and Medicine. New York: Elsevier 32. Morrison DF. Multivariate Statistical Methods. New York:
Science; 1989:672pp. McGraw-Hill; 1967.
10. Silberstein RB. Neuroelectric measures of mind. In: Nunez PL, ed. 33. van Rotterdam, 1970.
Neocortical Dynamics and Human EEG Rhythms. Oxford 34. Donchin E. Discriminant analysis in average evoked response
University Press; 1995:591–627. studies: the study of single trial data. Electroencephalogr Clin
11. Gevins AS, Cutillo BA. Neuroelectric measures of mind. In: Nunez Neurophysiol. 1969;27:311–314.
PL, ed. Neocortical Dynamics and Human EEG Rhythms. Oxford 35. Donchin E, Herning RI. A simulation study of the efficacy of step-
University Press; 1995:304–338. wise discriminant analysis in the detection and comparison of
12. Dawson GD. A summation technique for detecting small signals in event related potentials. Electroencephalogr Clin Neurophysiol.
a large irregular background. J Physiol. 1951;115:2–3. 1975;38:51–68.
13. Dawson GD. A summation technique for the detection of small 36. John ER, Walker P, Cawood D, et al. Factor analysis of evoked
evoked potentials. Electroencephalogr Clin Neurophysiol. 1954;6: potentials. Electroencephalogr Clin Neurophysiol. 1973;33:33–34.
153–154. 37. Rebert CS. Neuroelectric measures of lateral specialization in rela-
14. Barlow JS. An electronic method for detecting evoked responses of tion to performance. Electroencephalogr Clin Neurophysiol Suppl.
the brain and for reproducing their average waveforms. 1978;34:231–238.
Electroencephalogr Clin Neurophysiol. 1957;9(2):340–343. 38. Donchin E, Cohen L. Average evoked potentials and intramodality
15. Brazier MAB. Some uses of computers in experimental neurology. selective attention. Electroencephalogr Clin Neurophysiol.
Exp Neurol. 1960;2:123–143. 1967;22:537–546.
16. Clark WA. Average response computer (ARC1). Prog Rep Res Lab 39. Freeman WJ. Use of digital adaptive filters for measuring pre-
Electron MIT. 1958;114–117. pyriform evoked potentials from cats. Exp Neurol. 1964;
17. Sayers P, Beagley HA, Hanshall WR. The mechanisms of auditory 10:475–492.
evoked EEG responses. Nature. 1974;247:481–483. 40. Freeman WJ. Mass Action in the Nervous System. New York:
18. McClelland RJ, Sayers BM. Towards fully objective evoked Academic Press; 1975.
response audiometry. Br J Audiol. 1983;17(4):263–270. 41. Spekreijse H, Estevez O, Reits D. Visual evoked potentials and the
19. Makeig S, Debener S, Onton J, et al. Mining event-related brain physiological analysis of visual processes in man. In: Desmedt JE,
dynamics. Trends Cogn Sci. 2004;8(5):204–210. ed. Visual Evoked Potentials in Man. Oxford: Clarendon Press;
20. Mazaheri A, Picton TW. EEG spectral dynamics during discrimi- 1976:3–15.
nation of auditory and visual targets. Brain Res Cogn Brain Res. 42. McGillem CD, Aunon JI. Measurement of signal components in
2005;24(1):81–96. single visually evoked brain potentials. IEEE Trans Biomed Eng.
21. Mäkinen V, Tiitinen H, May P. Auditory event related responses are 1977;24(3):232–241.
generated independently of ongoing brain activity. Neuroimage. 43. Steeger GH. Ein Bettrag zur Verbesserung der Messung akustisch
2005;24:961–1068. evozierter Potentiate variabler Latenz im ElektroenzERPhalogramm
22. Klimesch W, Hanslmayr S, Sauseng P, et al. Distinguishing the des Menschen [thesis]. Universitat Erlangen Nürnberg; 1979. ^
evoked response from phase reset: a comment to Makinen et al. 44. Steeger GH, Reinhardt J. Ein Mittelwertrechner fŸr reizausgel s ste
Neuroimage. 2006;29(3):808–811. neuroclektrische Signale varibler Latenz. Biomed Tech. 1977;
23. Mazaheri A, Jensen O. Posterior {alpha} activity is not phase-reset 22:68–74.
by visual stimuli. Proc Natl Acad Sci U S A. 2006;103(8):2948–2952. 45. Ten Hoopen M. Variance in average response computation: regu-
24. Lopes da Silva FH. Event-related neural activities: what about lar versus irregular stimulation. In: Rémond A, ed. Handbook of
phase? Prog Brain Res. 2006;159:3–17. Electroencephalography and Clinical Neurophysiology, Part A. Vol 8.
25. Shah AS, Bressler SL, Knuth KH, et al. Neural dynamics and the Amsterdam: Elsevier; 1975;151–158.
fundamental mechanisms of event-related brain potentials. Cereb 46. Arnal D, Gerin P. Étude du bruit résiduel des potentials évoqués
Cortex. 2004;14:476–483. moyens. Electroencephalogr Clin Neurophysiol. 1969;27:315–321.
26. Nolte G, Bai O, Wheaton L, et al. Identifying true brain interaction 47. Ruchkin DS. An analysis of average response computations based
from EEG data using the imaginary part of coherency. Clin upon aperiodic stimuli. IEEE Trans Biomed Eng. 1965;12:87–94.
Neurophysiol. 2004;115:2292–2307. 48. Woody CD. Characterization of an adaptive filter for the analysis
27. John ER, Ruchkin DS, Vidal JJ. Measurement of event-related of variable latency neuroelectric signals. Med Biol Eng.
potentials. In: Callaway E, Tueting P, Koslow SH, eds. Event- 1967;5:539–553.
related Brain Potentials in Man. New York: Academic Press; 1978: 49. Helstrom CW. Statistical Theory of Signal Detection. 2nd ed.
93–138. Oxford: Pergamon Press; 1968.
28. Donchin E. A multivariate approach to the analysis of average 50. Pfurtscheller G, Cooper R. Selective averaging of the intracerebral
evoked potentials. IEEE Trans Biomed Eng. 1966;13:131–139. click evoked responses in man: an improved method of measuring
29. John ER, Ruchkin DS, Villegas J. Signal analysis and behavioral latencies and amplitudes. Electroencephalogr Clin Neurophysiol.
correlates of evoked potentials configurations in cats. Ann N Y 1975;38:187–190.
Acad Sci. 1964;112:362–420. 51. Coppola R, Tabor R, Buchsbaum MS. Signal to noise ratio and
30. Ruchkin DS, Villegas J, John ER. An analysis of average evoked response variability measurements in single trial evoked poten-
potentials making use of least mean square techniques. Ann N Y tials. Electroencephalogr Clin Neurophysiol. 1978;44:214–222.
Acad Sci. 1964;15:799–826. 52. Li R, Keil A, Principe JC. Single-trial P300 estimation with a spa-
31. Streeter DN, Raviv J. Research on Advanced Computer Methods for tiotemporal filtering method. J Neurosci Methods. 2009;177(2):
Biological Data Processing. Rept. ASTIA, Doc. AD 637452. 488–496.
Chapter 44 ■ Event-Related Potentials: General Aspects of Methodology and Quantification 933

53. Poolman P, Frank RM, Luu P, et al. A single-trial analytic frame- 75. Macpherson H, Pipingas A, Silberstein R. A steady state visually
work for EEG analysis and its application to target detection and evoked potential investigation of memory and ageing. Brain Cogn.
classification. Neuroimage. 2008;42(2):787–798. 2009;69(3):571–579.
54. Wiener N. Extrapolation, Interpolation and Smoothing of 76. Rees A, Green GG, Kay RH. Steady-state evoked responses to sinu-
Stationary Time Series. New York: John Wiley & Sons; 1949. soidally amplitude-modulated sounds recorded in man. Hear Res.
55. Walter DO. A posteriori “Wiener filtering” of averaged evoked 1986;23(2):123–133.
responses. Electroencephalogr Clin Neurophysiol Suppl. 1968;27: 77. Picton TW, Skinner CR, Champagne SC, et al. Potentials evoked by
61–70. the sinusoidal modulation of the amplitude or frequency of a tone.
56. Doyle DJ. Some comments on the use of Wiener-filtering for the J Acoust Soc Am. 1987;82(1):165–178.
estimation of evoked potentials. Electroencephalogr Clin Neuro- 78. Picton TW, John MS, Purcell DW, et al. Human auditory steady-
physiol. 1975;38:533–534. state responses: the effects of recording technique and state of
57. Albrecht V, Lansky P, Indra M, et al. Wiener filtration versus aver- arousal. Anesth Analg. 2003;97(5):1396–1402.
aging of evoked responses. Biol Cybern. 1977;27:147–154. 79. Ross B, Picton TW, Herdman AT, et al. The effect of attention on
58. Strackee J, Cerri SA. Some statistical aspects of digital Wiener fil- the auditory steady-state response. Neurol Clin Neuroph

You might also like