You are on page 1of 824

Academic Press is an imprint of Elsevier

525 B Street, Suite 1900, San Diego, CA 92101-4495, USA


30 Corporate Drive, Suite 400, Burlington, MA 01803, USA
32 Jamestown Road, London, NW1 7BY, UK
Radarweg 29, PO Box 211, 1000 AE Amsterdam, The Netherlands

Second edition 2009

Copyright 2009 Elsevier Ltd. All rights reserved.

No part of this publication may be reproduced, stored in a retrieval system


or transmitted in any form or by any means electronic, mechanical, photocopying,
recording or otherwise without the prior written permission of the publisher

Permissions may be sought directly from Elseviers Science & Technology Rights
Department in Oxford, UK: phone (44) (0) 1865 843830; fax (44) (0) 1865 853333;
email: permissions@elsevier.com. Alternatively you can submit your request
online by visiting the Elsevier web site at http://elsevier.com/locate/permissions,
and selecting obtaining permission to use Elsevier material

Notice

No responsibility is assumed by the publisher for any injury and/or damage to persons
or property as a matter of products liability, negligence or otherwise, or from any use
or operation of any methods, products, instructions or ideas contained in the material
herein. Because of rapid advances in the medical sciences, in particular, independent
verication of diagnoses and drug dosages should be made

Medicine is an ever-changing eld. Standard safety precautions must be followed, but as


new research and clinical experience broaden our knowledge, changes in treatment and
drug therapy may become necessary or appropriate. Readers are advised to check the most
current product information provided by the manufacturer of each drug to be administered
to verify the recommended dose, the method and duration of administrations, and
contraindications. It is the responsibility of the treating physician, relying on experience
and knowledge of the patient, to determine dosages and the best treatment for each
individual patient. Neither the publisher nor the authors assume any liability for any
injury and/or damage to persons or property arising from this publication

British Library Cataloguing-in-Publication Data


A catalogue record for this book is available from the British Library

Library of Congress Cataloging-in-Publication Data


A catalog record for this book is available from the Library of Congress

ISBN: 978-0-12-374001-4

For information on all Academic Press publications


visit our website at elsevierdirect.com

Printed and bound in USA

09 10 11 12 10 9 8 7 6 5 4 3 2 1
Preface to the 2nd Edition

In the 6 years since the publication of the rst edition of Asthma and COPD there has been
substantial progress in understanding the biological basis of both of these obstructive diseases. In
contrast both our understanding of the physiological basis of these conditions and their treatment
has improved very little. We rmly believe that the progress made in understanding the basic disease
biology that has accrued since 2002 will lead, in the years to come to new and better treatments of
these crippling obstructive airway diseases. As our knowledge advances, we now appreciate the areas
of overlap and distinction between these two conditions which share many physiological similari-
ties. Most of the chapters of this book highlight both the similarities and dierences between these
conditions.
In this era of widely available information on the internet it is reasonable to ask what value is
served by a book of this type? The answer is simple. The editors have assembled the worlds experts
on these topics and commissioned them to write succinct reviews focusing on their particular area
of expertise. For the student of the biology or treatment of these conditions, this compilation is an
important resource of up-to-date information. For the researcher or clinician, the investigative or
clinical problems that each of face on a daily basis are clearly illuminated by topic experts. We hope
that you will nd this summary of value to you in understanding these two highly variegated, closely
similar but clearly distinct entities at the biological and clinical level.

Peter J. Barnes
Jeery M. Drazen
Stephen I. Rennard
Neil C. Thomson

ix
About the Editors

Peter J Barnes, DM, DSc, FRCP, FMedSci, the Scientic Committee of the British Lung
FRS, is a Professor of Thoracic Medicine at Foundation. He has co-edited several textbooks
the National Heart and Lung Institute, Head on asthma and COPD and published over 150
of Respiratory Medicine at Imperial College, peer-reviewed papers on asthma. His current
and Honorary Consultant Physician at Royal research interests include corticosteroid insen-
Brompton Hospital, London. He qualied at sitivity in smokers with asthma, biomarkers in
Cambridge and Oxford Universities and was asthma and COPD and assessment of novel
appointed to his present post in 1987. He has treatments for asthma.
published over 1000 peer-review papers on
asthma, COPD, and related topics and has Jerey M Drazen, MD, was born in St. Louis,
edited over 40 books. He is also amongst the Missouri. He graduated from Tufts University
top 50 most highly cited researchers in the with a major in Physics, and from Harvard
world and has been the most highly cited clini- Medical School. He served his medical intern-
cal scientist in the United Kingdom and the ship and residency at Peter Bent Brigham
most highly cited respiratory researcher in the Hospital in Boston and was a Clinical Fellow
world over the last 20 years. He was elected a and Research Fellow at Harvard Medical
Fellow of the Royal Society in 2007, the rst School and Harvard School of Public Health.
respiratory researcher for over 150 years. He is Thereafter, he joined the Pulmonary Divisions
currently a Member of the Scientic Committee of the Harvard hospitals and served for many
of the WHO/NIH global guidelines on asthma years as Chief of the Combined Pulmonary
(GINA) and COPD (GOLD). He also serves Divisions at Beth Israel and Brigham and
on the Editorial Board of over 30 journals and Womens Hospitals.
is currently an Associate Editor of Chest and Currently, he is a Senior Physician at
Respiratory Editor of PLoS Medicine. He has the Brigham and Womens Hospital, and the
given several prestigious lectures, including the Distinguished Parker B. Francis Professor of
Amberson Lecture at the American Thoracic Medicine at Harvard Medical School, as well
Society and the Sadoul Lecture at the European as Professor of Physiology, Harvard School of
Respiratory Society. Public Health, and adjunct Professor of Medicine
at Boston University School of Medicine.
Neil C Thomson, MD, FRCP, is a Professor He has served on the NIH Respiratory
of Respiratory Medicine at the University and Applied Physiology Study Section, the
of Glasgow, Head of Respiratory Medicine NIH Pulmonary Disease Advisory Council,
within the Division of Immunology, Infection the NIH Lung Biology and Pathology Study
& Inammation, and Honorary Consultant Section, and the NHLBI Advisory Council.
at Gartnavel General Hospital, Glasgow. He Through his research, he dened the role
graduated from the University of Glasgow and of novel endogenous chemical agents in asthma.
undertook postgraduate training in Glasgow, This led to four new licensed pharmaceuticals
London and McMaster University, Canada. for asthma used in the treatment of millions of
He is a former Member of the Committee people worldwide. He has published nearly 500
for Safety of Medicine and former Chair of papers and edited 6 books.

xi
About the Editors

He has been a member of the Editorial Boards of many Institutes of Health where he remained for seven years,
prestigious journals, including the Journal of Applied Physiology, conducting research in the cell biology of lung disease.
American Journal of Physiology, Pulmonary Pharmacology, He joined the University of Nebraska in 1984 as Chief
Experimental Lung Research, Journal of Clinical Investigation, of Pulmonary and Critical Care, a position he retained
American Journal of Respiratory Cell and Molecular Biology, and until 1997. He was the Director of the Nebraska Oce of
the American Journal of Medicine. In addition, he has been an Tobacco Control and Research from 1997 until 2008.
associate editor of the Journal of Clinical Investigation and the Dr. Rennard currently serves on the Board of
American Review of Respiratory Disease. Directors of the COPD Foundation and the Alpha-1
In 2000, he assumed the post of Editor-in-Chief of Foundation. He is a member of the American Thoracic
the New England Journal of Medicine. During his tenure, Society Committee on Corporate Relations and the
the journal has published major papers advancing the sci- National Heart Lung Education Program Executive
ence of medicine, including the rst descriptions of SARS Committee. He is an external advisor to the Thomas Petty
and papers modifying the treatment of cancer, heart disease, Aspen Lung Conference and the University of California-
and lung disease. The journal, which has more than half a Davis Pulmonary Training Grant.
million readers every week, has the highest impact factor of Dr. Rennard is active in several professional societies
any medical journal publishing original research. and has previously served on the Board of Directors for the
American Thoracic Society, the Council of the American
Stephen I Rennard, MD, is Larson Professor of Medicine Lung Association and was a Governor for the American
in the Pulmonary and Critical Care Medicine Section of College of Chest Physicians. He served on the American
the Department of Internal Medicine at the University Board of Internal Medicine, Pulmonary Section and was a
of Nebraska Medical Center in Omaha, Nebraska, and member of the expert panel which prepared the global GOLD
courtesy Professor of the Department of Pathology and guidelines for COPD for the WHO/NHLBI. He has served
Microbiology and the Department of Genetics, Cell on several task force committees, including the ATS/ERS task
Biology and Anatomy. He received an AB with honors in force that prepared the ATS/ERS joint COPD Standards.
Folklore and Mythology from Harvard University and an Professor Rennard maintains an active program of
MD with honors from the Baylor College of Medicine, clinical investigation in COPD and smoking cessation and
Houston, Texas. He completed internal medicine train- a program of basic research in the mechanisms of lung tis-
ing at Barnes Hospital, Washington University, St. Louis, sue repair and remodeling, including the role of stem cells
Missouri and trained in Pulmonary Diseases at the National in disease pathogenesis and repair.

xii
List of Contributors

Ian M. Adcock (31) Joan Albert Barber (20)


National Heart and Lung Institute, Imperial Servei de Pneumologia, Hospital Clinic,
College School of Medicine, London, UK IDIBAPS, Universitat de Barcelona, Barcelona,
Spain
Alvar Agusti (44)
Hospital University Son Dureta, Servi Peter J. Barnes (32, 33, 34, 40, 49, 50, 51, 52, 61)
Pneumologia, Palma de Majorca, Spain National Heart and Lung Institute (NHLI),
Fundacion Caubet-CIMERA Clinical Studies Unit, Imperial College,
CIBER Enfermedades Respiratorias London, UK

Jennifer A. Alison (38) Bianca Begh (41)


Discipline of Physiotherapy, Faculty of Health Department of Respiratory Diseases,
Sciences, University of Sydney, Lidcombe, University of Modena and Reggio Emilia,
NSW, Australia Modena, Italy

Yassine Amrani (18)


Richard C. Boucher (16)
Department of Infection, Immunity and
Department of Medicine, The University of
Inammation, University of Leicester, UK
North Carolina at Chapel Hill, Chapel Hill,
NC, USA
Sandra D. Anderson (38)
Department of Respiratory Medicine,
Royal Prince Alfred Hospital, Camperdown, Andrew Briggs (62)
NSW, Australia Section of Public Health and Health Policy,
University of Glasgow, Glasgow, Scotland, UK
Kjetil Ask (29)
Centre for Gene Therapeutics, Department of Guy G. Brusselle (10)
Pathology and Molecular Medicine, McMaster Department of Pulmonary Medicine, Erasmus
University, Hamilton, ON, Canada University Medical Center, Rotterdam,
The Netherlands
Jon Ayres (39)
Department of Environmental and A. Sonia Buist (1)
Occupational Medicine, Liberty Safe Work Division of Pulmonary and Critical Care
Research Centre, University of Aberdeen, Medicine, Oregon Health Sciences
Aberdeen, UK University, Portland, OR, USA

xiii
List of Contributors

Kimberlie Burns (16) Christopher J. Corrigan (55)


Department of Medicine, The University of North Carolina Department of Asthma, Allergy and Respiratory Science,
at Chapel Hill, Chapel Hill, NC, USA Guys Hospital, London, UK
MRC and Asthma UK Centre in Allergic Mechanisms of
Andrew Bush (65) Asthma Kings College London, UK
Department of Respiratory Pediatrics, Imperial
College of Science, Technology and Manuel G. Cosio (13)
Medicine, Royal Brompton Hospital and Meakins-Christie Laboratories and the Respiratory
National Heart and Lung Institute, London, UK Division, Department of Medicine, McGill University,
Montreal, Quebec, Canada
Carlos A. Camargo Jr. (64)
Department of Emergency Medicine, Massachusetts Harvey O. Coxson (43)
General Hospital, Harvard Medical School, Boston, MA, Department of Radiology, James Hogg iCAPTURE Centre
USA for Cardiovascular and Pulmonary Research, University of
British Columbia, Vancouver, BC, Canada
Gaetano Caramori (31)
Adnan Custovic (46)
National Heart and Lung Institute, Imperial College
University of Manchester, Manchester, UK
School of Medicine, London, UK
Marc Decramer (58)
George H. Caughey (9) Respiratory Rehabilitation and Respiratory Division,
Cardiovascular Research Institute and Department of University Hospital, Leuven, Belgium
Medicine, University of California at San Francisco, USA
Faculty of Kinesiology and Rehabilitation Sciences,
Bartolome R. Celli (54) Department of Rehabilitation Sciences, Katholieke
Division of Pulmonary and Critical care, Universiteit Leuven, Leuven, Belgium
Caritas -St. Elizabeths Medical Center, Tufts University,
Dawn L. DeMeo (2)
Boston, MA, USA
Channing Laboratory, Brigham and Womens Hospital,
Harvard Medical School, Boston, MA, USA
Michael A. Chandler (47)
University of Nebraska Medical Center, Omaha, NE, USA Jerey M. Drazen (24, 33)
Department of Medicine, Pulmonary Division,
Richard N. Channick (56)
Birgham and Womens Hospital, Boston, MA, USA
Division of Pulmonary and Critical Care Medicine,
University of California, San Diego School of Medicine, Harvard Medical School, Editorial Oce New England
La Jolla, CA, USA Journal of Medicine, Boston, MA, USA

Leonardo M. Fabbri (41)


Moira Chan-Yeung (36)
Department of Respiratory Diseases, University of Modena
Department of Medicine, Respiratory Division, University
and Reggio Emilia, Modena, Italy
of British Columbia, Vancouver, BC, Canada
Jack Gauldie (29)
Kian Fan Chung (27, 52) Centre for Gene Therapeutics, Department of Pathology
National Heart and Lung Institute (NHLI), and Molecular Medicine, McMaster University, Hamilton,
Imperial College, London, UK ON, Canada

Donald W. Cockcroft (35) Maurice Godfrey (22)


Division of Respirology, Critical Care and Sleep Medicine, Center for Human Molecular Genetics, Munroe Meyer
Department of Medicine, University of Sasketchewan, Institute, University of Nebraska Medical Center, Omaha,
Royal University Hospital, Saskatoon, Saskatchewan, NE, USA
Canada
Stefano Guerra (3)
Lorenzo Corbetta (41) Arizona Respiratory Center and Mel and Enid Zuckerman
Department of Respiratory Diseases, University of Firenze, College of Public Health, University of Arizona,Tucson,
Firenze, Italy AZ, USA

xiv
List of Contributors

Ian P. Hall (48) Onn Min Kon (49)


Division of Therapeutics and Molecular Medicine, National Heart and Lung Institute (NHLI), Clinical
University Hospital of Nottingham, Nottingham, UK Studies Unit, Imperial College, London, UK

Trevor T. Hansel (49) Samuel L. Krachman (57)


National Heart and Lung Institute (NHLI), Clinical Section of Pulmonary and Critical Care Medicine,
Studies Unit, Imperial College, London, UK Temple University School of Medicine, Philadelphia,
PA, USA
James C. Hogg (6)
UBC McDonald Research Laboratories, St. Pauls Hospital, Vera P. Krymskaya (18)
University of British Columbia, Vancouver, BC, Canada Pulmonary, Allergy and Critical Care Division, Airways
Biology Initiative, University of Pennsylvania,
Stephen T. Holgate (7) Philadelphia, PA, USA
School of Medicine, University of Southampton,
Southampton, UK Bart N. Lambrecht (10)
Department of Respiratory Medicine, University Hospital
Gabor Horvath (19) Ghent, Belgium
Department of Pulmonology, Semmelweiss University, Department of Pulmonary Medicine, Erasmus University,
Budapest, Hungary Rotterdam, The Netherlands

Charles G. Irvin (5) Georey J. Laurent (15)


Department of Medicine and Physiology, University of UCLMS, Center for Respiratory Research, University
Vermont College of Medicine, Burlington, VT, USA St/Rayne Institute, London, UK

Kazuhiro Ito (30) Bruce D. Levy (24)


Section of Airway Disease, National Heart and Lung Department of Medicine, Pulmonary Division,
Institute, Imperial College, London, UK Brigham and Womens Hospital, Boston, MA, USA

Fabrizio Luppi (41)


Wim Janssens (58)
Department of Respiratory Diseases, University of
Respiratory Rehabilitation and Respiratory Division,
Modena and Reggio Emilia, Modena, Italy
University Hospital, Leuven, Belgium
Jean-Luc Malo (36)
Sebastian L. Johnston (37) Department of Chest Medicine, Sacr-Coeur Hospital,
Department of Respiratory Medicine, National Heart and Montreal, Canada
Lung Institute, Imperial College London, London, UK
James G. Martin (13)
Susan Kennedy (36) Meakins-Christie Laboratories and the Respiratory
School of Environmental Health, University of British Division, Department of Medicine, McGill University,
Columbia, Vancouver, BC, Canada Montreal, Quebec, Canada

Sergei A. Kharitonov (30, 42) Fernando D. Martinez (3)


Section of Airway Disease, National Heart and Lung Arizona Respiratory Center and Mel and Enid Zuckerman
Institute, Imperial College, London, UK College of Public Health, University of Arizona, Tucson,
AZ, USA
Darryl A. Knight (15)
Department of Pharmacology and Therapeutics, Simon D. Message (37)
University of British Columbia, Vancouver, BC, Canada Department of Respiratory Medicine, National Heart and
Lung Institute, Imperial College London, London, UK
Martin Kolb (29)
Centre for Gene Therapeutics, Department of Pathology Lynne A. Murray (15)
and Molecular Medicine, McMaster University, Manager of Pharmocology Promedior, Inc.
Hamilton, ON, Canada Malvern, PA, USA

xv
List of Contributors

Paul M. OByrne (23) Stephen I. Rennard (34, 47, 66)


Firestone Institute for Respiratory Health, St. Josephs Pulmonary and Critical Care Medicine, University of
Healthcare and Department of Medicine, McMaster Nebraska Medical Center, Omaha, NE, USA
University, Hamilton, ON, Canada
Roberto Rodrguez-Roisin (20)
Reynold A. Panettieri Jr. (18) Servei de Pneumologia, Hospital Clinic, IDIBAPS,
Pulmonary, Allergy and Critical Care Division, Airways Universitat de Barcelona, Barcelona, Spain
Biology Initiative, University of Pennsylvania, Philadelphia,
PA, USA Duncan F. Rogers (17)
Section of Airway Disease, National Heart and Lung
Martyn R. Partridge (68) Institute, Imperial College, London, UK
The Faculty of Medicine, Imperial College, London, UK
Brian H. Rowe (64)
Rodolfo M. Pascual (63)
Department of Emergency Medicine, University of
Section on Pulmonary, Critical Care, Allergy and
Alberta, and Capital Health, Edmonton, AB, Canada
Immunologic Diseases, Department of Internal Medicine,
Wake Forest University School of Medicine, Medical
Lewis J. Rubin (56)
Center Boulevard, Winston-Salem, NC, USA
Division of Pulmonary and Critical Care Medicine,
University of California, San Diego School of Medicine,
Ian D. Pavord (42)
La Jolla, CA, USA
Department of Respiratory Medicine and Thoracic Surgery,
Institute for Lung Health, Gleneld Hospital,
Elizabeth Sapey (14)
Leicester, UK
Department of Medicine, Birmingham University,
Birmingham, UK
James E. Pease (26)
Leukocyte Biology Section, National Heart and
Sanjay Sethi (53)
Lung Institute, Imperial College London, London, UK
Division of Pulmonary, Critical Care and Sleep Medicine,
Department of Medicine, University of Bualo, State
Stephen P. Peters (63)
University of New York, and Veterans Aairs Western
Section on Pulmonary, Critical Care, Allergy and
New York Health Care System, Bualo, New York, USA
Immunologic Diseases, Department of Internal Medicine,
Wake Forest University School of Medicine, Medical
Center Boulevard, Winston-Salem, NC, USA Pallav L. Shah (59)
Royal Brompton Hospital and National Heart and
Michael I. Polkey (59) Lung Institute, London, UK
Royal Brompton Hospital and National Heart and
Lung Institute, London, UK Steven D. Shapiro (8, 28)
Department of Medicine, University of Pittsburgh,
Dirkje S. Postma (4) Pittsburgh, PA, USA
Department of Pulmonology, University Medical Center
Groningen, University of Groningen, Groningen, The Dean Sheppard (21)
Netherlands Department of Medicine and Lung Biology Center,
University of California, San Francisco, CA, USA
Irfan Rahman (25)
Department of Environmental Medicine, Lung Biology Stephanie A. Shore (8)
and Disease Program, University of Rochester Medical Molecular and Integrative Physiological Sciences Program,
Center, Rochester, NY, USA Harvard School of Public Health, Boston, MA, USA

Scott H. Randell (16) Edwin K. Silverman (4)


Departments of Cell and Molecular Physiology and Channing Laboratory and Division of Pulmonary and
Medicine, The University of North Carolina at Chapel Hill, Critical Care Medicine, Brigham and Womens Hospital,
NC, USA Harvard Medical School, Boston, MA, USA

xvi
List of Contributors

Dagmar Simon (12) Galen B. Toews (11)


Department of Dermatology, Inselspital, University of Division of Pulmonary and Critical Care Medicine,
Bern, Bern, Switzerland University of Michigan Health System, Ann Arbor,
MI, USA
Hans-Uwe Simon (12)
Department of Pharmacology, University of Bern, Bern,
Thierry Troosters (58)
Switzerland
Respiratory Rehabilitation and Respiratory Division,
University Hospital, Leuven, Belgium
Michael W. Sims (18)
Pulmonary, Allergy and Critical Care Division, Airways Faculty of Kinesiology and Rehabilitation Sciences,
Biology Initiative, University of Pennsylvania, Philadelphia, Department of Rehabilitation Sciences, Katholieke
PA, USA Universiteit Leuven, Leuven, Belgium

Don D. Sin (45) Adam Wanner (19)


The James Hogg iCAPTURE Center for Cardiovascular Division of Pulmonary and Critical Care Medicine,
and Pulmonary Research, St. Pauls Hospital, Vancouver, University of Miami Miller School of Medicine, Miami,
BC, Canada FL, USA

Helen Starkie (62)


Section of Public Health and Health Policy, University of Jadwiga A. Wedzicha (67)
Glasgow, Glasgow, Scotland, UK Department of Respiratory Medicine, University College
of London, London, UK
Robert A. Stockley (14)
Lung Investigation Unit, University Hospital Birmingham, Scott T. Weiss (2)
NHS Foundation Trust, Edgbaston, Birmingham, UK Channing Laboratory, Brigham and Womens Hospital,
Harvard Medical School, Boston, MA, USA
Andrew J. Tan (49)
National Heart and Lung Institute (NHLI), Clinical
Studies Unit, Imperial College, London, UK Timothy J. Williams (26)
Leukocyte Biology Section, National Heart and Lung
Neil C. Thomson (32, 40, 60) Institute, Imperial College London, London, UK
Department of Respiratory Medicine, Division of
Immunology, Infection and Inammation, University of Ashley Woodcock (46)
Glasgow, Glasgow, UK, University of Manchester, Manchester, UK

Omar Tliba (18)


Olivia Wu (62)
Pulmonary, Allergy and Critical Care Division, Airways
Section of Public Health and Health Policy,
Biology Initiative, University of Pennsylvania,
University of Glasgow, Glasgow, Scotland, UK
Philadelphia, PA, USA

Martin J. Tobin (57) Zhou Xing (29)


Loyola University of Chicago, Stritch School of Medicine Centre for Gene Therapeutics, Department of Pathology
and Hines Veterans Administration Hospital, Maywood, and Molecular Medicine, McMaster University, Hamilton,
IL, USA ON, Canada

xvii
Definitions,
1
PART

Epidemiology, and
Genetics of Asthma
and COPD
Definitions
1
CHAPTER

Denitions of diseases evolve over time as our of the GINA Guidelines [1] proposes an oper-
understanding of them changes. For example, ational description of asthma as:
A. Sonia Buist
until recently, the presence or absence of revers- Division of Pulmonary and Critical Care
ibility was considered to be the key distinction a chronic inammatory disorder of the air- Medicine, Oregon Health and Sciences
between asthma and chronic obstructive pul- ways in which many cells and cellular ele-
University, Portland, OR, USA
monary disease (COPD) with reversible air- ments play a role. The chronic inammation
ow obstruction the hallmark of asthma, and is associated with airway hyperresponsiveness
irreversible airow obstruction the hallmark of that leads to recurrent episodes of wheezing,
COPD. Better understanding of both diseases breathlessness, chest tightness, and coughing,
has brought new denitions that acknowl- particularly at night or in the early morn-
edge the overlap and highlight the similarities ing. These episodes are usually associated with
and dierences between them. The important widespread, but variable, airow obstruction
change in our understanding is the recognition within the lung that is often reversible either
that chronic inammation underlies both dis- spontaneously or with treatment.
eases. The nature of the inammation diers,
however, as does the response to anti-inam- This is very similar to the denition pro-
matory medications, as described in detail in posed by the National Asthma Education and
later chapters. This chapter draws heavily on the Prevention Program in their 1997 guidelines [3].
latest information on asthma and COPD that Both denitions imply that asthma is one disor-
is included in the guidelines on the diagnosis der, rather than multiple complex disorders and
and management of these diseases from two syndromes a notion that is receiving increasing
widely respected global initiatives, the Global attention [4, 5].
Initiative for Asthma (GINA) [1] and the
Global Initiative for Chronic Obstructive Lung
Disease (GOLD) [2], as updated in 2006. COPD
Until quite recently, denitions of COPD used
to include the terms chronic bronchitis and
DEFINITIONS emphysema. The GOLD Guidelines, rst
published in 2002 [6], and revised in 2006
[2], the American Thoracic Society/European
Asthma Respiratory Society (ATSERS) Guidelines
published in 2004 [7], and the NICE
Most of the denitions for asthma have empha- Guidelines [8, 9] published in 2004 deliberately
sized the characteristics of uctuations over omitted these terms and used just the umbrella
time in bronchoconstriction and the reversible term COPD. The main reason for this is that
nature of the disease [1, 3]. As the pathophysi- the use of many dierent terms for COPD has
ological basis of asthma became clearer, deni- led to confusion on the part of health-care pro-
tions began to include a statement about the viders and the public. This in turn has stood in
pathological characteristics. The 2006 revision the way of COPD becoming widely recognized.

3
Asthma and COPD: Basic Mechanisms and Clinical Management

The 2006 revision of the GOLD guidelines [2]denes COPD belonging to a spectrum of diseases that all cause
COPD as: airow obstruction, to the concept of them as very dier-
ent diseases, and most recently to them both being inam-
a preventable and treatable disease with some signicant
matory diseases with important similarities and dierences,
extrapulmonary eects that may contribute to severity in
a theme that is taken up more fully in subsequent chapters.
individual patients. Its pulmonary component is charac-
The present thinking is illustrated in Fig. 1.1 from the 2006
terized by airow limitation that is not fully reversible.
GOLD guidelines, which shows both diseases involving an
The airow limitation is usually progressive and associ-
inammatory response that causes airow limitation, but
ated with an abnormal inammatory response of the lung
through geneenvironment interactions with dierent sen-
to noxious particles of gases.
sitizing agents, cell populations, and mediators. The airow
This denition includes the phrase preventable and limitation ranges from completely reversible (the asthma end
treatable to emphasize the importance of a positive attitude of the spectrum) to completely irreversible (the COPD end
to outcome and encourage a more active approach to man- of the spectrum). It is important to emphasize that COPD
agement. This denition also stresses that extrapulmonary and asthma often coexist, so the clinical picture may reect
eects [10, 11] are an integral part of COPD and need to be both conditions which may complicate the diagnostic proc-
taken into consideration in the diagnosis and management. ess and the pathological features. For example, some patients
with COPD have features of asthma and a mixed popula-
tion of inammatory cells; some patients with longstanding
asthma develop the pathological features of COPD.
SIMILARITIES AND DIFFERENCES Although the similarities between the diseases are
striking, it is the dierences in the inammatory processes
between the two diseases that dene their natural histories,
Over the past 30 years, the thinking about asthma and clinical presentations, and approaches to management. The
COPD has swung between the concept of asthma and major dierences in the pulmonary inammation between

Asthma COPD

Allergens Cigarette smoke

Epithelial cells Mast cell Alveolar macrophage Epithelial cells

CD4 cell CD8 cell


Eosinophils Neutrophils
(Th2) (Tc1)

Bronchoconstriction and Small airway fibrosis and


airway hyperresponsiveness alveolar destruction

Not fully
Reversible Airflow limitation
reversible
FIG. 1.1 Inflammatory cascade in
COPD and asthma (adapted from
Ref. [2] with permission).

4
Definitions 1
TABLE 1.1 Differences in pulmonary inflammation in COPD and asthma.

COPD Asthma Severe asthma

Cells Neutrophils  Eosinophils  Neutrophils 


Macrophages  Macrophages  Macrophages
CD8 T-cells (Tc1) CD4 T-cells (Th2) CD4 T-cells (Th2), CD8 T-cells (Tc1)

Key mediators IL-8 Eotaxin IL-8


TNF-, IL-1, IL-6 IL-4, IL-5, IL-13 IL-5, IL-13
NO  NO  NO 

Oxidative stress   

Site of disease Peripheral airways Proximal airways Proximal airways


Lung parenchyma Peripheral airways
Pulmonary vessels

Consequences Squamous metaplasia Fragile epithelium


Mucous metaplasia Mucous metaplasia
Small airway fibrosis Basement membrane
ParenchymaI destruction Bronchoconstriction
Pulmonary vascular remodeling

Response to therapy Small b/d response Large b/d response Smaller b/d response
Poor response to steroids Good response to steroids Reduced response to steroids

Source: From Ref. [2] with permission.


Notes: NO: nitric oxide; b/d: bronchodilator.

asthma and COPD are described in Table 1.1 from the TABLE 1.2 Classification of asthma severity by clinical features before
2006 GOLD report. These dierences are further explored treatment.
in later chapters.
Intermittent
Symptoms less than once a week
Severity classification Brief exacerbations
Nocturnal symptoms not more than twice a month
Both asthma and COPD are often stratied by severity, FEV1 or PEF  80% predicted
but the reason for this stratication is dierent for the two PEF or FEV1 variability  20%
conditions. In the 2006 GINA guidelines, the emphasis has
changed from using a severity classication to guide man- Mild persistent
agement to using a classication based on asthma control to Symptoms more than once a week but less than once a day
guide management. The GINA guidelines now recommend Exacerbations may affect activity and sleep
that asthma classication based on severity should now only Nocturnal symptoms more than twice a month
be used for research purposes. Tables 1.2 and 1.3 show the FEV1 or PEF  80% predicted
GINA severity classications based on lung function and PEF or FEV1 variability  2030%
asthma control, respectively.
For COPD, a severity classication based on spirometry Moderate persistent
is recommended for educational purposes, but not for guiding
Symptoms daily
management (Table 1.4). The current recommendation is that
Exacerbations may affect activity and sleep
treatment of COPD should be driven by the need to reduce
Nocturnal symptoms more than once a week
and control symptoms, not by level of lung function.
Daily use of inhaled short-acting 2-agonist
FEV1 or PEF 6080% predicted
PEF or FEV1 variability  30%
Differentiating between asthma
and COPD Severe persistent
Symptoms daily
It would be easy to dierentiate between asthma and COPD
Frequent exacerbations
if the latter occurred only in smokers and asthma in non-
Frequent nocturnal asthma symptoms
smokers. In fact, there is a clear diagnostic bias on the part of
Limitation of physical activities
physicians, with COPD more likely to be diagnosed in men
FEV1 or PEFs 60%  predicted
and asthma in women [12]. It is important to emphasize
PEF or FEV1 variability  30%
that both conditions may coexist in an individual, so many
will have the clinical and pathophysiological features of both Source: From Ref. [1] with permission.

5
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 1.3 Levels of asthma control.

Controlled (all of the Partly controlled (any measure


Characteristic following) present in any week) Uncontrolled

Daytime symptoms None (twice or less/week) More than twice/week

Limitations of activities None Any

Nocturnal symptoms/awakening None Any


Three or more features of partly
Meed for reliever/rescue treatment None (twice or less/week) More than twice/week controlled asthma present in any week

Lung function (PEF or FEV1)a Normal 80% predicted or personal best


(if known)

Exacerbations None One or more/yearb Once in any weekc

Source: From Ref. [1] with permission.


a
Lung function is not a reliable test for children 5 years and younger.
b
Any exacerbation should prompt review of maintenance treatment to ensure that it is adequate.
c
By definition, an exacerbation in any week makes that an uncontrolled asthma week.

TABLE 1.4 Spirometric classification of COPD severity based on post- TABLE 1.5 Clinical features of COPD and asthma.
bronchodilator FEV1.
Diagnosis Suggestive features
Stage 1: Mild FEV1/FVC  0.70
FEV1  80% predicted COPD Onset in mid-life
Symptoms slowly progressive
Stage II: Moderate FEV1/FVC  0.70
50%  FEV1  80% predicted Long history of tobacco smoking
Dyspnea during exercise
Stage III: Severe FEV1/FVC  0.70
Largely irreversible airflow limitation
30%  FEV1  50% predicted
Asthma Onset early in life (often childhood)
Stage IV: Very severe FEV1/FVC  0.70
Symptoms vary from day to day
FEV1  30% predicted or FEV1 
50% predicted plus chronic Symptoms at night/early morning
respiratory failure Allergy, rhinitis, and/or eczema also present
Family history of asthma
Source: From Ref. [2] with permission.
Notes: FEV1: forced expiratory volume in 1 s; FVC: forced vital capacity: respiratory Largely reversible airflow limitation
failure: arterial partial pressure of oxygen (PaQ2) less than 8.0 kPa (60 mmHg) with
Source: From Ref. [2] with permission.
or without arterial partial pressure of CO2 (PaCO2) greater than 6.7 kPa (50 mmHg)
while breathing air at sea level.

diseases. This makes dierentiating the diseases sometimes where other risk factors, such as heavy outdoor and indoor/
challenging for the clinician, especially in older adults who occupational air pollution, may be important risk fac-
are or have been smokers. tors that are causally related to COPD [2]. The relation-
The clinician can be guided by information in the clin- ship between asthma and smoking is complex. Individuals
ical history such as smoking history, age of onset of symp- with asthma may be non-smokers, smokers, or ex-smokers.
toms, history of atopic conditions, and description of acute Since asthma genes and genes leading to the susceptibility
episodes of shortness of breath (see Table 1.5). to develop airow obstruction with smoking are common in
Asthma usually has its onset in early childhood. the population, the likelihood that an individual may have
However, adult-onset asthma does exist, and many are una- both is high. Likewise, COPD may occur in lifetime non-
ble to remember childhood events that would provide a clue smokers [1315]. In some, this may be longstanding asthma
to the early stages of asthma. Therefore, unless symptoms that may have been undiagnosed. In others, genes that are
are continuous from childhood, the onset of asthma symp- as yet unrecognized may be responsible. Alpha-1 antitrypsin
toms in adult life may be hard to interpret, especially in the deciency is the model for such a genotype.
presence of other risk factors such as smoking. COPD typi- Pulmonary function tests can also provide guidance.
cally becomes clinically apparent in the sixth and seventh Both diseases are characterized by airow obstruction except
decades of life. If an individual is physically active, he or she in the early or mild stages. In asthma, lung function either
may notice reduced exercise tolerance earlier. remains normal or can be normalized with treatment in
COPD in developed countries is mostly a disease of patients with mild intermittent or mild persistent disease [1].
smokers. This is not necessarily true in developing countries COPD, in comparison with asthma, is dened by irreversible

6
Definitions 1
airow limitation, and this becomes progressively greater
as the disease advances. Lung function in asthma is char- LIMITATIONS OF THE DEFINITIONS
acterized by reversibility and variability. Reversibility refers
to the short-term response to an inhaled bronchodilating Denitions for both asthma and COPD have limitations since
agent, and is preferably measured with spirometry or peak they can reect only our current understanding of the diseases,
ow (acceptable but not as helpful as spirometry). Variability which is quite limited. Both diseases will continue to be rede-
refers to the improvement and deterioration in symptoms ned as our understanding of them deepens, and as new eec-
and lung function over time (both short- and long-time tive preventive strategies and treatments become available.
periods) that is characteristic of asthma. The GINA guide-
lines [1] recommend that reversibility be dened as a 20%
improvement post-bronchodilator in the 1-second forced References
expiratory volume (FEV1) or peak ow (PEF). GINA fur-
ther denes variability as a diurnal variation in PEF of more 1. Global Strategy for the Diagnosis, Management, and Prevention of chronic
than 20% (with twice daily reading, more than 10%). Obstructive Pulmonary Disease. Global Initiative for Chronic Obstructive
Lung Disease (GOLD), 2006; Available from: URL: www.goldcopd.org
The GOLD COPD guidelines [2] recommend that
2. Global Strategy for Asthma Management and Prevention. Global
spirometry be used to establish that the airow limitation is Initiative for Asthma (GOLD), 2006; Available from: URL: www.
not fully reversible (dened as a post-bronchodilator ginasthma.org
FEV1/FVC ratio of 0.7 and FEV1  80% predicted), 3. National Asthma Education and Prevention Program Expert Panel
and to stage the severity of the disease. However, GOLD Report 2: Guidelines for the Diagnosis and Management of Asthma,
emphasizes that bronchodilator reversibility testing does National Institute of Health, National Heart, Lung, and Blood
not predict a patients response to treatment or predict dis- Institute. NIH Publication 974051, 1997.
ease progression. 4. Wenzel SE. Asthma: Dening of the persistent adult phenotypes.
Lancet 368: 80413, 2006.
5. A plea to abandon asthma as a disease concept. Editorial, Lancet
368:705, 2006.
6. Pauwels RA, Buist AS, Calverley MA, Jenkins CR, Hurd SS. Global
OVERLAP BETWEEN ASTHMA AND COPD strategy for the diagnosis, management and prevention of chronic
obstructive pulmonary disease. NHLBI/WHO Global Initiative for
Chronic Obstructive Lung Disease (GOLD) Workshop Summary.
Not acknowledged in the denitions is the fact that long- Am J Respir Crit Care Med 163: 125676, 2001.
standing asthma can lead to airway remodeling and partly 7. Celli BR, MacNee W and committee members, Standards for the
irreversible airow obstruction [16]. So, in many (but not diagnosis and treatment of patients with COPD: A summary of the
all) with longstanding asthma, there is an appreciable com- ATS/ERS position paper. Eur Respir J 23: 93246, 2004.
ponent of chronic irreversible airow obstruction with 8. National Institute for Clinical Excellence (NICE). Chronic obstruc-
reduced lung function and incomplete response (or at least, tive pulmonary disease. National clinical guideline on management of
chronic obstructive pulmonary disease in adults in primary and second-
not complete reversibility) to a short-acting bronchodilator
ary care. Thorax 59(Suppl 1): 1232, 2004.
or to oral or inhaled corticosteroids [17]. This complicates 9. National Institute for Clinical Excellence (NICE) Guideline available
the diagnosis of asthma in older adults, and requires that at URL: www.nice.org.uk/CG012niceguideline
the goals of treatment be modied since maintenance of 10. Soriano JB, Visick GT, Muellerova H, Payvandi N, Hansell AL.
normal lung function can no longer be a realistic goal. Patterns of comorbidities in newly diagnosed COPD and asthma in
Whether longstanding asthma with remodeling can primary care. Chest 128(4): 2099107, 2005.
be called COPD is intensely controversial. In so far as there 11. Agusti AG. Systemic eects of chronic obstructive pulmonary disease.
is irreversible or poorly reversible airow obstruction in the Proc Am Thorac Soc 2(4): 36770, 2005.
remodeled lungs, the term seems appropriate. Conceptually 12. Dodge R, Cline MG, Burrows. B. Comparisons of asthma, emphy-
and practically, the recognition that remodeling is a feature sema, and chronic bronchitis diagnoses in a general population sample.
Am Rev Respir Dis 133: 98186, 1986.
of longstanding asthma in many (but not all) reinforces the
13. Celli BR, Halbert RJ, Nordyke RJ, Schan. B. Airway obstruction in
notion that these diseases constitute a spectrum of disease, never smokers: Results from the Third National Health and Nutrition
as illustrated in Fig. 1.1, ranging from fully reversible to Examination Survey. Am J Med 118: 136472, 2005.
fully irreversible. 14. Menezes AM, Perez-Padilla R, Jardim JR, Muino A, Lopez MV,
Valdivia G et al. Chronic obstructive pulmonary disease in ve Latin
American cities (the PLATINO study): A prevalence study. Lancet
366(9500): 187581, 2005.
EXACERBATIONS 15. Buist AS, McBurnie MA, Vollmer WM, Gillespie S, Burney P,
Mannino DM, Menezes AMB, Sullivan SD, Lee TA, Weiss KB, Jensen
RL, Marks GB, Gulsvik A, Nizankowska-Mogilnicka E. International
The denition of asthma highlights the importance of exac- Variation in the prevalence of chronic obstructive pulmonary disease
(The BOLD Study): a population-based prevalence study. Lancet 2007;
erbations as a feature of asthma, and emphasizes the uctua-
370:74150.
tions of the disease [1, 3]. The denition of COPD does not 16. James AL, Wenzel SE. Clinical relevance of airway remodelling in
include any mention of exacerbations [2, 68]. Nevertheless, airway diseases. Number 4 in Series Airway Remodelling: from Basic
they may be as important in the natural history of COPD as Science to Clinical Practice. Eur Respir J 30: 13455, 2007.
they are in asthma [1] and account for approximately 70% 17. Fish JE, Peters SP. Airway remodeling and persistent airway obstruc-
of the COPD-related costs in the United States [2]. tion in asthma. J Allergy Clin Immunol 104: 50916, 1999.

7
Epidemiology
2
CHAPTER

DEFINING THE DISEASES However, this denition is non-specic


Dawn L. DeMeo and
and does not provide absolute criteria to facili-
tate dierentiating asthma from other airway Scott T. Weiss
Asthma diseases [5].
Beyond denitions, there are dierences Channing Laboratory, Brigham and
The study of asthma epidemiology has been between languages for the words used to describe Womens Hospital, Harvard Medical
plagued by lack of consensus regarding standards asthma symptoms. A novel approach to this School, Boston, MA, USA
for diagnosis. Most denitions have included var- problem has been used in the International Study
iable airow obstruction, but asthma is a clinical of Asthma and Allergies in Children (ISAAC),
syndrome, without a widely accepted case deni- which includes an asthma video questionnaire
tion. Epidemiology studies have used question- demonstrating clinical signs of asthma, as an
naires to assess for the presence of disease, but are attempt to improve uniformity in surveying for
limited by recall and misclassication bias. Some asthma [6]. This multi-modality approach rep-
have suggested that symptoms should be assessed resents an improvement in asthma surveillance
in conjunction with airway hyperresponsiveness and has captured more individuals with asthma.
[1]. Others argue that airway hyperresponsive- However, diagnostic heterogeneity remains, as
ness and symptoms should be analyzed separately even within the ISAAC project, prevalence esti-
owing to the poor correlation between clinical mates of current asthma may vary depending
asthma and hyperresponsiveness [2]. Population- upon the assessment tool used (i.e. video versus
based epidemiology studies have demonstrated a questionnaire).
low sensitivity of airway hyperresponsiveness for
detecting individuals with physician-diagnosed
asthma versus a sensitivity of greater than 90% in Chronic obstructive pulmonary
clinic studies [3]. A standard denition of asthma
is as a chronic inammatory disease of the air-
disease
ways. A commonly referenced formal deni-
COPD includes chronic bronchitis, small airways
tion is that promulgated in the Global Initiative
disease and emphysema, and is characterized by
for Asthma (GINA) guideline, which denes
airway obstruction that is xed or only partially
asthma as
reversible. The diagnosis of COPD attached to
a chronic inammatory disorder of the airways a given patient varies depending on the degree
in which many cells and cellular elements play a of airow obstruction considered diagnostic
role. The chronic inammation causes an associ- within a given set of guidelines. Thus the long-
ated increase in airway hyper-responsiveness standing lack of international standardization of
that leads to recurrent episodes of wheezing, criteria for diagnosis of COPD makes under-
breathlessness, chest tightness and coughing, standing relative incidence and prevalence quite
particularly at night or in the early morning. challenging. This is well illustrated by a study by
These episodes are usually associated with wide- Lindberg et al. [7], who compared COPD prev-
spread but variable airow obstruction that is alence rates using European Respiratory Society
often reversible either spontaneously or with (ERS), British Thoracic Society (BTS), Global
treatment [4]. Initiative for Chronic Obstructive Lung Disease

9
Asthma and COPD: Basic Mechanisms and Clinical Management

(GOLD), and clinical and spirometric American Thoracic Global burden of disease estimates suggest that at least
Society (ATS) criteria. In this study, ERS criteria revealed a 300 million people worldwide have asthma, with the poten-
14.0% prevalence of COPD, BTS criteria 7.6% prevalence, tial for an additional 100 million more cases by the year 2025
GOLD criteria 14.1% prevalence, clinical ATS criteria [15]. In childhood, incidence rates for asthma are highest
12.2% prevalence, and spirometric ATS criteria 34.1%. among the youngest age groups and among male children
This example highlights the diculty of comparison until puberty [1621]. In a study of an adult Swedish popu-
between international studies/standards and the eorts to lation, Toren and Hermansson [22] found the incidence
understand COPD epidemiology on a global scale. Similar rate for adult-onset asthma to be highest among females of
prevalence dierences depending upon reference criteria all ages greater than 20, with an incidence of 1.3 per 1000
have been observed by other authors [8, 9], further highlight- person-years; among women 1620 years of age the rate was
ing the challenges inherent in making comparisons between 3 per 1000 person-years. Analysis of data from a prospec-
populations. tive cohort study in Finland demonstrated no increase in
Recently, an international committee convened to asthma incidence from 1982 to 1990 in adults aged 1845
rene and promote standardization of a COPD denition. years [23]. Early investigation into the increasing preva-
The GOLD has dened COPD as lence of asthma in the United States was noted in a review
of medical records from Olmsted County, Minnesota, where
a preventable and treatable disease with some signicant
the annual incidence of asthma was found to increase from
extra pulmonary eects that may contribute to the severity
183 per 100,000 in 1964 to 284 per 100,000 in 1983. The
in individual patients. Its pulmonary component is char-
most signicant increase was in children aged 114 years,
acterized by airow limitation that is not fully reversible.
suggesting a potential cohort eect early in life. Despite this
The airow limitation is usually progressive and associ-
increased incidence in asthma among children from 1964 to
ated with an abnormal inammatory response of the lungs
1983, constant rates were observed among adults [16].
to noxious particles or gases [10].
Although these data indicate that asthma incidence
GOLD stages are dened using post-bronchodilator has been increasing, minimal information has been available
spirometry, with an FEV1/FVC ratio of less than 0.7 den- for trends in COPD incidence. One challenge of studying
ing the presence of obstruction, and the FEV1 reduction COPD incidence is that the disease is usually moderately
translating into the severity of COPD. A key element of advanced at diagnosis, and thus true incidence rates have
this updated denition is that it includes capturing features remained elusive. However, a recent study observed that cough
of extra-pulmonary co-morbidities which have relevance and phlegm may identify a group at high risk for incident
for disease severity [11]. The GOLD initiative has aimed to COPD [24]. This study by de Marco and colleagues focused
promote studies to understand the increasing prevalence of on a cohort of 5002 subjects without asthma and with nor-
COPD worldwide, as well as to standardize the collection mal lung function. The observed incidence rate of COPD was
of data for international comparison of research [12]. 2.8 cases/1000/year; the incidence rate ratio for chronic cough
In summary, for many years both asthma and COPD and phlegm was 1.85 (95% CI 1.172.93) with a 10-year
have lacked widely accepted case denitions, but international cumulative incidence of COPD in the overall cohort of 2.8%.
eorts have been developed to rectify this. Denitions of both Two other studies have reported on incidence rates of COPD
diseases include both symptoms and spirometric features. using GOLD criteria, with a 10-year cumulative estimate of
Rening the standardization of clinical diagnosis will facili- 13.5% noted by Lindberg et al. [25], and a 9-year cumulative
tate comparability in epidemiological studies. However, given incidence of 6.1% in a study by Johannessen et al. [26].
the wide variability in disease denitions for both diseases,
comparisons of studies of asthma and COPD between popu-
lations and between countries must be viewed in the light of
dierences in criteria used for disease diagnosis. Although
the GINA and GOLD guidelines have brought some stand-
PREVALENCE
ardization and consensus to diagnosing asthma and COPD
respectively, diagnostic heterogeneity remains and further Trends in the prevalence of obstructive lung disease have
renement of these guidelines is an important goal. been suggested by an analysis of the Third National Health
and Nutrition Examination Survey (NHANES III) [27].
This survey included subjects with asthma, chronic bron-
chitis, and emphysema (Fig. 2.1). In this cohort, the case
INCIDENCE denitions were a physician diagnosis of chronic bronchitis,
asthma or emphysema, respiratory symptoms, and low lung
function. Of note, the investigators dened low lung func-
Incidence rates for asthma and COPD vary depending on tion as present when the FEV1/FVC ratio was 0.70 and
the age of the population under consideration. For exam- the FEV1 was less than 80% of predicted. Of the investigated
ple, asthma is commonly diagnosed in early childhood and population of 20,050 adults, 6.8% had low lung function as
COPD is commonly diagnosed after age 60. Interestingly, thus dened; 7.2% of the population had an FEV1/FVC
cases of adult-onset asthma and early-onset COPD pre- ratio less than 0.70 with an FEV1 greater than 80% pre-
dominate in women, suggesting that there may be a common dicted, and were not included as having low lung function.
hormonal inuence on the age-of-onset of these diseases Of the entire population, 8.5% reported obstructive lung
[13, 14]. disease. Importantly, 63.3% of those with documented low

10
Epidemiology 2
25
Current COPD and asthma Current COPD
Current asthma Past chronic bronchitis or asthma

20
People with indicated condition (%)

15

10

5
FIG. 2.1 Age-specific percentage of individuals,
stratified by race and sex, with COPD and
asthma, current COPD, current asthma, and
past chronic bronchitis or asthma. Reproduced
0 from National Center for Health Statistics, Plan
and Operation of the Third National Health and
1724
2544
4564
6574
7584
85

1724
2544
4564
6574
7584
85

1724
2544
4564
6574
7584
85

1724
2544
4564
6574
7584
85
Age Nutrition Examination Survey, 19881994, US
Department of Health and Human Services
Black female Black male White female White male publication PHS 941308, 1994, with permission.

lung function had no current or prior doctor diagnosis of and the main source of prevalence data in adults includ-
obstructive lung disease. In addition to prevalence informa- ing the European Community Respiratory Health Survey
tion regarding low lung function, NHANES III data suggest (ECRHS). Higher prevalences of asthma (10%) have been
that there is still a signicant proportion of disease that goes noted in developed countries including Canada, the United
undiagnosed in the mild stages, leading to an underestima- States, United Kingdom, New Zealand, and The Republic
tion of the true prevalence of obstructive lung disease. of Ireland [15]. In parallel with urbanization, the rates of
asthma have increased in developing countries as well.
The ISAAC has as its aim to describe, across 155
Asthma centers, the prevalence and severity of asthma in children
in 56 countries [6]. Phase 1 of this trial has demonstrated
Data from the United States have suggested an increase in a large variation in the prevalence of asthma symptoms in
prevalence of asthma in children as well as in older adults children throughout the world, with the highest preva-
(Fig. 2.2) [28]. During the last several decades studies have lence in centers from Australia, New Zealand, the United
suggested an increase in prevalence worldwide of 56% Kingdom, and Ireland [3235]. While the prevalence of
per year. Data from the National Health Interview Survey allergic rhinitis has been noted to be scattered in the groups
(NHIS) reveal a 75% increase in self-reported asthma rates with the highest prevalence of asthma, the lowest preva-
from 1980 to 1994. This trend was demonstrated in all age lence for rhinitis has been found in countries where the
and race strata as well as in both genders. The most signi- asthma prevalence was lowest, such as in Eastern Europe,
cant increase demonstrated by the NHIS was among chil- Indonesia, Greece, and India. In addition to dening preva-
dren 04 years of age (increase of 160%) and persons 514 lence rates, the ISAAC study represents an eort to estab-
years of age (increase of 74%) [29]. lish an international standard to facilitate comparability of
Repeatedly, the prevalence of asthma amongst inner- data from epidemiological studies of asthma. The rising
city children has been demonstrated to be much higher asthma prevalence rates in children heralds rising asthma
than among similar aged children living outside the inner rates for adults, as two-thirds of children with asthma still
city [2931]. It has been suggested that a doctors diagno- have symptoms by early adulthood [36].
sis of asthma is made less frequently than asthma symptom
reporting, raising concern that despite increasing prevalence
there is still a tendency to under-diagnose asthma, and con- Chronic obstructive pulmonary disease
sequently underestimate true prevalence values [32].
The GINA project [15] has compiled asthma preva- COPD is best understood by understanding rst the trends
lence data for 20 regions of the world, with the main for smoking in populations. Although projected smoking rates
sources of data in children including the ISAAC project, throughout the world have increased, smoking prevalence in

11
Asthma and COPD: Basic Mechanisms and Clinical Management

Australia Australia
40 United States 40 United States
Canada Switzerland
Switzerland Germany
Germany The Netherlands
35 35
United Kingdom United Kingdom
Norway Sweden
Finland Norway
30 Estonia 30 Estonia

Prevalence of asthma symptoms (%)


Poland Italy
Prevalence of asthma (%)

Italy Spain
25 Spain 25 Singapore
Israel Hong Kong
Singapore Korea
20 Hong Kong 20
Taiwan
Korea
15 15

10 10

5 5

0 0
1965 1970 1975 1980 1985 1990 1995 2000 2005 1965 1970 1975 1980 1985 1990 1995 2000 2005

FIG. 2.2 Changes in the prevalence of asthma and asthma symptoms in children and young adults. Reproduced from Ref. [28], with permission.

the United States has been decreasing. Estimates from the 14.1 million with chronic bronchitis and 1.8 with emphy-
year 2005 suggest that 21% of Americans smoke cigarettes, sema. By these measures, there was no change in the preva-
with cigarette smoking prevalence rates varying by both sex lence of emphysema from 1982 to 1996, although from 1983
and race. Recent prevalence estimates [37] for smoking were to 1995 the prevalence of chronic bronchitis continued to
highest for American Indian/Alaska Natives (37.5% men and increase. In a study of the Canadian population, prevalence
26.8% women). The prevalence of current smoking is 20.0% rates of COPD were 4.6% in the 5564 age group, 5.0% in
in white women, 17.3% in African American women, 24.0% the 6574 age group, and 6.8% in the greater than 75 age
in white men, and 26.7% in African American men. group [41]. These data may be an underestimation, as there
However, in the context of COPD, susceptibility is a suggestion that COPD prevalence rates are underesti-
to cigarette smoke is not uniform and varies as a function mated in the elderly, especially in those with lower incomes
of the amount of cigarette smoking [38]. Stang et al. [39] [42]. By 2001, in distinction to the many asthma prevalence
utilized smoking rates to create a mathematical model for studies, only 32 prevalence surveys of COPD were reported
estimating COPD prevalence. Using their model, they esti- (Fig. 2.3) [9] with comparability somewhat limited by
mated that 15.3 million people in the United States aged COPD case denitions. The Burden of Obstructive Lung
40 years or more have COPD; this was a reasonable esti- Disease (BOLD) project is a large-scale study of COPD,
mate compared to the spirometric prevalence of 17.1 mil- with one goal of measuring COPD prevalence using stand-
lion as estimated by the NHANES III. Using this model, ard GOLD criteria. A recent report from the study in 1258
they also predicted the prevalence of COPD in Germany subjects reported a 26.1% prevalence of at least GOLD
(2.7 million), the United Kingdom (3.0 million), Spain (1.5 Stage 1 COPD, and 10.7% for GOLD Stage 2 or higher
million), Italy (2.6 million), and France (2.6 million), and [44]. Pooled estimates based on GOLD criteria have sug-
suggested smoking rates as a useful surrogate for estimating gested a prevalence of GOLD Stage 1 or higher COPD of
COPD prevalence. 5.5% and 9.8% for GOLD Stage 2 or higher [9].
The World Health Organization prediction is that by COPD is likely under-diagnosed in both North
2020 COPD will rise from being the twelfth to the fth American and European populations. For example, the
most prevalent disease worldwide, and from being the sixth IBERPOC Project (Estudio Epidemiologico de la EPOC
most common cause of death to the third most common en Espaa) was a population-based study of prevalence of
[40]. Prevalence estimates of COPD in the United States in COPD in Spain [45]. The prevalence of COPD in this
1996, before widespread implementation of GOLD criteria, population (26% current smokers, 24% ex-smokers, 76%
indicated approximately 15 million people have COPD with men), dened according to ERS criteria, was 9.1%. Only

12
Epidemiology 2
Prevalence of COPD States; the utilization increase has been concomitant with the
20 documented increase in asthma prevalence [4648].
Increased hospital visits have been documented
Prevalence per 100

15 worldwide, including in England, New Zealand, the United


States, Greece, Australia, and Canada [4956]. In capturing
10 the epidemiology trend from the 1960s to the 1980s, Evans
et al. reported on a 200% increase in rates of hospitaliza-
5
tion of adults with asthma (and a 50% increase for children
with asthma) [57]. Overall, hospitalizations with asthma
as a primary diagnosis increased most steeply in the 1970s
0
25 35 45 55 65 75 85
until the mid-1980s and then remained constant; this is in
contrast to asthma as a secondary diagnosis which increased
Midpoint of age-range of population studied
until 1997. From 1975 to 1995, oce visits for asthma more
Opinion Diagnosis Symptoms Spirometry than doubled, from 4.6 to 10.4 million [29].
Based on the National Ambulatory Medical Care
FIG. 2.3 Prevalence of COPD using estimates from expert opinion, Survey and the National Hospital Ambulatory Medical Care
physician diagnosis symptoms and spirometry. Data from Ref. [9] as Survey, in 1998 more than 14 million visits were made to
reproduced in Ref. [43], with permission. physicians for diagnoses related to COPD, and this was an
increase from 5.5 million visits reported in 1980. In 2000,
there were approximately 726,000 hospitalizations coded
22% of those diagnosed had a prior diagnosis, while 48% as COPD or allied conditions [58]. Care must be taken in
had prior respiratory symptoms. More recently, despite interpretation of these data because of non-uniform case
prevalence estimates for GOLD Stage 1 or higher COPD denitions; more than half of the discharge diagnoses were
in the BOLD study in Salzburg, only 5.6% of subjects non-specically coded as COPD or allied conditions.
reported ever receiving a doctor diagnosis of COPD [44]; In summary, increased health service utilization has
the prevalence has yet to translate into widespread use of occurred for asthma and COPD during the last two decades.
spirometry for COPD screening. Overall, hospital admissions and discharges have increased
The high-prevalence estimates for both asthma and for both diseases.
COPD have translated into signicant morbidity associ-
ated with both diseases. In 2001, asthma was ranked as the
25th leading contributor to disability adjusted life years
(DALYs) worldwide; COPD was ranked 12th. In this con-
text, together with the high-prevalence estimates for both
MORBIDITY AND MORTALITY
disease, the global burden of disease and economic costs for
both diseases are high. Asthma
In summary, the prevalences of both asthma and
COPD are increasing in both Western developed and devel- The New Zealand epidemic of asthma in the 1970s prompted
oping countries. As both asthma and COPD are both likely a review of asthma deaths in Western countries; among such
under-diagnosed, the prevalence estimates underestimate the countries there was a notable increase of 1.5- to 2-fold in the
true burden of these diseases. Variability in denitions of both asthma mortality rates between the mid-1970s and the mid-
asthma and COPD contribute to inexact prevalence estimates 1980s [59]. The highest mortality rates in the United States
and problems with comparisons of prevalence data. ISAAC have been in the inner-city regions, with particularly high-
(for childhood asthma), ECRHS (for adult asthma), and risk populations studied in East Harlem, New York City,
GOLD (for COPD) represent international eorts under- and Cook County, Chicago [31, 60]. One study observed
way to standardize the denitions used in studies to enhance that socioeconomic and racial disparities were attributable to
comparisons of incidence, prevalence, and burden of disease. higher incidence of asthma exacerbations among inner-city
children, with no excess utilization of medical resources [61].
International comparisons of mortality rates have
been limited by dierences in recording statistics of cause of
HEALTHCARE UTILIZATION AND death [62]. There is variability in the mortality rates ascribed
HOSPITALIZATION TRENDS to asthma, with signicant variation by region of the world
(Fig. 2.4), with the highest asthma case fatality rates in
China (36.7/100,000 asthmatics) [15]. Mortality rates for
In the United States, the estimated cost for year 2000 for asthma have decreased over the past 20 years, likely due to
asthma was projected to be 12.7 billion dollars (8.1 for direct improvements in pharmacotherapy, although it has been sug-
cost, 2.6 related to morbidity, 2.0 related to mortality) and 30.4 gested that most deaths from asthma now occur in adults,
billion dollars for COPD (14.7 for direct cost, 6.5 related to potentially consequent to medical non-compliance [63].
morbidity, 9.2 related to mortality). Utilization of health serv- Comparison of mortality rates is dicult also because of the
ices has continued to increase for both diseases. An increase lack of standardized denitions for the disease, and because
in health service use has been documented in many countries, of environmental, genetic, socioeconomic, and occupational
including the United Kingdom, Canada, and the United inuences unique to a given population.

13
Asthma and COPD: Basic Mechanisms and Clinical Management

Countries shaded according to case fatality rate (per 100,000 asthmatics)

10.0 05.0 5.110.0 No standardized data available

FIG. 2.4 Asthma case fatality rate in individuals 534 years of age calculated as the number of deaths per 100,000 cases of asthma. Reproduced from Ref. [15],
with permission.

Rate/100,000 population Rate/100,000 population


40 30 20 10 0 0 10 20 30 40 50 60 70 80
Hungary (95)
Ireland (95)
Romania (96)
Scotland (97)
New Zealand (94)
United Kingdom (97)
England/Wales (97)
United States (97)
Northern Ireland (97)
Australia (95)
Spain (95)
Poland (96)
The Netherlands (95)
Germany (97)
Bulgaria (94)
Portugal (96)
Norway (95)
Canada (95)
Austria (97)
Italy (93)
France (95)
Sweden (96)
Israel (95)
Japan (94) Women Men
Greece (96)

FIG. 2.5 Age-adjusted death rates for COPD by country and sex, for individuals aged 3577. The year of data is shown in parentheses. Reproduced from Ref.
[12], with permission.

Chronic obstructive pulmonary disease [64]. In longitudinal follow-up of 13,756 individuals in the
Atherosclerosis Risk in Communities (ARIC) study, rapid
Since 1960 mortality associated with COPD has continued lung function decline over a 3-year period was associated
to rise. In the year 2000, the number of women who died with an increased risk for COPD-related hospitalization
from COPD surpassed the number of men [58], and stand- and death [65].
ardized mortality rates have been observed to be higher in International mortality trends demonstrate high rates
women versus men with COPD (4.8 versus 2.7 respectively) of deaths for COPD in many countries. These dierences
14
Epidemiology 2
may be accounted for in part by dierent smoking behav- 8% reduction in FEV1. The Vlagtwedde/Vlaardingen study
iors, which include tobacco type, along with other environ- [68] demonstrated a large eect of cigarette smoking in
mental, infectious, and genetic factors. Dierences among decreasing maximal lung function in individuals less than
these death rates are striking (Fig. 2.5), but again lack of age 20; this eect exceeded the eect of cigarette smoking
standardization in coding practices and death certication on lung function decline seen in older subjects.
as well as practice dierences and quality of care are rele- Smoking is a notable risk factor for both asthma and
vant when comparing estimates [12]. COPD in children and adults, and active smoking perpetu-
Although overall asthma mortality remains low com- ates pulmonary (and likely systemic) inammation. Overall,
pared with COPD, mortality rates for both asthma and smoking is associated with an increase in asthma incidence
COPD have increased in the last decade. Dierences in [69, 70], as well as an increased symptom severity, increased
death rates for asthma and COPD between countries are morbidity and lung function decline, and an increased
multifactorial (genetic, environmental, occupational, socioe- risk of death [7174]. Passive exposure to cigarette smoke
conomic), but dierential coding of cause-of-death statistics increases the risk for the development of asthma and allergic
hinders accuracy of estimates for both diseases. sensitization [7577]. There has also been a suggestion that
non-specic airways responsiveness is increased by environ-
mental and personal smoke exposure [78].
SMOKING Maternal smoking is a risk factor for the development
of asthma in children up to 1 year of age [79]. In a case-
control study of children whose mothers were heavy smok-
Burrows and colleagues [38] have demonstrated that, ers, one group demonstrated an odds ratio of 2.15 among
for a given level of tobacco smoke exposure, FEV1 varies 34-year olds for the development of asthma; these data
substantially (Fig. 2.6). In addition, the doseeect relation- were controlled for family history, past infections, gender,
ship between cigarette smoking and FEV1 decline depends and other demographic variables [80]. In a 6-year follow-up,
on the year of life when an individual is exposed. Dose and the odds ratio for asthma among those exposed to maternal
timing of tobacco smoke exposure have a dierential eect smoking was 3.8 [81].
on FEV1 depending on the stage of the life cycle (Table 2.1). As noted above, the single most signicant risk fac-
Cunningham et al. [66] observed that maternal smoking tor for COPD is tobacco smoking. Although an oft quoted
during pregnancy resulted in a 1.3% reduction in FEV1 when estimate is that only 1015% of smokers actually go on to
children were 812 years old. Tager et al. [67] found that develop obstructive lung disease, this is a marked underes-
adolescents who smoke when aged 1520 have an estimated timate, with many researchers suggesting that all individuals

0 pack-years 1 S.D. Mean 1 S.D.


20
10
Median
0
020 pack years
20
10
0
% of Population

2140 pack years


20
10
0
4160 pack years
20
10
0
61 pack years
20
10
0

40 60 80 100 120 140 160


% FEV1

FIG. 2.6 Distribution of percentage predicted forced expiratory volume in 1 s (FEV1) in adults with varying smoking histories as measured in pack-years. The
proportion of smokers with normal flow decreased with increasing pack-year histories. Yet, many have near-normal FEV1 with extensive smoking history.
Subjects with respiratory trouble before age 16 were excluded. Medians and means 1 SD are shown for each group in the abscissae. Note that among the
425 persons with 20 pack-years, only 15% have an FEV1 of 60% of predicted or less. Reproduced from Ref. [38], with permission.

15
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 2.1 Effects of cigarette smoking at different stages of the life cycle. immunoglobulin by sensitized lymphocytes. Elevations in
specic IgE and/or total IgE, total eosinophil counts, and
FEV1 reduction skin test reactivity to specic allergens have been used clini-
Life phase Total FEV1 (ml/year per cally to detect allergic individuals. As measured by skin test
(gender) Cigarette dose reduction packs/day) reactivity, allergy increases with age until about age 15, at
which point it is maximal. The decline in skin test reactivity
In utero Variable exposure 27.3 mla 36
after age 35 confounds the measurement of this phenotype
(M and F) for 9 months
in older individuals. This reported association between skin
Adolescence 15 cigarettes/day 390 ml 104 test reactivity and decline in FEV1 is not consistent in the
(M) for 5 yearsb literature. In retrospective studies, Taylor et al. [87] and Frew
et al. [88] demonstrated no relation of skin test positivity to
Adolescence 10 cigarettes/day 340 ml 136 decline in FEV1. However, Gottlieb et al. [89] investigated
(F) for 5 yearsb this prospectively in the Normative Aging Study and found
Adult (M) Variable N/A 13c
that skin test positivity predicted increased annual rates of
decline in both FEV1 and FEV1/FVC ratio.
Adult (F) Variable N/A 7c Allergic inammation is characteristic of asthma;
8090% of childhood asthmatics are atopic, and the degree
Source: Adapted from Weiss ST, Silverman EK. Risk factors for the development of chronic
of atopy appears to be associated with prognosis in child-
obstructive pulmonary disease. In: Severe Asthma, New York: Marcel Dekker, 2000.
hood asthma [90]. Studies have demonstrated that the asth-
Notes: M: male; F: female.
a
Adjusted for gender and maternal smoking in the past year; based on 1.3% reduction
matic phenotype is associated with elevated serum IgE levels
and mean FEV1 2.1 l, 1 pack/day in smoking mothers during pregnancy is assumed more so than skin test positivity [91], and that increased air-
for relative FEV1 reduction (Ref. [65]). ways responsiveness is related to total serum IgE levels [92].
b
Median values for cigarette smoking (Ref. [66]). Weiss has suggested that immediate type I hyper-
c
Estimated values (Ref. [67]). sensitivity is a risk factor for the development of chronic
obstructive lung disease, and suggests that atopy may inu-
ence childhood asthma and limit maximal lung function,
accelerate FEV1 decline, and potentially enhance interaction
who smoke will go on to develop COPD. Among those with cigarette smoking to result in FEV1 decline progressing
smokers already with a decreased FEV1, lung injury and sub- to the development of COPD [93]. Hargreave and Leigh
sequent decrements in lung function secondary to cigarette [94] demonstrated, in a subset of COPD patients, that eosi-
smoking are more dramatic. In the Lung Health Study, nophilic inammation is important in COPD exacerbations,
middle-aged smokers (with FEV1 between 55% and 90%) and potentially leads to a decline in lung function. As well,
who continued smoking for 5 years had further losses of an inverse association of serum IgE with the FEV1/FVC
several hundred milliliters of FEV1 [82]. However, COPD ratio has been observed [95]. These data indicate that aller-
has been identied in non-smokers as well. Prevalence has gen sensitization may represent an intermediate phenotype
been noted to increase with age, to be higher in women which needs to be considered in understanding disease onset
than in men, to be particularly high in Hispanic individuals, and progression in both asthma and COPD.
and to be higher in low income versus auent individuals
[83]. Recent data from NHANES III suggests that 25% of
cases of COPD in the United States occurs in lifelong non- Airways responsiveness
smokers [84], a nding that is echoed by estimates of 23%
in the United Kingdom [85] and 23% in Spain [86]. These Airways responsiveness to methacholine and histamine has
observations reinforce that although cigarette smoking is the been used in population-based studies to help dene indi-
most important risk factor for COPD, it is not necessary for viduals susceptible to the development of obstructive lung
the development of irreversible airow obstruction. disease. This intermediate phenotype is a feature of both
In summary, smoking has a lifetime inuence on asthma and a subset of patients with COPD. Baseline levels
asthma and COPD. This starts in utero and continues into of lung function, allergy, age, and cigarette smoking history
older age. Smoking is associated with enhanced airway all inuence airways responsiveness.
responsiveness, both in asthma and COPD. Smoking is suf- Airways hyperresponsiveness has been demonstrated to
cient but not necessary for the development of COPD. predict accelerated decline in lung function and the develop-
Smoking is only one of several risk factors for asthma. ment of COPD [96]. More recently, airways responsiveness
has been demonstrated to predict COPD mortality [97].
Airways responsiveness has been demonstrated to pre-
dict the development of asthma [98]. The prevalence of air-
INTERMEDIATE PHENOTYPES way hyperresponsiveness exceeds the prevalence of asthma;
the former is about 20% in the general population. Data
from the Childhood Respiratory Disease Study demonstrate
Allergy that increased airway responsiveness predicts the develop-
ment of asthma in children and young adults with a 2- to 3-
Allergy represents immediate hypersensitivity to an antigen fold risk [99]. Some have observed this risk to be increased
and is associated with an increased production of a specic as much as 5-fold [100].

16
Epidemiology 2
Airways hyperresponsiveness in COPD patients may more susceptible to develop severe early-onset COPD [14],
be demonstrated in more than two-thirds of individuals in have worse quality of life [114], and higher mortality from
situations where it is actually measured [101]. Some individ- COPD [115] than men. These gender/sex dierences most
uals who develop COPD have an allergic asthma phenotype, likely represent inuences of both dose of tobacco exposure
as suggested by the Dutch hypothesis [102]. Alternatively and underlying genetic and hormonal susceptibilities, and is
the hyperresponsiveness may be a consequence of COPD. a topic of active research.
Results from a 25-year longitudinal study in the Netherlands In summary, in the adult years women have a higher
revealed that increased airways responsiveness is an inde- prevalence of asthma. The prevalence of COPD in women
pendent risk factor for FEV1 decline [103]. Among individu- is increasing, in keeping with the historical trends of ciga-
als with early-onset COPD, the degree of baseline airways rette smoking and disease. The gender and sex dierences
responsiveness determined the response to cigarette smoking; between asthma and COPD highlight the importance of
those with early-onset COPD who have increased airways hormonal and genetic inuences relevant to disease expres-
responsiveness appeared more sensitive to the eects of ciga- sion in men versus women.
rette smoke and had an accelerated decline in FEV1 [104].
Thus, airway hyperresponsiveness, together with features of
allergy and smoking, likely represent common risk factors for
both asthma and COPD. DEMOGRAPHICS

Gender/sex-related influences In the United States, morbidity from asthma has been
demonstrated in multiple studies to be greater in children of
The impact of gender and sex-biology-related features on African American descent. In the United States, physician-
airway disease are complex, and include both social para- diagnosed asthma has been reported in 13.4% of African
digms (gender-related) as well as hormonal and genetic (sex- American children and 9.7% of white children [116].
related) inuences. African American children have also been reported to have
The epidemiology of asthma is characterized by age- greater limitation on activity due to asthma, with more hos-
related sex dierences. Asthma and wheezing have been pital admissions and fewer doctors visits when compared
demonstrated to be more prevalent in young boys than with white children [117]. Mortality from asthma has been
young girls [20]. This trend disappears during puberty [105]. higher for African American children when compared
A recent analysis of the European Respiratory Health Survey with children of other races since the mid-1980s [47, 57,
[106] found that girls had a lower risk of developing asthma 118121]. Data from the NHANES II and the Hispanic
than did boys during childhood; about the time of puberty Health and Nutrition Survey have noted that the preva-
the risk was equal. After puberty, the risk in young women lence of childhood asthma was higher in children of Puerto
was higher than in young men, and this was a consistent Rican versus African American descent (20.1% versus 9.1%
trend in the 16 countries included in this study. respectively) [122]. An extensive review on the topic has
Women older than 20 years have higher prevalence noted that amongst the dierent ethnic groups living in the
and morbidity rates from asthma, and women are more like United States, the prevalence of asthma is highest in Puerto
to present to the emergency department and be admitted Ricans [123].
with a diagnosis of asthma [107, 108]. In the multicenter Studies in Chicago have demonstrated socioeconomic
Asthma Collaboration Study [109], women were more likely gradients and diering outcomes by race. In 1996, asthma
to be admitted to the hospital and report ongoing symptoms hospitalization rates were more than twice as high as the
at follow-up, although overall men had less outpatient care United States rates overall. Age-adjusted mortality was 4.7
and lower pulmonary function. Although it is unclear why times higher in non-Hispanic African Americans than in
women have more severe disease and higher mortality from non-Hispanic whites [124]. An association with poverty has
asthma, hormonal, genetic, environmental, and social factors been suggested [125], and it has also been suggested that
are likely contributory [110]. severe asthma may occur more frequently in poorer com-
For many years COPD has traditionally been con- munities [126, 127]. The association of lower socioeconomic
sidered a disease of men, as men have been noted to have status with increased asthma prevalence is most likely mul-
a higher prevalence of COPD at the population level. Yet, tifactorial: the eects of indoor air pollution, passive ciga-
Prescott et al. [111] have suggested that women are more rette smoke exposure, allergen exposure, and reduced access
susceptible to the development of COPD, and observed to medical care may all be relevant.
that smoking was associated with a greater decrement in Some have suggested, based on the use of education
FEV1 per pack-year of cigarette smoked, when compared as a surrogate for lower socioeconomic status, that there is
to male smokers. Mannino et al. [112] analyzed data for an association of socioeconomic status with the develop-
deaths from obstructive lung disease from 1979 until 1993 ment of obstructive lung disease. Bakke et al. [128] dem-
and found that the mortality rates for men with COPD onstrated that completion of only primary schooling was
have started to stabilize but were continuing to increase associated with an odds ratio of 2.9 for the development
among women, likely reecting smoking trends. In the year of obstructive lung disease when compared with those who
2000, the number of women dying from COPD has sur- achieved university level education. Exposure to smoking
passed the number of men. Women have been observed to and occupational hazards decreased with increasing educa-
be more susceptible to the eects of tobacco smoking [113], tional status.

17
Asthma and COPD: Basic Mechanisms and Clinical Management

that individuals cannot have both reversible and xed airow


AGE obstruction, and hence asthma and COPD at the same time.
The importance of early life and in utero events for the
Infants born prematurely, when compared to those born at subsequent development of disease is a theme that is com-
term, have a risk for asthma that is increased approximately mon to a variety of complex traits. These same issues also
4-fold [129]; reduced lung function measured with birth has present themselves with disorders of the airways. The major
been associated with incident asthma by age 10 [130]. There barrier to applying a life-cycle approach to disease risk fac-
are data that breastfeeding is protective against asthma and, tors and natural history relates to the problem of recall bias
as previously noted, the risk for asthma increases in chil- and potentially missing or inadequate information about
dren exposed to cigarette smoke both in utero and during past events in both childhood and adult life, which may
childhood. Asthma that begins after age 50 is thought to be then distort the clinical picture. It is only through careful
more severe and less reversible than asthma that is incident analysis of longitudinal cohort data that the true history of
in childhood [131]. In childhood, the remission of asthma the relationship between the major environmental expo-
before adulthood has been suggested to be about 50% [69, sures and disease natural history can be deduced. We need
132, 133]. Less information is available on the epidemi- to continue to gather such data, particularly data to link
ology of asthma in the middle-aged and elderly, yet some childhood asthma with adult COPD.
suggest that older patients are more severely aected than
younger patients [134].
Some data support the proposition that individuals References
may outgrow asthma (with remission rates decreasing with
increasing age) [135]. Other data suggest that remission of 1. Toelle BG, Peat JK, Salome CM, Mellis CM, Woolcock AJ. Toward a
denition of asthma for epidemiology. Am Rev Respir Dis 146(3): 633
asthma and respiratory symptoms are uncommon [136].
37, 1992.
Aging has been associated with increased airway obstruc- 2. Pekkanen J, Pearce N. Dening asthma in epidemiological studies.
tion overall [137] and asthma is a risk factor for COPD in Eur Respir J 14(4): 95157, 1999.
adult lifetime never smokers [84]. The association of aging 3. Boushey HA, Holtzman MJ, Sheller JR, Nadel JA. Bronchial hyper-
with the development of COPD most likely represents the reactivity. Am Rev Respir Dis 121(2): 389413, 1980.
cumulative insult of a lifetime of smoking and environmen- 4. Global Initiative for Asthma. Global strategy for asthma management
tal exposure interacting with a susceptible host. and prevention. Bethesda, MD: NHLBI/WHO Workshop Report,
2004.
5. Hargreave FE, Parameswaran K. Asthma, COPD and bronchitis are
just components of airway disease. Eur Respir J 28(2): 26467, 2006.

CONCLUSION 6. Asher MI, Keil U, Anderson HR et al. International Study of Asthma


and Allergies in Childhood (ISAAC): Rationale and methods. Eur
Respir J 8(3): 48391, 1995.
7. Lindberg A, Jonsson AC, Ronmark E, Lundgren R, Larsson LG,
Ninety percent of all childhood asthma is diagnosed before
Lundback B. Prevalence of chronic obstructive pulmonary disease
the age of 6 years. Since there is a crude inverse relationship according to BTS, ERS, GOLD and ATS criteria in relation to doc-
between respiratory symptoms and level of lung function, it tors diagnosis, symptoms, age, gender, and smoking habits. Respiration
is not surprising that as lung function increases in childhood 72(5): 47179, 2005.
respiratory symptoms decrease and often disappear. Thus, 8. Viegi G, Pedreschi M, Pistelli F et al. Prevalence of airways obstruction
a large number of individuals are left with the intermedi- in a general population: European Respiratory Society vs American
ate phenotypes of increased airways responsiveness and/or Thoracic Society denition. Chest 117(5 Suppl 2): 339S345S, 2000.
allergy at the time that they reach their maximally attained 9. Halbert RJ, Natoli JL, Gano A, Badamgarav E, Buist AS, Mannino
level of lung function between the ages of 15 and 30. These DM. Global burden of COPD: Systematic review and meta-analysis.
intermediate phenotypes represent denable host character- Eur Respir J 28(3): 52332, 2006.
10. Global Initiative for Chronic Obstructive Lung Disease. A collabora-
istics that confer increased susceptibility to a variety of envi-
tive project of the National Heart, Lung and Blood Institute and the
ronmental exposures encountered in adult life, such as viral World Health Organization. National Institutes of Health, National
respiratory illness, occupation, allergen exposure, and perhaps Heart, Lung and Blood Institute, 2006; Accessed June 30, 2007, at
most importantly, cigarette smoking. www.goldcopd.com.
The most clearly dened (non-genetic) susceptibility 11. Celli BR, Cote CG, Marin JM et al. The body-mass index, airow
factors for premature or early-onset COPD are cigarette obstruction, dyspnea, and exercise capacity index in chronic obstructive
smoking, childhood asthma, increased airways responsive- pulmonary disease. N Engl J Med 350(10): 100512, 2004.
ness, and allergy. It is now clear that most airways hyperre- 12. Hurd SS. International eorts directed at attacking the problem of
sponsiveness in adults antedates, precedes, and predicts the COPD. Chest 117(5 Suppl 2): 336S338S, 2000.
development of COPD. Obviously, this construct suggests 13. Salam MT, Wenten M, Gilliland FD. Endogenous and exogenous sex
steroid hormones and asthma and wheeze in young women. J Allergy
that most genes that predict susceptibility to asthma may
Clin Immunol 117(5): 10017, 2006.
also be important genetic predictors of COPD susceptibil- 14. Silverman EK, Weiss ST, Drazen JM et al. Gender-related dierences
ity, and this is a topic of ongoing research. in severe, early-onset chronic obstructive pulmonary disease. Am J
Both asthma and COPD are dened as syndromes, Respir Crit Care Med 162(6): 215258, 2000.
and the denitions are loose; thus it is not surprising that 15. Masoli M, Fabian D, Holt S, Beasley R. The global burden of asthma:
there is substantial overlap between the conditions at any Executive summary of the GINA Dissemination Committee report.
given age. Indeed, there are no substantive data to suggest Allergy 59(5): 46978, 2004.

18
Epidemiology 2
16. Yunginger JW, Reed CE, OConnell EJ, Melton LJ, OFallon WM, year-old children from 9 Spanish populations. The Spanish Group of
Silverstein MD. A community-based study of the epidemiology the ISAAC Study (International Study of Asthma and Allergies in
of asthma. Incidence rates, 19641983. Am Rev Respir Dis 146(4): Childhood). (published erratum appears in Med Clin (Barc) 112(13):494,
88894, 1992. April 17, 1999)]. Med Clin (Barc) 112(5): 17175, 1999.
17. McWhorter WP, Polis MA, Kaslow RA. Occurrence, predictors, and 36. Gerritsen J, Koeter GH, Postma DS, Schouten JP, Knol K. Prognosis
consequences of adult asthma in NHANESI and follow-up survey. of asthma from childhood to adulthood. Am Rev Respir Dis 140(5):
Am Rev Respir Dis 139(3): 72124, 1989. 132530, 1989.
18. Broder I, Higgins MW, Mathews KP, Keller JB. Epidemiology of asthma 37. Centers for Disease Control and Prevention. Tobacco use among adults
and allergic rhinitis in a total community, Tecumseh, Michigan. III. United States, 2005. MMWR Morb Mortal Wkly Rep 55(42): 11458, 2006.
Second survey of the community. J Allergy Clin Immunol 53(3): 12738, 38. Burrows B, Knudson RJ, Cline MG, Lebowitz MD. Quantitative rela-
1974. tionships between cigarette smoking and ventilatory function. Am Rev
19. Broder I, Higgins MW, Mathews KP, Keller JB. Epidemiology of Respir Dis 115(2): 195205, 1977.
asthma and allergic rhinitis in a total community, Tecumseh, Michigan. 39. Stang P, Lydick E, Silberman C, Kempel A, Keating ET. The preva-
IV. Natural history. J Allergy Clin Immunol 54(2): 10010, 1974. lence of COPD: Using smoking rates to estimate disease frequency in
20. Dodge RR, Burrows B. The prevalence and incidence of asthma and the general population. Chest 117(5 Suppl 2): 354S359S, 2000.
asthma-like symptoms in a general population sample. Am Rev Respir 40. Lopez AD, Murray CC. The global burden of disease, 19902020.
Dis 122(4): 56775, 1980. Nat Med 4(11): 124143, 1998.
21. Martinez FD, Wright AL, Taussig LM, Holberg CJ, Halonen M, 41. Lacasse Y, Brooks D, Goldstein RS. Trends in the epidemiology of
Morgan WJ. Asthma and wheezing in the rst six years of life. The COPD in Canada, 1980 to 1995. COPD and Rehabilitation Committee
Group Health Medical Associates. N Engl J Med 332(3): 13338, 1995. of the Canadian Thoracic Society. Chest 116(2): 30613, 1999.
22. Toren K, Hermansson BA. Incidence rate of adult-onset asthma in 42. Enright PL, Kronmal RA, Higgins MW, Schenker MB, Haponik EF.
relation to age, sex, atopy and smoking: A Swedish population-based Prevalence and correlates of respiratory symptoms and disease in the
study of 15813 adults. Int J Tuberc Lung Dis 3(3): 19297, 1999. elderly. Cardiovascular Health Study. Chest 106(3): 82734, 1994.
23. Huovinen E, Kaprio J, Laitinen LA, Koskenvuo M. Incidence and prev- 43. Mannino DM. Epidemiology and global impact of chronic obstructive
alence of asthma among adult Finnish men and women of the Finnish pulmonary disease. Semin Respir Crit Care Med 26(2): 20410, 2005.
Twin Cohort from 1975 to 1990, and their relation to hay fever and 44. Schirnhofer L, Lamprecht B, Vollmer WM et al. COPD prevalence
chronic bronchitis. Chest 115(4): 92836, 1999. in Salzburg, Austria: Results from the Burden of Obstructive Lung
24. de Marco R, Accordini S, Cerveri I et al. Incidence of chronic obstruc- Disease (BOLD) Study. Chest 131(1): 2936, 2007.
tive pulmonary disease in a cohort of young adults according to the 45. Sobradillo V, Miravitlles M, Jimenez CA et al. [Epidemiological study
presence of chronic cough and phlegm. Am J Respir Crit Care Med of chronic obstructive pulmonary disease in Spain (IBERPOC):
175(1): 3239, 2007. Prevalence of chronic respiratory symptoms and airow limitation].
25. Lindberg A, Jonsson AC, Ronmark E, Lundgren R, Larsson LG, Arch Bronconeumol 35(4): 15966, 1999.
Lundback B. Ten-year cumulative incidence of COPD and risk factors for 46. Fleming DM, Crombie DL. Prevalence of asthma and hay fever in
incident disease in a symptomatic cohort. Chest 127(5): 154452, 2005. England and Wales. BMJ (Clin Res Ed) 294(6567): 27983, 1987.
26. Johannessen A, Omenaas E, Bakke P, Gulsvik A. Incidence of GOLD- 47. Gerstman BB, Bosco LA, Tomita DK, Gross TP, Shaw MM.
dened chronic obstructive pulmonary disease in a general adult popu- Prevalence and treatment of asthma in the Michigan Medicaid patient
lation. Int J Tuberc Lung Dis 9(8): 92632, 2005. population younger than 45 years, 19801986. J Allergy Clin Immunol
27. Mannino DM, Gagnon RC, Petty TL, Lydick E. Obstructive lung dis- 83(6): 103239, 1989.
ease and low lung function in adults in the United States: Data from 48. Manfreda J, Becker AB, Wang PZ, Roos LL, Anthonisen NR. Trends
the National Health and Nutrition Examination Survey, 19881994. in physician-diagnosed asthma prevalence in Manitoba between 1980
Arch Intern Med 160(11): 168389, 2000. and 1990. Chest 103(1): 15157, 1993.
28. Eder W, Ege MJ, von Mutius E. The asthma epidemic. N Engl J Med 49. Mitchell EA. International trends in hospital admission rates for
55(21): 222635, 2006. asthma. Arch Dis Child 60(4): 37678, 1985.
29. Mannino DM, Homa DM, Pertowski CA et al. Surveillance for asthma 50. Anderson HR, Bailey P, West S. Trends in the hospital care of acute
United States, 19601995. Mor Mortal Wkly Rep CDC Surveill Summ childhood asthma 19708: A regional study. BMJ 281(6249): 119194,
47(1): 127, 1998. 1980.
30. Crain EF, Weiss KB, Bijur PE, Hersh M, Westbrook L, Stein RE. An 51. Jackson RT, Mitchell EA. Trends in hospital admission rates and
estimate of the prevalence of asthma and wheezing among inner-city drug treatment of asthma in New Zealand. N Z Med J 96(740):
children. Pediatrics 94(3): 35662, 1994. 72830, 1983.
31. Persky VW, Slezak J, Contreras A et al. Relationships of race and socio- 52. Halfon N, Newacheck PW. Trends in the hospitalization for acute
economic status with prevalence, severity, and symptoms of asthma in childhood asthma, 197084. Am J Public Health 76(11): 130811, 1986.
Chicago school children. Ann Allergy Asthma Immunol 81(3): 26671, 53. Priftis K, Anagnostakis J, Harokopos E, Orfanou I, Petraki M, Saxoni-
1998. Papageorgiou P. Time trends and seasonal variation in hospital admis-
32. Sole D, Yamada E, Vana AT, Costa-Carvalho BT, Naspitz CK. sions for childhood asthma in the Athens region of Greece: 197888.
Prevalence of asthma and related symptoms in school-age children Thorax 48(11): 116869, 1993.
in Sao Paulo, Brazil International Study of Asthma and Allergies in 54. Carman PG, Landau LI. Increased paediatric admissions with asthma in
Children (ISAAC). J Asthma 36(2): 20512, 1999. Western Australia a problem of diagnosis?. Med J Aust 152(1): 2326,
33. Asher MI, Weiland SK. The International Study of Asthma and 1990.
Allergies in Childhood (ISAAC). ISAAC Steering Committee. Clin 55. Kun HY, Oates RK, Mellis CM. Hospital admissions and attendances
Exp Allergy 28(Suppl 5): 5266, 1998. discussion 901 for asthma a true increase? Med J Aust 159(5): 31213, 1993.
34. Duhme H, Weiland SK, Rudolph P, Wienke A, Kramer A, Keil U. 56. Wilkins K, Mao Y. Trends in rates of admission to hospital and death
Asthma and allergies among children in West and East Germany: A from asthma among children and young adults in Canada during the
comparison between Munster and Greifswald using the ISAAC phase 1980s. Can Med Assoc J 148(2): 18590, 1993.
I protocol. International Study of Asthma and Allergies in Childhood. 57. Evans Rd, Mullally DI, Wilson RW et al. National trends in the mor-
Eur Respir J 11(4): 84047, 1998. bidity and mortality of asthma in the US. Prevalence, hospitalization
35. Aguinaga Ontoso I, Arnedo Pena A, Bellido J, Guillen Grima F, Suarez and death from asthma over two decades: 19651984. Chest 91(6
Varela MM. [The prevalence of asthma-related symptoms in 1314- Suppl): 65S74S, 1987.

19
Asthma and COPD: Basic Mechanisms and Clinical Management

58. Mannino DM, Homa DM, Akinbami LJ, Ford ES, Redd SC. Chronic the rate of decline of FEV1. The Lung Health Study. JAMA 272(19):
obstructive pulmonary disease surveillance United States, 19712000. 1497505, 1994.
MMWR Surveill Summ 51(6): 116, 2002. 83. Whittemore AS, Perlin SA, DiCiccio Y. Chronic obstructive pulmo-
59. Jackson R, Sears MR, Beaglehole R, Rea HH. International trends in nary disease in lifelong nonsmokers: Results from NHANES. Am J
asthma mortality: 1970 to 1985. Chest 94(5): 91418, 1988. Public Health 85(5): 7026, 1995.
60. Carr W, Zeitel L, Weiss K. Variations in asthma hospitalizations and 84. Behrendt CE. Mild and moderate-to-severe COPD in nonsmokers:
deaths in New York City. Am J Public Health 82(1): 5965, 1992. Distinct demographic proles. Chest 128(3): 123944, 2005.
61. McConnochie KM, Russo MJ, McBride JT, Szilagyi PG, Brooks AM, 85. Birring SS, Brightling CE, Bradding P et al. Clinical, radiologic, and
Roghmann KJ. Socioeconomic variation in asthma hospitalization: induced sputum features of chronic obstructive pulmonary disease in
Excess utilization or greater need?. Pediatrics 103(6): e75, 1999. nonsmokers: A descriptive study. Am J Respir Crit Care Med 166(8):
62. Thom TJ. International comparisons in COPD mortality. Am Rev 107883, 2002.
Respir Dis 140(3 Pt 2): S2734, 1989. 86. Pena VS, Miravitlles M, Gabriel R et al. Geographic variations in
63. Burr ML, Davies BH, Hoare A et al. A condential inquiry into prevalence and underdiagnosis of COPD: Results of the IBERPOC
asthma deaths in Wales. Thorax 54(11): 98589, 1999. multicentre epidemiological study. Chest 118(4): 98189, 2000.
64. Ringbaek T, Seersholm N, Viskum K. Standardised mortality rates 87. Taylor RG, Joyce H, Gross E, Holland F, Pride NB. Bronchial reac-
in females and males with COPD and asthma. Eur Respir J 25(5): tivity to inhaled histamine and annual rate of decline in FEV1 in male
89195, 2005. smokers and ex-smokers. Thorax 40(1): 916, 1985.
65. Mannino DM, Reichert MM, Davis KJ. Lung function decline and 88. Frew AJ, Kennedy SM, Chan-Yeung M. Methacholine responsiveness,
outcomes in an adult population. Am J Respir Crit Care Med 173(9): smoking, and atopy as risk factors for accelerated FEV1 decline in
98590, 2006. male working populations. Am Rev Respir Dis 146(4): 87883, 1992.
66. Cunningham J, Dockery DW, Speizer FE. Maternal smoking during 89. Gottlieb DJ, Sparrow D, OConnor GT, Weiss ST. Skin test reactivity
pregnancy as a predictor of lung function in children. Am J Epidemiol to common aeroallergens and decline of lung function. The Normative
139(12): 113952, 1994. Aging Study. Am J Respir Crit Care Med 153(2): 56166, 1996.
67. Tager IB, Munoz A, Rosner B, Weiss ST, Carey V, Speizer FE. Eect 90. Nelson HS. The importance of allergens in the development of asthma
of cigarette smoking on the pulmonary function of children and ado- and the persistence of symptoms. J Allergy Clin Immunol 105(6 Pt 2):
lescents. Am Rev Respir Dis 131(5): 75259, 1985. S628S632, 2000.
68. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking, changes in smoking 91. Burrows B, Martinez FD, Halonen M, Barbee RA, Cline MG.
habits, and rate of decline in FEV1: New insight into gender dier- Association of asthma with serum IgE levels and skin-test reactivity
ences. Eur Respir J 7(6): 105661, 1994. to allergens. N Engl J Med 320(5): 27177, 1989.
69. Strachan DP, Butland BK, Anderson HR. Incidence and prognosis 92. Sears MR, Burrows B, Flannery EM, Herbison GP, Hewitt CJ,
of asthma and wheezing illness from early childhood to age 33 in a Holdaway MD. Relation between airway responsiveness and serum
national British cohort. BMJ 312(7040): 119599, 1996. IgE in children with asthma and in apparently normal children.
70. Kaplan BA, Mascie-Taylor CG. Smoking and asthma among 23-year- N Engl J Med 325(15): 106771, 1991.
olds. J Asthma 34(3): 21926, 1997. 93. Weiss ST. Atopy as a risk factor for chronic obstructive pulmonary
71. Ulrik CS, Frederiksen J. Mortality and markers of risk of asthma death disease. Epidemiological evidence. Am J Respir Crit Care Med 162(3
among 1,075 outpatients with asthma. Chest 108(1): 1015, 1995. Pt 2): S134S136, 2000.
72. Althuis MD, Sexton M, Prybylski D. Cigarette smoking and asthma 94. Hargreave FE, Leigh R. Induced sputum, eosinophilic bronchitis,
symptom severity among adult asthmatics. J Asthma 36(3): 25764, and chronic obstructive pulmonary disease. Am J Respir Crit Care Med
1999. 160(5 Pt 2): S53S57, 1999.
73. Lange P, Parner J, Vestbo J, Schnohr P, Jensen G. A 15-year follow- 95. Sherrill DL, Lebowitz MD, Halonen M, Barbee RA, Burrows B.
up study of ventilatory function in adults with asthma. N Engl J Med Longitudinal evaluation of the association between pulmonary
339(17): 1194200, 1998. function and total serum IgE. Am J Respir Crit Care Med 152(1):
74. Siroux V, Pin I, Oryszczyn MP, Le Moual N, Kaumann F. 98102, 1995.
Relationships of active smoking to asthma and asthma severity in the 96. Rijcken B, Schouten JP, Weiss ST, Speizer FE, van der Lende R. The
EGEA study. Epidemiological study on the Genetics and Environment association of airways responsiveness to respiratory symptom preva-
of Asthma. Eur Respir J 15(3): 47077, 2000. lence and to pulmonary function in a random population sample. Bull
75. Halken S, Host A, Nilsson L, Taudorf E. Passive smoking as a risk fac- Eur Physiopathol Respir 23(4): 39194, 1987.
tor for development of obstructive respiratory disease and allergic sen- 97. Hospers JJ, Postma DS, Rijcken B, Weiss ST, Schouten JP. Histamine
sitization. Allergy 50(2): 97105, 1995. airway hyper-responsiveness and mortality from chronic obstructive
76. Wartenberg D, Ehrlich R, Lilienfeld D. Environmental tobacco smoke pulmonary disease: A cohort study. Lancet 356(9238): 131317, 2000.
and childhood asthma: Comparing exposure metrics using probability 98. Xu X, Rijcken B, Schouten JP, Weiss ST. Airways responsiveness and
plots. Environ Res 64(2): 12235, 1994. development and remission of chronic respiratory symptoms in adults.
77. Cook DG, Strachan DP. Health eects of passive smoking. 3. Parental Lancet 350(9089): 143134, 1997.
smoking and prevalence of respiratory symptoms and asthma in school 99. Carey VJ, Weiss ST, Tager IB, Leeder SR, Speizer FE. Airways
age children. Thorax 52(12): 108194, 1997. responsiveness, wheeze onset, and recurrent asthma episodes in young
78. Weiss ST, Utell MJ, Samet JM. Environmental tobacco smoke expo- adolescents. The East Boston Childhood Respiratory Disease Cohort.
sure and asthma in adults. Environ Health Perspect 107(Suppl 6): 891 Am J Respir Crit Care Med 153(1): 35661, 1996.
95, 1999. 100. Weiss ST, Tosteson TD, Segal MR, Tager IB, Redline S, Speizer FE.
79. Weil CM, Wade SL, Bauman LJ, Lynn H, Mitchell H, Lavigne J. The Eects of asthma on pulmonary function in children. A longitudinal
relationship between psychosocial factors and asthma morbidity in population-based study. Am Rev Respir Dis 145(1): 5864, 1992.
inner-city children with asthma. Pediatrics 104(6): 127480, 1999. 101. OConnor GT, Sparrow D, Weiss ST. The role of allergy and non-
80. Infante-Rivard C. Childhood asthma and indoor environmental risk specic airway hyperresponsiveness in the pathogenesis of chronic
factors. Am J Epidemiol 137(8): 83444, 1993. obstructive pulmonary disease. Am Rev Respir Dis 140(1): 22552,
81. Infante-Rivard C, Gautrin D, Malo JL, Suissa S. Maternal smoking 1989.
and childhood asthma. Am J Epidemiol 150(5): 52831, 1999. 102. Sluiter HJ, Koeter GH, de Monchy JG, Postma DS, de Vries K, Orie
82. Anthonisen NR, Connett JE, Kiley JP et al. Eects of smoking inter- NG. The Dutch hypothesis (chronic non-specic lung disease) revis-
vention and the use of an inhaled anticholinergic bronchodilator on ited. Eur Respir J 4(4): 47989, 1991.

20
Epidemiology 2
103. Rijcken B, Schouten JP, Xu X, Rosner B, Weiss ST. Airway hyper- 119. Nelson DA, Johnson CC, Divine GW, Strauchman C, Joseph CL,
responsiveness to histamine associated with accelerated decline in Ownby DR. Ethnic dierences in the prevalence of asthma in middle
FEV1. Am J Respir Crit Care Med 151(5): 137782, 1995. class children. Ann Allergy Asthma Immunol 78(1): 2126, 1997.
104. Tashkin DP, Altose MD, Connett JE, Kanner RE, Lee WW, 120. Joseph CL, Foxman B, Leickly FE, Peterson E, Ownby D. Prevalence
Wise RA. Methacholine reactivity predicts changes in lung function of possible undiagnosed asthma and associated morbidity among
over time in smokers with early chronic obstructive pulmonary disease. urban schoolchildren. J Pediatr 129(5): 73542, 1996.
The Lung Health Study Research Group. Am J Respir Crit Care Med 121. Weitzman M, Gortmaker SL, Sobol AM, Perrin JM. Recent trends
153(6 Pt 1): 180211, 1996. in the prevalence and severity of childhood asthma. JAMA 268(19):
105. Venn A, Lewis S, Cooper M, Hill J, Britton J. Questionnaire study of 267377, 1992.
eect of sex and age on the prevalence of wheeze and asthma in ado- 122. Carter-Pokras OD, Gergen PJ. Reported asthma among Puerto
lescence. BMJ 316(7149): 194546, 1998. Rican, Mexican-American, and Cuban children, 1982 through 1984.
106. de Marco R, Locatelli F, Sunyer J, Burney P. Dierences in incidence Am J Public Health 83(4): 58082, 1993.
of reported asthma related to age in men and women. A retrospective 123. Hunninghake GM, Weiss ST, Celedon JC. Asthma in Hispanics.
analysis of the data of the European Respiratory Health Survey. Am J Am J Respir Crit Care Med 173(2): 14363, 2006.
Respir Crit Care Med 162(1): 6874, 2000. 124. Thomas SD, Whitman S. Asthma hospitalizations and mortal-
107. Skobelo EM, Spivey WH, St. Clair SS, Schostall JM. The inuence ity in Chicago: An epidemiologic overview. Chest 116(4 Suppl 1):
of age and sex on asthma admissions. JAMA 268(24): 343740, 1992. 135S141S, 1999.
108. Baibergenova A, Thabane L, Akhtar-Danesh N, Levine M, Gafni A, 125. Duran-Tauleria E, Rona RJ. Geographical and socioeconomic varia-
Leeb K. Sex dierences in hospital admissions from emergency depart- tion in the prevalence of asthma symptoms in English and Scottish
ments in asthmatic adults: A population-based study. Ann Allergy children. Thorax 54(6): 47681, 1999.
Asthma Immunol 96(5): 66672, 2006. 126. Mielck A, Reitmeir P, Wjst M. Severity of childhood asthma by soci-
109. Singh AK, Cydulka RK, Stahmer SA, Woodru PG, Camargo CA oeconomic status. Int J Epidemiol 25(2): 38893, 1996.
Jr. Sex dierences among adults presenting to the emergency depart- 127. Strachan DP, Anderson HR, Limb ES, ONeill A, Wells N. A national
ment with acute asthma. Multicenter Asthma Research Collaboration survey of asthma prevalence, severity, and treatment in Great Britain.
Investigators. Arch Intern Med 159(11): 123743, 1999. Arch Dis Child 70(3): 17478, 1994.
110. Melgert BN, Ray A, Hylkema MN, Timens W, Postma DS. Are there 128. Bakke PS, Hanoa R, Gulsvik A. Educational level and obstruc-
reasons why adult asthma is more common in females? Curr Allergy tive lung disease given smoking habits and occupational airborne
Asthma Rep 7(2): 14350, 2007. exposure: A Norwegian community study. Am J Epidemiol 141(11):
111. Prescott E, Bjerg AM, Andersen PK, Lange P, Vestbo J. Gender dif- 108088, 1995.
ference in smoking eects on lung function and risk of hospitalization 129. von Mutius E, Nicolai T, Martinez FD. Prematurity as a risk factor
for COPD: Results from a Danish longitudinal population study. Eur for asthma in preadolescent children. J Pediatr 123(2): 22329, 1993.
Respir J 10(4): 82227, 1997. 130. Haland G, Carlsen KC, Sandvik L et al. Reduced lung function at
112. Mannino DM, Brown C, Giovino GA. Obstructive lung disease birth and the risk of asthma at 10 years of age. N Engl J Med 355(16):
deaths in the United States from 1979 through 1993. An analy- 168289, 2006.
sis using multiple-cause mortality data. Am J Respir Crit Care Med 131. Vergnenegre A, Antonini MT, Bonnaud F, Melloni B, Mignonat G,
156(3 Pt 1): 81418, 1997. Bousquet J. Comparison between late onset and childhood asthma.
113. Gan WQ, Man SF, Postma DS, Camp P, Sin DD. Female smokers Allergol Immunopathol (Madr) 20(5): 19096, 1992.
beyond the perimenopausal period are at increased risk of chronic 132. Jonsson JA, Boe J, Berlin E. The long-term prognosis of childhood
obstructive pulmonary disease: A systematic review and meta-analysis. asthma in a predominantly rural Swedish county. Acta Paediatr Scand
Respir Res 7: 52, 2006. 76(6): 95054, 1987.
114. Martinez FJ, Curtis JL, Sciurba F et al. Gender dierences in severe pul- 133. Kelly WJ, Hudson I, Phelan PD, Pain MC, Olinsky A. Childhood
monary emphysema. Am J Respir Crit Care Med 176(3): 2223, 2007. asthma in adult life: A further study at 28 years of age. BMJ (Clin Res
115. Machado MC, Krishnan JA, Buist SA et al. Sex dierences in survival Ed) 294(6579): 105962, 1987.
of oxygen-dependent patients with chronic obstructive pulmonary 134. Burrows B, Barbee RA, Cline MG, Knudson RJ, Lebowitz MD.
disease. Am J Respir Crit Care Med 174(5): 52429, 2006. Characteristics of asthma among elderly adults in a sample of the
116. Taylor WR, Newacheck PW. Impact of childhood asthma on health. general population. Chest 100(4): 93542, 1991.
Pediatrics 90(5): 65762, 1992. 135. Panhuysen CI, Vonk JM, Koeter GH et al. Adult patients may out-
117. Coultas DB, Gong H Jr., Grad R et al. Respiratory diseases in minor- grow their asthma: A 25-year follow-up study. Am J Respir Crit Care
ities of the United States. Am J Respir Crit Care Med 149(3 Pt 2): Med 155(4): 126772, 1997. [published erratum appears in Am J
S93131, 1994. [published erratum appears in Am J Respir Crit Care Respir Crit Care Med 156(2 Pt 1): 674, August 1997]
Med 150(1):290, July 1994] 136. Ronmark E, Jonsson E, Lundback B. Remission of asthma in the
118. Clark NM, Feldman CH, Evans D, Levison MJ, Wasilewski Y, middle aged and elderly: Report from the Obstructive Lung Disease
Mellins RB. The impact of health education on frequency and cost in Northern Sweden study. Thorax 54(7): 61113, 1999.
of health care use by low income children with asthma. J Allergy Clin 137. Wise RA. Changing smoking patterns and mortality from chronic
Immunol 78(1 Pt 1): 10815, 1986. obstructive pulmonary disease. Prev Med 26(4): 41821, 1997.

21
Natural History
3
CHAPTER

and COPD share common risk factors or, alter-


INTRODUCTION natively, by the possible progression of persistent Stefano Guerra and
asthma into chronic airow limitation, the clini- Fernando D. Martinez
There is increasing interest in developing a better cal hallmark of COPD [4]. It is often not pos-
Arizona Respiratory Center and Mel
understanding of the natural history of the two sible to determine whether persons with airow
limitation have asthma or COPD. However, and Enid Zuckerman College of Public
most frequent chronic lower respiratory diseases,
understanding the natural history of the disease Health, University of Arizona, Tucson,
asthma and chronic obstructive pulmonary dis-
ease (COPD). The main reason for this interest is will have critical implications for identifying AZ, USA
the shift in the therapeutic approach to both con- optimal prevention and treatment strategies.
ditions. In the case of asthma, a growing body of
evidence suggests that, although several current
treatments for the disease are extremely eec-
tive in controlling symptoms, none can change
METHODOLOGICAL APPROACH
its natural course. It is thus evident that new
approaches to the primary and secondary pre- Any discussion about the natural history of a
vention of asthma are needed. These approaches chronic condition ought to address the dierent
will require a thorough understanding of the fac- hypothetical phases that such condition under-
tors that determine the inception of asthma and goes during its lifetime course (Fig. 3.1).
its progression with age. In the case of COPD, There is rst a pre-illness period during
it is apparent that, although smoking is the main which subjects without overt disease have the sus-
demonstrated cause of this condition, a simple ceptibility for the development of the condition,
strategy of discouraging tobacco consumption because of a genetic predisposition or because
has proven insucient to prevent the enormous of injuries or developmental variations that have
impact of nicotine addiction on public health. mutated one or more of the individuals disease-
A new understanding of the factors that increase associated phenotypes. During this pre-illness
the risk of COPD in smokers seems a more real- phase, the susceptible individual is not yet des-
istic approach, and could perhaps contribute to tined to develop the disease. More likely, he or
palliate the increasing social toll of COPD. she may be exposed to environmental factors that,
When discussing the natural history of in the presence of genetic variants that predispose
asthma and COPD, an important issue is the to the chronic condition, can further modify their
overlap between these two conditions [1]. It has phenotype. This model applies to both asthma
been conclusively shown that, at the population and COPD, as they are both likely to represent
level, adults who report asthma are more likely to heterogeneous diseases with multiple ways to
also report chronic bronchitis and/or emphysema reach nal common pathways and an array of
as compared with subjects with no asthma [2] diverse pathogenetic mechanisms giving rise to
and recent epidemiological evidence shows that more or less reversible airway obstruction.
active asthma is a strong risk factor for acquir- It would obviously be of great help for any
ing a subsequent diagnosis of COPD [3]. This prevention strategy if markers of susceptibility
overlap may be explained by the fact that asthma were identied that could be measured during

23
Asthma and COPD: Basic Mechanisms and Clinical Management

Inception Transient early Nonatopic IgE-associated


n Chronic
wheezers wheezers wheeze/asthma
sio disease
es
ogr
Pr

Wheezing prevalence
Protection Intermittent
No disease Initial phase
disease

Re
mi
ss
ion
Triggers No disease

FIG. 3.1 Hypothetical representation of the natural history of a chronic


condition. For explanation see text.

this pre-illness phase. But the absence of a marker in a spe- 0 3 6 11


cic individual may not exclude susceptibility if that is not the Age (years)
pathway that may be active in that individual. Thus, because
FIG. 3.2 Hypothetical yearly peak prevalence of wheezing for three
the marker phenotype may be inuenced by determinants
different phenotypes in childhood. Prevalence for each age interval should
other than the factors that inuence asthma or COPD risk, be the sum of the areas under each curve. The dashed curves suggest
ascertainment of a marker may be fraught with diculties. wheezing can present different curve shapes due to many different factors,
This context-dependency [5] of the phenotypes of markers including overlap of groups. Adapted from Ref. [15].
of disease creates a further layer of complexity for the identi-
cation of susceptible individuals.
The above discussion points to the importance of
manifestations of asthma occur during the rst 5 years of
determining the true time of incidence of the disease under
life [6, 11, 13]. Studies have ascertained this age of onset of
study. Although the event that is easiest to ascertain is the age
symptoms either by asking parents about the prevalence of
of initiation of symptoms, this event is modied by patients
wheezing or other asthma-like symptoms in their children
perceptions. What is important is that time of onset ought
at dierent ages, or by reviewing previous clinical charts in
to be ascertained before the disease has become chronic, that
subjects who present to outpatient clinics with signs and
is before the individual has evidence of physiological or ana-
symptoms of asthma at any age. Although these studies
tomical changes caused by the disease itself and that predis-
strongly suggest that asthma started at an early age in these
pose to persistent or recurrent symptoms. During this initial
individuals, a simple connection between these two phe-
phase of the disease, the aected individual has some clinical
nomena is not warranted. Many children have asthma-like
manifestations, but the condition has not yet fully developed.
symptoms during viral infections in their preschool years,
Moreover, some individuals may not go beyond this initial
but most have transient conditions that subside with age,
phase, because remission or stabilization may be fostered by
and only a minority will go on to have persistent asthma
the presence of environmental or genetic protective factors.
[14]. Thus it is dicult to distinguish who will go on to
During this initial phase secondary prevention is possible,
develop chronic asthma from those who will not.
but again knowledge of the natural history becomes essential.
Studies of this issue have suggested that there are at
least three quite distinct groups of children with asthma-like
symptoms coexisting up to the adolescent years (Fig. 3.2).
NATURAL HISTORY OF ASTHMA PHENOTYPES The great majority of infants who wheeze during the rst
12 years of life do so during viral infections, especially
those caused by the respiratory syncytial virus (RSV) and
Asthma is a heterogeneous condition, and one of the main by rhinovirus. Most of these children will have one or only
obstacles in understanding its natural history has been a few episodes of wheezing, with no further symptoms
the lack of well-dened markers for the dierent disease beyond the age of 23 years. This condition, which has been
phenotypes grouped under this common label. This hurdle identied as transient wheezing of infancy [16], is the most
has been addressed lately by a series of longitudinal studies frequent form of recurrent airway obstruction in this age
that have assessed incidence and prevalence of asthma group, aecting over two-thirds of all infants with asthma-
at dierent ages or dened the outcome of persons with like symptoms. The main predisposing factors for transient
asthma-like symptoms enrolled at dierent ages and espe- wheezing of infancy are maternal smoking during preg-
cially during childhood [613]. nancy and lower levels of lung function, as assessed during
the very rst months of life and before any wheezing epi-
sode has occurred. Interestingly, infants (especially females)
Most cases of chronic, persistent asthma whose mothers smoke during pregnancy do have lower
start in early life levels of lung function early in life than those whose moth-
ers do not smoke [17]. This has suggested the possibility
Several longitudinal studies have conrmed that, in most that both inherited and acquired characteristics of the lung,
cases of chronic, persistent asthma, the initial clinical the airways, or both may create the structural conditions for

24
Natural History 3
the development of airway obstruction during lower respi- 40
ratory illnesses [18]. Alternaria negative asthma
The factors that determine the remission of early life Alternaria positive asthma
wheezing in these children are not well understood, but
it is likely that growth of the airways may outpace that of 30

Percentage of asthma group


the lung parenchyma after age 25 months, the period of
the highest incidence of wheezing episodes [19]. Since the
airways are growing faster than the parenchyma, the condi-
tions required for airow to be dynamically obstructed may
not be easily reached and noisy breathing is less likely [19]. 20
It is also possible that changes in the regulation of airway
tone may result in decreased bronchial hyperresponsiveness
and decreased likelihood of respiratory symptoms with age
[20, 21]. However, despite remission of wheezing with age 10
in this group, children with early transient wheezing appear
to have lower levels of lung function, in terms of FEF2575,
FEV1, and FEV1/FVC ratio values, throughout childhood
as compared with their peers who did not wheeze in the 0
rst 6 years of life [22]. It is established that individuals 0
01 12 23 34 45 56 67
who enter adulthood with lung function decits are more Age at which asthma diagnosed (years)
likely to develop COPD during the late adult years, sug-
gesting that children with early transient wheezing may be FIG. 3.3 Age of diagnosis of asthma for children who are skin-test positive
particularly predisposed to COPD in adult life if exposed for Alternaria or skin-test negative for Alternaria at age 6 in the Tucson
to cigarette smoking. Whether this is the case is at present Childrens Respiratory Study. Adapted from Ref. [24].
unknown.
The factors that determine enduring or newly devel-
oped abnormalities in lung function during the rst years with a history of RSV-LRI than in those with no such
of life are the subject of intense scrutiny, because they seem history. However, the risk decreased with age and was not
to be clearly associated with the risk of persistent asthma. statistically signicant by early adolescence. There was also
Zeiger et al. [23] showed an inverse relationship between no association between RSV-LRI and subsequent risk of
duration of symptoms and level of lung function in school- sensitization to local aeroallergens either at age 6 or at age
age children with asthma, and this suggests that the ear- 11 years. The only factor that was strongly associated with
lier onset of asthma symptoms is associated with greater RSV-LRI in early life was diminished baseline levels of
losses in lung function. Sensitization to local aeroaller- FEV1, as measured at age 11. Interestingly, these decits
gens is strongly associated with increased risk of chronic were reversed by use of a bronchodilator, suggesting that
asthma-like symptoms into adult life. Using this knowledge, they were likely to be due to increased bronchomotor tone.
Halonen et al. [24] divided children who had a diagnosis of Longitudinal studies performed in the early 1980s had also
asthma by age 6 years into two main groups: those who at suggested that the outcome of RSV-LRIs is usually benign
age 6 were skin-test positive for Alternaria, the main aeroal- and unrelated to increased allergic sensitization [26].
lergen associated with asthma in the study area [25], and It thus appears that at least three dierent forms of
those who were not. They observed that, in the majority of asthma coexist during infancy and early childhood (Fig. 3.2).
children in both groups, symptoms had started before age Transient infant wheezing is quite frequent, is usually trig-
3 years. However, among children with asthma who were gered by viruses (especially RSV), and, although it is con-
skin-test negative to Alternaria, inception of the disease had ned to the rst 3 years of life, may be associated with
occurred mainly during the rst year of life; while in those long-term lung function decits. Other children who
who were skin-test positive to Alternaria, peak incidence wheeze during viral infections in early life continue to have
occurred during the second and third years of life (Fig. 3.3). recurrent respiratory symptoms during the early school
The fact that young children who will go on to years. This form of nonatopic wheezing is associated with
develop chronic, atopy-related asthma by age 6 are diag- lower levels of lung function during the school years, but
nosed 1 or 2 years later than those with nonatopic asthma these lower levels seem to be reversible after use of a bron-
suggests that the mechanisms of disease are likely to be chodilator. Finally, children who will develop atopy-related
dierent in these two groups. Support for this contention asthma start having symptoms mainly during the second
comes from a slightly dierent analysis of the data from and third years of life.
the same cohort on which Halonen and coworkers based
their report quoted above. Stein et al. [14] assessed the out-
come of children with conrmed lower respiratory tract ill- Chronic asthma is most often related
nesses due to RSV (RSV-LRI), after adjusting for all other to atopy
known risk factors for subsequent asthma, including skin
test reactivity, maternal history of asthma and birth weight, There is now strong evidence indicating that, as a group,
among others. They found that, as previously reported, risk individuals with chronic asthma at any age between the ele-
of wheezing during the school years was higher in children mentary school years and mid-adult life are either sensitized

25
Asthma and COPD: Basic Mechanisms and Clinical Management

to local aeroallergens, have elevated total levels of circulat- the immune system and, in turn, they may lead to susceptibil-
ing IgE, or both [27]. Although the association between ity to viral infections, favor a Th2 skewed polarization of the
asthma and total and specic IgE is well established, the immune system, and aect airway growth. Whether decits
nature of the association is not well understood. For years of early IFN responses can explain the link between asthma
it was thought that sensitization to specic allergens, espe- and atopy or simply represent one of multiple pathways that
cially in early life, was a cause of asthma [28], and that are shared by these two phenotypes remains unknown.
development of specic IgE against these allergens was the
rst step in the natural history of the disease. The strong
association between risk of having asthma and sensitiza- Outcome of childhood asthma in adolescence
tion to the allergens of house dust mites in coastal regions
seemed to argue in favor of this hypothesis, and strategies and adult life: Remission of mild asthma and
for the primary prevention of asthma based on avoidance persistence of severe disease
of exposure to these allergens were proposed [29]. However,
several studies performed in desert areas with low exposure Only a few population-based studies have addressed the
to house dust mites showed that the prevalence of asthma outcome of childhood asthma in adolescence and adult life.
was either similar or even higher in these regions than These studies have used longitudinal assessment of asthma
that observed in zones where mite infestation rates were symptoms and lung function in children who were enrolled
high [25, 30]. In desert regions, the allergens of the mold at birth [12] or during the early school years [36, 37] or
Alternaria appeared to be strongly associated with asthma. who were part of birth cohorts for which assessment of
Moreover, studies in northern Sweden, where indoor expo- asthma symptoms in early life was made through retrospec-
sure to either dust mites or molds is very low, showed that tive questionnaires [7, 8, 10, 38].
the prevalence of school-age asthma is very similar to that The transition of asthma from childhood into adoles-
observed in southern Sweden, where exposure to house dust cence has been studied in the birth cohort of the Tucson
mites is high [31]. Interestingly, only 50% of all schoolchil- Childrens Respiratory Study [12]. In that study, most
dren with asthma are sensitive to known aeroallergens in children who experienced only infrequent wheezing dur-
northern Sweden (especially cat and dog), compared with ing childhood had remission of wheezing after puberty.
over 90% of children in more temperate regions, but skin- However, almost 60% of children with frequent wheezing
test negative children with asthma have signicantly higher and/or a physician-conrmed diagnosis of asthma expe-
IgE levels than nonasthmatic children [31]. These obser- rienced wheezing at some point during adolescence. These
vations suggest that, regardless of the nature of the asso- results challenge the commonly held view that most chil-
ciation between elevated IgE and asthma, this link is not dren with asthma outgrow the disease in adolescence and
allergen-specic. suggest that complete remission of the disease may be the
Recent epidemiological studies have challenged the exception rather than the rule, at least among cases of mod-
view of a unidirectional link between atopy and asthma. erate to severe asthma.
Our group found that parental asthma is a strong predictor This link between severity and persistence of asthma
not only for asthma in the child (as one would expect), but has been conrmed by long-term prospective studies that
also for the childs total IgE levels and skin test reactivity have followed cohorts of children into mid-adult life. The
[32]. These relationships remained signicant after adjusting longest such ongoing study is that initiated by Williams and
for the intensity and other characteristics of parental atopy, McNicol in Melbourne, Australia [39]. Children with a his-
supporting independent eects of parental asthma on the tory of recurrent episodes of wheezing were enrolled at the
development of atopy in the child. Previous studies had also age of 7 years, together with a small group of controls with-
shown that, although the level of allergen exposure early in out such a history. These individuals were then periodically
life predicts the development of allergic sensitization and reassessed, and the latest published data refer to information
allergic sensitization is associated with subsequent develop- collected when their mean age was 42 years. Information
ment of asthma, no direct link between allergen exposure about the age upon initiation of symptoms was obtained
early in life and subsequent development of asthma could by use of questionnaires and, as a consequence, data on a
be demonstrated. crucial period of life for the development of asthma was
All these studies suggest that the essential causal possibly biased by preferential recall [36, 40]. The sec-
mechanism associated with asthma is not sensitization to ond study was the British 1958 birth cohort, in which
any specic allergen, but more likely is an alteration in the over 18,000 subjects born between March 3 and March 9,
regulation of responses to many dierent antigens with 1958 were enrolled [8]. Of these persons, 31% contributed
the potential of eliciting IgE responses, and especially to information for ages 7, 11, 16, 23, and 33 years. As with
aeroallergens. IFN production might be among these the Melbourne study, information for the rst 7 years of
altered immunological responses. In the Tucson cohorts of life was obtained retrospectively at age 7. Similarly, in the
the Childrens Respiratory Study [33, 34] and the Infant Dunedin Multidisciplinary Health and Development Study
Immune Study [35], our group found strong associations (New Zealand) parents completed rst respiratory ques-
between reduced IFN production from polyclonally stimu- tionnaires when children were already 9 years old. To date,
lated peripheral blood mononuclear cells in the rst year of participants have been followed up to age 26 with repeated
life and the subsequent development of both allergic sensi- questionnaires and lung function measurements [7]. Finally,
tization and asthma-related respiratory symptoms. Reduced the Tasmanian asthma survey enrolled over 8000 chil-
IFN responses may be related to a delayed maturation of dren born in 1961 who were also rst contacted at age 7

26
Natural History 3
[10]. Two thousand randomly chosen individuals were lung function as measured in early school age was a sig-
re-examined when they were 2932 years of age. nicant predictor of subsequent persistent wheezing. In the
The most important ndings of all four studies were Melbourne study, lung function for each subgroup of sub-
that asthma remits, at least transiently, in early adulthood jects classied according to their wheezing history by age 7
in a sizable proportion of asthmatic children, and that the was repeatedly assessed up to the age of 42 years. Children
severity of asthma tracks signicantly with age. This second with severe asthma at age 10 had very low initial levels of
point is important because it reects a stability in the dis- lung function, and this was to be expected because decits
ease: most children with severe symptoms still have severe in lung function were used to classify them as having severe
symptoms as adults, and asthmatic children with mild symp- asthma in the rst place. When compared with their peers,
toms either have no asthma or have mild asthma as adults. these children did not show further decits in lung func-
tion growth with age, and by age 42 their position relative
Children enrolled in the Melbourne study were divided to subjects with milder symptoms or with no asthma was
at the time of enrollment into ve groups according to substantially unchanged. A similar pattern of relatively sta-
their previous history of wheezing: a control group (no ble lung function growth was observed for patients with
wheezing), a group with mild wheezy bronchitis (less mild asthma who had shown loss of lung function at enroll-
than ve lifetime episodes of wheezing associated with ment, albeit much less pronounced than persons with severe
colds), a group with wheezy bronchitis (ve or more such asthma. Similarly, in the Dunedin study children with per-
episodes), a group with asthma (wheezing apart from sistent wheezing had consistently lower values of the FEV1/
colds), and a group with severe asthma (selected at age FVC ratio from age 9 up to age 26, as compared with con-
10 based on severe impairment of lung function). At age trols. The mean decit was almost 7%, but no signicant
42 years, less than a quarter of children with either form dierences in the slopes of change in the FEV1/FVC ratio
of wheezy bronchitis showed frequent asthma episodes were found between persistent wheezers and controls. Thus,
(wheezing during the previous 3 months, but less than persistent wheezers had decreased values of FEV1/FVC, but
once a week) or persistent asthma (once a week or more). not steeper slopes of decline. These ndings are very rele-
However, 52% of children with asthma and 76% of vant for our understanding of the natural history of COPD,
those with severe asthma at the time of enrollment had and will be discussed in more detail below.
frequent or persistent wheeze at age 42 [40]. The potential role of bronchial hyperresponsiveness in
Similar results have been reported based on the British school age has been explored in two of the above referenced
1958 study: 27% of all children whose parents reported cohorts. In the Dunedin study, the presence at 9 years of
they had wheezed before age 7 reported wheezing during age of either a value of methacholine PC20  8 mg/ml or
the previous year at age 33 [8]. an increase of FEV1  10% from baseline in response to a
bronchodilator was associated with fourfold increased odds
In the Dunedin study, 37% of children who ever for persistent wheezing and with almost sevenfold increased
wheezed had wheezing that persisted from childhood to odds for relapse of wheezing [7]. In that study, bronchial
adulthood or that relapsed after remission [7]. hyperresponsiveness remained signicantly associated with
For the Tasmanian study, 25.6% of subjects with asthma both outcomes after adjusting for other risk factors. In the
or wheezy breathing by age 7 reported current asthma Tucson Childrens Respiratory Study, we found 68% of the
at the age of 2932 years, compared with only 10.8% of children whose asthma persisted after the onset of puberty
subjects without parental reports of childhood asthma to have had a positive test for bronchial hyperresponsiveness
[10]. As with the Melbourne study, those who had a at age 11, as compared with only 37% of children whose
history of more than 10 attacks of asthma by age 7 were asthma remitted during adolescence [12]. Another interest-
almost twice as likely to have persistent wheezing as ing nding of that study was the strong association between
adults than those who did not. childhood obesity and persistence of asthma after the onset
of puberty. Multiple recent reports have consistently linked
A consistent factor associated with persistent asthma obesity not only to persistence but also to incidence of
in all four cohorts was evidence of an allergic predisposition. asthma [4144]. The nature of this association remains at
In the Melbourne study, severe asthma in early life was present not fully understood and warrants further research.
associated with signicantly higher prevalence of allergic
rhinitis at age 35. In the British cohort, allergic symptoms
(e.g. allergic rhinitis or eczema) were signicantly associ- Relapse of asthma symptoms in patients
ated with persistence of symptoms into adult life. In the whose asthma remitted in childhood
Dunedin cohort, skin sensitization to house dust mites
increased more than twice the odds for both persistence and Little is known about prevalence and risk factors for relapses
relapse of wheezing, independent of other predisposing fac- of asthma in adult life among subjects whose symptoms
tors [7]. Finally, in the Tasmanian cohort, having a history remitted during childhood. Taylor and colleagues found
of eczema in early life was also signicantly associated with one-third of subjects who had childhood asthma that was in
persistent wheezing at ages 2932. remission at age 18 to have relapses of the disease in young
Childhood decits in lung function are another fac- adulthood [45]. In the British cohort study, a group of over
tor that has been consistently associated with persistence 1300 persons with a history of wheezing illnesses from
of asthma into adult life across dierent cohorts. In the birth to age 16 years had symptom remission by age 23. This
Tasmanian, the Melbourne, and the Dunedin studies, low group was 50% more likely to report wheezing at age 33

27
Asthma and COPD: Basic Mechanisms and Clinical Management

than those with no history of asthma or wheezy bronchitis 100

FEV1 (% normal level at age 20)


during childhood [8]. These ndings suggest that complete (c)
remission of asthma in adulthood may be less common (a)
80 (d)
than previously thought and are consistent with results
from clinical studies supporting the presence of active,
subclinical forms of the disease among asthmatics in clini- 60
cal remission. In a Dutch cohort of 119 patients who were (b)
diagnosed with asthma during school age and re-examined 40
30 years later (at age 3242), bronchial hyperresponsiveness
and/or reduced lung function were present among 57% of 20
those who were in clinical remission as adults [46]. Previous
studies have shown that indicators of airway inammation,
such as eosinophils and IL-5 in bronchial biopsies, eosi- 0
0 10 20 30 40 50 60 70 80
nophil percent in bronchoalveolar lavage, and exhaled nitric Age (years)
oxide levels, are higher among children and adolescents in
clinical remission of asthma, as compared with their peer FIG. 3.4 Hypothetical mechanisms that may lead to a critically low level
controls [4749]. Bronchial hyperresponsiveness and other of lung function in adult life and to chronic airway obstruction (horizontal
clinical abnormalities among these apparently remitting line): (a) normal growth and decline; (b) impaired lung growth with a lower
asthma cases may pose these subjects at increased risk for plateau phase but a normal rate of decline compared to (a); (c) normal
obstructive lung disease in adult life and their potential role plateau with rapid initial decline in lung function and a subsequent normal
as determinants of COPD will be discussed below. rate of decline; (d) normal plateau with normal initial rate of decline but a
subsequent accelerated loss in lung function. Adapted from Ref. [51].

NATURAL HISTORY OF COPD of lung function at the beginning of adult life as in individ-
uals who already have a diminished level of lung function to
begin with.
According to the Global Initiative for Chronic Obstructive The essential determinants of the natural history of
Lung Disease (GOLD) [50], the pulmonary component COPD are the rate of growth of lung function up to late
of COPD is characterized by airow limitation that is not childhood, the timing and length of the plateau phase, and
fully reversible, usually progressive, and associated with the rate of decline of lung function during adult life.
an abnormal inammatory response of the lung to nox-
ious particles or gases. Thus, the natural history of COPD
overlaps extensively with that of the level of lung function
(usually FEV1, expressed as percent of predicted value in Growth of lung function during childhood
relation to a certain height) during adulthood. Three are the
main factors that can determine the level of lung function The three most important determinants of growth of lung
achieved at any age during adult life (Fig. 3.4). function during childhood are:
First, the individual may either start life with a low level the level of lung function at birth,
of lung function or show a signicant decline in lung func-
tion growth during the rst years of life (line b in Fig. 3.4). the incidence of lower respiratory illnesses during the
The individuals level of lung function will fall after a certain rst years of life,
age, at the same rate as normal peers, but at a lower level the development of persistent asthma-like symptoms
overall. The level of lung function attained by late adoles- during childhood.
cence or early adult life will thus be lower. Through the nor-
mal process of aging, decline in lung function will naturally
occur, and this decline, although parallel to that occurring Level of lung function at birth
in all the population, will nevertheless predispose to the The availability of lung function tests that can be performed
attainment of a level of lung function (expressed as percent- shortly after birth, and which appear to show good correlation
age of normal level in early adulthood) that will be lower with more invasive tests of airway function [52], has allowed
than the threshold for the expression of clinical symptoms us to study the potential role of the development of the lung
of airway obstruction. in utero on subsequent levels of lung and airway function
A second potential mechanism is that of an early attained by the individual. Since these methods have been
decline in lung function occurring either at the end of ado- available only for the last 25 years, it is not possible to deter-
lescence or in early adult life (line c in Fig. 3.4). Again, the mine the role of airway function in the postnatal period on
main result of such a mechanism would be the attainment events occurring beyond young adult life. Nevertheless, it is
of lower level of lung function in these individuals as com- now apparent that there is quite signicant tracking between
pared with the rest of the population. the level of lung function measured shortly after birth and
A third mechanism is that of a faster rate of decline that measured during childhood [53, 54], adolescence [22],
of lung function during adult life (line d in Fig. 3.4). This and up to young adulthood [55]. This may explain, at least in
may obviously occur both in individuals with a normal level part, the strong association between lower respiratory illness

28
Natural History 3
in early life and level of lung function in late childhood and of lung function [63]. This hypothesis seemed attractive,
early adulthood (see the discussion below). because it suggested a potential strategy for the prevention
The factors that determine growth of lung and air- of early losses in lung function that could predispose to
ways in utero are only partly understood. The two main COPD. However, several studies in which the techniques
determinants of the level of lung function measured shortly to assess lung function in infants, described earlier, were
after birth are gender and exposure to tobacco smoke prod- used showed that children who developed lower respira-
ucts in utero [56]. Boys have been found to have consist- tory symptoms during viral infections in early life had
ently lower maximal ows at functional residual capacity diminished pre-illness levels of lung function [18, 56, 59].
(VmaxFRC) throughout the rst year of life in comparison The hypothesis was thus suggested that lower levels of lung
with girls. Indeed, females have larger airway size, rela- function observed after lower respiratory illnesses in early
tive to the size of lungs and to body size, as compared with life could be explained by preexisting diminished lung func-
males both at birth and during childhood, up to the plateau tion, the latter being therefore the link between early life
of lung growth [57]. However, the role of these gender dif- episodes of airway obstruction and subsequent decits in
ferences in inuencing airway function and predisposing to lung function.
obstructive lung disease is not well understood. Maternal Unfortunately, the number of infants in whom lung
smoking during pregnancy is associated with lower levels function has been ascertained in early life and who have
of VmaxFRC in both genders in comparison with unexposed been followed for a number of years after birth is rather
infants [56]. Postnatal exposure to tobacco smoke appears small. Therefore, the possibility that lower respiratory ill-
to be less important than prenatal exposure, at least in most nesses by themselves may alter lung and airway growth in
developed countries [58]. The eects of intrauterine expo- groups of susceptible individuals cannot be excluded. It is
sure to tobacco smoke products persist for the rst year unlikely that pre-illness lung function may explain all forms
of life [56], and even beyond the rst year up to the early of infection-associated wheezing during early childhood,
adolescent years [59]. and it is legitimate to surmise that immune responses to the
Independent of what factors determine in utero lung viruses themselves may also play a signicant role.
growth, evidence has been accumulating that lung func- Evidence from studies in the United Kingdom would
tion at birth aects lung health far beyond infancy. In addi- support the validity of both hypotheses [6466]. In a cohort
tion to the well-established association between reduced of children enrolled in 1964 and reassessed in 2001 for cur-
VmaxFRC shortly after birth and wheezing in the rst years rent lung function, smoking status, and respiratory symp-
of life [18, 56, 59, 60], recent studies have associated lung toms, a signicant linear trend was found between birth
function measured in the rst month of life to persistent weight and FEV1 at age 4550 years after adjusting for
wheezing and asthma at 1011 years [54, 61]. Turner et al. confounders, including smoking [65]. This association was
found wheezing at 11 years to be associated with a reduced conrmed by a recent meta-analysis [66] of eight stud-
mean z score for VmaxFRC at 1 month of age [54], whereas a ies reporting a pooled increase of 48 ml in FEV1 per 1 kg
reduced fraction of expiratory time to peak tidal expiratory in birth weight. As modest as this association may appear,
ow to total expiratory time (tPTEF/tE) as assessed 3 days these ndings support a direct inuence of in utero growth
after birth predicted asthma and bronchial hyperrespon- on lung function in adulthood. However, another very long-
siveness at age 10 years in a Norwegian birth cohort [61]. term study in England provides evidence in support of addi-
Of note, in the latter study tPTEF/tE at birth did not cor- tional eects of childhood respiratory infections on adult
relate with FEV1 or FVC at 10 years of age. These ndings lung function [64]. In this study, men born between 1911
are complemented by results from the Tucson Childrens and 1930, whose birth weights, weights at 1 year, and child-
Respiratory Study, in which we found VmaxFRC measured hood respiratory illnesses were recorded in early life, were
at 2 month of age to correlate strongly with FEV1 and the studied at ages 5970 years. Death from chronic obstruc-
FEV1/FVC ratio assessed at age 11, 16, and 22 years [55]. tive disease, FEV1, and respiratory symptoms were the main
Participants in the lowest quartile for infant VmaxFRC had outcome variables. The main early life determinants of the
a 5% mean decit in the FEV1/FVC ratio through age 22 level of FEV1 in old age were birth weight and history of
compared to the upper three quartiles and this association bronchitis or pneumonia in infancy, and these eects were
was independent of wheezing and asthma status. Taken independent of smoking habit and social class. These data
together, this evidence suggests that lung function at birth would thus suggest that both intrauterine growth (and
accounts for part of the lung function levels attained in presumably lung development) and lower respiratory ill-
young adult life and, in turn, may inuence predisposition nesses during the rst years of life exert independent eects
to subsequent obstructive lung disease. on the level of lung function attained late during adult
life, and thus may be important determinants of the risk
of COPD.
Role of lower respiratory illnesses in early life
It has been known for decades that children who have
lower respiratory illnesses during the rst years of life have Persistent asthma-like symptoms during
lower levels of lung function during childhood and into childhood
adult life [62]. One possible explanation for this association Epidemiological evidence supports that persistent asthma-
is that viral infections, which are the main etiological fac- like symptoms are a signicant risk factor for the devel-
tor for lower respiratory illnesses in early life, may damage opment of lower levels of lung function in childhood and
lung and airways, and this may predispose for lower levels young adult life [7, 36] and, in turn, may predispose to the

29
Asthma and COPD: Basic Mechanisms and Clinical Management

development of COPD. One proposed explanation for of lung function is reached in early adult life. Xuan et al.
these associations has been that asthma is a progressive [72] observed that, between the ages of 17 and 19 years,
disease [67]. It has thus been proposed that the presence of when growth in height had stopped, FEV1 continued to
chronic airway inammation is associated with signicant grow in both males and females. However, children who
lung remodeling and that the latter fosters a signicant had recent episodes of wheeze and those with evidence of
alteration in lung growth. bronchial hyperresponsiveness showed a reduced rate of
Although some studies were able to show impair- growth in airway caliber. Unfortunately, lung function was
ment in the development of lung function in children not assessed after use of a bronchodilator, and it is thus not
with asthma [67, 68], the main outcome variable used in possible from these data to assess whether the changes are
these studies was pre-bronchodilator lung function. The due to increased airway tone or to irreversible alterations
Childhood Asthma Management Program (CAMP) study in airway structure. In addition to active wheezing and
was specically designed to test the hypothesis that mild to bronchial hyperresponsiveness, early initiation to cigarette
moderate childhood asthma is associated with signicant smoking has been associated with detrimental eects on the
deterioration in airway growth and that treatment with plateau level of lung function. Holberg et al. [73] showed
inhaled anti-inammatory therapy could reverse this dete- that, at age 16, the level of lung function achieved by males
rioration in lung function [69]. Children aged 612 years who had started smoking was signicantly lower than
were treated for 46 years with either an inhaled corticos- that of those who had not. No such eect was observed in
teroid (budesonide), nedocromil, or a placebo. Post-bron- females.
chodilator FEV1 was considered the outcome variable,
because pre-bronchodilator FEV1 could be aected by the
degree of activity of the disease at the time of testing. The Determinants of increased slope of lung
results of the study showed that, although children with function decline
asthma have levels of lung function that are signicantly
lower than those of children without the disease at any time Several longitudinal studies have addressed the role of dif-
between the ages of 6 and 15, these levels do not further ferent intrinsic and extrinsic factors on the rate of decline
deteriorate even among subjects who are not systematically of lung function after the plateau phase. The role of ciga-
treated with anti-inammatory therapy. Moreover, system- rette smoking has been clearly and consistently established.
atic treatment with anti-inammatory therapy improves Camilli et al. [74] examined changes in FEV1 in over 1700
bronchial hyperresponsiveness, but is not associated with a adults enrolled in a prospective study of a general popula-
signicant improvement in lung function with time. Based tion sample. Individuals who smoked more than 10 ciga-
on these ndings, more recently the Prevention of Early rettes per day had excessive rates of decline in FEV1 as
Asthma in Kids (PEAK) trial was designed to determine compared with nonsmokers. The excess decline of smokers
whether the lack of sustained eects of inhaled corticoster- was age dependent, particularly in men: much of the excess
oids on the natural history of childhood asthma was attrib- loss of lung function occurred between 50 and 70 years of
utable to the initiation of the intervention too late in life. In age. Interestingly, ex-smokers showed declines in FEV1 val-
fact, the PEAK trial failed to detect any long-term eects ues that were similar to those of nonsmokers. The authors
of a 2-year treatment with inhaled corticosteroids on the also examined the eect of quitting smoking on the decline
natural history of asthma in a group of high-risk preschool of FEV1. In subjects younger than 35 years, quitting smok-
children, despite the temporary benecial eects of the ing during follow-up was associated with an actual increase
treatment on disease symptoms and exacerbations [70]. in FEV1. In men above 50 years, smoking cessation early in
An interpretation of these results is that, in chil- the study led to a return to normal rate of functional decline
dren with a diagnosis of asthma, most of the deterioration during follow-up. In addition to cigarette smoking, exposure
in lung function observed during the school years occurs to other toxic gases and particles including environmental
early in life. The results of studies by our group suggest that tobacco smoke, indoor and outdoor pollution, occupational
children who go on to have persistent asthma-like symp- dust, gases, and fumes has been consistently associated
toms during childhood start life with levels of lung func- with COPD and a steeper decline of lung function [75, 76].
tion that are slightly lower than those of children who will However, the observation that only a more [77] or less
not have these persistent symptoms [71]. It is also possi- [78] sizable fraction of smokers develop COPD strongly
ble that persistent childhood asthma aects lung function suggests the existence of intrinsic factors that can modify
through mechanisms that are at least partly unrelated to the eect of smoking and other deleterious exposures on
airway inammation and, therefore, insensitive to corticos- the rate of decline of lung function. Bronchial hyperre-
teroid treatment. Finding the answer to these questions will sponsiveness has been long hypothesized to be one of these
have crucial implications for the prevention of long-term factors [79, 80]. Indeed, bronchial hyperresponsiveness has
obstructive lung disease among these children. been consistently shown to be a strong and independent
risk factor for accelerated decline of lung function in pro-
spective studies among smokers with early COPD as well
Determinants of early losses in lung as asymptomatic individuals [8183]. Findings from these
function epidemiological studies have been conrmed in the clinical
setting. For instance, Postma and coworkers [84] assessed
Very little is known about the factors that determine early the course of lung function after 221 years of follow-up
losses in lung function at the age in which the plateau level in 81 nonallergic patients with considerable lung function

30
Natural History 3
impairment (55% FEV1/FVC ratio) at the beginning of
the study. They reported that a more favorable rate of change INTERSECTION OF THE NATURAL HISTORY OF
in FEV1 was not only associated with fewer pack-years of ASTHMA AND COPD
smoking but also with less nonspecic bronchial hyperreac-
tivity and a higher degree of reversibility of airow obstruc- It is not uncommon in the clinical setting to observe
tion. These eects were independent of baseline FEV1 value, patients with asthma showing COPD-like phenotypes,
both in smokers and in ex-smokers. Whether the association and vice versa. Consistently, it has been long known that
between bronchial hyperresponsiveness and decline in lung diagnoses of asthma, chronic bronchitis, and emphysema
function is the consequence of an ongoing COPD-related are frequently associated at the population level [2]. This
inammatory process and to what extent this inammatory evidence indicates that the natural history of asthma and
process diers from that associated with bronchial hyperre- COPD can converge in some cases during adult life. A pos-
sponsiveness in asthma remain at present open questions. sible explanation for these observations is that a signicant
Airway inammation and its structural sequelae may proportion of cases with severe and/or persistent asthma can
be also involved in the causal mechanisms linking acceler- develop in the long-term COPD-like phenotypes. Support
ated decline of lung function in smokers with two other risk for this contention is provided by another report from the
factors: chronic bronchitis (i.e. chronic cough and phlegm) TESAOD study [19], in which adult subjects with active
and lower airway colonization/infection. Although the pre- asthma were found to have a 12 times higher risk of acquir-
dictive value of chronic bronchitis for the development of ing a diagnosis of COPD over time than subjects with no
COPD remains somewhat controversial [85, 86], this phe- asthma, after adjusting for covariates including smoking.
notype has been consistently associated with steeper rates of Since chronic airow limitation is the central hall-
FEV1 decline [87, 88]. It has been suggested that the clini- mark of COPD, much attention has been paid to under-
cal impact of chronic bronchitis may be dependent upon stand the natural history of lung function of patients who
the stage of COPD, with this phenotype having stronger develop COPD as a sequela of asthma. Much like for indi-
eects on the severe forms of COPD when lower airways viduals from the general population (Fig. 3.4), in patients
become colonized and/or infected [89]. Acute exacerbations with asthma FEV1 decits can develop mainly as a result
and lower airway colonization are strongly associated with of either lower FEV1 levels at the beginning of adult age
decline of lung function among smokers and patients with or an accelerated FEV1 decline during adulthood [51].
COPD. In a prospective study [90], patients with frequent Interestingly, most birth cohort studies including the
exacerbations (mean number of episodes per year 2.9) had Dunedin Multidisciplinary Health and Development Study
a signicantly faster decline of FEV1 than infrequent exac- [7], the Melbourne Asthma Study [36], and the British
erbators. Lower respiratory illnesses had a similar deleteri- 1958 Birth Cohort [94] have shown that, in a signicant
ous eect on decline of FEV1 over 5 years among smokers proportion of cases of persistent childhood asthma, lung
in the Lung Health Study [91]. function decits are established before the early adult years,
Of note, airway infections may also aect lung func- and track over time. These ndings, together with the obser-
tion decline through mechanisms that are independent of vation that, among children who start life with low levels
(and may indeed precede) the presence of COPD. Burrows of lung function, expiratory ows remain lower than those
and coworkers reported that smokers who recalled a history of their peers throughout childhood and adolescence [22],
of respiratory trouble before 16 years of age had signi- point toward the importance of early events in aecting
cantly steeper rates of decline in lung function as compared lung growth and/or airway remodeling in childhood and, in
with those with no such history [92]. This eect was inde- turn, in inuencing early predisposition to COPD. As for
pendent of a current or past diagnosis of asthma. It thus the second potential mechanism of lung function impair-
appears that intrinsic factors that modify the eects of ment (i.e. accelerated decline of lung function) in asthmat-
smoking may be related to events occurring during the rst ics, prospective cohort studies on adult populations have
years of life. This conclusion is compatible with the obser- provided somewhat inconsistent ndings. In the Busselton
vation by Barker and coworkers quoted earlier [64], that Health Study [95] and in the Copenhagen City Heart
elderly individuals with a history of lower respiratory illness Study [96], asthmatics showed both initial FEV1 decits
early in life were more likely to have lower levels of lung at age 20 years and an increased slope of FEV1 decline in
function than those with no such history. adulthood. However, the increase in FEV1 decline associ-
Although it is not well established how the eects of ated with asthma was only 4 ml/year in the Busselton Study.
these intrinsic and extrinsic factors on the rate of decline A possible reconciliation for these inconsistent results
of lung function are modulated, genetics are likely to play a is provided by a recent study from the TESAOD cohort.
major role. In the large cohort of the Tucson Epidemiological We identied adult participants who had FEV1/FVC ratio
Study of Airway Obstructive Disease (TESAOD), our consistently lower than 70% (as a hallmark of persistent air-
group observed signicant intra-family correlation in the ow limitation) and compared the natural history of their
rate of decline of lung function within smokers [93], sug- lung function based on the presence of asthma. Patients
gesting that these rates of decline have an important genetic with asthma accounted for about one-third of all cases of
component. However, the identication of the genetic com- persistent airow limitation. Persistent airow limitation
ponents of COPD, as well as of those of asthma, is proving was strongly associated with smoking among nonasthmat-
a challenging task and at present deciency of 1-antit- ics and with eosinophilia among subjects who had asthma
rypsin remains the only established genetic risk factor for onset  25 years. Most importantly, the natural course of
this disease.

31
Asthma and COPD: Basic Mechanisms and Clinical Management

FIG. 3.5 Natural history of lung function among participants


in the TESAOD study who had persistent airflow limitation
with or without asthma. The black line refers to subjects with
no asthma and persistent airflow limitation. The red line refers
to subjects with asthma onset  25 years and persistent
airflow limitation. The gold line refers to subjects with asthma
onset 25 years and persistent airflow limitation. Predicted
values for subjects with no asthma and no airflow limitation
(healthy controls, green line) are also reported for comparison.
Depicted values represent predicted values for a 175-cm
tall male from the best-fitting random coefficients model.
Adapted from Ref. [96].

lung function diered substantially between the two groups of their lung function impairment is already established.
(Fig. 3.5), with subjects with asthma onset  25 years Similarly, the dierent trajectories by which patients with
having lower FEV1 levels at age 25 but not steeper FEV1 traditional COPD and patients with asthma may develop
decline in adulthood as compared with expected values chronic airow limitation in their adult life support the phe-
from healthy controls. In contrast, subjects who developed notypic heterogeneity that has emerged from clinical stud-
persistent airow limitation but had no asthma showed only ies on these two groups [97]. Future research is required to
moderate FEV1 decits at age 25 years but had greater than determine whether multiple functional, morphological, and
expected FEV1 loss between age 25 and 75. Interestingly, immunological assessments can be developed to capture the
the natural course of lung function of subjects who had heterogeneity of these diseases and to identify optimal pre-
adult-onset asthma (after age 25 years) included both mod- vention and treatment strategies for these patients.
erate FEV1 decits in young adulthood and accelerated
FEV1 decline thereafter.
Taken together, ndings from this and previous stud- References
ies suggest that: (1) asthma accounts for a sizable fraction
of COPD cases at the population level; (2) while develop- 1. Burrows B, Bloom JW, Traver GA, Cline MG. The course and progno-
ment of traditional COPD is strongly associated with expo- sis of dierent forms of chronic airways obstruction in a sample from
sure to noxious agents (e.g. cigarette smoking), patients with the general population. N Engl J Med 317(21): 130914, 1987.
asthma may develop persistent airow limitation through 2. Soriano JB, Davis KJ, Coleman B, Visick G, Mannino D, Pride NB.
pathways that are independent of smoking and related to The proportional Venn diagram of obstructive lung disease: Two
allergic and eosinophilic inammation; (3) among sub- approximations from the United States and the United Kingdom.
jects who had severe and/or persistent asthma in childhood Chest 124(2): 47481, 2003.
signicant decits in lung function are already present in 3. Silva GE, Sherrill DL, Guerra S, Barbee RA. Asthma as a risk factor
early adult life; (4) during adulthood accelerated decline of for COPD in a longitudinal study. Chest 126(1): 5965, 2004.
4. Pauwels RA, Buist AS, Calverley PM, Jenkin CR, Hurd SS. Global
FEV1 might be present for subgroups of asthmatics, but this
strategy for the diagnosis, management, and prevention of chronic
accelerated decline is likely to account for a limited propor- obstructive pulmonary disease. NHLBI/WHO Global Initiative for
tion of the asthma-related decits in lung function up to Chronic Obstructive Lung Disease (GOLD) Workshop summary.
mid-adult life; (5) childhood versus adult-onset asthma may Am J Respir Crit Care Med 163(5): 125676, 2001.
be associated with a dierent natural history of lung func- 5. Martinez FD. Context dependency of markers of disease. Am J Respir
tion that might explain part of the inconsistencies of results Crit Care Med 162(2 Pt 2): S56S57, 2000.
on FEV1 decline in asthmatics across dierent studies. 6. Barbee RA, Dodge R, Lebowitz ML, Burrows B. The epidemiology of
The implications of these conclusions for prevention asthma. Chest 87(1 Suppl): 21S25S, 1985.
and treatment are crucial because they indicate that the tim- 7. Sears MR, Greene JM, Willan AR et al. A longitudinal, population-
based, cohort study of childhood asthma followed to adulthood. N Engl
ing and nature of prevention strategies for COPD-like phe-
J Med 349(15): 141422, 2003.
notypes need to be dierent for subjects with and without 8. Strachan DP, Butland BK, Anderson HR. Incidence and prognosis
asthma. While smoking prevention and cessation is vital of asthma and wheezing illness from early childhood to age 33 in a
against inception of traditional COPD, any intervention to national British cohort. BMJ 312(7040): 119599, 1996.
prevent development of persistent airow limitation among 9. Oswald H, Phelan PD, Lanigan A, Hibbert M, Bowes G, Olinsky A.
patients with childhood asthma will need to target these Outcome of childhood asthma in mid-adult life. BMJ 309(6947):
patients before they enter adult life because by then the bulk 9596, 1994.

32
Natural History 3
10. Jenkins MA, Hopper JL, Bowes G, Carlin JB, Flander LB, Giles most province of Sweden: Schools as a major site of exposure. J Allergy
GG. Factors in childhood as predictors of asthma in adult life. BMJ Clin Immunol 103(6): 101824, 1999.
309(6947): 9093, 1994. 32. Crestani E, Guerra S, Wright AL, Halonen M, Martinez FD. Parental
11. Anderson HR, Pottier AC, Strachan DP. Asthma from birth to age 23: asthma as a risk factor for the development of early skin test sensitiza-
Incidence and relation to prior and concurrent atopic disease. Thorax tion in children. J Allergy Clin Immunol 113(2): 28490, 2004.
47(7): 53742, 1992. 33. Martinez FD, Stern DA, Wright AL, Holberg CJ, Taussig LM,
12. Guerra S, Wright AL, Morgan WJ, Sherrill DL, Holberg CJ, Martinez Halonen M. Association of interleukin-2 and interferon-gamma pro-
FD. Persistence of asthma symptoms during adolescence: Role of obes- duction by blood mononuclear cells in infancy with parental allergy
ity and age at the onset of puberty. Am J Respir Crit Care Med 170(1): skin tests and with subsequent development of atopy. J Allergy Clin
7885, 2004. Immunol 96(5 Pt 1): 65260, 1995.
13. Yunginger JW, Reed CE, OConnell EJ, Melton LJ III, OFallon WM, 34. Stern DA, Guerra S, Halonen M, Wright AL, Martinez FD. Low
Silverstein MD. A community-based study of the epidemiology of asthma. IFN-gamma production in the rst year of life as a predictor of wheeze
Incidence rates, 19641983. Am Rev Respir Dis 146(4): 88894, 1992. during childhood. J Allergy Clin Immunol 120(4):83541.
14. Stein RT, Sherrill D, Morgan WJ et al. Respiratory syncytial virus 35. Guerra S, Lohman IC, Halonen M, Martinez FD, Wright AL.
in early life and risk of wheeze and allergy by age 13 years. Lancet Reduced interferon gamma production and soluble CD14 levels in
354(9178): 54145, 1999. early life predict recurrent wheezing by 1 year of age. Am J Respir Crit
15. Stein RT, Holberg CJ, Morgan WJ et al. Peak ow variability, metha- Care Med 169(1): 7076, 2004.
choline responsiveness and atopy as markers for detecting dierent 36. Phelan PD, Robertson CF, Olinsky A. The Melbourne Asthma Study:
wheezing phenotypes in childhood. Thorax 52(11): 94652, 1997. 19641999. J Allergy Clin Immunol 109(2): 18994, 2002.
16. Martinez FD, Helms PJ. Types of asthma and wheezing. Eur Respir J 37. Horak E, Lanigan A, Roberts M et al. Longitudinal study of childhood
Suppl 27: 3s8s, 1998. wheezy bronchitis and asthma: Outcome at age 42. BMJ 326(7386):
17. Tager IB, Ngo L, Hanrahan JP. Maternal smoking during pregnancy. 42223, 2003.
Eects on lung function during the rst 18 months of life. Am J Respir 38. Marossy AE, Strachan DP, Rudnicka AR, Anderson HR. Childhood
Crit Care Med 152(3): 97783, 1995. chest illness and the rate of decline of adult lung function between ages
18. Martinez FD, Morgan WJ, Wright AL, Holberg CJ, Taussig LM. 35 and 45 years. Am J Respir Crit Care Med 175(4): 35559, 2007.
Diminished lung function as a predisposing factor for wheezing respi- 39. Williams H, McNicol KN. Prevalence, natural history, and relationship
ratory illness in infants. N Engl J Med 319(17): 111217, 1988. of wheezy bronchitis and asthma in children. An epidemiological study.
19. Martinez FD. Sudden infant death syndrome and small airway occlu- BMJ 4(5679): 32125, 1969.
sion: Facts and a hypothesis. Pediatrics 87(2): 19098, 1991. 40. Phelan PD. Asthma in Children and Adolescents: An Overview. .
20. Montgomery GL, Tepper RS. Changes in airway reactivity with age London: Baillire Tindall, 1995.
in normal infants and young children. Am Rev Respir Dis 142(6 Pt 1): 41. Camargo CA Jr., Weiss ST, Zhang S, Willett WC, Speizer FE.
137276, 1990. Prospective study of body mass index, weight change, and risk of
21. Burrows B, Sears MR, Flannery EM, Herbison GP, Holdaway MD, adult-onset asthma in women [see comments]. Arch Intern Med
Silva PA. Relation of the course of bronchial responsiveness from age 159(21): 258288, 1999.
9 to age 15 to allergy. Am J Respir Crit Care Med 152(4 Pt 1): 13028, 42. Castro-Rodriguez JA, Holberg CJ, Morgan WJ, Wright AL, Martinez
1995. FD. Increased incidence of asthmalike symptoms in girls who become
22. Morgan WJ, Stern DA, Sherrill DL et al. Outcome of asthma and overweight or obese during the school years. Am J Respir Crit Care Med
wheezing in the rst 6 years of life: Follow-up through adolescence. 163(6): 134449, 2001.
Am J Respir Crit Care Med 172(10): 125358, 2005. 43. Beuther DA, Sutherland ER. Overweight, obesity, and incident
23. Zeiger RS, Dawson C, Weiss S. Relationships between duration of asthma: A meta-analysis of prospective epidemiologic studies. Am J
asthma and asthma severity among children in the Childhood Asthma Respir Crit Care Med 175(7): 66166, 2007.
Management Program (CAMP). J Allergy Clin Immunol 103(3 Pt 1): 44. Guerra S, Sherrill DL, Bobadilla A, Martinez FD, Barbee RA. The
37687, 1999. relation of body mass index to asthma, chronic bronchitis, and emphy-
24. Halonen M, Stern DA, Lohman C, Wright AL, Brown MA, Martinez sema. Chest 122(4): 125663, 2002.
FD. Two subphenotypes of childhood asthma that dier in maternal 45. Taylor DR, Cowan JO, Greene JM, Willan AR, Sears MR. Asthma
and paternal inuences on asthma risk. Am J Respir Crit Care Med in remission: can relapse in early adulthood be predicted at 18 years of
160(2): 56470, 1999. age? Chest 127(3): 84550, 2005.
25. Halonen M, Stern DA, Wright AL, Taussig LM, Martinez FD. 46. Vonk JM, Postma DS, Boezen HM et al. Childhood factors associated with
Alternaria as a major allergen for asthma in children raised in a desert asthma remission after 30 year follow up. Thorax 59(11): 92529, 2004.
environment. Am J Respir Crit Care Med 155(4): 135661, 1997. 47. van Den Toorn LM, Prins JB, Overbeek SE, Hoogsteden HC,
26. Pullan CR, Hey EN. Wheezing, asthma, and pulmonary dysfunction de Jongste JC. Adolescents in clinical remission of atopic asthma have
10 years after infection with respiratory syncytial virus in infancy. BMJ elevated exhaled nitric oxide levels and bronchial hyperresponsiveness.
(Clin Res Ed) 284(6330): 166569, 1982. Am J Respir Crit Care Med 162(3 Pt 1): 95357, 2000.
27. Burrows B, Martinez FD, Halonen M, Barbee RA, Cline MG. 48. van den Toorn LM, Overbeek SE, de Jongste JC, Leman K,
Association of asthma with serum IgE levels and skin-test reactivity to Hoogsteden HC, Prins JB. Airway inammation is present during
allergens. N Engl J Med 320(5): 27177, 1989. clinical remission of atopic asthma. Am J Respir Crit Care Med 164(11):
28. Peat JK, Tovey E, Toelle BG et al. House dust mite allergens. A major 210713, 2001.
risk factor for childhood asthma in Australia. Am J Respir Crit Care 49. Warke TJ, Fitch PS, Brown V et al. Outgrown asthma does not mean
Med 153(1): 14146, 1996. no airways inammation. Eur Respir J 19(2): 28487, 2002.
29. Dust mite allergens and asthmaa worldwide problem. J Allergy Clin 50. Rabe KF, Hurd S, Anzueto A et al. Global strategy for the diagnosis,
Immunol 83(2 Pt 1): 41627, 1989. management, and prevention of COPD-2006 Update. Am J Respir Crit
30. Peat JK, Tovey E, Mellis CM, Leeder SR, Woolcock AJ. Importance Care Med, 2007.
of house dust mite and Alternaria allergens in childhood asthma: An 51. Weiss ST, Ware JH. Overview of issues in the longitudinal analysis of
epidemiological study in two climatic regions of Australia. Clin Exp respiratory data. Am J Respir Crit Care Med 154(6 Pt 2): S208S211,
Allergy 23(10): 81220, 1993. 1996.
31. Perzanowski MS, Ronmark E, Nold B, Lundback B, Platts-Mills TA. 52. Stocks J. Lung function testing in infants. Pediatr Pulmonol Suppl 18:
Relevance of allergens from cats and dogs to asthma in the northern- 1420, 1999.

33
Asthma and COPD: Basic Mechanisms and Clinical Management

53. Le Souef P, Turner S, Rye P et al. Pulmonary function at four weeks 74. Camilli AE, Burrows B, Knudson RJ, Lyle SK, Lebowitz MD.
correlates with pulmonary function at 6 and 12 years. Am J Respir Crit Longitudinal changes in forced expiratory volume in one second in
Care Med 163: A541, 2001. adults. Eects of smoking and smoking cessation. Am Rev Respir Dis
54. Turner SW, Palmer LJ, Rye PJ et al. The relationship between infant 135(4): 79499, 1987.
airway function, childhood airway responsiveness, and asthma. Am J 75. Pauwels RA, Rabe KF. Burden and clinical features of chronic obstruc-
Respir Crit Care Med 169(8): 92127, 2004. tive pulmonary disease (COPD). Lancet 364(9434): 61320, 2004.
55. Stern DA, Morgan WJ, Wright AL, Guerra S, and Martinez FD. Poor 76. Anto JM, Vermeire P, Vestbo J, Sunyer J. Epidemiology of chronic
aiway function in early infancy and lung function by age 22 years: a non- obstructive pulmonary disease. Eur Respir J 17(5): 98294, 2001.
selective longitudinal cohort study. Lancet 370(9589): 75864, 2007. 77. Rennard SI, Vestbo J. COPD: The dangerous underestimate of 15%.
56. Young S, Arnott J, OKeee PT, Le Souef PN, Landau LI. The associa- Lancet 367(9518): 121619, 2006.
tion between early life lung function and wheezing during the rst 2 78. Fletcher CM, Peto CM, Tinker CM, Spizer FE. The Natural History
yrs of life. Eur Respir J 15(1): 15157, 2000. of Chronic Bronchitis and Emphysema. New York: Oxford University
57. Pagtakhan RD, Bjelland JC, Landau LI et al. Sex dierences in growth Press, 1976.
patterns of the airways and lung parenchyma in children. J Appl Physiol 79. Orie NGM, Sluiter HJ, de Vries K, Tammeling GJ, Witkop J. The
56(5): 120410, 1984. host factor in bronchits. In: Orie NGM, Sluiter HJ (eds). Bronchitis an
58. Stein RT, Holberg CJ, Sherrill D et al. Inuence of parental smoking International Symposium. Assen, Netherlands: Royal vanGorcum, 1961.
on respiratory symptoms during the rst decade of life: The Tucson 80. Sluiter HJ, Koeter GH, de Monchy JG, Postma DS, de Vries K, Orie NG.
Childrens Respiratory Study. Am J Epidemiol 149(11): 103037, The Dutch hypothesis (chronic non-specic lung disease) revisited. Eur
1999. Respir J 4(4): 47989, 1991.
59. Tager IB, Hanrahan JP, Tosteson TD et al. Lung function, pre- and 81. Tashkin DP, Altose MD, Connett JE, Kanner RE, Lee WW, Wise RA.
post-natal smoke exposure, and wheezing in the rst year of life. Methacholine reactivity predicts changes in lung function over time in
Am Rev Respir Dis 147(4): 81117, 1993. smokers with early chronic obstructive pulmonary disease. The Lung
60. Murray CS, Pipis SD, McArdle EC, Lowe LA, Custovic A, Woodcock Health Study Research Group. Am J Respir Crit Care Med 153(6 Pt 1):
A. Lung function at one month of age as a risk factor for infant res- 180211, 1996.
piratory symptoms in a high risk population. Thorax 57(5): 38892, 82. Brutsche MH, Downs SH, Schindler C et al. Bronchial hyperrespon-
2002. siveness and the development of asthma and COPD in asymptomatic
61. Haland G, Carlsen KC, Sandvik L et al. Reduced lung function at individuals: SAPALDIA cohort study. Thorax 61(8): 67177, 2006.
birth and the risk of asthma at 10 years of age. N Engl J Med 355(16): 83. Rijcken B, Schouten JP, Xu X, Rosner B, Weiss ST. Airway hyperre-
168289, 2006. sponsiveness to histamine associated with accelerated decline in FEV1.
62. Burrows B, Knudson RJ, Lebowitz MD. The relationship of childhood Am J Respir Crit Care Med 151(5): 137782, 1995.
respiratory illness to adult obstructive airway disease. Am Rev Respir 84. Postma DS, de Vries K, Koeter GH, Sluiter HJ. Independent inuence
Dis 115(5): 75160, 1977. of reversibility of air-ow obstruction and nonspecic hyperreactivity
63. Samet JM, Tager IB, Speizer FE. The relationship between respiratory on the long-term course of lung function in chronic air-ow obstruc-
illness in childhood and chronic air-ow obstruction in adulthood. tion. Am Rev Respir Dis 134(2): 27680, 1986.
Am Rev Respir Dis 127(4): 50823, 1983. 85. Vestbo J, Lange P. Can GOLD Stage 0 provide information of prog-
64. Barker DJ, Godfrey KM, Fall C, Osmond C, Winter PD, Shaheen SO. nostic value in chronic obstructive pulmonary disease? Am J Respir Crit
Relation of birth weight and childhood respiratory infection to adult Care Med 166(3): 32932, 2002.
lung function and death from chronic obstructive airways disease. BMJ 86. de Marco R, Accordini S, Cerveri T et al. Incidence of chronic obstruc-
303(6804): 67175, 1991. tive pulmonary disease in a cohort of young adults according to the
65. Edwards CA, Osman LM, Godden DJ, Campbell DM, Douglas JG. presence of chronic cough and phlegm. Am J Respir Crit Care Med
Relationship between birth weight and adult lung function: Controlling 175(1): 3239, 2007.
for maternal factors. Thorax 58(12): 106165, 2003. 87. Sherman CB, Xu X, Speizer FE, Ferris BG Jr, Weiss ST, Dockery DW.
66. Lawlor DA, Ebrahim S, Davey Smith G. Association of birth weight Longitudinal lung function decline in subjects with respiratory symp-
with adult lung function: Findings from the British Womens Heart toms. Am Rev Respir Dis 146(4): 85559, 1992.
and Health Study and a meta-analysis. Thorax 60(10): 85158, 2005. 88. Vestbo J, Prescott E, Lange P. Association of chronic mucus hyperse-
67. Peat JK, Woolcock AJ, Cullen K. Rate of decline of lung function in cretion with FEV1 decline and chronic obstructive pulmonary disease
subjects with asthma. Eur J Respir Dis 70(3): 17179, 1987. morbidity. Copenhagen City Heart Study Group. Am J Respir Crit Care
68. Agertoft L, Pedersen S. Eects of long-term treatment with an inhaled Med 153(5): 153035, 1996.
corticosteroid on growth and pulmonary function in asthmatic chil- 89. Vestbo J, Hogg JC. Convergence of the epidemiology and pathology of
dren. Respir Med 88(5): 37381, 1994. COPD. Thorax 61(1): 8688, 2006.
69. Long-term eects of budesonide or nedocromil in children with 90. Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA.
asthma. The Childhood Asthma Management Program Research Relationship between exacerbation frequency and lung function decline
Group. N Engl J Med 343(15): 105463, 2000. in chronic obstructive pulmonary disease. Thorax 57(10): 84752, 2002.
70. Guilbert TW, Morgan WJ, Zeiger RS et al. Long-term inhaled corti- 91. Kanner RE, Anthonisen NR, Connett JE. Lower respiratory illnesses
costeroids in preschool children at high risk for asthma. N Engl J Med promote FEV(1) decline in current smokers but not ex-smokers with
354(19): 198597, 2006. mild chronic obstructive pulmonary disease: results from the lung
71. Martinez FD, Wright AL, Taussig LM, Holberg CJ, Halonen M, health study. Am J Respir Crit Care Med 164(3): 35864, 2001.
Morgan WJ. Asthma and wheezing in the rst six years of life. The 92. Burrows B, Knudson RJ, Cline MG, Lebowitz MD. A reexamination
Group Health Medical Associates. N Engl J Med 332(3): 13338, of risk factors for ventilatory impairment. Am Rev Respir Dis 138(4):
1995. 82936, 1988.
72. Xuan W, Peat JK, Toelle BG, Marks GB, Berry G, Woolcock AJ. 93. Kurzius-Spencer M, Sherrill DL, Holberg CJ, Martinez FD,
Lung function growth and its relation to airway hyperresponsiveness Lebowitz MD. Familial correlation in the decline of forced expiratory
and recent wheeze. Results from a longitudinal population study. Am J volume in one second. Am J Respir Crit Care Med 164(7): 126165,
Respir Crit Care Med 161(6): 182024, 2000. 2001.
73. Holberg CJ, Stern DA, Sherrill DL, Wright AL, Martinez FD, 94. Marossy AE, Strachan DP, Rudnicka AR, Anderson HR. Childhood
Morgan WJ. Eect of smoking on the development of lung function in Chest Illness and the Rate of Decline of Adult Lung Function
adolescence. Am J Respir Crit Care Med 163: A260, 2001. Between Ages 35 and 45 Years. Am J Respir Crit Care Med, 2006.

34
Natural History 3
95. James AL, Palmer LJ, Kicic E et al. Decline in lung function in the 97. Fabbri LM, Romagnoli M, Corbetta L et al. Dierences in airway
Busselton Health Study: The eects of asthma and cigarette smoking. inammation in patients with xed airow obstruction due to asthma
Am J Respir Crit Care Med 171(2): 10914, 2005. or chronic obstructive pulmonary disease. Am J Respir Crit Care Med
96. Guerra S, Sherrill DL, Kurzius-Spencer M, Venker C, Halonen M, 167(3): 41824, 2003.
Quan SF, Martinez FD. The course of persistent airow limitation in
subjects with and without asthma. Respir Med (in press).

35
Genetics of Asthma and COPD
4
CHAPTER

INTRODUCTION if they smoke cigarettes. However, the develop- Dirkje S. Postma1 and
ment of COPD among PI ZZ subjects is highly Edwin K. Silverman2
variable, and environmental, developmental, and
It is now a well-established fact that genetic fac- genetic factors likely contribute to this variabil- 1
Department of Pulmonology,
tors contribute to the development of asthma and ity. DeMeo and colleagues recently conrmed University Medical Center Groningen,
chronic obstructive pulmonary disease (COPD). that male gender and asthma (especially in University of Groningen, Groningen,
This chapter sets out the current knowledge childhood), as well as cigarette smoking, are risk The Netherlands
focusing on the heritability of asthma and factors for COPD in PI ZZ subjects [3]. 2
Channing Laboratory and Division of
COPD and the genes found by positional clon- Evidence that COPD unrelated to AAT Pulmonary and Critical Care Medicine,
ing and association studies. It also focuses on par- deciency is also inuenced by genetic factors
ticular genes to provide insight into the current Brigham and Womens Hospital, Harvard
has been provided by many familial aggregation
knowledge on genetic approaches to understand- Medical School, Boston, MA, USA
studies. Pulmonary function levels in the general
ing diseases like asthma and COPD. population cluster in families [4]. Moreover, stud-
ies of relatives of COPD subjects have conrmed
familial aggregation of COPD, probably related
to gene-by-smoking interactions. Silverman and
HERITABILITY OF ASTHMA AND COPD colleagues found that rst-degree relatives of
severe, early-onset COPD subjects who smoke,
had an approximately threefold increased risk
It has been known for centuries that asthma of airow obstruction and chronic bronchitis
clusters in families (familial aggregation). [5]; nonsmoking relatives had increased risk for
The rst segregation studies in families abnormalities in sensitive spirometric measures
with an asthmatic proband already showed that such as FEF25-75 but not for severe airow
no single-gene accounts for a major part of the obstruction [6]. McCloskey and colleagues con-
expression of the disease. A polygenic model rmed that COPD showed familial aggregation,
with some evidence of oligogenic loci (i.e. a which was only apparent in smokers [7]. Multiple
handful of loci being responsible for most of genes likely contribute to the susceptibility for
the genetic control) provided the best t to the COPD and its phenotypic expression.
data [1]. In the studies of the heritability of asthma
For COPD, a role for genetic factors has and COPD, the absence of unambiguous crite-
been less obvious, because cigarette smoking ria for their diagnosis and the fact that asthma
is such a major environmental risk factor. The and COPD are not one disease entity but are
identication of alpha 1-antitrypsin (AAT) both heterogeneous diseases, for example
deciency in the 1960s proved that genes could COPD can encompass both chronic bronchitis
inuence COPD susceptibility [2]. AAT de- and emphysema and the latter occurs homoge-
ciency is largely caused by homozygosity for neously or locally in the lung, have been major
the Z allele (PI ZZ) at the SERPINA1 locus, obstacles to pinpointing the exact polymor-
although multiple other rare deciency alle- phisms in genes contributing to disease devel-
les have been identied. PI ZZ subjects are at opment. Another explanation for the failure to
markedly increased risk for COPD, especially identify loci may lie in the genetic component

37
Asthma and COPD: Basic Mechanisms and Clinical Management

of the disease. For instance, dierent genes can be involved Candidate genes of unknown function or unknown
in the same phenotype (genetic heterogeneity) or the role in disease pathogenesis can also be selected for genetic
same genotype may result in dierent phenotypes (pleiot- analysis based on their dierential expression in diseased
ropy). Mutated genes do not always express the phenotype versus normal tissue [10, 11]. Another approach is to select
(incomplete penetrance) and vice versa. The specic phe- a gene that is a proven cause of a monogenic syndrome
notype under study can be expressed without the genetic that has the disease of interest as a component of its syn-
mutation (phenocopy). drome constellation. The hypothesis that can be tested is
that mutations in the gene with a milder functional eect
can contribute to the development of a complex genetic
disorder in the general population. The gene SPINK5 that
ASSESSING GENES FOR ASTHMA AND COPD encodes the serine protease inhibitor LEKTI on chromo-
some 5q32 as the cause of Netherton syndrome is such an
example [12]. Netherton syndrome is a severe autosomal
The human genome consists of approximately 3 billion base recessive disorder with a congenital skin disease associ-
pairs. The sequence of the whole human genome has recently ated with defective cornication and severe atopic mani-
been published [8]. The genetic distances are expressed in festations. A common coding polymorphism of SPINK5,
centiMorgans (cM). One cM is about 100,000 base pairs on E420K, has been shown to be associated with atopy and
a physical map and corresponds to 1% recombination during atopic dermatitis in two independent family cohorts [13],
meiosis. This means that one crossing-over event between although replication attempts have had varying success [14,
two loci that are one cM apart occurs in every hundred 15]. In COPD, cutis laxa is a rare dermatological syndrome
meioses. The estimation of the total number of genes in the related to abnormal connective tissue elasticity; some cases
human genome is about 20,000 [9]. Most genes in the pop- are caused by mutations in the distal part of the elastin
ulation have multiple locations where more than one variant (ELN) gene. Emphysema often occurs in cutis laxa at a very
is commonly found; the variants at these polymorphic loca- early age. Kelleher and colleagues found a rare mutation
tions are known as alleles. Only a minority of human DNA in the rst base of the last exon of ELN in an early-onset
is responsible for the coding for a biological product. COPD subject [16]. This variant, which was not a private
The main strategies to identify susceptibility genes are mutation in that pedigree, interfered with the assembly of
positional cloning and the candidate gene approach. the elastic ber, changed the proteolytic pattern of the ELN
Positional cloning starts with the investigation of protein, and altered cellular adhesion of the ELN molecule.
families without a predetermined hypothesis regarding the Thus, in both asthma and COPD, the assessment of can-
location or identity of the underlying susceptibility gene or didate genes from monogenic syndromes has led to some
genes. Markers are randomly spaced throughout the entire insights into the disease etiology.
genome and tested for linkage (i.e. coinheritance) with a Both the positional cloning and the candidate gene
disease phenotype. After the nding of linkage between a approaches have their own limitations. Population associa-
particular marker and a phenotype, further ne spaced typ- tion between a disease and a genetic marker can arise as an
ing of genetic markers (ne mapping) is required to pin- artifact of the population structure. Linkage studies with
point the exact gene causing the linkage. The approach is modest numbers of aected sib pairs may be underpowered
time consuming, as in-depth analysis of a particular region and fail to detect linkage, especially if there is genetic het-
of linkage that still can cover a large part of a chromosome, erogeneity. Furthermore, although linkage analysis has been
requires considerable molecular analysis. successful to identify genes underlying single-gene disor-
An alternative approach is to select candidate genes ders, in complex diseases it is frequently very dicult to
that putatively contribute to the underlying pathological suciently narrow a region of linkage to just a single gene.
process of the disease. The gene is screened for polymor- Nevertheless positional cloning in asthma has identied
phisms, which are tested for association with the disease some genes.
or phenotype in question. The results can be interpreted in A truly comprehensive genetic association study must
three ways, as shown in Box 4.1. consider all putative causal alleles in a gene of interest or
Replication of any genetic study is required to exclude in the entire human genome if resources are available. Until
spurious ndings especially if multiple genes are involved in recently, this was practically impossible. With the comple-
the disease process, like in asthma and COPD. tion of the International HapMap Project [17], it is now

Box 4.1. POTENTIAL CAUSES OF GENOTYPEPHENOTYPE ASSOCIATION


1. The trait of interest is due to a genotyped variant in the candidate gene.
2. The trait is determined by one or more genetic variants in linkage disequilibrium with a genotyped variant; that is, a
genotyped variant is very close to the disease gene.
3. The association is the result of population admixture, that is, a certain trait has a higher prevalence in a specic ethnic
subgroup within a mixed population. Any allele with a higher frequency within this subgroup will show association
with the trait.

38
Genetics of Asthma and COPD 4
possible to target a large proportion of the genetic variation the linkage between the markers and a hypothetical disease
across the genome, either directly or indirectly (via LD). susceptibility locus is determined. The evidence for linkage
HapMap is a freely available reference panel of genotype is often expressed as an LOD score, which is the logarithm
data from dierent worldwide populations (http://www. to the base 10 of the odds for linkage. Depending on the
hapmap.org). This resource can be used to guide the design study design, LOD scores above 3.3 (for extended pedi-
of disease association studies and prioritization of single grees) or 3.6 (for sib pairs) correspond to signicant linkage
nucleotide polymorphism (SNP) genotyping assays. With [23]. Although the location of genes for classic Mendelian
this dataset, it is possible to study genetic variants for any disorders can usually be determined quite accurately by
locus of interest. The HapMap dataset has clearly demon- linkage analysis, the locations of complex disease suscepti-
strated the existence of correlations between nearby variants. bility loci are typically less precisely localized due to genetic
By taking advantage of these correlations, one can select heterogeneity, incomplete penetrance, and environmental
informative SNPs (tagging SNPs) that provide information phenocopies. Linkage can be assessed for quantitative phe-
about neighboring variants that are not genotyped. Only a notypes as well as disease aection status.
small fraction of SNPs need to be genotyped to capture the A large number of genome scan linkage studies have
full information in a specic region. If a causal variant is not been performed in asthma, which were summarized by
genotyped, its eect can be indirectly tested with the corre- Celedon and colleagues [24]. Based on follow-up studies
lated tag SNP that has been genotyped. after linkage analyses, six potential susceptibility genes for
The single-gene approach described earlier can also asthma have been reported using positional cloning [2530].
be extended to the entire genome. Genome-wide asso- The only published genome scan linkage studies in COPD
ciation scans are emerging as powerful tools to identify have been performed in the Boston Early-Onset COPD
genes involved in complex diseases. On the basis of phase Study, implicating chromosomes 2q, 12p, and 19q as likely
I HapMap data, it was shown that approximately 250,000 locations of COPD susceptibility genes [31, 32]. Of interest,
500,000 SNPs are required to capture all common SNPs a linkage study of spirometric phenotypes in a set of families
in human populations. Although these numbers appear unselected for respiratory disease identied chromosome
impressive, current technologies can evaluate 1,000,000 2q, a location of signicant linkage to COPD, and chromo-
SNPs simultaneously [18, 19]. By selecting SNPs properly, some 5q, a location of signicant linkage to asthma, as likely
we can now interrogate the entire genome in one assay. This locations for genetic inuences on FEV1/FVC in the gen-
allows us to undertake genome-wide association studies, eral population [33]. In an adjacent region on chromosome
combining the ne mapping and power of association anal- 2q, Postma and colleagues found signicant evidence for
ysis and the ease of casecontrol cohort recruitment with linkage to FEV1/VC in families ascertained through asth-
a genome-wide hypothesisindependent approach. These matic probands [34]. Thus, there may be genetic inuences
promising tools will certainly have a major impact on our on asthma, COPD, and pulmonary function in the general
understanding of the genetic basis of complex diseases such population located within chromosome 2q.
as asthma and COPD. Genome-wide association stud-
ies have already led to important and novel insights into
the genetic architecture of a rapidly expanding list of com-
plex diseases, including age-related macular degeneration
and adult-onset diabetes mellitus [20, 21]. Although not ASSOCIATION STUDIES IN ASTHMA AND COPD
yet reported in COPD, Moatt and colleagues have per-
formed a genome-wide association analysis in asthma [22]. A large number of SNPs in the promoters and coding
They identied a region of highly signicant association to regions of a wide range of candidate genes have been exam-
asthma on chromosome 17q, which was replicated in sev- ined for genetic association in asthma. There are now over
eral other study populations. Using microarray gene expres- 500 studies that have examined polymorphisms in over 200
sion data, they observed that the expression of one of the genes for association with atopy and allergic disease pheno-
genes in that region, ORMDL3, was strongly associated types [3537]. These studies have provided us with increas-
with the same SNPs that were associated with asthma ing insight into genetic susceptibility to asthma, the role of
suggesting that asthma susceptibility is related to ORMDL3 geneenvironment interaction and the role of genetic varia-
gene expression. tion in inter-individual response to treatment.
In this part we will focus on ve of the candidate genes
that have the best evidence for involvement in asthma sus-
ceptibility (Table 4.1); these genes have been identied in
LINKAGE ANALYSIS OF ASTHMA AND COPD various ways, including positional cloning (ADAM33) and
positional and/or biological candidates (IL-13, IL-4, IL-4R,
CD-14). We will also review an example from asthma phar-
Linkage analysis includes a group of genetic epidemiologi- macogenetics (ADRB2).
cal methods that are used to identify chromosomal regions Subsequently, we will consider ve of the COPD
that are likely to contain one or more genetic determinants candidate genes with the strongest evidence for associa-
inuencing a phenotype of interest. A panel of genetic tion to COPD susceptibility, including positional candi-
markers is genotyped within family units, which can be dates (TGFB1 and SERPINE2) and biological candidates
as small as sib pairs or as large as extended pedigrees, and (GSTP1, EPHX1, and SOD3) (Table 4.2).

39
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 4.1 Summary of positive and negative association studies in the genes ADAM33, IL-3, IL-4, and IL-4R with asthma and its related phenotypes.

Studies supporting Studies refuting


Gene association association Comments

A disintegrin and [25] Caucasian [38] Puerto Rican,


metalloprotease Mexican
33 (ADAM33) [39] African Americans, [40] Raby children
Caucasian, Hispanic, Dutch
[41] Japanese [42] Chinese Cheng allergic rhinitis
[43] ) Korean [44] German Schedel children. Positive and negative associations with various phenotypes
[45] Caucasian Jongepier association with FEV1 decline
[46] Caucasian
[47] Caucasian Simpson low lung function in children
[48]
[49] Japanese Noguchi children

IL-13, IL-4, IL-4R [50] [51] Celedon IL-13 83 trios children


Deichmann IL-13, IL-4R
[52] IL-13
[53] IL-13
[54] IL-13  Children
[55] IL-13  Children
[56] IL-4R /IL-4Children
[57] IL-13 /IL-4R
[56] IL-4R
[53] IL-13
[58] IL-13, IL-4 Interaction
[59] IL-13 Children
[60] IL-13
[61] IL-4R
[62] IL-13/IL-4R Interaction
[63] IL-13 Children
[64] IL-13/IL-4 Children
[65] IL-13/Il-4/IL-4R Children, Interaction
[66] IL-4/IL-4R Interaction
[67] IL-13 Children
[68) IL-4/ IL-4R Children
[69] Children
[70] IL-13
[71]
[72] IL-13/IL-4/IL-4RInteraction Children
[73] IL-4R/IL-13 Interaction  Children
[74] IL-13 Children

SNPs in the IL13/IL4 pathway have been reported as single gene association. When interaction was present, this has been assigned as interaction, and when interaction was
absent, this has been assigned as interaction .

ASTHMA GENETIC ASSOCIATION STUDIES [25]. Multiple SNPs within ADAM33 were signicantly
associated with asthma and BHR in a US or UK popula-
tion, or with these two populations combined. The nomen-
Gene found by positional cloning: ADAM33 clature typically used for ADAM33 SNPs is unusual; the
exons have been assigned sequential letters, and the SNPs
ADAM33 was discovered by positional cloning in 460 have been sequentially numbered within each exon (e.g. S_2
Caucasian families from the US and UK on chromosome is the second SNP in exon S). SNPs occurring in the intron
20p. It was associated with asthma, specically when bron- before an exon are designated with a minus sign (e.g. Q-1
chial hyperresponsiveness (BHR) was present, and there is an intronic SNP before exon Q), and SNPs occurring in
were variations in total and as well as specic IgE levels an intron immediately after an exon are designated with

40
Genetics of Asthma and COPD 4
TABLE 4.2 Summary genetic association evidence for five candidate genes in COPD.

Studies supporting Studies refuting


Gene association association Comments

Microsomal epoxide hydrolase (EPHX1) [75] [76] Yoshikawa reported association with COPD severity
[77] [78]
[79] [80]
[81] [81] Hersh found association in their case-control study but
not their family study
[82] Cheng reported association with COPD severity

Glutathione S-transferase P1 (GSTP1) [83] [84]


[85] [81] He found increased risk for Val105 as opposed to Ile105
[86] [82] Cheng found increased COPD risk for combination of
Ile105 with GSTM1 and EPHX1 variants

Extracellular superoxide dismutase (SOD3) [87] Arg213Gly variant has low frequency
[88]

Transforming growth factor beta 1 (TGFB1) [89] [90]


[91] [92]
[93]
[94] Van Diemen found association with COPD affection
status but not FEV1 decline

Serpin peptidase inhibitor, Clade E, Member [95] [96]


2 (SERPINE2)
[97]

a plus sign (e.g. F  1 is an intronic SNP after exon F). level [99]. A total of 1390 subjects from a Dutch general
The majority of associated SNPs were located in the 3
population cohort were genotyped for eight asthma-
half of the gene, extending from exon Q to the last exon, associated SNPs. Individuals homozygous for the minor
exon V. Subsequently, there have been a large number of alleles of SNPs S_2 and Q-1 and heterozygous for the
casecontrol and family-based association studies focused SNP S_1 had a signicantly accelerated FEV1 decline over
on ADAM33, with the majority [39, 43, 45, 46, 48, 49, 82] 25 years follow-up of 4.9, 9.6, and 3.6 ml/year, respectively,
but not all [38, 40, 42, 44], conrming the original nding when compared with the wild-type allele. A further analysis
(Table 4.1). For example, the Collaborative Study on the demonstrated a higher prevalence of the SNPs F  1, S_1,
Genetics of Asthma, comprising eight US genetic cent- S_2, ST  5, and T_2 in subjects with COPD at GOLD
ers, demonstrated positive association between SNPs in (Global Initiative for Chronic Obstructive Lung Disease)
ADAM33 and asthma as well as BHR in African-American, stage II or higher. Thus, in addition to asthma, it seems that
Hispanic, and white populations [39]. In a further study polymorphic variation in ADAM33 also inuences the rate
involving 1299 asthma cases, 1665 control subjects, and of decline of lung function at a population level, which may
4561 family members, Blakey and colleagues [48] applied then lead to COPD. Subsequently, the authors conrmed
a literature-based meta-analysis, which supplemented the association with COPD in a second cohort [100] and
the database with new asthma cases from populations in interestingly found that the same ST_5 SNP was associ-
Iceland and the United Kingdom, and demonstrated sig- ated with the severity of BHR in COPD patients, as well
nicant association for 4 of the 13 SNPs tested. as to the number of sputum cells and CD8 cells in bron-
It is well recognized that in patients with chronic per- chial biopsies. This links the increase in cells in COPD with
sistent asthma, baseline lung function declines more rapidly hyperresponsiveness and ADAM33.
over time when compared with that of normal individuals Finally, the Manchester Asthma and Allergy Study,
[98]. Jongepier et al. showed in 200 patients with chronic a prospective cohort study of the development of asthma
asthma who were studied annually for 25 years [45] that and allergies in children, investigated the relation between
the rare allele of the S_2 polymorphism (minor allele fre- SNPs in ADAM33 and lung function at ages 3 and 5 [47].
quency 0.25) was signicantly associated with excess At the age of 5 years, four of the SNPs were associated
decline in FEV1 over time and concluded that this variant with reduced FEV1 (F  1, N  1, T1, and T2; p  0.04).
of ADAM33 was not only important in the development Linkage disequilibrium mapping of ADAM33 pointed to
of asthma but also in disease progression, possibly related functional SNPs lying between F  1 and B  1.
to enhanced airway remodeling. A further study by the Taken together, these studies support the notion that
same group investigated whether SNPs in ADAM33 could ADAM33 may be a gene involved in lung development
also predict an accelerated FEV1 decline at a population and further remodeling over a lifetime (Fig. 4.1). This may

41
Asthma and COPD: Basic Mechanisms and Clinical Management

then contribute to an accelerated loss of lung function and from gene expression studies and transcription factor
contribute as well to BHR. Given the associations with binding analysis on C-1112T strongly support a functional
dierent SNPs, ADAM33 may have dierential function role of this polymorphism in gene expression [101]. Genetic
inducing dierent phenotypes in men. variations in IL-13 have been associated with asthma and
related phenotypes in almost all studies that assess SNPs in
IL-13 (Table 4.1), and this is present in ethnically diverse
Genegene interaction: populations living in variable environmental circumstances
IL-13, IL-4, and IL-4R [5055, 5760, 6265, 67, 70, 7274, 102].
In the coding region of the IL-4Ralpha gene, at least
Il-13 and IL-4 are cytokines produced by Th2 cells that are 14 polymorphisms have been identied and for some of the
capable of inducing isotype class switching of B-cells to pro- more frequent genetic variations, functional data is available.
duce IgE. They also share a receptor component, IL-4R, SNP A148G alters an amino acid in the extracellular part of
which is an important factor in the development or expres- the receptor (I50V) which leads to increased IgE produc-
sion of atopy and asthma. The IL-13 receptor consists of one tion in B cells [102]. Polymorphisms T1432C and A1652G
IL-4R subunit and either a low-anity IL-13R1 or a lead to amino acid changes Ser478Pro and Gln551Arg in
high-anity IL-13R2 subunit. The complete receptor for the intracellular domain of the IL-4Ra chain, respectively.
IL-4 is composed of one IL-4R subunit and an IL-4R Dierent studies have assessed the eect of these polymor-
subunit, or IL-13 subunit (Fig. 4.2). IL-4 and IL-13 are phisms on atopic diseases [50, 56, 59, 61, 62, 64, 66, 68,
located on chromosome 5q and share the IL-4 receptor 72, 73, 103]. However, overall, the associations between
-chain, which is located at chromosome 16p12. IL-4Ralpha polymorphisms and the diagnosis of asthma as
In IL-13 seven frequent polymorphisms exist of well as serum IgE levels were only minor. In 2002, Howard
which at least three could be of functional relevance: et al. published a combined analysis of two polymorphisms
Polymorphism G2044A alters an amino acid in the protein in the IL-4/ IL-13 pathway [62]. In a longitudinal popula-
domain involved with receptor binding (Arg 130Gln) [71]. tion of Dutch adults with asthma, one polymorphism in the
This SNP and two other polymorphisms in the promoter promoter of IL-13 (C-1112T) and one in the IL-4Ra gene
(A-1512C and C-1112T) showed association with asthma leading to an amino acid change (Ser478Pro) were assessed.
and IgE regulation in dierent populations [54, 65]. Results The SNP in IL-13 (C-1112T) was previously found to be
associated with BHR in this population. The SNP in IL-4Ra
was associated with higher levels of IgE. As both traits are
associated with asthma the authors studied interaction of
ADAM33 these genes. When individuals with polymorphic alleles in
both locations were compared to individuals with wild-type
Lung function (FEV1)

Asthma alleles in both locations the risk for asthma increased vefold.
ADAM33
Also, the interaction between the polymorphism in
the coding region of IL-13 (G2044A) and the promoter
ADAM33 polymorphism C-589T in IL-4 has been studied [64]. The
ADAM33
IL-13 G2044A polymorphism and haplotypes consisting of
COPD IL-13 G2044A and IL-4 C-589T were associated with the
ADAM33 development of atopy and atopic dermatitis. As children in
this analysis were only followed up to the age of 24 months,
no information on asthma was available.
Age (years) Three polymorphisms in the IL-4 receptor alpha gene
(Arg551Glu, Ile50Val, and Pro478Ser) have been investigated
FIG. 4.1The phases of lung function growth and decline: growth, plateau
and decline phase. In all three phases ADAM33 may contribute to the
in combination with a promoter polymorphism in the IL-4
course of lung functions over a lifetime. gene (C-589T) [68]. The risk for asthma increased up to an

Type I IL-4 Type II IL-4 IL-13 receptor


receptor receptor

IL-4 IL-4
IL-13

Membrane

IL-13R IL-13R

IL-4R IL-4R IL-4R


C

FIG. 4.2 Interaction between IL4 and IL13 and their respective receptors, that have some similarities.

42
Genetics of Asthma and COPD 4
odds ratio (OR) of 1.97 (95% CI 1.073.71) in individuals directing the adaptive immune response. The CD14 gene
with the Arg allele at position 551 while the other two SNPs contains several SNPs, the most important one being the
in the IL-4Ra gene did not show any signicant eects. The CD14/-159 (also called CD14/-260). Functional genomic
combination of the IL-4Ralpha Arg allele with the IL-4 studies showed that monocytic cells with the T allele are
589T allele increased the OR to 3.70 (95% CI 1.07 transcriptionally more active [103].
12.78). This study extends the initial ndings by Howard in The initial study by Baldini et al. [104] demonstrated
the sense that a second combination of genes seems to inter- that the C-159T polymorphism was associated with elevated
act in the IL-4/ IL-13 pathway. In a similar British study, soluble CD14 levels, and reduced total serum IgE levels in
two polymorphisms in the IL-4Ra gene (Arg551Glu and children who were skin prick test positive to common aeroal-
Ile50Val, respectively) and two in the IL-4 gene (the highly lergens. Not all studies have been consistent however, rais-
linked SNPs C-589T and C-33T, respectively) were ana- ing the possibility that the level of environmental exposure
lyzed concomitantly. While transmission disequilibrium tests to endotoxin may alter the eect of CD14 polymorphisms
and haplotype analysis showed signicant associations with [105]. A number of recent studies have supported this notion.
asthma for both genes individually, the authors state that no Endotoxin. The hygiene hypothesis proposes that
interaction between the two genes was observed. However, microbial exposure during early life development reduces
no data and no information on how interaction was tested, is the development of asthma and allergic disease [106] and
provided [66]. has been the subject of many genetic association studies
Other researchers also failed to replicate interactions [107]. The timing, dose, and route of microbial exposures,
between polymorphisms in the IL-4/IL-13 pathway. Liu such as endotoxin, are likely to interact with genetic inu-
et al. investigated six polymorphisms in IL-4, IL-13, and ences, thus altering the response to these exposures [106].
IL-4Ra in almost 1000 children who were followed from Exposure to endotoxin is known to occur indoors from
birth. The eects of genetic variation in the IL-4/IL-13 contact with house dust. In the Barbados Asthma Genetics
pathway on specic and total serum IgE levels were ana- Study, subjects with low house dust endotoxin exposure,
lyzed [65]. The authors conrmed a role of IL-13 polymor- who had the CD14-159TT genotype had a reduced risk of
phisms in the regulation of total and specic IgE, but SNPs asthma whereas high exposure increased the risk of asthma
in IL-4 and IL4-Ra were only associated with specic IgE with this genotype [107]. Taken together, these studies pro-
responses in that study population. Although the interac- vide support for an endotoxin switch in which there is a
tion analysis is not shown, the authors state that no signi- dose-dependent response to endotoxin exposure for specic
cant interactions between polymorphisms in the three genes risk genotypes [108]. Exposure to endotoxin is also encoun-
were found. A further study on IL-4, IL-4Ra, IL-13, and tered in occupational settings such as farming. Adult farm-
IL-13Ra1 provided association of the IL-13 G2044A poly- ers with the CD14-159TT and -1691GG genotypes had
morphism with asthma in two populations, without signi- signicantly lower lung function and increased wheezing
cant interaction [53]. In contrast, recent studies by Kabesch compared with other genotypes, possibly due to increased
et al. [72] and Chan et al. [73] found signicant interactions soluble CD14 levels interacting with inhaled endotoxin
between the genes in the IL-4/IL-13 pathway. from the agricultural environment [103].
Dierences in observations may result from small Animal exposure. The type of microbial exposure
study population sizes and from characteristics of the popu- during immune system maturation may inuence the
lation under study, for example the dierences due to age, development of atopy and asthma. In children with the
gender, prevalence of atopy, high- and low-risk population CD14-159C allele who had regular contact with pets,
and so on. Thus the ndings need more study. However, it is serum IgE levels were higher than with the T allele [109].
shown that genegene interaction may contribute to asthma The opposite occurred in children with regular contact with
and atopy development. Analyses of genes in pathways that stable animals where the C allele was associated with lower
are relevant to atopy, asthma and or its subphenotypes like IgE levels. In another study, early life farm environment and
BHR need further attention. the CD14-159TT genotype combined to give the lowest
risk of nasal allergies and atopy [110].
Environmental tobacco smoke. Exposure to environ-
Geneenvironment interaction: CD14 mental tobacco smoke may increase the risk of asthma
in susceptible individuals. In a study of Puerto Rican
Innate immunity genes provide the interface between the and Mexican families, people with asthma who had the
immune system and microbial products. Many microbes CD14  1437GG or GC genotypes and exposure to envi-
have specic pattern molecules on their surfaces and these ronmental tobacco smoke had a mean forced expiratory
molecules interact with pattern-recognition receptors. volume in 1 s that was lower by 8.6% of that predicted as
CD14 is such a receptor. CD14 is part of the receptor com- compared with GG or GC subjects that were not exposed
plex for lipopolysaccharide (LPS, endotoxin) together with to environmental tobacco smoke [111]. In addition, people
toll-like receptors (TLR) 2 and 4. CD14 does not have a with asthma with the CD14-159TT genotype and expo-
transmembrane domain and thus does not signal itself but sure to environmental tobacco smoke had lower serum IgE
contributes to the anity of the interaction between the levels. The mechanism for this interaction could involve
microbial products and TLRs by formation of a receptor exposure to endotoxin found in cigarettes. Sex dierences
complex. Downstream eects include the upregulation of may also exist in response to tobacco smoke. Girls whose
accessory molecules and release of cytokines, such as IL-10 mothers had smoked during pregnancy or whose parents
and IL-12. These cytokines are potentially important in had asthma had lower mean soluble CD14 levels [112].

43
Asthma and COPD: Basic Mechanisms and Clinical Management

Asthma pharmacogenetics: The treatment-related changes in lung function. Israel et al.


beta2-adrenergic receptor [123] matched asthmatic individuals homozygous Arg/Arg
(n 37) to those Gly/Gly (n 41), by level of FEV1. The
The therapeutic response to beta2-agonists is heterogeneous genotype-stratied individuals were then randomized in a
in asthmatic subjects [113], a nding suggested to be due double-blind, cross-over study of regularly scheduled qid
in part to genetic variation in the beta2-adrenergic receptor albuterol therapy versus placebo over two 16-week periods.
gene (ADRB2). A number of ADRB2 polymorphisms have Again, those with the Arg/Arg genotype had lower morn-
been described [114], with the greatest attention devoted to ing PEF versus placebo (10 l/min, p 0.02), while those
the SNPs causing amino acid substitutions at positions 16 with Gly/Gly had higher PEF (14 l/min, p 0.02). The dif-
and 27, namely the Gly16Arg and the Gln27Glu polymor- ference between Arg/Arg and Gly/Gly genotypes was sig-
phisms [115, 116]. Many of the early studies on the acute nicant for morning PEF (24 l/min, p 0.0003), evening
bronchodilator response to a beta-agonist diered substan- PEF, FEV1, morning symptom score, and need for rescue
tially from one another with respect to study design, type of medication.
agonist utilized, and primary outcome assessed [117, 118]. Further studies have focused on asthma exacerbations
Not surprisingly, there was marked inconsistency in results as an outcome. With regular use of both short- and long-
between trials. Subsequently, larger studies have demon- acting beta-agonists, presence of at least one Gly16 allele is
strated that bronchodilator responses were higher and more protective against exacerbations [124]. Moreover, in a meta-
rapid among Arg16 homozygotes as compared to Gly16 analysis evaluating long-acting beta-agonist usage, subjects
homozygotes and heterozygotes [119, 120]. with at least one Arg allele demonstrated signicant decre-
The second phenotypic outcome evaluated has been ments in PC20 when compared to placebo, suggesting that
the potential for downregulation of beta-agonist receptor the downregulation of the receptor accompanying tachyph-
responsivity (tachyphylaxis) with the chronic administration ylaxis may result in decreased bronchoprotection [125].
of beta-agonists in association with ADRB2 genetic vari- Several studies have assessed the eects of long-
ants. In a key study, 255 asthmatics of mild severity were acting beta-agonists [126128]. Wechsler et al. showed
randomized to either regular (180 g QID) or as-needed decreased responses to salmeterol in Arg/Arg subjects com-
albuterol use and were assessed over a 16-week period for pared with Gly/Gly subjects [126]. In the Salmeterol or
evidence of tachyphylaxis and clinical deterioration (as Corticosteroids (SOCS) trial, morning peak ow worsened
measured by fall in AM peak expiratory ow rate (PEF)). in Arg/Arg asthmatic subjects (n 12) who were treated
No dierence in PEF variation was observed between treat- with salmeterol alone after inhaled corticosteroids (ICS)
ment groups [121]. However, Arg16 homozygotes sig- withdrawal when compared with subjects who received
nicantly decreased their pre-albuterol PEF with regular placebo; no decrease in morning peak ow was observed
utilization of albuterol therapy, compared to both Arg16 in Gly/Gly subjects (n 13). In the Salmeterol and/or
homozygotes receiving as-needed albuterol and Gly16 Inhaled Corticosteroid (SLIC) trial, Arg/Arg (n 8) asth-
homozygotes in either treatment group [121]. The dier- matic subjects did not show a sustained improvement in
ence in evening PEF between these groups was 31.6  10.2 lung function as compared with Gly/Gly (n 22) subjects
l/min comparing Arg16 homozygous regular users ver- on salmeterol, regardless of whether subjects were admin-
sus Gly16 homozygous regular users (p 0.002) and istered concomitant ICS. As these studies were all of small
31.1  13.0 l/min versus the Arg16 homozygous as needed size that may confound the outcome of the analyses, larger
group (p 0.02) [122]. Similar dierences in morning PEF studies are required to rmly establish whether polymor-
were noted (Fig. 4.3). phisms in the ADRB2 gene do aect the benecial clinical
2AR desensitization was further evaluated in a eects of long-acting beta2-antagonists (LABA) treatment
prospective, genotype-stratied study of Arg16Gly on when added to inhaled steroids. The size of the study popu-
lation is of importance, particularly given the variability of
individual responses to pharmacotherapy both between sub-
jects and within subjects on a day-to-day basis.
10 Finally, previous studies have illustrated that rela-
Arg/Arg-as needed
Gly/Gly-regular tively few ABRB2 haplotypes account for the majority of
5
the haplotypic diversity of this gene, and that the frequency
AM PEF (l /min)

0 of common haplotypes diers between population groups


[115, 129]. An eect of ADRB2 haplotype on acute bron-
5
chodilator responses to albuterol has been demonstrated in
10 some studies [128] but not in others [115, 130].
15
Arg/Arg-regular Studies on ADRB2 provide evidence that the gene
is likely to be associated with bronchodilator response but
20 that genetic prediction of this response may vary by eth-
25 nic group. Additionally, individuals homozygous for Arg16
0 5 10 15 20 demonstrate signicant 2AR desensitization and these
Run out individuals (approximately 15% of population) are at risk of
Weeks after randomization
clinical deterioration with the regular use of beta-agonists.
FIG. 4.3 Change in morning PEF (Peak Expiratory Flow) with beta-agonist Future studies have to assess whether this is still the case if
(albuterol) treatment, according to ADRB2 genetic variants. patients are being treated with ICs.

44
Genetics of Asthma and COPD 4
most rapid FEV1 decline and 308 subjects who had no meas-
COPD GENETIC ASSOCIATION STUDIES urable FEV1 decline over 5 years. EPHX1 haplotypes were
associated with rate of FEV1 decline, and this association
Biological candidate gene: Microsomal was more signicant among individuals who reported a fam-
ily history of COPD potentially related to other COPD
epoxide hydrolase genetic risk factors, which act additively with EPHX1 or
which interact with EPHX1 to increase COPD risk.
Microsomal epoxide hydrolase is a xenobiotic enzyme that Hersh and colleagues studied a total of eight EPHX1
detoxies highly reactive epoxides, which are created by cig- SNPs, which included the two widely studied nonsynony-
arette smoking. Thus, variation in the activity of this enzyme mous SNPs above, in a family-based study and a casecon-
has been hypothesized to contribute to variable COPD trol study [81]. The family-based Boston Early-Onset
susceptibility. In an early COPD genetic association study, COPD Study subjects included 127 extended pedigrees
Smith and Harrison studied two genetic variants in the (949 total subjects) ascertained through a proband with
EPHX1 gene [75], one of which had been reported to cause severe, early-onset COPD; the casecontrol study included
reduced activity of this xenobiotic enzyme (Tyr113His) 304 severe COPD subjects from the National Emphysema
and the other which had been reported to cause increased Treatment Trial (NETT) and 441 control smokers from the
enzyme activity (His139Arg). They analyzed two small case Normative Aging Study. None of the EPHX1 SNPs were
groups that included 68 subjects with spirometrically proven associated with quantitative or qualitative airway obstruc-
COPD and 94 patients with both pathologically proven tion phenotypes in the early-onset COPD families under an
emphysema and lung cancer. Their control group of 203 additive model. However, the Arg139 allele showed modest
blood donors was not phenotyped. Both their COPD and evidence for association to COPD in the casecontrol study.
emphysema/lung cancer case groups had signicantly higher Overall, there is reasonable but not uniformly con-
frequencies of homozygosity for the His113 slow allele sistent evidence for association between EPHX1 SNPs and
associated with reduced EPHX1 enzyme activity. Somewhat COPD. The small sample sizes of many of these studies
paradoxically, a higher frequency of the Arg139 fast allele could contribute to inconsistent replication. A further expla-
associated with increased EPHX1 enzyme activity was also nation might be dierences in ethnicities; however, another
found in the COPD cases compared to controls. potential contribution to these inconsistent results is phe-
In a study of 180 former poison gas factor workers notypic heterogeneity within the COPD cases. COPD
in Japan, followed to determine the impact of these toxic includes both emphysema and airway disease in varying
exposures on the development of lung disease, Yoshikawa proportions between dierent subjects, and emphysema dis-
and colleagues compared the distributions of the Tyr113His tribution also varies widely between subjects. DeMeo and
and His139Arg variants in 40 subjects with COPD colleagues studied emphysema distribution in 282 of the
(FEV1  70% predicted) and the remaining 140 subjects 304 NETT Genetics Ancillary Study participants studied
[76]. No signicant dierences were observed in allele fre- by Hersh and colleagues who had chest CT scans with den-
quencies between these groups. However, when they sub- sitometric and radiologist assessment of emphysema sever-
divided their 40 COPD cases into 20 mild (FEV1  60% ity and distribution [131]. The EPHX1 His139Arg SNP
predicted) and 20 moderate/severe (FEV1  60% pre- was signicantly associated with emphysema distribution
dicted) subjects, a higher frequency of the slow His113 assessed by densitometry (p 0.005) and by radiologist
allele was observed in the more severely aected subjects. scoring (p 0.01). To determine whether utilizing a more
Thus, they speculated that EPHX1 variants were associated homogeneous set of COPD cases would improve the abil-
with COPD severity rather than susceptibility. ity to detect genetic association, the association results using
Two other small casecontrol genetic association all NETT COPD cases were compared to the results using
studies, performed in Asian populations and published in only 171 upper lobe predominant emphysema cases; the
2000, found no association between the Tyr113His and same set of 441 control subjects was used for both com-
His139Arg variants and COPD [78, 80]. In a subsequent parisons. Despite including a smaller number of COPD
study of 184 COPD cases and 212 control smokers from cases, the upper lobe predominant emphysema group asso-
Taiwan, the genotype frequencies for the Tyr113His and ciation analysis indicated that the Arg139 fast allele was
His139Arg SNPs did not dier between the cases and more protective (OR 0.60 with p 0.005) compared to
controls [41]. However, using the genotypes at both posi- the association analysis using all COPD cases (OR 0.73
tions to assign two functional EPHX1 categories (slow/very with p 0.02). These results suggest that phenotypic het-
slow versus normal/fast), did lead to a signicant dierence erogeneity could contribute to the inconsistent replication
between cases and controls. Moreover, homozygosity for of EPHX1 (and other COPD candidate gene SNPs) asso-
the His113 allele was more prevalent in severe versus mild ciations with COPD; more comprehensive phenotyping of
COPD cases. More recently, Brogger and colleagues stud- COPD cases, including chest CT scans, should be consid-
ied 244 subjects meeting spirometric criteria for COPD ered for future COPD genetic studies.
and 248 control subjects [77]. They found that the His113
variant was associated with COPD.
Sandford and colleagues studied the impact of haplo- Another xenobiotic enzyme gene: GSTP1
types formed by the two widely studied EPHX1 SNPs on
rate of decline in FEV1 among continuing smokers in the Like EPHX1, glutathione S-transferase P1 (GSTP1)
Lung Health Study [79]. They selected 283 subjects with the detoxies oxygen radicals produced by cigarette smoke; it

45
Asthma and COPD: Basic Mechanisms and Clinical Management

has been extensively studied as a COPD candidate gene. colleagues genotyped the Arg213Gly variant in 230 COPD
Ishii and colleagues studied two nonsynonymous SNPs in cases and 210 control smokers [87]. Importantly, as the Gly213
GSTP1 in 53 COPD cases and 50 control subjects from variant was hypothesized to confer resistance to COPD among
Japan [81]. A higher frequency of Ile105 homozygotes smokers, their control subjects had normal spirometry with a
at the Ile105Val SNP was observed in COPD cases (79% heavy smoking history (mean pack-years 42). The allele fre-
versus 52%); some evidence suggests that the Ile105 variant quency of Gly213 was only 1% in the COPD cases but it was
confers reduced functional activity of the GSTP1 enzyme 5% in the control smokers (p 0.02). Carriers of at least one
[132]. In a Korean population of 89 COPD cases and 94 Gly213 allele were signicantly more common among controls
smoking controls, Yim and colleagues found no association than cases (p 0.005).
of the Ile105Val SNP with COPD [84]. In the Lung Health In the Copenhagen City Heart Study, Juul and col-
Study, He and his colleagues compared 544 subjects with leagues studied the Arg213Gly variant in 9258 Danish sub-
the highest baseline FEV1 values (mean FEV1 91.8% jects [88]. They conrmed the marked eect of this variant
predicted) to 554 subjects with the lowest baseline FEV1 on EC-SOD plasma levels, with mean values of 142 ng/ml
values (mean FEV1 62.6% predicted); all of these subjects in Arg213 homozygotes, 1278 ng/ml in Arg213/Gly213
had COPD based on reduced FEV1/FVC ratio [85]. They heterozygotes, and 4147 ng/ml in Gly213 homozygotes.
observed a higher frequency of Val105 homozygotes among In a multivariate model adjusting for gender, age, smoking,
the low lung function group (13.2% versus 9.3%), as well as and occupational dust exposure, smokers who were hetero-
a faster rate of FEV1 decline in high lung function subjects zygous (Arg213/Gly213) had substantially lower risk for
who were Val105 homozygotes. COPD than Arg213 homozygotes (OR 0.4, with 95% CI
There are multiple potential explanations for the incon- 0.20.8). Among nonsmokers, no protective eect of the
sistent results of these previous studies, including population Gly213 variant for COPD was observed. Only two Gly213
dierences (ethnicity, COPD status), failure to analyze the homozygous subjects were identied in the entire cohort.
actual functional variant in GSTP1, and small samples in sev- This conrms the potentially protective eect of the Gly213
eral of the studies. However, it is also possible that genegene allele among smokers, and it emphasizes the utility of large
interactions need to be considered to identify key COPD sample sizes especially when studying relatively uncom-
susceptibility genes. Studies of combinations of GSTP1 vari- mon genetic variants.
ants with other potential susceptibility genes have also been
performed. Calikoglu and colleagues did nd a higher fre-
quency of Ile105 homozygotes in a male Turkish population
of 149 COPD cases (61%) compared to 150 control subjects TGFB1: Convergence of biological and
(38%) [86]. They also examined the impact of including vari- positional candidate gene approaches
ants in GSTT1 and GSTM1 in combination with GSTP1
and found a markedly increased risk of COPD when a com- Transforming growth factor beta 1 (TGFB1) is a cytokine
bination of variants in these three genes was considered. In that acts as a central regulator of the inammatory response;
addition to EPHX1, Cheng and colleagues also studied the thus, it is a logical biological candidate gene for COPD.
GSTP1 Ile105Val variant in their set of 184 COPD cases A potentially functional variant has been identied at
and 212 control subjects from Taiwan [41]. Although the codon 10, which changes a leucine to a proline (Leu10Pro);
Ile105Val SNP was not associated with COPD when ana- individuals that carry at least one proline allele have higher
lyzed individually, the combination of at least one EPHX1 TGFB1 mRNA in peripheral blood mononuclear cells and
slow allele (His113), homozygosity for the GSTM1 Null higher serum TGFB1 protein levels [134]. Wu and col-
allele, and homozygosity for the Ile105 allele was signicantly leagues genotyped this nonsynonymous SNP in Caucasian
more common in COPD cases (36%) versus controls (8.5%). subjects from New Zealand. They included 165 COPD
Formal tests of interaction in large samples will be required cases and two control groups 76 control smokers and 140
to replicate these ndings and to determine whether these unphenotyped blood donors [89]. The Pro10 allele was less
results represent the combination of susceptibility alleles act- common in COPD cases (33%) than in either the blood
ing independently or signicant genegene interactions. donor controls (45%) or smoking controls (45%).
Celedon and colleagues identied TGFB1 as a posi-
tional candidate gene on chromosome 19q after performing
linkage analysis in 72 extended pedigrees (585 individuals)
Association with a relatively uncommon ascertained through severe, early-onset COPD probands [91].
allele: SOD3 The initial evidence for linkage of FEV1 to 19q was mod-
est (LOD score 1.40). However, genotyping additional
Although genetic association studies typically analyze common short tandem repeat markers to increase the information
genetic variants with allele frequencies above 10%, it is quite available for linkage analysis, as well as performing a strati-
likely that less common variants contribute to the susceptibility, ed analysis in smokers only (to identify genomic regions
and resistance, to complex diseases like COPD. Extracellular likely inuenced by gene-by-smoking interactions), led to
superoxide dismutase (EC-SOD or SOD3) detoxies oxy- more substantial evidence for linkage (LOD score 3.30).
gen radicals by scavenging superoxide anions. A variant in this TGFB1 is located within the linkage region, and ve SNPs
gene, Arg213Gly, has been shown to have a functional eect were genotyped in both Boston Early-Onset COPD Study
on the processing of the EC-SOD protein, leading to substan- pedigrees for family-based association analysis and in a set of
tially elevated plasma levels of the protein [133]. Young and 304 COPD cases and 441 smoking control subjects. Three

46
Genetics of Asthma and COPD 4
SNPs were signicantly associated with COPD phenotypes were associated with airow obstruction phenotypes in the
in each population, and one SNP in the promoter region of Boston Early-Onset COPD families; a SNP-by-smoking
TGFB1 (rs2241718) replicated in both study populations. interaction term was included in those analyses. Five of
The Leu10Pro variant was only signicantly associated in the these 18 SNPs demonstrated replicated associations in the
casecontrol analysis, but a lower Pro10 allele frequency was casecontrol cohort. Although Chappell and colleagues
noted in COPD cases indicating the same directionality of wrote a Letter to the Editor indicating that they could not
eect as in the study by Wu and colleagues. replicate these associations in their casecontrol cohort [96],
Following these initially supportive studies using the Zhu and colleagues recently reported their analyses of 25
biological and positional candidate gene approaches, further SERPINE2 SNPs in both a family-based cohort of 1910
replication eorts of TGFB1 in COPD susceptibility have subjects and a casecontrol cohort from Norway includ-
had mixed results. In a small study comparing 84 COPD ing 973 COPD cases and 956 control smokers [97]. The
cases and 97 control subjects from China, signicant dif- family-based association analysis demonstrated association
ferences in both allele and genotype frequencies were noted of ve SNPs to COPD, with p values as low as 0.002. The
for two TGFB1 promoter SNPs [93]. However, association casecontrol analysis only found one SNP with p  0.05
analysis of the Leu10Pro SNP and two promoter SNPs for COPD aection status; however, ve SNPs were associ-
in 102 COPD cases and 159 control smokers from Korea ated with FEV1/FVC in COPD cases; these were the same
showed no evidence for association [90]. Furthermore, in ve SNPs associated with COPD in the family-based asso-
a comparison of 283 continuing smokers with rapid FEV1 ciation analysis. Thus, reasonable evidence for replication
decline versus 307 continuing smokers with slow FEV1 has been found, supporting the utility of future studies of
decline in the Lung Health Study, Ogawa and colleagues SERPINE2 association and function to conrm whether it
found no association between three TGFB1 SNPs (includ- is a valid COPD susceptibility gene.
ing the Leu10Pro SNP) and FEV1 decline [92]. van Diemen
and colleagues studied three TGFB1 SNPs including one
promoter SNP (rs1800469, C-509T), one nonsynonymous PERSPECTIVES ON GENETICS
SNP (rs1982073, Leu10Pro), and one 3
untranslated region
SNP (rs6957) in a general population sample of 1390 indi- OF ASTHMA AND COPD
viduals in the Netherlands who were followed longitudinally
for 25 years [94]. Although they found no association of As demonstrated in this chapter, the research on genetics of
TGFB1 SNPs with FEV1 decline, rs6957 was signicantly asthma and COPD has become a promising new eld. The
more common in 188 subjects meeting criteria for GOLD main goal of the industries and the research institutes is to
Stage II COPD than among the remainder of the cohort nd the genes that increase susceptibility to develop asthma
(p 0.001). Additional studies of TGFB1 in COPD will and COPD.
be required, including larger numbers of SNPs in larger Both in asthma and in COPD the heterogene-
study populations, to denitively conrm or refute the role ity of the diseases with complex hereditary traits and many
of TGFB1 genetic variants in COPD susceptibility. (sub)phenotypes are obstacles to nd the right genetic infor-
mation easily. Although a rapidly increasing number of genes
are being implicated in asthma and COPD pathogenesis,
SERPINE2: Integrating genomics and the functional variants in these genes have not been deni-
genetics in COPD tively identied; this is a major area requiring investigation.
In the pathogenesis of asthma and COPD it is reasonable to
Although genetic linkage studies can point to chromo- suspect that the interactions between multiple genes and
somal regions that are likely to include susceptibility genes multiple environmental triggers are important. This will
for a complex disease, such regions typically contain many require collaboration between many researchers combining
genes. In addition to the approaches of selecting a biologi- their cohorts that have been carefully phenotyped for asthma
cally plausible positional candidate gene (as for TGFB1) and COPD. By combining large cohorts with systematic
or dense genotyping of SNP markers (not yet reported for analyses of environmental risk factors, comprehensive assess-
COPD) within linkage regions, assessment of gene expres- ment of genetic polymorphisms (including genome-wide
sion can point to promising candidate genes that would SNP genotyping), and genetic analysis with advanced statis-
not have been suspected based on our current knowledge tical techniques, we will better understand the genetic deter-
of disease pathobiology. DeMeo and colleagues integrated minants and heterogeneity of asthma and COPD. Hopefully
gene expression microarray results from two data sets one this will lead to new treatments, intervention strategies, and
data set analyzed mouse lung development and the other nally prevention of asthma and COPD.
data set compared gene expression in human lung tissue
for the genes located within their linkage region to airway
obstruction on chromosome 2q [95]. Based on these gene
expression results, SERPINE2 (Serpin Peptidase Inhibitor,
ACKNOWLEDGEMENTS
Clade E, Member 2) was selected as a candidate gene, and
48 SNPs were genotyped in a family-based cohort (949 Research on asthma and COPD in Groningen is supported
individuals from 127 Boston Early-Onset COPD Study by the Netherlands Asthma Foundation, ZonMW and
pedigrees) and a casecontrol cohort (304 COPD cases KNAW. Dr. Silverman was supported by NIH grants R01
and 441 control smokers). Eighteen SERPINE2 SNPs HL075478 and HL68926.

47
Asthma and COPD: Basic Mechanisms and Clinical Management

References 19. Engle LJ, Simpson CL, Landers JE. Using high-throughput SNP
technologies to study cancer. Oncogene 25: 1594601, 2006.
20. Dewan A, Liu M, Hartman S, Zhang SS, Liu DT, Zhao C, Tam PO,
1. Holberg CJ, Elston RC, Halonen M, Wright AL, Taussig LM, Morgan Chan WM, Lam DS, Snyder M, Barnstable C, Pang CP, Hoh J.
WJ, Martinez FD. Segregation analysis of physician-diagnosed asthma HTRA1 promoter polymorphism in wet age-related macular degen-
in hispanic and non-hispanic white families. Am J Respir Crit Care Med eration. Science 314: 98992, 2006.
154: 14450, 1996. 21. Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, Boutin P,
2. Laurell CB, Eriksson S. The electrophoretic 1-globulin pattern of serum Vincent D, Belisle A, Hadjadj S, Balkau B, Heude B, Charpentier G,
in 1-antitrypsin deciency. Scand J Clin Lab Invest 15: 13240, 1963. Hudson TJ, Montpetit A, Pshezhetsky AV, Prentki M, Posner BI,
3. DeMeo DL, Sandhaus RA, Barker AF, Brantly ML, Eden E, Balding DJ, Meyre D, Polychronakos C, Froguel P. A genome-wide
McElvaney NG, Rennard S, Burchard EG, Stocks JM, Stoller JK, association study identies novel risk loci for type 2 diabetes. Nature
Strange C, Turino GM, Campbell EJ, Silverman EK. Determinants 445: 88185, 2007.
of airow obstruction in severe alpha 1-antitrypsin deciency. Thorax 22. Moatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S,
62: 80613. 2007. Depner M, von Berg A, Bufe A, Rietschel E, Heinzmann A, Simma B,
4. Lewitter FI, Tager IB, McGue M, Tishler PV, Speizer FE. Genetic Frischer T, Willis-Owen SA, Wong KC, Illig T, Vogelberg C, Weiland
and environmental determinants of level of pulmonary function. Am J SK, von Mutius E, Abecasis GR, Farrall M, Gut IG, Lathrop GM,
Epidemiol 120: 51829, 1984. Cookson WO. Genetic variants regulating ORMDL3 expression con-
5. Silverman EK, Chapman HA, Drazen JM, Weiss ST, Rosner B, tribute to the risk of childhood asthma. Nature 448: 47073, 2007.
Campbell EJ, ODonnell WJ, Reilly JJ, Ginns L, Mentzer S, Wain J, 23. Lander E, Kruglyak L. Genetic dissection of complex traits: Guidelines
Speizer FE. Genetic epidemiology of severe, early-onset chronic for interpreting and reporting linkage results. Nat Genet 11: 24147, 1995.
obstructive pulmonary disease: Risk to relatives for airow obstruction 24. Celedon JC, Raby B, Weiss ST et al. (eds). Asthma genetics. In:
and chronic bronchitis. Am J Respir Crit Care Med 157: 177078, 1998. Silverman EK, Shapiro SD, Lomas DA, and Weiss ST. Respiratory
6. DeMeo DL, Carey VJ, Chapman HA, Reilly JJ, Ginns LC, Speizer Genetics, pp. 21996. London: Hodder Arnold, 2005.
FE, Weiss ST, Silverman EK. Familial aggregation of FEF(25-75) and 25. Van Eerdewegh P et al. Association of the ADAM33 gene with asthma
FEF(25-75)/FVC in families with severe, early onset COPD. Thorax and bronchial hyperresponsiveness. Nature 418: 42630, 2002.
59: 396400, 2004. 26. Allen M et al. Positional cloning of a novel gene inuencing asthma
7. McCloskey SC, Patel BD, Hinchlie SJ, Reid ED, Wareham NJ, from chromosome 2q14. Nat Genet 35: 25863, 2003.
Lomas DA. Siblings of patients with severe chronic obstructive pulmo- 27. Zhang Y, Leaves NI, Anderson GG, Ponting CP, Broxholme J, Holt R,
nary disease have a signicant risk of airow obstruction. Am J Respir Edser P, Bhattacharyya S, Dunham A, Adcock IM, Pulleyn L,
Crit Care Med 164: 141924, 2001. Barnes PJ, Harper JI, Abecasis G, Cardon L, White M, Burton J,
8. Pennisi E. Human genome. Finally, the book of life and instructions Matthews L, Mott R, Ross M, Cox R, Moatt MF, Cookson WO.
for navigating it. Science 288: 23047, 2000. Positional cloning of a quantitative trait locus on chromosome 13q14
9. Pennisi E. Genetics. Working the (gene count) numbers: Finally, a rm that inuences immunoglobulin E levels and asthma. Nat Genet 34:
answer?. Science 316: 1113, 2007. 18186, 2003.
10. Schmidt-Weber CB. Gene expression proling in allergy and asthma. 28. Laitinen T, Polvi A, Rydman P, Vendelin J, Pulkkinen V, Salmikangas P,
Chem Immunol Allergy 91: 18894, 2006. Makela S, Rehn M, Pirskanen A, Rautanen A, Zucchelli M, Gullsten H,
11. Nomura I, Gao B, Boguniewicz M, Darst MA, Travers JB, Leung DY. Leino M, Alenius H, Petays T, Haahtela T, Laitinen A, Laprise C,
Distinct patterns of gene expression in the skin lesions of atopic Hudson TJ, Laitinen LA, Kere J. Characterization of a common sus-
dermatitis and psoriasis: A gene microarray analysis. J Allergy Clin ceptibility locus for asthma-related traits. Science 304: 3004, 2004.
Immunol 112: 1195202, 2003. 29. Nicolae D, Cox NJ, Lester LA, Schneider D, Tan Z, Billstrand C,
12. Chavanas S, Garner C, Bodemer C, Ali M, Teillac DH, Wilkinson J, Kuldanek S, Donfack J, Kogut P, Patel NM, Goodenbour J, Howard T,
Bonafe JL, Paradisi M, Kelsell DP, Ansai S, Mitsuhashi Y, Larregue M, Wolf R, Koppelman GH, White SR, Parry R, Postma DS, Meyers D,
Leigh IM, Harper JI, Taieb A, Prost Y, Cardon LR, Hovnanian A. Bleecker ER, Hunt JS, Solway J, Ober C. Fine mapping and positional
Localization of the Netherton syndrome gene to chromosome 5q32, candidate studies identify HLA-G as an asthma susceptibility gene on
by linkage analysis and homozygosity mapping. Am J Hum Genet 66: chromosome 6p21. Am J Hum Genet 76: 34957, 2005.
91421, 2000. 30. Noguchi E, Yokouchi Y, Zhang J, Shibuya K, Shibuya A, Bannai M,
13. Walley AJ, Chavanas S, Moatt MF, Esnouf RM, Ubhi B, Lawrence R, Tokunaga K, Doi H, Tamari M, Shimizu M, Shirakawa T, Shibasaki M,
Wong K, Abecasis GR, Jones EY, Harper JI, Hovnanian A, Cookson Ichikawa K, Arinami T. Positional identication of an asthma suscep-
WO. Gene polymorphism in Netherton and common atopic disease. tibility gene on human chromosome 5q33. Am J Respir Crit Care Med
Nat Genet 29: 17578, 2001. 172: 18388, 2005.
14. Kabesch M, Carr D, Weiland SK, von Mutius E. Association between 31. Silverman EK, Palmer LJ, Mosley JD, Barth M, Senter JM, Brown A,
polymorphisms in serine protease inhibitor, kazal type 5 and asthma Drazen JM, Kwiatkowski DJ, Chapman HA, Campbell EJ,
phenotypes in a large German population sample. Clin Exp Allergy 34: Province MA, Rao DC, Reilly JJ, Ginns LC, Speizer FE, Weiss ST.
34045, 2004. Genomewide linkage analysis of quantitative spirometric phenotypes
15. Jongepier H, Koppelman GH, Nolte IM, Bruinenberg M, in severe early-onset chronic obstructive pulmonary disease. Am J Hum
Bleecker ER, Meyers DA, te Meerman GJ, Postma DS. Polymorphisms Gen 70: 122939, 2002.
in SPINK5 are not associated with asthma in a Dutch population. 32. DeMeo DL, Celedon JC, Lange C, Reilly JJ, Chapman HA, Sylvia JS,
J Allergy Clin Immunol 115: 48692, 2005. Speizer FE, Weiss ST, Silverman EK. Genome-wide linkage of forced
16. Kelleher CM, Silverman EK, Broekelmann T, Litonjua AA, Hernandez mid-expiratory ow in chronic obstructive pulmonary disease. Am J
M, Sylvia JS, Stoler J, Reilly JJ, Chapman HA, Speizer FE, Weiss ST, Respir Crit Care Med 170: 1294301, 2004.
Mecham RP, Raby BA. A functional mutation in the terminal exon of 33. Malhotra A, Peier AP, Ryujin DT, Elsner T, Kanner RE, Leppert MF,
elastin in severe, early-onset chronic obstructive pulmonary disease. Hasstedt SJ. Further evidence for the role of genes on chromosome 2
Am J Respir Cell Mol Biol 33: 35562, 2005. and chromosome 5 in the inheritance of pulmonary function. Am J
17. Altshuler D, Brooks LD, Chakravarti A, Collins FS, Daly MJ, Respir Crit Care Med 168: 55661, 2003.
Donnelly P. A haplotype map of the human genome. Nature 437: 34. Postma DS, Meyers DA, Jongepier H, Howard TD, Koppelman GH,
1299320, 2005. Bleecker ER. Genomewide screen for pulmonary function in 200 families
18. Syvanen AC. Toward genome-wide SNP genotyping. Nat Genet ascertained for asthma. Am J Respir Crit Care Med 172: 44652, 2005.
37(Suppl): S510, 2005.

48
Genetics of Asthma and COPD 4
35. Ober C, Hoffjan S. Asthma genetics 2006: The long and winding road 53. Heinzmann A et al. Genetic variants of IL-13 signalling and human
to gene discovery. Genes Immun 7: 95100, 2006. asthma and atopy. Hum Mol Genet 9: 54959, 2000.
36. Hoffjan S, Nicolae D, Ober C. Association studies for asthma and atopic 54. Graves PE, Kabesch M, Halonen M, Holberg CJ, Baldini M, Fritzsch
diseases: A comprehensive review of the literature. Respir Res 4: 14, 2003. C, Weiland SK, Erickson RP, von Mutius E, Martinez FD. A cluster
37. Kere J, Laitinen T. Positionally cloned susceptibility genes in allergy of seven tightly linked polymorphisms in the IL-13 gene is associ-
and asthma. Curr Opin Immunol 16: 68994, 2004. ated with total serum IgE levels in three populations of white children.
38. Lind DL, Choudhry S, Ung N, Ziv E, Avila PC, Salari K, Ha C, J Allergy Clin Immunol 105: 50613, 2000.
Lovins EG, Coyle NE, Nazario S, Casal J, Torres A, Rodriguez- 55. Liu X, Nickel R, Beyer K, Wahn U, Ehrlich E, Freidho LR, Bjorksten B,
Santana JR, Matallana H, Lilly CM, Salas J, Selman M, Boushey HA, Beaty TH, Huang SK. An IL13 coding region variant is associated with
Weiss ST, Chapela R, Ford JG, Rodriguez-Cintron W, Silverman EK, a high total serum IgE level and atopic dermatitis in the German mul-
Sheppard D, Kwok PY, Gonzalez Burchard E. ADAM33 is not associ- ticenter atopy study (MAS-90). J Allergy Clin Immunol 106: 16770,
ated with asthma in Puerto Rican or Mexican populations. Am J Respir 2000.
Crit Care Med 168: 131216, 2003. 56. Takabayashi A, Ihara K, Sasaki Y, Suzuki Y, Nishima S, Izuhara K,
39. Howard TD, Postma DS, Jongepier H, Moore WC, Koppelman GH, Hamasaki N, Hara T. Childhood atopic asthma: Positive association
Zheng SL, Xu J, Bleecker ER, Meyers DA. Association of a disintegrin with a polymorphism of IL-4 receptor alpha gene but not with that of
and metalloprotease 33 (ADAM33) gene with asthma in ethnically IL-4 promoter or Fc epsilon receptor I beta gene. Exp Clin Immunogenet
diverse populations. J Allergy Clin Immunol 112: 71722, 2003. 17: 6370, 2000.
40. Raby BA, Silverman EK, Kwiatkowski DJ, Lange C, Lazarus R, 57. Shirakawa I, Deichmann KA, Izuhara I, Mao I, Adra CN, Hopkin
Weiss ST. ADAM33 polymorphisms and phenotype associations in JM. Atopy and asthma: Genetic variants of IL-4 and IL-13 signalling.
childhood asthma. J Allergy Clin Immunol 113: 107178, 2004. Immunol Today 21: 6064, 2000.
41. Cheng SL, Yu CJ, Chen CJ, Yang PC. Genetic polymorphism of 58. Noguchi E, Nukaga-Nishio Y, Jian Z, Yokouchi Y, Kamioka M,
epoxide hydrolase and glutathione S-transferase in COPD. Eur Respir J Yamakawa-Kobayashi K, Hamaguchi H, Matsui A, Shibasaki M,
23: 81824, 2004. Arinami T. Haplotypes of the 5 region of the IL-4 gene and SNPs in
42. Wang P, Liu QJ, Li JS, Li HC, Wei CH, Guo CH, Gong YQ. Lack of the intergene sequence between the IL-4 and IL-13 genes are associ-
association between ADAM33 gene and asthma in a Chinese popula- ated with atopic asthma. Hum Immunol 62: 125157, 2001.
tion. Int J Immunogenet 33: 3036, 2006. 59. Leung TF, Tang NL, Chan IH, Li AM, Ha G, Lam CW. A polymor-
43. Lee JH, Park HS, Park SW, Jang AS, Uh ST, Rhim T, Park CS, Hong phism in the coding region of interleukin-13 gene is associated with
SJ, Holgate ST, Holloway JW, Shin HD. ADAM33 polymorphism: atopy but not asthma in Chinese children. Clin Exp Allergy 31: 151521,
Association with bronchial hyper-responsiveness in Korean asthmatics. 2001.
Clin Exp Allergy 34: 86065, 2004. 60. Howard TD, Whittaker PA, Zaiman AL, Koppelman GH, Xu J, Hanley
44. Schedel M, Depner M, Schoen C, Weiland SK, Vogelberg C, MT, Meyers DA, Postma DS, Bleecker ER. Identication and associa-
Niggemann B, Lau S, Illig T, Klopp N, Wahn U, von Mutius E, Nickel R, tion of polymorphisms in the interleukin-13 gene with asthma and atopy
Kabesch M. The role of polymorphisms in ADAM33, a disintegrin in a Dutch population. Am J Respir Cell Mol Biol 25: 37784, 2001.
and metalloprotease 33, in childhood asthma and lung function in two 61. Wjst M, Kruse S, Illig T, Deichmann K. Asthma and IL-4 receptor
German populations. Respir Res 7: 91, 2006. alpha gene variants. Eur J Immunogenet 29: 26368, 2002.
45. Jongepier H, Boezen HM, Dijkstra A, Howard TD, Vonk JM, 62. Howard TD, Koppelman GH, Xu J, Zheng SL, Postma DS, Meyers
Koppelman GH, Zheng SL, Meyers DA, Bleecker ER, Postma DS. DA, Bleecker ER. Genegene interaction in asthma: IL4RA and IL13
Polymorphisms of the ADAM33 gene are associated with accelerated in a Dutch population with asthma. Am J Hum Genet 70: 23036, 2002.
lung function decline in asthma. Clin Exp Allergy 34: 75760, 2004. 63. DeMeo DL, Lange C, Silverman EK, Senter JM, Drazen JM,
46. Werner M, Herbon N, Gohlke H, Altmuller J, Knapp M, Heinrich J, Barth MJ, Laird N, Weiss ST. Univariate and multivariate family-based
Wjst M. Asthma is associated with single-nucleotide polymorphisms association analysis of the IL- 13 ARG130GLN polymorphism in the
in ADAM33. Clin Exp Allergy 34: 2631, 2004. Childhood Asthma Management program. Genet Epidem 23: 33548,
47. Simpson A, Maniatis N, Jury F, Cakebread JA, Lowe LA, Holgate 2002.
ST, Woodcock A, Ollier WE, Collins A, Custovic A, Holloway JW, 64. He JQ, Chan-Yeung M, Becker AB, Dimich-Ward H, Ferguson AC,
John SL. Polymorphisms in a disintegrin and metalloprotease 33 Manfreda J, Watson WT, Sandford AJ. Genetic variants of the IL13
(ADAM33) predict impaired early-life lung function. Am J Respir Crit and IL4 genes and atopic diseases in at-risk children. Genes Immun 4:
Care Med 172: 5560, 2005. 38589, 2003.
48. Blakey J, Halapi E, Bjornsdottir US, Wheatley A, Kristinsson S, 65. Liu X, Beaty TH, Deindl P, Huang SK, Lau S, Sommerfeld C, Fallin
Upmanyu R, Stefansson K, Hakonarson H, Hall IP. Contribution of MD, Kao WH, Wahn U, Nickel R. Associations between total serum
ADAM33 polymorphisms to the population risk of asthma. Thorax 60: IgE levels and the 6 potentially functional variants within the genes
27476, 2005. IL4, IL13, and IL4RA in German children: The German Multicenter
49. Noguchi E, Ohtsuki Y, Tokunaga K, Yamaoka-Sageshima M, Ichikawa Atopy Study. J Allergy Clin Immunol 112: 38288, 2003.
K, Aoki T, Shibasaki M, Arinami T. ADAM33 polymorphisms are 66. Beghe B, Barton S, Rorke S, Peng Q, Sayers I, Gaunt T, Keith TP,
associated with asthma susceptibility in a Japanese population. Clin Clough JB, Holgate ST, Holloway JW. Polymorphisms in the inter-
Exp Allergy 36: 6028, 2006. leukin-4 and interleukin-4 receptor alpha chain genes confer susceptibil-
50. Deichmann KA, Heinzmann A, Forster J, Dischinger S, Mehl C, ity to asthma and atopy in a Caucasian population. Clin Exp Allergy 33:
Brueggenolte E, Hildebrandt F, Moseler M, Kuehr J. Linkage and 111117, 2003.
allelic association of atopy and markers anking the IL4-receptor gene. 67. Hummelshoj T, Bodtger U, Datta P, Malling HJ, Oturai A, Poulsen LK,
Clin Exp Allergy 28: 15155, 1998. Ryder LP, Sorensen PS, Svejgaard E, Svejgaard A. Association between
51. Celedon JC, Soto-Quiros ME, Palmer LJ, Senter J, Mosley J, Silverman an interleukin-13 promoter polymorphism and atopy. Eur J Immunogenet
EK, Weiss ST. Lack of association between a polymorphism in the 30: 35559, 2003.
interleukin-13 gene and total serum immunoglobulin E level among 68. Lee SG, Kim BS, Kim JH, Lee SY, Choi SO, Shim JY, Hong TJ,
nuclear families in Costa Rica. Clin Exp Allergy 32: 38790, 2002. Hong SJ. Genegene interaction between interleukin-4 and inter-
52. van der Pouw Kraan TC, van Veen A, Boeije LC, van Tuyl SA, de Groot leukin-4 receptor alpha in Korean children with asthma. Clin Exp
ER, Stapel SO, Bakker A, Verweij CL, Aarden LA, van der Zee JS. Allergy 34: 12028, 2004.
An IL-13 promoter polymorphism associated with increased risk of 69. Hoffjan S, Ostrovnaja I, Nicolae D, Newman DL, Nicolae R,
allergic asthma. Genes Immun 1: 6165, 1999. Gangnon R, Steiner L, Walker K, Reynolds R, Greene D, Mirel D,

49
Asthma and COPD: Basic Mechanisms and Clinical Management

Gern JE, Lemanske RF Jr., Ober C. Genetic variation in immu- 87. Young RP, Hopkins R, Black PN, Eddy C, Wu L, Gamble GD, Mills
noregulatory pathways and atopic phenotypes in infancy. J Allergy Clin GD, Garrett JE, Eaton TE, Rees MI. Functional variants of antioxi-
Immunol 113: 51118, 2004. dant genes in smokers with COPD and in those with normal lung
70. Nieters A, Linseisen J, Becker N. Association of polymorphisms in function. Thorax 61: 39499, 2006.
Th1, Th2 cytokine genes with hayfever and atopy in a subsample of 88. Juul K, Tybjaerg-Hansen A, M S, Lange P, Nordestgaard BG.
EPIC-Heidelberg. Clin Exp Allergy 34: 34653, 2004. Genetically increased antioxidant protection and decreased chronic
71. Vladich FD, Brazille SM, Stern D, Peck ML, Ghittoni R, Vercelli D. obstructive pulmonary disease. Am J Respir Crit Care Med, 173: 858
IL-13 R130Q, a common variant associated with allergy and asthma, 64, 2006.
enhances eector mechanisms essential for human allergic inamma- 89. Wu L, Chau J, Young RP, Pokorny V, Mills GD, Hopkins R, McLean L,
tion. J Clin Invest 115: 74754, 2005. Black PN. Transforming growth factor-beta1 genotype and susceptibility
72. Kabesch M, Schedel M, Carr D, Woitsch B, Fritzsch C, Weiland SK, von to chronic obstructive pulmonary disease. Thorax 59: 12629, 2004.
Mutius E. IL-4/IL-13 pathway genetics strongly inuence serum IgE 90. Yoon HI, Silverman EK, Lee HW, Yoo CG, Lee CT, Chung HS,
levels and childhood asthma. J Allergy Clin Immunol 117: 26974, 2006. Kim YW, Han SK, Shim YS, Yim JJ. Lack of association between
73. Chan IH, Leung TF, Tang NL, Li CY, Sung YM, Wong GW, Wong COPD and transforming growth factor-b1 (TGFB1) genetic poly-
CK, Lam CW. Genegene interactions for asthma and plasma total morphisms in Koreans. Int J Tuberc Lung Dis, 10: 50409, 2006.
IgE concentration in Chinese children. J Allergy Clin Immunol 117: 91. Celedon JC, Lange C, Raby BA, Litonjua AA, Palmer LJ, DeMeo
12733, 2006. DL, Reilly JJ, Kwiatkowski DJ, Chapman HA, Laird N, Sylvia JS,
74. Hunninghake GM, Soto-Quiros ME, Avila L, Su J, Murphy A, Hernandez M, Speizer FE, Weiss ST, Silverman EK. The transform-
Demeo DL, Ly NP, Liang C, Sylvia JS, Klanderman BJ, Lange C, ing growth factor-beta1 (TGFB1) gene is associated with chronic
Raby BA, Silverman EK, Celedon JC. Polymorphisms in IL13, total obstructive pulmonary disease (COPD). Hum Mol Genet 13: 164956,
IgE, eosinophilia, and asthma exacerbations in childhood. J Allergy Clin 2004.
Immunol 120: 8490, 2007. 92. Ogawa E, Ruan J, Connett JE, Anthonisen NR, Pare PD, Sandford AJ.
75. Smith CAD, Harrison DJ. Association between polymorphism in gene Transforming growth factor-beta1 polymorphisms, airway responsive-
for microsomal epoxide hydrolase and susceptibility to emphysema. The ness and lung function decline in smokers. Respir Med 101: 93843,
Lancet 350: 63033, 1997. 2007.
76. Yoshikawa M, Hiyama K, Ishioka S, Maeda H, Maeda A, Yamakido 93. Su ZG, Wen FQ, Feng YL, Xiao M, Wu XL. Transforming growth
M. Microsomal epoxide hydrolase genotypes and chronic obstructive factor-beta1 gene polymorphisms associated with chronic obstruc-
pulmonary disease in Japanese. Int J Mol Med 5: 4953, 2000. tive pulmonary disease in Chinese population. Acta Pharmacol Sin 26:
77. Brogger J, Steen VM, Eiken HG, Gulsvik A, Bakke P. Genetic asso- 71420, 2005.
ciation between COPD and polymorphisms in TNF, ADRB2 and 94. van Diemen CC, Postma DS, Vonk JM, Bruinenberg M, Nolte IM,
EPHX1. Eur Respir J 27: 68288, 2006. Boezen HM. Decorin and TGF-beta1 polymorphisms and develop-
78. Yim JJ, Park GY, Lee CT, Kim YW, Han SK, Shim YS, Yoo CG. ment of COPD in a general population. Respir Res 7: 89, 2006.
Genetic susceptibility to chronic obstructive pulmonary disease in 95. DeMeo D, Mariani TJ, Lange C, Srisuma SS, Litonjua AA,
Koreans: Combined analysis of polymorphic genotypes for microsomal Celedon JC, Lake SL, Reilly JJ, Chapman HA, Mecham BH, Haley
epoxide hydrolase and glutathione S-transferase M1 and T1. Thorax 55: KJ, Sylvia JS, Sparrow D, Spira A, Beane J, Pinto-Plata V, Speizer FE,
12125, 2000. Shapiro SD, Weiss ST, Silverman EK. The SERPINE2 gene is asso-
79. Sandford AJ, Chagani T, Weir TD, Connett JE, Anthonisen NR, Pare ciated with chronic obstructive pulmonary disease. Am J Hum Genet
PD. Susceptibility genes for rapid decline of lung function in the Lung 178: 25364, 2006.
Health Study. Am J Respir Crit Care Med 163: 46973, 2001. 96. Chappell S, Daly L, Morgan K, Baranes TG, Roca J, Rabinovich R,
80. Takeyabu K, Yamaguchi E, Suzuki I, Nishimura M, Hizawa N, Millar A, Donnelly SC, Keatings V, MacNee W, Stolk J, Hiemstra PS,
Kamakami Y. Gene polymorphism for microsomal epoxide hydrolase Miniati M, Monti S, OConnor CM, Kalsheker N. The SERPINE2
and susceptibility to emphysema in a Japanese population. Eur Respir J gene and chronic obstructive pulmonary disease. Am J Hum Genet 79:
15: 89194, 2000. 18486, 2006, author reply 1867.
81. Hersh CP, Demeo DL, Lange C, Litonjua AA, Reilly JJ, Kwiatkowski 97. Zhu G, Warren L, Aponte J, Gulsvik A, Bakke P, ICGN
D, Laird N, Sylvia JS, Sparrow D, Speizer FE, Weiss ST, Silverman Investigators, Anderson WH, Lomas DA, Silverman EK, Pillai SG.
EK. Attempted replication of reported chronic obstructive pulmo- The SERPINE2 gene is associated with chronic obstructive pulmo-
nary disease candidate gene associations. Am J Respir Cell Mol Biol 33: nary disease in two large populations. Am J Respir Crit Care Med 176:
7178, 2005. 16773, 2007.
82. Cheng L, Enomoto T, Hirota T, Shimizu M, Takahashi N, Akahoshi 98. Dijkstra A, Vonk JM, Jongepier H, Koppelman GH, Schouten JP,
M, Matsuda A, Dake Y, Doi S, Enomoto K, Yamasaki A, Fukuda S, ten Hacken NH, Timens W, Postma DS. Lung function decline in
Mao XQ, Hopkin JM, Tamari M, Shirakawa T. Polymorphisms in asthma: Association with inhaled corticosteroids, smoking and sex.
ADAM33 are associated with allergic rhinitis due to Japanese cedar Thorax 61: 10510, 2006.
pollen. Clin Exp Allergy 34: 1192201, 2004. 99. van Diemen CC, Postma DS, Vonk JM, Bruinenberg M, Schouten JP,
83. Ishii T, Matsuse T, Teramoto S, Matsui H, Miyao M, Hosoi T, Boezen HM. A disintegrin and metalloprotease 33 polymorphisms
Takahashi H, Fukuchi Y, Ouchi Y. Glutathione S-transferase P1 and lung function decline in the general population. Am J Respir Crit
(GSTP1) polymorphism in patients with chronic obstructive pulmo- Care Med 172: 32933, 2005.
nary disease. Thorax 54: 69396, 1999. 100. Gosman MM, Boezen HM, van Diemen CC, Snoeck-Stroband JB,
84. Yim JJ, Yoo CG, Lee CT, Kim YW, Han SK, Shim YS. Lack of associ- Lapperre TS, Hiemstra PS, Ten Hacken NH, Stolk J, Postma DS.
ation between glutathione S-transferase P1 polymorphism and COPD A disintegrin and metalloprotease 33 and chronic obstructive pulmo-
in Koreans. Lung 180: 11925, 2002. nary disease pathophysiology. Thorax 62: 24247, 2007.
85. He JQ, Connett JE, Anthonisen NR, Pare PD, Sandford AJ. 101. Vercelli D. Genetics of IL-13 and functional relevance of IL-13 vari-
Glutathione S-transferase variants and their interaction with smoking ants. Curr Opin Allergy Clin Immunol 2: 38993, 2002.
on lung function. Am J Respir Crit Care Med 170: 38894, 2004. 102. Kruse S, Braun S, Deichmann KA. Distinct signal transduction proc-
86. Calikoglu M, Tamer L, Ates Aras N, Karakas S, Ercan B. The esses by IL-4 and IL-13 and inuences from the Q551R variant of
association between polymorphic genotypes of glutathione S-trans- the human IL-4 receptor alpha chain. Respir Res 3: 24, 2002.
ferases and COPD in the Turkish population. Biochem Genet 44: 103. LeVan TD, Von Essen S, Romberger DJ, Lambert GP,
30719, 2006. Martinez FD, Vasquez MM, Merchant JA. Polymorphisms in the

50
Genetics of Asthma and COPD 4
CD14 gene associated with pulmonary function in farmers. Am J 121. Drazen JM, Israel E, Boushey HA, Chinchilli VM, Fahy JV, Fish JE,
Respir Crit Care Med 171: 77379, 2005. Lazarus SC, Lemanske RF, Martin RJ, Peters SP, Sorkness C, Szeer
104. Baldini M, Lohman IC, Halonen M, Erickson RP, Holt PG, SJ. Comparison of regularly scheduled with as-needed use of albuterol
Martinez FD. A Polymorphism* in the 5 anking region of the CD14 in mild asthma. Asthma Clinical Research Network. N Engl J Med 33:
gene is associated with circulating soluble CD14 levels and with total 84147, 1996.
serum immunoglobulin E. Am J Respir Cell Mol Biol 20: 97683, 1999. 122. Israel E, Drazen JM, Liggett SB, Boushey HA, Cherniack RM,
105. Martinez FD. Geneenvironment interactions in asthma and aller- Chinchilli VM, Cooper DM, Fahy JV, Fish JE, Ford JG, Kraft M,
gies: A new paradigm to understand disease causation. Immunol Kunselman S, Lazarus SC, Lemanske RF, Martin RJ, McLean DE,
Allergy Clin North Am 25: 70921, 2005. Peters SP, Silverman EK, Sorkness CA, Szeer SJ, Weiss ST, Yandava
106. Liu AH, Leung DY. Renaissance of the hygiene hypothesis. J Allergy CN. The eect of polymorphisms of the beta(2)-adrenergic receptor
Clin Immunol 117: 106366, 2006. on the response to regular use of albuterol in asthma. Am J Respir Crit
107. Zambelli-Weiner A, Ehrlich E, Stockton ML, Grant AV, Zhang S, 3are Med 162: 7580, 2000.
Levett PN, Beaty TH, Barnes KC. Evaluation of the CD14/-260 122. Israel E, Chinchilli VM, Ford JG, Boushey HA, Cherniack R, Craig
polymorphism and house dust endotoxin exposure in the Barbados TJ, Deykin A, Fagan JK, Fahy JV, Fish J, Kraft M, Kunselman SJ,
Asthma Genetics Study. J Allergy Clin Immunol 115: 12039, 2005. Lazarus SC, Lemanske RF Jr., Liggett SB, Martin RJ, Mitra N,
108. Yang IA, Fong KM, Holgate ST, Holloway JW. The role of Toll- Peters SP, Silverman E, Sorkness CA, Szeer SJ, Wechsler ME,
like receptors and related receptors of the innate immune system in Weiss ST, Drazen JM. Use of regularly scheduled albuterol treatment
asthma. Curr Opin Allergy Clin Immunol 6: 2328, 2006. in asthma: Genotype-stratied, randomised, placebo-controlled cross-
109. Eder W, Klimecki W, Yu L, von Mutius E, Riedler J, Braun- over trial. The Lancet 364: 150512, 2004.
Fahrlander C, Nowak D, Martinez FD. Opposite eects of CD 14/- 124. Taylor DR, Drazen JM, Herbison GP, Yandava CN, Hancox RJ, Town
260 on serum IgE levels in children raised in dierent environments. GI. Asthma exacerbations during long term beta agonist use: Inuence
J Allergy Clin Immunol 116: 6017, 2005. of beta(2) adrenoceptor polymorphism. Thorax 55: 76267, 2000.
110. Leynaert B, Guilloud-Bataille M, Soussan D, Benessiano J, Guenegou 125. Lee DK, Currie GP, Hall IP, Lima JJ, Lipworth BJ. The arginine-16
A, Pin I, Neukirch F. Association between farm exposure and atopy, beta2-adrenoceptor polymorphism predisposes to bronchoprotective
according to the CD14 C-159T polymorphism. J Allergy Clin subsensitivity in patients treated with formoterol and salmeterol. Br J
Immunol 118: 65865, 2006. Clin Pharmacol 57: 6875, 2004.
111. Choudhry S, Avila PC, Nazario S, Ung N, Kho J, Rodriguez-Santana 126. Wechsler ME, Lehman E, Lazarus SC, Lemanske RF Jr., Boushey HA,
JR, Casal J, Tsai HJ, Torres A, Ziv E, Toscano M, Sylvia JS, Alioto M, Deykin A, Fahy JV, Sorkness CA, Chinchilli VM, Craig TJ, DiMango E,
Salazar M, Gomez I, Fagan JK, Salas J, Lilly C, Matallana H, Castro Kraft M, Leone F, Martin RJ, Peters SP, Szeer SJ, Liu W, Israel E.
RA, Selman M, Weiss ST, Ford JG, Drazen JM, Rodriguez-Cintron beta-Adrenergic receptor polymorphisms and response to salmeterol.
W, Chapela R, Silverman EK, Burchard EG. CD14 tobacco gene- Am J Respir Crit Care Med 173: 51926, 2006.
environment interaction modies asthma severity and immunoglobu- 127. Bleecker ER, Yancey SW, Baitinger LA, Edwards LD, Klotsman M,
lin E levels in Latinos with asthma. Am J Respir Crit Care Med 172: Anderson WH, Dorinsky PM. Salmeterol response is not aected by
17382, 2005. beta2-adrenergic receptor genotype in subjects with persistent asthma.
112. Lodrup Carlsen KC, Lovik M, Granum B, Mowinckel P, Carlsen J Allergy Clin Immunol 118: 80916, 2006.
KH. Soluble CD14 at 2 yr of age: Gender-related eects of tobacco 128. Nelson HS, Weiss ST, Bleecker ER, Yancey SW, Dorinsky PM. The
smoke exposure, recurrent infections and atopic diseases. Pediatr Salmeterol Multicenter Asthma Research Trial: A comparison of
Allergy Immunol 17: 30412, 2006. usual pharmacotherapy for asthma or usual pharmacotherapy plus sal-
113. Sin DD, Man J, Sharpe H, Gan WQ, Man SF. Pharmacological man- meterol. Chest 129: 1526, 2006.
agement to reduce exacerbations in adults with asthma: A systematic 129. Drysdale CM, McGraw DW, Stack CB, Stephens JC, Judson RS,
review and meta-analysis. JAMA 292: 36776, 2004. Nandabalan K, Arnold K, Ruano G, Liggett SB. Complex promoter
114. Hawkins GA, Tantisira K, Meyers DA, Ampleford EJ, Moore WC, and coding region beta 2-adrenergic receptor haplotypes alter recep-
Klanderman B, Liggett SB, Peters SP, Weiss ST, Bleecker ER. Sequence, tor expression and predict in vivo responsiveness. Proc Natl Acad Sci
haplotype, and association analysis of ADRbeta2 in a multiethnic asthma USA 97: 1048388, 2000.
casecontrol study. Am J Respir Crit Care Med 174: 11019, 2006. 130. Taylor DR, Epton MJ, Kennedy MA, Smith AD, Iles S, Miller AL,
115. Contopoulos-Ioannidis DG, Manoli EN, Ioannidis JP. Meta-analysis Littlejohn MD, Cowan JO, Hewitt T, Swanney MP, Brassett KP,
of the association of beta2-adrenergic receptor polymorphisms with Herbison GP. Bronchodilator response in relation to beta2-adreno-
asthma phenotypes. J Allergy Clin Immunol 115: 96372, 2005. ceptor haplotype in patients with asthma. Am J Respir Crit Care Med
116. Green SA, Turki J, Innis M, Liggett SB. Amino-terminal polymor- 172: 7003, 2005.
phisms of the human beta-2 adrenergic receptor impart distinct agonist- 131. DeMeo DL, Hersh CP, Homan EA, Litonjua AA, Lazarus R,
promoted regulatory properties. Biochemistry 33: 941419, 1994. Sparrow D, Benditt JO, Criner GJ, Make B, Martinez FJ, Scanlon PD,
117. Ohe M, Munakata M, Hizawa N, Itoh A, Doi I, Yamaguchi E, Sciurba FC, Utz JP, Reilly JJ, Silverman EK. Genetic determinants of
Homma Y, Kawakami Y. Beta 2 adrenergic receptor gene restriction emphysema distribution in the National Emphysema Treatment Trial.
fragment length polymorphism and bronchial asthma. Thorax 50: Am J Respir Crit Care Med 176: 4248, 2007.
35359, 1995. 132. Sundberg K, Johansson AS, Stenberg G, Widersten M, Seidel A,
118. Aziz I, McFarlane LC, Lipworth BJ. Comparative trough eects Mannervik B, Jernstrom B. Dierences in the catalytic eciencies of
of formoterol and salmeterol on lymphocyte beta2-adrenoceptor allelic variants of glutathione transferase P1-1 towards carcinogenic
regulation and bronchodilatation. Eur J Clin Pharmacol 55: 43136, diol epoxides of polycyclic aromatic hydrocarbons. Carcinogenesis 19:
1999. 43336, 1998.
119. Martinez FD, Graves PE, Baldini M, Solomon S, Erickson R. 133. Olsen DA, Petersen SV, Oury TD, Valnickova Z, Thogersen IB,
Association between genetic polymorphisms of the beta-2 adrenocep- Kristensen T, Bowler RP, Crapo JD, Enghild JJ. The intracellular pro-
tor and response to albuterol in children with and without a history of teolytic processing of extracellular superoxide dismutase (EC-SOD)
wheezing. J Clin Invest 100: 318488, 1997. is a two-step event. J Biol Chem 279: 2215257, 2004.
120. Lima JJ, Thomason DB, Mohamed MH, Eberle LV, Self TH, 134. Suthanthiran M, Li B, Song JO, Ding R, Sharma VK, Schwartz JE,
Johnson JA. Impact of genetic polymorphisms of the beta2-adren- August P. Transforming growth factor-beta 1 hyperexpression in
ergic receptor on albuterol bronchodilator pharmacodynamics. Clin African-American hypertensives: A novel mediator of hypertension
Pharmacol Ther 65: 51925, 1999. and/or target organ damage. Proc Natl Acad Sci USA 97: 347984, 2000.

51
Physiology and 2
PART

Pathology of
Asthma and COPD
Pulmonary Physiology
5
CHAPTER

INTRODUCTION volume which distinguish these airway disor-


ders from restrictive processes of the respiratory Charles G. Irvin
system that reduce lung volumes. Lung volumes
Asthma is an inammatory disease principally have important bearing on the degree of dis- Department of Medicine and
of the small airways but current understanding is ease since increases in the residual volume (RV) Physiology, University of Vermont
that all the airways are involved [1]. The physi- occur relatively early even with mild airway dis- College of Medicine, Burlington, VT, USA
ological manifestations of lung inammation are ease whereas increases in the total lung capacity
reversible airow limitation and airow limita- (TLC) are usually an indication of more severe
tion that uctuates widely with time; asthma or long-standing disease (Fig. 5.1) [46].
can also result in persistent loss of lung function. The most useful lung volumes and capaci-
In contrast, airow obstruction in patients with ties to measure are those that assess the physical
chronic obstructive pulmonary disease (COPD) limits of the lung and chest wall and dene the
presents with both reversible and irreversible air- extremes of the vital capacity (VC); TLC and
ow limitation, the latter linked to loss of static RV. The volume in the lung at end expiratory
elastic recoil due in part to the destruction of the position, the functional residual capacity (FRC),
architecture of the lung. The airow limitation is important because of the inuence that lung
of COPD has much less temporal or periodic volume has on airway caliber and in turn on
variation over the short term but patients with airow resistance during eupnea. TLC, FRC,
COPD usually exhibit steady consistent losses of and RV are boundary conditions of respiration
lung function over a period of years. In reality the and accordingly provide the most information
clinical presentation of both COPD and asthma about the capacity and function of the respira-
is often quite variable and the pathophysiologi- tory system [6]. Assessing the changes in these
cal data alone do not allow a denitive diagnosis. lung volumes to evaluate disease severity and
This confusing presentation of patients with fea- response to therapy allows one to distinguish
tures of both asthma and COPD is in keeping between obstructive lung disease and restrictive
with the overlap observed in the epidemiology lung disease (Table 5.1) [8, 9].
of these diseases [2, 3]. The focus of this chap- Obstructive lung diseases such as chronic
ter is on the pathophysiological presentation of bronchitis, asthma, and emphysema all result
COPD and asthma and the features that distin- in increases in lung volume. An increase in
guish each disease as well as the similarities. This TLC usually represents more chronic, severe
chapter will also examine the physiological proc- disease, and in the case of COPD it is related
esses that characterize airways disease. to remodeling of the chest wall leading to the
so-called barrel chest. The increase in TLC
may have another more important role; that is
the preservation of forced vital capacity (FVC)
LUNG VOLUMES AND ELASTIC RECOIL in face of a rise in RV that would then result
in more pronounced falls in FEV1 if the rise in
TLC had not ameliorated the fall in FVC [4,
Lung volumes and capacities 7]. The increase in TLC in parallel with each
increase in RV can occur until the chest wall
In diseases of the airway such as asthma and reaches some structural limit which probably
COPD there are characteristic increases in varies from person to person [7, 10] and acutely
55
Asthma and COPD: Basic Mechanisms and Clinical Management

(B) Structural limit


TLC
(A)

VC
IRV IC

Volume
VC
FRC
Volume

TV
FRC
RV
ERV

RV

Time RV Mild Moderate Severe


Disease severity

FIG. 5.1 (A) The figure presents a trace of volume displacement as a function of time during quiet breathing followed by a maximal inspiration to TLC, the
subject then exhales the VC until no more air exits the lung. The volume of gas remaining in the lung at this point is RV. FRC is the volume left in the lung at
the end of each normal breath. Expiratory reserve (ERV) and inspiratory reserve volume (IRV) are as noted. The inspiratory capacity (IC) is the volume of air
inspired from FRC to TLC and is sometimes used as a surrogate for TLC. The three boundary conditions TLC, RV, FRC provide the most useful information as to
the functioning of the lung (see text). (B) The figure present the changes in lung volumes and capacities that occur with increased airway disease severity as
during an asthma attack or during the progression of COPD. The first (and last) change is usually an increase in RV (mild) as airway closure increases the RV
continues to rise but FVC does not fall if the TLC rises in concert [4, 7]. However, at some point in the disease process (severe) or depending on the flexibility
of the chest wall a structural limit is reached where the TLC cannot increase further. At this point the further rise in RV results in a fall in VC, FVC, and FEV1
(see text).

TABLE 5.1 Boundaries of lung volume and physical determinants. balance of elastic forces of the respiratory system (see
below). Increases in FRC observed in airways disease may
Volume or represent a loss of recoil of the lung as occurs in emphysema
capacity TLC (%) Determinants or an increase in outward recoil of the chest wall. In the case
of the latter, the inspiratory respiratory muscle in eect reset
TLC: total lung Respiratory muscle function
the equilibrium point of the respiratory system to a higher
capacity Structural limits of chest wall distortion
volume such as occurs during an asthma attack [6, 20]. For
Compliance of chest wall and lung
patients with asthma or COPD, the FRC increases by sev-
FRC: function 50 Compliance of chest wall and lung eral mechanisms including persistent activity of the respira-
residual capacity Expiratory time constants tory muscles [20], increased expiratory time constants [20],
Respiratory muscle activity or alterations in recoil. Severe increases in FRC occur in
patients with more severe forms of COPD because all these
RV: residual 2030 Airway closure mechanisms occur in concert (Fig. 5.2).
volume Adequate expiratory effort
Children (18 years): chest wall
compliance
Elastic recoil
Note: TLC (%): total lung capacity for normal persons.
Pressurevolume (PV) relationships are used to quantita-
tively display the elastic recoil of the components of the res-
during an exacerbation of disease (Fig. 5.1B). The cause piratory system: the chest wall and the lungs. Elastic recoil
of the increase in TLC is unknown but the possibilities is very important in a number of respects. First, the strength
include either a loss of an inhibitory reex that activates of the chest wall muscle when coupled to the elastic recoil of
the muscles of the chest wall as TLC is reduced or sim- the chest wall and lungs serve to determine the static lung
ple structural overdistension [1012]. For patients with volumes, specically the maximal excursion of the chest wall
COPD it would appear that the increase in TLC may as assessed by TLC and RV [6]. Second, the elastic recoil
also be due to remodeling of the chest wall and is com- as inuenced by changes in lung volume, alters intrapulmo-
monly observed in X-ray or CT images. Assessing changes nary airway caliber [1315, 21, 22]. Thirdly, elastic recoil is
in FRC is of particular importance because resting lung the most important load to smooth muscle contraction as
volume has such an important bearing on airway caliber changes in lung volumes have a profound eect on ASM
and airway smooth muscle (ASM) contractility [1319]. performance [18, 22]. Lastly, elastic recoil is the major driv-
Determination of FRC provides a reasonable alternative ing force for eort-independent maximal airow, inuencing
to the placement of an esophageal balloon to measure the peak ow and in particular FEV1 [23, 24].

56
Pulmonary Physiology 5
125 B Chest volume increases or decreases the airway caliber increases
wall or decreases [1315]. Since, the contraction of ASM is
A
dependent upon muscle ber length and constitutes the
100 load against which ASM must contract, lung volume sets
Lung
the starting load. The observation explains why at low lung
(% Predicted TLC)

volumes the response to an inhaled bronchoconstricting


75 FRC A  B agent is enhanced [16, 18, 19]. Hyperination diminishes
Volume

luminal narrowing caused by ASM activation as shown by


FRC A or B
animal [16, 17] or human studies [18, 19].
50 FRC

25
DETERMINANTS OF AIRWAY CALIBER

0 10 20 30 The size of an intrapulmonary airway lumen at any given


Pressure (cmH2O) lung volume (e.g. FRC) is generally thought to depend on:
(1) contents of the lumen (such as mucous plugs or liquid
FIG. 5.2 Pressurevolume (PV) relationships in a normal subject (heavy accumulation), (2) the structure of the airway both inter-
solid lines), asthmatic patient (chestwall shifts to B) and in a patient with nal and external to the smooth muscle, (3) activation state
emphysema (where the lung PV shift to A and chest wall to B: A  B). or bronchomotor tone of the airways smooth muscle, (4)
The heavy solid line to the left is the PV curve of the chest wall showing compliance or stiness of the airway wall, and (5) tether-
outward recoil up until 66% of TLC and the line to the right is the PV curve
ing forces of the attached alveolar wall or lung volume.
of the lung. For the normal person the equilibrium point (FRC) where
the outward recoil is equal but opposite to the inward recoil occurs at
Transmural pressure (Ptm), which is the pressure inside
about 50% of the TLC. However, for the patient with loss of recoil of the minus pressure outside of the airway, represents the sum-
lung (dashed line A) or chest (dashed line B) FRC rises because the inward mation of forces working to open or close the airway and is
recoil of the lung is less or the chest is more and a new equilibrium (FRC resisted by the structure (or compliance) of the airway wall.
A or B) is reached. However, when both the chest wall (B) and lung PV (A) In both asthma and COPD there is a considerable change
relationship change (both dashed lines) the FRC is increased further (FRC referred to as remodeling of these structural components of
A  B). the airway wall (Fig. 5.3).

Elastic recoil is diminished in all patients with Contents of the lumen


emphysema whereas patients with asthma or the bronchitic
form of COPD exhibit, on average, more normal recoil The biological processes that lead to obstructive lung disease
[2527]. A caveat is that the loss of static elastic recoil in leave the airway lumen lled with liquid, cells, and mucus;
asthmatic patients has been reported by several work- this accumulation is even more marked during acute exac-
ers [2729]. In asthma the loss in elastic recoil can occur erbations [35]. Indeed, this pathologic feature may be the
acutely [30], and can be reversed with bronchodilator treat- most physiologically signicant as obstruction of the airway
ment [28, 31]. Unlike emphysema where the loss of elas- causes much of the mismatch of ventilation to perfusion
tic recoil is thought to be due to microscopic destruction of and in turn results in hypoxemia (see below). Moreover
parenchyma [32], these more acute losses in elastic recoil in removal of such an obstruction is anything but straight-
some patients with asthma may relate to alterations in sur- forward as the contents change from being serous to more
face tension due to extravasation of plasma proteins from mucoid in nature. Acutely lumenal obstructions appear in
the inammation or as a result of airway luminal brin for- the peripheral branches of the dependent portions of the
mation within the airway lumen [33]. trachealbronchial tree [36, 37]. These pathological changes
Determination of FRC coupled with an assessment in the distal lung are associated with 10-fold increase in
of the chest wall (e.g. visual inspection) allows one to assess peripheral resistance [38, 39] that increases further with
the likely position of the lung PV relationship without the disease severity [4042]. Yet even with this 10-fold increase
issues associated with placement of an esophageal balloon. in peripheral resistance in the mild asthmatic, spirometry
For example, nding a low FRC in a patient that was not can still be within normal limits.
overweight or obese would indicate a restrictive process;
whereas an increase in measured FRC in a patient with nor-
mal appearing chest wall is suggestive of either persistent Structure of the airway wall
activity of the inspiratory muscles (especially if the patient
has labored breathing) or a loss of inward elastic recoil of Inammation of the airway wall characterizes all forms of
the lung or chest wall [6, 8]. obstructive airway disease. Structural alterations include
Elastic recoil has a profound eect on airway caliber. increased extracellular deposits of the extracellular matrix
As lung volume increases there is outward pull on intrapul- that include bronectin, collagen, vitronectin, and a host
monary airways since these airways are embedded in the of other components [43]. The apparent thickening of the
alveolar matrix of the parenchyma [34]. Hence, as lung basement membrane below the airway epithelium is due

57
Asthma and COPD: Basic Mechanisms and Clinical Management

(A) Normal Airways disease

1 plugging
2 folds
BM
6 3 liquid (B) Airway disease Normal
5
ASM
4 4 BM
5 ASM

ium 6 breaks
el
ith
Ep 7 Pel
Pel
1
7 8 thickening
Pbr
2
Palv 3 8

thickness wall stiffness

ASM contractility uncoupling parallel load ASM

series load ASM series load ASM

shortening, velocity barrier function

clearance

FIG. 5.3 (A) A schematic representation of the cross-section of an airway in a normal person (left-hand side of A) and a patient with obstructive airways
disease (right-hand side of A). The changes that occur during airway remodeling in obstructive lung disease that could either cause airway narrowing and
airway hyperresponsiveness or prevent airway hyperresponsiveness are presented. In asthma and COPD several factors are operational but the relationship
of these structural changes to lung function is anything but clear. The mechanisms that cause or prevent airway narrowing are several and include: (1) Debris
plugging of airway lumen, (2) decreased number of folds, (3) liquid in luminal folds of epithelium, (4) thickness of the basement membrane, (5) increased
in contractility and/or size of ASM, (6) breaks in the alveolar tethers, (7) loss of elastic recoil, and (8) thickening of airway wall. Pel: elastic recoil forces; Palv:
alveolar pressure; Pbr: pressure in the lumen of the bronchiole; Transmural pressure: the difference in Pbr Palv; ASM: airway smooth muscle. (B) Balance of
forces favoring airway narrowing or airway opening.

to type I and III collagen deposition and is observed even Compliance of the airway wall
in mild disease [44]. ASM is also increased both in volume
(hypertrophy) and in number (hyperplasia) [43]. The func- The structural components of the airway wall represent the
tional consequence of remodeling of the airway is uncertain; load to smooth muscle shortening, thus an increase in com-
indeed, there is little proof that there is a functional conse- pliance would amplify ASM contraction and a decrease in
quence. Arguments have been made that airway remodeling compliance would be expected to antagonize ASM shorten-
can both promote and protect the size of airway lumen [45] ing. Cartilage, an important structural element of the airway
(Fig. 5.3B). wall, would serve to protect the airway lumen and has been
shown to soften with disease [43, 48], an eect thought to
be due to inammatory mediators and the release of metal-
Activation state of the ASM loprotineases. Folding or buckling of the mucosa has been
speculated to either prevent airway narrowing or enhance
Bronchomotor tone has two consequences: rst, depending airway luminal narrowing by changing wall compliance
on the compliance of the airway wall, smooth muscle tone [46, 49]. In this situation as the wall is compressed by ASM
narrows airways and reduces airow but, secondly, smooth contraction, the layer internal to the ASM folds or buck-
muscle tone also stiens the airway wall as it contracts. les up. The folds reinforce each other which serve to prevent
A decrease of airway wall compliance would serve to pre- further narrowing with an apparent increased load to the
vent further collapse [45, 46]. This is best appreciated when ASM. However, if the folds are few in number [49] and/or
ASM is activated at low lung volumes. Several studies have interstices of the folds become lled with liquid because of
shown that when methacholine is inhaled in the supine the small radius of curvature of an individual fold and the
posture, compared to the upright posture, more severe high surface tension found in the presence of inadequate
airway narrowing occurs [18, 19] and there is a marked surfactant [50], the airway lumen area would be narrowed
increase in the maximal level of airway resistance. However, or even obstructed.
a decrease in bronchomotor tone will increase airway wall Direct in vivo measurements of airway wall compli-
compliance and lead to dynamic airway narrowing during ance yield conicting results. Measurements from autopsy
maximal expiratory ow [31, 47]. specimens of patients with COPD showed reduced

58
Pulmonary Physiology 5
compliance only in very small (1 mm) airways [51]. typically seen in patients with emphysema [57, 58]. For
Normal lungs show age-related decrease in wall compliance patients with COPD or severe long-standing asthma this
which complicates the interpretation of the losses observed profound alteration in the function and structure of the
in patients with COPD who are normally older [52, 53]. In airway accounts for the sustained and therapeutically resist-
asthma both increases [53, 54] and decreases [21] in wall ant increase in airway resistance or decrease in FEV1. The
stiness have been reported. Decreased wall compliance or dilemma for the clinician is to ascertain what portion of
stiening is likely the result of inammation and/or wall the loss of structure and function is reversible and what
thickening and hence would be related to asthma severity portion is not.
and disease status. Another potential mechanism of airway
narrowing from the increase in wall compliance results from
a decrease in forces tethering the airway open. These forces
are decreased through two mechanisms. First, there are few
alveolar attachments to the outer mucosa of the airway AIRFLOW RESISTANCE
wall and second scar tissue has less tensile strength than
normal tissue [55].
Chronic obstructive pulmonary disease
Tethering forces or the coupling of lung A common feature of the patient with COPD is airow
volume and resistance limitation; this is most often assessed by the fall in FEV1.
The current Global Initiative for Chronic Obstructive Lung
The attachments of the alveolar wall to the external wall of Disease (GOLD) classication system for COPD uses
the intrapulmonary airway are the anatomical link between post-bronchodilator FEV1 as a major factor in classica-
the alveoli of the parenchyma and airway. These attachments tion of disease severity and in practice this variable makes
form the physical linkage system that result in airways wid- up a signicant part of the case denition for COPD [59].
ening and the fall in airways resistance as lung volumes Airow limitation in COPD arises in the small (2 mm
increase. This tethering force is the single most important diameter at FRC) airways which exhibit neutrophilic
factor stabilizing and inuencing airway lumen integrity inammation even in asymptomatic smokers [60, 61].
[1315]. This fact is evidenced by the profound eect lung When patients have mild, asymptomatic disease there are
volume has on airow. As noted above, for a given degree of subtle changes in airow limitation [60] with changes in
smooth muscle activation greater airway narrowing occurs distribution and homogeneity of ventilation. As the severity
at low versus high lung volumes. For example, the simple of COPD increases there is increased involvement of more
act of assuming the supine position, which is not associ- central airways but even when FEV1 has fallen to 5060%
ated with a change in smooth muscle activation, markedly of predicted, more than half of the increased resistance can
increases airway resistance [15, 18]. The eects of lung vol- be attributed to narrowed intrapulmonary (3 mm) airways
ume on airways resistance are very sensitive to even small [42]. Airway dimensions in COPD are reduced, even when
losses (1 cmH2O) in lung elastic recoil pressure [16, 17, 35]. the lungs are inated to a standard ination pressure (typi-
This process is not limited to changes within the parameter cally 30 cmH2O) so one must conclude that the structural
of the airway as denoted by ASM, since changes external remodeling of collagen, mucous gland hyperplasia, general
to the ASM can also uncouple the lung volumeairway wall thickening, and loss of tethering elastic forces contrib-
resistance relationship. utes to the airway narrowing [62].
Changing from the upright to the supine position These alterations in structure and function lead to
leads to airway hyperresponsiveness (AHR) in asthmatics profound ventilatory defects and a loss of collateral ven-
but such changes also occur in normals [18, 56]. The mech- tilation [63, 64]. In turn there is a trapping of gas due to
anism for this phenomenon is most likely the loss of the both long expiratory time constants and check valve-like
tethering forces, as estimated by Pel. These forces pull open processes within the obstructed and emphysematous spaces.
the airway wall as lung volume increases, thus a loss of out- Re-establishment of collateral pathways with intrabronchial,
ward recoil tethering the airway open is termed uncoupling. one-way valves, much like the spiracles of insects, leads to
Airway resistance and lung volume uncoupling occurs rap- deation of these overextended spaces [65] and is currently
idly with sleep onset in the patient with nocturnal asthma being explored as a therapeutic option to the COPD patient
and further uncoupling during the night probably as a result to relieve this trapped gas [66].
of inammatory events, resultant airway wall thickening, In patients with severe COPD expiratory airow lim-
and mechanical uncoupling [21]. In COPD the loss of elas- itation is observed at all lung volumes and often occurs even
tic recoil (Pel) is both greater and more persistent than the when there is a minimal expiratory eort; in these patients
age-dependent loss of recoil that occurs in normal persons, expiratory airow limitation occurs on expiration with each
and when considered in regards to loss of Pel with aging, breath [67]. In more mild cases, the airow limitation is
accounts for the progressive loss of FEV1 over the years. observed only on forced expiration because: (1) ow and
The loss of Pel, together with breaks in alveolar attach- volume during eupnea do not reach their structural limits
ments, results in the increased airway narrowing noted on due to the reserve in the respiratory system and (2) dynamic
forced expiration in patients with obstructive airway dis- hyperination, which increases elastic recoil by increasing
ease [47]. Smooth muscle contraction contributes to severe lung volume, further prevents reaching these limits during
airway narrowing and loss of bronchodilation response eupnea. The pathogenesis of airow limitation in patents

59
Asthma and COPD: Basic Mechanisms and Clinical Management

(A) (B) A
A
100
A

Volume (%TLC)
5

Flow (lps)
A B
3
C
2 D
1

0 10 20 30 0 5 10 15
Flow (lps) Pressure (cmH2O) Pressure (cmH2O)

FIG. 5.4 (A) The role of the loss of lung recoil and airway wall collapse in affecting a fall in maximal expiratory flow (FEV1) can be assessed by constructing
a MFSR curve [73, 74]. (B) The conductance (resistance) of airways downstream of the point (equal pressure point) of airflow limitation is determined as the
slope of V/Pel relationship. This V/Pel relationship or MFSR curve comes from selecting points from the both flowvolume and pressurevolume relationships
at points of isovolume (horizontal dashed lines in A). The dotted line, flowvolume loop is from volume displacement of body plethysmograph whereas
the solid line flowvolume loop is integrated volume from the mouth flow signal. The shaded area in (B) is from normal subjects [75]. MFSR A is the V/Pel
without the effect of gas (airway wall compression) whereas MFSR A includes the effect of compression; the resistance is increased. MFSR B is a patient with
increased resistance (e.g. airway narrowing) of the airways. MFSR C is a patient with emphysema where the sole abnormality is a loss of recoil, airway wall
collapse and lower flow rates. MFSR D is a patient with increased resistance, loss of recoil, and increase pressure (volume) at which the airways close due to
collapse and/or airway dynamic compression shifting the V/Pel relationship to the right.

with COPD or more severe forms of bronchitis and asthma variability and reproducibility of the FEV1 as an outcome
accordingly is quite complex and is a reection of the inter- measure is robust and for this reason the FEV1 is a prin-
play between airway caliber, respiratory muscle function, cipal outcome variable for clinical research. Unfortunately
lung volume, elastic recoil, and airway wall compliance a low FEV1 is not specic for any single cause of airow
[6, 14, 23, 24, 27, 62, 68]. limitation, and other assessments of the loss in airow limi-
The severity of COPD is dened as the post- tation must be made.
bronchodilator FEV1 [59], so it is useful to consider what a Useful insight into the causes of airow limitation can
fall in FEV1 signies. The forced expiratory volume in 1 s be gained by the construction of isovolume pressureow
the FEV1 is a measurement taken from the volumetime curves or maximum ow static recoil (MFSR) relationships
relationship, known as a spirogram, during a forced exhala- which are determined from owvolume and PV relation-
tion from TLC. It is important to remember that the FEV1 ships (Fig. 5.4). To assess artifacts from compression of the
is a volume (liters) not a measure of airow (liters per sec- gas in thorax, lung volume is measured from the volume
ond) but it reects the average airow over the rst second displacement of the chest wall and is obtained by having
of a forced exhalation. A decrease in the FEV1, as a percent- the patient sit in a variable-volume, body plethysmograph.
age of its predicted value, indicates that the patient is unable To measure static recoil an esophageal balloon attached to
to exhale a volume of air in 1 s similar in magnitude to that a pressure transducer is positioned in the lower esophagus
which could be exhaled by a person of the same sex, age, to estimate pleural pressure. The subject then inhales to
and height without lung disease. A considerable number of TLC and expires slowly to construct a deation PV curve
factors inuence the magnitude of the FEV1, but only one (Fig. 5.4A) and then the subject performs a maximal ow
of these factors is airway luminal size. The magnitude of the volume curve. By taking a series of data pairs of ow and
FEV1 is very much inuenced by the size of the inspira- pressure from points of isovolume on the PV and ow
tion prior to beginning the forced expiration (TLC), elas- volume relationship, the MFSR curve can be constructed
tic recoil, airway caliber, airway wall compliance, and size of (Fig. 5.4B).
the lung that is open. In the case of the latter as lung vol- The slope (V/P) of the MFSR curve is resist-
ume is lost by airway closure, lling or destruction, the VC, ance (or its inverse, conductance) of the segment of lung
FVC, and hence FEV1, decreases. It is important to realize upstream of the point and is known as the equal pressure
that FEV1 is a polyvalent index; where polyvalent indices point, that is the point along the airway which Ptm is zero.
are ones in which many factors contribute to the exact value Because there is still elastic recoil of the lung at RV, the
of the index [69]; indeed most measures of lung function MFSR relationship does not intersect with zero pressure.
are polyvalent. Accordingly it is unclear what a low FEV1 Patients with airow limitation will exhibit dierent MFSR
might signify, the cause of the decrease or even the best curves depending upon the cause of the airow limitation.
treatment needed to reverse the loss in FEV1. For patients where airow limitation is solely the result of
Empirically the FEV1 is a very useful measure of loss of static elastic recoil, the slope of the curve is normal;
overall lung function since it relates to morbidity and mor- that is upstream resistance is normal and the loss of airow
tality of COPD, even death due to all causes [70]. The is due solely to the loss of static elastic recoil. However, in
techniques used to measure FEV1 have been well codied a patient with airway narrowing due to inammation, col-
[71, 72]. The general performance and in particular the lagen deposition and/or collapse, the slope is decreased, that

60
Pulmonary Physiology 5
is resistance is elevated. By comparing owvolume curves TABLE 5.2 Criteria recommended for acute bronchodilator responses in
constructed from measuring both lung volumes by integrat- adults [79, 81].
ing the ow at the mouth to lung volume measured by the
chest wall, the role of airway wall compression on the loss of FEV1 (%) FVC (%) Comments
airow can be assessed [76]. The owvolume curve derived ACCP 1525 1525 % of baseline in 2 of 3 tests
from the ow and volume exiting the lung is displaced such
that peak ow occurs at a higher volume but, after this ini- Intermountain 15 15 % of baseline
tial peak, ow is lower at any given volume due to dynamic ATS
airway wall compression. In essence this analysis assesses
the delay in gas exiting the lung; the more dynamic com- ATS 1991 12 12 % of baseline and 200 ml
pression, the more temporal delay. ATS/ERS 2000 12 12 % of baseline and 200 ml
In most patients with COPD, a combination of fac-
tors contributes to airow limitation [7376]. Determining ACCP: American College of Chest Physicians; ATS: American Thoracic Society [79]; ERS:
MSFR relationships can suggest which component might European Respiratory Society [81].
be amenable to treatment; bronchodilation for airway nar-
rowing, pursed lip breathing for airway collapse and lung
volume reduction surgery for improving static elastic recoil. -agonist; however, this enhances the response if the start-
In the case of the latter several studies using this analysis ing baseline of FEV1 or FVC is low. The use of a deni-
have shown that the eect of lung reduction surgery is a tion that includes both an absolute change (e.g. 200 ml)
reduction of RV and the resultant increase in the VC [77] or change expressed as a percent of predicted addresses this
or a more complex response where both a change in volume dilemma.
and a change in resistance occurred [78]. There are several approaches used to deliver bron-
chodilators to patients for assessing reversibility but two to
four pus of albuterol with or without a spacer is the most
common. However, combinations of bronchodilators with
BRONCHIAL RESPONSIVENESS or without an anticholinergic agent or the use of nebulizer
treatments are sometimes used. In this case one adminis-
ters the bronchodilator until side eects are observed or the
Bronchial responsiveness includes either the acute response total dose delivered is used [71, 72, 7981]. A wait period is
to a one-time treatment with a bronchodilator usually a then imposed to allow for the drug to exhibit its physiologi-
2-receptor antagonist or the response to a constrictor cal eects, for example 1520 min in the case of albuterol.
bronchial challenge. Determination of bronchial responsive- Unfortunately techniques to determine bronchodilator
ness better denes and classies patients with airways dis- responsiveness are highly variable between studies and labo-
ease. Bronchial responsiveness assessments are often used as ratories making direct comparison of study results dicult.
major outcome variables in clinical investigations and treat- Bronchodilator responsiveness is commonly meas-
ment trials because of the precision of the measure, its asso- ured as a change in the FEV1 but can also be assessed by
ciation to underlying inammation, the relevance to asthma expiratory owvolume loops, specic airway conductance
triggers, and the changes in bronchial responsiveness as the as determined with a body plethysmograph, or with the
result of treatment. forced oscillation technique. Each technique measures dif-
ferent aspects of lung function and the result obtained may
not be exactly equivalent. Moreover the pattern of the bron-
Bronchodilator responses chodilator response can vary with some patients having a
more central airways response (SGaw), whereas others have
Bronchodilator responsiveness to inhaled bronchodilators a more peripheral lung response as measured with FVC or
is among the factors used to dierentiate between whether the FEV1 [4, 81, 82]. Such ndings are of great interest in
a given patient is to be categorized as COPD or asthma. determining the inferred site of the functional defect such
Professional Society guidelines and position statements as central airways versus peripheral airways and have obvi-
for interpretation of spirometric results [72, 79] discuss ous ramications to drug delivery.
the various consensus criteria used to derive the currently
used bronchodilator criterion that identies a signicant
response as one where a 12% or greater change in the post- Bronchoconstriction responses
bronchodilator FEV1 or FVC from pre-bronchodilator val-
ues and a 200 ml absolute change occurs (Table 5.2). AHR is either a bronchoconstricting response (fall in
Establishing a criterion that indicates a signicant FEV1) to a stimulus that normally does not cause a con-
bronchodilator response has been controversial because of striction or is a heightened bronchoconstriction to the
uncertainty as to just what constitutes a signicant response stimulus. Examples of the former are exercise, cold air, or
[79, 80]. Various authors have suggested criteria that range bradykinin where examples of the latter are histamine and
from 10% to 15% change from baseline, some suggest using methacholine. Airways hyperresponsiveness is often used
a change in percent of predicted or an exact volume change to dene the diagnosis of asthma, however, the presence of
(e.g. 400 ml). Bronchodilator responses are reported as a AHR is not specic since many lung diseases are associated
percent change from the volumes obtained prior to use of a with hyperresponsiveness including COPD [83].

61
Asthma and COPD: Basic Mechanisms and Clinical Management

Over the last 50 years the proposed mechanisms to 60


explain AHR included anything from enhanced choliner-

FEV1, fall (% baseline)


gic tone of the airways to the current focus on pathologic 50 Asthma : Severe
alterations (remodeling) of the airway wall. While airways
smooth muscle usually has a central role in most theories, 40
recent experimental evidence suggests that hypercontrac-
30
tility of the ASM is not always required [84]. Bronchial Asthma : Mild
thermoplasty in which radio frequency energy is applied 20
to airways through the bronchoscope to abate the ASM, Normal
improves asthma symptoms with only modest improve- 10
ments in AHR [85]. This interesting result suggests that the COPD
ASM may not be as essential to AHR as previously thought 0
or may serve other purposes [86]. 0.1 1.0 10 100
AHR is functionally dened as a shift of the dose Methacholine dose
response relationship to an inhaled bronchoconstrictor ago-
nist, for example, methacholine, such that lower amount FIG. 5.5 Dose response curves for the dose of inhaled methacholine
of the agent yields equivalent biological response, that is a (mg/ml) versus FEV1 expressed as a percent decrease from the starting
leftward shift of the doseresponse curve [83]. The inhaled baseline. Data points are individual values up until there is a plateau in
dose of bronchial constricting agent is plotted against the the response or 50% fall in FEV1 has occurred. The dashed line is the PC20
fall in lung function and from this relationship the PC20 that is interpolated. Both mild asthma and COPD patients exhibit leftward
(hyperresponsiveness) shifts of the curve without increase in the maximal
is then measured. The PC20 is the interpolated dose of
response plateau. The patients with severe asthma show marked shift to
inhaled agonist that causes FEV1 to fall exactly 20%. The the left without a plateau. Modified and redrawn from Ref. [91].
PC20, decreases as asthma severity increases [83, 87]. The
relationship between the amount of inhaled bronchocon-
strictor agent and FEV1 often has a plateau in which fur-
Lung volume also inuences AHR based on recent
ther doses of drug illicit smaller and smaller physiological
studies on the eects of deep inspiration (DI). Large TLC
responses. In the case of severe asthmatics often a plateau of
breaths are known to be eective in some but not all asth-
response cannot be demonstrated [8891]. This loss of max-
matics [98]. In some patients DI can even cause bronchoc-
imal response may signify the uncoupling, loss of protec-
onstriction and such a response is associated with the degree
tive mechanisms with resultant severe bronchoconstriction
of airway inammation [99]. A DI can also provide protec-
[90]. In clinical, and most research practice, doseresponse
tion to a subsequent bronchoconstricting response when
relationships are usually not carried out until a plateau is
the DI is performed prior to the inhalation of methacholine
revealed thus the PC20 only assesses the position (sensitiv-
[100]. Loss of this bronchoprotective eect of DI occurs in
ity) of the airways responsiveness relationship (Fig. 5.5).
asthmatic patients with mild disease whereas reversibility
Although there is considerable variability among
of an induced constriction by a DI is only lost as asthma
subjects, AHR is considered present when the PC20, using
becomes more severe [101, 102]. In patents with COPD
methacholine chloride as the bronchoconstrictor agonist, is
the eects of DI are lost and this loss has been shown to be
less than 8 mg/ml [83, 87, 90]. Asthmatics will often dem-
related to the loss of alveolar attachments [103].
onstrate a reduced PC20 even though the FEV1 is normal
at the onset of the test; moreover, the PC20 in most studies
does not correlate with the FEV1 [88, 91]. As methacholine
is well known to stimulate serous secretions from airway
glands and cells, it seems reasonable to suggest the AHR of GAS EXCHANGE
patients with mild to moderate asthma could be viewed as a
measure of increased tendency for airway closure to occur. The respiratory system provides oxygen (O2) and removes
In contrast among patients with COPD there is carbon dioxide (CO2) from the body and failure of this
a correlation of FEV1 to PC20 which suggests more of critical role of gas exchange leads to CO2 retention, hyper-
a structural mechanism for AHR consistent with the capnia, and/or hypoxemia. For ideal gas exchange to occur
observed airway wall thickening [44, 9193]. Moreover the ventilation must be matched with the vascular perfusion
patients with -1-antitrypsin deciency demonstrated an within the individual alveolar gas exchange units. The diu-
elevated response in FEV1 in concert with the loss of elastic sion capacity (DLCO) or transfer factor assesses the passive
recoil [94]. The mechanism of the fall in PC20 in COPD transfer of a test gas, carbon monoxide (CO), from alveolus
also seems unlikely to be the same as asthma since inhaled to the blood. The DLCO test is a frequently used measure-
corticosteroids do not lead to an improvement in AHR for ment in the pulmonary function laboratory and is quite use-
COPD patients [95] whereas inhaled steroids do improve ful in categorizing the dierent types of airway disease.
AHR in asthmatic patients [96]. AHR has important
prognostic value because as AHR increases (PC20 decreases)
there is accelerated decline in FEV1 in patients with COPD Ventilation/perfusion relationship
[97]. Taken together the data provide strong evidence that
the physiological mechanisms that lead to AHR in COPD The concept of ventilation to perfusion matching VA/Q
and asthma are probably quite dierent. is commonly used in understanding how airway disease

62
Pulmonary Physiology 5
pathology aects gas exchange. There are four mechanisms exchange units yet this is not the case [109112]. The
used to explain gas exchange abnormalities and include: (1) apparent explanation would include, preserved uploading
alveolar hypoventilation, (2) impaired O2/CO2 diusion, (3) of oxygen on the hemoglobin due to more than adequate
shunt, and (4) VA/Q mismatch, where the latter mechanism capillary transit time, hypoxic vasoconstriction, redirecting
is by far and away the most common cause of gas exchange blood away from aected areas, and adequate collateral ven-
abnormalities in patients with obstructive lung disease. tilation thus bypassing the closed airway.
During an asthma exacerbation or status asthmati-
cus the presence of a bimodal pattern emerges but even in
Chronic obstructive pulmonary disease this situation little pure shunt is observed. Treatment of the
asthma attack returns this pattern to normal [112] even at
The obstruction of airow to specic regions of the lung time of discharge from the emergency department [114].
parenchyma and widespread airway narrowing coupled with The relative preservation of arterial blood gases up until
the well-described pulmonary vascular abnormalities leads respiratory failure is attributed to the mechanisms covered
to marked VA/Q abnormalities and abnormal arterial blood above and the high ventilatory and cardiac output that
gases. As respiratory failure ensues, alveolar hypoventilation occurs during an attack. Bronchial challenge produces simi-
further contributes to the observed hypercapnia and hypox- lar patterns but it has been noted that the disturbances in
emia [104, 105]. Intrapulmonary shunt is not a major factor gas exchange lag behind the recovery of airow rates [115,
contributing to hypoxemia nor surprisingly is there a signif- 116]. This pattern of recovery does not occur with leuko-
icant contribution of reduced gas diusion probably because triene challenge, presumably because LTD4 causes a more
there is little ventilation of the areas that have emphysema. selective central airway constriction [117]. However, follow-
VA/Q mismatching appears to fully explain the hypoxemia ing exercise-induced bronchospasm airow normalization
of patients with COPD [105, 106]. In the case of very mild occurs after the return of VA/Q to normal, consistent with
disease in the peripheral airways as evidenced by FEV1 of dierent mechanisms of action in contrast to a naturally
80% predicted or greater, patients have an abnormal slope occurring asthma exacerbation [118]. Taken together these
of phase III on the single breath N2 test, a widened A-a O2 ndings suggest that our understanding of gas exchange in
gradient and mild VA/Q mismatch but with a normal PaO2 asthma, unlike COPD, is unclear.
[107]. During an exacerbation of COPD the VA/Q mis- Therapy has a marked eect on gas exchange.
match increases and accounts for about half of the observed Paradoxical hypoxemia has long been noted as a result of
hypoxemia whereas the remainder is low mixed venous O2 acute -agonist administration [119] and has been attrib-
due to increased O2 consumption of the body presumably uted to eects of -agonists on pulmonary blood ow. VA/Q
due to metabolic requirements of the disease [105, 107]. The measurement made after isoproterenol [110] showed tran-
A-a O2 gradient is correlated to emphysema score [108]; sient (5 min) alterations in blood ow to areas low in VA/Q;
hence, the abnormal gas exchange of COPD is explained by however, studies with a more selective -agonist, salbuterol
the pathology of the disease. were without this eect [112, 120].

Asthma Diffusion capacity (transfer factor)


Early studies with the multiple inert gas elimination tech- The diusion capacity, DLCO, is a measure of the disappear-
nique (MIGET) in stable, mild asthmatics showed a bimo- ance of CO, from the alveolus over a 10 s breath hold and is
dal distribution of pulmonary blood ow with about 25% of taken as an index of the total area of potential gas exchange
the blood ow going to units with low (0.1) VA/Q ratios. surface area. Because CO combines at the same site as oxy-
There was no evidence of areas of shunt or areas of high gen on the hemoglobin molecule, blood ow, hematocrit,
VA/Q or increased dead space [109]. However, later studies and cardiac output signicantly inuence the magnitude
showed less of this bimodal distribution and only some of of DLCO. The addition of helium to the test gas mixture
the patients showed this bimodal pattern [110, 111]; whereas inhaled allows for the calculation of the starting concentra-
most patients showed widening of the distribution of blood tion of CO and the determination of the communicating
ow. Patients in this latter study were receiving treatment gas volume by inert gas dilution. The standard single breath
with inhaled glucocorticosteroids which could explain the DLCO yields two useful measurements; the DLCO and a
dierences between studies. The gas exchange abnormali- measurement of TLC (VA) [121].
ties of patients with mild disease were thought to be due to
abnormalities in the periphery and consistent with abnormal
peripheral lung function such as in RV or frequency depend- Asthma & DLCO
ence of compliance. Asthma severity is associated with wid- The DLCO in patients with asthma is either within normal
ening of the VA/Q relationship [112] but most patients still limits or high depending on several factors. In asthmatic
exhibit a near normal PaO2 until they reach the point of patients with preserved lung function, the DLCO is typically
seeking medical assistance (Fig. 5.6) [113]. normal [122]. In moderate to severe asthma the DLCO is
Given that measurements of lung mechanisms show usually elevated and will also increase with bronchodila-
marked increase in peripheral resistance due to airway clo- tor treatment. The high DLCO values [123, 124] have been
sure, one might expect to observe considerable shunting explained by hyperination, increased intrathoracic pressure,
as blood ows by these closed airways and unventilated and a more likely cause, increases in pulmonary capillary

63
Asthma and COPD: Basic Mechanisms and Clinical Management

Normal Episodic asthma


1.5
0.9

1.0
0.6

0.5 0.2

Shunt
0.0 0.0
0.0 0.1 1.0 10 100 0.0 0.01 1.0 100

Chronic severe asthma Acute severe asthma

1.2
Ventilation and blood flow (l/min)

0.6 1.0

0.8
0.4
0.6

0.4
0.2

0.2

0.0 0.0
0.0 0.01 1.0 100 0.0 0.1 1.0 10 100

COPD type A COPD type B


2.0
1.2

0.8
1.0

0.4

0.0
0.0 0.01 0.1 1.0 10 100 0.0 0.01 0.1 1.0 10 100
Ventilation/blood flow ratio

FIG. 5.6 The VA/Q relationship as determined by the multiple inert gas elimination technique (MIGET). Note the different scaling for both axes in the various
examples. The normal distribution of ventilation (open circles) and blood flow (solid circles) is over a narrow range of VA/Q ratios and is unimodal. Patients
with mild or chronic asthma have wider distributions but still exhibit unimodal distributions whereas acute severe asthma is bimodal. In emphysema (COPD,
type A) there is a bimodal distribution in ventilation whereas chronic bronchitis (COPD, type B) exhibits bimodal blood flow. Shunt is not present in any of
the examples. Reproduced from Refs [105, 111] with permission.

blood volume or extravasation of red blood cells into particularly those who are more severe [125]. Increased vac-
the alveolus. Although not frequently commented on or ularization of the lung and airways might also increase the
because current practice is to lyse the lavage sample, the red apparent pulmonary capillary blood volume and account for
blood cell content is high in lavage samples of asthmatics, the elevated DLCO [43].

64
Pulmonary Physiology 5
Chronic obstructive pulmonary disease & DLCO TABLE 5.3 Physiological features of asthma contrasted with COPD.
In contrast to asthma patients with emphysema exhibit
Physiological
a reduction in DLCO [121, 126]. This reduction in DLCO
assessment COPD Asthma
has been shown to be closely related to evidence of patho-
logic emphysema [126] or emphysema as measured by CT Airflow limitation Present Sometimes present
[127, 128]. In asymptomatic smokers there is a detectable
10% decrease in DLCO that reverses with smoking cessa- Loss of elastic recoil Frequently present Sometimes present
tion likely due to reduced pulmonary capillary volume and
Bronchodilator  Usually present
CO back pressure [129, 130]. However, it should be noted
response
that the correlation of the reduction in DLCO to disease
severity in the individual patients is poor, perhaps reecting AHR  
the heterogeneity of the pathology of a given patient. The
DLCO is typically within normal limits for COPD patients Exercise-induced AHR  
with primary chronic bronchitis. One study showed a sig- VA/Q Widened Widened
nicant relationship between the DLCO and 3-year survival
[131], although another study did not nd this to be the DL CO * WNL or
case [132].
Notes: AHR: airways hyperresponsiveness; VA/Q: ventilation to perfusion ration; DL CO:
The measurement of alveolar volume (VA) provides
diffusion capacity of the lung.
additional insight into the physiological derangements * For emphysema, chronic bronchitis would be WNL.
present in the patient with airways obstruction. Because VA
is a single breath dilution of an inspired gas, it is aected
by both the presence of non-communicating lung volume,
TABLE 5.4 Pattern of changes in DL CO and VA in patients with obstructive
for example emphysematous bullae and the long-time con-
lung disease.
stants caused by airways narrowing. In normal subjects, and
when expressed as the volume of gas at body temperature, Clinical condition FEV1 VA DLCO
pressure and fully saturated, the measure of VA will agree
(200 ml) with TLC by body plethysmograph; however, Emphysema
patients with asthma or COPD will exhibit greater than
200 ml dierence between TLC and VA [133]. The degree Bronchitis WNL
of dierence between TLC and VA provides an approxi- Asthma WNL or
mate estimate of the total size of the non-communicating
bullae and alveoli [126, 134] or gas maldistribution. Notes: DL CO: diffusion capacity of carbon monoxide; FEV1: forced expiratory volume in
1 s; WNL: within normal limit.

CONCLUSION coupled with the measurement of spirometry and the


response to a bronchodilator [135137] (Table 5.4).
The clinical presentation of patients with obstructive lung
disease often presents a confusing clinical picture (Table
5.3). Consider, for example, the patient who reports asthma- References
like symptoms to common triggers but who also smokes and
has a greater than 20 pack per year smoking history. Such a 1. Bachert C, van Cauwenberge P, Khaltaev N. Allergic rhinitis and its
patient might be expected to exhibit physiological features impact on asthma. In collaboration with the world health organiza-
of both disorders. This is because the physiological mecha- tion. Executive Summary of the Workshop Report. December 710, 1999,
Geneva, Switzerland. Allergy 57: 841855, 2002.
nisms of asthma and COPD that dier in the details of
2. Soriano JB, Davis KJ, Coleman B, Visick G, Mannino D, Pride NB. The
the inammatory processes, cells, mediators, and pathology proportional Venn diagram of obstructive lung disease: Two approxi-
may be very similar physiologically in terms of hyperina- mations from the United States and the United Kingdom. Chest 124:
tion, loss of static recoil, and expiratory airow limitation. 47481, 2003.
Bronchodilator responsiveness is also not unique to asthma 3. Viegi G, Matteelli G, Angino A, Scognamiglio A, Baldacci S, Soriano JB,
since patients with COPD often demonstrate reversibility Carrozzi L. The proportional Venn diagram of obstructive lung disease
of airow limitation. Bronchial responsiveness is also present in the Italian general population. Chest 126: 1093101, 2004.
in both entities but the response to bronchoconstrictors is 4. Brown RH, Pearse DB, Pyrgos G, Liu MC, Togias A, Permutt S. The
more related to lung function and structure in COPD and structural basis of airways hyperresponsiveness in asthma. J Appl Physiol
the patient with COPD generally does not respond to exer- 101: 3039, 2006.
5. Kraft M, Cairns CB, Ellison MC, Pak J, Irvin C, Wenzel S.
cise by exhibiting bronchospasm. Gas exchange in general
Improvements in distal lung function correlate with asthma symptoms
is similar between both diseases with increased VA/Q mis- after treatment with oral montelukast. Chest 130: 172632, 2006.
match but with little evidence of shunt or global hypoven- 6. Irvin C, Cherniack R. Pathophysiology and physiologic assessment of
tilation. The DLCO represents an important distinguishing the asthmatic patient. Sem Resp Med 8(3): 20115, 1987.
feature between emphysema and other airways diseases and 7. Irvin CG. Lessons from structure-function studies in asthma: Myths
provides a means of discriminating the disease process when and truths about what we teach. J Appl Physiol 101: 79, 2006.

65
Asthma and COPD: Basic Mechanisms and Clinical Management

8. Irvin C. Guide to the evaluation of pulmonary function. In: Hamid Q, 31. De Troyer A, Yernault JC, Rodenstein D. Inuence of beta-2 agonist
Shannon J, Martin J, Hamilton J (eds). Physiologic Basis of Respiratory aerosols on pressurevolume characteristics of the lungs. Am Rev Respir
Disease. Ontario: BC Decker, 2005. Dis 118: 98795, 1978.
9. Wanger J, Clausen JL, Coates A, Pedersen OF, Brusasco V, 32. Eidelman DH, Ghezzo H, Kim WD, Hyatt RE, Cosio MG. Pressure
Burgos F, Casaburi R, Crapo R, Enright P, van der Grinten CP et al. volume curves in smokers. Comparison with alpha-1-antitrypsin de-
Standardisation of the measurement of lung volumes. Eur Respir J 26: ciency. Am Rev Respir Dis 139: 145258, 1989.
51122, 2005. 33. Wagers SS, Norton RJ, Rinaldi LM, Bates JH, Sobel BE, Irvin CG.
10. Whittaker LA, Irvin CG. Going to extremes of lung volume. J Appl Extravascular brin, plasminogen activator, plasminogen activator
Physiol 102: 83133, 2007. inhibitors, and airway hyperresponsiveness. J Clin Invest 114: 10411,
11. Loring SH, ODonnell CR, Butler JP, Lindholm P, Jacobson F, 2004.
Ferrigno M. Transpulmonary pressures and lung mechanics with glos- 34. Mead J, Takishima T, Leith D. Stress distribution in lungs: A model of
sopharyngeal insuation and exsuation beyond normal lung volumes pulmonary elasticity. J Appl Physiol 28: 596608, 1970.
in competitive breath-hold divers. J Appl Physiol 102: 1846, 2007. 35. Carroll NG, Mutavdzic S, James AL. Increased mast cells and neu-
12. Peress L, Sybrecht G, Macklem PT. The mechanism of increase in total trophils in submucosal mucous glands and mucus plugging in patients
lung capacity during acute asthma. Am J Med 61: 16569, 1976. with asthma. Thorax 57: 67782, 2002.
13. Butler J, Caro CG, Alcala R, Dubois AB. Physiological factors aect- 36. Lundblad LK, Thompson-Figueroa J, Allen GB, Rinaldi L,
ing airway resistance in normal subjects and in patients with obstruc- Norton RJ, Irvin CG, Bates JH. Airway hyperresponsiveness in allergi-
tive respiratory disease. J Clin Invest 39: 58491, 1960. cally inamed mice: The role of airway closure. Am J Respir Crit Care
14. Vincent NJ, Knudson R, Leith DE, Macklem PT, Mead J. Factors Med 175: 76874, 2007.
inuencing pulmonary resistance. J Appl Physiol 29: 23643, 1970. 37. Venegas JG, Winkler T, Musch G, Vidal Melo MF, Layeld D,
15. Briscoe WA, Dubois AB. The relationship between airway resistance, Tgavalekos N, Fischman AJ, Callahan RJ, Bellani G, Harris RS. Self-
airway conductance and lung volume in subjects of dierent age and organized patchiness in asthma as a prelude to catastrophic shifts.
body size. J Clin Invest 37: 127985, 1958. Nature 434: 77782, 2005.
16. Bates JH, Schuessler TF, Dolman C, Eidelman DH. Temporal dynam- 38. Wagner EM, Liu MC, Weinmann GG, Permutt S, Bleecker ER.
ics of acute isovolume bronchoconstriction in the rat. J Appl Physiol 82: Peripheral lung resistance in normal and asthmatic subjects. Am Rev
5562, 1997. Respir Dis 141: 58488, 1990.
17. Nagase T, Martin JG, Ludwig MS. Comparative study of mechanical 39. Kaminsky DA, Irvin CG, Gurka DA, Feldsien DC, Wagner EM,
interdependence: Eect of lung volume on raw during induced con- Liu MC, Wenzel SE. Peripheral airways responsiveness to cool, dry air
striction. J Appl Physiol 75: 25005, 1993. in normal and asthmatic individuals. Am J Respir Crit Care Med 152:
18. Ding DJ, Martin JG, Macklem PT. Eects of lung volume on maximal 178490, 1995.
methacholine-induced bronchoconstriction in normal humans. J Appl 40. Kraft M, Pak J, Martin RJ, Kaminsky D, Irvin CG. Distal lung dys-
Physiol 62: 132430, 1987. function at night in nocturnal asthma. Am J Respir Crit Care Med 163:
19. Meinero M, Coletta G, Dutto L, Milanese M, Nova G, 155156, 2001.
Sciolla A, Pellegrino R, Brusasco V. Mechanical response to metha- 41. Ohrui T, Yanai M, Sekizawa K, Morikawa M, Sasaki H, Takishima T.
choline and deep inspiration in supine men. J Appl Physiol 102: 26975, Eective site of bronchodilation by beta-adrenergic and anticholinergic
2007. agents in patients with chronic obstructive pulmonary disease: Direct
20. Martin J, Powell E, Shore S, Emrich J, Engel LA. The role of respira- measurement of intrabronchial pressure with a new catheter. Am Rev
tory muscles in the hyperination of bronchial asthma. Am Rev Respir Respir Dis 146: 8891, 1992.
Dis 121: 44147, 1980. 42. Yanai M, Sekizawa K, Ohrui T, Sasaki H, Takishima T. Site of airway
21. Irvin CG, Pak J, Martin RJ. Airway-parenchyma uncoupling in noc- obstruction in pulmonary disease: Direct measurement of intrabron-
turnal asthma. Am J Respir Crit Care Med 161: 5056, 2000. chial pressure. J Appl Physiol 72: 101623, 1992.
22. Irvin CG. Lung volume: A principle determinant of airway smooth 43. James AL, Wenzel S. Signicance of airway remodeling in airway dis-
muscle function. Eur Respir J 22: 35, 2003. eases. Eur J Respir Dis 30: 142041, 2007.
23. Mead J, Turner JM, Macklem PT, Little JB. Signicance of the rela- 44. Roche WR, Beasley R, Williams JH, Holgate ST. Subepithelial brosis
tionship between lung recoil and maximum expiratory ow. J Appl in the bronchi of asthmatics. Lancet 1: 52024, 1989.
Physiol 22: 95108, 1967. 45. McParland BE, Macklem PT, Pare PD. Airway wall remodeling:
24. Black LF, Hyatt RE, Stubbs SE. Mechanism of expiratory airow Friend or foe? J Appl Physiol 95: 42634, 2003.
limitation in chronic obstructive pulmonary disease associated with 46. Wiggs BR, Hrousis CA, Drazen JM, Kamm RD. On the mechanism
1-antitrypsin deciency. Am Rev Respir Dis 105: 89199, 1972. of mucosal folding in normal and asthmatic airways. J Appl Physiol 83:
25. Gibson GJ, Pride NB, Davis J, Schroter RC. Exponential descrip- 181421, 1997.
tion of the static pressurevolume curve of normal and diseased lungs. 47. Krowka MJ, Enright PL, Rodarte JR, Hyatt RE. Eect of eort on
Am Rev Respir Dis 120: 799811, 1979. measurement of forced expiratory volume in one second. Am Rev
26. Finucane KE, Colebatch HJ. Elastic behavior of the lung in patients Respir Dis 136: 82933, 1987.
with airway obstruction. J Appl Physiol 26: 33038, 1969. 48. Tandon MK, Campbell AH. Bronchial cartilage in chronic bronchitis.
27. Schlueter DP, Immekus J, Stead WW. Relationship between maximal Thorax 24: 60712, 1969.
inspiratory pressure and total lung capacity (coecient of retraction) in 49. Lambert RK, Codd SL, Alley MR, Pack RJ. Physical determinants of
normal subjects and in patients with emphysema, asthma, and diuse bronchial mucosal folding. J Appl Physiol 77: 120616, 1994.
pulmonary inltration. Am Rev Respir Dis 96: 65665, 1967. 50. Yager D, Butler JP, Bastacky J, Israel E, Smith G, Drazen JM.
28. Gold WM, Kaufman HS, Wadd JA. Elastic recoil of the lungs in Amplication of airway constriction due to liquid lling of airway
chronic asthmatic patients before and after therapy. J Appl Physiol 23: interstices. J Appl Physiol 66: 287384, 1989.
43338, 1967. 51. Wilson AG, Massarella GR, Pride NB. Elastic properties of airways in
29. Woolcock AJ, Read J. The static elastic properties of the lungs in human lungs post mortem. Am Rev Respir Dis 110: 71629, 1974.
asthma. Am Rev Respir Dis 98: 78894, 1968. 52. Maisel JC, Silvers GW, Mitchell RS, Petty TL. Bronchial atrophy and
30. Mansell A, Dubrawsky C, Levison H, Bryan AC, Langer H, dynamic expiratory collapse. Am Rev Respir Dis 98: 98897, 1968.
Collins-Williams C, Orange RP. Lung mechanics in antigen-induced 53. Brackel HJ, Pedersen OF, Mulder PG, Overbeek SE, Kerrebijn KF,
asthma. J Appl Physiol 37: 297301, 1974. Bogaard JM. Central airways behave more stiy during forced expiration

66
Pulmonary Physiology 5
in patients with asthma. Am J Respir Crit Care Med 162: 896904, 74. Pride NB, Permutt S, Riley RL, Bromberger-Barnea B. Determinants
2000. of maximal expiratory ow from the lungs. J Appl Physiol 23: 64662,
54. Wilson JW, Li X, Pain MC. The lack of distensibility of asthmatic air- 1967.
ways. Am Rev Respir Dis 148: 8069, 1993. 75. Leaver DG, Tattereld AE, Pride NB. Contributions of loss of lung
55. Saetta M, Ghezzo H, Kim WD, King M, Angus GE, Wang NS, Cosio recoil and of enhanced airways collapsibility to the airow obstruc-
MG. Loss of alveolar attachments in smokers. A morphometric cor- tion of chronic bronchitis and emphysema. J Clin Invest 52: 211728,
relate of lung function impairment. Am Rev Respir Dis 132: 894900, 1973.
1985. 76. Ingram RH Jr., Schilder DP. Eect of gas compression on pulmonary
56. Skloot G, Permutt S, Togias A. Airway hyperresponsiveness in asthma: pressure, ow, and volume relationship. J Appl Physiol 21: 182126,
A problem of limited smooth muscle relaxation with inspiration. J Clin 1966.
Invest 96: 2393403, 1995. 77. Ingenito EP, Evans RB, Loring SH, Kaczka DW, Rodenhouse JD,
57. Opazo Saez AM, Seow CY, Pare PD. Peripheral airway smooth mus- Body SC, Sugarbaker DJ, Mentzer SJ, DeCamp MM, Reilly JJ Jr..
cle mechanics in obstructive airways disease. Am J Respir Crit Care Med Relation between preoperative inspiratory lung resistance and the out-
161: 91017, 2000. come of lung-volume-reduction surgery for emphysema. N Engl J Med
58. Pare PD, Wiggs BR, James A, Hogg JC, Bosken C. The compara- 338: 118185, 1998.
tive mechanics and morphology of airways in asthma and in chronic 78. Gelb AF, Brenner M, McKenna RJ Jr., Fischel R, Zamel N, Schein MJ.
obstructive pulmonary disease. Am Rev Respir Dis 143: 118993, Serial lung function and elastic recoil 2 years after lung volume reduc-
1991. tion surgery for emphysema. Chest 113: 1497506, 1998.
59. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS. Global 79. ATS. Lung function testing: Selection of reference values and interpre-
strategy for the diagnosis, management, and prevention of chronic tive strategies. Am Rev Respir Dis 144: 120218, 1991.
obstructive pulmonary disease. NHLBI/WHO global initiative for 80. Pellegrino R, Viegi G, Brusasco V, Crapo RO, Burgos F, Casaburi
chronic obstructive lung disease (GOLD) workshop summary. Am J R, Coates A, van der Grinten CP, Gustafsson P, Hankinson J
Respir Crit Care Med 163: 125676, 2001. et al. Interpretative strategies for lung function tests. Eur Respir J 26:
60. Cosio M, Ghezzo H, Hogg JC, Corbin R, Loveland M, Dosman J, 94868, 2005.
Macklem PT. The relations between structural changes in small airways 81. Smith HR, Irvin CG, Cherniack RM. The utility of spirometry in the
and pulmonary-function tests. N Engl J Med 298: 127781, 1978. diagnosis of reversible airways obstruction. Chest 101: 157781, 1992.
61. Niewoehner DE, Kleinerman J, Rice DB. Pathologic changes in the 82. ODonnell DE, Lam M, Webb KA. Spirometric correlates of improve-
peripheral airways of young cigarette smokers. N Engl J Med 291: ment in exercise performance after anticholinergic therapy in chronic
75558, 1974. obstructive pulmonary disease. Am J Respir Crit Care Med 160: 54249,
62. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, 1999.
Cherniack RM, Rogers RM, Sciurba FC, Coxson HO et al. The nature 83. Crapo RO, Casaburi R, Coates AL, Enright PL, Hankinson JL,
of small-airway obstruction in chronic obstructive pulmonary disease. Irvin CG, MacIntyre NR, McKay RT, Wanger JS, Anderson SD et al.
N Engl J Med 350: 264553, 2004. Guidelines for methacholine and exercise challenge testing-1999. This
63. Hogg JC, Macklem PT, Thurlbeck WM. The resistance of collateral ocial statement of the American thoracic society was adopted by
channels in excised human lungs. J Clin Invest 48: 42131, 1969. the ATS board of directors, July 1999. Am J Respir Crit Care Med 161:
64. Terry PB, Traystman RJ, Newball HH, Batra G, Menkes HA. 30929, 2000.
Collateral ventilation in man. N Engl J Med 298: 1015, 1978. 84. Wagers S, Lundblad LK, Ekman M, Irvin CG, Bates JH. The allergic
65. Choong CK, Macklem PT, Pierce JA, Lefrak SS, Woods JC, mouse model of asthma: Normal smooth muscle in an abnormal lung?
Conradi MS, Yablonskiy DA, Hogg JC, Chino K, Cooper JD. Transpleural J Appl Physiol 96: 201927, 2003.
ventilation of explanted human lungs. Thorax 62: 62330, 2007. 85. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC,
66. Wood DE, McKenna RJ Jr., Yusen RD, Sterman DH, Ost DE, Olivenstein R, Pavord ID, McCormack D, Chaudhuri R et al. Asthma
Springmeyer SC, Gonzalez HX, Mulligan MS, Gildea T, Houck WV control during the year after bronchial thermoplasty. N Engl J Med 356:
et al. A multicenter trial of an intrabronchial valve for treatment of 132737, 2007.
severe emphysema. J Thorac Cardiovasc Surg 133: 6573, 2007. 86. Solway J, Irvin CG. Airway smooth muscle as a target for asthma ther-
67. Eltayara L, Becklake MR, Volta CA, Milic-Emili J. Relationship apy. N Engl J Med 356: 136769, 2007.
between chronic dyspnea and expiratory ow limitation in patients 87. Hargreave FE, Ryan G, Thomson NC, OByrne PM, Latimer K,
with chronic obstructive pulmonary disease. Am J Respir Crit Care Med Juniper EF, Dolovich J. Bronchial responsiveness to histamine or meth-
154: 172634, 1996. acholine in asthma: Measurement and clinical signicance. J Allergy
68. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of airway Clin Immunol 68: 34755, 1981.
obstruction in chronic obstructive lung disease. N Engl J Med 278: 88. Ryan G, Latimer KM, Dolovich J, Hargreave FE. Bronchial respon-
135560, 1968. siveness to histamine: Relationship to diurnal variation of peak ow
69. Mead J. Problems in interpreting common tests of pulmonary mechan- rate, improvement after bronchodilator, and airway calibre. Thorax 37:
ical function. In: Macklem P, Permutt S (eds). The Lung in Transition 42329, 1982.
Between Death and Disease. New York: Marcel Dekker Inc, 1979. 89. Gibbons WJ, Sharma A, Lougheed D, Macklem PT. Detection of
70. Ferguson GT, Enright PL, Buist AS, Higgins MW. Oce spirometry excessive bronchoconstriction in asthma. Am J Respir Crit Care Med
for lung health assessment in adults: A consensus statement from the 153: 58289, 1996.
national lung health education program. Chest 117: 114661, 2000. 90. Sterk PJ, Bel EH. The shape of the doseresponse curve to inhaled
71. Enright PL, Johnson LR, Connett JE, Voelker H, Buist AS. Spirometry bronchoconstrictor agents in asthma and in chronic obstructive pulmo-
in the lung health study. I: Methods and quality control. Am Rev Respir nary disease. Am Rev Respir Dis 143: 143337, 1991.
Dis 143: 121523, 1991. 91. Woolcock AJ, Anderson SD, Peat JK, Du Toit JI, Zhang YG,
72. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R, Coates Smith CM, Salome CM. Characteristics of bronchial hyperresponsive-
A, Crapo R, Enright P, van der Grinten CP, Gustafsson P et al. ness in chronic obstructive pulmonary disease and in asthma. Am Rev
Standardisation of spirometry. Eur Respir J 26: 31938, 2005. Respir Dis 143: 143843, 1991.
73. Fry DL, Hyatt RE. Pulmonary mechanisms: A unied analysis of the 92. Taylor RG, Joyce H, Gross E, Holland F, Pride NB. Bronchial reactiv-
relationship between pressure, volume and gas ow in the lungs of nor- ity to inhaled histamine and annual rate of decline in FEV1 in male
mal and diseased subjects. Am J Physiol 26: 67289, 1960. smokers and ex-smokers. Thorax 40: 916, 1985.

67
Asthma and COPD: Basic Mechanisms and Clinical Management

93. Tashkin DP, Altose MD, Bleecker ER, Connett JE, Kanner RE, 100% oxygen and salbutamol. Am Rev Respir Dis 141: 55862,
Lee WW, Wise R. The lung health study: Airway responsiveness to 1990.
inhaled methacholine in smokers with mild to moderate airow limi- 113. Mountain RD, Sahn SA. Clinical features and outcome in patients
tation. The Lung Health Study Research Group. Am Rev Respir Dis with acute asthma presenting with hypercapnia. Am Rev Respir Dis
145: 30110, 1992. 138: 53539, 1988.
94. Cheung D, Schot R, Zwinderman AH, Zagers H, Dijkman JH, 114. Roca J, Ramis L, Rodriguez-Roisin R, Ballester E, Montserrat JM,
Sterk PJ. Relationship between loss in parenchymal elastic recoil Wagner PD. Serial relationships between ventilationperfusion ine-
pressure and maximal airway narrowing in subjects with alpha1- quality and spirometry in acute severe asthma requiring hospitaliza-
antitrypsin deciency. Am J Respir Crit Care Med 155: 13540, 1997. tion. Am Rev Respir Dis 137: 105561, 1988.
95. Watson A, Lim TK, Joyce H, Pride NB. Failure of inhaled corticos- 115. Rodriguez-Roisin R, Ferrer A, Navajas D, Agusti AG, Wagner PD,
teroids to modify bronchoconstrictor or bronchodilator responsive- Roca J. Ventilationperfusion mismatch after methacholine chal-
ness in middle-aged smokers with mild airow obstruction. Chest lenge in patients with mild bronchial asthma. Am Rev Respir Dis 144:
101: 35055, 1992. 8894, 1991.
96. Sont JK, Willems LN, Bel EH, van Krieken JH, Vandenbroucke JP, 116. Echazarreta AL, Gomez FP, Ribas J, Achaval M, Barbera JA, Roca
Sterk PJ. Clinical control and histopathologic outcome of asthma J, Chung KF, Rodriguez-Roisin R. Eects of inhaled furosemide on
when using airway hyperresponsiveness as an additional guide to platelet-activating factor challenge in mild asthma. Eur Respir J 14:
long-term treatment. The AMPUL study group. Am J Respir Crit Care 61621, 1999.
Med 159: 104351, 1999. 117. Echazarreta AL, Dahlen B, Garcia G, Agusti C, Barbera JA, Roca J,
97. Tashkin DP, Altose MD, Connett JE, Kanner RE, Lee WW, Dahlen SE, Rodriguez-Roisin R. Pulmonary gas exchange and spu-
Wise RA. Methacholine reactivity predicts changes in lung function tum cellular responses to inhaled leukotriene D-4 in asthma. Am J
over time in smokers with early chronic obstructive pulmonary dis- Respir Crit Care Med 164: 2026, 2001.
ease. The Lung Health Study Research Group. Am J Respir Crit Care 118. Young IH, Corte P, Schoeel RE. Pattern and time course of
Med 153: 180211, 1996. ventilationperfusion inequality in exercise-induced asthma. Am Rev
98. Orehek J, Charpin D, Velardocchio JM, Grimaud C. Bronchomotor Respir Dis 125: 30411, 1982.
eect of bronchoconstriction-induced deep inspirations in asthmatics. 119. Knudson RJ, Constantine HP. An eect of isoproterenol on ventila-
Am Rev Respir Dis 121: 297305, 1980. tionperfusion in asthmatic versus normal subjects. J Appl Physiol 22:
99. Lim TK, Pride NB, Ingram RH Jr.. Eects of volume history dur- 4026, 1967.
ing spontaneous and acutely induced air-ow obstruction in asthma. 120. Ballester E, Reyes A, Roca J, Guitart R, Wagner PD, Rodriguez-
Am Rev Respir Dis 135: 59196, 1987. Roisin R. Ventilationperfusion mismatching in acute severe asthma:
100. Scichilone N, Kapsali T, Permutt S, Togias A. Deep inspiration- Eects of salbutamol and 100% oxygen. Thorax 44: 25867, 1989.
induced bronchoprotection is stronger than bronchodilation. Am J 121. Cotton DJ, Graham BL. Single-breath carbon monoxide diusion
Respir Crit Care Med 162: 91016, 2000. capacity or transfer factor. In: Hamid Q, Shannon J, Martin J (eds).
101. Assefa D, Amin N, Dozor AJ. Eect of deep inspiration on airway Physiologic Basis of Respiratory Disease. Hamilton: BC Decker, 2005.
caliber in children with asthma. Pediatr Pulmonol 38: 40612, 2004. 122. Saydain G, Beck KC, Decker PA, Cowl CT, Scanlon PD. Clinical
102. Scichilone N, Marchese R, Soresi S, Interrante A, Togias A, signicance of elevated diusing capacity. Chest 125: 44652, 2004.
Bellia V. Deep inspiration-induced changes in lung volume decrease 123. Keens TG, Mansell A, Krastins IR, Levison H, Bryan AC,
with severity of asthma. Respir Med 101: 95156, 2007. Hyland RH, Zamel N. Evaluation of the single-breath diusing
103. Scichilone N, Bruno A, Marchese R, Vignola AM, Togias A, Bellia V. capacity in asthma and cystic brosis. Chest 76: 4144, 1979.
Association between reduced bronchodilatory eect of deep inspira- 124. Collard P, Njinou B, Nejadnik B, Keyeux A, Frans A. Single breath
tion and loss of alveolar attachments. Respir Res 6: 55, 2005. diusing capacity for carbon monoxide in stable asthma. Chest 105:
104. Begin P, Grassino A. Inspiratory muscle dysfunction and chronic 142629, 1994.
hypercapnia in chronic obstructive pulmonary disease. Am Rev Respir 125. Diaz P, Galleguillos FR, Gonzalez MC, Pantin CF, Kay AB.
Dis 143: 90512, 1991. Bronchoalveolar lavage in asthma: The eect of disodium cromogly-
105. Agusti AG, Barbera JA. Contribution of multiple inert gas elimi- cate (cromolyn) on leukocyte counts, immunoglobulins, and comple-
nation technique to pulmonary medicine. 2. Chronic pulmonary ment. J Allergy Clin Immunol 74: 4148, 1984.
diseases: Chronic obstructive pulmonary disease and idiopathic 126. McLean A, Warren PM, Gillooly M, MacNee W, Lamb D.
pulmonary brosis. Thorax 49: 92432, 1994. Microscopic and macroscopic measurements of emphysema: Relation
106. Hlastala MP, Robertson HT. Inert gas elimination characteristics of to carbon monoxide gas transfer. Thorax 47: 14449, 1992.
the normal and abnormal lung. J Appl Physiol 44: 25866, 1978. 127. Gould GA, MacNee W, McLean A, Warren PM, Redpath A,
107. Barbera JA, Roca J, Ferrer A, Felez MA, Diaz O, Roger N, Best JJ, Lamb D, Flenley DC. CT measurements of lung density
Rodriguez-Roisin R. Mechanisms of worsening gas exchange dur- in life can quantitate distal airspace enlargement An essential den-
ing acute exacerbations of chronic obstructive pulmonary disease. ing feature of human emphysema. Am Rev Respir Dis 137: 38092,
Eur Respir J 10: 128591, 1997. 1988.
108. Barbera JA, Roca J, Ramirez J, Wagner PD, Ussetti P, Rodriguez- 128. Gevenois PA, De Vuyst P, de Maertelaer V, Zanen J, Jacobovitz D,
Roisin R. Gas exchange during exercise in mild chronic obstructive Cosio MG, Yernault JC. Comparison of computed density and micro-
pulmonary disease. Correlation with lung structure. Am Rev Respir scopic morphometry in pulmonary emphysema. Am J Respir Crit Care
Dis 144: 52025, 1991. Med 154: 18792, 1996.
109. Wagner PD, Dantzker DR, Iacovoni VE, Tomlin WC, West JB. 129. Sansores RH, Pare P, Abboud RT. Eect of smoking cessation on
Ventilationperfusion inequality in asymptomatic asthma. Am Rev pulmonary carbon monoxide diusing capacity and capillary blood
Respir Dis 118: 51124, 1978. volume. Am Rev Respir Dis 146: 95964, 1992.
110. Wagner PD, Hedenstierna G, Bylin G. Ventilationperfusion ine- 130. Watson A, Joyce H, Hopper L, Pride NB. Inuence of smoking habits
quality in chronic asthma. Am Rev Respir Dis 136: 60512, 1987. on change in carbon monoxide transfer factor over 10 years in middle
111. Rodriguez-Roisin R, Roca J. Contributions of multiple inert gas aged men. Thorax 48: 11924, 1993.
elimination technique to pulmonary medicine. III: Bronchial asthma. 131. Dubois P, Machiels J, Smeets F, Delwiche JP, Lulling J. CO trans-
Thorax 49: 102733, 1994. fer capacity as a determining factor of survival for severe hypoxae-
112. Ballester E, Roca J, Ramis L, Wagner PD, Rodriguez-Roisin R. mic COPD patients under long-term oxygen therapy. Eur Respir J 3:
Pulmonary gas exchange in severe chronic asthma. Response to 104247, 1990.

68
Pulmonary Physiology 5
132. Anthonisen NR, Wright EC, Hodgkin JE. Prognosis in chronic 136. Klein JS, Gamsu G, Webb WR, Golden JA, Muller NL. High-
obstructive pulmonary disease. Am Rev Respir Dis 133: 1420, 1986. resolution CT diagnosis of emphysema in symptomatic patients
133. Graham BL, Mink JT, Cotton DJ. Eect of breath-hold time on with normal chest radiographs and isolated low diusing capacity.
DLCO(sb) in patients with airway obstruction. J Appl Physiol 58: Radiology 182: 81721, 1992.
131925, 1985. 137. Sciurba FC. Physiologic similarities and dierences between COPD
134. Wade JF 3rd, Mortenson R, Irvin CG. Physiologic evaluation of bul- and asthma. Chest 126: 117S124S, 2004, discussion 159S161S.
lous emphysema. Chest 100: 115154, 1991.
135. Gelb AF, Gold WM, Wright RR, Bruch HR, Nadel JA. Physiologic
diagnosis of subclinical emphysema. Am Rev Respir Dis 107:
5063, 1973.

69
Airway Pathology
6
CHAPTER

NORMAL ANATOMY normal lung because of their much smaller total


James C. Hogg
cross-sectional area [3, 4].
The conducting airways are lined by epi- UBC McDonald Research Laboratories,
The normal human bronchogram (Fig. 6.1) shows thelium and surrounded by an adventitial layer St. Pauls Hospital, University of British
that the length of pathways from the trachea to [5]. The submucosa between the epithelium and Columbia, Vancouver, BC, Canada
the terminal airways diers depending on the outer edge of the muscle layer is often referred
pathway followed and it can take as few as 8 or to as the lamina propria, but this term is tech-
as many as 24 divisions of airway branching to nically incorrect because many airways are not
reach the gas-exchanging surface [1]. The small completely surrounded by muscle [6]. Bronchi are
bronchi and bronchioles 2 mm in diameter are dened by the presence of a layer of brocartilage
spread out from the fourth to the fourteenth external to the smooth muscle and tubuloalveo-
generation of airway branching [2]. The total air- lar glands, which communicate with the airway
way cross-sectional area expands rapidly beyond lumen via ducts [7]. Bronchioles lack both carti-
the 2 mm airways to provide for rapid diusion lage and glands and become respiratory bronchi-
of gas between the distal conducting airways and oles when alveoli open directly into their lumen
the gas-exchanging surface. The central conduct- [7]. The lining of the trachea and major bronchi
ing airways larger than 2 mm internal diameter consists of pseudostratied, ciliated columnar epi-
are the major site of resistance to airow in the thelium which gradually becomes more cuboidal

FIG. 6.1 Postmortem bronchogram from


an otherwise normal 19-year-old man who
died suddenly for reasons unrelated to the
lung. Note the difference in airway length
depending upon the pathway that is followed.

71
Asthma and COPD: Basic Mechanisms and Clinical Management

FIG. 6.2 Photomicrograph of a bronchiole from a patient who died of asthma. This shows the location of submucosal capillaries (white arrow), connecting
vessels passing between muscle bundles (dark arrow) to larger postcapillary venules located outside the muscle layer. On the right, the same features are
shown at a higher magnification.

with fewer ciliated cells as the alveolar surface is approached the third intercostal artery, from the right internal mammary
[68]. Light microscopy reveals that the basal aspect of air- artery, or from the right subclavian artery [8, 15]. Millers
way epithelial cells is attached to a thin basement membrane classic anatomical account [8] showed that two to three
(8090 nm width) that contains primarily type IV collagen arterial branches accompany each of the larger bronchi and
and elastin [9]. Transmission electron microscopy shows that that anastomoses between these branches form an arterial
a true basement membrane or basal lamina can be readily dis- plexus in the outer wall of the airway. Small branches of this
tinguished from the connective tissue observed with the light plexus penetrate the smooth muscle layer to form a capillary
microscope [6, 7]. Quantitative studies [1012] have shown network below the epithelium. Short connecting branches
that the trachea and mainstem bronchi of normal subjects extend from this plexus through the muscle layer to form a
consist of by volume: second plexus of venules along the outer surface of the airway
30% cartilage; smooth muscle (Fig. 6.2). In some species, this outer plexus
contains large venous sinuses that can extend into the sub-
15% mucous glands; mucosal layer of the major bronchi, where there is no smooth
5% smooth muscle; and muscle between the cartilage and epithelium [16].
The venous blood from the rst two or three subdivi-
50% connective tissue matrix containing the bronchial sions of the bronchial tree drains into the azygous and hemi-
arterial, venous, and lymphatic vessels. azygous venous systems that empty into the vena cava. The
With progression toward the periphery of the bronchial remainder of the bronchial venous ow drains directly into
tree, the amount of cartilage and glands decrease, and the the pulmonary circulation, although there is debate as to how
percentage of smooth muscle increases to account for approx- much enters at the precapillary, capillary, and postcapillary
imately 20% of the total wall thickness in the bronchioles. levels [8, 15]. Airway disease increases the anastomotic ow
The degree to which the smooth muscle surrounds between the pulmonary and the bronchial vascular systems;
the airway lumen varies according to site. In the trachea and in diseases such as bronchiectasis, injection of the bron-
and mainstem bronchi, the airway smooth muscle is located chial arteries can result in rapid lling of the entire pulmo-
within the posterior membranous sheath, whereas, in the nary vascular tree right back to the pulmonic valve [15].
bronchioles, it surrounds the entire lumen of the airway [7, The bronchial circulation accounts for about 1% of car-
8, 13]. Consequently, the same degree of muscle shortening diac output and an average cardiac output of 5 l/min deliv-
has a smaller eect on the caliber of the trachea and cen- ers approximately 72 l of blood to the conducting airways
tral airways than on the distal bronchi and bronchioles [14]. over 24 h. Each liter of blood contains between 4 and
The adventitial layer consists of loose bundles of collagen 11 109 white blood cells, made up of approximately 60%
admixed with blood vessels, lymphatics, and nerves. In the neutrophils, 30% lymphocytes, 5% monocytes, 3% eosi-
peripheral conducting airways, this layer interacts with the nophils, and 2% basophils. The neutrophils do not divide after
surrounding lung parenchyma through alveolar attachments they leave the marrow have a relatively short half life within
that are distributed along the circumference of the adventi- the circulation and remain within the vascular space unless
tia. These alveolar attachments have the ability to limit the an inammatory response is present. Although much less is
amount of airway narrowing produced by smooth muscle known about eosinophils and basophil kinetics they are pre-
contraction, particularly at higher lung volumes [14]. dominately a tissue cell, indicating that their transit times
The systemic arterial supply to the bronchial tree origi- through the tissue compartment is much longer than that for
nates from the ventral side of the upper thoracic aorta in the migrating neutrophils. Monocytes on the other hand leave the
left hemithorax, while on the right the origin of the bronchial vascular space even in the absence of an inammatory stimu-
vessels is more variable. They may originate from the rst to lus and divide in the tissue to form the alveolar macrophages

72
Airway Pathology 6

FIG. 6.3 (A) Shows a small bronchiole with a collection of lymphocytes containing a germinal center. (B) Shows another airway containing a lymphoid
follicle where the germinal center stains strongly for B-cells. (C) Shows the same airway as in B where the CD4 cells appear at the edge of the follicle. (D)
Shows yet another area where the wall is thickened and fibrosed and the arrow pints to the smooth muscle in the airway wall. Adapted from Ref. [17] with
permission.

that are removed via the airways with very little if any re-entry of the follicle and the CD4 and CD8 T-cells are located
into the tissue. On the other hand lymphocytes leave the cir- around the edges of the germinal center. Thus dendritic cells
culation regularly with the assistance of specialized high migrating from the epithelium and subepithelium circulate
endothelial cells and re-entering the circulating blood with through the T- and B-cell rich regions of the follicle and can
the lymph as it drains into the venous system. present antigen to uncommitted T and B lymphocytes moving
A study of lung tissue obtained from patients at all through these regions in the lymph (Fig. 6.3). Moreover the
ve Global Initiative for Chronic Obstructive Lung Disease recirculation of lymphocytes though the follicle and back into
(GOLD) categories of chronic obstructive pulmonary dis- the circulation enhances the opportunity for T Helper cells
ease (COPD) severity has shown an association between the and B-cells that have recognized the same antigen to interact
decrease in FEV1 with both the extent of the accumulation of with each other and initiate an adaptive immune response. The
these cells (i.e. the percentage of airways containing cells) and appearance of lymphoid follicles with germinal centers in the
severity of this accumulation (i.e. the total accumulated vol- tissue provides histological evidence that an adaptive immune
ume of cells) within small airway tissue [17]. The accumulat- response has been mounted within the peripheral lung. Some
ing lymphocytes form follicles with germinal centers in both investigators have begun to study this response in detail [18].
the small conducting airways [17] and the parenchyma [18] of
the peripheral lung in both COPD [17, 18] and asthma [19].
These accumulations of lymphocytes (Fig. 6.3) are part of the
bronchial associated lymphoid system or bronchial-alveolar THE PATHOLOGY OF ASTHMA
lymphoid tissue (BALT) and dier from the regional lymph
nodes in that they have no capsule and no aerent lymphat-
ics. They are similar in structure to the tonsils and adenoids, Postmortem studies
the Peyers patches in the small bowel, and the appendix of
the cecum which are also part of the mucosal immune system. At postmortem, the lungs of patients who have died in sta-
They are organized in the same way as other lymph follicles tus asthmaticus remain markedly hyperinated after the
(Fig. 6.3) in that the B-cells are found in the germinal center thorax is opened. This hyperination is due to air trapping

73
Asthma and COPD: Basic Mechanisms and Clinical Management

caused by widespread plugging of the segmental, subseg- these cells can be divided according to the cytokine mes-
mental, and the smaller conducting airways by mucus and senger RNA (mRNA) and proteins that they produce [35].
cellular debris [20]. Although this luminal content may These experiments established that one type of T-cell clone
extend to the respiratory bronchioles, it usually stops short (Th-1) produced IL-2 and interferon- but no IL-4 or
of these structures and does not ll the alveolar airspaces. IL-5; whereas the second (Th-2) produced IL-4 and IL-5,
Examination of the cut surface of the lung reveals the but no IL-2 or interferon-. Both clones produced IL-3
plugged airways, but in contrast to parenchymal destruc- and GMCSF, and interactions between Th-1 and Th-2
tion seen in hyperinated emphysematous lungs the subtypes allowed one type of clone to inhibit the other. For
parenchyma of the asthmatic lung remains intact. example, IL-4 is a mast cell growth factor that also stimu-
Huber and Koesslers [21] classic 1922 paper on the lates IgE production, and IL-5 promotes the dierentiation
pathology of asthma reviewed 15 published cases and pro- and survival of eosinophils. Robinson and associates [36]
vided new data on 6 more. They noted that the pathology put forward the hypothesis that asthma was the result of a
consisted of common features that allowed asthma to be Th-2 response based on bronchoalveolar lavage and bron-
distinguished from other conditions. They emphasized the chial biopsy ndings. And subsequent studies of surgically
presence of intraluminal mucus secretion, airway epithelial resected lung specimens from asthmatic patients estab-
desquamation, and repair (e.g. goblet cell metaplasia), and lished that a similar inammatory process was present in
airway inammatory inltrates consisting of an admixture the smaller airways [37].
of mononuclear cells and eosinophils and the presence of a It has now become clear that the structural features of
thickened, hyalinized subepithelial basement membrane. the airways from asthmatic patients result from an inam-
Later studies based on electron microscopy and immuno- matory process involving tissue with a mucus-secreting
histochemistry showed that this feature of the basement surface. This response appears to be driven by a subset of
membrane was due to deposition of collagen brils and CD4 T lymphocytes producing cytokines that result in
extracellular matrix below the true basement membrane, an excess of eosinophils and an overproduction of IgE. The
rather than thickening of the basal lamina. Huber and end result is abnormal airway function, characterized by
Koesslers report noted that the tenacious plugs that ll excessive airway narrowing in response to external stimuli,
the airway lumen consist of an exudate of plasma contain- reversible airways obstruction, and gas trapping. Although
ing inammatory cells, particularly eosinophils, mixed with the majority of the symptoms produced by the process can
epithelial cells that had sloughed from the airway surface. be rapidly reversed with appropriate treatment, the process
Using an eyepiece micrometer to measure the external air- can be life threatening and result in sudden death.
way diameters, they concluded that the walls of bronchi and
bronchioli of more than 2 mm outside diameter were thick-
ened compared with non-asthmatic persons, and that this The relationship between airway structure
dierence was due to an increased thickness of all of the and function
components of the airway wall.
Over the next several decades, other reports con- The concept that the same degree of smooth muscle short-
rmed and extended these ndings [2229]. Comparing ening will cause greater reduction in airway caliber when
Floreys [30] basic studies of the inammatory process with the wall is thickened by disease has been suggested by sev-
these pathological ndings show that the structural changes eral authors [38, 39]. Moreno and associates [39] calculated
associated with asthma are consistent with an inamma- that the thickening of the airway wall observed in asthma
tory process involving a mucus-secreting surface. However, would have only a minor eect on the caliber of the lumen
this knowledge had relatively little impact on the allergists, of a fully dilated airway. However, when the smooth mus-
pulmonary physicians, and physiologists until the 1970s cle in the airway shortens, the increased tissue between
[31], because they were preoccupied with the concept that the muscle and lumen causes an excess reduction in airway
asthma was due to IgE sensitized mast cells releasing medi- caliber. Subsequent studies by James et al. [40] showed that
ators that caused excessive contraction of airway smooth the increase in wall thickness observed in the small airways
muscle following specic antigen challenge. of asthmatics was sucient to close the lumen of these
airways, even when smooth muscle shortening remained
within the accepted normal range. This suggests that normal
Bronchoscopic studies smooth muscle shortening may act in series with an abnor-
mally thickened airway wall to narrow the airway lumen
The nature of the airway pathology in persons with asthma and it follows that the reduction in airway caliber pro-
was further revealed by studies of tissue obtained through the duced by this mechanism would be rapidly reversed when
rigid and exible bronchoscope. These techniques allowed the smooth muscle relaxed. This showed that an important
investigators to obtain cells from living asthmatic patients by feature of the pathology of asthma was the change in the
both bronchoalveolar lavage and bronchial biopsies [3134]. structure of the airway wall produced by the remodeling
This brought physiologists and clinicians into closer agree- of the tissue that occurs in relation to the inammatory
ment with the pathologists view that the inammatory process. The important point is that these structural changes
process was important to the pathogenesis of both bronchial could result in excess airway narrowing with normal smooth
hyperresponsiveness and reversible airow obstruction. muscle contraction and that excessive smooth muscle short-
A very important conceptual development based on ening will enhance the eect of these structural changes on
the discovery that murine CD4 T-cell clones showed that the airway lumen.

74
Airway Pathology 6
(A) 20 Wiggs et al. [41, 42] extended this concept using a
18
computer model to test the eect of these structural changes
Asthma on airway function. Their analysis (Fig. 6.4) showed that
Total
16 maximum stimulation of the smooth muscle caused air-
14 way resistance to increase and reach a plateau in the normal
lung. This nding was consistent with previous observations
Resistance

12
10
by Woolcock et al. [43], who reported that the changes in
Control
the maximum volume of air that can be expired from the
8
lung in 1 s (FEV1) reached a plateau in normal subjects
6 when maximally stimulated by inhaled bronchoconstrictors.
4 However, when the data on airway structure were changed
2 from normal values to those found in asthmatic lungs, a
similar degree of airway smooth shortening resulted in a
0
sustained rapid increase in airway resistance without a pla-
0 0.5 1 1.5 2 2.5 3 3.5 4 4.5 5
teau. When the eect of disease on the central and periph-
(B) 20 eral airway function was examined separately, they found
18 Asthma that the increase in airway resistance was primarily due
Central to the eect of disease on the peripheral airways, where
16
smooth muscle shortening produced widespread airway clo-
14
sure. The concept that the peripheral airways are the major
Resistance

12 site of obstruction in asthma has now been conrmed by


10 Control direct measurements in living asthmatic patients reported
8 by Yanai et al. [44].
The changes that are produced in the airway tis-
6
sue also reduce the function of the conducting airways.
4 Lambert [45] was the rst to systematically study the nor-
2 mal folding pattern of the bronchial mucosa, and showed
0 that in asthma the multiple mucosal folds that occur when
0 0.5 1 1.5 2 2.5 3 3.5 4 4.5 5 normal airways narrow were replaced with fewer and larger
(C) 20 folds that reduced airway caliber. Both Lambert and Wiggs
18
et al. [46] suggested that the mucosal folding pattern was
Asthma controlled by the stiness of the subepithelial layer rela-
Peripheral
16 tive to that of the surrounding airway tissue. This analysis
14 suggested that changes in the subepithelial connective tis-
sue might play a key role in determining the pattern of
Resistance

12
10
mucosal folding. Lambert and Wiggs et al. argued that the
Control
formation of a large number of folds in the normal airway
8 placed a load on the airway smooth muscle that tended
6 to prevent airway closure at low lung volumes. It followed
4 that a change in the mucosal folding pattern in asthma may
2 be one way in which this disease causes peripheral airway
dysfunction.
0
The caliber of the airway lumen is also inuenced by
0 0.5 1 1.5 2 2.5 3 3.5 4 4.5 5
airway surface tension that is normally low in the small air-
Log (dose)
ways because they are lined with surfactant [47]. Exudation
FIG. 6.4 Data from Wiggs et al. (Refs. [38, 39]) showing results from a
of plasma and the secretion of mucus on to the small airway
computer model of the airways. The structural data from normal lungs lumen surface should increase surface tension and cause the
were used to obtain the control measurements. These show that airway airways to narrow. The analysis of induced sputum has con-
resistance increases from about 1 cmH2O per liter per second to reach a rmed that plasma proteins, mucus, and inammatory cells
plateau of 12 cmH2O per liter per second, with maximum contraction of are present in the airway lumen even in mild asthma, sug-
the airways smooth muscle. (A) When the structural features of asthmatic gesting that some of the abnormalities in airway function of
airways were used in the same computer model, the airway resistance asthmatics might result from the presence of this material in
continued to increase and did not reach a plateau at any physiologically the lumen. Kuyper and colleagues [48] recently re-assessed
meaningful value. (B) Data obtained from the model when the structural the importance of the occlusion of the airway lumen by
changes caused by asthma were limited to the central airway (i.e. those
inammatory exudates in deaths attributed to asthma. By
2 mm diameter). Note that asthmatic changes in the central airways
increased total resistance and resulted in a plateau slightly greater than the
examining the airway wall and luminal content of 275 air-
control lungs. (C) However, when the asthmatic changes were placed in ways from 93 patients with fatal asthma aged 1049 years.
the peripheral airway, the resistance increased without reaching a plateau. Compared with control airways obtained from persons that
These data suggest that asthmatic changes in the peripheral airways have died of causes unrelated to the lungs, the asthmatic airways
a much greater effect on overall airway function than changes that occur in showed much more extensive lumenal occlusion, by mucus
the central airways. Reproduced from Ref. [38] with permission. and cells. They concluded that widespread airway occlusion

75
Asthma and COPD: Basic Mechanisms and Clinical Management

was the major cause of death from asthma and death due to
closure of empty airways by excessive bronchoconstriction
must be a rare event. Moreover the extensive nature of the
airway occlusion observed at autopsy in these cases suggests
that bronchoconstriction superimposed on airways that
have become partially occluded is the most probable cause
of sudden death.

THE PATHOLOGY OF COPD

The inammatory process contributes to the pathogenesis of


chronic cough and sputum production [49], peripheral air-
ways obstruction [3, 17, 50], and emphysematous destruc-
tion of the lung surface [51] that dene COPD. As tobacco
smoking produces lung inammation in everyone, and only
1520% of heavy smokers develop COPD, clinical disease
from cigarette smoke-induced airway inammation must
develop in this minority of people because it amplied by
either genetic or environmental risk factors [52].

Chronic bronchitis
Cough and sputum production are the features of airways FIG. 6.5 Low-power photomicrograph of a normal bronchus showing
disease that dene chronic bronchitis [53] and these symp- the opening of a bronchial gland duct into the lumen and its connection
toms can be present either with or without airways obstruc- with the submucosal gland. The bronchial muscle and cartilage are clearly
tion [49]. Figure 6.5 shows the histology of a normal labeled. The inflammatory process associated with chronic bronchitis
bronchus at low power where with the connection between involves the mucosa, gland ducts, and glands of the bronchi that are
between 2 and 4 mm internal diameter. Original photograph taken by the
the epithelial lining of the bronchial lumen to the mucus
late Dr. William Thurlbeck.
duct and gland is clearly seen. Reid [54] used the relative
size of the mucus glands to the airway wall as a yardstick for
measuring chronic bronchitis and downplayed the inuence
of inammation in driving mucus production. However, a The site of airways obstruction
reevaluation of this problem some years later showed that
chronic bronchitis was associated with inammation of The site of airways obstruction in COPD is in the smaller
the airway mucosal surface, the submucosal glands, and conducting airways that include bronchi and bronchioles of
gland ducts, particularly in the smaller bronchi between less than 2 mm diameter [3]. Direct measurements of air-
2 and 4 mm in diameter [49]. The nature of the inam- ways resistance in dogs [4] and in similar measurements
matory process present in these airways is now quite well in postmortem human lungs [3] established, in the normal
established with several studies reporting that CD8 lym- lung, that the small peripheral airways oer very little resist-
phocytes are present in excess numbers in smokers with ance to air ow. Although van Brabant et al. [63] subse-
chronic bronchitis [55, 56]. quently argued that these peripheral airways account for a
Sputum production represents the clearance of a larger proportion of the total airways resistance in the nor-
mucoid inammatory exudate from the lumen of the bronchi. mal lung, there is general agreement that the peripheral air-
This exudate contains plasma proteins, inammatory cells, ways are the major site of obstruction in COPD [3, 44, 63].
and small amounts of mucus added from goblet cells on the The causes of the reduced forced expiratory ow that
surface epithelium and the epithelial glands. Although the dene COPD include destruction of alveolar support of
size of the bronchial mucous glands tends to increase [49] the peripheral airways [64], the loss of elastic recoil in the
in chronic bronchitis, Thurlbeck and Angus [57] showed parenchyma supporting the airways [65], a decrease in the
that the Reid index was normally distributed with no clear elastic force available to drive ow out of the lung [66], and
separation between patients with chronic bronchitis and structural narrowing of the airway lumen by a remodeling
controls. Chronic bronchitis also results in an increase in process [3, 17, 67]. Comparison of the histological appear-
airway smooth muscle, a generalized increase in the con- ance of peripheral airways at dierent degrees of severity of
nective tissue in the airway wall, degenerative changes COPD shows a progressive increase in the magnitude of the
in the airway cartilage, and a shift in epithelial cell type inammatory process with thickening of the airway walls
that increases the number of goblet and squamous cells accompanied by occlusion of the airway lumen by inamma-
[5862]. tory mucous exudates [17]. However, multivariate analysis of

76
Airway Pathology 6

FIG. 6.6 (A) Shows a small bronchiole from a patient with severe (GOLD-4) COPD the lumen is nearly fully occluded and the mucosa is folded because it
was fixed in a collapsed state. (B) Shows a redrawing of the same airway where the lumen occlusion is less severe because the airway has been fully inflated
by removing the folds from the mucosa using a computer (C) Compares the frequency distribution of the ratio of luminal content area to the total area of
the lumen for 562 airways from 42 patients with GOLD-4 COPD before and after the expansion of the lumen shown in A and B. As we do not breathe at
either full expansion of the lung or at minimal volume many airways must have been partially or fully occluded in the normal breathing range. (D) Shows the
relationship between the severity of the occlusion of the airway lumen with the level of FEV1. Adapted from Ref. [17] with permission.

these data indicated that thickening of the airway walls and Emphysema
the occlusion of the lumen by inammatory exudates con-
taining mucus explained more of the variance in the decline Emphysema has been dened as abnormal permanent
in FEV1 than the inltration of the airway tissue by any of enlargement of airspaces distal to terminal bronchioles,
the inammatory cell types examined [17]. Interestingly the accompanied by destruction of their walls without obvi-
severity of the occlusion of the airways lumen by inamma- ous brosis [79]. This denition emphasizes the destruction
tory exudates containing mucus (Fig. 6.6) has also associated of the alveolar surface with a minimal reparative response in
with premature death in persons with severe (GOLD-3) and the lung matrix and the ability of this destructive process to
very severe (GOLD-4) disease [68]. reduce the gas-exchanging surface of the lung. The centrilob-
The lung inammatory changes that are associated ular form of emphysema (Fig. 6.7) resulting from dilatation
with cigarette smoking have been documented in autopsy and destruction of the respiratory bronchioles in the center of
studies [6973], resected lung specimens [4951, 74] lung secondary lobule of Miller has been most closely associated
biopsies [75], and indirectly by examining bronchoalveolar with smoking [80, 82]. The panacinar form of emphysema, on
lavage uid [7678]. Collectively these data support the the other hand, results from a uniform destruction of all of
concept that cigarette smoke-induced lung inammation the acini in the entire secondary lobule and is characteristic
is present in all smokers, including those with normal lung of the lesions found in the lungs in 1-antitrypsin deciency
function and that it persists long after people have stopped [83]. The terms distal acinar, mantle, or periseptal emphy-
smoking [17]. The reasons for both the amplication of the sema are used to describe lesions that occur in the periphery
smoking-related inammatory process in individual that of the lobule and along the lobular septae particularly in the
develop COPD, the persistence of this response after the subpleural region. These lesions have been associated with
smoking has stopped and the precise relationship between spontaneous pneumothorax in young adults and bullous lung
the inammatory response and the remodeling process disease in older individuals [84]. In far-advanced parenchy-
needs further clarication. mal destruction such as that frequently observed in end-stage

77
Asthma and COPD: Basic Mechanisms and Clinical Management

(A) (B)

(C) (D)

CLE
TB

FIG. 6.7 (A) Normal lung photographed from the pleural surface after a postmortem bronchogram. The connective tissue outlining the secondary lobule
is clearly seen (solid arrow) and a terminal bronchiole (TB) indicated by a clear arrow supplies a single acinus. (B) Photomicrograph of an acinus showing a
terminal bronchiole (TB), respiratory bronchiole (RB), and alveolar ducts (AD). Their structure is represented by the fine spray of contrast at the end of the TB
shown in the bronchogram in (A). (C) Diagram of the lesion in centrilobular emphysema, showing the dilation and destruction of the respiratory bronchioles.
(D) Postmortem bronchogram showing a centrilobular emphysematous lesion (CLE) outlined by bronchographic material. Parts (C) and (D) reproduced from
Refs [80, 81], with permission.

COPD, these descriptive terms are less helpful because centri- arrhythmia, pulmonary embolism, pneumothorax, postop-
lobular disease eventually destroys the entire lung lobule and erative complications, and lung cancer [86], with no cause
destroyed lobules can coalesce to form much larger lesions. being established in 303 (25%) of these 1205 admissions.
The destruction of the lung by centrilobular emphy- Early studies by Stuart-Harris and associates [87, 88] showed
sema results in a loss of elastic recoil with subsequent that COPD patients with acute upper respiratory infec-
hyperination and a change in the pressurevolume rela- tions are more likely to have signs of infection in the lower
tionship of the emphysematous lung tissue compared to airways than normal controls. Probably because acute viral
the surrounding normal lung such that at any volume recoil upper respiratory tract infections increase the risk of aspira-
pressure is less [81]. This decrease in lung elastic recoil tion of mucoid exudate containing large numbers of bacte-
diminishes expiratory ow by reducing the pressure avail- ria from the upper airways, but also because viral infection
able to drive air out of the lung [66], and interferes with gas reduces both the mucociliary clearance and bacterial kill-
exchange by reducing lung surface area [85]. ing in the lower airways [8991]. Streptococcus pneumonia,
Staphylococcus aureus, and Hemophilus inuenzae are the most
common bacteria infecting the lower airways during an acute
Acute exacerbations of COPD viral infection [90, 91], and these infections account for the
positive eect of antibiotics in some exacerbations of COPD
In a hospital-based study of 1205 admissions to ve hospi- [92]. Two important early studies established an association
tals for acute exacerbations of COPD, infections accounted between acute illness and serologic laboratory evidence of
for 520 (43%), heart failure for 260 (22%), and 122 (10%) viral infection in patients with COPD [93, 94]. The devel-
cases were attributed to a variety of etiologies that included opment of polymerized chain reaction (PCR)-based viral

78
Airway Pathology 6
diagnostic techniques have improved the sensitivity of the References
diagnosis of viral infection and the use of real time PCR to
measure viral copy numbers will likely improve its specicity. 1. Horseld K, Cumming G. Morphology of the bronchial tree in man.
Early reports of studies based on these techniques indicate J Appl Physiol 24: 37383, 1968.
the importance of rhinoviral infection in causing acute exac- 2. Weibel ER. Morphometry of the Human Lung. New York: Academic
erbations in an outpatient setting with inuenza A and the Press, 1963.
corona virus OC-43 being more prominent causes of exacer- 3. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of airways
bations that require admission to hospital [9597]. obstruction in chronic obstructive lung disease. N Engl J Med 48:
The pathological features of the lung during acute 42131, 1968.
4. Macklem PT, Mead J. Resistance of central and peripheral airways
exacerbations of COPD are incompletely dened. Primarily
measured by a retrograde catheter. J Appl Physiol 22: 395401, 1967.
because postmortem studies of patients that die during 5. Bai A, Eidelman DH, Hogg JC et al. Proposed nomenclature for
exacerbations are complicated by terminal events and biopsy quantifying subdivisions of the bronchial wall. J Appl Physiol 77:
studies of patients with severe (GOLD-3 and very severe 101114, 1994.
GOLD-4 disease during an exacerbation cannot be car- 6. Neutra M, Podykula H, Weiss L Histology, Cell and Tissue Biology, 5th
ried out without excess risk to the patient. One exception edn, pp. 658706. Amsterdam: Elsevier Biomedical, 1983.
is an important study by Saetta et al. [98] in less severely 7. Weiss L (ed.), Histology, Cell and Tissue Biology, 5th edn, pp. 788868.
ill patients with GOLD class 2 disease and a mean FEV1 Amsterdam: Elsevier Biomedical, 1983.
of 62  7% predicted implicated the eosinophil as a cell of 8. Miller WS. The blood vessels in the lung. The Lung, 3rd edn, pp. 7384.
interest. Clearly more studies especially those that can make Springeld, IL: Charles Thomas, 1943.
9. Roche WR, Beasley R, Williams JH, Holgate ST. Subepithelial brosis
use of non-invasive or less invasive techniques than biopsy
in the bronchi of asthmatics. Lancet i: 52024, 1989.
are needed to help clarify this important area of COPD. 10. Dunnill MS. The pathology of asthma with special reference to changes
in the bronchial mucosa. J Clin Pathol 13: 2733, 1960.
11. Dunnill MS. Quantitative methods in the study of pulmonary pathol-
ogy. Thorax 17: 32028, 1962.
12. Takizawa T, Thurlbeck WM. Muscle and mucous gland size in the
SUMMARY major bronchi of patients with chronic bronchitis, asthma and asth-
matic bronchitis. Am Rev Respir Dis 104: 33136, 1971.
13. Plopper CG. Ten Have-Opbroek AAW Anatomical and histologi-
Asthma cal classication of the bronchioles. In: Epler GR (ed.), Diseases of the
Bronchioles, pp. 1525. New York: Raven Press, 1994.
The pathology of asthma is dominated by widespread plug- 14. Macklem PT. Bronchial hyporesponsiveness. Chest 87: 158S159S,
ging of the segmental, subsegmental, and smaller conducting 1985.
airways that leads to hyperination but not destruction of 15. Cudkowicz L. The Human Bronchial Circulation in Health and
the parenchyma. These airway plugs are a manifestation of Disease. . Baltimore: Williams & Wilkins, 1968.
the uid and cellular exudative phase of an inammatory 16. Hill P, Goulding D, Webber SE, Widdicombe JG. Blood sinuses in the
process based in the airway tissue (Fig. 6.6). The cytokine submucosa of the large airways of sheep. J Anat 162: 23547, 1989.
and cellular content of this process is consistent with a Th- 17. Hogg JC, Chu F, Utokaparch S et al. The nature of small-airway
2 type of immune response [36, 37], and quantitative post- obstruction in chronic obstructive pulmonary disease. N Engl J Med
mortem studies have established that the airway occlusion 350: 264553, 2004.
18. van der Strate BW, Postma DS, Brandsma CA et al. Cigarette smoke-
by plugs of this exudate is the major determinant of death
induced emphysema: A role for the B cell?. Am J Respir Crit Care Med
from asthma [48]. 173: 75158, 2006, Epub 2006 Jan 6.
19. Elliot JG, Jensen CM, Mutavdzic S, Lamb JP, Carroll NG, James AL.
COPD Aggregations of lymphoid cells in the airways of nonsmokers, smokers,
The chronic bronchitis of COPD is dened by excess cough and subjects with asthma. Am J Respir Crit Care Med 169: 71218, 2004.
with sputum production and is associated with an inam- 20. Rigler LG, Koucky R. Roentgen studies of pathological physiology of
bronchial asthma. Am J Roentgenol 39: 35362, 1938.
matory process located in the mucosa, gland ducts, and
21. Huber HL, Koessler KK. The pathology of bronchial asthma. Arch
glands of intermediate-sized bronchi between 2 and 4 mm Intern Med 30: 689760, 1922.
internal diameter. The airway obstruction in COPD is the 22. Cardell BS, Pearson RSB. Death in asthmatics. Thorax 14: 34152, 1959.
result of a similar inammatory response in the smaller 23. Earle BV. Fatal bronchial asthma: A series of 15 cases with a review of
bronchi and bronchioles under 2 mm internal diameter, the literature. Thorax 8: 195206, 1953.
where the repair process associated with chronic inam- 24. Houston JC, de Navasquez S, Trounce JR. A clinical and pathological
mation thickens the airway wall and narrows the lumen to study of fatal cases of status asthmaticus. Thorax 8: 20713, 1953.
cause xed airway obstruction. Emphysematous destruction 25. Dunnill MS, Massarella GR, Anderson JA. A comparison of the
of the lung surface contributes to the decline in FEV1 by quantitative anatomy of the bronchi in normal subjects, in status asth-
reducing the elastic recoil force available to drive air out of maticus, in chronic bronchitis and in emphysema. Thorax 24: 17679,
1969.
the lung and is responsible for reduced gas exchange. The
26. MacDonald IG. The local and constitutional pathology of bronchial
acute exacerbations of COPD that occur with increas- asthma. Ann Intern Med 6: 25377, 1933.
ing frequency as the disease progresses have several known 27. Messer J, Peters GA, Bennet WA. Cause of death and pathological
causes including infection, right heart failure, and pulmo- ndings in 304 cases of bronchial asthma. Dis Chest 38: 61624, 1960.
nary embolism but it is discouraging that in up to 25% of 28. Richards W, Patrick JR. Death from asthma in children. Am J Dis Child
cases admitted to hospital the cause is unknown. 110: 421, 1965.

79
Asthma and COPD: Basic Mechanisms and Clinical Management

29. Saetta M, di Stefano A, Rosina C, Thiene G, Fabbri LM. Quantitative relationship of CD-8  T lymphocytes with FEV1. Am J Respir Crit
structural analysis of peripheral airways and arteries in sudden fatal Care Med 155: 38287, 1997.
asthma. Am Rev Respir Dis 143: 13843, 1991. 56. Saetta M, Di Stefano A, Turato G et al. T lymphocytes in the periph-
30. Florey H. The secretion of mucus and inammation in mucus mem- eral airways of smokers with chronic obstructive pulmonary disease.
branes. In: Florey H (ed.), General Pathology, 3rd edn, pp. 16796. Am J Respir Crit Care Med 157: 82226, 1998.
London: Lloyd-Luke Medical Books, 1962. 57. Thurlbeck WM, Angus GE. The distribution curve for chronic bron-
31. Ciba Symposium on the Identication of Asthma. January 1971. chitis. Thorax 19: 43642, 1964.
32. Djukanovic R, Wilson JW, Lai CKW, Holgate ST, Howarth PH. The 58. Jamal K, Cooney TP, Fleetham JA, Thurlbeck WM. Chronic bronchi-
safety aspects of beroptic bronchoscopy, bronchoalveolar lavage, and tis: Correlation of morphological ndings and sputum production and
endobronchial biopsy in asthma. Am Rev Respir Dis 143: 77277, 1991. ow rates. Am J Respir Dis 129: 71922, 1984.
33. Djukanovic R, Roche WR, Wilson JW et al. Mucosal inammation in 59. Carlile A, Edwards C. Structural variations in the main bronchi of the
asthma. Am Rev Respir Dis 142: 43457, 1990. left lung: a morphometric study. Br J Dis Chest 77: 34448, 1983.
34. Glynn AA, Michaels L. Bronchial biopsy in chronic bronchitis and 60. Mackenzie HI, Outhred KG. Chronic bronchitis in coal miners:
asthma. Thorax 15: 14253, 1960. Antimortem/postmortem comparisons. Thorax 24: 52735, 1969.
35. Mosmann TR, Coman RL. TH1 and TH2 cells: Dierent patterns 61. Haraguchi M, Shemura S, Shirata K. Morphologic analysis of bron-
of lymphokine secretion lead to dierent functional properties. Annu chial cartilage in chronic obstructive pulmonary disease and bronchial
Rev Immunol 7: 14573, 1989. asthma. Am J Respir Crit Care Med 159: 100513, 1999.
36. Robinson DS, Hamid Q, Ying S et al. Predominant TH2-like bron- 62. Thurlbeck WM, Pun R, Toth J, Fraser RG. Bronchial cartilage in
choalveolar T-lymphocyte population in atopic asthma. N Engl J Med chronic obstructive lung disease. Am Rev Respir Dis 109: 7380, 1974.
326: 298304, 1992. 63. van Braband T, Cauberghs M, Verbeken E et al J Appl Physiol: Respir.
37. Hamid Q, Song Y, Kotsimbos TC et al. Small airways inammation in Environ. Exercise Physiol. 55: 173342, 1983.
asthma. J Allergy Clin Immunol 100: 4451, 1997. 64. Dayman H. Mechanics of airow in health and emphysema. J Clin
38. Friedman BJ. Functional anatomy of the bronchi. Bull Pathophysiol Invest 3031: 117590, 1951.
Respir 8: 54551, 1972. 65. Butler J, Caro C, Alkaler R, Dubois AB. Physiological factors aecting
39. Moreno R, Hogg JC, Par PD. Mechanics of airway narrowing. Am airway resistance in normal subjects and in patients with obstructive
Rev Respir Dis 133: 117180, 1986. airways disease. J Clin Invest 39: 58491, 1960.
40. James AL, Par PD, Hogg JC. The mechanics of airway narrowing in 66. Mead J, Turner JM, Macklem PT, Little J. Signicance of the relation-
asthma. Am Rev Respir Dis 139: 24246, 1989. ship between lung recoil and maximum expiratory ow. J Appl Physiol
41. Wiggs BR, Moreno R, Hogg JC, Hilliam C, Par PD. A model of the 22: 95108, 1967.
mechanics of airway narrowing. J Appl Physiol 69: 84960, 1990. 67. Matsuba K, Thurlbeck WM. The number and dimensions of small air-
42. Wiggs BR, Bosken C, Par PD, James A, Hogg JC. A model of air- ways in emphysematous lungs. Am J Pathol 67: 26575, 1972.
way narrowing in asthma and chronic obstructive pulmonary disease. 68. Hogg JC, Chu FS, Tan WC et al. Survival following lung volume
Am Rev Respir Dis 145: 125158, 1992. reduction in COPD: Insights from small airway pathology. Am J Respir
43. Woolcock AJ, Salome CM, Yan K. The shape of the dose-response Crit Care Med 16, 2007.
curve to histamine in asthmatic and normal subjects. Am Rev Respir 69. McLean KA. Pathogenesis of pulmonary emphysema. Am J Med 25:
Dis 130: 7175, 1984. 6274, 1958.
44. Yanai M, Sekizawa K, Ohrui T, Sasaki H, Takishima T. Site of airway 70. Niewoehner DE, Kleinerman J, Reisst DB. Pathologic changes in
obstruction in pulmonary disease: Direct measurements of intrabron- the peripheral airways of young cigarette smokers. N Engl J Med 291:
chial pressure. J Appl Physiol 72: 101623, 1992. 75558, 1974.
45. Lambert R. The role of the bronchial basement membrane and airway 71. Auerbach O, Garnkle L, Hammond EC. Relation of smoking and
collapse. J Appl Physiol 71: 66673, 1991. age to ndings in lung parenchyma: A microscopic study. Chest 65:
46. Wiggs BR, Hrousis CA, Drazen JM, Kamm RD. The implications of 2935, 1974.
airway wall buckling in asthmatic airways. Am J Respir Crit Care Med 72. Auerbach O, Hammond EC, Garnkle L, Benante C. Relation of
149: A585, 1994. smoking and age to emphysema: Whole lung section study. N Engl J
47. Macklem PT, Proctor DF, Hogg JC. The stability of peripheral airways. Med 286: 85357, 1972.
Respir. Physiol. 8: 191203, 1970. 73. Petty TL, Silverds GW, Stanford RE, Baird ME, Mitchell MS. Small
48. Kuyper LM, Par PD, Hogg JC, Lambert RK, Ionescu D, Woods R, airway pathology is related to increased closing capacity and abnor-
Bai TR. Characterization of airway plugging in fatal asthma. Am J Med mal slope of phase III in excised human lungs. Am Rev Respir Dis 121:
115: 611, 2003. 44956, 1980.
49. Mullen JBM, Wright JL, Wiggs B, Par PD, Hogg JC. Reassessment 74. Wright JL, Lawson LM, Par et al. Morphology of peripheral airways in
of inammation in the airways of chronic bronchitis. BMJ 291: 1235 current smokers and ex-smokers. Am Rev Respir Dis 127: 47477, 1983.
39, 1985. 75. Ollerenshaw SL, Woolcock AJ. Characteristics of the inammation in
50. Cosio M, Ghezzo M, Hogg JC et al. The relation between structural biopsies from large airways in subjects with asthma and chronic airow
changes in small airways and pulmonary function tests. N Engl J Med limitation. Am Rev Respir Dis 145: 92227, 1992.
298: 127781, 1978. 76. Gadek JE, Fells JA, Crystal RG. Cigarette smoking induces a func-
51. Retamales I, Elliott M, Meshi B et al. Amplication of inammation tional antiprotease deciency in the lower respiratory tract of humans.
in emphysema and its association with latent adenoviral infection. Science 206: 31516, 1979.
Am J Respir Crit Care Med 164: 46973, 2001. 77. Hunninghake GW, Crystal RG. Cigarette smoking and lung destruc-
52. Speizer FE, Tager IB. Epidemiology of chronic mucus hypersecretion tion: Accumulation of neutrophils in the lungs of cigarette smokers.
and obstructive airways disease. Epidemiol Rev 1: 12442, 1979. Am Rev Respir Dis 128: 83338, 1983.
53. Ciba Guest Symposium Report. Terminology, denitions and clas- 78. Stone DJ, Galor JD, McGowan SE et al. Functional 1-protease inhibitor
sications of chronic pulmonary emphysema and related conditions. in lower respiratory tract of cigarette smokers is not decreased. Science
Thorax 14: 28699, 1959. 221: 118789, 1983.
54. Reid L. Measurement of the bronchial mucous gland layer: A diagnos- 79. Snider GL, Kleinerman JL, Thurlbeck WM, Bengally ZH. Denition
tic yardstick in chronic bronchitis. Thorax 15: 13241, 1960. of emphysema. Report of a National Heart, Lung and Blood Institute,
55. OShaughnessy TC, Ansari TW, Barnes NC, Jeery PK. Inammation Division of Lung Disease Workshop. Am Rev Respir Dis 132: 18285,
in bronchial biopsies of subjects with chronic bronchitis: Inverse 1985.

80
Airway Pathology 6
80. Leopold JG, Goe J. Centrilobular form of hypertrophic emphysema 91. Lauria DB, Blumeneld HL, Ellis JT, Kilbourne ED, Rogers DE.
and its relation to chronic bronchitis. Thorax 12: 21935, 1957. Studies on inuenza in the pandemic of 195758. II: Pulmonary com-
81. Hogg JC, Macklem PT, Thurlbeck WM. The elastic properties of plication of inuenza. J Clin Invest 38: 21365, 1959.
the centrilobular emphysematous space. J Clin Invest 48: 130612, 1969. 92. Anthonisen NR, Manfreda J, Warren CP et al. Antibiotic therapy in
82. Anderson AE, Hernandez JA, Holmes WL, Foraker AG. Pulmonary exacerbations of COPD. Ann Intern Med 106: 196204, 1987.
emphysema: Prevalence, severity and anatomical patterns with respect 93. Monto AS, Higgins MW, Ross HW. The Tecumseh study of respira-
to smoking habits. Arch Environ Health 12: 56977, 1966. tory illness. VIII: Acute infection in chronic respiratory disease and
83. Pratt PC, Kilborn KH. A modern concept of emphysemas based on comparison groups. Am Rev Respir Dis 111: 2736, 1975.
correlations of structure and function. Hum Pathol 1: 44553, 1970. 94. Smith CB, Golden CA, Canner RE, Renzetti AD. Association of viral
84. Laurenzi GA, Toreno GM, Fishman AP. Bullous disease of the lung. and mycoplasmal pneumonia infections with acute respiratory illness
Am J Med 36: 36178, 1962. in patients with COPD. Am Rev Respir Dis 121: 22532, 1980.
85. McLean A, Warren PM, Gilooly M, Lamb D. Microscopic and mac- 95. Seemungal T, Harper-Owen R, Bhowmik A et al. Respiratory viruses,
roscopic measurements of emphysema: Relation to carbon monoxide symptoms, and inammatory markers in acute exacerbations and sta-
gas transfer. Thorax 47: 14449, 1992. ble chronic obstructive pulmonary disease. Am J Respir Crit Care Med
86. Connors AF, Dawson NV, Thomas C et al. Outcomes following acute 164(9): 161823, 2001.
exacerbation of severe chronic obstructive lung disease. Am J Respir Crit 96. Ko FW, Ip M, Chan PK et al. Viral etiology of acute exacerbations of
Care Med 154: 95967, 1996. chronic obstructive pulmonary disease in Hong Kong. Chest 15, 2007. ,
87. Stuart-Harris CH. The role of bacterial and viral infection in chronic Epub ahead of print
bronchitis. Arch Environ Health 16: 58695, 1968. 97. Tan WC, Xiang X, Qiu D, Ng TP, Lam SF, Hegele RG. Epidemiology
88. Stuart-Harris CH. Infection, the environment and chronic bronchitis. of respiratory viruses in patients hospitalized with near-fatal asthma,
J Roy Coll Physicians Lond 5: 35161, 1971. acute exacerbations of asthma, or chronic obstructive pulmonary dis-
89. Knipe DN. Virushost interactions. In: Fields BM, Knipe DN, ease. Am J Med 115(4): 27277, 2003.
Howley PM (eds). Fields Virology, pp. 27399. New York: Lippincott- 98. Saetta M, Di Stefano A, Maestrelli P et al. Airway eosinophilia in
Raven, 1996. chronic bronchitis during exacerbations. Am J Respir Crit Care Med
90. Hers JF, Masurel N, Mulder J. Bacteriology and histopathology of the 150: 164652, 1994.
respiratory tract in fatal Asian inuenza. Lancet ii: 114143, 1958.

81
Airway Remodeling
7
CHAPTER

Airway remodeling may be dened as a process of consequence of long-term airways disease, stud-
sustained disruption and modication of structural ies have revealed early manifestations in asth-
Stephen T. Holgate
cells and tissues leading to the development of a matics [7] and young smokers [8], suggesting School of Medicine, University of
new airway-wall structure and as a consequence, that remodeling maybe as much part of the pri- Southampton, Southampton, UK
new functions. Although this process was noted mary pathology of the disease rather than simply
as long ago as 1922 [1], interest in the underly- being a result of chronic inammation. However,
ing in asthma has risen only in recent years, the a considerable degree of variability in susceptibil-
debate about asthma causation largely being ity to remodel the changes exists in both patient
driven by concepts in immunology and inam- groups. Computerized tomography (CT) shows
mation. In chronic obstructive pulmonary disease increased airway wall thickening in proportion
(COPD), although airway structural changes are to disease severity in both children and adults
well recognized [2], the underlying pathogenesis with asthma [9, 10], whereas in highly labile
has received much less attention, possibly due to brittle asthma, this change is notably absent
the more peripheral location of pathology and the [11]. At a population level, asthma is associated
overshadowing interest in the adjacent emphyse- with a greater than normal decay in lung func-
matous tissue destruction. tion over time and associated loss of corticoster-
In both conditions, a number of remod- oid responsiveness [12]. In a small proportion of
eling processes occur; but in proportion, the asthma patients with severe chronic disease the
changes observed are quite dierent. Principal decline in lung function is more progressive and
among these changes are airway brosis, an ele- associated with persistence of symptoms despite
vation in smooth muscle mass, mucous meta- corticosteroid therapy, thereby resembling a
plasia, and glandular hypertrophy, in addition COPD-like state [13], although the pathology
to less well dened alterations of the bronchial that underlies this in the two disorders diers
vasculature and nerves to create an abnormal [14], The accelerated decline in lung function in
airway wall. In asthma, segmental and subseg- asthma may reect not only the presence of air-
mental bronchial walls are thickened over their way structural modication, but also the inade-
entire size range [3]. In COPD, only the inner quacy of currently available treatments to modify
wall area of the large airways is convincingly the remodeling processes.
thicker [4]. The peripheral airways (2 mm Airway remodeling may have a number
diameter), normally devoid of either supporting of clinical consequences for both asthma and
cartilage or bronchial glands, are also conspicu- COPD. For example, bronchial hyperresponsive-
ously remodeled in COPD [5, 6]. ness has been postulated to be a function of the
physical eects of overall airway wall thickening,
in addition to airway inammation and smooth
NATURAL HISTORY AND CLINICAL muscle reactivity [15]. Of interest, however is
IMPORTANCE the inverse relationship between airway wall
thickening demonstrated by CT and by endo-
bronchial ultrasound and airway hyperrespon-
The natural history of remodeling is not well siveness [16, 17]. Individual remodeling changes,
understood. Though it is surmised to be a such as an increased smooth muscle bulk or an

83
Asthma and COPD: Basic Mechanisms and Clinical Management

enhanced mucus producing facility, have their own implica- are causally involved, with their eects being augmented
tions for both symptom severity and airway function. by the airway inammatory responses induced by these
agents. Thus, the generation of reactive oxygen species and
the products of inltrating inammatory cells including
arginine-rich eosinophilic proteins [29], mast cell tryptase
MECHANISMS OF REMODELING [30], neutrophil elastase, and metalloproteases derived from
eosinophils and mast cells [31, 32], neutrophils and macro-
phages all induce epithelial injury. The result in asthma is
Epithelial damage, airway inflammation, a fragile bronchial epithelium with weakening of junctional
and epithelial repair adhesion structures, whereas in COPD the epithelium pro-
liferates and thickens. Consequently, a feature of asthma is
Responses of the airway epithelial barrier to injury and the shedding of columnar epithelial cells from their basal
manipulation and abnormalities in the ensuing processes cell attachments [3336]. In COPD, although atrophy and
of repair are the most likely causes of remodeling. In both shedding of the epithelium may occur [37], focal squamous
adult and children with chronic asthma, there is a good metaplasia is much more common [38, 39].
evidence that following repeated epithelial injury by envi- Altered airway adhesion molecule expression, a hall-
ronmental factors (e.g. allergens, viruses, and pollutants) mark of an active repair process, has been demonstrated in
the repair processes involve intercellular signaling between asthma and COPD. This includes enhanced expression of
the damaged and regenerating epithelium and the underly- CD44 [40], the integrins [41], and E cadherin [42] in asthma,
ing subepithelial (myo)broblast sheet, to inuence matrix while in both asthma and COPD there is augmented expres-
synthesis and the mass and composition of structures lying sion of ICAM-1 [43]. A recent important nding in asthma
beneath [18]. Under these conditions, the asthmatic airway is the incomplete formation of tight junctions at the apex of
is behaving like a chronic wound with release of proinam- the columnar epithelial cells which is preserved in tissue cul-
matory cytokines and multiple growth factors leading to ture of dierentiated asthmatic epithelium many weeks after
some permanent changes to airway wall morphology. These removal from the patients airways [44, 45] (Fig. 7.1).
epithelialmesenchymal interactions have invited parallels Since tight junctions are crucial in controlling para-
with the intercellular communication found during branch- cellular transport, their disruption in asthma could help to
ing morphogenesis, and remodeling has been suggested to explain why inhaled allergens and other inhaled environ-
reect a reactivation of these early life processes (discussed mental agents initiate and maintain a chronic inammatory
later). response at this site. Epithelial expression of the epidermal
In contrast to asthma, COPD is thought to be due growth factor receptor (EGFR) family is also increased
almost exclusively to the toxic eects of tobacco smoke. in asthma, and reects an injury and repair response seen
Repetitive attempts by the epithelium to protect itself and in other epithelial tissues by interacting with six distinct
repair the injury induced by this noxious agent leads to ligands:
marked structural changes to the epithelium with thickening
and squamous metaplasia accompanied by an enhanced epidermal growth factor (EGF)
mesenchymal response at some sites and alveolar destruc- transforming growth factor (TGF-)
tion at others. The airway obstruction that accompanies amphiregulin (AR)
these changes is resistant to corticosteroids and in this
respect becomes permanent or xed. heparin-binding EGF-like growth factor (HB-EGF)
The importance of airway inammation, integral and betacellulin (BTC)
intimately linked to the process of epithelial injury, is rec-
epiregulin.
ognized in both diseases [19]. In asthma, much interest has
focused on the CD4 T-lymphocyte that is skewed toward Activation of the EGFR promotes both migration
a Th2 type phenotype as the orchestrator of an immune and proliferation of epithelial cells [46], and there is in vitro
response to allergens [20]. The predominant pattern of Th2 and in vivo evidence supporting a role for this receptor in
cytokine expression results in an inammatory phenotype bronchial epithelial repair [47, 48]. However, in asthma,
where eosinophils and mast cells are prominent, but macro- increased EGFR expression does not appear to be coupled
phages, broblasts, and dendritic cells are also involved [21]. to an appropriate proliferative response by the repairing
The inammatory milieu diers considerably in COPD. epithelium [49]. This impairment may explain why EGFR
Eosinophils may play a role in a subset that has features in expression, as a marker of tissue injury, correlates positively
common with asthma and also during acute exacerbations with both asthma severity, neutrophil inux [50], and the
[22, 23], but their signicance in the majority is outweighed extent of subepithelial brosis [51]. The impaired epithelial
by the contribution of macrophages and neutrophils with repair response in asthma may be well linked to the actions
a CD8 T-cell preponderance [2426]. Indeed CD8 of the probrogenic transforming growth factor beta
T-cells are assuming increasing importance in orchestrating (TGF-) family of cytokines, which are known inhibitors
the neutrophil-mediated injury in COPD possibly linked of epithelial proliferation [52] and whose level is markedly
to chronic infection. Known perpetrators of epithelial dam- increased in asthmatic airways [53].
age in COPD include tobacco smoke toxins, viruses, and Although there are substantial changes in epithelial
bacteria, whereas in asthma house dust mite allergen [27], structure in COPD, relatively little is known of the factors
fungal and pollen enzymes [28], and toxic air pollutants driving this. Cigarette smoke results in enhanced EGFR

84
Airway Remodeling 7
(A)
Solutes

ZO-1 Actin
Occludin ZO-2
ZO-3
Claudins ZO-1

Cingulin
JAM
E-cadherin

Catenin

Cell 1 Cell 2

(B) Non-atopic normal Atopic asthma


XY

ZO-1

FIG. 7.1 Schematic representation


37.5 m 37.5 m of airway epithelial tight junction
showing their localisation at the apex
XZ of epithelial cells (A). Confocal images
of airway biopsies from atopic normal
ZO-1 and asthmatic subjects stained by
immunoflurescence for the tight junction
Nuclei protein ZO-1. Note the disrupted staining
25 m 25 m pattern in asthma (B).

expression in airways epithelium [54] and augmented basic broblast growth factor (FGF1)
expression of both EGF and TGF- have been observed in acidic broblast growth factor (FGF2)
subjects with chronic bronchitis [53]. Thus in both asthma
and COPD, there occurs a potential imbalance between insulin-like growth factor (IGF)
proliferative and anti-proliferative signaling in the air- platelet-derived growth factor (PDGF)
ways in the former leading to a frustrated and incomplete
response and in the latter an exuberant response. transforming growth factor (TGF-)
The relationship between epithelial injury and brosis Endothelin-1 (ET-1) [55].
has been examined using co-cultures of bronchial epithelial
cells and broblasts. In these studies, damage inicted on In addition to these, a range of other potentially
the epithelial cells that resulted in enhanced myobroblast important epithelial-derived products such as nerve growth
proliferation and collagen gene expression secondary to the factor (NGF), nitric oxide, metalloproteinases, bronectin,
release of a number of growth factors including (an extracellular matrix component with potent chemotactic

85
Asthma and COPD: Basic Mechanisms and Clinical Management

properties for broblasts [56]) and a range of proinamma- bronectin has been observed to cause selective recruitment
tory cytokines [5760] are produced by epithelial cells dur- of myobroblasts from a population of normal fetal lung
ing the repair process. broblasts [70]. In COPD, studies investigating the poten-
tial role of myobroblasts suggest an increase in peripheral
Remodeling of the extracellular matrix airways that show the greatest tendency to remodel [71].

The extracellular matrix is composed of a network of brous The role of growth factors and
and structural proteins embedded in a hydrated polysaccha-
ride gel to form the strong, resilient framework of the air- inflammatory mediators
way wall. Alteration in both the mass and the composition
of this structure is one of the primary aspects of remodeling The use of transgenic animal models of mediators over-
in both asthma and COPD, with enhanced broblast activ- expression has provided further evidence to link epithelial
ity being most directly responsible. growth factor and inammatory cytokine generation with
The repair environment in the airways produces a matrix remodeling. Specically, TGF-, over-expressed
number of changes in cytokine and growth factor expression in the rat airway epithelium by adenoviral gene transfer
to which the broblast is receptive. Studies in asthma have results in severe interstitial and pleural brosis [72], while
indicated that myobroblasts, a cell population with morpho- over-expression of TGF-, IL-6, or IL-11 as transgenes in
logical and biochemical features intermediate between those the airway epithelium induces subepithelial brosis in the
of broblasts and smooth muscle cells [61], are responsible lungs of mouse models [73, 74]. Mouse models expressing
for the particular pattern of matrix deposition in this disease. the Th2 cytokines IL-5 [75], IL-9 [76], and IL-13 [77] all
These cells, lying below and adjacent to the lamina reticularis developed remodeling responses in their airways, although
in a layer referred to as the attenuated broblast sheath are interestingly in mice over-expressing IL-4 the results are
major producers of collagen and proteoglycans implicated in inconsistent [78, 79]. Interleukin-13 is over-expressed in
matrix accumulation, as well as wound contraction [62]. In asthmatic airways [80] and is being pursued as an impor-
the absence of specic immunohistochemical markers for this tant new therapeutic target in asthma because of its involve-
cell type, myobroblasts are identied by ultrastructural cri- ment in B-cell IgE isotype switching, Th2 inammation,
teria and the expression of -smooth muscle actin (-SMA) mucous metaplasia and remodeling eects [81]. The TGF-
which is also found in abundance in smooth muscle cells. superfamily of cytokines (that include the bone morpho-
Though myobroblast hyperplasia has been demonstrated in genic proteins (BMPs)) are among the most studied with
the airway subepithelium in asthma [61, 63], with their num- respect to airway brosis and remodeling. TGF- is a mul-
bers correlating with the extent of subepithelial collagen thick- tifunctional probrotic growth factor and a key component
ness [63, 64], the signicance of this nding in relation to the in the regulation of tissue growth and dierentiation dur-
remodeling response involving the full thickness of the airway ing branching morphogenesis and wound repair as well as
wall is uncertain, however, their increased number is highly a promoting smooth muscle dierentiation [82]. It stimulates
characteristic of asthma. Myobroblasts (or bromyocyte as broblast growth and is responsible for the dierentiation
they have also been called) appear early after allergen chal- of broblasts into myobroblasts [55, 83, 84]. TGF- pro-
lenge, perhaps implying a quiescent precursor cell that dier- motes synthesis of extracellular matrix components by a
entiates to acquire myobroblast features [61]. Candidates for number of cells including broblasts, smooth muscle, epi-
this include broblasts, smooth muscle cells, or perhaps other thelial cells, and macrophages [85], and it blocks matrix
primitive mesenchymal or structural cells. Recent interest in degradation by inhibiting proteolytic enzyme synthesis and
the recruitment of CD32 bone marrow-derived brocytes augmenting the action of protease inhibitors with increased
from the circulation into the airways following allergen chal- expression both in asthma and COPD [53, 86, 87]. Tissue
lenge is another possible source of these cells [65]. A further macrophage TGF- production is elevated in both dis-
source of myobroblasts is through epithelialmesenchymal eases, while in asthma eosinophils and epithelial cells are
transition involving TGF- as described in pulmonary bro- also important sources [53, 88, 89], a signicant correla-
sis [66]. A number of factors have been identied which alter tion being demonstrated between the level of expression of
broblast -SMA expression: TGF- and the extent of subepithelial brosis and numbers
of broblasts [53]. TGF- levels in asthmatic airways are
TGF- [67] unaected by corticosteroid treatment.
Another growth factor cluster is platelet-derived
Heparin [68]
growth factor (PDGF). This is produced mainly in the air-
Interferon- ways by macrophages, epithelium, and most airway inam-
granulocyte macrophage colony stimulating factor matory cell types including mast cells and eosinophils
(GM-CSF). as important sources of in asthma [90, 91]. PDGF pro-
motes broblast chemotaxis, brosis, and smooth muscle
Proliferation and migration of resident progenitors mitogenesis [92] and would seem to be another very plau-
and/or myobroblasts already present in the airways may sible mediator of airway remodeling with asthmatic bron-
contribute to the increase in these cells beneath the epi- chial broblasts showing enhanced responsiveness [93].
thelium [63]. For example, smooth muscle mitogens and Nevertheless, it has been dicult to show increased expres-
broblast growth factors have been detected in bronchoal- sion of PDGF in the airways either in asthma or in COPD
veolar lavage (BAL) uid after allergen challenge [69], and [94, 95], so that its relevance in these diseases has yet to be

86
Airway Remodeling 7
clearly demonstrated. A family of probrotic mediators are muscle mitogen, is dependent on MMP3-induced cleav-
the endothelins (1 and 2) whose levels are increased in both age of its transmembrane precursor [42]. Mast cell tryptase,
diseases [9698]. These agents, in addition to their well acting via cellular protease activated receptor (PAR)2 acts
described vasoconstrictor and bronchoconstrictor proper- directly on broblasts and smooth muscle cells to promote
ties, the endothelins are potent activators of broblasts for both mitogenesis and collagen secretion [112] and is also
enhanced matrix production [99]. mitogenic for epithelialial cells and enhances probrogenic
Although in asthma epithelial EGFR expression has growth factor release [113].
been shown to correlate with the extent of subepithelial
brosis, the EGFR as an eector of matrix remodeling mes-
enchymal cells is also important, along with platelet-derived Effects of the altered matrix on remodeling
growth factor (PDGF), insulin-like growth factor, brob-
last growth factors and TGF-. Stimulation of broblast- The capacity of the extracellular matrix to directly and indi-
bound EGF and PDGF receptors works in concert with rectly inuence cell migration, proliferation, and matura-
binding of 1 integrins also present on the surface of these tion is indicative of a highly dynamic function in addition
cells by matrix components such as laminin or bronec- to provide structural support [85, 114]. Matrix components
tin to stimulate broblast chemotaxis and migration [100, such as decorin, versican, and bronectin provide stimuli
101]. Thus, alterations in growth factor and receptor expres- for inammatory, epithelial, and stromal cells by serving
sion, matrix composition, and adhesion molecule binding all as ligands for adhesion molecules [115] and by acting as a
have the potential to inuence broblast activity in the air- reservoir for release of cytokines, chemokines, and growth
way repair response. Although EGF immunoreactivity has factors such as TGF- [116]. Glycosaminoglycans, such as
been demonstrated in the airway mucosa of both asthmatic hyaluronic acid (HA), found to be increased in the BAL
and COPD patients, the number of EGF-expressing cells uid of asthmatics [117], facilitate cell migration, and pro-
did not correlate with either basement membrane thickness liferation during injury and repair, [118] by interacting with
or underlying broblast number [53]. Increased epithelial receptors such as CD44 [119], while survival of eosinophils
expression of TGF-, another ligand of the EGF family is prolonged by interaction with bronectin and laminin,
results in lung brosis, but the eect is mediated via the epi- which inhibit apoptosis in part through the autocrine eect
thelium to enhance other broblast growth factor secretion of GM-CSF [120]. Indeed, bronectin is a matrix glyco-
rather than through a direct action on broblasts [46]. protein that has received considerable attention regarding
its role in repair [85, 121]. It is produced by bronchial epi-
thelial cells [122] broblasts, smooth muscle cells, and mac-
The role of proteolytic enzymes rophages [123] and is upregulated by growth factors such
as TGF- [124] and integrin ligation [125]. Fibronectin is
Alteration in matrix turnover is a critical factor in the incorporated into the provisional matrix that forms after
remodeling process involving a range of proteolytic injury, where it acts as a chemoattractant for both epithelial
enzymes. Serine proteases and matrix metalloproteases that cells and broblasts [126]. Elevated quantities of bronectin
are produced by a variety of inammatory and stromal cells have been detected in bronchial lavage uid in COPD and
can digest all the major components of the extracellular in asthma where it is also found abnormally deposited in
matrix [102]. Examples include MMP3 (stromolysin) and the subbasement membrane along with tenascin C, another
MMP9 in asthma [103] [104] and MMP1, MMP9, and matrix glycoprotein that is chemotactic for leukocytes and
MMP12 in COPD (with regard to their potential to induce repair cells [63, 127, 128].
emphysema) [105, 106]. In emphysema it is believed that a
protease/anti-protease imbalance exists favoring the exces-
sive proteolytic digestion of lung parenchyma and especially
elastin, although there is now strong evidence that deposi- RESULTS OF MATRIX REMODELING IN
tion of new matrix is an equally important component of
COPD involving the more peripheral airways and contrib-
ASTHMA AND COPD
uting to increased airway stiness [107]. Thus, although
both MMP9 and neutrophil elastase levels are elevated in The pattern and distribution of matrix deposition in the
induced sputum of asthmatic and COPD patients, the aug- remodeled airway diers between asthma and COPD.
mented presence of these proteases is outweighed by a pro- Interest in asthma has centered mainly on the larger air-
portionally larger increase in their natural inhibitors TIMP1 ways, whereas in COPD it is involvement of small air-
and 1-antitrypsin [108, 109] which would favor brosis. ways where remodeling dominates the pathology. Matrix
The release of growth factors that are encrypted in remodeling may be roughly subdivided into thickening of
the extracellular matrix or from their cell membrane-bound the subepithelial basement membrane (lamina reticularis or
precursors is another way that proteases may contribute to basal lamina), submucosal thickening, matrix deposition in
remodeling. For example, both FGF and TGF- are bound airway smooth muscle and new matrix deposited in the air-
as inactive forms to heparan sulfate and decorin respec- way adventitia outside the smooth muscle layer. In addition,
tively [110, 111] and can be released from these matrix the proliferation of new blood vessels, nerves, and muscle as
stores by the proteolytic activity of plasmin and MMPs. well as mucous metaplasia and formation of larger and more
In contrast, release of heparin-binding EGF (hb-EGF), a numerous submucous glands are all part of the remodeling
further member of the EGF family and a potent smooth response.

87
Asthma and COPD: Basic Mechanisms and Clinical Management

Subepithelial basement membrane matrix MUCOUS METAPLASIA


deposition in asthma and COPD
In normal lung, submucosal mucous glands and epithe-
Deposition of protein beneath the true epithelial basement
lial goblet cells are distributed throughout the cartilagen-
membrane is characteristic of asthma. Electron microscopic
ous airways in. In asthma and COPD, epithelial mucous
and immunohistochemical analysis of bronchial biopsy
metaplasia and hyperplasia occur with hypertrophy of the
specimens has shown that the true basement membrane,
submucosal gland mass. An increase in both the number
made up of the lamina rara and lamina densa, is normal in
and the size of mucus-secreting cells leads to enlargement
both size and composition and that the changes occur in
of these tracheobronchial glands in both diseases [146,
the lamina reticularis. In this region, abnormal deposition
147]; however, a variable degree of replacement of serous
of interstitial repair collagen subtypes I, III, and V takes
with mucous acini occurs in COPD, but not in asthma
place [129] in addition to non-collagenous matrix compo-
[148].
nents that include bronectin [129], laminin 2 [130], and
Epithelial mucous cell metaplasia is observed in
tenascin C [131]. Classical epithelial-derived membrane
both central and peripheral airways in asthma [149, 150]
subtypes, such as collagens IV and VII, are absent from
but there exists a substantial degree of individual vari-
this abnormal subepithelial matrix layer. Recent studies
ation. In those who have died due to asthma goblet cells
in asthma using the blocking anti-IL-5 monoclonal anti-
are found in the peripheral airways [151], compatible with
body, mepolizumab, have shown that depletion of IL-5 and
most asthma deaths occurring in association with exces-
partial depletion of airway eosinophils results in reduced
sive mucous occlusion of airways [152]. In COPD, mucous
immunostaining for tenascin C, lumican, and collagen III
metaplasia and hyperplasia are observed both centrally and
in the lamina reticularis [132], but, interestingly, this is not
peripherally [2, 153, 154] resulting in a more even distri-
paralleled by remission of asthma [133]. Similar eects on
bution of secretary cells throughout the airways. Thus, the
the matrix component of the late phase allergen response
smaller (400 m diameter) airways which are normally
have also been reported [134]. It is possible that more pro-
populated with very few goblet cells become important
longed anti-IL-5 treatment in more severe asthma might be
contributors to the excess mucous which characterizes this
more clinically eective since mepolizumab is highly eec-
disease [155, 156]. Indeed, the mucus produced in asthma
tive in eosinophilic oesophagitis and nasal polyposis [135].
and COPD is qualitatively and quantitatively abnormal
Studies of COPD have not revealed an equivalent to
with major alterations in its cellular and molecular compo-
the subepithelial brosis as observed in asthma, although in
sition. The elevated ratio of mucous/serous acini in COPD
post-transplant obliterative bronchiolitis, this change occurs
results in secretion of a more gel-like thicker mucus, which
secondary to epithelial damage, epithelial-mesenchymal
is also lacking in anti-proteases [157]. In severe asthma,
transition, and proliferation of myobroblasts [136, 137].
bronchial obstruction from mucus plugging occurs in both
Studies of COPD show reveal normal basement membrane
central and peripheral airways [151, 158]. In COPD, par-
thickness [37, 138], although in a subset of patients with
tial or complete occlusion of the small (2 mm diameter)
COPD who display overlap with the asthmatic phenotype
airways with mucous plugs is common [5]. In both diseases,
thickening of the lamina reticularis is apparent especially in
reduced levels of surfactant lining the small airways results
the presence of eosinophils [139]. These individuals, who
in increased surface tension, airway collapse, and diculty
have a BAL eosinophilia and signicant corticosteroid
in re-expansion [159].
reversibility, also show demonstrable basement membrane
Expression of mucin genes, which encode the mucin
thickening, but the composition remains unknown [140].
glycoproteins, is the principal factor governing the dif-
Further studies are needed to properly dene the clinical
ferentiation of epithelial cells into goblet cells [160]. Both
and pathological characteristics of this subgroup and any
environmental and host factors, acting on the epithelium,
overlap with late onset intrinsic asthma.
have been shown to stimulate mucin gene upregulation and
mucin secretion. Environmental factors include infectious
Diffuse matrix deposition in asthma agents [161] and environmental pollutants [162] while
and COPD host factors include inammatory mediators [163] and cell
degranulation products [164, 165]. Acrolein, a low molecu-
Studies evaluating matrix deposition deep to the lamina lar weight component of cigarette smoke, induces epithelial
reticularis in asthma have been few. An excess of collagen MUC5AC gene expression, and mucous metaplasia in rats
including types III and V have been found in large airway in vivo [166] ,while the Th2 cytokines IL-4, IL-9, and IL-13
samples by some investigators [141, 142], but not by others have been closely linked to both augmented mucin gene
[129, 143]. Other matrix proteins and proteoglycans found expression, particularly the MUC5AC and MUC2 genes,
in excess in this region in asthma include decorin, lumican, and goblet cell dierentiation both in vitro and in vivo
biglycan, versican, and bronectin [108]. Excess matrix dep- [167170].
osition in COPD has been identied predominantly in the A central role has been postulated for the neutrophil
peripheral, noncartilaginous airways (2 mm diameter) [39, in stimulation of airway mucin production. This is based
144]. However, one immunohistochemical investigation has on the observation of augmented MUC5AC expression
demonstrated reduced decorin and biglycan in the periph- by cultured epithelium when exposed to neutrophils [171],
eral airways, with staining patterns for type IV collagen and possibly through neutrophil elastase and oxidative stress,
laminin similar to those observed in control lungs [145]. each of which has been shown to augment both epithelial

88
Airway Remodeling 7
MUC5AC mRNA and protein expression [171174]. The An extensive list of smooth muscle mitogens has been
EGFR is involved in regulation of airway mucin synthesis identied:
[175, 176] as mucin gene expression results from ligand- inammatory mediators
independent transactivation of the EGFR in response to
oxidative stress [177] that can be blocked by selective inhib- growth factors
itors of the EGFR tyrosine kinase [131]. Both EGF and enzymes
EGFR expression are elevated in bronchial glandular tissue
in asthmatic subjects compared with controls [178] sug- components of the extracellular matrix [192].
gesting a similar role for the EGFR in asthma. IL-13 may Group 1 are mediators, such as EGF and PDGF, which
cause induction of mucin gene expression via an EGFR- activate tyrosine kinase receptors. Group 2 includes those
dependent pathway. receptors coupled to GTP-binding proteins such as thrombin
and mast cell tryptase [193]. Although ET-1 and LTD4 are
smooth muscle mitogens in animal models, their eects on
human smooth is controversial, although recent studies sug-
SMOOTH MUSCLE REMODELING gest an interaction with tyrosine kinase receptors possibly
through the activation of metalloproteases and the mobiliza-
tion of growth factors such as EGF ligands from their cell-
The presence of an augmented airway smooth muscle bound precursors [193, 194]. TGF- and PDGF isoforms are
bulk in asthma and COPD is well established. In asthma, both dierentiation factors for airway smooth muscle, but at
smooth muscle mass is increased in both large [179] and the time of writing their role in smooth muscle development
peripheral [180, 181] airways, although debate contin- in asthmatic and COPD airways remains speculative.
ues over whether hypertrophy or hyperplasia dominate. A further association between inammation and
Autopsy studies suggest the existence of two distinct pat- smooth muscle function has been suggested by the
terns. The type 1 pattern has increased muscle mass due enhanced expression of adhesion molecules such as ICAM-
to hyperplasia restricted to large central airways, while in 1 and VCAM-1 on airway smooth muscle cells in response
type 2 there is smooth muscle thickening throughout the to TNF-, IL-1, LPS, and IFNs [195]. Furthermore,
bronchial tree caused predominantly by hypertrophy, par- the adherence of T-cells to smooth muscle cells results in
ticularly in the small airways, with a mild degree of hyper- stimulation of DNA synthesis [195]. The capacity of these
plasia in the larger airways [182]. The situation is further ASM to express adhesion molecules, synthesize ECM
complicated by the considerable degree of heterogeneity components, and release inammatory cytokines and chem-
present. An increase in smooth muscle mass is not always okines including RANTES [196], eotaxin, and IL-8 [192]
demonstrated in mild asthma [183] or in post-mortem indicates that smooth muscle remodeling may itself be part
studies of asthmatics whom have died from other causes of the inammatory process. Towards this end, there is now
[184]. Why dierent patterns of smooth muscle enlarge- good evidence that in asthma smooth muscle cells and their
ment should exist in asthma or what factors predispose associated matrix support a distinct mast cell population
some but not other asthmatics to the development of either that maybe highly relevant to the pathogenesis of hyperre-
one is unknown. Nor is it known whether smooth muscle sponsiveness [197, 198].
changes occur before, in parallel with or as a consequence In asthma and COPD, polymorphism of the dis-
of the inammation. integrin and metalloprotease 33 (ADAM33) molecule is
The peripheral airways are also the location of an strongly associated with BHR and an accelerated decline
increase in smooth muscle mass in COPD [183, 185187], in lung function over time [199, 200]. ADAM33 is local-
though it is suggested that the changes are less marked ized to airway smooth muscle and is implicated in vascu-
than those in asthma [183]. Studies often describe these logenesis through release of its soluble form [201]. There
changes under the heading of hypertrophy, but the extent to is also much interest in the factors that are chemotractant
which hyperplasia plays a role is again unclear. Estimates of for smooth muscle cells, their relationship to myobroblasts
smooth muscle enlargement in the larger airways in COPD and whether mechanisms for removal of muscle could be
have varied [188]. Some investigators have observed no therapeutically benecial. Towards this end, bronchoscopic
abnormality in smooth muscle area [179], while others thermoplasty is looking very promising as an entirely new
report up to a twofold increase in bronchial smooth muscle approach to asthma treatment [202, 203].
thickness attributable to both hyperplasia and hypertrophy
[189]. A clinical correlation with the presence of wheeze
has also been suggested [190].
The mechanisms leading to the increase in smooth
VASCULAR AND NEURAL ALTERATIONS
muscle in asthma (and COPD) are largely speculative and
include the growth promoting potential of an enriched Elevated airway-wall blood-vessel area has been demon-
plasma environment due to microvascular leakage [191]. strated in adult and childrens asthma and is greater than in
The mitogenic inuences of mediators involved in the controls or subjects with COPD [180, 183, 204]. However,
inammatory response and an intrinsic abnormality of it is unclear whether this vascular remodeling is prima-
smooth muscle itself. However, most knowledge of factors rily due to the formation of new blood vessels [205] or to
that promote smooth muscle growth has come from in vitro enlargement of the existing microvasculature. A study of
cell culture studies [192]. the membranous bronchioles of subjects with asthma and

89
Asthma and COPD: Basic Mechanisms and Clinical Management

COPD has suggested the latter [183]. Indeed fatal asthma inammation. Another possibility is that the structural
is known to be associated with dilatation of bronchial changes either precede or occur in parallel rather than
mucosal blood vessels, congestion, and wall edema [206]. sequential to inammation. While in vitro studies suggest
However, Li and Wilson [207] have discovered changes possible inhibitory eects of bronchodilator drugs such as
suggestive of new vessel formation in mild asthma, and the 2-adrenoceptor agonists and xanthines (e.g. theophyl-
microenvironment in asthma has been shown to possess the line) on smooth muscle and broblast proliferation [217,
potential for angiogenesis. 218], similar eects on these measures in vivo are far from
Mediators such as histamine, heparin, and tryptase all established.
possess angiogenic properties, while expression of vascular It has been proposed that a more eective strat-
endothelial growth factor (VEGF), a potent vascular growth egy to reduce airway smooth muscle in asthma would be
factor, is upregulated by agents such as TGF-, TNF-, and to interfere with growth factor and other receptor mecha-
TGF- that are involved in the inammatory milieu of the nisms. Suggested approaches include blocking the actions
asthmatic airway. Of particular importance is VEGF which of TGF- in an attempt to limit smooth muscle dieren-
is released from epithelial cells as well as a broad range of tiation as well as brosis. Monoclonal antibodies targeted
inammatory cells including mast cells. The pro-angiogenic to TGF- are in clinical trials in diuse brotic disorders
function of soluble ADAM33 is also relevant here. such as scleroderma, but concerns over safety have so far
There is evidence in severe asthma of increased air- prevented their exploration in asthma. The other functions
way neural networks. This is most likely the consequence of this growth factor such as the development of anti-
of nerve growth factor (and related neurotrophins) release inammatory T-regulatory cells need to be taken into
from epithelial and inammatory cells. Circulating levels account as possible alternative on target side eects [219].
of NGF relate [208] strongly to asthma severity and may Whether targeting other mechanisms such as thrombin
prove to be a useful biomarker of this more severe pheno- inhibitors, protease activated receptors themselves, endothe-
type [209]. In addition, in both asthma and COPD, the lin and its receptors and other growth factors such as PDGF
compromised epithelial barrier allows a greater exposure of are very much in the early experimental stage. So far, bron-
nerve endings to environmental stimuli. Thus, the poten- chial thermoplasty seems the most promising approach
tial for release of neurotransmitters such as the tachykinins although even with this, side eects of delayed airway
substance P and neurokinin A is increased. These agents, brosis is a concern. One exciting possibility, however, is
in addition to their eects on vascular and smooth muscle the inhibition of IL-13, a cytokine with multiple eects on
homeostasis, can contribute to local inammation with the inammation, remodeling, and mucous metaplasia. After
attraction and activation of inammatory cells. Neurogenic successfully pass in phase I safety studies, a number of
inammation is one way sensory information received human and humanized monoclonal antibodies targeting the
from the inhaled environment could contribute to ongoing IL-13 pathway are in clinical development in asthma. The
inammation, although this has been dicult to prove in clear advantage here is that blockade of this pathway may
asthma or COPD but is easily shown in animal models of inuence several interacting aspects of chronic asthma.
airway inammation [210]. Attempts are in progress to reduce mucus secretion.
While inhibiting EGFR signaling would be one route the
downstream consequences of this on epithelial tissue repair
responses would be serious, especially since epithelial repair
EFFECTS OF THERAPY in asthma is already known to be defective [220]. However,
the chloride channel Gob 5 (hCLCA1) as an IL-13 sen-
sitive mechanism that inuences the secretion of mucus,
Whether any of the current anti-inammatory strategies is a promising target for small molecular weight inhibitors
can signicantly alter the course of remodeling is unknown. [221].
Long-term corticosteroid therapy has been shown to slow A protective role for basement membrane thicken-
the annual rate of decline in lung function in adult asthmat- ing has been postulated also in restricting inammatory cell
ics [211]. However, other studies have suggested that a neg- passage to the epithelial layer above, while smooth muscle
ative relationship exists between response to treatment and hypertrophy may possibly help to maintain bronchiolar
duration of disease [212]. Added to this are reports showing caliber in severe emphysema [164]. Thus if a new anti-
persistent airow obstruction in some patients, despite both remodeling agent were to emerge, the benecial eects
oral and inhaled therapy [208]. The implication is that early would have to be weighed against the consequences of
therapy may, to a limited extent, prevent remodeling, but interfering with what may, in some ways, be a valuable
established structural change is steroid-insensitive. Several defence mechanism.
recent birth cohort studies [213, 214] in childhood asthma
have shown that inhaled corticosteroids modify asthma
symptoms while being continuously administered, but they
have little or no eect on the natural history of asthma.
Similar ndings have been reported in the large CAMP and
CONCLUDING COMMENTS
START inhaled corticosteroid intervention studies in older
asthmatic children [215, 216]. This is somewhat surpris- While airway wall remodeling undoubtedly occurs in both
ing if it is thought that airway remodeling and the increase asthma and COPD, its physiological signicance in health
in smooth muscle is the direct consequence of chronic and disease remains speculative. Of concern is the lack

90
Airway Remodeling 7
Increased susceptibility
of epithelium to damage by environmental
agents and inflammatory cell products

Epidermal growth factor

TGF-
PDGF
FGFs
IGFs
ET-1
VEGF
(Myo) fibroblasts

FIG. 7.2 Schematic representation of the


Blood vessels
epithelial mesenchymal trophic unit (EMTU)
in asthma that becomes reactivated with
the secretion of many growth factors by the
epithelium and underlying myofibroblasts
sheath.

of knowledge connecting inammation to the structural 7. Pohunek P. Eosinophilic inammation in the bronchial mucosa of chil-
changes. In asthma an overarching hypothesis is emerging dren with bronchial asthma. Eur Respir J 19(25), 1997.
that involves aberrant communication between a chroni- 8. Niewoehner DE, Kleinerman J, Rice DB. Pathologic changes in the
cally damaged epithelium and the underlying mesenchyme peripheral airways of young cigarette smokers. N Engl J Med 291(15):
75558, 1974.
reminiscent of reactivation of the epithelial mesenchymal
9. Vignola AM, Paganin F, Capieu L, Scichilone N, Bellia M, Maakel L
trophic unit (EMTU) involved in fetal branching morpho- et al. Airway remodelling assessed by sputum and high-resolution com-
genesis of the lung [222, 223] (Fig. 7.2). The inability of an puted tomography in asthma and COPD. Eur Respir J 24(6): 91017,
ecient epithelial wound healing response is responsible for 2004.
the production of a large number of growth factors impli- 10. De BJ, Scheinmann P. The use of imaging techniques for assessing
cated in brosis, vascuogenesis and new nerve formation, severe childhood asthma. J Allergy Clin Immunol 119(4): 80810, 2007.
but whether this process is also involved in smooth muscle 11. Boulet L, Belanger M, Carrier G. Airway responsiveness and
enhancement in asthma remains conjectural. The chronically bronchial-wall thickness in asthma with or without xed airow
damaged/repairing EMTU is also the source of a range of obstruction. Am J Respir Crit Care Med 152(3): 86571, 1995.
cytokines and growth factors to support chronic inamma- 12. Peat JK, Woolcock AJ, Cullen K. Rate of decline of lung function in
subjects with asthma. Eur J Respir Dis 70(3): 17179, 1987.
tion. Thus directing new therapies to increase resistance of
13. Brown PJ, Greville HW, Finucane KE. Asthma and irreversible airow
the asthmatic airways to environmental insults may create a obstruction. Thorax 39(2): 13136, 1984.
new approach to treatment. 14. Mauad T, Dolhniko M. Pathologic similarities and dierences
between asthma and chronic obstructive pulmonary disease. Curr Opin
Pulm Med 14(1): 3138, 2008.
References 15. Moreno RH, Hogg JC, Pare PD. Mechanics of airway narrowing.
Am Rev Respir Dis 133(6): 117180, 1986.
1. Huber H. The pathology of bronchial asthma. Arch Int Med 30: 16. Park JW, Hong YK, Kim CW, Kim DK, Choe KO, Hong CS. High-
689760, 1992. resolution computed tomography in patients with bronchial asthma:
2. Nagai A, West WW, Paul JL, Thurlbeck WM. The National Institutes Correlation with clinical features, pulmonary functions and bronchial
of Health Intermittent Positive-Pressure Breathing trial: Pathology hyperresponsiveness. J Investig Allergol Clin Immunol 7(3): 18692,
studies. I. Interrelationship between morphologic lesions. Am Rev 1997.
Respir Dis 132(5): 93745, 1985. 17. Shaw TJ, Wakely SL, Peebles CR, Mehta RL, Turner JM, Wilson SJ
3. James AL, Pare PD, Hogg JC. The mechanics of airway narrowing in et al. Endobronchial ultrasound to assess airway wall thickening:
asthma. Am Rev Respir Dis 139(1): 24246, 1989. Validation in vitro and in vivo. Eur Respir J 23(6): 81317, 2004.
4. Tiddens HA, Pare PD, Hogg JC, Hop WC, Lambert R, de Jongste JC. 18. Holgate ST, Holloway J, Wilson S, Bucchieri F, Puddicombe S, Davies DE.
Cartilaginous airway dimensions and airow obstruction in human Epithelial-mesenchymal communication in the pathogenesis of chronic
lungs. Am J Respir Crit Care Med 152(1): 26066, 1995. asthma. Proc Am Thorac Soc 1(2): 9398, 2004.
5. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of airway 19. Jeery PK. Structural and inammatory changes in COPD: A com-
obstruction in chronic obstructive lung disease. N Engl J Med 278(25): parison with asthma. Thorax 53(2): 12936, 1998.
135560, 1968. 20. Holgate ST, Wilson JR, Howarth PH. New insights into airway
6. Macklem PT, Thurlbeck WM, Fraser RG. Chronic obstructive disease inammation by endobronchial biopsy. Am Rev Respir Dis 145(2 Pt 2):
of small airways. Ann Intern Med 74(2): 16777, 1971. S26, 1992.

91
Asthma and COPD: Basic Mechanisms and Clinical Management

21. Djukanovic R, Roche WR, Wilson JW, Beasley CR, Twentyman OP, 43. Vignola AM, Campbell AM, Chanez P, Bousquet J, Paul-Lacoste P,
Howarth RH et al. Mucosal inammation in asthma. Am Rev Respir Michel FB et al. HLA-DR and ICAM-1 expression on bronchial
Dis 142(2): 43457, 1990. epithelial cells in asthma and chronic bronchitis. Am Rev Respir Dis
22. Saha S, Brightling CE. Eosinophilic airway inammation in COPD. 148(3): 68994, 1993.
Int J Chron Obstruct Pulmon Dis 1(1): 3947, 2006. 44. Holgate ST. Epithelium dysfunction in asthma. J Allergy Clin Immunol
23. Chanez P, Vignola AM, OShaugnessy T, Enander I, Li D, Jeery PK 120(6): 123344, 2007.
et al. Corticosteroid reversibility in COPD is related to features of 45. de Boer WI,Sharma HS,Baelemans SM,Hoogsteden HC,Lambrecht BN,
asthma. Am J Respir Crit Care Med 155(5): 152934, 1997. Braunstahl GJ. Altered expression of epithelial junctional proteins in
24. Saetta M, Di SA, Maestrelli P, Ferraresso A, Drigo R, Potena A et al. atopic asthma: Possible role in inammation. Can J Physiol Pharmacol
Activated T-lymphocytes and macrophages in bronchial mucosa of sub- 86(3): :10512, 2008.
jects with chronic bronchitis. Am Rev Respir Dis 147(2): 3016, 1993. 46. Davies DE, Polosa R, Puddicombe SM, Richter A, Holgate ST.
25. Keatings VM, Collins PD, Scott DM, Barnes PJ. Dierences in The epidermal growth factor receptor and its ligand family: Their
interleukin-8 and tumor necrosis factor-alpha in induced sputum potential role in repair and remodelling in asthma. Allergy 54(8): 771
from patients with chronic obstructive pulmonary disease or asthma. 83, 1999.
Am J Respir Crit Care Med 153(2): 53034, 1996. 47. Madtes DK, Busby HK, Strandjord TP, Clark JG. Expression of trans-
26. OShaughnessy TC, Ansari TW, Barnes NC, Jeery PK. Inammation forming growth factor-alpha and epidermal growth factor receptor is
in bronchial biopsies of subjects with chronic bronchitis: Inverse rela- increased following bleomycin-induced lung injury in rats. Am J Respir
tionship of CD8 T lymphocytes with FEV1. Am J Respir Crit Care Cell Mol Biol 11(5): 54051, 1994.
Med 155(3): 85257, 1997. 48. Van Winkle LS, Isaac JM, Plopper CG. Distribution of epidermal
27. Herbert CA, King CM, Ring PC, Holgate ST, Stewart GA, Thompson PJ growth factor receptor and ligands during bronchiolar epithelial repair
et al. Augmentation of permeability in the bronchial epithelium by from naphthalene-induced Clara cell injury in the mouse. Am J Pathol
the house dust mite allergen Der p1. Am J Respir Cell Mol Biol 12(4): 151(2): 44359, 1997.
36978, 1995. 49. Demoly P, Simony-Lafontaine J, Chanez P, Pujol JL, Lequeux N,
28. Hassim Z, Maronese SE, Kumar RK. Injury to murine airway epithe- Michel FB et al. Cell proliferation in the bronchial mucosa of asthmat-
lial cells by pollen enzymes. Thorax 53(5): 36871, 1998. ics and chronic bronchitics. Am J Respir Crit Care Med 150(1): 21417,
29. Frigas E, Loegering DA, Gleich GJ. Cytotoxic eects of the guinea pig 1994.
eosinophil major basic protein on tracheal epithelium. Lab Invest 42(1): 50. Hamilton LM, Torres-Lozano C, Puddicombe SM, Richter A, Kimber I,
3543, 1980. Dearman RJ et al. The role of the epidermal growth factor receptor in
30. Redington AE, Polosa R, Walls AF, Howarth PH, Holgate ST. Role of sustaining neutrophil inammation in severe asthma. Clin Exp Allergy
mast cells and basophils in asthma. Chem Immunol 62: 2259, 1995. 33(2): 23340, 2003.
31. Shute JK, Parmar J, Holgate ST, Howarth PH. Urinary gly- 51. Puddicombe SM, Polosa R, Richter A, Krishna MT, Howarth PH,
cosaminoglycan levels are increased in acute severe asthmaa role for Holgate ST et al. Involvement of the epidermal growth factor receptor
eosinophil-derived gelatinase B? Int Arch Allergy Immunol 113(1-3): in epithelial repair in asthma. FASEB J 14(10): 136274, 2000.
36667, 1997. 52. Moses HL, Yang EY, Pietenpol JA. Regulation of epithelial prolifera-
32. Ohno I, Ohtani H, Nitta Y, Suzuki J, Hoshi H, Honma M et al. tion by TGF-beta. Ciba Found Symp 157: 6674, 1991.
Eosinophils as a source of matrix metalloproteinase-9 in asthmatic air- 53. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo A
way inammation. Am J Respir Cell Mol Biol 16(3): 21219, 1997. et al. Transforming growth factor-beta expression in mucosal biopsies in
33. Lackie PM. The impact of allergen on the airway epithelium. Clin Exp asthma and chronic bronchitis. Am J Respir Crit Care Med 156(2 Pt 1):
Allergy 27(12): 138386, 1997. 59199, 1997.
34. Ollerenshaw SL, Woolcock AJ. Characteristics of the inammation in 54. Barsky SH, Roth MD, Kleerup EC, Simmons M, Tashkin DP.
biopsies from large airways of subjects with asthma and subjects with Histopathologic and molecular alterations in bronchial epithelium in
chronic airow limitation. Am Rev Respir Dis 145(4 Pt 1): 92227, 1992. habitual smokers of marijuana, cocaine, and/or tobacco. J Natl Cancer
35. Laitinen LA, Heino M, Laitinen A, Kava T, Haahtela T. Damage of Inst 90(16): 1198205, 1998.
the airway epithelium and bronchial reactivity in patients with asthma. 55. Zhang S, Smartt H, Holgate ST, Roche WR. Growth factors secreted
Am Rev Respir Dis 131(4): 599606, 1985. by bronchial epithelial cells control myobroblast proliferation:
36. Jeery PK, Wardlaw AJ, Nelson FC, Collins JV, Kay AB. Bronchial An in vitro co-culture model of airway remodeling in asthma. Lab
biopsies in asthma. An ultrastructural, quantitative study and correla- Invest 79(4): 395405, 1999.
tion with hyperreactivity. Am Rev Respir Dis 140(6): 174553, 1989. 56. Shoji S, Rickard KA, Ertl RF, Robbins RA, Linder J, Rennard SI.
37. Ollerenshaw SL, Woolcock AJ. Characteristics of the inammation in Bronchial epithelial cells produce lung broblast chemotactic factor:
biopsies from large airways of subjects with asthma and subjects with Fibronectin. Am J Respir Cell Mol Biol 1(1): 1320, 1989.
chronic airow limitation. Am Rev Respir Dis 145(4 Pt 1): 92227, 1992. 57. Cromwell O, Hamid Q, Corrigan CJ, Barkans J, Meng Q, Collins PD
38. Jeery PK. Structural and inammatory changes in COPD: A com- et al. Expression and generation of interleukin-8, IL-6 and granulocyte-
parison with asthma. Thorax 53(2): 12936, 1998. macrophage colony-stimulating factor by bronchial epithelial cells
39. Cosio M, Ghezzo H, Hogg JC, Corbin R, Loveland M, Dosman J and enhancement by IL-1 beta and tumour necrosis factor-alpha.
et al. The relations between structural changes in small airways and Immunology 77(3): 33037, 1992.
pulmonary-function tests. N Engl J Med 298(23): 127781, 1978. 58. Laberge S, Ernst P, Ghaar O, Cruikshank WW, Kornfeld H, Center DM
40. Lackie PM, Baker JE, Gunthert U, Holgate ST. Expression of CD44 et al. Increased expression of interleukin-16 in bronchial mucosa of sub-
isoforms is increased in the airway epithelium of asthmatic subjects. jects with atopic asthma. Am J Respir Cell Mol Biol 17(2): 193202, 1997.
Am J Respir Cell Mol Biol 16(1): 1422, 1997. 59. Kwon OJ, Jose PJ, Robbins RA, Schall TJ, Williams TJ, Barnes PJ.
41. Peroni DG, Djukanovic R, Bradding P, Feather IH, Montefort S, Glucocorticoid inhibition of RANTES expression in human lung epi-
Howarth PH et al. Expression of CD44 and integrins in bronchial thelial cells. Am J Respir Cell Mol Biol 12(5): 48896, 1995.
mucosa of normal and mildly asthmatic subjects. Eur Respir J 9(11): 60. Aoki Y, Qiu D, Uyei A, Kao PN. Human airway epithelial cells express
223642, 1996. interleukin-2 in vitro. Am J Physiol 272(2 Pt 1): L27686, 1997.
42. Bullock GR. Expression of E-cadherin and alpha, beta and gamma 61. Gizycki MJ, Adelroth E, Rogers AV, OByrne PM, Jeery PK.
catenin is associated with epithelial shedding in human lung and nose. Myobroblast involvement in the allergen-induced late response in
J Clin Immunol 101(1 part 2): 36470, 1998. mild atopic asthma. Am J Respir Cell Mol Biol 16(6): 66473, 1997.

92
Airway Remodeling 7
62. Gabbiani G, Le LM, Bailey AJ, Bazin S, Delaunay A. Collagen 81. Izuhara K, Arima K, Kanaji S, Ohta S, Kanaji T. IL-13: A promis-
and myobroblasts of granulation tissue. A chemical, ultrastructural and ing therapeutic target for bronchial asthma. Curr Med Chem 13(19):
immunologic study. Virchows Arch B Cell Pathol 21(2): 13345, 1976. 229198, 2006.
63. Brewster CE, Howarth PH, Djukanovic R, Wilson J, Holgate ST, 82. Broide DH. Immunologic and inammatory mechanisms that drive
Roche WR. Myobroblasts and subepithelial brosis in bronchial asthma progression to remodeling. J Allergy Clin Immunol 121(3):
asthma. Am J Respir Cell Mol Biol 3(5): 50711, 1990. 56070, 2008.
64. Hoshino M, Nakamura Y, Sim J, Shimojo J, Isogai S. Bronchial subepi- 83. Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. Transforming
thelial brosis and expression of matrix metalloproteinase-9 in asthmatic growth factor-beta 1 induces alpha-smooth muscle actin expression
airway inammation. J Allergy Clin Immunol 102(5): 78388, 1998. in granulation tissue myobroblasts and in quiescent and growing
65. Schmidt M, Sun G, Stacey MA, Mori L, Mattoli S. Identication cultured broblasts. J Cell Biol 122(1): 10311, 1993.
of circulating brocytes as precursors of bronchial myobroblasts in 84. Zhang HY, Gharaee-Kermani M, Zhang K, Karmiol S, Phan SH.
asthma. J Immunol 171(1): 38089, 2003. Lung broblast alpha-smooth muscle actin expression and contractile
66. Willis BC, duBois RM, Borok Z. Epithelial origin of myobroblasts phenotype in bleomycin-induced pulmonary brosis. Am J Pathol
during brosis in the lung. Proc Am Thorac Soc 3(4): 37782, 2006. 148(2): 52737, 1996.
67. Ronnov-Jessen L, Petersen OW. Induction of alpha-smooth muscle 85. Rennard SI. Repair mechanisms in asthma. J Allergy Clin Immunol
actin by transforming growth factor-beta 1 in quiescent human breast 98(6 Pt 2): S27886, 1996.
gland broblasts. Implications for myobroblast generation in breast 86. Rennard SI. Repair mechanisms in asthma. J Allergy Clin Immunol
neoplasia. Lab Invest 68(6): 696707, 1993. 98(6 Pt 2): S27886, 1996.
68. Desmouliere A, Rubbia-Brandt L, Grau G, Gabbiani G. Heparin 87. de Boer WI, van SA, Sont JK, Sharma HS, Stolk J, Hiemstra PS
induces alpha-smooth muscle actin expression in cultured broblasts et al. Transforming growth factor beta1 and recruitment of macro-
and in granulation tissue myobroblasts. Lab Invest 67(6): 71626, 1992. phages and mast cells in airways in chronic obstructive pulmonary
69. Naureckas ET, Ndukwu IM, Halayko AJ, Maxwell C, Hershenson MB, disease. Am J Respir Crit Care Med 158(6): 195157, 1998.
Solway J. Bronchoalveolar lavage uid from asthmatic subjects is 88. Minshall EM, Leung DY, Martin RJ, Song YL, Cameron L, Ernst P
mitogenic for human airway smooth muscle. Am J Respir Crit Care et al. Eosinophil-associated TGF-beta1 mRNA expression and air-
Med 160(6): 206266, 1999. ways brosis in bronchial asthma. Am J Respir Cell Mol Biol 17(3):
70. Kawamoto M, Matsunami T, Ertl RF, Fukuda Y, Ogawa M, Spurzem JR 32633, 1997.
et al. Selective migration of alpha-smooth muscle actin-positive myo- 89. Ohno I, Nitta Y, Yamauchi K, Hoshi H, Honma M, Woolley K et al.
broblasts toward bronectin in the Boydens blindwell chamber. Clin Transforming growth factor beta 1 (TGF beta 1) gene expression by
Sci (Lond) 93(4): 35562, 1997. eosinophils in asthmatic airway inammation. Am J Respir Cell Mol
71. Hogg JC, Pierce RA. Remodelling of peripheral lung tissue in COPD. Biol 15(3): 4049, 1996.
Eur Respir J 31(5): 91314, 2008. 90. Ohno I, Nitta Y, Yamauchi K, Hoshi H, Honma M, Woolley K
72. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector- et al. Eosinophils as a potential source of platelet-derived growth
mediated gene transfer of active transforming growth factor-beta1 factor B-chain (PDGF-B) in nasal polyposis and bronchial asthma.
induces prolonged severe brosis in rat lung. J Clin Invest 100(4): Am J Respir Cell Mol Biol 13(6): 63947, 1995.
76876, 1997. 91. Taylor IK, Sorooshian M, Wangoo A, Haynes AR, Kotecha S,
73. Tang W, Geba GP, Zheng T, Ray P, Homer RJ, Kuhn C III et al. Mitchell DM et al. Platelet-derived growth factor-beta mRNA in
Targeted expression of IL-11 in the murine airway causes lymphocytic human alveolar macrophages in vivo in asthma. Eur Respir J 7(11):
inammation, bronchial remodeling, and airways obstruction. J Clin 196672, 1994.
Invest 98(12): 284553, 1996. 92. Krymskaya VP, Homan R, Eszterhas A, Ciocca V, Panettieri RA Jr.
74. DiCosmo BF, Geba GP, Picarella D, Elias JA, Rankin JA, Stripp BR TGF-beta 1 modulates EGF-stimulated phosphatidylinositol 3-
et al. Airway epithelial cell expression of interleukin-6 in transgenic kinase activity in human airway smooth muscle cells. Am J Physiol
mice. Uncoupling of airway inammation and bronchial hyperreactivity. 273(6 Pt 1): L122027, 1997.
J Clin Invest 94(5): 202835, 1994. 93. Chanez P, Vignola M, Stenger R, Vic P, Michel FB, Bousquet J.
75. Lee JJ, McGarry MP, Farmer SC, Denzler KL, Larson KA, Carrigan PE Platelet-derived growth factor in asthma. Allergy 50(11): 87883,
et al. Interleukin-5 expression in the lung epithelium of transgenic 1995.
mice leads to pulmonary changes pathognomonic of asthma. J Exp 94. Chanez P, Vignola M, Stenger R, Vic P, Michel FB, Bousquet J.
Med 185(12): 214356, 1997. Platelet-derived growth factor in asthma. Allergy 50(11): 87883,
76. Temann UA, Geba GP, Rankin JA, Flavell RA. Expression of inter- 1995.
leukin 9 in the lungs of transgenic mice causes airway inammation, 95. Aubert JD, Hayashi S, Hards J, Bai TR, Pare PD, Hogg JC. Platelet-
mast cell hyperplasia, and bronchial hyperresponsiveness. J Exp Med derived growth factor and its receptor in lungs from patients with
188(7): 130720, 1998. asthma and chronic airow obstruction. Am J Physiol 266(6 Pt 1):
77. Zhu Z, Homer RJ, Wang Z, Chen Q, Geba GP, Wang J et al. L65563, 1994.
Pulmonary expression of interleukin-13 causes inammation, mucus 96. Springall DR, Howarth PH, Counihan H, Djukanovic R, Holgate
hypersecretion, subepithelial brosis, physiologic abnormalities, and ST, Polak JM. Endothelin immunoreactivity of airway epithelium in
eotaxin production. J Clin Invest 103(6): 77988, 1999. asthmatic patients. Lancet 337(8743): 697701, 1991.
78. Jain-Vora S, Wert SE, Temann UA, Rankin JA, Whitsett JA. Interleukin-4 97. Mattoli S, Soloperto M, Marini M, Fasoli A. Levels of endothelin in
alters epithelial cell dierentiation and surfactant homeostasis in the post- the bronchoalveolar lavage uid of patients with symptomatic asthma
natal mouse lung. Am J Respir Cell Mol Biol 17(5): 54151, 1997. and reversible airow obstruction. J Allergy Clin Immunol 88(3 Pt 1):
79. Rankin JA, Picarella DE, Geba GP, Temann UA, Prasad B, DiCosmo B 37684, 1991.
et al. Phenotypic and physiologic characterization of transgenic mice 98. Chalmers GW, MacLeod KJ, Sriram S, Thomson LJ, McSharry C,
expressing interleukin 4 in the lung: Lymphocytic and eosinophilic Stack BH et al. Sputum endothelin-1 is increased in cystic bro-
inammation without airway hyperreactivity. Proc Natl Acad Sci U S A sis and chronic obstructive pulmonary disease. Eur Respir J 13(6):
93(15): 782125, 1996. 128892, 1999.
80. Saha SK, Berry MA, Parker D, Siddiqui S, Morgan A, May R et al. 99. Takuwa Y, Yanagisawa M, Takuwa N, Masaki T. Endothelin, its
Increased sputum and bronchial biopsy IL-13 expression in severe diverse biological activities and mechanisms of action. Prog Growth
asthma. J Allergy Clin Immunol 121(3): 68591, 2008. Factor Res 1(4): 195206, 1989.

93
Asthma and COPD: Basic Mechanisms and Clinical Management

100. Li J, Lin ML, Wiepz GJ, Guadarrama AG, Bertics PJ. Integrin- 120. Anwar AR, Moqbel R, Walsh GM, Kay AB, Wardlaw AJ. Adhesion
mediated migration of murine B82L broblasts is dependent on the to bronectin prolongs eosinophil survival. J Exp Med 177(3): 83943,
expression of an intact epidermal growth factor receptor. J Biol Chem 1993.
274(16): 1120919, 1999. 121. Rennard SI. Inammation and repair processes in chronic obstructive
101. Sakai T, de la Pena JM, Mosher DF. Synergism among lysophos- pulmonary disease. Am J Respir Crit Care Med 160(5 Pt 2): S1216, 1999.
phatidic acid, beta1A integrins, and epidermal growth factor or 122. Shoji S, Ertl RF, Linder J, Romberger DJ, Rennard SI. Bronchial
platelet-derived growth factor in mediation of cell migration. J Biol epithelial cells produce chemotactic activity for bronchial epithelial
Chem 274(22): 1548086, 1999. cells. Possible role for bronectin in airway repair. Am Rev Respir Dis
102. OConnor CM, FitzGerald MX. Matrix metalloproteases and lung 141(1): 21825, 1990.
disease. Thorax 49(6): 6029, 1994. 123. Vignola AM, Chanez P, Chiappara G, Merendino A, Zinnanti
103. Ohno I, Ohtani H, Nitta Y, Suzuki J, Hoshi H, Honma M et al. E, Bousquet J et al. Release of transforming growth factor-beta
Eosinophils as a source of matrix metalloproteinase-9 in asthmatic air- (TGF-beta) and bronectin by alveolar macrophages in airway dis-
way inammation. Am J Respir Cell Mol Biol 16(3): 21219, 1997. eases. Clin Exp Immunol 106(1): 11419, 1996.
104. Dahlen B, Shute J, Howarth P. Immunohistochemical localisation of 124. Ignotz RA, Massague J. Transforming growth factor-beta stimulates
the matrix metalloproteinases MMP-3 and MMP-9 within the air- the expression of bronectin and collagen and their incorporation
ways in asthma. Thorax 54(7): 59096, 1999. into the extracellular matrix. J Biol Chem 261(9): 433745, 1986.
105. Finlay GA, ODriscoll LR, Russell KJ, DArcy EM, Masterson JB, 125. Zhang Q, Sakai T, Nowlen J, Hayashi I, Fassler R, Mosher DF.
FitzGerald MX et al. Matrix metalloproteinase expression and pro- Functional beta1-integrins release the suppression of bronectin
duction by alveolar macrophages in emphysema. Am J Respir Crit Care matrix assembly by vitronectin. J Biol Chem 274(1): 36875, 1999.
Med 156(1): 24047, 1997. 126. Shoji S, Ertl RF, Linder J, Romberger DJ, Rennard SI. Bronchial
106. Yoshioka A, Betsuyaku T, Nishimura M, Miyamoto K, Kondo T, epithelial cells produce chemotactic activity for bronchial epithelial
Kawakami Y. Excessive neutrophil elastase in bronchoalveolar lav- cells. Possible role for bronectin in airway repair. Am Rev Respir Dis
age uid in subclinical emphysema. Am J Respir Crit Care Med 152 141(1): 21825, 1990.
(6 Pt 1): 212732, 1995. 127. Roche WR, Beasley R, Williams JH, Holgate ST. Subepithelial bro-
107. Kranenburg AR, Willems-Widyastuti A, Moori WJ, Sterk PJ, sis in the bronchi of asthmatics. Lancet 1(8637): 52024, 1989.
Alagappan VK, de Boer WI et al. Enhanced bronchial expression of 128. Trebaul A, Chan EK, Midwood KS. Regulation of broblast migra-
extracellular matrix proteins in chronic obstructive pulmonary disease. tion by tenascin-C. Biochem Soc Trans 35(Pt 4): 69597, 2007.
Am J Clin Pathol 126(5): 72535, 2006. 129. Roche WR, Beasley R, Williams JH, Holgate ST. Subepithelial bro-
108. Vignola AM, Riccobono L, Mirabella A, Prota M, Chanez P, Bellia V sis in the bronchi of asthmatics. Lancet 1(8637): 52024, 1989.
et al.Sputum metalloproteinase-9/tissue inhibitor of metalloproteinase-1 130. Altraja A, Laitinen A, Virtanen I, Kampe M, Simonsson BG,
ratio correlates with airow obstruction in asthma and chronic bron- Karlsson SE et al. Expression of laminins in the airways in various
chitis. Am J Respir Crit Care Med 158(6): 194550, 1998. types of asthmatic patients: A morphometric study. Am J Respir Cell
109. Vignola AM, Bonanno A, Mirabella A, Riccobono L, Mirabella F, Mol Biol 15(4): 48288, 1996.
Prota M et al. Increased levels of elastase and alpha1-antitrypsin 131. Laitinen A, Altraja A, Kampe M, Linden M, Virtanen I, Laitinen LA.
in sputum of asthmatic patients. Am J Respir Crit Care Med 157(2): Tenascin is increased in airway basement membrane of asthmatics
50511, 1998. and decreased by an inhaled steroid. Am J Respir Crit Care Med 156(3
110. Redington AE, Roche WR, Holgate ST, Howarth PH. Co-localization Pt 1): 95158, 1997.
of immunoreactive transforming growth factor-beta 1 and decorin 132. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A,
in bronchial biopsies from asthmatic and normal subjects. J Pathol Ludwig MS et al. Anti-IL-5 treatment reduces deposition of ECM
186(4): 41015, 1998. proteins in the bronchial subepithelial basement membrane of mild
111. Ornitz DM. FGFs, heparan sulfate and FGFRs: Complex interac- atopic asthmatics. J Clin Invest 112(7): 102936, 2003.
tions essential for development. Bioessays 22(2): 10812, 2000. 133. Flood-Page P, Swenson C, Faiferman I, Matthews J, Williams M,
112. Cairns JA, Walls AF. Mast cell tryptase stimulates the synthe- Brannick L et al. A study to evaluate safety and ecacy of mepoli-
sis of type I collagen in human lung broblasts. J Clin Invest 99(6): zumab in patients with moderate persistent asthma. Am J Respir Crit
131321, 1997. Care Med 176(11): 106271, 2007.
113. Cairns JA, Walls AF. Mast cell tryptase is a mitogen for epithe- 134. Phipps S, Flood-Page P, Menzies-Gow A, Ong YE, Kay AB.
lial cells. Stimulation of IL-8 production and intercellular adhesion Intravenous anti-IL-5 monoclonal antibody reduces eosinophils and
molecule-1 expression. J Immunol 156(1): 27583, 1996. tenascin deposition in allergen-challenged human atopic skin. J Invest
114. Raghow R. The role of extracellular matrix in postinammatory Dermatol 122(6): 140612, 2004.
wound healing and brosis. FASEB J 8(11): 82331, 1994. 135. Stein ML, Collins MH, Villanueva JM, Kushner JP, Putnam PE,
115. Miyake K, Underhill CB, Lesley J, Kincade PW. Hyaluronate can Buckmeier BK et al. Anti-IL-5 (mepolizumab) therapy for eosi-
function as a cell adhesion molecule and CD44 participates in hyalur- nophilic esophagitis. J Allergy Clin Immunol 118(6): 131219, 2006.
onate recognition. J Exp Med 172(1): 6975, 1990. 136. Ward C, De SA, Fisher AJ, Pritchard G, Forrest I, Corris P.
116. Redington AE, Roche WR, Holgate ST, Howarth PH. Co-localization A descriptive study of small airway reticular basement membrane
of immunoreactive transforming growth factor-beta 1 and decorin thickening in clinically stable lung transplant recipients. J Heart Lung
in bronchial biopsies from asthmatic and normal subjects. J Pathol Transplant 24(5): 53337, 2005.
186(4): 41015, 1998. 137. Sato M, Liu M, Anraku M, Ogura T, DCruz G, Alman BA et al.
117. Bousquet J, Chanez P, Lacoste JY, Enander I, Venge P, Peterson C Allograft airway brosis in the pulmonary milieu: A disorder of tissue
et al. Indirect evidence of bronchial inammation assessed by titra- remodeling. Am J Transplant 8(3): 51728, 2008.
tion of inammatory mediators in BAL uid of patients with asthma. 138. Saetta M, Di SA, Maestrelli P, Ferraresso A, Drigo R, Potena A
J Allergy Clin Immunol 88(4): 64960, 1991. et al. Activated T-lymphocytes and macrophages in bronchial mucosa
118. Hamann KJ, Dowling TL, Neeley SP, Grant JA, Le AR. Hyaluronic of subjects with chronic bronchitis. Am Rev Respir Dis 147(2): 3016,
acid enhances cell proliferation during eosinopoiesis through the 1993.
CD44 surface antigen. J Immunol 154(8): 407380, 1995. 139. Jeery PK. Remodeling and inammation of bronchi in asthma
119. Rothenberg ME. CD44 A sticky target for asthma. J Clin Invest and chronic obstructive pulmonary disease. Proc Am Thorac Soc 1(3):
111(10): 146062, 2003. 17683, 2004.

94
Airway Remodeling 7
140. Chanez P, Vignola AM, OShaugnessy T, Enander I, Li D, Jeery PK 159. Nagai A, West WW, Thurlbeck WM. The National Institutes of
et al. Corticosteroid reversibility in COPD is related to features of Health Intermittent Positive-Pressure Breathing trial: Pathology
asthma. Am J Respir Crit Care Med 155(5): 152934, 1997. studies. II. Correlation between morphologic ndings, clinical nd-
141. Wilson JW, Li X. The measurement of reticular basement membrane ings, and evidence of expiratory air-ow obstruction. Am Rev Respir
and submucosal collagen in the asthmatic airway. Clin Exp Allergy Dis 132(5): 94653, 1985.
27(4): 36371, 1997. 160. Rogers DF. Airway goblet cells: Responsive and adaptable front-line
142. Chu HW, Halliday JL, Martin RJ, Leung DY, Szeer SJ, Wenzel SE. defenders. Eur Respir J 7(9): 1690706, 1994.
Collagen deposition in large airways may not dierentiate severe 161. Li JD, Feng W, Gallup M, Kim JH, Gum J, Kim Y et al.
asthma from milder forms of the disease. Am J Respir Crit Care Med Activation of NF-kappaB via a Src-dependent Ras-MAPK-pp90rsk
158(6): 193644, 1998. pathway is required for Pseudomonas aeruginosa-induced mucin
143. Godfrey RW, Lorimer S, Majumdar S, Adelroth E, Johnston PW, overproduction in epithelial cells. Proc Natl Acad Sci U S A 95(10):
Rogers AV et al. Airway and lung elastic bre is not reduced in 571823, 1998.
asthma nor in asthmatics following corticosteroid treatment. Eur 162. Borchers MT, Wert SE, Leikauf GD. Acrolein-induced MUC5ac
Respir J 8(6): 92227, 1995. expression in rat airways. Am J Physiol 274(4 Pt 1): L57381, 1998.
144. Adesina AM, Vallyathan V, McQuillen EN, Weaver SO, Craighead JE. 163. Howarth PH, Babu KS, Arshad HS, Lau L, Buckley M, McConnell W
Bronchiolar inammation and brosis associated with smoking. A et al. Tumour necrosis factor (TNFalpha) as a novel therapeutic tar-
morphologic cross-sectional population analysis. Am Rev Respir Dis get in symptomatic corticosteroid dependent asthma. Thorax 60(12):
143(1): 14449, 1991. 101218, 2005.
145. van Straaten JF, Coers W, Noordhoek JA, Huitema S, Flipsen JT, 164. Breuer R, Christensen TG, Niles RM, Stone PJ, Snider GL. Human
Kauman HF et al. Proteoglycan changes in the extracellular matrix neutrophil elastase causes glycoconjugate release from the epithelial
of lung tissue from patients with pulmonary emphysema. Mod Pathol cell surface of hamster trachea in organ culture. Am Rev Respir Dis
12(7): 697705, 1999. 139(3): 77982, 1989.
146. Mitchell RS, Stanford RE, Johnson JM, Silvers GW, Dart G, George MS. 165. Kim KC, Wasano K, Niles RM, Schuster JE, Stone PJ, Brody JS.
The morphologic features of the bronchi, bronchioles, and alveoli in Human neutrophil elastase releases cell surface mucins from primary
chronic airway obstruction: A clinicopathologic study. Am Rev Respir cultures of hamster tracheal epithelial cells. Proc Natl Acad Sci U S A
Dis 114(1): 13745, 1976. 84(24): 93048, 1987.
147. Dunnill MS, Massarella GR, Anderson JA. A comparison of the 166. Borchers MT, Wert SE, Leikauf GD. Acrolein-induced MUC5ac
quantitative anatomy of the bronchi in normal subjects, in status expression in rat airways. Am J Physiol 274(4 Pt 1): L57381, 1998.
asthmaticus, in chronic bronchitis, and in emphysema. Thorax 24(2): 167. Temann UA, Prasad B, Gallup MW, Basbaum C, Ho SB, Flavell RA
17679, 1969. et al. A novel role for murine IL-4 in vivo: Induction of MUC5AC
148. Glynn AA, Michaels L. Bronchial biopsy in chronic bronchitis and gene expression and mucin hypersecretion. Am J Respir Cell Mol Biol
asthma. Istanbul Tip Fak Mecmuasi 15: 14253, 1960. 16(4): 47178, 1997.
149. Aikawa T, Shimura S, Sasaki H, Ebina M, Takishima T. Marked goblet 168. Dabbagh K, Takeyama K, Lee HM, Ueki IF, Lausier JA, Nadel JA.
cell hyperplasia with mucus accumulation in the airways of patients who IL-4 induces mucin gene expression and goblet cell metaplasia in
died of severe acute asthma attack. Chest 101(4): 91621, 1992. vitro and in vivo. J Immunol 162(10): 623337, 1999.
150. Shimura S, Andoh Y, Haraguchi M, Shirato K. Continuity of airway 169. Louahed J, Toda M, Jen J, Hamid Q, Renauld JC, Levitt RC et al.
goblet cells and intraluminal mucus in the airways of patients with Interleukin-9 upregulates mucus expression in the airways. Am J
bronchial asthma. Eur Respir J 9(7): 1395401, 1996. Respir Cell Mol Biol 22(6): 64956, 2000.
151. Aikawa T, Shimura S, Sasaki H, Ebina M, Takishima T. Marked 170. Zuhdi AM, Piazza FM, Selby DM, Letwin N, Huang L, Rose MC.
goblet cell hyperplasia with mucus accumulation in the airways Muc-5/5ac mucin messenger RNA and protein expression is a marker
of patients who died of severe acute asthma attack. Chest 101(4): of goblet cell metaplasia in murine airways. Am J Respir Cell Mol Biol
91621, 1992. 22(3): 25360, 2000.
152. Sheehan JK, Richardson PS, Fung DC, Howard M, Thornton DJ. 171. Takeyama K, Dabbagh K, Jeong SJ, Dao-Pick T, Ueki IF, Nadel JA.
Analysis of respiratory mucus glycoproteins in asthma: A detailed Oxidative stress causes mucin synthesis via transactivation of epider-
study from a patient who died in status asthmaticus. Am J Respir Cell mal growth factor receptor: Role of neutrophils. J Immunol 164(3):
Mol Biol 13(6): 74856, 1995. 154652, 2000.
153. Adesina AM, Vallyathan V, McQuillen EN, Weaver SO, Craighead JE. 172. Kim KC, Wasano K, Niles RM, Schuster JE, Stone PJ, Brody JS.
Bronchiolar inammation and brosis associated with smoking. Human neutrophil elastase releases cell surface mucins from primary
A morphologic cross-sectional population analysis. Am Rev Respir Dis cultures of hamster tracheal epithelial cells. Proc Natl Acad Sci U S A
143(1): 14449, 1991. 84(24): 93048, 1987.
154. Mitchell RS, Stanford RE, Johnson JM, Silvers GW, Dart G, 173. Voynow JA, Young LR, Wang Y, Horger T, Rose MC, Fischer BM.
George MS. The morphologic features of the bronchi, bronchioles, Neutrophil elastase increases MUC5AC mRNA and protein expression
and alveoli in chronic airway obstruction: A clinicopathologic study. in respiratory epithelial cells. Am J Physiol 276(5 Pt 1): L83543, 1999.
Am Rev Respir Dis 114(1): 13745, 1976. 174. Fischer B, Voynow J. Neutrophil elastase induces MUC5AC mes-
155. Niewoehner DE, Kleinerman J, Rice DB. Pathologic changes in the senger RNA expression by an oxidant-dependent mechanism. Chest
peripheral airways of young cigarette smokers. N Engl J Med 291(15): 117(5 Suppl 1): 317S320S, 2000.
75558, 1974. 175. Takeyama K, Dabbagh K, Lee HM, Agusti C, Lausier JA, Ueki IF
156. Cosio MG, Hale KA, Niewoehner DE. Morphologic and morpho- et al. Epidermal growth factor system regulates mucin production in
metric eects of prolonged cigarette smoking on the small airways. airways. Proc Natl Acad Sci U S A 96(6): 308186, 1999.
Am Rev Respir Dis 122(2): 22165, 1980. 176. Lee HM, Takeyama K, Dabbagh K, Lausier JA, Ueki IF, Nadel JA.
157. Kramps JA, Franken C, Meijer CJ, Dijkman JH. Localization of low Agarose plug instillation causes goblet cell metaplasia by activat-
molecular weight protease inhibitor in serous secretory cells of the ing EGF receptors in rat airways. Am J Physiol Lung Cell Mol Physiol
respiratory tract. J Histochem Cytochem 29(6): 71219, 1981. 278(1): L18592, 2000.
158. Kim KC, McCracken K, Lee BC, Shin CY, Jo MJ, Lee CJ et al. 177. Goldkorn T, Balaban N, Matsukuma K, Chea V, Gould R, Last J et al.
Airway goblet cell mucin: Its structure and regulation of secretion. EGF-Receptor phosphorylation and signaling are targeted by H2O2
Eur Respir J 10(11): 264449, 1997. redox stress. Am J Respir Cell Mol Biol 19(5): 78698, 1998.

95
Asthma and COPD: Basic Mechanisms and Clinical Management

178. Amishima M, Munakata M, Nasuhara Y, Sato A, Takahashi T, associated with accelerated lung function decline in asthma. Clin Exp
Homma Y et al. Expression of epidermal growth factor and epidermal Allergy 34(5): 75760, 2004.
growth factor receptor immunoreactivity in the asthmatic human air- 200. Gosman MM, Boezen HM, van Diemen CC, Snoeck-Stroband JB,
way. Am J Respir Crit Care Med 157(6 Pt 1): 190712, 1998. Lapperre TS, Hiemstra PS et al. A disintegrin and metalloprotease 33
179. Dunnill MS, Massarella GR, Anderson JA. A comparison of the quan- and chronic obstructive pulmonary disease pathophysiology. Thorax
titative anatomy of the bronchi in normal subjects, in status asthmaticus, 62(3): 24247, 2007.
in chronic bronchitis, and in emphysema. Thorax 24(2): 17679, 1969. 201. Puxeddu I, Pang YY, Harvey A, Haitchi HM, Nicholas B, Yoshisue H
180. Saetta M, Di SA, Rosina C, Thiene G, Fabbri LM. Quantitative et al. The soluble form of a disintegrin and metalloprotease 33 pro-
structural analysis of peripheral airways and arteries in sudden fatal motes angiogenesis: Implications for airway remodeling in asthma.
asthma. Am Rev Respir Dis 143(1): 13843, 1991. J Allergy Clin Immunol, 2008.
181. Carroll N, Elliot J, Morton A, James A. The structure of large and 202. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC
small airways in nonfatal and fatal asthma. Am Rev Respir Dis 147(2): et al. Asthma control during the year after bronchial thermoplasty. N
40510, 1993. Engl J Med 356(13): 132737, 2007.
182. Ebina M, Takahashi T, Chiba T, Motomiya M. Cellular hypertro- 203. Pavord ID, Cox G, Thomson NC, Rubin AS, Corris PA, Niven RM
phy and hyperplasia of airway smooth muscles underlying bronchial et al. Safety and ecacy of bronchial thermoplasty in symptomatic,
asthma. A 3-D morphometric study. Am Rev Respir Dis 148(3): severe asthma. Am J Respir Crit Care Med 176(12): 118591, 2007.
72026, 1993. 204. Barbato A, Turato G, Baraldo S, Bazzan E, Calabrese F, Panizzolo C
183. Kuwano K, Bosken CH, Pare PD, Bai TR, Wiggs BR, Hogg JC. et al. Epithelial damage and angiogenesis in the airways of children
Small airways dimensions in asthma and in chronic obstructive pul- with asthma. Am J Respir Crit Care Med 174(9): 97581, 2006.
monary disease. Am Rev Respir Dis 148(5): 122025, 1993. 205. Battegay EJ. Angiogenesis: Mechanistic insights, neovascular diseases,
184. Sobonya RE. Quantitative structural alterations in long-standing and therapeutic prospects. J Mol Med 73(7): 33346, 1995.
allergic asthma. Am Rev Respir Dis 130(2): 28992, 1984. 206. Dunnill MS, Massarella GR, Anderson JA. A comparison of the
185. Mitchell RS, Stanford RE, Johnson JM, Silvers GW, Dart G, George quantitative anatomy of the bronchi in normal subjects, in status
MS. The morphologic features of the bronchi, bronchioles, and alve- asthmaticus, in chronic bronchitis, and in emphysema. Thorax 24(2):
oli in chronic airway obstruction: A clinicopathologic study. Am Rev 17679, 1969.
Respir Dis 114(1): 13745, 1976. 207. Li X, Wilson JW. Increased vascularity of the bronchial mucosa in
186. Cosio MG, Hale KA, Niewoehner DE. Morphologic and morpho- mild asthma. Am J Respir Crit Care Med 156(1): 22933, 1997.
metric eects of prolonged cigarette smoking on the small airways. 208. Backman KS, Greenberger PA, Patterson R. Airways obstruction in
Am Rev Respir Dis 122(2): 22165, 1980. patients with long-term asthma consistent with irreversible asthma.
187. Hale KA, Ewing SL, Gosnell BA, Niewoehner DE. Lung disease Chest 112(5): 123440, 1997.
in long-term cigarette smokers with and without chronic air-ow 209. Olgart HC, de BF, Oster JP, Duvernelle C, Kassel O, Pauli G et al.
obstruction. Am Rev Respir Dis 130(5): 71621, 1984. Nerve growth factor levels and localisation in human asthmatic bron-
188. Thurlbeck WM. Pathology of chronic airow obstruction. Chest 97 chi. Eur Respir J 20(5): 111016, 2002.
(2 Suppl): 6S10S, 1990. 210. Groneberg DA, Quarcoo D, Frossard N, Fischer A. Neurogenic
189. Hossain S, Heard BE. Hyperplasia of bronchial muscle in chronic mechanisms in bronchial inammatory diseases. Allergy 59(11):
bronchitis. J Pathol 101(2): 17184, 1970. 113952, 2004.
190. Takizawa T, Thurlbeck WM. Muscle and mucous gland size in the 211. Grol MH, Gerritsen J, Vonk JM, Schouten JP, Koeter GH, Rijcken B
major bronchi of patients with chronic bronchitis, asthma, and asth- et al. Risk factors for growth and decline of lung function in asthmatic
matic bronchitis. Am Rev Respir Dis 104(3): 33136, 1971. individuals up to age 42 years. A 30-year follow-up study. Am J Respir
191. Shiels IA, Bowler SD, Taylor SM. Airway smooth muscle prolif- Crit Care Med 160(6): 183037, 1999.
eration in asthma: The potential of vascular leakage to contribute to 212. Agertoft L, Pedersen S. Eects of long-term treatment with an
pathogenesis. Med Hypotheses 45(1): 3740, 1995. inhaled corticosteroid on growth and pulmonary function in asth-
192. Hirst SJ. Airway smooth muscle as a target in asthma. Clin Exp matic children. Respir Med 88(5): 37381, 1994.
Allergy 30(Suppl 1): 5459, 2000. 213. Guilbert TW, Morgan WJ, Zeiger RS, Mauger DT, Boehmer SJ,
193. Panettieri RA Jr. Cellular and molecular mechanisms regulating air- Szeer SJ et al. Long-term inhaled corticosteroids in preschool chil-
way smooth muscle proliferation and cell adhesion molecule expres- dren at high risk for asthma. N Engl J Med 354(19): 198597, 2006.
sion. Am J Respir Crit Care Med 158(5 Pt 3): S13340, 1998. 214. Murray CS, Woodcock A, Langley SJ, Morris J, Custovic A.
194. Cohen MD, Ciocca V, Panettieri RA Jr. TGF-beta 1 modulates Secondary prevention of asthma by the use of Inhaled Fluticasone
human airway smooth-muscle cell proliferation induced by mitogens. propionate in Wheezy INfants (IFWIN): Double-blind, randomised,
Am J Respir Cell Mol Biol 16(1): 8590, 1997. controlled study. Lancet 368(9537): 75462, 2006.
195. Lazaar AL, Albelda SM, Pilewski JM, Brennan B, Pure E, 215. The Childhood Asthma Management Program Research Group.
Panettieri RA Jr. T lymphocytes adhere to airway smooth muscle Long-term eects of budesonide or nedocromil in children with
cells via integrins and CD44 and induce smooth muscle cell DNA asthma. N Engl J Med 343: 105463, 2000.
synthesis. J Exp Med 180(3): 80716, 1994. 216. Busse WW, Pedersen S, Pauwels RA, Tan WC, Chen YZ, Lamm CJ
196. John M, Hirst SJ, Jose PJ, Robichaud A, Berkman N, Witt C et al. et al. The Inhaled Steroid Treatment As Regular Therapy in Early
Human airway smooth muscle cells express and release RANTES in Asthma (START) study 5-year follow-up: Eectiveness of early
response to T helper 1 cytokines: Regulation by T helper 2 cytokines intervention with budesonide in mild persistent asthma. J Allergy Clin
and corticosteroids. J Immunol 158(4): 184147, 1997. Immunol 121(5): 116774, 2008.
197. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, 217. Tomlinson PR, Wilson JW, Stewart AG. Inhibition by salbutamol of
Pavord ID. Mast-cell inltration of airway smooth muscle in asthma. the proliferation of human airway smooth muscle cells grown in cul-
N Engl J Med 346(22): 1699705, 2002. ture. Br J Pharmacol 111(2): 64147, 1994.
198. Begueret H, Berger P, Vernejoux JM, Dubuisson L, Marthan R, 218. Levi-Schaer F, Touitou E. Xanthines inhibit 3T3 broblast prolif-
Tunon-de-Lara JM. Inammation of bronchial smooth muscle in eration. Skin Pharmacol 4(4): 28690, 1991.
allergic asthma. Thorax 62(1): 815, 2007. 219. Pyzik M, Piccirillo CA. TGF-beta1 modulates Foxp3 expression
199. Jongepier H, Boezen HM, Dijkstra A, Howard TD, Vonk JM, and regulatory activity in distinct CD4 T cell subsets. J Leukoc Biol
Koppelman GH et al. Polymorphisms of the ADAM33 gene are 82(2): 33546, 2007.

96
Airway Remodeling 7
220. Holgate ST. The airway epithelium is central to the pathogenesis of exposure of infant monkeys to allergen and/or ozone. Toxicol Appl
asthma. Allergol Int 57(1): 110, 2008. Pharmacol 214(3): 23743, 2006.
221. Long AJ, Sypek JP, Askew R, Fish SC, Mason LE, Williams CM 223. Holgate ST, Davies DE, Powell RM, Howarth PH, Haitchi HM,
et al. Gob-5 contributes to goblet cell hyperplasia and modulates pulmo- Holloway JW. Local genetic and environmental factors in asthma dis-
nary tissue inammation. Am J Respir Cell Mol Biol 35(3): 35765, 2006. ease pathogenesis: Chronicity and persistence mechanisms. Eur Respir
222. Joad JP, Kott KS, Bric JM, Peake JL, Plopper CG, Schelegle ES et al. J 29(4): 793803, 2007.
Structural and functional localization of airway eects from episodic

97
Asthma and COPD: Animal Models
8
CHAPTER

OVERVIEW are housed which eliminate many environmental Stephanie A. Shore1 and
modiers, and some dierences in the immune
system (mice have a more polarized T-lym- Steven D. Shapiro2
Animal models have been used extensively in phocyte response than humans). These dis- 1
Molecular and Integrative Physiological
the discovery of the biological processes that advantages have been documented elsewhere Sciences Program, Harvard School of
underlie asthma and COPD. For example, the [37] and may limit the utility of these mod- Public Health, Boston, MA, USA
demonstration by Gross et al. [1] that instilla- els for studying human disease in some cases. 2
Department of Medicine, University of
tion of papain into the lungs of rats resulted in a Mouse models of asthma and COPD will be
Pittsburgh, Pittsburgh, PA, USA
syndrome similar to emphysema, contributed to reviewed separately below.
the elastase/anti-elastase hypothesis regarding
the pathogenesis of emphysema. Animal mod-
els have also been used to guide research into
pharmacological interventions for the treatment
of asthma and COPD. For example, the current MOUSE MODELS OF ASTHMA
use of leukotriene receptor antagonists for the
treatment of asthma had its foundation partly Human asthma is characterized by intermittent
in the demonstration of the marked broncho- reversible airway obstruction, AHR, and airway
constrictor potency of cysteinyl leukotrienes in inammation. Pathologically, airway remodeling
guinea pigs [2]. While many species, including including increased airway smooth muscle mass,
dogs, cats, monkeys, horses, pigs, sheep, guinea subepithelial brosis, mucous hypersecretion,
pigs, and rats have been used to study asthma and increased airway vascularity are observed.
and COPD, most current investigations involve Asthma does not typically develop in most
the use of mice. Consequently, this review will non-human species. Consequently, many inves-
focus on models developed using this species. tigators have chosen to develop animal models
The advantages of using mice include cost of in which an asthma-like phenotype is induced
housing, a short breeding period, the availability by some intervention. The etiology of asthma is
of inbred species with known characteristics, a complex, and still poorly understood, and it is
well characterized immune system and genome, clear that multiple factors including exposure to
and the ability to either overexpress or delete allergens, exercise, exposure to air pollution, cer-
specic genes, sometimes conditionally, that is, tain viral infections, and the onset of obesity can
at specic times or in particular cells. This abil- also initiate symptoms. Because of the extensive
ity to genetically manipulate mice is a powerful nature of this topic, we have chosen to focus on
tool that permits very mechanistic experiments only two of these factors: allergen and obesity.
that cannot be accomplished in human subjects
and has yielded some important advances in our
understanding of lung disease. The disadvan- Modeling allergic asthma
tages of using mice include dierences in lung
anatomy, lung development, pattern and route Many investigators have used models of allergic
of breathing (mice are obligate nose breathers), asthma in which mice are rst sensitized to an
the pathogen-free facilities in which most mice allergen and then challenged with that allergen

99
Asthma and COPD: Basic Mechanisms and Clinical Management

via the lungs. The sensitization and the challenge protocols Murine models of allergic airways disease have been
vary widely among investigators, especially depending on used to examine the role of many cells, cytokines, chemok-
whether AHR or airway remodeling are being targeted. The ines, enzymes, and other factors in the pathology of asthma.
sensitization is usually i.p. injection of the allergen with an These studies are too numerous to document here. Instead,
adjuvant. Allergens used have included ovalbumin (OVA), we focus on three issues. One, the role of lymphocytes, can
picryl chloride, sheep erythrocytes, short ragweed extracts, be seen as a major success of these models. Two others, the
and house dust mite allergen. Although not environmen- role of IgE and mast cells, and the role of eosinophils are
tally very relevant, most investigators have used OVA as instances where the models do not appear to reproduce the
the allergen, perhaps because of its relatively low cost and human ndings.
easy availability. Mice are usually assessed 13 days after the Role of T-lymphocytes in allergen-induced airway
last of several daily airway challenges and the characteristics responses: A major accomplishment of studies using these
monitored are AHR, airway inammation, and serum IgE. murine models was in establishing a role for lymphocytes in
Consistent features of the model are increased OVA- the development of allergic asthma. For example, following
specic IgE and IgG1, eosinophilia (intraluminal, peribron- allergen sensitization and challenge, AHR does not develop
chial, and perivascular), and recruitment of lymphocytes to in athymic mice or in mice with severe combined immuno-
the airways. Increases in Th2-type cytokines either in bron- deciency, but adoptive transfer of lymphocytes from
choalveolar lavage (BAL) uid, in lung tissue, or in lymph wild-type sensitized and challenged mice restores AHR in
nodes draining the lungs also occur. These changes are gen- athymic mice [18, 19]. A particular role for CD4 T-lym-
erally similar to those observed in the airways of asthmatics phocytes was established by experiments demonstrating
after allergen challenge, although the kinetics of the recruit- that MHC Class II decient mice lacking mature CD4
ment of eosinophils and lymphocytes dier: in humans, the T-lymphocytes and mice depleted of CD4 T-cells do
peak in T-lymphocyte recruitment occurs about 24 h after not develop AHR or airway inammation following aller-
challenge and precedes the peak of eosinophil recruitment, gen sensitization and challenge [20, 21]. The requirement
whereas in mice, T-cell recruitment continues long after for CD4 T-cells was also underscored by the observation
eosinophils have peaked and returned to baseline [8]. that reconstitution of CD4 T-cells alone is sucient to
AHR is often, though not always observed in aller- restore allergen-induced AHR to RAG/ mice which lack
gen sensitized and challenged mice. There are few reports of both B- and T-lymphocytes [22]. It has since been estab-
comparisons of the various protocols in terms of their abil- lished that other T-cell subsets including -T-cells, iNKT
ity to induce AHR, but those available indicate that both cells, and regulatory T-cells may have roles in enhancing or
systemic sensitization and pulmonary challenge are required attenuating allergic airways disease in these mouse models
to induce AHR [9]. For example, multiple airway chal- [2326].
lenges without systemic sensitization can result in pulmo- A major focus of research using these models over the
nary eosinophilia, but in vivo AHR is not observed under last decade has been to establish how T-lymphocytes con-
these conditions [10]. The route of airway challenge (nasal tribute to allergic airway responses. Early on, it was estab-
or intratracheal versus aerosol) may also be a factor [11]. lished that the ability of these cells to generate cytokines
Mouse strain also has an important impact on allergen- was likely to be important. Initial work focused on IL-4,
induced AHR. AHR is relatively easy to induce in Balb/c IL-5, and IL-13 [19, 20, 22, 2731]. More recently, other
and A/J mice, but more dicult to induce in other strains, cytokines, including IL-17, have received increasing atten-
including C57BL/6 mice even though airway inammation tion [3234].
is robust in this strain [9, 1214]. The relative resistance of Role of IgE and mast cells in allergen-induced AHR:
C57BL/6 mice to allergen-induced AHR is unfortunate, Mast cells express receptors (Fc RI) on their surface that
since many of the knockout mice that could be used to bind the Fc portion of IgE with high anity. Crosslinking
study the mechanistic basis for allergen-induced AHR are Fc RI receptors upon binding of allergen to IgE results in
originally developed on this background. the secretion of a panel of mediators including histamine,
Recently, investigators have begun using repeated aller- eicosanoids, and cytokines, many of which have the capac-
gen challenges over several weeks to months to model the ity to cause bronchoconstriction and to elicit AHR, though
airway remodeling that characterizes human asthma [15]. In it is important to note that IgE independent activation of
general, such challenges often result in tolerance, although mast cells can occur in response to complement proteins,
in the A/J strain, eosinophilia and AHR are preserved over neuropeptides, and other moieties. In humans, activation
several months, and these mice develop subepithelial bro- of mast cells by IgE is important for both early and late
sis and airway smooth muscle hypertrophy [12]. However, responses to allergen inhalation: treatment with the human-
modications to the protocols may improve the utility of ized anti-IgE antibody, omalizumab, markedly reduces both
mouse models for studies of airway remodeling. A recent the rapid and delayed declines in FEV1 that occur following
report by Yu et al. [16] indicates that elimination of adjuvant inhalation of allergen [35]. An important drawback of using
during the sensitization phase results in preserved AHR, mice is that such airway obstruction is not easily observed
eosinophilia, and subepithelial brosis even after 9 weeks of [36]. Small increases in pulmonary resistance (RL) (less than
repeated weekly intranasal challenges in mice on a C57BL/6 50% increase) can be observed following systemic admin-
background. Others have observed subepithelial brosis and istration of activating anti-IgE antibodies [37], but RL
increases in airway smooth muscle mass after chronic chal- changes little after airway challenge with allergen [36]. This
lenge over several months with very low concentrations of is in marked contrast to many other species which develop
OVA delivered intranasally, even in C57BL/6 mice [17]. marked increases in RL after inhalation of allergen [3840].

100
Asthma and COPD: Animal Models 8
Mast cells and IgE also appear to play a role in the cytokines involved. Renements in these models involv-
airway inammation that occurs after allergen challenge in ing chronic challenges may ultimately allows us to use such
asthmatics [41], but the ability of mouse models to repro- models to study the airway remodeling that characterizes
duce this phenomenon has not been consistent. Kung et al. asthma, but are currently in their infancy. The protocol used
[42] reported that aerosol OVA challenge of systemically to sensitize and challenge the mice, and the choice of mouse
sensitized mast cell decient mice resulted in fewer eosi- strain are key issues and can inuence outcome.
nophils in BAL uid and lung tissue compared to congenic
controls, whereas others [10, 4345] have failed to observe
any eect of mast cell deciency or IgE deciency on the Modeling obesity and asthma
recruitment of eosinophils. At least part of this discrepancy
is likely related to the precise protocol used to sensitize and Obesity is a risk factor for asthma. Both in adults and
challenge the mice. Kobayashi et al. used two distinct pro- children, the prevalence and incidence of asthma increase
tocols to sensitize and challenge mast cell decient mice with body mass index (BMI) (see recent reviews [5558]).
and found reduced eosinophils compared to strain matched Obesity also appears to worsen asthma control and may
controls when relatively mild protocols for airway challenge increase asthma severity. In addition, both surgical and
that resulted in relatively few eosinophils were employed, diet-induced weight loss improve many asthma out-
but no dierence between mast cell decient and control comes. The ecacy of many standard asthma medications
mice when more frequent and robust airway challenges that is altered in the obese [58, 59], suggesting that additional
produced marked increases in eosinophils were used [43]. therapeutic strategies may be necessary in this population.
Role of eosinophils in allergen-induced AHR: Eosinophils Development of these strategies will require an understand-
contain cationic proteins such as major basic protein (MBP) ing of the mechanistic underpinnings of the relationship
that have the capacity to induce AHR [46] and experiments between obesity and asthma. To that end, mice have been
in mice in the late 1990s provided strong evidence that eosi- used to model the relationship between obesity and asthma.
nophils may be responsible for the AHR associated with These studies have established that obese mice exhibit a
allergic asthma. For example, in mice, transgenic overexpes- pulmonary phenotype that includes both innate AHR,
sion of IL-5, a dierentiation, chemoattractant, and survival as well as increased responses to ozone (O3) and allergen
factor for eosinophils, led to accumulations of eosinophils [6065], and suggest a role for adipokines in the obesity
and AHR even in the absence of allergen challenge [47]. asthma relationship [66, 67].
Following OVA sensitization and challenge, both accumula- Several types of obese mice have been used in these
tion of eosinophils in the airways and AHR were prevented studies (see Ref. [68] for details). Ob/ob mice are geneti-
in IL-5 decient compared to wild-type mice and recon- cally decient in leptin, a satiety hormone synthesized by
stitution of IL-5 in these knockout mice restored pulmo- adipocytes and released into blood in proportion to adipose
nary eosinophilia and AHR [48, 49]. Similar results were tissue mass. In the absence of leptin, ob/ob mice eat exces-
obtained using anti-IL-5 antibodies [50]. The observation sively, have a low resting metabolic rate, and by 8 weeks
of a good correlation between airway eosinophils and AHR of age weigh at least twice as much as wild-type controls
across multiple strains of mice, all sensitized and challenged [69]. The increased body weight is entirely the result of an
in the same way, also argued for a relationship between these increase in fat mass. Db/db mice are genetically decient in
factors [13]. Data such as those cited above were strong the long form of the leptin receptor, which is required for
enough to initiate clinical trials for anti-IL-5 in human leptins eect on satiety and metabolism [69]. Thus, db/db
asthma. However, the results of such trials have been dis- mice are similar to ob/ob mice in many respects: they over-
appointing. For example, a recent fairly large study showed eat, are hypothermic and inactive, and massively obese.
that while anti-IL-5 antibody treatment results in marked However, short forms of the leptin receptor with truncated
suppression of both blood and sputum eosinophils, it does or absent cytoplasmic domains are expressed in these mice,
not impact pulmonary function, -agonist use, symptom and are capable of some types of signaling. Hence, there can
or quality of life scores in patients with persistent asthma be subtle dierences between ob/ob and db/db mice, includ-
symptoms despite inhaled corticosteroid use [51], although ing dierences in their pulmonary phenotype [60]. Cpe fat
it has been argued that the study population (one of dicult mice are genetically decient in carboxypeptidase E (Cpe),
to control asthma) could have biased the study [52]. Did our an enzyme involved in processing of neuropeptides involved
mouse models lead us astray? There are certainly data from in eating behaviors. Obesity develops more slowly in these
mouse models arguing against a role for eosinophils in the mice than in ob/ob or db/db mice, but can be quite marked
AHR associated with allergen sensitization and challenge [69]. Dietary obesity can also be induced by feeding wean-
[19, 53, 54]. Mouse strain and the strength of the allergen ling C57BL/6 mice a diet in which 45% or 60% of calories
challenge appear to impact whether a role for eosinophils are derived from fat (predominantly in the form of lard).
are observed or not: experiments performed in Balb/c mice Diet-induced obesity (DIO) develops slowly and is much
in which milder challenges are used indicate little role for less marked than in the genetic obesities. All of these obese
eosinophils in OVA-induced AHR [5]. mice are hyperglycemic, hyperinsulinemic, and hyperlipi-
Summary: Mice are not useful for modeling the early demic to some extent [68, 69].
or late responses to allergen, but models in which mice are Innate AHR: Airway responsiveness to intrave-
sensitized and then challenged with allergen have been use- nous methacholine is increased in ob/ob, db/db, and Cpe fat
ful in establishing a role for T-lymphocytes in asthma and mice, and in mice with DIO [6065], indicating that it is
are currently being used to investigate the T-cell subsets and a common feature of murine obesity. Consistent with the

101
Asthma and COPD: Basic Mechanisms and Clinical Management

non-specic nature of human asthma, AHR is observed Responses to allergen: Some, but not all, aspects of
following both methacholine and serotonin challenges [60]. the response to allergen are augmented in obese mice. For
Both the magnitude and the duration of obesity appear to example, following OVA sensitization and challenge, splen-
play some role in the development of AHR in obese mice. ocyte proliferation, IL-2 production, and mast cell numbers
For example, AHR is more marked in ob/ob and db/db mice are increased in mice with DIO compared to lean controls
than in Cpe fat mice or mice with DIO. Mice do not exhibit [78]. Increased OVA-induced AHR is also observed in
AHR when they are raised on high-fat diets for approxi- obese mice [63]. However, this increase in airway respon-
mately 16 weeks, but AHR is observed after 30 weeks on siveness occurs in the absence of any dierences in Th2
the diet, even though body weight averages about 40% more cytokines and in the face of decreases in BAL eosinophils
than low-fat fed controls at both time points [61]. compared to lean controls, suggesting that other factors
The mechanistic basis for the AHR observed in obese must contribute. Interestingly, OVA-induced increases in
mice is not known. However, the mice do not have any overt serum IgE are also greater in ob/ob and db/db mice than
cellular inammation in their lungs [60, 63]. All measure- in lean controls. As described above, IgE and mast cells
ments were made in open-chested mice, mechanically venti- do not appear to contribute prominently in most studies
lated at a xed positive end-expiratory pressure (PEEP) and of allergen-induced airways disease in mice. However, the
a xed tidal volume, so changes in absolute or tidal volume expression of 5-lipoxygenase activating protein is substan-
likely do not play a role. In ob/ob and db/db mice, there are tially increased in the adipose tissue of ob/ob mice [79]. This
eects on lung development: despite their massive obesity, could lead to greater leukotriene synthesis and subsequent
these mice have small lungs [60, 65]. However, lung size is release from activated mast cells and thus enhance airway
not aected in Cpe fat mice and mice with DIO, but these responsiveness. In this context, Peters-Golden et al. recently
mice still exhibit AHR [61, 62]. Instead, it is likely that reported a more benecial eect of the leukotriene antago-
some aspect of the chronic low-grade systemic inamma- nist montelukast in obese versus lean asthmatics [80].
tion that characterizes the obese state [70] contributes to the Adipokines: Several hypotheses have been proposed
AHR observed in obese mice. For example, preliminary data to explain the association between obesity and asthma
in both db/db and Cpe fat mice indicate that treatment with (Fig. 8.1). For example, obesity results in changes in a
anti-TNF- antibodies reduces airway responsiveness [71]. variety of adipose tissue derived factors (adipokines) that
Responses to O3: Exposure to O3, a common air pollut- may contribute to the relationship between obesity and
ant, is a trigger for asthma. Hospital admissions for asthma asthma. Serum leptin is increased in obesity and two cross-
are higher on days of high ambient O3 concentrations [72] sectional studies indicate that leptin is also increased in
and in children, O3 increases asthmatic symptoms even at asthmatics [81, 82]. Numerous reports indicate that lep-
concentrations below the US Environmental Protection tin is pro-inammatory (see recent review [83]), and these
Agency standard [73]. O3 causes airway inammation, as inammatory eects could exacerbate asthma in the obese.
well as AHR, both of which may contribute to the ability Leptin treatment has been shown to augment allergen-
of O3 to exacerbate asthma. induced AHR in lean mice, without aecting eosinophil
Obesity impacts the eects of O3 in the lung. Both the inux or Th2 cytokine expression [66]. Indeed, rather than
changes in pulmonary function and the AHR induced by O3 modifying adaptive immunity, leptin could be acting on the
exposure are increased in obese versus lean human subjects innate immune system: exogenous administration of lep-
[74, 75]. Similar results are observed in obese mice regard- tin to lean mice increases their subsequent inammatory
less of the nature of their obesity. Acute exposure to O3 response to acute O3 exposure [65], a response character-
(2 ppm for 3 h) increases RL in obese but not lean mice and ized by release of acute phase cytokines and chemokines,
induces greater AHR in obese versus lean mice [60, 62, 65]. and dependent to some extent on toll-like receptor acti-
Compared to lean controls, O3-induced injury and inam- vation [84]. However, there must be factors in addition to
mation are also greater in ob/ob mice, db/db mice, Cpe fat
mice, and mice with DIO [6062, 65]. However, the factors
that contribute to the innate AHR of obese mice appear to
be dierent from those that contribute to their increased Mechanisms proposed to explain
response to acute O3: increased O3-induced inammation the relationship between obesity and asthma
is observed as early as 7 weeks of age in Cpe fat mice [76],
whereas AHR is not observed until they are 10 weeks old. Common etiologies Effects of obesity
At least part of the increased response to O3 observed In utero conditions on lung mechanics
in obese mice appears to derive from dierences in IL-6 Genetics FRC
Tidal volume
signaling. Following acute O3 exposure, obese mice have
increased IL-6 release into BAL uid compared to lean Co-morbidities Adipokines
mice [60, 62, 65]. Treatment with anti-IL-6 antibodies Gastroesophageal reflux Cytokines
ablates the accelerated neutrophil inux that is observed fol- Sleep disorder breathing Chemokines
lowing acute O3 exposure in ob/ob mice and also attenuates Type-II diabetes Energy regulating hormones
the enhanced O3-induced epithelial injury that is observed Hypertension Acute phase reactants
Other factors
in these mice [77]. Pulmonary expression of signal trans-
ducer and activator of transcription-1 (STAT-1) is reduced FIG. 8.1 Potential mechanisms whereby obesity may be associated
in ob/ob mice [77] and may modify pulmonary responses to with asthma. Reproduced from Ref. [68] with permission of the American
IL-6 in these mice. Physiological Society.

102
Asthma and COPD: Animal Models 8
leptin that contribute to the augmented O3 responses of fact that elastin fragments themselves [101] are chemok-
obese mice, since such responses are observed not only in ines, and that the events following PPE instillation are very
Cpe fat and DIO mice [61, 62] that have marked increases in similar to the inammatory cell cascade that one observes
serum leptin, but also in ob/ob and db/db mice [60, 65], that in COPD following cigarette smoke exposure. That is, ini-
lack either leptin or the leptin receptor. tially PPE leads to acute airspace enlargement with elastin
In contrast to other adipokines, adiponectin, an insulin- destruction and release of fragments of elastin and other
sensitizing hormone, declines in obesity [85, 86]. In contrast matrix proteins. This results in subsequent neutrophilia fol-
to leptin, adiponectin has important anti-inammatory lowed within days by a predominant macrophage and T-cell
eects in obesity [86]. In lean Balb/c mice, exogenous admin- inux. These activated inammatory cells release endog-
istration of adiponectin results in an almost complete sup- enous elastases causing progressive airspace enlargement.
pression of OVA-induced AHR, airway inammation, and Environmental factors: Cigarette smoke is clearly the
Th2 cytokine expression in the lung [67]. Adiponectin has major etiologic factor for COPD. However, a variety of other
been shown to inhibit macrophage production of lymphocyte environmental agents have been applied to determine their
chemotactic factors [87]. An adiponectin-induced reduc- capacity to induce COPD. Nitrogen dioxide and ozone cause
tion in lymphocyte inux into the lungs would be expected mild lung injury suggesting that they may be modifying envi-
to reduce pulmonary concentrations of Th2 cytokines and ronmental factors but unlikely primary environmental factors
could thus be responsible for attenuating eosinophilia and leading to COPD [90]. Cadmium chloride [102], a constitu-
AHR following OVA challenge. Three adiponectin receptors ent of cigarette smoke, primarily causes interstitial brosis
have been cloned: adipoR1, adipoR2, and T-cadherin [88, tethering open adjoining airspaces simulating emphysema.
89]. Lung tissue expresses all three adiponectin receptors. While this mechanism diers from airspace enlargement
The expression of all three receptors declines in the lungs fol- secondary to matrix destruction that characterizes emphy-
lowing allergen sensitization and challenge in mice [67], sug- sema, excess collagen deposition in the context of loss of
gesting that asthma may be a state of adiponectin resistance. elastin is a feature of human centrilobular emphysema.
Coupled with obesity-related declines in adiponectin [85, Cigarette smoke [4, 94, 95]: The great strength of ciga-
86] and additional declines in serum adiponectin that occur rette smoke-induced models of COPD is that we know
with allergen challenge [67], the data suggest that the obese that this is the causative agent for COPD. Hence, path-
asthmatic is likely to have defects in this important immuno- ways leading to phenotypic changes of COPD mirror those
modulatory pathway that augment the eects of allergen of human COPD, limited only by dierences in smoke
challenge. exposure as well as biological dierences between mice and
humans. Over the years many species of mammals have
been exposed to cigarette smoke using a variety of smok-
ing chambers. Larger animals are limited by the long (years)
MOUSE MODELS OF COPD duration of exposure required for development of COPD.
Of small animals tested, the guinea pig is the most suscep-
tible species and rat the least. Several inbred mouse strains
Modeling COPD are also susceptible to cigarette smoke-induced COPD, and
hence they have dominated the literature due to our ability
Animal models were critical to the development of the to manipulate gene expression as well as other advantages
elastase:anti-elastase hypothesis generated 45 years ago. This discussed above.
hypothesis remains the cornerstone of our understanding of Following exposure to cigarette smoke, mice develop
COPD pathogenesis today. The major nding at that time changes similar to humans including acute inammation
was that instillation of the proteinase papain into experi- with neutrophils followed by subacute increases in macro-
mental animals caused airspace enlargement, the charac- phages, and CD8  CD4 T-cells [103]. Proteinases are
teristic feature of emphysema [1]. Since then animals have released and apoptosis of structural cells is observed. These
been exposed to a variety of molecular, chemical, and envi- processes ultimately lead to airspace enlargement that is
ronmental agents that lead to airspace enlargement [90]. In easily detectable in most strains within 6 months. With
particular elastases [9193], cigarette smoke [94, 95], and respect to the airway, mice lose cilia upon cigarette smoke
more recently inducers of apoptosis have been most inform- exposure, develop small airway brosis, and also develop
ative [96, 97]. Mouse genetic mutants have also been criti- goblet cell hypertophy [4, 104].
cal in furthering our understanding of the pathogenesis of Apoptosis: Recently investigators have found that
COPD [4, 95]. exposure to agents that initiate either endothelial cell death
Elastases: Ever since the initial ndings with papain, a (via VEGFRII inhibition) [96] or epithelial cell death
variety of proteinases have been instilled into experimental (via caspase 3 nodularin, ceramide delivery, and VEGF
animals. Of note, only those with elastolytic activity develop inhibition) [97, 105, 106] lead to non-inammatory air-
emphysema, collagenases do not [91]. Of most utility space enlargement. Clearly, to lose an acinar unit, one
has been porcine pancreatic elastase (PPE) [91, 98, 99], a must destroy both extracellular matrix (ECM) and struc-
potent elastase that quickly generates airspace enlargement. tural cells. Traditionally, we believed that inammatory
This model has also been useful to test agents that have the cell proteinases destroy ECM and cells unable to attach to
capacity to restore lung structure such as retinoic acid [100]. the ECM oat away and die. The apoptotic models sug-
A recent surprise has been the usefulness of PPE to model gest death of structural cells may be an initiating event,
events upstream from elastolysis. This is likely due to the with subsequent release of matrix degrading proteinases.

103
Asthma and COPD: Basic Mechanisms and Clinical Management

Whether this occurs in human COPD as a primary event is cells and mediators are involved. Investigators are now
uncertain, but does raise interesting testable possibilities. focused on teasing out mechanisms that lead to this com-
Genetic models: Natural genetic mutants, transgenic mice, plex network and the interactions between these cells and
and gene-targeted mice, may develop spontaneous airspace their mediators that leads to COPD.
enlargement of developmental origin. They may also develop Upon acute exposure to cigarette smoke one observes
progressive airspace enlargement acquired spontaneously over activation of macrophages and structural cells that leads
time that more closely reects the destruction of mature lung to a mild, acute, transient accumulation of neutrophils in
tissue characteristic of emphysema [4, 99]. Overexpression or the lung. Over days to weeks one also observes increased
underexpression of a protein during development that leads number of T-cells, particularly CD8 T-cells, and mac-
to failed alveogenesis and enlarged airspaces is very informa- rophages [107]. Neutrophils accumulate with each acute
tive regarding pathways required for normal lung develop- exposure, but due to the short-half life of the neutrophil do
ment and hence repair in emphysema. However, this is very not accumulate to high levels. The longer-lived macrophages
dierent from destruction and enlargement of normal mature and T-cells progressively accumulate with chronic smoke
alveoli that denes pulmonary emphysema. exposure. In humans, one observes B-cell accumulation in
If overexpression of a protein in transgenic mice leads bronchialalveolar lymphoid tissue (BALT) late in the dis-
to airspace enlargement in the adult, then if the protein is ease. While this could be secondary to bacterial coloniza-
overexpressed in the endogenous process it is likely to play tion in humans, it has also been observed in mice exposed
a role. For example, instillation of PPE mimics emphysema to cigarette smoke in the absence of infection [108].
quite well, but of course PPE does not travel from the gut An undoubtedly simplistic but unied pathway, sup-
to the lung in true emphysema. ported by data from cigarette smoke-exposed mice, can
Of most utility in dissecting the pathogenesis of account for much of the inammation observed in response
COPD are developmentally normal gene-targeted mice to cigarette smoke. Cigarette smoke, via its oxidant prop-
applied to cigarette smoke. Protection of a knockout with erties, can activate macrophages leading to TNF- pro-
smoking suggests that the protein deleted is involved in a duction with latent TNF- translocating to the surface
pathway that promotes COPD, while worsening COPD whereupon MMP-12 sheds active TNF- leading to
suggests a protective factor was deleted. There are many neutrophil recruitment and activation [109].
proteins that have been identied using these techniques as Release of MMP-12 from macrophages and colla-
recently reviewed [99]. Knowledge gained from these stud- genases MMP-8 and MMP-13 (MMP-1 not expressed in
ies will be synthesized below. mice) from macrophages and likely epithelial cells leads to
Limitations: The mouse lung structure is not identical generation of elastin and collagen fragments respectively.
to humans. Mice do not inhale cigarettes as humans would Elastin fragments serve as chemokines for macrophages
and these obligate nasal breathers bring the smoke through [101] and collagen fragments promote neutrophil recruit-
an extensive olfactory epithelium into the airway where ment [110]. Indeed, in humans CXC and CC chemokines
there are few submucosal glands, much less airway branch- may play a signicant role in inammatory cell migration,
ing, and no respiratory bronchioles the initial site of cen- particularly when colonized with bacteria. However, in mice
triacinar emphysema. Hence, mice do not develop small under sterile conditions, elastin fragments appear to be crit-
airway disease that is so important in human COPD. ical to the low-level macrophage accumulation one observes
Nevertheless, the opportunity exists to infect mouse airways in response to cigarette smoke.
with relevant human pathogens to learn more about airway T-lymphocytes, particularly CD8 T-cells, are
changes and exacerbations of COPD. increased in lungs of patients with COPD [111].
Mice require months of cigarette smoke exposure to Overexpression of the T-cell product IFN in transgenic
develop mild emphysema. These are more likely to reect mice, results in apoptosis, inammation, and emphysema
Global Initiative for Chronic Obstructive Lung Disease [112]. Moreover, IFN and IFN-IP-10 (interferon-
(GOLD) stage I and II not the severe stages of COPD (III gamma-inducible protein-10) have been shown to be
and IV). Moreover, there is a variety of smoking chambers increased in CD8 T-cells from patients with emphysema,
now available that dier with respect to the mode, rate, and and IP-10 was also shown to induce the production of
amount of smoke delivered. Particulate amounts, carboxy- MMP-12 in human alveolar macrophages [113]. In addi-
hemoglobin levels, and other surrogates of exposure are tion to this potentially indirect inuence of CD8 T-cells
often measured to assess how well these mimic human on emphysema, they may also directly cause cytotoxicity
exposure. The most important features are the resultant contributing to emphysema. The importance of the CD8
phenotypic changes in the exposed animals and their rela- T-cell in the pathogenesis of emphysema, was recently con-
tionship to humans. One note of caution is that excessive rmed by subjecting wild-type (C57BL/6 J), CD8 T-cell
acute exposure to smoke leads to acute lung injury with pro- decient (CD8/) mice, and CD4 T-cell decient (CD4/)
nounced neutrophilia, which is not likely to reect COPD. mice to a model of cigarette smoke-induced emphysema
[107]. In the absence of CD8 T-cells, but not CD4
T-cells, there was a marked inhibition in inammation and
Lessons from mouse models of COPD emphysema. Integrating these results into the network,
one can conclude that CD8 T-cells via production of IP-
Inammatory cell network in COPD: Animal models have 10, signal through macrophage CXCR3 to activate the mac-
taught us that emphysema is not caused by a single cell type rophage leading to proteinase and cytokine production. The
or proteinase. Rather, multiple inammatory and immune macrophage then recruits neutrophils, and their proteinases

104
Asthma and COPD: Animal Models 8
then degrade ECM, particularly elastin and collagen, con- Lung-specic transgenic mice expressing either the Th2
tinuing the inammatory cascade. Hence, CD8 T-cells cytokine IL-13 (Dutch mice) [121] or the Th1 cytokine
may be the master regulator of inammation in COPD. IFN- (British mice) [112] both develop proteinase-
Of note, these results are entirely consistent with data from dependent emphysema. IFN- overexpressors develop a
human COPD. cysteine proteinase/apoptosis mediated form of emphysema,
Defects in adaptive immunity are also likely to play an while IL-13 overexpression induces inammation with
important role with respect to colonization of microorganisms MMP-12- and MMP-9-dependent airspace enlargement.
in the airway and exacerbations. Prolonged cigarette smoke IL-13 transgenic mice also develop small airway remodeling
exposure (1 month) in mice results in a decrease in lung den- that is believed to be due to MMP-9-dependent activation
dritic cell (DC) number, but DC migration to regional tho- of TGF- with airway brosis [122]. The role of TGF- in
racic lymph nodes remains intact. Once in the lymph nodes, emphysema was also highlighted with avb6/ mice, that
however, cigarette smoking suppresses maturation of DCs as are unable to activate TGF- in the airspace, develop mac-
demonstrated by the reduced expression of co-stimulatory rophage inammation and MMP-12-dependent emphy-
molecules. Moreover, these cigarette smoke exposed DC cells sema [123]. TGF- regulation is complex as total absence
have impaired capacity to activate CD4 T-cells with less of TGF- releases the brake on inammation and destruc-
IL-2 production and diminished T-cell proliferation [114]. tion in the airspace while too much TGF- leads to brosis
Impaired CD4 T-cell activation would predispose patients in the airways.
to colonization with infectious microorganisms. Other gene-targeted mice that develop emphysema
As discussed, B-cells and BALT are present in include surfactant protein D/ (SP-D/) mice, which
COPD lung tissue, however, their role in the disease process exhibit macrophage activation, MMP production, and con-
remains to be determined. Of note, recent animal [115] and sequent emphysema [124]. TIMP-3 deciency leads to a
human [116] studies have implicated autoimmune processes combination of developmental airspace enlargement com-
in the pathogenesis of COPD. In fact, elastin fragments bined with progressive destructive emphysema in adults
themselves were shown to be an important autoimmunue [125], supporting the role of MMPs in COPD. There is also
target in COPD in human studies [116]. Other cell types evidence from animal models that neutrophil elastase (NE),
including eosinophils, mast cells, and natural killer (NK) a serine proteinase long thought to cause COPD, is also
cells are also likely to be involved in this inammatory cell involved in experimental emphysema. NE/ mice develop
network, but to date, they have not been carefully addressed. only 40% as much airspace enlargement as wild-type mice
Role of oxidative stress in COPD: Cigarette smoke, exposed to long-term cigarette smoke [126]. These stud-
loaded with 1017 oxidant molecules per pu leads to ies have uncovered several interactions between NE and
increased oxidative stress in smokers and in patients with MMPs. MMPs degrade 1-AT and NE degrades TIMPs,
COPD. Oxidative stress, in part through inactivation of each potentiating the others proteinase activity. Moreover,
histone deacetylases, exposes DNA sequences and activates NE mediates monocyte migration and as discussed, acute
transcription factors for inammatory genes, such as nuclear neutrophil inammation secondary to smoking is related to
factor-B (NF-B) and activator protein (AP)-1 [117]. This MMP-12-dependent TNF-shedding [109].
nding, initially in humans, has been conrmed in rodents Role of apoptosis in COPD: As discussed above, induc-
exposed to cigarette smoke [118]. Oxidants also have a sig- tion of structural cell apoptosis leads to airspace enlarge-
nicant role in promoting apoptosis, and although oxidants ment. However, there are some aspects of these models that
do not directly degrade ECM proteins, they might modify do not mimic human disease. They are non-inammatory
these proteins making them more susceptible to proteo- and transient, which is likely related to their lack of matrix
lytic cleavage. Oxidative stress also can alter the structure degradation. Most importantly, however, closer pathologic
of pro-MMPs leading to autolytic cleavage and activation and physiologic examination of at least one model of epi-
[119]. Gene deletion of Nrf-2 a master transcription fac- thelial cell apoptosis demonstrates that this model is not
tor for antioxidant genes also leads to enhanced emphysema emphysema per se, but acute lung injury with collapsed alve-
in response to cigarette smoke exposure highlighting the oli tethering of open adjoining airspaces to appear enlarged
importance of oxidative stress in COPD [120]. (Mouded M and Shapiro SD, unpublished observations).
Role of proteinases in COPD: As mentioned at the Thus, like other traditional models of epithelial apoptosis,
beginning of this chapter and reviewed in depth in the bleomycin being the classic, the result is acute lung injury
Matrix Degrading Proteinase (Chapter 28), ECM and if severe, brosis. Endothelial apoptosis could have a
destruction by proteinases is the cornerstone of emphysema. dierent outcome, but it is likely that apoptosis does not
Animal models have played an important role in dening initiate emphysema. Apoptosis, however, is likely to be a
the contribution of dierent elastolytic enzymes in this critically important modier in the pathogenesis of COPD.
process. In addition to the original instillation of papain by For example, as in humans, apoptosis is observed in animal
Gross [1], in one of the rst applications of gene-targeted models following cigarette smoke exposure, likely due to
mice to lung disease, it was shown that macrophage elastase loss of basement membrane attachment / oxidant eects
(MMP-12) decient mice were protected from the develop- of cigarette smoke. The fate of the apoptotic cell is criti-
ment of emphysema [95]. As in several other models of dis- cal to disease progression. Clearance by macrophages leads
ease, MMP-12 is pro-inammatory, which in part is related to anti-inammatory TGF- production and release of
to the generation of chemotactic elastin fragments [101]. growth factors that promote repair [127]. Inecient clear-
Subsequently, several other animal models have ance leads to secondary necrosis, augmenting inammation
demonstrated the importance of proteinases in COPD. and destruction.

105
Asthma and COPD: Basic Mechanisms and Clinical Management

Role of repair in COPD: The ability of the adult lung References


to repair damaged alveoli appears limited. This is likely due
to the structural and functional dependence upon the elas- 1. Gross P, Ptzer E, Tolker M. Experimental emphysema: Its produc-
tic ber architecture combined with the diculty in repair- tion with papain in normal and silicotic rats. Arch Environ Health 11:
ing elastic bers. Whether one can temporally and spatially 5058, 1965.
bring together the multiple microbrillar components of an 2. Drazen JM, Israel E, OByrne PM. Treatment of asthma with drugs
elastic ber let alone coordinate matrix repair with endothe- modifying the leukotriene pathway. N Engl J Med 340: 197206, 1999.
lial and epithelial migration and dierentiation over an 3. Shapiro SD. Animal models of asthma: Pro: Allergic avoidance of ani-
injured matrix is unknown. mal (model[s]) is not an option. Am J Respir Crit Care Med 174: 1171
73, 2006.
With the emergence of regenerative medicine and
4. Shapiro SD. Transgenic and gene-targeted mice as models for chronic
stem cell biology, there is hope that we can one day cre- obstructive pulmonary disease. Eur Respir J 29: 37578, 2007.
ate new functional lung tissue. However, given the com- 5. Boyce JA, Austen KF. No audible wheezing: Nuggets and conundrums
plex three-dimensional nature of the lung, its regeneration from mouse asthma models. J Exp Med 201: 186973, 2005.
presents a special challenge relative to other organs. It is 6. Wenzel S, Holgate ST. The mouse trap: It still yields few answers in
currently not known whether there is a master stem cell, asthma. Am J Respir Crit Care Med 174: 117376, 2006. Discussion
separate progenitors for each cell type, nor what their repar- 117678.
ative capacity is. More importantly, given the collapse of the 7. Zosky GR, Sly PD. Animal models of asthma. Clin Exp Allergy 37:
elastic ber cable network, it is unclear whether stem cells 97388, 2007.
alone could regenerate a functional alveolar unit. 8. Lommatzsch M, Julius P, Kuepper M, Garn H, Bratke K, Irmscher
S, Luttmann W, Renz H, Braun A, Virchow JC. The course of aller-
There has been some evidence that one can repair
gen-induced leukocyte inltration in human and experimental asthma.
emphysematous lung tissue in animal models. The study J Allergy Clin Immunol 118: 9197, 2006.
by Massaro et al. [100] discussed above, showing that all- 9. Zhang Y, Lamm WJ, Albert RK, Chi EY, Henderson WR Jr., Lewis DB.
trans retinoic acid reversed PPE-mediated emphysema, has Inuence of the route of allergen administration and genetic back-
led to hope that we might be able to promote lung repair ground on the murine allergic pulmonary response. Am J Respir Crit
[100]. Use of retinoic acid did not have signicant eects in Care Med 155: 66169, 1997.
human trials, but has opened the door to mechanisms that 10. Hamelmann E, Tadeda K, Oshiba A, Gelfand EW. Role of IgE
could be reparative. in the development of allergic airway inammation and airway
There have been attempts to add growth factors, such hyperresponsiveness a murine model. Allergy. 54: 297305, 1999.
as keratinocyte growth factor (KGF) to elastase-treated 11. Shore SA. Modeling airway remodeling: The winner by a nose? Am J
Respir Crit Care Med 168: 91011, 2003.
lungs. The results demonstrate the ability to protect from
12. Shinagawa K, Kojima M. Mouse model of airway remodeling: Strain
injury, but not repair damaged tissue [128]. The advent of dierences. Am J Respir Crit Care Med 168: 95967, 2003.
lineage tagged and lineage ablated genetically engineered 13. Brewer JP, Kisselgof AB, Martin TR. Genetic variability in pulmonary
mice will be helpful to dene progenitor cells and their physiological, cellular, and antibody responses to antigen in mice. Am J
reparative capacity in the future. Respir Crit Care Med 160: 115056, 1999.
14. Ewart SL, Kuperman D, Schadt E, Tankersley C, Grupe A,
Shubitowski DM, Peltz G, Wills-Karp M. Quantitative trait loci con-
trolling allergen-induced airway hyperresponsiveness in inbred mice.
Am J Respir Cell Mol Biol 23: 53745, 2000.
SUMMARY 15. Lloyd CM. Building better mouse models of asthma. Curr Allergy
Asthma Rep 7: 23136, 2007.
16. Yu M, Tsai M, Tam SY, Jones C, Zehnder J, Galli SJ. Mast cells can
It is important to keep in mind that there are important promote the development of multiple features of chronic asthma in
structural dierences in the lungs of mice and humans and mice. J Clin Invest 116: 163341, 2006.
to interpret animal data with caution. In some instances, 17. Lim DH, Cho JY, Miller M, McElwain K, McElwain S, Broide DH.
murine models of allergic asthma have not recapitulated Reduced peribronchial brosis in allergen-challenged MMP-9-decient
the human ndings, and have led to some costly failures mice. Am J Physiol Lung Cell Mol Physiol 291: L265271, 2006.
in drug development. Nevertheless, animal models have 18. Garssen J, Nijkamp FP, Van Der Vliet H, Van Loveren H. T-cell-
mediated induction of airway hyperreactivity in mice. Am Rev Respir
been extremely useful in the evolution of current concepts
Dis 144: 93138, 1991.
regarding the etiology of asthma and COPD and geneti- 19. Corry DB, Folkesson HG, Warnock ML, Erle DJ, Matthay MA,
cally engineered mice continue to drive the generation and Wiener-Kronish JP, Locksley RM. Interleukin 4, but not interleukin
testing of new hypotheses. 5 or eosinophils, is required in a murine model of acute airway hyper-
reactivity. J Exp Med 183: 10917, 1996.
20. Brusselle GG, Kips JC, Tavernier JH, van der Heyden JG, Cuvelier
CA, Pauwels RA, Bluethmann H. Attenuation of allergic airway
inammation in IL-4 decient mice. Clin Exp Allergy 24: 7380, 1994.
ACKNOWLEDGEMENTS 21. Gavett SH, Chen X, Finkelman F, Wills-Karp M. Depletion of murine
CD4 T lymphocytes prevents antigen-induced airway hyperreactivity
and pulmonary eosinophilia. Am J Respir Cell Mol Biol 10: 58793, 1994.
The authors work is supported by National Institute of 22. Corry DB, Grunig G, Hadeiba H, Kurup VP, Warnock ML, Sheppard D,
Environmental Health Sciences Grants ES-013307 and Rennick DM, Locksley RM. Requirements for allergen-induced air-
ES-00002, and National Heart, Lung, and Blood Institute way hyperreactivity in T and B cell-decient mice. Mol Med 4: 34455,
Grants HL-084044 and HL-082541. 1998.

106
Asthma and COPD: Animal Models 8
23. Zuany-Amorim C, Rue C, Haile S, Vargaftig BB, Pereira P, 42. Kung TT, Stelts D, Zurcher JA, Jones H, Umland SP, Kreutner W,
Pretolani M. Requirement for gammadelta T cells in allergic airway Egan RW, Chapman RW. Mast cells modulate allergic pulmonary
inammation. Science 280: 126567, 1998. eosinophilia in mice. Am J Respir Cell Mol Biol 12: 4049, 1995.
24. Randolph DA, Stephens R, Carruthers CJ, Chaplin DD. Cooperation 43. Kobayashi T, Miura T, Haba T, Sato M, Serizawa I, Nagai H, Ishizaka K.
between Th1 and Th2 cells in a murine model of eosinophilic airway An essential role of mast cells in the development of airway hyperre-
inammation. J Clin Invest 104: 102129, 1999. sponsiveness in a murine asthma model. J Immunol 164: 385561, 2000.
25. Meyer EH, DeKruy RH, Umetsu DT. T cells and NKT cells in the 44. MacLean JA, Sauty A, Luster AD, Drazen JM, De Sanctis GT.
pathogenesis of asthma. Annu Rev Med 59: 28192, 2008. Antigen-induced airway hyperresponsiveness, pulmonary eosinophilia,
26. Strickland DH, Stumbles PA, Zosky GR, Subrata LS, Thomas JA, and chemokine expression in B cell-decient mice. Am J Respir Cell
Turner DJ, Sly PD, Holt PG. Reversal of airway hyperresponsiveness Mol Biol 20: 37987, 1999.
by induction of airway mucosal CD4 CD25 regulatory T cells. J Exp 45. Takeda K, Hamelmann E, Joetham A, Shultz LD, Larsen GL, Irvin CG,
Med 203: 264960, 2006. Gelfand EW. Development of eosinophilic airway inammation and
27. Lukacs NW, Strieter RM, Chensue SW, Kunkel SL. Interleukin-4- airway hyperresponsiveness in mast cell-decient mice. J Exp Med 186:
dependent pulmonary eosinophil inltration in a murine model of 44954, 1997.
asthma. Am J Respir Cell Mol Biol 10: 52632, 1994. 46. Gleich GJ. The eosinophil and bronchial asthma: Current understanding.
28. Kips JC, Brusselle GG, Joos GF, Peleman RA, Devos RR, Tavernier JH, J Allergy Clin Immunol 85: 42236, 1990.
Pauwels RA. Importance of interleukin-4 and interleukin-12 in allergen- 47. Lee JJ, McGarry MP, Farmer SC, Denzler KL, Larson KA,
induced airway changes in mice. Int Arch Allergy Immunol 107: 11518, Carrigan PE, Brenneise IE, Horton MA, Haczku A, Gelfand EW,
1995. Leikauf GD, Lee NA. Interleukin-5 expression in the lung epithe-
29. Grunig G, Warnock M, Wakil AE, Venkayya R, Brombacher F, lium of transgenic mice leads to pulmonary changes pathognomonic of
Rennick DM, Sheppard D, Mohrs M, Donaldson DD, Locksley RM, asthma. J Exp Med 185: 214356, 1997.
Corry DB. Requirement for IL-13 independently of IL-4 in experi- 48. Foster PS, Hogan SP, Ramsay AJ, Matthaei KI, Young IG. Interleukin
mental asthma [see comments]. Science 282: 226163, 1998. 5 deciency abolishes eosinophilia, airways hyperreactivity, and lung
30. Hamelmann E, Wahn U, Gelfand EW. Role of the Th2 cytokines in damage in a mouse asthma model [see comments]. J Exp Med 183:
the development of allergen-induced airway inammation and hyper- 195201, 1996.
responsiveness. Int Arch Allergy Immunol 118: 9094, 1999. 49. Hamelmann E, Takeda K, Haczku A, Cieslewicz G, Shultz L, Hamid Q,
31. Wills-Karp M, Luyimbazi J, Xu X, Schoeld B, Neben TY, Karp CL, Xing Z, Gauldie J, Gelfand EW. Interleukin (IL)-5 but not immu-
Donaldson DD. Interleukin-13: Central mediator of allergic asthma noglobulin E reconstitutes airway inammation and airway hyper-
[see comments]. Science 282: 225861, 1998. responsiveness in IL-4-decient mice. Am J Respir Cell Mol Biol 23:
32. He R, Oyoshi MK, Jin H, Geha RS. Epicutaneous antigen exposure 32734, 2000.
induces a Th17 response that drives airway inammation after inhala- 50. Hogan SP, Mould A, Kikutani H, Ramsay AJ, Foster PS. Aeroallergen-
tion challenge. Proc Natl Acad Sci USA 104: 1581722, 2007. induced eosinophilic inammation, lung damage, and airways hyperre-
33. Karwot R, Maxeiner JH, Schmitt S, Scholtes P, Hausding M, Lehr HA, activity in mice can occur independently of IL-4 and allergen-specic
Glimcher LH, Finotto S. Protective role of nuclear factor of activated immunoglobulins. J Clin Invest 99: 132939, 1997.
T cells 2 in CD8() long-lived memory T cells in an allergy model. 51. Flood-Page P, Swenson C, Faiferman I, Matthews J, Williams M,
J Allergy Clin Immunol 121(4): 99299, 2008. Brannick L, Robinson D, Wenzel S, Busse W, Hansel TT, Barnes NC.
34. Pichavant M, Goya S, Meyer EH, Johnston RA, Kim HY, A study to evaluate safety and ecacy of mepolizumab in patients with
Matangkasombut P, Zhu M, Iwakura Y, Savage PB, DeKruy RH, moderate persistent asthma. Am J Respir Crit Care Med 176: 106271,
Shore SA, Umetsu DT. Ozone exposure in a mouse model induces air- 2007.
way hyperreactivity that requires the presence of natural killer T cells 52. OByrne PM. The demise of anti IL-5 for asthma, or not. Am J Respir
and IL-17. J Exp Med 205: 38593, 2008. Crit Care Med 176: 105960, 2007.
35. Fahy JV, Fleming HE, Wong HH, Liu JT, Su JQ, Reimann J, 53. Hessel EM, Van Oosterhout AJ, Van Ark I, Van Esch B, Hofman G,
Fick RB Jr., Boushey HA. The eect of an anti-IgE monoclonal anti- Van Loveren H, Savelkoul HF, Nijkamp FP. Development of airway
body on the early- and late-phase responses to allergen inhalation in hyperresponsiveness is dependent on interferon-gamma and independ-
asthmatic subjects. Am J Respir Crit Care Med 155: 182834, 1997. ent of eosinophil inltration. Am J Respir Cell Mol Biol 16: 32534,
36. Zosky GR, Larcombe AN, White OJ, Burchell JT, Janosi TZ, Hantos Z, 1997.
Holt PG, Sly PD, Turner DJ. Ovalbumin-sensitized mice are good 54. Humbles AA, Lloyd CM, McMillan SJ, Friend DS, Xanthou G,
models for airway hyperresponsiveness but not acute physiological McKenna EE, Ghiran S, Gerard NP, Yu C, Orkin SH, Gerard C. A
responses to allergen inhalation. Clin Exp Allergy 38(6): 82938, 2007. critical role for eosinophils in allergic airways remodeling. Science 305:
37. Martin TR, Galli SJ, Katona IM, Drazen JM. Role of mast cells in 177679, 2004.
anaphylaxis. Evidence for the importance of mast cells in the cardi- 55. Beuther DA, Weiss ST, Sutherland ER. Obesity and asthma. Am J
opulmonary alterations and death induced by anti-IgE in mice. J Clin Respir Crit Care Med 174: 11219, 2006.
Invest 83: 137583, 1989. 56. Ford ES. The epidemiology of obesity and asthma. J Allergy Clin
38. Chand N, Nolan K, Soa RD, Diamantis W. Changes in aeroallergen- Immunol 115: 897909, 2005. , quiz 910
induced pulmonary mechanics in actively sensitized guinea pig: 57. Shore SA, Johnston RA. Obesity and asthma. Pharmacol Ther 110:
Inhibition by azelastine. Ann Allergy 64: 15154, 1990. 83102, 2006.
39. Eidelman DH, Belloore S, Martin JG. Late airway responses to anti- 58. Shore SA. Obesity and asthma: Implications for treatment. Curr Opin
gen challenge in sensitized inbred rats. Am Rev Respir Dis 137: 1033 Pulm Med 13: 5662, 2007.
37, 1988. 59. Boulet LP, Franssen E. Inuence of obesity on response to uticasone
40. Abraham WM, Oliver W Jr., King MM, Yerger L, Wanner A. Eect with or without salmeterol in moderate asthma. Respir Med 101: 2240
of pharmacologic agents on antigen-induced decreases in specic lung 47, 2007.
conductance in sheep. Am Rev Respir Dis 124: 55458, 1981. 60. Lu FL, Johnston RA, Flynt L, Theman TA, Terry RD, Schwartzman
41. Djukanovic R, Wilson SJ, Kraft M, Jarjour NN, Steel M, Chung IN, Lee A, Shore SA. Increased pulmonary responses to acute ozone
KF, Bao W, Fowler-Taylor A, Matthews J, Busse WW, Holgate ST, exposure in obese db/db mice. Am J Physiol Lung Cell Mol Physiol 290:
Fahy JV. Eects of treatment with anti-immunoglobulin E antibody L856l865, 2006.
omalizumab on airway inammation in allergic asthma. Am J Respir 61. Johnston RA, Theman TA, Lu FL, Terry RD, Williams ES Shore SA
Crit Care Med 170: 58393, 2004. (in press) Diet-induced obesity causes innate airway hyperresponsiveness

107
Asthma and COPD: Basic Mechanisms and Clinical Management

to methacholine and enhances ozone induced pulmonary inammation. 83. Fantuzzi G. Adipose tissue, adipokines, and inammation. J Allergy
J Appl Physiol 104: 172735, 2008. Clin Immunol 115: 91119, 2005.
62. Johnston RA, Theman TA, Shore SA. Augmented Responses to Ozone 84. Williams AS, Leung SY, Nath P, Khorasani NM, Bhavsar P, Issa R,
in Obese Carboxypeptidase E-Decient Mice. Am J Physiol Regul Mitchell JA, Adcock IM, Chung KF. Role of TLR2, TLR4, and
Integr Comp Physiol 290: R126R133, 2006. MyD88 in murine ozone-induced airway hyperresponsiveness and
63. Johnston RA, Zhu M, Rivera-Sanchez YM, Lu FL, Theman TA, neutrophilia. J Appl Physiol 103: 118995, 2007.
Flynt L, Shore SA. Allergic airway responses in obese mice. Am J 85. Kadowaki T, Yamauchi T, Kubota N. The physiological and patho-
Respir Crit Care Med 176: 65058, 2007. physiological role of adiponectin and adiponectin receptors in the
64. Rivera-Sanchez YM, Johnston RA, Schwartzman IN, Valone J, peripheral tissues and CNS. FEBS Lett 582: 7480, 2008.
Silverman ES, Fredberg JJ, Shore SA. Dierential eects of ozone on 86. Fantuzzi G. Adiponectin and inammation: Consensus and contro-
airway and tissue mechanics in obese mice. J Appl Physiol 96: 22006, versy. J Allergy Clin Immunol 121: 32630, 2008.
2004. 87. Okamoto Y, Folco EJ, Minami M, Wara AK, Feinberg MW,
65. Shore SA, Rivera-Sanchez YM, Schwartzman IN, Johnston RA. Sukhova GK, Colvin RA, Kihara S, Funahashi T, Luster AD, Libby P.
Responses to ozone are increased in obese mice. J Appl Physiol 95: 938 Adiponectin Inhibits the Production of CXC Receptor Chemokine
45, 2003. Ligands in Macrophages and Reduces T-Lymphocyte Recruitment in
66. Shore SA, Schwartzman IN, Mellema MS, Flynt L, Imrich A, Atherogenesis. Circ Res 121, 2007.
Johnston RA. Eect of leptin on allergic airway responses in mice. 88. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S,
J Allergy Clin Immunol 115: 1039, 2005. Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K,
67. Shore SA, Terry RD, Flynt L, Xu A, Hug C. Adiponectin attenuates Ohteki T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y,
allergen-induced airway inammation and hyperresponsiveness in Terauchi Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T,
mice. J Allergy Clin Immunol 118: 38995, 2006. Shimizu T, Nagai R, Kadowaki T. Cloning of adiponectin receptors
68. Shore SA. Obesity and asthma: Lessons from animal models. J Appl that mediate antidiabetic metabolic eects. Nature 423: 76269, 2003.
Physiol 102: 51628, 2007. 89. Hug C, Wang J, Ahmad NS, Bogan JS, Tsao TS, Lodish HF.
69. Leibel RL, Chung WK, Chua SC Jr. The molecular genetics of rodent T-cadherin is a receptor for hexameric and high-molecular-weight forms
single gene obesities. J Biol Chem 272: 3193740, 1997. of Acrp30/adiponectin. Proc Natl Acad Sci USA 101: 1030813, 2004.
70. Scherer PE. Adipose tissue: From lipid storage compartment to endo- 90. Snider GL, Lucey EC, Stone PJ. Animal models of emphysema. Am
crine organ. Diabetes 55: 153745, 2006. Rev Respir Dis 133: 14969, 1986.
71. Lang JE, Williams ES, Shore SA. TNF contributes to innate airway 91. Kuhn C, Yu SY, Chraplyvy M, Linder HE, Senior RM. The induc-
hyperresponsiveness in murine obesity. Am J Respir Crit Care Med 175: tion of emphysema with elastase. II: Changes in connective tissue.
A912, 2007. Lab Invest 34: 37280, 1976.
72. Tolbert PE, Mulholland JA, MacIntosh DL, Xu F, Daniels D, 92. Jano A, Sloan B, Weinbaum G, Damiano V, Sandhaus RA, Elias J,
Devine OJ, Carlin BP, Klein M, Dorley J, Butler AJ, Nordenberg DF, Kimbel P. Experimental emphysema induced with puried human
Frumkin H, Ryan PB, White MC. Air quality and pediatric emergency neutrophil elastase: Tissue localization of the instilled protease. Am
room visits for asthma in Atlanta, Georgia, USA. Am J Epidemiol 151: Rev Respir Dis 115: 46178, 1977.
798810, 2000. 93. Kao RC, Wehner NG, Skubitz KM, Gray BH, Hoidal JR. Proteinase
73. Gent JF, Triche EW, Holford TR, Belanger K, Bracken MB, 3. A distinct human polymorphonuclear leukocyte proteinase that
Beckett WS, Leaderer BP. Association of low-level ozone and ne par- produces emphysema in hamsters. J Clin Invest 82: 196373, 1988.
ticles with respiratory symptoms in children with asthma. JAMA 290: 94. Wright JL, Churg A. Cigarette smoke causes physiologic and mor-
185967, 2003. phologic changes of emphysema in the guinea pig. Am Rev Respir Dis
74. Bennett WD, Hazucha MJ, Folinsbee LJ, Bromberg PA, Kissling GE, 142: 142228, 1990.
London SJ. Acute pulmonary function response to ozone in young 95. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD.
adults as a function of body mass index. Inhal Toxicol 19: 114754, Requirement for macrophage elastase for cigarette smoke-induced
2007. emphysema in mice. Science 277: 20024, 1997.
75. Alexee SE, Litonjua AA, Suh H, Sparrow D, Vokonas PS, Schwartz J. 96. Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF.
Ozone Exposure and Lung Function: Eect Modied by Obesity and Endothelial cell death and decreased expression of vascular endothe-
Airways Hyperresponsiveness in the VA Normative Aging Study. Chest lial growth factor and vascular endothelial growth factor receptor 2 in
132: 189097, 2007. emphysema. Am J Respir Crit Care Med 163: 73744, 2001.
76. Johnston RA, Williams ES, Shore SA. Airway responses to ozone dur- 97. Aoshiba K, Yokohori N, Nagai A. Alveolar wall apoptosis causes lung
ing the development of obesity. Proc Am Thorac Soc 3: A528, 2006. destruction and emphysematous changes. Am J Respir Cell Mol Biol
77. Lang JE, Williams ES, Mizgerd JP Shore SA (in press) Eect of obes- 28: 55562, 2003.
ity on pulmonary inammation induced by acute ozone exposure: Role 98. Lucey EC, Keane J, Kuang PP, Snider GL, Goldstein RH. Severity
of interleukin-Am J Physiol Lung Cell Mol Physiol 294: L101320, 2008. of elastase-induced emphysema is decreased in tumor necrosis factor-
78. Mito N, Kitada C, Hosoda T, Sato K. Eect of diet-induced obesity alpha and interleukin-1beta receptor-decient mice. Lab Invest 82:
on ovalbumin-specic immune response in a murine asthma model. 7985, 2002.
Metabolism 51: 124146, 2002. 99. Mahadeva R, Shapiro SD. Animal models of pulmonary emphysema.
79. Back M, Sultan A, Ovchinnikova O, Hansson GK. 5-Lipoxygenase- Curr Drug Targets Inamm Allergy 4: 66573, 2005.
activating protein: A potential link between innate and adaptive immu- 100. Massaro GD, Massaro D. Retinoic acid treatment abrogates elastase-
nity in atherosclerosis and adipose tissue inammation. Circ Res 100: induced pulmonary emphysema in rats. Nat Med 3: 67577, 1997.
94649, 2007. 101. Houghton AM, Quintero PA, Perkins DL, Kobayashi DK, Kelley
80. Peters-Golden M, Swern A, Bird SS, Hustad CM, Grant E, Edelman DG, Marconcini LA, Mecham RP, Senior RM, Shapiro SD. Elastin
JM. Inuence of body mass index on the response to asthma controller fragments drive disease progression in a murine model of emphysema.
agents. Eur Respir J 27: 495503, 2006. J Clin Invest 116: 75359, 2006.
81. Guler N, Kirerleri E, Ones U, Tamay Z, Salmayenli N, Darendeliler F. 102. Snider GL, Lucey EC, Faris B, Jung-Legg Y, Stone PJ, Franzblau C.
Leptin: Does it have any role in childhood asthma? J Allergy Clin Cadmium-chloride-induced air-space enlargement with interstitial
Immunol 114: 25459, 2004. pulmonary brosis is not associated with destruction of lung elastin.
82. Sood A, Ford ES, Camargo CA Jr. Association between leptin and Implications for the pathogenesis of human emphysema. Am Rev
asthma in adults. Thorax 61: 3005, 2006. Respir Dis 137: 91823, 1988.

108
Asthma and COPD: Animal Models 8
103. Shapiro SD. COPD unwound. N Engl J Med 352: 201619, 2005. 117. Ito K, Ito M, Elliott WM, Cosio B, Caramori G, Kon OM, Barczyk A,
104. Wright JL, Postma DS, Kerstjens HA, Timens W, Whittaker P, Hayashi S, Adcock IM, Hogg JC, Barnes PJ. Decreased histone
Churg A. Airway remodeling in the smoke exposed guinea pig model. deacetylase activity in chronic obstructive pulmonary disease. N Engl
Inhal Toxicol 19: 91523, 2007. J Med 352: 196776, 2005.
105. Medler TR, Petrusca DN, Lee PJ, Hubbard WC, Berdyshev EV, 118. Marwick JA, Kirkham PA, Stevenson CS, Danahay H, Giddings J,
Skirball J, Kamocki K, Schuchman E, Tuder RM, Petrache I. Butler K, Donaldson K, Macnee W, Rahman I. Cigarette smoke
Apoptotic sphingolipid signaling by ceramides in lung endothelial alters chromatin remodeling and induces proinammatory genes in
cells. Am J Respir Cell Mol Biol 38(6): 63946, 2008. rat lungs. Am J Respir Cell Mol Biol 31: 63342, 2004.
106. Tang K, Rossiter HB, Wagner PD, Breen EC. Lung-targeted VEGF 119. Parks WC, Shapiro SD. Matrix metalloproteinases in lung biology.
inactivation leads to an emphysema phenotype in mice. J Appl Physiol Respir Res 2: 1019, 2001.
97: 155966, 2004. Discussion 1549. 120. Rangasamy T, Cho CY, Thimmulappa RK, Zhen L, Srisuma SS,
107. Maeno T, Houghton AM, Quintero PA, Grumelli S, Owen CA, Kensler TW, Yamamoto M, Petrache I, Tuder RM, Biswal S. Genetic
Shapiro SD. CD8 T Cells are required for inammation and ablation of Nrf2 enhances susceptibility to cigarette smoke-induced
destruction in cigarette smoke-induced emphysema in mice. emphysema in mice. J Clin Invest 114: 124859, 2004.
J Immunol 178: 809096, 2007. 121. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, Riese RJ Jr.,
108. van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Chapman HA Jr., Shapiro SD, Elias JA. Inducible targeting of IL-13
Luinge MA, Geerlings M, Hylkema MN, van den Berg A, Timens W, to the adult lung causes matrix metalloproteinase- and cathepsin-
Kerstjens HA. Cigarette smoke-induced emphysema: A role for the B dependent emphysema. J Clin Invest 106: 108193, 2000.
cell? Am J Respir Crit Care Med 173: 75158, 2006. 122. Lee CG, Homer RJ, Zhu Z, Lanone S, Wang X, Koteliansky V,
109. Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, Shapiro SD, Shipley JM, Gotwals P, Noble P, Chen Q, Senior RM, Elias JA.
Wright JL. Macrophage metalloelastase mediates acute cigarette Interleukin-13 induces tissue brosis by selectively stimulating and
smoke-induced inammation via tumor necrosis factor-alpha release. activating transforming growth factor beta(1). J Exp Med 194: 80921,
Am J Respir Crit Care Med 167: 108389, 2003. 2001.
110. Weathington NM, van Houwelingen AH, Noerager BD, Jackson 123. Morris DG, Huang X, Kaminski N, Wang Y, Shapiro SD, Dolganov G,
PL, Kraneveld AD, Galin FS, Folkerts G, Nijkamp FP, Blalock JE. Glick A, Sheppard D. Loss of integrin alpha(v)beta6-mediated TGF-
A novel peptide CXCR ligand derived from extracellular matrix deg- beta activation causes Mmp12-dependent emphysema. Nature 422:
radation during airway inammation. Nat Med 12: 31723, 2006. 16973, 2003.
111. Saetta M. Airway inammation in chronic obstructive pulmonary 124. Wert SE, Yoshida M, LeVine AM, Ikegami M, Jones T, Ross GF,
disease. Am J Respir Crit Care Med 160: S1720, 1999. Fisher JH, Korfhagen TR, Whitsett JA. Increased metalloproteinase
112. Wang Z, Zheng T, Zhu Z, Homer RJ, Riese RJ, Chapman HA Jr., activity, oxidant production, and emphysema in surfactant protein D
Shapiro SD, Elias JA. Interferon gamma induction of pulmonary gene-inactivated mice. Proc Natl Acad Sci USA 97: 597277, 2000.
emphysema in the adult murine lung. J Exp Med 192: 1587600, 125. Leco KJ, Waterhouse P, Sanchez OH, Gowing KL, Poole AR,
2000. Wakeham A, Mak TW, Khokha R. Spontaneous air space enlargement
113. Grumelli S, Corry DB, Song LZ, Song L, Green L, Huh J, in the lungs of mice lacking tissue inhibitor of metalloproteinases-3
Hacken J, Espada R, Bag R, Lewis DE, Kheradmand F. An immune (TIMP-3). J Clin Invest 108: 81729, 2001.
basis for lung parenchymal destruction in chronic obstructive pulmo- 126. Shapiro SD, Goldstein NM, Houghton AM, Kobayashi DK,
nary disease and emphysema. PLoS Med 1: e8, 2004. Kelley D, Belaaouaj A. Neutrophil elastase contributes to cigarette
114. Robbins C, Franco F, Cernandes M Shapiro SD (in press) Cigarette smoke-induced emphysema in mice. Am J Pathol 163: 232935, 2003.
smoke exposure impairs dendritic cell maturation and T cell prolifera- 127. Henson PM, Cosgrove GP, Vandivier RW. State of the art. Apoptosis
tion in thoracic lymph nodes of mice. J Immunol 180: 66238, 2008. and cell homeostasis in chronic obstructive pulmonary disease. Proc
115. Taraseviciene-Stewart L, Scerbavicius R, Choe KH, Moore M, Am Thorac Soc 3: 51216, 2006.
Sullivan A, Nicolls MR, Fontenot AP, Tuder RM, Voelkel NF. An 128. Plantier L, Marchand-Adam S, Antico VG, Boyer L, De Coster C,
animal model of autoimmune emphysema. Am J Respir Crit Care Med Marchal J, Bachoual R, Mailleux A, Boczkowski J, Crestani B.
171: 73442, 2005. Keratinocyte growth factor protects against elastase-induced pul-
116. Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S, monary emphysema in mice. Am J Physiol Lung Cell Mol Physiol 293:
Green L, Hacken-Bitar J, Huh J, Bakaeen F, Coxson HO, Cogswell S, L12301239, 2007.
Storness-Bliss C, Corry DB, Kheradmand F. Antielastin autoimmunity
in tobacco smoking-induced emphysema. Nat Med 13: 56769, 2007.

109
Inflammatory Cells
3
PART

and Extracellular
Matrix
Mast Cells and Basophils
9
CHAPTER

Mast cells and basophils rst came to attention George H. Caughey


over a century ago due to their ample stocks ORIGIN AND FATE
of intracellular granules with unusual staining Cardiovascular Research Institute and
characteristics. For many years their origins, nor- Department of Medicine, University of
Mast cells and basophils have shared origins
mal functions, and roles in disease were obscure, but distinct distributions and fates. Mast cells, California at San Francisco, USA
and in some respects remain so. Nonetheless, but not basophils, are normal residents of unin-
knowledge of their biology increased tremen- amed airways. Mature mast cells rarely appear
dously in the past two decades [15]. in blood, whereas mature basophils circulate
Researchers once focused on mast cell and are recruited from blood to sites of allergic
and basophil release of histamine and eicosa- inammation. Both cell types originate from
noids in acute allergic events, which were con- shared progenitors in bone marrow; see Lee
sidered to be a corruption of hypothesized and Krilis [12] for a review. Immature mast cells
normal function of defending against invasion released from marrow circulate briey, exit the
by parasites such as worms and ticks. However, bloodstream to multiple tissue destinations, then
studies in mice now suggest that mast cells dierentiate, adopting a phenotype determined
contribute to innate immune defense against by their microenvironment. Basophils, on the
airway bacteria like mycoplasma [6] and pro- other hand, mature in the marrow, circulate, then
tection from immunologically nonspecic home to sites of inammation if recruited to do
injury, as from venoms [7]. Mast cells and so. Tissue mast cells are not xed in location,
basophils also have the demonstrated potential for they migrate toward airway epithelium after
to inuence immune system development, reg- antigen challenge and trac to lymph nodes
ulation, and initiation of the immune response from sites of antigen exposure. Mast cells prob-
[811]. In some contexts, the overall contri- ably also proliferate in tissues. Conditions such
bution of mast cells is anti-inammatory [6]. as gut parasitosis lead to large local increases in
These roles deviate from traditional concepts mast cells.
of mast cell and basophil participation in adap- Both types of cell survive degranulation
tive responses involving antigen recognition and can restock their secretory granules with
by IgE. mediators. Life span in vivo has not been estab-
In asthma investigations, attention is shift- lished, but in vitro studies predict that basophils
ing from roles of these cells in acute responses are short-lived compared to mast cells, which
to aeroallergen to roles in promoting persist- survive for weeks in culture. In humans, the
ent inammation and remodeling in chronic aggregate mast cell mass is much larger than
disease. Their roles in other obstructive lung that of basophils, which usually comprise 1%
diseases have received less attention, but they of circulating leukocytes. Among mammals,
may indeed contribute to conditions apart from basophil numbers vary widely, ranging from
asthma. numerous in guinea pigs, few in humans and
This chapter summarizes current think- mice, to nearly nonexistent in dogs, in which
ing about roles of mast cells and basophils in they are arguably unimportant. There are genetic
obstructive airway disease. variants of mice with almost no mast cells but

113
Asthma and COPD: Basic Mechanisms and Clinical Management

none with an inherited, selective decit of basophils; thus, it RECRUITMENT


is easier to assess involvement of mast cells than of basophils
in mouse disease models; see Galli [5] for a review.
In asthmatics, increased numbers of basophils and activated
mast cells appear in sputum after allergen challenge [17],
which may reect migration from submucosal sites and blood-
stream, respectively. As noted, mast cell precursors home to
DEVELOPMENT AND HETEROGENEITY tissues even without inammation. Constitutive homing and
epithelial migration may involve responses to proteins such as:
Paucigranular, mast cell-committed progenitors are released C5a;
from marrow expressing surface receptor tyrosine kinase
c-kit and low anity IgG receptor (FcRII) but not high RANTES;
anity IgE receptor, FcRI. In vitro, cells with mature char- IL-8;
acteristics, including FcRI and protease-rich granules, dif-
MIP-1;
ferentiate from progenitors under the inuence of IL-6 and
kit ligand. Presumably, similar events occur in vivo, with kit MCP-1;
ligand produced by endothelial, stromal, and epithelial cells VEGF;
being critical for mast cell survival. Mice with defective
c-kit or its ligand lack mast cells. Their importance to mast fractalkine;
cell development is underscored by gain-of-function c-kit CXCL10;
mutations in systemic mastocytosis and mast cell malig- kit ligand.
nancy, and development of generalized mastocytosis in
response to exogenous kit ligand. The importance of local These may orchestrate movement singly or in combi-
production of kit ligand is suggested by the nding in mice nation. Cultured human mast cells express a broad repertoire
that intratracheal kit ligand provokes mast cell-dependent of chemokine receptors, which diminishes as cells mature,
hyperreactivity [13]. thus limiting mast cell movement after dierentiation at a
In vitro, a variety of cytokines (especially IL-3, -6, -9 tissue site.
and -10, and TGF-1) determines phenotype of matur- Kit ligand is notable in that it is specic for mast
ing mouse mast cells. In the case of IL-9 (a candidate mouse cells in comparison with other leukocytes and is produced
asthma gene), airway overexpression in transgenic mice by airway cells. Mast cell migration into tissues depends
causes eosinophilic inammation and hyperresponsiveness on integrins and other adhesion molecules [18]. Basophils
with mast cell hyperplasia [14]. express an array of chemokine receptors. Chemokines bind-
Mast cells vary in features such as: ing to CCR3 (such as RANTES and eotaxins) may be
especially important [19, 20]. The multiplicity of chemoat-
proteoglycan and protease content; tractants predicts redundant recruiting pathways. Several
metachromasia; chemoattractants also prime or activate one or both types of
cells [20, 21], enhancing their role in asthma pathogenesis
granule ultrastructure; beyond that of recruitment alone.
responses to degranulating stimuli, such as substance P.

These phenotypic variations appear reversible in a given


cell and changeable in cell populations in response to infec-
tion and injury. Classically, rodent mast cells are divided into ACTIVATION
mucosal and connective tissue groups, although the phe-
notype distribution is not strictly bimodal. Human mast cells
are sorted into subsets based on content of granule proteases IgE-dependent activation
[15]. MCT cells express tryptases but not chymase, whereas
MCTC express tryptases and chymase. Occasionally, chymase- Classic mast cell and basophil activation involves docking
only MCC mast cells are seen. Bronchi contain a mixture of of allergens to IgE via FcRI expressed as an assemblage of
types, whereas alveolar interstitium contains predominantly subunits (2) in the plasma membrane. FcRI expres-
MCT. Because mast cells develop and are stimulated in tis- sion is strongly inuenced by the serum level of IgE itself
sues, some variation is due to dierences in maturation, acti- [22]. Aeroallergens with repeating epitopes attach to recep-
vation, or recovery from degranulation. tor chain-bound IgE, bridging receptors. Allergen-driven
Less is known of factors inuencing basophil dier- cross-linking initiates intracellular signaling characterized
entiation. c-Kit appears less important than for mast cells initially by phosphorylation of intracellular immunoreceptor
because mature basophils normally express little or no c-kit tyrosine-based activation motif (ITAM) domains of receptor
and because levels are largely unaected by defects in c-kit and chains. In turn, these recruit and activate nonrecep-
causing profound mast cell decits in mice. Nonetheless, tor tyrosine kinases, especially lyn, syk, and btk, which access
c-kit human basophil-like cells circulate in asthma and pathways leading to exocytosis and synthesis of eicosanoids
atopy and manifest phenotypic changes in which c-kit could and cytokines. Some of these activation pathway proteins
play a role [16]. have emerged as targets for new types of anti-allergic drugs

114
Mast Cells and Basophils 9
[23, 24]. For example, inhibition of syk blocks allergic air- bacterial products (e.g. E. coli FimH);
way inammation in mice [25]. Intriguingly, syk protein is chemokines and lymphokines (e.g. IL-4);
decient in some humans with nonreleaser basophils [26],
although the exact relationship between the nonreleaser phe- kit ligand.
notype and the asthma (or lack thereof ) is not yet clear [27]. These inputs are immunologically nonspecic and
Furthermore, dexamethasone depresses syk activity, which provide the means by which products of activation partici-
may contribute corticosteroid ecacy in asthma [28]. The pate in neurogenic inammation and innate host defense
FcRI chain is not essential in humans but amplies the sig- [31]. They also augment responses to allergen-specic mast
nal. FcRI signals are damped by inhibitory receptors, such as cell activation (see below).
FcRII and gp49, which possess intracellular, immunorecep- Thus, innate and adaptive responses are not mutually
tor tyrosine-based inhibition motif (ITIM) domains [29]. exclusive. C3a, an agent of innate immunity, is an example,
Phosphorylated ITIMs attract tyrosine phos- for C3a receptor-null mice are protected from sequelae of
phatases, such as SHIP, which may inhibit Fc RI signaling airway allergen challenge [32].
by dephosphorylating activated proteins in the signaling It should be noted that human mast cell subpopula-
pathway. tions do not respond uniformly to all stimuli. For example,
FcRIIs importance is particularly compelling [30]. lung mast cells tend to be less responsive than skin mast
When IgG and IgE antibodies are raised against polyvalent cells to substance P [33].
antigen, heterotypic cross-linking of FcRII and Fc RI by
allergen bound to IgG and IgE inhibits signaling by Fc RI.
This may be a means by which blocking antibodies reduce
atopic symptoms after allergen desensitization.
PRIMING AND INHIBITION
IgE-independent activation
Priming describes the response to a substance that does not
Mast cells are activated by multiple nonimmunological release mediators by itself but enhances the eect of another
inputs (Fig. 9.1). Physiological activators include: stimulus, such as cross-linked FcRI. In cultured human
mast cells, priming occurs with allergic cytokines, such as
neuropeptides (e.g. substance P); IL-4 and IL-5 [34], and also with kit ligand and adenos-
purines (adenosine and ATP); ine [35]. Mechanisms of priming may be stimulus-specic
and aect mediator synthesis and release in dierent ways.
byproducts of complement activation (e.g. C3a); Interactions between primers are potentially complex, and, in
eosinophil toxins; the case of IL-4 and -5, may involve autocrine stimulation.

Input Output
Antigen-specific /
Adaptive
Histamine
Antigen Proteases
AgIgG
nonspecific/ Heparin Tryptases, tPA
Innate AgIgE Chymases, Cathepsin G
MHC II
Gelatinases A & B
Substance P Carboxypeptidase A
C3a,C5a Dipeptidylpeptidase I
Escherichia coli FimH Cytokines
Endothelin IL-4, IL-13
IL-5, IL-6, TNF-
Kit ligand Chemokines
IL-8, MIP-1
NGF
Growth factors
ATP bFGF, VEGF, NGF
Adenosine CD40L Eicosanoids
PGD2, LTC4
IL-4
IL-5

Mast cell
FIG. 9.1 Mast cell inputs and outputs. This contains a partial listing of factors influencing human mast cell production of mediators with postulated roles in
acute and chronic airway inflammation in asthma. Inputs are divided into those directly involving immunoglobulin-mediated adaptive responses and those
involving immunologically nonspecific innate/natural immunity.

115
Asthma and COPD: Basic Mechanisms and Clinical Management

In basophils, priming in vitro is especially impressive with heparin delivered into airways in pharmacological doses is
IL-3, which augments release of histamine in response to anti-asthmatic, heparin released from stimulated mast cells
MCP-4 and IL-4 in response to allergen, and is enhanced may attenuate inammatory eects of mediators released
by eotaxin [21]. Basophils harvested from allergic asthmat- from mast cells and other eector cells [45].
ics are primed [36], presumably by exposure to cytokines There is little evidence that human basophils express
in vivo. mediators not also present in mast cells. However, proteins
Priming may serve to activate cells when (and only of unknown function (such as basogranulin) are recognized
when) necessary to protect the host; in asthmatics, dys- by monoclonal antibodies and may be basophil specic
regulated priming may contribute to the pathology of aller- [16, 46].
gic inammation. Fc RI-mediated activation can also be
opposed by physiological inuences, including adrenergic
agonists and inhibitory receptors with ITIMs, as discussed Cytokines and chemokines
above. Inhibition of activation is the basis of emerging strat-
egies to combat allergic disease, including use of adenosine Activated human mast cells express cytokines similar to
receptor antagonists, cytokine and chemokine antagonists, those of TH2 cells, including IL-4, -5, -13, -16, and TNF-
and activators of ITIM containing co-receptors [37]. . In airways in allergic rhinitis or asthma, mast cells are
a substantial fraction of leukocytes expressing these TH2
cytokines [47].
The relative importance of mast cells and basophils
MEDIATORS versus lymphocytes as a source of these cytokines is unclear.
However, mast cells dier from lymphocytes in stor-
ing cytokines in secretory granules. In the case of TNF-
Eicosanoids, histamine, and proteases , release from mast cell stores is a critical determinant of
survival from peritonitis [48]. Asthmatic basophils produce
Stimulated mast cells and basophils release an astonishing prodigious amounts of IL-4 and IL-13 after antigen-specic
variety of stored and newly synthesized mediators. These activation [49]. In mice, basophils may be one of the IL-4-
include prostanoids (Chapter 23), leukotrienes (Chapter producing cell types that skew the immune environment
24), proteases, chemokines (Chapter 26), lymphokines, in the lung toward allergic, Th2-type responses [50, 51].
growth factors (Chapter 29), and nitric oxide (Chapter 30), The importance of IL-4 and IL-13 is strongly supported
the properties and pharmacology of which are considered in by studies in genetically modied mice, which suggest that
other cited chapters and are not reviewed extensively here. airway overproduction causes epithelial hypertrophy, mucus
The principal eicosanoids synthesized after stimula- metaplasia, eosinophilic inammation, and hyperrespon-
tion are PGD2 (whose importance in allergic airway inam- siveness. Cytokine-dependent proliferation of mast cells is
mation is demonstrated in PGD receptor-null mice [38]) complex and is subject to autocrine regulation, as from self-
and LTC4, which is the major target of 5-lipoxygenase stimulation by other types of mediators, such as leukotrienes
inhibitors and leukotriene receptor antagonists. The major [52]. Sustained overproduction of allergic cytokines by mast
granule-associated constituents of human mast cells are cell and basophils may heighten and perpetuate asthmatic
histamine, serine proteases, and proteoglycans (heparin and inammation. Chemokines expressed and secreted by mast
chondroitin). Histamines importance is clearer in upper air- cells include IL-8 and MIP-1, which may recruit eector
way allergic disease, because antihistamines tend to be more leukocytes to sites of inammation [53].
eective in treating rhinitis than asthma. Proteases are the
most abundant proteins in secretory granules [39], though
this may not be true of normal basophils. As noted, human Growth factors
mast cells vary in expression of tryptases and chymase. In
atopic and asthmatic individuals, circulating cells resembling Known cellular growth factors expressed by mast cells
hybrids of basophils and mast cells may express one or both include bFGF [54], VEGF, [55] and nerve growth factor
types of protease [16]. [56]. Less known growth factors are tryptases, which are
Human tryptases are a polymorphic family of trypsin- mitogens for airway broblasts, smooth muscle, and epithe-
like proteases implicated in asthma based on in vitro degrada- lial cells [57] and may inuence endothelial cells to form
tion of bronchodilating peptides, enhancement of bronchial vessels, thereby promoting angiogenesis [58]. Mast cell
contraction, promotion of airway smooth muscle and suben- IL-4 is brogenic when presented in the context of cell-
dothelial broblast growth and collagen production, and pro- to-cell contact [59]. These mediators suggest mechanisms
inammatory properties [40]. Studies in sheep and guinea by which mast cell and basophil mediators promote airway
pigs suggest that inhibitors of tryptases reduce allergen- remodeling in the setting of persistent airway inammation.
induced airway inammation and bronchoconstriction [41]. Recent data from asthmatics suggest that inltration of air-
Chymase is postulated to play roles in asthma by way smooth muscle by mast cells (mastitis) is a phenome-
stimulating gland secretion and promoting airway remod- non specic for asthma, which is to say that it is not seen in
eling via production of angiotensin II and activation of related inammatory airway disorders such as eosinophilic
matrix metalloproteinases [39, 42, 43]. Apart from helping bronchitis [(which unlike asthma is not associated with air-
to package and stabilize proteases [44], the role of heparin way hyperresponsiveness (AHR)] and smoking-associated
and other proteoglycans is less clear. However, because bronchitis [60]. Brightling and colleagues propose that

116
Mast Cells and Basophils 9
airway smooth muscle mastocytosis in asthma is caused by a Anti-IgE joins a growing list of anti-asthmatic drugs inu-
unique asthmatic smooth muscle phenotype, which includes encing mast cells, basophils, and their products. These include:
production of mast cell chemoattractants like CXCL10
[61]. In turn, tryptase and other growth factors produced by corticosteroids (which decrease mast cell numbers) [75];
mast cells recruited to airway smooth muscle may promote cromones;
myocyte hyperplasia or hypertrophy [62, 63]. On the other
hand, Wenzel and colleagues report a positive correlation -adrenergic agonists;
between the chymase-positive subset of distal airway mast theophylline;
cells and lung function [64].
heparin (which inhibits degranulation);
IL-4 antagonists (which inhibit priming) (58);
leukotriene pathway inhibitors.
ROLES IN HOMEOSTASIS Others, eective in animal models, include tryptase
inhibitors [76].
Basophils and mast cells are presumed to exist for reasons Interestingly, mast cell desensitization to chronic
other than to promote sneezing, itching, and wheezing. -agonists used by asthmatics without corticosteroids may
Lacking informative animal models or natural deciency contribute to clinical deterioration [77]. Furthermore, corti-
states, investigators have few direct clues regarding normal costeroids may protect mast cells from desensitization [78].
basophil function. However, compelling evidence of roles Pharmacological evidence of roles for mast cells cor-
for mast cells has emerged from studies by using mast cell- relates with studies indicating activation in allergic airway
decient mice, which suggest critical roles in innate as well as disease [1, 79]. Mast cells release histamine and tryptase
adaptive immune responses, including defense against bacterial into bronchi following allergen challenge [80]. They appear
peritonitis and pneumonia [48, 65]. In similar mouse models degranulated in asthmatic airway, even in stable disease, [81,
(reviewed in [5]), mast cells promote a variety of immunologi- 82] and the percentage of mast cells expressing cytokines
cally nonspecic forms of inammation, for example, ozone- IL-4, IL-5, and TNF- increases [47]. Mast cell and
inicted lung injury [66], providing further evidence that they basophil numbers rise in asthmatic airways and correlate
are activated by IgE-independent pathways. with hyperresponsiveness to acetylcholine [83]. However,
fewer basophils are seen in asthmatic bronchial biopsies
than mast cells or eosinophils, and they are more promi-
nent in cutaneous than airway late-phase reactions [84].
Nonetheless, their arrival coincides with development of
ROLES IN ASTHMA late-phase bronchoconstriction [85]. Airway basophils are
thought to be a source of the late histamine release after
allergen exposure, because this occurs without a correspond-
Animal models ing peak in tryptase, which is more abundant in mast cells.
On the other hand, other granulated leukocytes, including
Several experiments in mice support a role for mast cells in polymorphonuclear neutrophils, also can be sources of air-
asthma-like allergic inammation, including eosinophilia, way histamine [86].
hyperresponsiveness, and epithelial remodeling [14, 6770]. More basophils appear in asthmatic sputum with
Other studies suggest that IgE and mast cells do not make late-phase responses to allergen than in those without such
major contributions (reviewed in [71, 72]). Together, these responses, and their numbers correlate with methacholine
studies reveal that IgE- and mast cell-dependence vary with responsiveness [17]. This supports the hypothesis that mast
strain of mouse, choice of antigen, mode of sensitization and cells and basophils are important in early- and late-phase
challenge, and choice of physiological endpoints. Mast cell responses, respectively. The further hypothesis that these
dependence is easier to detect in mice sensitized and chal- cells promote chronic, persistent asthma is speculative.
lenged locally than systemically, using lower (more physi- However, this notion is supported by the studies summa-
ological) amounts of antigen, without adjuvants. Overall, rized above suggesting that proteases, cytokines, and growth
murine studies suggest that mast cells and IgE are not factors from chronically activated mast cells and basophils
essential for development of eosinophilic inammation but promote airway remodeling and sustain TH2-assisted IgE
do inuence kinetics and magnitude of its expression. production and allergic inammation.

Humans
Notwithstanding some studies questioning its importance ROLES IN CHRONIC OBSTRUCTIVE
in mice, IgEs long-suspected contributions to human
asthma are supported by trials by using anti-IgE antibodies,
PULMONARY DISEASE
which reduce circulating IgE to nearly undetectable levels
and decrease symptoms and corticosteroid use in moder- Several lines of evidence suggest connections between mast
ate, steroid-dependent asthmatics, [73] and rhinitics [74]. cells and chronic obstructive pulmonary disease (COPD) [87].

117
Asthma and COPD: Basic Mechanisms and Clinical Management

Elevated levels of histamine and tryptase in smokers lavage Langerhans cells in response to bacterial peptidoglycan. J Immunol 177:
uid [88] imply that mast cells are activated by smoke. Studies 175562, 2006.
in mice suggest that mast cells promote airway injury and epi- 11. Min B, Le Gros G, Paul WE. Basophils: A potential liaison between
innate and adaptive immunity. Allergol Int 55: 99104, 2006.
thelial remodeling in response to ozone [66], which could
12. Li L, Krilis SA. Mast-cell growth and dierentiation. Allergy 54:
explain some human responses to smoke. Most mast cells 30612, 1999.
close to human bronchial glands express chymase [89], which 13. Campbell E, Hogaboam C, Lincoln P, Lukacs NW. Stem cell factor-
stimulates gland cell secretion [42] and may promote sputum induced airway hyperreactivity in allergic and normal mice. Am J Pathol
production in bronchitis. 154: 125965, 1999.
Conceivably, mast cells contribute to emphysema- 14. Temann UA, Geba GP, Rankin JA, Flavell RA. Expression of inter-
tous lung destruction by secreting matrix altering proteases, leukin 9 in the lungs of transgenic mice causes airway inammation,
for example chymase, tryptases, gelatinases, plasminogen mast cell hyperplasia, and bronchial hyperresponsiveness. J Exp Med
activator, and dipeptidyl peptidase I [39]. Some investiga- 188: 130720, 1998.
tors nd increased mast cell numbers in proximal airways 15. Irani AA, Schechter NM, Craig SS et al. Two types of human mast
cells that have distinct neutral protease compositions. Proc Natl Acad
of asymptomatic smokers with mild COPD [90], although
Sci USA 83: 446468, 1986.
the relationship to lung function is less clear. Overall, evi- 16. Li L, Li Y, Reddel SW et al. Identication of basophilic cells that
dence of mast cell involvement in asthma is more compel- express mast cell granule proteases in the peripheral blood of asthma,
ling, although further investigation of connections between allergy, and drug-reactive patients. J Immunol 161: 507986, 1998.
mast cells and COPD is warranted. 17. Gauvreau GM, Lee JM, Watson RM et al. Increased numbers of
both airway basophils and mast cells in sputum after allergen inhala-
tion challenge of atopic asthmatics. Am J Respir Crit Care Med 161:
147378, 2000.
18. Hamawy MM, Mergenhagen SE, Siraganian RP. Adhesion molecules
SUMMARY as regulators of mast-cell and basophil function. Immunol Today 15:
6266, 1994.
19. Uguccioni M, Mackay CR, Ochensberger B et al. High expression of
Mast cells protect from certain types of infection and injury the chemokine receptor CCR3 in human blood basophils. Role in acti-
by contributing to innate and adaptive immune responses. vation by eotaxin, MCP-4, and other chemokines. J Clin Invest 100:
The homeostatic roles of basophils are less clear. Both cell 113743, 1997.
types participate in the pathology of atopy and asthma by 20. Ochensberger B, Tassera L, Bifrare D et al. Regulation of cytokine expres-
deploying an arsenal of inammatory mediators, including sion and leukotriene formation in human basophils by growth factors,
proteases, growth factors, chemokines, and TH2 cytokines. chemokines and chemotactic agonists. Eur J Immunol 29: 1122, 1999.
Mast cells, being permanent airway residents, are more likely 21. Devouassoux G, Metcalfe DD, Prussin C. Eotaxin potentiates antigen-
to encounter aeroallergens rst and to participate in acute dependent basophil IL-4 production. J Immunol 163: 287782, 1999.
22. Saini SS, Klion AD, Holland SM et al. The relationship between serum
responses. Basophils, because of the time lag of recruitment,
IgE and surface levels of FcR on human leukocytes in various diseases:
may be more important in late-phase responses. Correlation of expression with FcepsilonRI on basophils but not on
Support is mounting for the hypothesis that both types monocytes or eosinophils. J Allergy Clin Immunol 106: 51420, 2000.
of cells magnify the pathology of persistent inammation in 23. Oliver JM, Kepley CL, Ortega E, Wilson BS. Immunologically medi-
chronic asthma, including stimulation of IgE production, ated signaling in basophils and mast cells: nding therapeutic targets
recruitment of eosinophils, and remodeling of epithelium. for allergic diseases in the human FcR1 signaling pathway. Immuno-
pharmacology 48: 26981, 2000.
24. Barnes PJ. Novel signal transduction modulators for the treatment of
airway diseases. Pharmacol Ther 109: 23845, 2006.
References 25. Matsubara S, Li G, Takeda K et al. Inhibition of spleen tyrosine kinase
prevents mast cell activation and airway hyperresponsiveness. Am J
1. Kaliner M. Asthma and mast cell activation. J Allergy Clin Immunol 83: Respir Crit Care Med 173: 5663, 2006.
51020, 1989. 26. Kepley CL. Antigen-induced reduction in mast cell and basophil func-
2. Schulman ES. The role of mast cells in inammatory responses in the tional responses due to reduced Syk protein levels. Int Arch Allergy
lung. Crit Rev Immunol 13: 3570, 1993. Immunol 138: 2939, 2005.
3. Metcalfe DD, Baram D, Mekori YA. Mast cells. Physiol Rev 77: 27. Youssef LA, Schuyler M, Gilmartin L et al. Histamine release from the
103379, 1997. basophils of control and asthmatic subjects and a comparison of gene
4. Dvorak AM. Cell biology of the basophil. Int Rev Cytol 180: 87236, expression between releaser and nonreleaser basophils. J Immunol
1998. 178: 458494, 2007.
5. Galli SJ. Mast cells and basophils. Curr Opin Hematol 7: 3239, 2000. 28. Hiragun T, Peng Z, Beaven MA. Cutting edge: dexamethasone nega-
6. Xu X, Zhang D, Lyubynska N et al. Mast cells protect mice from myc- tively regulates Syk in mast cells by up-regulating SRC-like adaptor
oplasma pneumonia. Am J Respir Crit Care Med 173: 21925, 2006. protein. J Immunol 177: 204750, 2006.
7. Metz M, Piliponsky AM, Chen CC et al. Mast cells can enhance 29. Ott VL, Cambier JC. Activating and inhibitory signaling in mast cells:
resistance to snake and honeybee venoms. Science 313: 52630, 2006. New opportunities for therapeutic intervention?. J Allergy Clin Immunol
8. Nakae S, Suto H, Kakurai M et al. Mast cells enhance T cell activation: 106: 42940, 2000.
Importance of mast cell-derived TNF. Proc Natl Acad Sci USA 102: 30. Takai T, Ono M, Hikida M et al. Augmented humoral and anaphylac-
6467472, 2005. tic responses in FcRII-decient mice. Nature 379: 34649, 1996.
9. Lu LF, Lind EF, Gondek DC et al. Mast cells are essential intermedi- 31. Galli SJ, Wershil BK. The two faces of the mast cell. Nature 381: 2122,
aries in regulatory T-cell tolerance. Nature 442: 9971002, 2006. 1996.
10. Jawdat DM, Rowden G, Marshall JS. Mast cells have a pivotal role in 32. Humbles AA, Lu B, Nilsson CA et al. A role for the C3a anaphylatoxin
TNF-independent lymph node hypertrophy and the mobilization of receptor in the eector phase of asthma. Nature 406: 9981001, 2000.

118
Mast Cells and Basophils 9
33. Church MK, Lowman MA, Robinson C et al. Interaction of neuropep- 56. Nilsson G, Forsberg-Nilsson K, Xiang Z et al. Human mast cells
tides with human mast cells. Int Arch Allergy Immunol 88: 7078, 1989. express functional TrkA and are a source of nerve growth factor. Eur J
34. Ochi H, De Jesus NH, Hsieh FH et al. IL-4 and -5 prime human mast Immunol 27: 2295301, 1997.
cells for dierent proles of IgE-dependent cytokine production. Proc 57. Cairns JA, Walls AF. Mast cell tryptase is a mitogen for epithelial cells-
Natl Acad Sci U. S A 97: 1050913, 2000. Stimulation of IL-8 production and intercellular adhesion molecule-1
35. Marquardt DL. Adenosine. In: Barnes PJ, Grunstein MM, Le AR, expression. J Immunol 156: 27583, 1996.
Woolcock AJ (eds). Asthma, Philadelphia: Lippincott-Raven, 1997. 58. Coussens LM, Raymond WW, Bergers G et al. Inammatory mast
36. Lie WJ, Knol EF, Mul FP et al. Basophils from patients with allergic cells upregulate angiogenesis during squamous epithelial carcinogen-
asthma show a primed phenotype. J Allergy Clin Immunol 104: 10007, esis. Genes Dev 13: 138297, 1999.
1999. 59. Trautmann A, Krohne G, Brocker EB, Klein CE. Human mast cells
37. Bingham CO III, Austen KF. Mast-cell responses in the development augment broblast proliferation by heterotypic cell-cell adhesion and
of asthma. J Allergy Clin Immunol 105: S527S534, 2000. action of IL-4. J Immunol 160: 505357, 1998.
38. Matsuoka T, Hirata M, Tanaka H et al. Prostaglandin D2 as a mediator 60. Brightling CE, Bradding P, Symon FA et al. Mast-cell inltration of
of allergic asthma. Science 287: 201317, 2000. airway smooth muscle in asthma. N Engl J Med 346: 1699705, 2000.
39. Caughey GH (ed.), Mast Cell Proteases in Immunology and Biology. New 61. Brightling CE, Ammit AJ, Kaur D et al. The CXCL10/CXCR3 axis
York: Marcel Dekker, Inc, 1995. mediates human lung mast cell migration to asthmatic airway smooth
40. Caughey GH. Of mites and men: Trypsin-like proteases in the lungs. muscle. Am J Respir Crit Care Med 171: 110308, 2005.
Am J Respir Cell Mol Biol 16: 62128, 1997. 62. Brown JK, Jones CA, Rooney LA, Caughey GH. Mast cell tryptase
41. Clark JM, Abraham WM, Fishman CE et al. Tryptase inhibitors block activates extracellular-regulated kinases (p44/p42) in airway smooth-
allergen-induced airway and inammatory responses in allergic sheep. muscle cells: Importance of proteolytic events, time course, and role in
Am J Resp Crit Care Med 152: 207683, 1995. mediating mitogenesis. Am J Respir Cell Mol Biol 24: 14654, 2001.
42. Sommerho CP, Caughey GH, Finkbeiner WE et al. Mast cell chy- 63. Caughey GH. Mast cell tryptases and chymases in inammation and
mase. A potent secretagogue for airway gland serous cells. J Immunol host defense. Immunol Rev 217: 14154, 2007.
142: 245056, 1989. 64. Balzar S, Chu HW, Strand M, Wenzel S. Relationship of small airway
43. Fang KC, Raymond WW, Blount JL, Caughey GH. Dog mast cell chymase-positive mast cells and lung function in severe asthma. Am J
-chymase activates progelatinase B by cleaving the Phe88-Phe89 Respir Crit Care Med 171: 43139, 2005.
and Phe91-Glu92 bonds of the catalytic domain. J Biol Chem 272: 65. Malaviya R, Ikeda T, Ross E, Abraham SN. Mast cell modulation of
2562835, 1997. neutrophil inux and bacterial clearance at sites of infection through
44. Humphries DE, Wong GW, Friend DS et al. Heparin is essential for TNF-. Nature 381: 7780, 1996.
the storage of specic granule proteases in mast cells. Nature 400: 769 66. Longphre M, Zhang LY, Harkema JR, Kleeberger SR. Mast cells con-
72, 1999. tribute to O3-induced epithelial damage and proliferation in nasal and
45. Ahmed T, Syriste T, Mendelssohn R et al. Heparin prevents antigen- bronchial airways of mice. J Appl Physiol 80: 132230, 1996.
induced airway hyperresponsiveness: Interference with IP3-mediated 67. Martin TR, Takeishi T, Katz HR et al. Mast cell activation enhances
mast cell degranulation? J Appl Physiol 76: 893901, 1994. airway responsiveness to methacholine in the mouse. J Clin Invest 91:
46. McEuen AR, Buckley MG, Compton SJ, Walls AF. Development and 117682, 1993.
characterization of a monoclonal antibody specic for human basophils 68. Kung TT, Stelts D, Zurcher JA et al. Mast cells modulate allergic pul-
and the identication of a unique secretory product of basophil activa- monary eosinophilia in mice. Am J Respir Cell Mol Biol 12: 4049, 1995.
tion. Lab Invest 79: 2738, 1999. 69. Kobayashi T, Miura T, Haba T et al. An essential role of mast cells in
47. Bradding P, Roberts JA, Britten KM et al. Interleukin-4, -5, and -6 and the development of airway hyperresponsiveness in a murine asthma
tumor necrosis factor- in normal and asthmatic airways: Evidence for model. J Immunol 164: 385561, 2000.
the human mast cell as a source of these cytokines. Am J Repir Cell Mol 70. Williams CM, Galli SJ. Mast cells can amplify airway reactivity and
Biol 10: 47180, 1994. features of chronic inammation in an asthma model in mice. J Exp
48. Echtenacher B, Mnnel DN, Hltner L. Critical protective role of mast Med 192: 45562, 2000.
cells in a model of acute septic peritonitis. Nature 381: 7577, 1996. 71. Hamelmann E, Tadeda K, Oshiba A, Gelfand EW. Role of IgE in the
49. Devouassoux G, Foster B, Scott LM et al. Frequency and characteri- development of allergic airway inammation and airway hyperrespon-
zation of antigen-specic IL-4- and IL-13- producing basophils and sivenessa murine model. Allergy 54: 297305, 1999.
T cells in peripheral blood of healthy and asthmatic subjects. J Allergy 72. Galli SJ. Commentary: Complexity and redundancy in the pathogen-
Clin Immunol 104: 81119, 1999. esis of asthma: Reassessing the roles of mast cells and T cells. J Exp
50. Min B, Prout M, Hu-Li J et al. Basophils produce IL-4 and accumu- Med 186: 34347, 1997.
late in tissues after infection with a Th2-inducing parasite. J Exp Med 73. Milgrom H, Fick RB Jr., Su JQ et al. Treatment of allergic asthma with
200: 50717, 2004. monoclonal anti-IgE antibody. N Engl J Med 341: 196673, 1999.
51. Voehringer D, Shinkai K, Locksley RM. Type 2 immunity reects 74. Adelroth E, Rak S, Haahtela T et al. Recombinant humanized mAb-
orchestrated recruitment of cells committed to IL-4 production. E25, an anti-IgE mAb, in birch pollen-induced seasonal allergic rhini-
Immunity 20: 26777, 2004. tis. J Allergy Clin Immunol 106: 25359, 2000.
52. Jiang Y, Kanaoka Y, Feng C, Nocka K, Rao S, Boyce JA. Cutting edge: 75. Djukanovic R, Wilson JW, Britten KM et al. Quantitation of mast
Interleukin 4-dependent mast cell proliferation requires autocrine/ cells and eosinophils in the bronchial mucosa of symptomatic atopic
intracrine cysteinyl leukotriene-induced signaling. J Immunol 177: asthmatics and healthy control subjects using immunohistochemistry.
275559, 2006. Am Rev Respir Dis 142: 86371, 1990.
53. Moller A, Lippert U, Lissmann D et al. Human mast cell produce 76. Clark JM, Moore WR, Fishman CE et al. A novel tryptase inhibitor,
IL-8. J Immunol 151: 326166, 1993. APC 366, inhibits allergen-induced airway and inammatory responses
54. Powers MR, Qu Z, LaGesse PC et al. Expression of basic broblast in allergic sheep. Am J Respir Crit Care Med 152: 207683, 1995.
growth factor in nasal polyps. Ann Otol Rhinol Laryngol 107: 89197, 77. Swystun VA, Gordon JR, Davis EB et al. Mast cell tryptase release and
1998. asthmatic responses to allergen increase with regular use of salbutamol.
55. Boesiger J, Tsai M, Maurer M et al. Mast cells can secrete vascular J Allergy Clin Immunol 106: 5764, 2000.
permeability factor/vascular endothelial cell growth factor and exhibit 78. Chong LK, Drury DEJ, Dummer JF et al. Protection by dexametha-
enhanced release after IgE-dependent upregulation of Fc epsilon recep- sone of the functional desensitization to 2-adrenoceptor-mediated
tor I expression. J Exp Med 188: 113545, 1998. responses in human lung mast cells. Br J Pharmacol 121: 71722, 1997.

119
Asthma and COPD: Basic Mechanisms and Clinical Management

79. Bousquet J, Jeery PK, Busse WW et al. Asthma. From bronchocon- 85. Guo CB, Liu MC, Galli SJ et al. Identication of IgE-bearing cells in
striction to airways inammation and remodeling. Am J Respir Crit the late-phase response to antigen in the lung as basophils. Am J Respir
Care Med 161: 172045, 2000. Cell Mol Biol 10: 38490, 1994.
80. Wenzel SE, Fowler A, Schwartz LB. Activation of pulmonary mast 86. Xu X, Zhang D, Zhang H et al. Neutrophil histamine contributes to
cells by bronchoalveolar allergen challenge. In vivo release of hista- inammation in mycoplasma pneumonia. J Exp Med 203: 290717,
mine and tryptase in atopic subjects with and without asthma. Am Rev 2006.
Respir Dis 137: 10028, 1998. 87. Pesci A, Rossi GA, Bertorelli G et al. Mast cells in the airway lumen
81. Beasley R, Roche WR, Roberts JA, Holgate ST. Cellular events in the and bronchial mucosa of patients with chronic bronchitis. Am J Respir
bronchi in mild asthma and after bronchial provocation. Am Rev Respir Crit Care Med 149: 131116, 1994.
Dis 139: 80617, 1989. 88. Kalenderian R, Raju L, Roth W et al. Elevated histamine and tryptase
82. Pesci A, Foresi A, Bertorelli G et al. Histochemical characteristics and levels in smokers bronchoalveolar lavage uid. Do lung mast cells con-
degranulation of mast cells in epithelium and lamina propria of bron- tribute to smokers emphysema? Chest 94: 11319, 1988.
chial biopsies from asthmatic and normal subjects. Am Rev Respir Dis 89. Matin R, Tam EK, Nadel JA, Caughey GH. Distribution of chymase-
147: 68489, 1993. containing mast cells in human bronchi. J Histochem Cytochem 40:
83. Koshino T, Arai Y, Miyamoto Y et al. Airway basophil and mast cell 78186, 1992.
density in patients with bronchial asthma: Relationship to bronchial 90. Ekberg-Jansson A, Amin K, Bake B et al. Bronchial mucosal mast
hyperresponsiveness. J Asthma 33: 8995, 1996. cells in asymptomatic smokers relation to structure, lung function and
84. Macfarlane AJ, Kon OM, Smith SJ et al. Basophils, eosinophils, and emphysema. Respir Med 99: 7583, 2005.
mast cells in atopic and nonatopic asthma and in late-phase allergic
reactions in the lung and skin. J Allergy Clin Immunol 105: 99107, 2000.

120
Dendritic Cells in Asthma and COPD
10
CHAPTER

INTRODUCTION situation and the therapeutic potential of novel Bart N. Lambrecht1,2


ndings will be discussed where possible.
and Guy G. Brusselle2
The lung contains many subsets of dendritic cells 1
Department of Respiratory Medicine,
(DCs) that are distributed in various anatomi- University Hospital Ghent, Belgium
cal compartments. In homeostatic conditions, a LUNG DC SUBSETS IN 2
Department of Pulmonary Medicine,
ne-tuned balance exists between plasmacytoid
and myeloid DCs necessary for maintaining
MOUSE AND MAN Erasmus University Medical Center,
Rotterdam, The Netherlands
tolerance to inhaled antigen and for avoiding
overt inammation. These subsets of DCs also It has long been established that the various
play important roles in establishment of airway lung compartments (conducting airways, lung
inammation seen in asthma and COPD. Based parenchyma, alveolar compartment, pleura) con-
on these new insights on airway DC biology, tain numerous DCs, of which the precise line-
several approaches that interfere with DC func- age or origin have been poorly dened. Recently
tion show potential as new intervention strate- however, many groups have rened the ways
gies for these ever increasing diseases. in which lung DCs should be studied, both in
Obstructive airway disease, broadly mouse [25] and in human [6, 7]. It is clear
divided clinically into asthma or COPD, is a that dierent DC subsets can be found in the
signicant cause of morbidity and mortality. In lung, each with functional specialization. In the
allergic asthma, allergen-specic T-helper type mouse, all of these express the integrin CD11c
2 (Th2) cells produce key cytokines like IL-4, and the subsets are further dened on the basis
IL-5, and IL-13 that regulate the synthesis of of the expression of the myeloid marker CD11b,
allergen-specic IgE and control tissue eosi- as well as anatomical location in the lung. The
nophilic airway inammation and remodeling trachea and large conducting airways have a
of the airways. In the lungs of COPD patients, well-developed network of intraepithelial DCs,
predominantly CD8 lymphocytes and neu- even in steady-state conditions. These cells in
trophilic airway inammation are seen, con- some way resemble skin Langerhans cells,
comitant with remodeling of small airways and and have been shown to express langerin and
the destruction of distal air spaces characteris- CD103 while lacking expression of CD11b [4,
tic of emphysema. It is increasingly clear that 8]. In the submucosa of the conducting airways,
DCs are essential for inducing activation and CD103CD11bCD11c myeloid DCs can be
dierentiation of not only nave but also eec- found, particularly under conditions of inam-
tor CD4 T- and CD8 T-cells in response to mation, and these cells are particularly suited for
inhaled antigen, and it has been well established priming and restimulating eector CD4 cells
that these cells play a pivotal role in the initia- in the lung [8, 9]. The lung interstitium that is
tion and maintenance phase of airway inam- accessible by enzymatic digestion also contains
mation [1]. In this chapter, we will highlight the CD11b and CD11b DCs that access the
recent discoveries in airway DC biology with alveolar lumen and migrate to the mediastinal
special emphasis on mouse models of asthma lymph nodes (MLN) [2, 5, 10]. It should be
and COPD. The applicability to the human noted that this population is contaminated with

121
Asthma and COPD: Basic Mechanisms and Clinical Management

DCs lining the small intrapulmonary bronchioles, as well as and then migrating to the MLN in a CCR7-dependent way
those lining the vessel walls. In the nearby alveolar lumen, [5, 8, 10]. It is still a matter of debate, however, whether the
CD11chi alveolar macrophages control the function of these uptake and transport of inhaled antigen occurs exclusively
interstitial DCs. Plasmacytoid DCs are CD11bCD11cint by alveolar wall DCs, by intraepithelial DCs lining the large
cells expressing SiglecH and bone marrow stromal Ag-1 conducting airways or by both [2, 5]. Another controversial
(recognized by the moAbs mPDCA-1 or 120G8). In the issue is the location and extent by which plasmacytoid DCs
lungs, pDCs are predominantly found in the lung intersti- take up inhaled antigen. Two reports describe that within
tium and produce large amounts of IFN in response to 2448 h following exposure of inhaled uorescently labeled
triggering by CpG motifs or viral infection ex vivo [11]. Ag, almost 5060% of pDCs are antigen-positive [11, 16],
The exact denition of the dierent DC subtypes in whereas another report saw only a minor percentage of Ag
the human lung is incomplete and a subject of controversy. uptake in this subset [5]. It remains to be demonstrated
Two groups identied myeloid DC and plasmacytoid DC if pDCs take up antigen in the periphery of the lung and
in single cell suspensions from digested human lung tis- subsequently migrate, whether they get their antigen from
sue by ow cytometry, using the blood dendritic cell anti- another migratory DC [17], or whether they take up free
gen markers (BDCA) 1 through 3 [7, 12]. The Cluster of aerent lymph while residing in the lymph node. How
Dierentiation (CD) nomenclature of these BDCA mark- much antigen crosses the epithelial barrier passively in the
ers is as follows: BDCA1 CD1c, BDCA2 CD303, and absence of DC uptake is unknown, but it heavily depends
BDCA3 CD141. Within the low autouorescent mono- on the molecular weight of the Ag, its dose, as well as the
nuclear cells of the lung digests, cells that were positive potential to disrupt epithelial tight junctions. Control of
for the lineage markers, that is, CD3, CD19, CD20, and epithelial barrier function could be under important genetic
CD56, were excluded. Within the lineage negative pulmo- control as well, as many of the gene polymorphisms associ-
nary mononuclear cells, myeloid DC type 1 were character- ated with atopy in humans control epithelial integrity (e.g.
ized as CD1c/HLA-DR cells, myeloid DC type 2 were Spink5, S100 family). It is similarly possible that Ag uptake
characterized as CD141/HLA-DR cells, and plasmacy- by lung pDCs would be facilitated by the presence of Ag-
toid DC were characterized as CD303/CD123 cells. In specic immunoglobulins acting on Fc receptors, thus
vitro studies suggest that Langerhans type DC (that are enhancing endocytosis [18].
Langerin, CD1a, and Birbeck granule positive) derive
from myeloid DC precursors that are CD1c and CD1a
[13], and others have shown that Langerhans-type DC can
be generated from monocytes under the inuence of inter-
leukin-15 [14]. It is unclear whether dierences in terms of ROLE OF DCS IN ASTHMA
expression of surface markers reect separate stages of dif-
ferentiation of the DC, rather than dierent sublineages. Outcome of antigen inhalation depends
It is tempting to speculate that, within these DC subsets,
carrying dierent surface markers, functional specialization on the functional state of myeloid and
does occur. Indeed, pulmonary myeloid DC types 1 and 2 plasmacytoid DCs
were shown to release proinammatory cytokines (TNF-,
IL-1, IL-6, and IL-8) in response to Toll-like receptor-2 The usual outcome of inhalation of harmless protein antigen
(TLR-2) and TLR-4 ligands, whereas plasmacytoid DC in the lungs is immunological tolerance (see Fig. 10.1 for a
released high amounts of interferon- in response to the model depicting cellular interactions). As a result, when the
TLR-9 ligand CpG oligonucleotides [6]. Moreover, mye- antigen is subsequently given to mice in an adjuvant setting
loid DC type 1 was shown to be strong inducers of T-cell (e.g. in combination with the Th2 adjuvant alum) it no longer
proliferation in a mixed leukocyte reaction, while plasmacy- induces an immunological response that leads to eector
toid DC induced little T-cell proliferation and myeloid DC cells causing inammation [11, 19]. Inhalational tolerance is
type 2 had an intermediate T-cell-stimulatory capacity [6]. mediated in part by deletion of Ag reactive T-cells as well as
How, where, and by which DC subset inhaled anti- induction and/or expansion of regulatory T-cells in the medi-
gen is sampled from the airway lumen has been a matter astinal nodes [17, 1921]. The latter type of tolerance is domi-
of debate. Jahnsen demonstrated that, analogous to that nant and can be transferred to other mice by adoptive transfer.
reported in the gut, a subset of rat airway intraepithelial Induction of tolerance to inhaled antigen is a function of lung
DCs extend their processes into the airway lumen. This DC subsets that migrate from the lung in a CCR7-dependent
periscope up function is constitutively expressed within way [17]. It is often claimed that induction of tolerance is a
the airway mucosal DC population, providing a mechanism function of immature DCs, meaning that these cells lack the
for continuous immune surveillance of the airway lumi- expression of high levels of major histocompatibility complex
nal surface in the absence of danger signals [15]. In the (MHC), adhesion, and co-stimulatory molecules, However,
mouse, CD103CD11b intraepithelial DCs express the Reis and Sousa recently argued that the term mature DC
tight junction proteins claudin-1, claudin-7, and zonula-2, should be reserved for those DCs that have the potential to
allowing the sampling of airway luminal contents whereas generate eector T-cells, and that expression of costimulatory
keeping the epithelial barrier function intact [4]. This subset molecules by DCs does not exclude the possibility that toler-
is also found in the alveolar septa, and DCs lining the alve- ance would be induced [22]. In the lungs, inhaled tolerance is
olar wall can take up inhaled harmless ovalbumin (OVA) or dependent on signals delivered by CD86 and/or ICOS-L on
bacterial anthrax spores by forming intra-alveolar extensions DCs, supporting this view [20].

122
Dendritic Cells in Asthma and COPD 10
Microbial
cofactors

Complement Other baseline


Harmless activation agonist
antigens

C5aR
Partially mature
? CD80/86/ICOSL
Abortive proliferation
MARCO Treg formation
SR-AI/II
mDC Naive
Cox-2-derived T
Mph
PGs Negative
signal

Treg
Immature
pDC high PDL-1
2,3-IDO
Negative
signal X
GITR Treg
CTLA4

FIG. 10.1 Dendritic cell function following inhalation of harmless antigen in homeostatic conditions. When an inhaled antigen reaches the deeper airways
and it is not cleared by mucociliary transport or by macrophages, it will be taken up by airway DCs. Both mDCs and pDCs take up antigen. mDCs seem to
do this in the periphery, whereas pDCs only do this in the mediastinal nodes. In baseline conditions, the mDCs that reach the nodes are only partly mature
and the T-cell response that they induce is characterized mainly by division but not by differentiation to effector cells. Eventually many dividing cells die.
Additionally, mDCs induce Treg cells that suppress inflammation. At the same time, the pDCs control the level of activity of the mDCs so that these cells are
kept in a quiescent state. This function of pDCs is controlled by Treg control occurring via GITRGIRL ligand and CLTA4CD80/86 interactions. The signals
involved are not precisely known but could involve IDO, or some surface expressed ligand on pDCs (programmed death ligand-1, PDL1). The function of
mDCs is constantly kept in check among others by tonic inhibition by cyclooxygenase-2 (Cox-2) derived prostaglandins (PGs), as well as by complement
activation acting on the C5a receptor (C5a). The precise ligand for the class A scavenger receptors (MARCO and SR-AI/II) are not known.

Conventional lung DCs (either CD11b or CD11b) When pDCs are depleted from the lungs, inhaled tol-
are necessary for tolerance induction [17] but are also erance is abolished, and consequently pDC/mDC balance
responsible for inducing Th2 sensitization, providing there is in the lung is tightly regulated among others by the cytokine
some form of activation (either LPS or TNF) [23] leading osteopontin as well as complement C5a [11, 16, 28, 29].
to functional DC dierentiation and their capacity to prime How exactly pDC depletion leads to sensitization is still
Th2 eector cells (see Fig. 10.2 for a model depicting cellular unsolved, but in vitro and in vivo data suggest that pDCs
interactions leading to Th2 sensitization in the airways). In directly suppress the potential of mDCs to generate eector
further support, Th2 sensitization can be induced by adop- T-cells [11, 29]. Plasmacytoid DCs can also stimulate the
tive intratracheal transfer of GM-CSF-cultured bone mar- formation of Treg cells, possibly in an ICOS-L-dependent
row DCs, most closely resembling mature monocyte-derived way [11, 30]. Tregs expressing GITR could also induce
CD11b DCs, but not by Flt3L-cultured bone marrow- the production of the tryptophan catabolizing enzyme
derived DCs that more resemble the immature steady-state indoleamine 2,3-dioxygenase (IDO) through reverse sign-
DCs resident in the lymph nodes and spleen [11]. As acti- aling in pDCs [31]. In mice depleted of pDCs, there was
vation of lung DCs is the common event leading to Th2 endogenous release of extracellular adenosine triphosphate
sensitization, it is likely that under homeostatic conditions, (ATP) responsible for inducing mDC maturation and
the degree of DC maturation is, therefore, constantly kept in Th2 skewing potential. Th2 sensitization to inhaled OVA
check. One such pathway of tonic DC suppression seems to was abolished when ATP signaling was blocked using the
involve COX-2-derived prostaglandins or their metabolites, broad spectrum P2X and P2Y receptor antagonist suramin.
most likely derived from nearby alveolar macrophages [24]. On the contrary, a non-degradable form of ATP was able
Chimeric mice in which the PGD2 receptor DP1 was selec- to break inhalation tolerance to OVA [32]. At present, it
tively deleted on hematopoietic cells, demonstrated sponta- is unclear how purinergic receptor triggering on DCs pro-
neous maturation of lung mDCs and subsequently, response motes Th2 development, but it could involve the formation
to harmless antigen was greatly enhanced, suggesting tonic of the inammasome, a multi-protein complex that leads to
inhibition of DC function by PGD2 in the lung [25]. When activation of caspase 1 and processing and release of IL-1,
exposed to selective PGD2 agonists, myeloid DCs induced IL-18, and possibly IL-33. One possibility is also that ATP
the formation of Foxp3 Ag-specic Tregs that subsequently indirectly inuences DC function via the modication of
suppressed airway inammation. A similar mechanism on mast cell and eosinophil function. Mast cells (e.g. through
DCs was found for stable PGI2 analogs [26, 27]. release of cytokines, PGD2 or sphingosine metabolites) [33]

123
Asthma and COPD: Basic Mechanisms and Clinical Management

Epithelial activation:
TLR agonist
Proteolytic allergen
Diesel, cigarette smoke

TSLP
GM-CSF
IL1
Inducing mDC maturation:
Proteolytic allergens
Low dose TLR agonist
Loss of Treg control (Run 3/) Mature
CCR4
Alum/diesel adjuvant high CD80/86
Th2 effectors
Viral infection CCL17/CCL22
Interleukin 17?
Endogenous danger signals
(ATP, TNF-, HMGB1 ?, uric acid ?)

mDC Naive
T
mDC

Mature
high CD80/86
pDC
Increasing mDC/pDC balance: pos
GM-CSF overexpression signal?
C5aR blockade IFN-
Osteopontin Loss of pDC tolerogenic function:
Cholera toxin Viral infection
Artificial injection of mDCs Injection of pDC depleting antibody
Osteopontin

FIG. 10.2 Model of dendritic cell function during Th2 sensitization. Several known risk factors for atopy have been shown to interfere with DC function in
the airways. Also, several experimental models have been developed in which sensitization occurs even after inhalation of harmless antigens to the lung,
providing there is some form of DC activation. In these models, respiratory tolerance is broken. Some models have induced a shift in the pDC/mDC balance,
and consequently mDCs induce priming because they are no longer suppressed by adequate numbers of pDCs. Activated mDCs also produce chemokines
like CCL17 or CCL22 to further attract Th2 cells into the response. Some adjuvants induce proper activation of mDCs (yet not sufficient to induce IL-12)
so that they now induce effector Th2 cells rather than regulatory T-cells. Some stimuli, like concomitant viral infection, might have an additional effect by
inducing maturation of pDCs and their production of IFN. This is a known maturation stimulus for mDCs and in this way, these cells might even contribute
to sensitization upon viral infections. Activation of epithelial cells by proteolytic allergens, virus infection, TLR ligands, or air pollutants is an indirect way of
activating and polarizing the DC network, through release of thymic stromal lymphopoeitin (TSLP) or granulocyte-macrophage colony stimulating factor
(GM-CSF) or interleukin 1 (IL-1). The precise source and role of endogenous danger signals such as ATP, tumor necrosis factor (TNF-), high mobility group
box 1 (HMGB1), or uric acid is currently being investigated.

and eosinophils (through release of leukotrienes and eosi- pDCs or whether its eects are mediated indirectly through
nophil-specic enzymes) also inuence DC function [34]. modication of any of the above interactions. For systemically
The conditions regulating ATP release in the lungs will have administered TLR agonists, like endotoxin, the activation
to be studied more carefully before we can conclude how of DCs occurs mainly through direct recognition by TLR4
important the pathway of purinergic signaling is in sensiti- expressed on the DC, but in epithelia, the response could be
zation to more common allergens, like house-dust mite. clearly dierent [35]. As an example, low dose endotoxin was
able to break inhalation tolerance to inhaled OVA by induc-
ing mDC maturation [23]. These eects could be mediated
Direct or indirect mechanisms of Th2 directly via TLR and Myd88-dependent pathways in DCs
sensitization to inhaled antigen but could also be mediated via TLRs on bronchial epithelial
cells [36]. Bronchial epithelial cells could produce chemok-
Induction of tolerance or immunity to inhaled antigen by DCs ines as well as crucial growth and dierentiation factors that
is tightly controlled by signals from alveolar macrophages, subsequently attract, activate, and polarize lung DCs to prime
Tregs, NKT cells, complement activation, nervous system Th2 responses. In this regard, the epithelial cytokines thymic
interactions, and epithelial activation. While studying the stromal lymphopoeitin (TSLP) and GM-CSF might be cru-
literature on particular substances that can break inhala- cial, as their overexpression in the lungs breaks inhalational
tion tolerance and induce Th2 priming, one needs to won- tolerance [37, 38]. On the contrary, neutralization of these
der, therefore, whether a stimulus acts directly on mDCs or cytokines, during priming regimens, eliminates much of the

124
Dendritic Cells in Asthma and COPD 10
adjuvant eects of diesel exhaust particles (DEP) [39, 40] or are given [50]. Increased levels of class A scavenger receptors
pro-allergic eects of house-dust mite [41]. Importantly, the (MARCO and SR-AI/II) were found in the lungs of asth-
production of these cytokines by bronchial epithelial cells in matic mice, possibly suppressing DC-driven inammation.
response to these triggers might be genetically regulated and These receptors are expressed on lung macrophages, DCs, and
this could be the explanation why some individuals become basophils. Receptor-decient mice had more eosinophilic air-
primed to inhaled antigen under the right environmental way inammation, airway hyperresponsiveness (AHR), and
exposure [42]. Under some conditions, predictions about sen- increased migration of DCs to the MLN [52].
sitizers or adjuvants can be made from in vitro experiments. The role of mDCs in the secondary immune response
Ambient particulate matter (APM) is ubiquitous in the envi- was further supported by the fact that their depletion at
ronment and is associated with allergic diseases in inner cities. the time of allergen challenge abrogated all the features of
In vitro, inhaled APM can act directly on human DCs as a asthma, including airway inammation, goblet cell hyperpla-
danger signal to direct a proallergic pattern of innate immune sia, and BHR [9]. Again the defect was restored by intrat-
activation, thus explaining why it acts as an adjuvant [43]. racheal injection of GM-CSF cultured CD11bCD11c
Likewise, DEP induce maturation of human DCs indirectly, mDCs, most closely resembling monocyte-derived inam-
via promoting GM-CSF production in bronchial epithelial matory DCs. The same eects were observed when DCs
cells in vitro [40]. In mice, DEP and APM induce altered were depleted in the nose in an animal model for allergic
DC maturation directly, via nuclear factor-erythroid 2 (NF- rhinitis [53]. It therefore seems that inammatory DCs
E2)-related factor 2-mediated signaling, implicating oxidative are both necessary and sucient for secondary immune
stress in the activation of DCs [44]. Whether enhancement responses to allergen. Upon allergen challenge, lung
of inammation in Nrf-2-decient mice, which are hyper- DCs upregulate the expression of CD40, CD80, CD86,
sensitive to oxidative stress, is also the result of overzealous ICOS-L, PD-L1, and PD-L2, particularly upon contact
DC activation remains to be shown [45]. Another known with Th2 cells [9, 11, 48, 49, 54]. Costimulatory molecules
sensitizer is cigarette smoke. When given concomitantly with might be involved in activation of eector T-cells in the tis-
harmless OVA, it induces Th2 responses, and this response sues or in regulation of Treg activity. In allergen-challenged
is associated with enhanced DC maturation and migration mice, DCs might also be a prominent source of the inam-
[46]. DCs developed in a nicotinic environment (nicDCs) matory chemokines CCL17 and CCL22, involved in
fail to support the terminal development of eector memory attracting CCR4 Th2 cells to the airways, and in produc-
Th1 cells due to their dierential expression of costimulatory ing eosinophil-selective chemokines [29, 49]. In helminth
molecules and lack of IL-12 production. In both human and infections, recruitment of Th2 cells and eosinophils depends
mouse, nicDCs promoted the development of Th2 responses on an IL4/IL13-responsive bone marrow-derived cell, most
[47]. As maternal cigarette smoking is a solid risk factor for likely a DC or alternatively an activated macrophage pop-
becoming sensitized in early life, it will be important to eluci- ulation [55]. A number of cytokines and innate immune
date how it leads to DC activation (e.g. whether any indirect response elements control the production of these chemok-
mechanisms acting via epithelial TLR4 contribute), as this ines. The pro-allergic cytokine TSLP induces the production
might provide novel intervention strategies. Another unsolved of large amounts of CCL17 by mDCs, thus contributing to
question is how the nearby nervous endings react to viruses the recruitment of Th2 cells to the airways, explaining how
or air pollution and how this could aect the way DCs react it may act to enhance inammation [37]. The complement
to inhaled allergen. In this regard, the remodeling that occurs factor C5a suppresses the production of CCL17 and
in the airway unmyelinated nerves following RSV virus could CCL22 [29]. A similar eect was seen with the cytokine
partly explain the subsequent enhanced risk of Th2 sensitiza- IL-17, explaining how it may suppress allergic inamma-
tion in mice. tion when given during allergen challenge. In vitro, IL-17
reduced CCL17 production and antigen uptake by DCs and
IL-5 and IL-13 production in regional lymph nodes in vivo.
Furthermore, IL-17 is regulated in an IL-4-dependent man-
Function of DCs in allergic inflammation ner as mice decient for IL-4R signaling showed a marked
and tissue remodeling increase in IL-17 concentration with inhibited eosinophil
recruitment [56]. Emerging evidence suggests that IL4R
Not only do DCs play a role in the primary immune response expression on lung DCs is an important feedback mecha-
to inhaled allergens, they are also crucial during the eector nism through which IL-4 producing cells (eector Th2 cells,
phase of asthma. The number of CD11b DCs is increased in eosinophils, basophils) might promote further Th2 polariza-
the conducting airways and lung interstitium of sensitized and tion in ongoing responses [57].
challenged mice during the acute phase of the response [9, As the number and activation status of lung CD11b
48, 49]. However, during the chronic phase of the pulmonary DCs during the secondary challenge seems critical for con-
response, induced by prolonged exposure to a large number trolling allergic inammation, studying the factors that
of aerosols, respiratory tolerance develops through unclear control recruitment, survival, or egress from the lung dur-
mechanisms. During this regulatory phase, the number of ing allergic inammation will be important, as this might
mDCs as well as their costimulatory molecules in the lungs reveal therapeutic targets. In an elegant study using mixed
steadily decreased, and this was associated with a reduction bone marrow chimeras in which half the hematopoietic
of bronchial hyperresponsiveness (BHR), possibly mediated cells were CCR2/ and half were CCR2/, it was shown
by the action of Treg cells [19, 50, 51]. Inammation how- by Robays et al. that CCR2 (and not CCR5 or CCR6) is
ever reappears when mature inammatory CD11b DCs crucial for releasing DC precursors from the bone marrow

125
Asthma and COPD: Basic Mechanisms and Clinical Management

and attracting them into allergically inamed lung. This cigarette smoke and by activated cells of the innate immune
was unexpected, as CCR6 is generally seen as the chemok- system, causing tissue damage), disturbance in the protease
ine receptor attracting immature DCs into peripheral tis- antiprotease balance (with an elevated production of proteases
sues [58]. Lung mDCs use CCR7 ligands and CCR8 for and/or decreased levels of antiproteases such as 1-anti-
emigration to the draining lymph nodes but not the leuko- trypsin and tissue inhibitors of matrixmetalloprotenases
triene C4 transporter multidrug-related protein-1 as they (TIMP)), increased programmed cell death and probrotic con-
do in the skin [59]. Unexpectedly, disruption of CCR7- ditions in the small airways. The exact pathogenetic mecha-
selective chemokines in paucity of lymphocyte T-cell (plt) nisms for ongoing pulmonary inammation and damage in
mutant mice, decient in CCL21 and CCL19, resulted in COPD, even after the initial inciting agent has disappeared
airway inammation and Th2 activity that were enhanced (i.e. after smoking cessation), are poorly understood. Latent
[60]. Still, increased numbers of mDCs could be found in viral respiratory infections, chronic bacterial colonization of
the draining lymph nodes of these mice. So, in addition to the lower airways, repetitive infectious exacerbations, auto-
CCR7 ligands, there are other factors involved in the migra- immune responses against changed epitopes in the lung,
tion of DCs to the draining LN, including other chemokine and genetic predisposition, are proposed as important driv-
receptors [59]. Eicosanoid lipid mediators like prostaglan- ing mechanisms for the persistent inammation in patients
dins and leukotrienes can also inuence the migration of with COPD [72].
lung DCs [61]. Leukotriene LTB4 promoted the migration The DCs present in the human lung and lymphoid
of immature and mature skin DCs but these eects seem to organs, linking innate and adaptive immune responses,
be indirect [62]. It will be important to study if well-known could be a key element in the pathogenesis of COPD. In the
inducers of LTB4 in the lungs, such as the environmen- remaining part of the chapter, we will discuss what is known
tal biopolymer chitin, derived from fungi, helminthes, and about the eects of cigarette smoke on DCs and how DC
insects, also induce DC migration [63]. Additional drugga- populations are altered in healthy smokers (without airway
ble factors promoting the migration of DCs to the drain- obstruction) and patients with COPD. We will also formu-
ing mediastinal nodes during inammatory responses could late several hypotheses about the role of DC in the patho-
be sphingosine-1-P and extracellular ATP [32, 64]. genesis of COPD.
In humans, allergen challenge leads to an accumula-
tion of myeloid, but not plasmacytoid DCs in the airways
of asthmatics, concomitant with a reduction in circulating DC populations in lungs of smokers and
CD11c cells, showing that these cells are recruited from COPD patients
the bloodstream in response to allergen challenge [65, 66]. A
recent report suggests that pDCs are also recruited into the There have been only a few studies addressing the number
bronchoalveolar lavage (BAL) uid but are poor antigen- and distribution of DCs in the lungs of smokers and
presenting cells (APCs) [66]. The exact role of plasmacytoid patients with COPD. Most of these descriptive studies have
DCs in ongoing allergen-specic responses in asthma is a cross-sectional design, so that the sequence of events can-
currently unknown. It was shown that pDCs accumulate in not be established and any evidence for causality is (very)
the nose, but not lungs, of allergen challenged atopics [67]. weak. At rst sight, some data in the literature appear dis-
When pDCs were pulsed with pollen allergens, they were as crepant or even contradictory, but this may be due to dif-
ecient as mDCs in inducing Th2 proliferation and eec- ferences in the area of interest (bronchial biopsies sampling
tor function [68]. Others have suggested, as in the mouse, large airways versus surgical resection specimens sampling
that pDCs might also confer protection against allergic small airways and parenchyma versus BAL sampling the
responses [16]. In children at high risk of developing atopic alveolar lumen) (Fig. 10.3), dierences in the examination
disease, the number of circulating pDCs was reduced. techniques (electron microscopy, owcytometry, or immu-
nohistochemistry) or dierences in the immunohistochemi-
cal markers used to identify and enumerate the DCs. It is
also critical to discriminate between the eects of smoking
ROLE OF DCS IN COPD per se on DC numbers, phenotypic markers, or functions
versus the disease-specic eects of COPD on DC, irre-
spective of the current smoking status (see Fig. 10.3).
The inflmmatory basis of COPD When studying pulmonary DC, it is important to take
into account not only the dierent DC subsets mentioned
COPD is an inammatory disease of the large and small earlier but also the dierent anatomic locations (i.e. distribu-
airways and the lung parenchyma, which is caused most tion) of DC within the lung. Moreover, the dierent com-
commonly due to the inhalation of noxious particles and partments of the lungs (large airways, small airways, lung
gases and is associated with an abnormal systemic inam- parenchyma interstitium and alveolar lumen) are sampled
matory response. The lungs of COPD patients are inltrated by dierent methods (bronchoscopy, surgical lung resec-
with cells of the innate immune system such as neutrophils tions, and BAL, respectively). In the large airways (trachea,
and macrophages [69], but there is also evidence for an bronchi), sampled by bronchoscopy with bronchial biopsies,
activated adaptive immune response with accumulation of the number of CD1a DC was evaluated in healthy smok-
CD8 T-cells, Bcells, and the presence of lymphoid fol- ing controls and current smoking COPD patients, showing
licles [70, 71]. Four key elements appear to be crucial in the no signicant dierences between groups [73, 74]. Others
pathogenesis of COPD: increased oxidative stress (caused by evaluated the number of DC in large airways using electron

126
Dendritic Cells in Asthma and COPD 10
never-smokers and healthy-smokers (without COPD), sug-
Cigarette smoke COPD gesting an accumulation of Langerhans-type DC in COPD
[77]. Moreover, the number of Langerin DC further
increased with the severity of the disease [77].
EM defined DC CD1a DC Studies sampling BAL uid evaluated the number of
(Rogers et al., 2008) (Verhoeven et al., 2002)
DCs between never-smokers and smokers without COPD,
Large showing a signicant increase in the expression of Langerin
airway
and CD1a (markers of Langerhans cells) on myeloid DC
[78] as well as an increase in Birbeck granule positive
Langerhans-type DC in smokers versus non-smokers [79].
Finally, in the alveolar parenchyma, the number of CD1a
DC was increased in smokers versus never-smokers, whereas
the number of CD1c DC was not dierent [76].
Taken together, evidence points toward an accumulation
of myeloid DC with Langerhans-type cell markers (CD207,
CD1a, and Birbeck granules) in the small airways and alveoli
of smokers and COPD patients. This is in agreement with
CD1a DC Small data from experimental models of COPD, in which mice are
CD1c DC airway Langerin DC chronically exposed to cigarette smoke and develop manifest
(Soler et al., 1989) (Demedts et al., 2007)
pulmonary inammation and emphysema. A clear accumula-
Langerin DC tion of myeloid DC was seen in the BAL uid and lungs of
CD1a DC these mice [80], exposed to relatively high doses of cigarette
CD1c DC smoke (with carboxyhemoglobin levels in serum compara-
Alveolus
(Bratke et al., 2008) ble to the levels obtained in human smokers who smoke 20
Birbeck DC cigarettes a day). However, exposing the mice to a lower dose
(Casolaro et al., 1988)
of cigarette smoke appears to decrease the number of DCs in
CD1a DC the lung in the absence of inammation [81].
CD1c DC Interstitium
(Soler et al., 1989)

FIG. 10.3 Distribution of DCs in the lungs and the impact of smoking or
Functional differences in DCs exposed to
COPD on different subsets of dendritic cells. Myeloid DCs can be retrieved cigarette smoke
in different compartments of the lung. Airway DCs are located as a network
immediately above and beneath the basement membrane, either in the Evidence from the mouse model of COPD and from
large airways (sampled by bronchial biopsies taken during bronchoscopy) human lung tissue suggests an activated macrophage
or in the small airways (present in lung resection specimens). Alveolar inammatory protein 3 (MIP3alpha)/CC-chemokine
DCs are present in the lumen of alveolar spaces and can be recovered by
ligand (CCL20)CC-chemokine receptor 6 (CCR6)-axis in
BAL, whereas interstitial DCs are located in the interstitium of the lung
parenchyma between alveolar spaces (also present in lung resection
COPD, responsible for the accumulation of myeloid DC in
specimens). The exact location of human plasmacytoid DCs has still to be the lung [77, 82]. At the epithelial surface, DCs are capable
elucidated. On the left side of the figure, studies evaluating the impact of sensing danger signals and take up antigens to process
of cigarette smoking per se on the number of different myeloid DCs are them. However, little is known about the inuence of ciga-
indicated and summarized; on the right side, the studies comparing the rette smoke on the expression and function of innate recep-
number of CD1a DC (in large airways) or Langerin DCs (in small airways) tors of DCs, including TLR and lectin-like receptors (such
between patients with COPD and healthy-smokers without airflow as Langerin and blood DC antigen-2 [83]). Once the DC
limitation are indicated. COPD: chronic obstructive pulmonary disease; has sampled the antigen, it will process the antigen, upregu-
DCs: dendritic cells. late the expression of CC-chemokine receptor 7 (CCR7),
and migrate toward the secondary lymphoid organs (i.e.
MLN) (Fig. 10.4).
Several in vitro studies have evaluated the eect of
microscopy, based upon morphologic criteria derived from nicotine and cigarette smoke extract (CSE) on the matu-
cultured CD1c DC, and showed a signicant decrease in ration process and T-cell stimulatory capacity of myeloid
the number of DCs in the large airways of smoking COPD DC. Human monocyte-derived DCs, which have a pre-
patients versus ex-smoking COPD patients and never smok- dominant interstitial phenotype, show increased maturation
ing controls [75]. and increased IL-10 and IL-12 production upon exposure
In the small airways (bronchioli), which is the main to a high dose nicotine [84], whereas a low dose of nico-
location responsible for airway obstruction in COPD, the tine results in a decreased immature DC-dependent T-cell
number of DC was evaluated using immunohistochemical proliferation and a decreased IL-12 production after LPS
staining for CD1a and CD1c (BDCA-1), showing no dif- stimulation [85]. These nicotine-exposed DCs are indeed
ference between smokers and non-smokers [76]. In patients less able to induce dierentiation of nave T-cells into
with COPD, however, a signicant increase in the number T-helper 1 cells [47, 86]. When CSE is added during the
of CD207 (Langerin) positive cells was found compared to last 18 h of a monocyte-derived DC culture, mimicking the

127
Asthma and COPD: Basic Mechanisms and Clinical Management

Airway epithelium

Alveoli
Antigen
MIP-3 sampling
Immature DC

CCR7
CCR6 CD80
MMP-12 CD28
CD86
Migration to Proteolytic
MHCII TCR
the lung activity Migration to
lymph nodes T-cell co-stimulation

TLR9 TLR2
TLR7 TLR4

Blood DC
precursor
pDC
mDC
IFN- IL-1/TNF-
IL-6/IL-8
Pathogen sensing IL-12 Mature DC

Blood circulation Lymph node

FIG. 10.4 Possible interrelationship between cigarette smoke, airway inflammation, and DCs in the pathogenesis of COPD. Injury to epithelial surfaces by
cigarette smoke and/or oxidative stress induces the release of numerous DC-attracting mediators, including macrophage inflammatory protein-3 (MIP-
3)/CC-chemokine ligand 20 (CCL20), which is the only chemokine ligand of CC-chemokine receptor 6 (CCR6). Blood DC precursors are recruited out of the
blood circulation, and they migrate to the site of antigen entry and tissue damage. Immature DCs display numerous TLR, including TLR2, TLR4, TLR7, and/or
TLR9, which enable myeloid and plasmacytoid DCs to sense multiple pathogen-associated molecular patterns (PAMPs), which may precipitate infectious
exacerbations in patients with COPD. After antigen uptake and activation, DCs lose all chemokine receptors except CC-chemokine receptor 7 (CCR7), which
mediates the migration of DCs toward the secondary lymphoid organs in response to the chemokines CCL19 and CCL21. In the lymph nodes, mature DCs
express high amounts of peptide-loaded MHC molecules and T-cell costimulatory molecules (such as CD40, CD80 and CD86). In BAL fluid, an increased
expression of antigen presentation markers such as CD80 and CD86 was observed on myeloid DCs of smokers compared to never-smokers, whereas the
expression of the lymph node homing receptor CCR7 was significantly decreased. CCL: CC-chemokine ligand; CCR: CC-chemokine receptor; DC(s): dendritic
cell(s); MIP-3: macrophage inflammatory protein-3; MHC: major histocompatibility complex; PAMPs: pathogen associated molecular patterns; TLR: Toll-like
receptors.

exposure to mainstream cigarette smoke, the DC-induced increased expression of maturation markers by myeloid DC
T-cell proliferation is signicantly reduced, with a reduction combined with a decrease of CCR7 expression in smokers
of IL-12 production and an increase of IL-10 production compared to non-smokers [78]. Although in vitro
by the DC. Moreover, the T-cell priming is skewed toward for a specic stimulus cigarette smoke exposed DC might
a T-helper 2 response and the LPS-induced maturation of mature less than non exposed DC, we hypothesize that in
CSE-exposed DC is inhibited, with a reduced upregulation vivo the overall inammatory response and danger signals
of costimulatory molecules and an inhibition of chemotac- due to the damaging eect of cigarette smoke in the lungs
tic CCR7, which is involved in migration of the DC toward will still enhance maturation of DC in smokers and patients
the secondary lymphoid organs. In summary, these in vitro with COPD.
data suggest that CSE impairs the maturation of intersti- Apart from the integration of danger signals and
tial type DC, skews the DC induced T-cell dierentiation antigens, DCs are also capable of producing substantial
toward a Th2 response, and could possibly alter the migra- amounts of proteinases, contributing to the proteinase
tion of DC toward the lymph nodes. In contrast, ex vivo antiproteinase imbalance in COPD. Mouse DCs show
data from human myeloid DC obtained from BAL show an indeed an increase of MMP-12 production upon cigarette

128
Dendritic Cells in Asthma and COPD 10
smoke exposure [87] and could thereby contribute to the A very similar mechanism was described for inhaled iloprost,
pathogenesis of emphysema. a prostacyclin analog acting on the IP receptor expressed
Further research evaluating the eects of CSE on the by lung DCs [27]. Extracellular ATP might be released by
dierentiation of DC and on the maturation of Langerhans- platelets upon allergen challenge. Neutralization of ATP
type DC compared to interstitial type DC is mandatory to via administration of the enzyme apyrase or the broad spec-
understand the role of the DC in smoking-related diseases. trum P2 receptor antagonist suramin reduced all the cardi-
Ideally, longitudinal studies sampling the large and small nal features of asthma by interfering with DC function [32].
airways should be performed in never-smokers, smokers A specic small molecule compound (VAF347) that blocks
without COPD, and patients with COPD to enumerate the function of B-cells and DCs was also shown to be eec-
the dierent DC subsets. Importantly, healthy-smokers and tive in suppressing allergic airway inammation in a mouse
smoking COPD patients should be studied longitudinally model of asthma [89]. Finally, specic inhibitors of syk
before and after smoking cessation, to be able to discern the kinase were shown to suppress DC function and eliminate
eects of smoking per se versus the disease-specic eects established inammation [90].
of COPD on DC numbers, phenotypic markers, and func-
tions. Moreover, the role of the plasmacytoid DC in COPD
has to be elucidated, both during stable disease and at exac-
erbations. As plasmacytoid DC have important antiviral CONCLUSION
and tolerogenic properties, they could play a crucial role in
the pathogenesis of COPD. In addition, impaired matu-
rational response of DC upon cigarette smoke exposure Our understanding of DC biology in the airways has grown
could lead to an increased susceptibility to viral and bacte- considerably. The concept that dierent subtypes of DCs
rial infections, contributing to the increased incidence of perform dierent functions not only during sensitization
lower respiratory tract infections (including pneumonia and but also during established inammation is a theme that
tuberculosis) in smokers and of infectious exacerbations in will persist in the coming years. Slowly, therapeutic strate-
patients with COPD. gies are emerging from these basic studies in animal models
of asthma and COPD, which can eventually reach clinical
application. However, detailed knowledge of DC biology in
human airways is still lacking.
DCS AS DRUG TARGETS IN OBSTRUCTIVE
AIRWAY DISEASES References

If DCs are crucial in mounting immune responses during 1. Lambrecht BN, Hammad H. Taking our breath away: Dendritic cells
ongoing inammation in the lung, then interfering with in the pathogenesis of asthma. Nat Rev Immunol 3: 9941003, 2003.
2. von Garnier C, Filgueira L, Wikstrom M, Smith M, Thomas JA,
their function could constitute a novel form of treatment
Strickland DH, Holt PG, Stumbles PA. Anatomical location deter-
for obstructive airway diseases. Additionally, pharmacologi- mines the distribution and function of dendritic cells and other APCs
cal modication of DCs might fundamentally reset the bal- in the respiratory tract. J Immunol 175: 160918, 2005.
ance of the pro-inammatory immune response in favor of 3. de Heer HJ, Hammad H, Kool M, Lambrecht BN. Dendritic cell sub-
regulatory T-cells and thus lead to a more long lasting eect sets and immune regulation in the lung. Semin Immunol 17: 295303,
on the natural course of allergic disease as well as progres- 2005.
sion in COPD. Glucocorticosteroids are currently the cor- 4. Sung SS, Fu SM, Rose CE Jr., Gaskin F, Ju ST, Beaty SR. A major
nerstone of anti-inammatory treatment in these diseases. lung CD103 (alphaE)-beta7 integrin-positive epithelial dendritic cell
Inhaled corticosteroids reduce the number of lung and nose population expressing Langerin and tight junction proteins. J Immunol
DCs in patients with allergic asthma and allergic rhinitis 176: 216172, 2006.
5. Wikstrom ME, Stumbles PA. Mouse respiratory tract dendritic cell
[88]. Corticosteroids might also interfere with a GITRL-
subsets and the immunological fate of inhaled antigens. Immunol Cell
driven induction of the enzyme IDO in plasmacytoid DCs, Biol 85: 18288, 2007.
thus broadly suppressing inammation [31]. 6. Demedts IK, Bracke KR, Maes T, Joos GF, Brusselle GG. Dierent
Recently, several other new molecules have surfaced roles for human lung dendritic cell subsets in pulmonary immune
that may alter DC function in allergic inammation and defense mechanisms. Am J Respir Cell Mol Biol 35: 38793, 2006.
thus treat disease. Many of these compounds were rst 7. Masten BJ, Olson GK, Tarleton CA, Rund C, Schuyler M, Mehran R,
discovered by their potential to interfere with DC-driven Archibeque T, Lipscomb MF. Characterization of myeloid and plasma-
Th2 sensitization. The sphingosine-1-P analog FTY720 cytoid dendritic cells in human lung. J Immunol 177: 778493, 2006.
is currently used in clinical trials for multiple sclerosis and 8. del Rio M-L, Rodriguez-Barbosa J-I, Kremmer E, Forster R. CD103-
transplant rejection. When given to the lungs of mice with and CD103 bronchial lymph node dendritic cells are specialized
in presenting and cross-presenting innocuous antigen to CD4 and
established inammation, it strongly reduced inammation
CD8 T cells. J Immunol 178: 686166, 2007.
by suppressing the T-cell stimulatory capacity and migratory 9. van Rijt LS, Jung S, Kleinjan A, Vos N, Willart M, Duez C,
behavior of lung DCs [64]. Also selective agonists of partic- Hoogsteden HC, Lambrecht BN. In vivo depletion of lung CD11c
ular prostaglandin series receptors might suppress DC func- dendritic cells during allergen challenge abrogates the characteristic
tion. The DP1 agonist BW245C strongly suppressed airway features of asthma. J Exp Med 201: 98191, 2005.
inammation and bronchial hyperreactivity when given to 10. Cleret A, Quesnel-Hellmann A, Vallon-Eberhard A, Verrier B, Jung S,
allergic mice by inhibiting the maturation of lung DCs [25]. Vidal D, Mathieu J, Tournier JN. Lung dendritic cells rapidly mediate

129
Asthma and COPD: Basic Mechanisms and Clinical Management

anthrax spore entry through the pulmonary route. J Immunol 178: cardinal features of asthma via inhibition of airway dendritic cell func-
79948001, 2007. tion. J Clin Invest 117: 46472, 2007.
11. De Heer HJ, Hammad H, Soullie T, Hijdra D, Vos N, Willart MA, 28. Oriss TB, Ostroukhova M, Seguin-Devaux C, Dixon-McCarthy B,
Hoogsteden HC, Lambrecht BN. Essential role of lung plasmacytoid Stolz DB, Watkins SC, Pillemer B, Ray P, Ray A. Dynamics of den-
dendritic cells in preventing asthmatic reactions to harmless inhaled dritic cell phenotype and interactions with CD4 T cells in airway
antigen. J Exp Med 200: 8998, 2004. inammation and tolerance. J Immunol 174: 85463, 2005.
12. Demedts IK, Brusselle GG, Vermaelen KY, Pauwels RA. Identication 29. Kohl J, Baelder R, Lewkowich IP, Pandey MK, Hawlisch H, Wang L,
and characterization of human pulmonary dendritic cells. Am J Respir Best J, Herman NS, Sproles AA, Zwirner J et al. A regulatory role for
Cell Mol Biol 32: 17784, 2005. the C5a anaphylatoxin in type 2 immunity in asthma. J Clin Invest 116:
13. Ito T, Inaba M, Inaba K, Toki J, Sogo S, Iguchi T, Adachi Y, Yamaguchi K, 78396, 2006.
Amakawa R, Valladeau J et al. A CD1a/CD11c subset of 30. Ito T, Yang M, Wang YH, Lande R, Gregorio J, Perng OA, Qin XF,
human blood dendritic cells is a direct precursor of Langerhans cells. Liu YJ, Gilliet M. Plasmacytoid dendritic cells prime IL-10-producing
J Immunol 163: 140919, 1999. T regulatory cells by inducible costimulator ligand. J Exp Med 204:
14. Mohamadzadeh M, Berard F, Essert G, Chalouni C, Pulendran B, 10515, 2007.
Davoust J, Bridges G, Palucka AK, Banchereau J. Interleukin 15 31. Grohmann U, Volpi C, Fallarino F, Bozza S, Bianchi R, Vacca C,
skews monocyte dierentiation into dendritic cells with features of Orabona C, Belladonna ML, Ayroldi E, Nocentini G et al. Reverse sig-
Langerhans cells. J Exp Med 194: 101320, 200 naling through GITR ligand enables dexamethasone to activate IDO
15. Jahnsen FL, Strickland DH, Thomas JA, Tobagus IT, Napoli S, in allergy. Nat Med 13: 57986, 2007.
Zosky GR, Turner DJ, Sly PD, Stumbles PA, Holt PG. Accelerated 32. Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA,
antigen sampling and transport by airway mucosal dendritic cells fol- Muskens F, Hoogsteden HC, Luttmann W, Ferrari D, Di Virgilio F
lowing inhalation of a bacterial stimulus. J Immunol 177: 586167, 2006. et al. Extracellular ATP triggers and maintains asthmatic airway
16. Xanthou G, Alissa T, Semitekolou M, Simoes DC, Economidou E, inammation by activating dendritic cells. Nat Med 13: 91319, 2007.
Gaga M, Lambrecht BN, Lloyd CM, Panoutsakopoulou V. 33. Bulanova E, Budagian V, Orinska Z, Hein M, Petersen F, Thon L,
Osteopontin has a crucial role in allergic airway disease through regu- Adam D, Bulfone-Paus S. Extracellular ATP induces cytokine expres-
lation of dendritic cell subsets. Nat Med 13: 57078, 2007. sion and apoptosis through P2 7 receptor in murine mast cells.
17. Hintzen G, Ohl L, del Rio ML, Rodriguez-Barbosa JI, Pabst O, J Immunol 174: 388090, 2005.
Kocks JR, Krege J, Hardtke S, Forster R. Induction of tolerance to 34. Idzko M, Panther E, Bremer HC, Sorichter S, Luttmann W,
innocuous inhaled antigen relies on a CCR7-dependent dendritic cell- Virchow CJ Jr., Di Virgilio F, Herouy Y, Norgauer J, Ferrari D.
mediated antigen transport to the bronchial lymph node. J Immunol 177: Stimulation of P2 purinergic receptors induces the release of eosi-
734654, 2006. nophil cationic protein and interleukin-8 from human eosinophils. Br J
18. Benitez-Ribas D, Adema GJ, Winkels G, Klasen IS, Punt CJ, Pharmacol 138: 124450, 2003.
Figdor CG, de Vries IJ. Plasmacytoid dendritic cells of melanoma 35. Nolte MA, Leibundgut-Landmann S, Jo re O, Sousa CR. Dendritic
patients present exogenous proteins to CD4 T cells after Fc gamma cell quiescence during systemic inammation driven by LPS stimula-
RII-mediated uptake. J Exp Med 203: 162935, 2006. tion of radioresistant cells in vivo. J Exp Med 204: 1487501, 2007.
19. Van Hove CL, Maes T, Joos GF, Tournoy KG. Prolonged inhaled 36. Noulin N, Quesniaux VF, Schnyder-Candrian S, Schnyder B,
allergen exposure can induce persistent tolerance. Am J Respir Cell Mol Maillet I, Robert T, Vargaftig BB, Ryel B, Couillin I. Both hemo-
Biol 36: 57384, 2007. poietic and resident cells are required for MyD88-dependent pul-
20. Akbari O, Freeman GJ, Meyer EH, Greeneld EA, Chang TT, monary inammatory response to inhaled endotoxin. J Immunol 175:
Sharpe AH, Berry G, DeKruy RH, Umetsu DT. Antigen-specic 686169, 2005.
regulatory T cells develop via the ICOS-ICOS-ligand pathway and 37. Zhou B, Comeau MR, De Smedt T, Liggitt HD, Dahl ME, Lewis DB,
inhibit allergen-induced airway hyperreactivity. Nat Med 8: 102432, Gyarmati D, Aye T, Campbell DJ, Ziegler SF. Thymic stromal lym-
2002. phopoietin as a key initiator of allergic airway inammation in mice.
21. Ostroukhova M, Seguin-Devaux C, Oriss TB, Dixon-McCarthy B, Nat Immunol 6: 104753, 2005.
Yang L, Ameredes BT, Corcoran TE, Ray A. Tolerance induced by 38. Lee HC, Ziegler SF. Inducible expression of the proallergic cytokine
inhaled antigen involves CD4() T cells expressing membrane-bound thymic stromal lymphopoietin in airway epithelial cells is controlled by
TGF-beta and FOXP3. J Clin Invest 114: 2838, 2004. NFkappaB. Proc Natl Acad Sci USA 104: 91419, 2007.
22. Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol 6: 39. Ohta K, Yamashita N, Tajima M, Miyasaka T, Nakano J, Nakajima M,
47683, 2006. Ishii A, Horiuchi T, Mano K, Miyamoto T. Diesel exhaust particulate
23. Piggott DA, Eisenbarth SC, Xu L, Constant SL, Huleatt JW, induces airway hyperresponsiveness in a murine model: Essential role
Herrick CA, Bottomly K. MyD88-dependent induction of allergic Th2 of GM-CSF. J Allergy Clin Immunol 104: 102430, 1999.
responses to intranasal antigen. J Clin Invest 115: 45967, 2005. 40. Bleck B, Tse DB, Jaspers I, Curotto de Lafaille MA, Reibman J. Diesel
24. Hashimoto K, Sheller JR, Morrow JD, Collins RD, Goleniewska K, exhaust particle-exposed human bronchial epithelial cells induce den-
ONeal J, Zhou W, Ji S, Mitchell DB, Graham BS et al. Cyclooxygenase dritic cell maturation. J Immunol 176: 743137, 2006.
inhibition augments allergic inammation through CD4-dependent, 41. Cates EC, Fattouh R, Wattie J, Inman MD, Goncharova S, Coyle AJ,
STAT6-independent mechanisms. J Immunol 174: 52532, 2005. Gutierrez-Ramos JC, Jordana M. Intranasal exposure of mice to house
25. Hammad H, Kool M, Soullie T, Narumiya S, Trottein F, Hoogsteden HC, dust mite elicits allergic airway inammation via a GM-CSF-mediated
Lambrecht BN. Activation of the D prostanoid 1 receptor suppresses mechanism. J Immunol 173: 638492, 2004.
asthma by modulation of lung dendritic cell function and induction of 42. Holgate ST. The epithelium takes centre stage in asthma and atopic
regulatory T cells. J Exp Med 204: 35767, 2007. dermatitis. Trends Immunol 28: 24851, 2007.
26. Zhou W, Hashimoto K, Goleniewska K, ONeal JF, Ji S, Blackwell TS, 43. Williams MA, Porter M, Horton M, Guo J, Roman J, Williams D,
Fitzgerald GA, Egan KM, Geraci MW, Peebles RS Jr. Prostaglandin I2 Breysse P, Georas SN. Ambient particulate matter directs nonclassic
analogs inhibit proinammatory cytokine production and T cell stimu- dendritic cell activation and a mixed TH1/TH2-like cytokine response
latory function of dendritic cells. J Immunol 178: 70210, 2007. by naive CD4 T cells. J Allergy Clin Immunol 119: 48897, 2007.
27. Idzko M, Hammad H, van Nimwegen M, Kool M, Vos N, 44. Chan RC, Wang M, Li N, Yanagawa Y, Onoe K, Lee JJ, Nel AE.
Hoogsteden HC, Lambrecht BN. Inhaled iloprost suppresses the Pro-oxidative diesel exhaust particle chemicals inhibit LPS-induced

130
Dendritic Cells in Asthma and COPD 10
dendritic cell responses involved in T-helper dierentiation. J Allergy 62. Del Prete A, Shao WH, Mitola S, Santoro G, Sozzani S, Haribabu B.
Clin Immunol 118: 45565, 2006. Regulation of dendritic cell migration and adaptive immune response
45. Rangasamy T, Guo J, Mitzner WA, Roman J, Singh A, Fryer AD, by leukotriene B4 receptors: A role for LTB4 in up-regulation of
Yamamoto M, Kensler TW, Tuder RM, Georas SN et al. Disruption of CCR7 expression and function. Blood 109: 62631, 2007.
Nrf2 enhances susceptibility to severe airway inammation and asthma 63. Reese TA, Liang HE, Tager AM, Luster AD, Van Rooijen N,
in mice. J Exp Med 202: 4759, 2005. Voehringer D, Locksley RM. Chitin induces accumulation in tissue
46. Moerloose KB, Robays LJ, Maes T, Brusselle GG, Tournoy KG, Joos GF. of innate immune cells associated with allergy. Nature 447: 9296,
Cigarette smoke exposure facilitates allergic sensitization in mice. 2007.
Respir Res 7: 49, 2006. 64. Idzko M, Hammad H, van Nimwegen M, Kool M, Muller T, Soullie T,
47. Nouri-Shirazi M, Tinajero R, Guinet E. Nicotine alters the biological Willart MA, Hijdra D, Hoogsteden HC, Lambrecht BN. Local appli-
activities of developing mouse bone marrow-derived dendritic cells cation of FTY720 to the lung abrogates experimental asthma by alter-
(DCs). Immunol Lett 109: 15564, 2007. ing dendritic cell function. J Clin Invest 116: 293544, 2006.
48. Huh JC, Strickland DH, Jahnsen FL, Turner DJ, Thomas JA, Napoli S, 65. Upham JW, Denburg JA, OByrne PM. Rapid response of circulating
Tobagus I, Stumbles PA, Sly PD, Holt PG. Bidirectional Interactions myeloid dendritic cells to inhaled allergen in asthmatic subjects. Clin
between antigen-bearing respiratory tract dendritic cells (DCs) and T Exp Allergy 32: 81823, 2002.
cells precede the late phase reaction in experimental asthma: DC acti- 66. Bratke K, Lommatzsch M, Julius P, Kuepper M, Kleine HD,
vation occurs in the airway mucosa but not in the lung parenchyma. Luttmann W, Christian Virchow J. Dendritic cell subsets in human bron-
J Exp Med 198: 1930, 2003. choalveolar lavage uid after segmental allergen challenge. Thorax 62:
49. Beaty SR, Rose CE Jr., Sung SS. Diverse and potent chemokine pro- 16875, 2007.
duction by lung CD11bhi dendritic cells in homeostasis and in allergic 67. Jahnsen FL, Lund-Johansen F, Dunne JF, Farkas L, Haye R,
lung inammation. J Immunol 178: 188295, 2007. Brandtzaeg P. Experimentally induced recruitment of plasmacytoid
50. Koya T, Kodama T, Takeda K, Miyahara N, Yang ES, Taube C, (CD123high) dendritic cells in human nasal allergy. J Immunol 165:
Joetham A, Park JW, Dakhama A, Gelfand EW. Importance of mye- 406268, 2000.
loid dendritic cells in persistent airway disease after repeated allergen 68. Farkas L, Kvale EO, Johansen FE, Jahnsen FL, Lund-Johansen F.
exposure. Am J Respir Crit Care Med 173: 4255, 2006. Plasmacytoid dendritic cells activate allergen-specic TH2 mem-
51. Lewkowich IP, Herman NS, Schleifer KW, Dance MP, Chen ory cells: Modulation by CpG oligodeoxynucleotides. J Allergy Clin
BL, Dienger KM, Sproles AA, Shah JS, Kohl J, Belkaid Y et al. Immunol 114: 43643, 2004.
CD4  CD25  T cells protect against experimentally induced asthma 69. Lapperre TS, Willems LN, Timens W, Rabe KF, Hiemstra PS,
and alter pulmonary dendritic cell phenotype and function. J Exp Med Postma DS, Sterk PJ. Small airways dysfunction and neutrophilic
202: 154961, 2005. inammation in bronchial biopsies and BAL in COPD. Chest 131:
52. Arredouani MS, Franco F, Imrich A, Fedulov A, Lu X, Perkins D, 5359, 2007.
Soininen R, Tryggvason K, Shapiro SD, Kobzik L. Scavenger receptors 70. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
SR-AI/II and MARCO limit pulmonary dendritic cell migration and Cherniack RM, Rogers RM, Sciurba FC, Coxson HO et al. The nature
allergic airway inammation. J Immunol 178: 591220, 2007. of small-airway obstruction in chronic obstructive pulmonary disease.
53. KleinJan A, Willart M, van Rijt LS, Braunstahl GJ, Leman K, Jung S, N Engl J Med 350: 264553, 2004.
Hoogsteden HC, Lambrecht BN. An essential role for dendritic cells 71. Gosman MM, Willemse BW, Jansen DF, Lapperre TS, van
in human and experimental allergic rhinitis. J Allergy Clin Immunol Schadewijk A, Hiemstra PS, Postma DS, Timens W, Kerstjens HA.
118: 111725, 2006. Increased number of B-cells in bronchial biopsies in COPD. Eur
54. Van Rijt LS, Vos N, Willart M, Kleinjan A, Coyle AJ, Hoogsteden HC, Respir J 27: 6064, 2006.
Lambrecht BN. Essential role of dendritic cell CD80/CD86 costimu- 72. Curtis JL, Freeman CM, Hogg JC. The immunopathogenesis of
lation in the induction, but not reactivation, of TH2 eector responses chronic obstructive pulmonary disease: Insights from recent research.
in a mouse model of asthma. J Allergy Clin Immunol 114: 16673, 2004. Proc Am Thorac Soc 4: 51221, 2007.
55. Voehringer D, Reese TA, Huang X, Shinkai K, Locksley RM. Type 2 73. Hoogsteden HC, Verhoeven GT, Lambrecht BN, Prins JB. Airway
immunity is controlled by IL-4/IL-13 expression in hematopoietic inammation in asthma and chronic obstructive pulmonary disease with
non-eosinophil cells of the innate immune system. J Exp Med 203: special emphasis on the antigen-presenting dendritic cell: Inuence of
143546, 2006. treatment with uticasone propionate. Clin Exp Allergy 29(Suppl 2):
56. Schnyder-Candrian S, Togbe D, Couillin I, Mercier I, Brombacher F, 11624, 1999.
Quesniaux V, Fossiez F, Ryel B, Schnyder B. Interleukin-17 is a negative 74. Verhoeven GT, Hegmans JP, Mulder PG, Bogaard JM, Hoogsteden HC,
regulator of established allergic asthma. J Exp Med 203: 271525, 2006. Prins JB. Eects of uticasone propionate in COPD patients with bron-
57. Webb DC, Cai Y, Matthaei KI, Foster PS. Comparative roles of IL-4, chial hyperresponsiveness. Thorax 57: 694700, 2002.
IL-13, and IL-4Ralpha in dendritic cell maturation and CD4 Th2 75. Rogers AV, Adelroth E, Hattotuwa K, Dewar A, Jeery PK. Bronchial
cell function. J Immunol 178: 21927, 2007. mucosal dendritic cells in smokers and ex-smokers with COPD: An
58. Robays LJ, Maes T, Lebecque S, Lira SA, Kuziel WA, Brusselle GG, electron microscopic study. Thorax 63: 10814, 2008.
Joos GF, Vermaelen KV. Chemokine receptor CCR2 but not CCR5 or 76. Soler P, Moreau F, Basset F, Hance AJ. Cigarette smoking-induced
CCR6 mediates the increase in pulmonary dendritic cells during aller- changes in the number and dierentiated state of pulmonary dendritic
gic airway inammation. J Immunol 178: 530511, 2007. cells/Langerhans cells. Am Rev Respir Dis 139: 1112, 1989.
59. Jakubzick C, Tacke F, Llodra J, van Rooijen N, Randolph GJ. 77. Demedts IK, Bracke KR, Van Pottelberge G, Testelmans D,
Modulation of dendritic cell tracking to and from the airways. Verleden GM, Vermassen FE, Joos GF, Brusselle GG. Accumulation
J Immunol 176: 357884, 2006. of dendritic cells and increased CCL20 levels in the airways of patients
60. Grinnan D, Sung SS, Dougherty JA, Knowles AR, Allen MB, Rose CE with chronic obstructive pulmonary disease. Am J Respir Crit Care Med
3rd., Nakano H, Gunn MD, Fu SM, Rose CE Jr.. Enhanced allergen- 175: 9981005, 2007.
induced airway inammation in paucity of lymph node T cell (plt) 78. Bratke K, Klug M, Bier A, Julius P, Kuepper M, Virchow JC,
mutant mice. J Allergy Clin Immunol 118: 123441, 2006. Lommatzsch M. Function-associated surface molecules on airway den-
61. Hammad H, Lambrecht BN. Lung dendritic cell migration. Adv dritic cells in cigarette smokers. Am J Respir Cell Mol Biol, 38: 65560
Immunol 93: 26578, 2007. 2008.

131
Asthma and COPD: Basic Mechanisms and Clinical Management

79. Casolaro MA, Bernaudin J-F, Saltini C, Ferrans VJ, Crystal RG. 85. Nouri-Shirazi M, Guinet E. Evidence for the immunosuppressive
Accumulation of Langerhans cells on the epithelial surface of the role of nicotine on human dendritic cell functions. Immunology 109:
lower respiratory tract in normal subjects in association with cigarette 36573, 2003.
smoking. Am Rev Respir Dis 137: 40611, 1988. 86. Nouri-Shirazi M, Guinet E. A possible mechanism linking ciga-
80. DHulst AI, Vermaelen KY, Brusselle GG, Joos GF, Pauwels RA. Time rette smoke to higher incidence of respiratory infection and asthma.
course of cigarette smoke-induced pulmonary inammation in mice. Immunol Lett 103: 16776, 2006.
Eur Respir J 26: 20413, 2005. 87. Bracke K, Cataldo D, Maes T, Gueders M, Noel A, Foidart JM,
81. Robbins CS, Dawe DE, Goncharova SI, Pouladi MA, Drannik AG, Brusselle G, Pauwels RA. Matrix metalloproteinase-12 and cathepsin
Swirski FK, Cox G, Stampi MR. Cigarette smoke decreases pulmonary D expression in pulmonary macrophages and dendritic cells of ciga-
dendritic cells and impacts antiviral immune responsiveness. Am J Respir rette smoke-exposed mice. Int Arch Allergy Immunol 138: 16979, 2005.
Cell Mol Biol 30: 20211, 2004. 88. Hammad H, Lambrecht BN. Recent progress in the biology of airway
82. Bracke KR, DHulst A I, Maes T, Moerloose KB, Demedts IK, dendritic cells and implications for understanding the regulation of
Lebecque S, Joos GF, Brusselle GG. Cigarette smoke-induced pulmo- asthmatic inammation. J Allergy Clin Immunol 118: 33136, 2006.
nary inammation and emphysema are attenuated in CCR6-decient 89. Ettmayer P, Mayer P, Kaltho F, Neruda W, Harrer N, Hartmann G,
mice. J Immunol 177: 435059, 2006. Epstein MM, Brinkmann V, Heusser C, Woisetschlager M. A novel
83. Arpinati M, Chirumbolo G, Urbini B, Martelli V, Stanzani M, low molecular weight inhibitor of dendritic cells and B cells blocks
Falcioni S, Bonifazi F, Bandini G, Tura S, Baccarani M et al. Use of allergic inammation. Am J Respir Crit Care Med 173: 599606, 2006.
anti-BDCA-2 antibody for detection of dendritic cells type-2 (DC2) 90. Matsubara S, Koya T, Takeda K, Joetham A, Miyahara N, Pine P,
in allogeneic hematopoietic stem cell transplantation. Bone Marrow Masuda ES, Swasey CH, Gelfand EW. Syk activation in dendritic
Transplant 29: 88791, 2002. cells is essential for airway hyperresponsiveness and inammation.
84. Aicher A, Heeschen C, Mohaupt M, Cooke JP, Zeiher AM, Dimmeler S. Am J Respir Cell Mol Biol 34: 42633, 2006.
Nicotine strongly activates dendritic cell-mediated adaptive immunity:
Potential role for progression of atherosclerotic lesions. Circulation 107:
60411, 2003.

132
Macrophages
11 CHAPTER

Macrophages are a family of mononuclear leu- monocytes express the cell surface protein Ly6c
kocytes that are widely distributed throughout (Gr-1), the chemokine receptor CCR2 and the
Galen B. Toews
most tissues. They vary considerably in pheno- adhesion molecule L-selectin and are selectively Division of Pulmonary and Critical Care
type depending on the local microenvironment recruited to inamed tissues and lymph nodes. Medicine, University of Michigan Health
[1]. Macrophages are involved in the scaveng- The second subset of monocytes (resting) are System, Ann Arbor, MI, USA
ing of dying cells, pathogens, and molecules found in resting and inamed tissues. This subset
through phagocytosis and endocytosis [2]. is dened by high expression of the chemokine
Macrophages secrete more than 100 substances. receptor CX3CR and LFA-1 and by the lack of
These secreted molecules expression of Gr-1, CCR2, and L-selectin. Two
monocyte subsets have also been identied in
induce cell movement;
humans; CD14, CD16 monocytes resemble
induce cell growth; murine inammatory monocytes and CD14low
induce cell death; and CD16 monocytes resemble mouse resi-
dent monocytes [3]. Interleukin-3 (IL-3), gran-
inuence cell dierentiation; ulocyte-macrophage colony stimulating factor
modify connective tissue structures; (GM-CSF), and macrophage colony stimulating
factor (M-CSF) stimulate a sequence of dieren-
regulate blood vessel growth.
tiation steps important in monocyte development
Macrophages within tissues make a vital by binding to specic receptors on progenitor
contribution to immune and inammatory cells. Cytokines released during acute inamma-
responses [2]. Macrophages are crucial for tory responses also regulate macrophage dieren-
tiation. Interleukin-1 (IL-1) and tumor necrosis
early recognition of microbes, particulates, factor (TNF-) enhance M-CSF and GM-CSF
and immunogens; production. Inhibitory inammatory cytokines
the initiation and regulation of inammatory involved in macrophage dierentiation include
responses and adaptive immunity; macrophage inammatory protein-1 (MIP-1)
the ingestion and killing of invading and transforming growth factor (TGF-) [4].
microbes. Monocytes/macrophages require continu-
ous reconstitution. This can be achieved by self
renewal of dierentiated cells, by proliferation
of bone marrow-derived precursors in peripheral
ORIGIN AND DISTRIBUTION tissues [5] or by the continuous extravasation
and dierentiation of circulating mononuclear
phagocyte precursors [6]. Alveolar macrophages
The mononuclear phagocyte system (MPS) is (AMs) are derived from blood monocytes and
composed of strikingly diverse dierentiated from proliferating macrophage precursors in the
cell types. Two subsets of murine monocytes interstitium of the lung. Approximately 1% of
arise from a common hematopoeitic progeni- the AM population in the normal lung is pro-
tor, the macrophage, and dendritic cell precur- liferating at any single time [7]. AMs have a life
sor (MDP, or monoblast) [3]. Inammatory span of months and perhaps years [8].

133
Asthma and COPD: Basic Mechanisms and Clinical Management

A [37], brinogen [38], and breakdown products of tissue


RECOGNITION OF MICROBES/MICROBIAL matrix, such as bronectin fragments [39] and hyaluronic
PRODUCTS acid oligosaccharides [40].
Activation of TLRs initiates signal transduction path-
Macrophages are a major cellular component of the innate ways that initiate and amplify inammatory responses in the
immune system [9]. The molecular and cellular processes lung and modulate adaptive immune responses. Molecules
of the innate immune response defend the host in the rst induced by PAMP-PRR interactions include
minutes or rst hours after exposure to microbes. The rec- signals that generate inammatory responses,
ognition of microbes is problematic because of their molec- including TNF, IL-1, IL-6, interferon (IFN)/ and
ular heterogeneity and their high mutation rates. The innate chemokines;
immune system uses a relatively small number of proteins
signals that function as co-stimulators of T-cell
encoded in the germ line to recognize a vast variety of
activation, B7.1 and B7.2;
molecular structures associated with microbes. The recep-
tors recognize a few, highly conserved structures present signals that regulate the dierentiation of lymphocytes,
in large groups of microorganisms. The receptors recognize including IL-4, IL-5, IL-10, IL-12, TGF- and
molecular patterns rather than particular structures and IFN- [41].
accordingly have been termed pattern-recognition recep- Activation of TLRs can also contribute to the induc-
tors (PRRs). The pathogen-associated molecular patterns tion of programmed cell death.
(PAMPs) recognized by PRR are chemically quite distinct
but share certain features. PAMPs are: (a) produced only
by microbes and not by eukaryote hosts, (b) essential for
survival or pathogenecity of the microbe, and (c) invariant
structures shared by classes of pathogens. Characteristic
HOMEOSTATIC REGULATION OF AM
PAMPs include lipopolysaccharides (LPSs) and teichoic FUNCTION
acids, shared by gram-negative and gram-positive bacte-
ria, respectively; mannans, conserved components of yeast Resident AMs are continually exposed to inhaled and aspi-
cell walls; and the unmethylated CpG motif characteris- rated particulates due to their position in the airway and
tic of bacterial but not mammalian DNA. PRRs expressed alveolar lumen. In spite of this exposure to particulates,
on macrophages include the mannose receptor, DEC205, resident AMs produce only low levels of cytokines and are
CD14, scavenger receptors, integrins, and toll-like receptors poorly phagocytic.
(TLRs). Recognition of pathogens results in the activation A mechanism by which the lung microenvironment
of various types of innate immune receptors [1015]. instructs AM innate and adaptive immune functions has
TLRs comprise a family of PRRs that recognize a recently been dened [42]. Under homeostatic conditions,
variety of conserved PAMPs displayed by microbes. Ten AMs closely adhere to alveolar epithelial cells (AECs). This
TLRs have been described (an eleventh is functional in cellcell interaction induces TGF- dependent expres-
mice but not in man [1622]). The principle role of TLRs sion of the integrin v6 on AEC. Localized activation of
appears to be induction of immune/inammatory responses TGF- in the vicinity of the macrophage suppresses mac-
to a broad range of pathogens (viruses, helminths, bacteria, rophage phagocytosis and cytokine production. This inhibi-
protozoa, and fungi). TLR4 is crucial for eective responses tion of macrophage function by the v6TGF- complex
to gram-negative LPS [23]. LPS delivery to TLR4 requires is unique to the lung.
the accessory proteins LPS-binding protein (LBP), CD14 The mechanism whereby microbial infection over-
and MD-2. Integrins such as CD11b/CD18 may facilitate comes suppression is also explained by AEC-macrophage-
responses to LPS [13, 24]. TLR4 also facilitates responses TLR responses. Infectious agents trigger TLRs which lead
to Streptococcus pneumoniae pneumolysin and proteins of to a rapid loss of contact with AECs which in turn induces
respiratory syncytial viruses [25, 26]. TLR2 mediates the rapid loss of expression of v6 on AEC. Under these con-
responses to lypoproteins and lipoteichoic acids (LTA) from ditions TGF- is no longer activated and macrophage activa-
gram-positive bacteria (S. pneumoniae) and Mycobacteria tion and innate immune functions are no longer suppressed.
[27, 28]. TLR2 plays a role in responses to Borrelia burgdor- Macrophages can now be primed to secrete pro-inammatory
feri, Aspergillus fumigatus, and Mycoplasma. TLR2 responses cytokines and to ingest and kill microbes. AMs lack a feed
are broadened by hetero-dimerization; TLR1/2 heterodim- forward mechanism of amplication mediated in other mono-
ers respond to a dierent panel than TLR2/6 heterodimers. nuclear phagocytes by autocrine secretion of IFN-. AMs
TLR2 and TLR4 respond to helminths. thus require exogenous IFN- for a robust response [43]
TLR9 mediates the responses through recognition of
CpG motifs in microbial DNA. TLR9 is activated by bac-
teria, Herpes viruses [29, 30], and Aspergillus [31]. TLR3
responds to double stranded viral RNA and TLR7 and
TLR8 mediate responses to single stranded RNA [32, 33].
GENERATION OF INFLAMMATORY RESPONSES
TLRs also respond to endogenous host molecules, includ-
ing defensins [34], reactive oxygen species (ROS) [35], Resident AMs can eectively ingest and kill invading
protein released from dying cells [36], surfactant protein microbes if the bacterial burden is low and the microbe is

134
Macrophages 11
minimally virulent [44, 45]. However, the recruitment of by cells that do not respond directly to bacterial products.
polymorphonuclear neutrophils (PMN) is essential for the Mononuclear phagocytes, neutrophils, and endothelial
eective containment of most virulent encapsulated bacte- cells produce CXC chemokines in response to LPS.
ria within the lung and for the eventual clearance of these Alternatively, airway and AECs, pulmonary broblasts, and
virulent microbes from the host. The generation of inam- pleural mesothelial cells produce IL-8 in response to spe-
mation in the lower respiratory tract is a dynamic process cic host-derived signals, such as TNF- or IL-1 [4850].
that involves the coordinated expression of both pro- and The importance of IL-1 and TNF- as key cytokines in
anti-inammatory cytokines. After the introduction of the initiation of this augmented inammatory response
microbes or microbial products, TLR-mediated signals is emphasized by the fact that all nucleated cells possess a
result in the production of TNF- and IL-1. TNF- and functional receptor for IL-1 and TNF-.
IL-1 stimulate the expression of adhesion molecules on Macrophages are also potent sources of bioactive
vascular endothelial cells. L-selectin on neutrophils interacts lipids, which are important in inammatory responses.
with its receptor/ligand (P- and E-selectin) on endothelial Leukotriene (LT) synthesis is dependent on three sequential
cells that lead to rolling. Intercellular adhesion molecule-1 enzymes: cytosolic phospholipase A (cPLA2), 5-lipoxygenase
(ICAM-1) expression is also induced on the surface of the (5-LO), and 5-LO-activating protein (FLAP). These three
endothelium; interactions between neutrophils and ICAM proteins co-localize at a single membrane site to form a
lead to rm adhesion [46, 47]. CXC chemokines and CC macromolecular complex termed a metabolon. There is now
chemokines are also rapidly produced in macrophages fol- abundant evidence documenting that the nuclear envelope
lowing microbial stimuli. CXC chemokines include IL-8, is the site at which this metabolon is assembled [51].
(CXCL8), GRO, (CXCL1) ENA-78, and (CXCL5) are The LT synthetic pathway is initiated when cPLA2
major PMN chemoattractants (Table 11.1). Thus, immedi- translocates from the cytosol to the nuclear envelope fol-
ately after the recognition of bacterial products in the alveo- lowing activation signals. Arachidonate is released from
lar environment, PMNs begin to accumulate. nuclear envelope phospholipids and is bound by FLAP, an
Macrophages also play a major role in amplifying the integral nuclear envelope protein. FLAP facilitates process-
inammatory response by stimulating cytokine production ing by 5-LO. On activation, 5-LO also translocates from
its resting locale(s) in the cytosol and/or nucleoplasm to
the nuclear envelope where it catalyzes the initial steps in
TABLE 11.1 The CXC chemokine/receptor family. LT synthesis [52, 53]. LTs, thus synthesized, are capable
of either entering into the nucleus or being exported out
Systematic Chemokine of the cell [53]. Monocytes release LTB4 and LTC4 after
name Human ligand Mouse ligand receptor(s) being exposed to non-immunologic stimuli or immunologic
stimuli. AMs produce a substantial excess of LTB4 com-
CXCL1 GRO/MGSA- GRO/KC? CXCR2  CXCR1 pared with LTC4 after stimulation [54]. LTB4 is a potent
CXCL2 GRO/MGSA- GRO/KC? CXCR2
chemotactic factor for PMNs and weaker chemotactic fac-
tor for eosinophils. LTB4 accounts for the majority of neu-
CXCL3 GRO/MGSA- GRO/KC? CXCR2 trophil chemotactic activity elaborated by AM immediately
following stimulation [54]. LTB4 also promotes adherence
CXCL4 PF4 PF4 Unknown of inammatory cells to the endothelium. Leukotrienes
CXCL5 ENA-78 LIX? CXCR2 also play a permissive role in inammation by promoting
the synthesis by macrophages of TNF-, IL-8, (CXCL8),
CXCL6 GCP-2 Ck-3 CXCR1, CXCR2 GRO, (CXCL1) ENA-78, (CXCL5), and IL-6.
An important implication of the metabolon concept
CXCL7 NAP-2 Unknown CXCR2
is that the site of macromolecular assembly has evolved in a
CXCL8 IL-8 Unknown CXCR1, CXCR2 manner to best serve the needs of the cell. The observation
that the LT metabolon is located within the nuclear enve-
CXCL9 Mig Mig CXCR3 lope suggests that the autocrine actions of bioactive lipid
mediators, including those potentially mediated within the
CXCL10 IP-10 IP-10 CXCR3
nucleus, may be of more importance than those paracrine
CXCL11 I-TAC Unknown CXCR3 actions that have been classically recognized. 5-LO metab-
olites are important modulators of mitogenesis, apoptosis,
CXCL12 SDF-1/ SDF-1 CXCR4 and the activation of various transcription factors. A solu-
CXCL13 BLC/BCA-1 BLC/BCA-1 CXCR5
ble nuclear receptor for LTB4 provides additional support
for such interactions; interestingly, this receptor is a mem-
CXCL14 BRAK/bolekine BRAK Unknown ber of a super-family of transcription factors and its ligation
induces gene transcription. Alternatively, reactive oxygen
(CXCL15) Unknown Lungkine Unknown species, which are a byproduct of arachidonate 5-lipoxygen-
A systematic name in parentheses means a human homologue has not been ation could exert nuclear actions by activating transcription
identified. factors or otherwise modifying nuclear constituents.
A question mark indicates that the mouse ligand homologue listed may not Monocytes exit the blood in inamed tissues in
correspond to the human ligand. response to specic chemotaxins. Monocyte recruitment is

135
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 11.2 The CC chemokine/receptor family. critically dependent on CC chemokines [29]. Twenty-seven
dierent CC chemokine ligands (CCL) have been described
Systematic Chemokine [55]. Chemokines that act mainly on monocytes are located
name Human ligand Mouse ligand receptor(s) on a cluster on human chromosome 17q11.2. Important
monocyte chemoattractant CC chemokines include MCP-1
CCL1 I-309 TCA-3/P500 CCR8
(CCL2), MIP-1 (CCL3), MIP-1 (CCL4), and RANTES
CCL2 MCP-1/MCAF JE? CCR2 (CCL5) (Table 11.2). CC chemokines are produced by
monocytes, alveolar macrophages, lymphocytes, neutrophils,
CCL3 MIP-1/LD78 MIP-1 CCR1/CCR5 epithelial cells, broblasts, smooth muscle cells, and endothe-
CCL4 MIP-1 MIP-1 CCR5
lial cells. These cells produce chemokines in response to a
variety of factors including cigarette smoke, viruses, bacte-
CCL5 RANTES RANTES CCR1/CCR3/ rial products, IL-1, TNF, C5a, LTB4, and INFs [5666].
CCR5 Bronchial epithelial cells release monocyte chemotaxins
in response to cigarette smoke [63]. TNF- and IL-1 are
(CCL6) Unknown C10/MRP-1 Unknown among the most potent stimuli for epithelial cell cytokine
CCL7 MCP-3 MARC? CCR1/CCR2/ production. Macrophage stimulation of epithelial cells by
CCR3 TNF or IL-1 likely enhances and perpetuates the initial
inammatory response. Increased epithelial expression of
CCL8 MCP-2 MCP-2? CCR3 MCP-1 has been observed by immunohistochemistry and
biopsy specimens from patients with atopic asthma, and
(CCL9/10) Unknown MRP-2/CCF18/ Unknown
increased levels of this chemokine are also seen in bron-
MIP-1
choalveolar lavage (BAL) uid of allergic asthmatics when
CCL11 Eotaxin Eotaxin CCR3 compared with normal subjects [65, 66].
The biological eects of chemokines are mediated by
(CCL12) Unknown MCP-5 CCR2 seven transmembrane domain receptors that are a subset of
the G-protein-coupled receptor super-family. Sixteen recep-
CCL13 MCP-4 Unknown CCR2/CCR3
tors have been identied. Redundancy and binding promis-
CCL14 HCC-1 Unknown CCR1 cuity exists between ligands and receptors (Tables 11.1 and
11.2). A single chemokine may bind to several receptors
CCL15 HCC-2/Lkn-1/ Unknown CCR1/CCR3 and a single chemokine receptor can transduce signals for
MIP-1 several chemokines [55].
CCL16 HCC-4/LEC LCC-1 CCR1
Monocytes also respond to formyl peptides, C5a, and
elastin fragments. In a murine model of cigarette smoke-
CCL17 TARC TARC CCR4 induced emphysema, CC and CXC chemokines are unde-
tectable but chemotactic extracellular matrix fragments,
CCL18 DC-CK1/PARC Unknown Unknown particularly elastin fragments, are present [6769].
AMAC-1

CCL19 MIP-3/ELC/ MIP-3/ELC/ CCR7


exodus-3 exodus-3
T-CELL INDEPENDENT MACROPHAGE
CCL20 MIP-3/LARC/
exodus-1
MIP-3/LARC/
exodus-1
CCR6
ACTIVATION
CCL21 6Ckine/SLC/ 6Ckine/SLC/ CCR7
The early nonantigen-specic activation of macrophages
exocus-2 exocus-2/
is one of the rst events in the innate immune response
TCA-4
and is often very eective in eliminating microbes. Innate
CCL22 MDC/STCP-1 ABCD-1 CCR4 immune macrophage activation lls a gap between micro-
bial entry into the host and the development of antigen-
CCL23 MPIF-1 Unknown CCR1 specic immunity. Innate immune macrophage activation is
a cytokine mediated macrophage-NK cell interaction [70].
CCL23 MPIF-2/ Unknown CCR3
Macrophages release IL-12 and TNF- following micro-
Eotaxin-2
bial recognition; these cytokines induce NK cell IFN-
CCL25 TECK TECK CCR9 which primes macrophages for microbicidal activity. IL-12
is regulated by both positive and negative feedback mecha-
CCL26 Eotaxin Unknown CCR3 nisms. IFN- activated macrophages produce much higher
CCL27 CTACK/ILC ALP/CTACK/ CCR10
levels of IL-12. IL-10, a product of macrophages and other
ILC ESkine
cell types, is a potent inhibitor of IL-12 production [71].
Expression of IL-10 is delayed compared with the expres-
A systematic name in parentheses means a human homologue has not been identified. sion IL-12 in vivo. IL-10 is a critical component of the
A question mark indicates that the mouse ligand homologue listed may not hosts natural defense against excessive production of IL-12
correspond to the human ligand. and its pathological consequences.

136
Macrophages 11
ROLE OF MACROPHAGES IN INITIATION OF to the lung might theoretically dilute the resident AM
population with recruited monocytes and convert a nor-
IMMUNE RESPONSES mally immunosuppressive tissue milieu into one that is sup-
portive of T-cell activation.
The activation of T-lymphocytes is a complex biological func-
tion that requires the participation of an antigen-presenting
cell (APC). Most cell types cannot perform these functions;
professional APCs (macrophages, dendritic cells (DCs), and
B-cells) are required. Antigen presentation involves the dis- INNATE IMMUNE CONTROL OF ADAPTIVE
play of an antigenic epitope in association with a major his-
tocompatibility complex (MHC) molecule. Three additional
IMMUNE RESPONSES
molecular interactions are crucial to the interaction between
an APC and a T-lymphocyte: Naive T-lymphocytes can dierentiate along dierent path-
ways to become distinct eector cells. The tissue micro-
1. adhesion molecules that promote the physical interaction environment in which the specic immune response is
between APC and T-cells; generated crucially regulates this dierentiation process via
the secretion of specic cytokine signals (Fig. 11.1). IL-12
2. co-stimulatory molecules which are membrane bound produced by macrophages during the early innate immune
growth/dierentiation molecules that produce T-cell response and IFN- induced by IL-12 create an environ-
activation [72]; ment in which antigen-specic CD4 and CD8 T-cells
3. soluble molecules such as TNF- and IL-1. are preferentially induced to dierentiate into T1 cells that
produce even higher levels of IFN- [78]. IL-4 is crucial
AMs are ineective in presenting antigen to T-cells to the development of T2 responses during the priming of
[7375]. AMs are less eective than monocytes in inducing naive T-cells. The crucial cellular source of early IL-4 pro-
proliferation of blood T-lymphocytes to soluble recall anti- duction is uncertain. Basophils, mast cells, cells, T2 lym-
gens. AMs can, however, restimulate recently activated T- phocytes, and an NK 1.1 CD4CD8 T-lymphocyte have
cells eectively. AMs fail to activate CD4 T-cells because all been reported to produce IL-4 [79].
of defective expression of B7 co-stimulatory cell surface The type of APC that presents the antigen may also
molecules. AMs activated with IFN- fail to express B7-1 or be crucial. DCs preferentially activate T2 cells in certain cir-
B7-2 antigens [76]. Resident pulmonary AMs also actively cumstances [80]. The mechanisms by which DCs favor the
suppress T-cell proliferation induced by antigen [73]. expression of T2 cells remains uncertain but may be related
The mechanisms whereby particulate antigens and to the ability of DCs to secrete IL-1, a co-stimulator for T2
microbial agents induce T-cell responses are being dened. cells but not T1 cells, and to the absence of IFN- produc-
DCs are present in the airways and in the interstitium of tion by DCs which would inhibit the development of T2
the lower respiratory tract [77]. DCs are clearly crucial for cells. Dierential expression of co-stimulatory molecules
activating naive T-cells for proliferation and clonal expan- may also be crucial to this polarization process. AM fails to
sion. Monocytes are also stimulatory to T-cell activation. express B7.1 or B7.2 even when stimulated with IFN-. B7.2
Thus, inammatory stimuli that recruit fresh monocytes is constitutively expressed on DCs and macrophages, whereas

Th1 cell

IL-10
Macrophage
IL-12
IFN-
MHC/Peptide/TCR
Bacteria Density FIG. 11.1 Innate immune control of
Affinity
Fungi APC Antigen focusing adaptive immune response. The tissue
NK cell B7
Protozoa CD28 microenvironment regulates adaptive
Bystander APC
Viruses Costimulation
TCR MHC
immune responses. DC migration and
Helminths CD28 B7-1, B7-2
functional differentiation state are crucial
Allergens Costimulation
Dendritic cell
Superantigens T-cell Nave B7-2 IL-4 DC subset
determinants of immune priming. IgE on
T helper cell State of differentiation the surface of DC may focus uptake and
processing of other antigens. Density and
IL-4
Antigen focusing
affinity of antigens regulates differential
signals following T-cell receptor engagement.
Basophil/mast cell
Co-stimulatory molecules influence
Th2 cell T-cell differentiation. Innate immune cell
cytokines are the major regulators of T-
cell differentiation. (Solid arrows indicate
NK 1.1; CD4, CD8 T-cells stimulators; broken arrows indicate inhibitor.)

137
Asthma and COPD: Basic Mechanisms and Clinical Management

B7.1 is not. The outcome of B7.1 and B7.2 co-stimulation is that contain the sequence GLU-LEU-ARG (ELR motif )
dierent. B7.2 co-stimulates the production of IL-4 as well are potent angiogenic factors. In contrast, CXC chem-
as IL-2 and IFN-. Thus, after B7.2 co-stimulation, an initial okines that lack the ELR motif (platelet factor 4, IFN-
source of IL-4 is available. However, B7.2 co-stimulation inducible protein 10) behave as potent angiostatic factors.
provides only a moderate signal for T2 cell dierentiation; Non-ELR-containing chemokines can inhibit angiogenic
additional signals are almost surely required to achieve high activity of both ELR-CXC chemokines and structurally
levels of IL-4 production [81]. B7 co-stimulatory signals can unrelated macrophage-derived angiogenic factor bFGF [87].
be delivered by bystander APC in vitro with the same e-
ciency as the APC engaging the TCR. Accordingly, T-cell Macrophages and chronic obstructive
activation in vivo may occur with MHC-TCR engagement
being provided by one APC, whereas co-stimulation is deliv- pulmonary disease
ered by a second bystander APC-type [82].
The tissue microenvironment in which the immune A marked increase in the number of macrophages and neu-
responses are generated is crucial in determining the type trophils in the airways of both humans and experimental
and intensity of T-lymphocyte responses. Cytokines secreted animals is the most consistent, early eect of exposure to
in the tissues, where the antigen is deposited, are also crucial cigarette smoke [88]. Histologic studies of bronchial biop-
for the maintenance and regulation of immune responses. sies and lung parenchyma obtained from cigarette smokers
demonstrate a predominance of macrophages and CD8
T-cells at sites of parenchymal destruction [89, 90]. The
paucity of neutrophils in lung parenchyma and airway biop-
sies suggests neutrophils trac rapidly from the blood into
ROLE OF MACROPHAGE-DERIVED the airway lumen. The inammatory process, once initiated,
GROWTH FACTORS IN TISSUE persists in ex-smokers [90, 91]. Surprisingly, the inamma-
tory responses persist in end-stage emphysema. Increased
REMODELING AND REPAIR number of macrophages, CD8 and CD4 Tlymphocytes,
and neutrophils are noted in lungs from patients undergoing
Fibrotic changes occur in airways and the lower respira- lung volume reduction surgery.
tory tract following certain injuries. Macrophages produce Macrophages are likely to play an important role in
numerous growth factors for broblasts including platelet- initiating the neutrophilic inammatory response in ciga-
derived growth factor (PDGF), TGF- and TGF-, and rette smokers. Macrophages may be activated by cigarette
insulin-like growth factor [8385]. smoke and other inhaled particulates. Elevated levels of the
CXC chemokines, IL-8, (CXCL8), GRO, and (CXCL1)
are noted in cigarette smokers. IL-8 and (CXCL8) lev-
PDGF induces broblast proliferation and collagen
els in sputum correlate with the magnitude of neutrophilic
production.
inammation and with percent predicted FEV1 [92]. LTB4
The wound healing eects of PDGF are macrophage is increased in the sputum of patients with chronic obstruc-
dependent in most models [85]. tive pulmonary disease (COPD); this potent neutrophilic
TGF- stimulates the closure of wounds induced in chemoattractant likely participates in the generation of the
cultures of type II AEC in vitro. neutrophilic inammatory response.
The inammatory response in COPD has several dis-
TGF- likely plays a role in epithelial cell repair of the tinguishing features:
lungs following injury. Macrophages also produce TGF-
, which has important eects on the turnover of matrix Resident AMs are activated upon exposure to cigarette
proteins and the proliferation of broblasts. smoke.
TGF- activates genes that favor the production of Neutrophils are rapidly recruited almost immediately
matrix proteins. TGF- also down-regulates production after cigarette smoke exposure in response to macrophage
of matrix metalloproteinases derived from PMNs and and epithelial cell-derived chemokines and leukotrienes.
macrophages that digest matrix in the interstitium
Macrophages and CD8 and CD4 T-lymphocytes
and alveolar spaces. In aggregate, these eects shift the
accumulate within days to weeks and continue to
balance in favor of matrix accumulation.
accumulate with time.
GM-CSF has a protective eect on repair processes The abnormal accumulation of inammatory cells
following bleomycin. This protective eect is mediated, in persists throughout the disease process, even when the
part, by GM-CSF-induced production of PGE2 by pulmo- inciting agent, cigarette smoke, is removed.
nary macrophages [86]. PGE2 has crucial down-regulatory
eects on broblasts, including decreasing broblast prolif-
eration and decreasing matrix produced by broblasts. Macrophage proteinases and COPD
Macrophages may also play critical roles in regulating
angiogenesis, which is a central biological event in repair Early changes of emphysema include subtle disruption of
and remodeling. CXC chemokines such as IL-8 or ENA-78 elastic bers, bronchiolar and alveolar distortion, and the

138
Macrophages 11
appearance of fenestrae. Destruction of the elastic frame- elastin, which are responsible for the monocyte recruit-
work leads to loss of the intra-alveolar septae and macro- ment in response to smoke. Macrophage-mediated lung
scopic appearance of spaces of more than 1mm in diameter. destruction after exposure to cigarette smoke is, at least in
This destructive process is accompanied by an increase in the part, directly related to the presence of MMP-12, which is
mass of collagen, suggesting that active alveolar wall brosis most likely required for direct degradation of lung tissue.
occurs in the tissues which remain in otherwise emphysema- The generation of chemotactic elastin fragments provides a
tous lungs [93]. The dominant, working hypothesis to explain positive feedback loop perpetuating macrophage accumula-
the pathogenesis of emphysema has postulated an imbalance tion in lung destruction.
between proteolytic enzymes and proteinase inhibitors in the Cigarette smoke induces a distinct pattern of AM
lung, favoring an excess of proteinases, particularly elastases. gene expression not seen in non-smokers or patients with
This hypothesis postulates that cigarette smoke, macro- asthma [102]. AM from smokers strongly express MMP-12,
phages, chemoattractants, neutrophils, elastases, and protei- but most of the 72 genes upregulated in smokers were not
nase inhibitors interact with lung connective tissue, primarily identied in two transgenic murine models of emphysema.
elastin, to cause repeated destruction and synthesis of pulmo-
nary matrix.
Pulmonary macrophages may play an important role
in this proteolytic process by releasing neutrophil chemotac-
tic factors, which recruit neutrophils to the respiratory tract. MACROPHAGES AND ADAPTIVE
Neutrophil elastase is believed to play an important role in IMMUNITY IN COPD
COPD. Neutrophil elastase is one of the most potent elastases
in the lung. Instillation of neutrophil elastase and protein-
ase 3 causes emphysema in animals [9496]. Neutrophils Cigarette smoking stimulates humoral and cellular compo-
are recruited into the lungs of animals and generate detect- nents of the adaptive immune response. This response may
able elastin fragments following exposure to cigarette smoke. provide exquisite specic memory for previous exposures
Neutrophil elastase is also a potent mucus secretagague. to foreign materials. The histologic hallmark of an adap-
Macrophages also secrete potent proteinases. Human tive immune response is the presence of lymphoid follicules
AMs produce the cysteine (thiol) proteinases, cathepsins with germinal centers. Bronchial-alveolar lymphoid tis-
B, H, L, and S. Cathepsins L and S have relatively indis- sue (BALT) collections are found in 5% of smokers with
criminate substrate specicities that include elastin and normal lung function (GOLD0) and in those with mild
other matrix components. Cysteine proteinases are involved (GOLD1) and moderate (GOLD2) severity. These collec-
in lung destruction in IL-13 and IFN- transgenic mice tions of BALT increase sharply in severe (GOLD3) and
[97, 98]. very severe (GOLD4) COPD perhaps in response to colo-
Pulmonary macrophages also produce matrix metallo- nization and infection of the lower airways [90].
proteinases (MMPs). Studies of human emphysematous lung
tissue have demonstrated the presence of several MMPs. A
correlation between MMP-1 and MMP-9 and emphysema
was noted when smokers with emphysema were compared MACROPHAGES AND APOPTOSIS IN COPD
with smokers without emphysema [99]. Studies using trans-
genic and gene-targeted mice also lend support for the role
There is growing evidence for a role for apoptosis in COPD
of MMPs in emphysema. Transgenic mice over-expressing
[103]. AMs show a markedly reduced capacity to ingest
human MMP-1 in the lung develop airspace enlargement
apoptotic cells relative to their avid ingestion of inert par-
[100]. Studies of MMP-12 knockout mice provide specic
ticles [104]. This capacity is further reduced in COPD
loss-of-function data that MMP-12 is importantly involved
with attendant increased accumulation of apoptotic cells.
in the development of emphysema in response to cigarette
Apoptotic cell recognition typically induces a unique anti-
smoke. Exposure of MMP-12 / mice to long-term ciga-
inammatory state; defective clearance might be a factor
rette smoke led to inammatory cell recruitment followed
encouraging lung inammation [105].
by alveolar space enlargement similar to that seen in humans
with emphysema. Mice decient in MMP-12 (MMP-
12 /) were protected from development of emphysema
despite long-term smoke exposure [101]. Interestingly,
MMP-12 / mice failed to recruit monocytes into the MACROPHAGES AND INITIATION OF
lung in response to cigarette smoke. MMP-12 / mice ANTIGEN-SPECIFIC T2 IMMUNE
could egress from the pulmonary vasculature in response to
MCP-1 instillation. However, even in the presence of MMP-
RESPONSES IN ASTHMA
12 / macrophages, the lungs of mice exposed to cigarette
smoke had no changes in the mean linear intercept in alveolar Resident pulmonary AMs actively suppress T-cell pro-
duct areas when compared to mice not exposed to smoke. liferation induced by antigen or polyclonal stimuli [73].
These ndings suggest that MMP-12 is expressed by Changes occur within the local inductive milieu of the lung
resident AMs after exposure to cigarette smoke. MMP-12 in patients with asthma. AM suppression is reduced after
generates monocyte chemotaxins, likely fragments of exposure to allergens [106108].

139
Asthma and COPD: Basic Mechanisms and Clinical Management

The tissue microenvironment is a crucial regulator of


specic immune response generation (Fig. 11.1). The pres-
MACROPHAGES AND AIRWAY RESPONSES TO
ence of IgE on APCs likely promotes the uptake and the VIRAL INFECTION
processing of allergens and their eventual presentation to
nave T-cells. DCs express both FcR I and FcR II. These Host innate immune responses to respiratory viral infec-
two receptors could function to capture allergen bound to tions may play a crucial role in the pathogenesis of asthma.
allergen-specic IgE and thus focus the immune response The site of initial viral replication is usually respiratory epi-
through facilitated antigen presentation [109]. thelial cells and macrophages. Host defense against virus
Antigens also deliver signals via quantitative vari- depends on the targeted death of infected cells and the sub-
ation in ligand density on APC. Peptide/MHC class II sequent clearance of the cellular corpses by macrophages.
complexes that interact strongly with the TCR favor T1 Macrophages must avoid viral-induced death for this proc-
responses, whereas weak interactions result in the priming of ess to be eective. CCL5CCR5 interactions are required
T2 responses. The overall binding anity can be varied by to prevent apoptosis of virus infected macrophages in vivo
modifying the peptide, which results in dierent signals. The and in vitro. Infected macrophages must resist cell death
mechanisms by which signals delivered via the TCR con- to eciently clear virus infected apoptotic cells from the
trol dierentiation is uncertain; dierential TCR aggrega- lung. If this clearance process is disrupted, pathogens are
tion may result in dierential intracellular signals that favor not cleared and residual apoptotic cells including activated
distinct cytokine gene expression or certain MHCTCR macrophages can cause further damage [116].
interactions may favor dierential co-receptor expression
[110]. As noted above, co-stimulatory molecules may direct
the polarization of T-cells into T1 or T2 cells; B7.2 pro- References
vides only a moderate signal for T2 cell dierentiation; and
co-stimulatory signals may be delivered either by the APC
1. Gordon S, Hughes DA. Macrophages and their origins: Heterogeneity
that presents the antigen or by the bystander APC. Thus, in relation to tissue microenvironment. In: Lipscomb M, Russell S
macrophages may serve as bystander APC and inuence (eds). Lung Macrophages and Dendritic Cells, pp. 331. New York:
DC-induced T-cell proliferation [111]. Soluble cytokines Marcel Dekker, 1997.
produced by cells of the innate immune response are likely 2. Gordon S. Pattern recognition receptors: Doubling up for the innate
the major regulators of T-cell dierentiation (see Innate immune response. Cell 111(7): 92730, 2002.
Control of Adaptive Immune Responses section). 3. Geissmann F, Jung Steen Littman DR. Blood monocytes consist of
two principal subsets with distinct migratory properties. Immunity 19:
7182, 2003.
4. Sie CA. Hematopoietic growth factors. J Clin Invest 79: 154957,
MACROPHAGES IN THE EFFECTOR PHASES OF 1987.
5. Takahashi K, Nalto M, Takeya M. Development and heterogeneity of
IMMUNE RESPONSES IN ASTHMA macrophages and their related cells through their dierentiation path-
ways. Pathol Int 46: 47385, 1996.
6. Kennedy DW, Abkowitz JL. Mature monocytic cells enter tissues and
Macrophages are likely the sources of cell-specic chem- engraft. Proc Natl Acad Sci USA 95: 1494449, 1998.
oattractants in patients with asthma. RANTES, (CCL5), 7. Bitterman PB, Saltzman LE, Adelberg S et al. Alveolar macrophage
MIP-1, and (CCL3) are chemotactic for macrophages, replication: One mechanism for the expansion of the mononuclear
eosinophils, and basophils. MCP-1 and (CCL1) are chem- phagocyte population in the chronically inamed lung. J Clin Invest 74:
otactic for macrophages and basophils. IL-8 and (CXCL8) 46069, 1984.
are chemotactic for neutrophils, basophils, and to lesser 8. Thomas ED, Rambergh RE, Sale GE et al. Direct evidence for
extent eosinophils. Additionally, macrophage-released early bone marrow origin of the alveolar macrophage in man. Science 192:
cytokines likely induce epithelial cells and broblasts to 101618, 1976.
9. Medzhitov R, Janeway CA. Jr. Innate immunity. N Eng J Med 343:
release chemoattractants and growth factors (GM-CSF, IL-
33844, 1992.
8, (CXCL8), MCP-1, (CCL1), MCP-3, (CCL7), MCP4, 10. Stahl PD. The mannose receptor and other macrophage lectins. Curr
(CCL13), MIP-1, (CCL3), RANTES, and (CCL9)) in Opin Immunol 4: 4952, 1992.
patients with asthma [112115]. 11. Jiang W, Swiggard WJ, Heuer C et al. The receptor DEC-2O5
Leukotrienes are perhaps the best studied macrophage- expressed by dendritic cells and thymic epithelial cells is involved in
produced mediators involved in the pathogenesis of asthma. antigen processing. Nature 375: 15153, 1995.
The role of macrophage-derived leukotrienes remains uncer- 12. Ulevitch RJ. Tobias PS. Receptor-dependent mechanisms of cell stimu-
tain. Cysteinyl leukotrienes (LTC4, D4, and E4) are minor lation by bacterial endotoxin. Annu Rev Immunol 13: 43757, 1995.
products of AMs when compared to LTB4 production [51, 13. Wright SD, Ramos RA, Tobias PS et al. CD14, a receptor for com-
53, 113]. This metabolic prole argues against a role for AMs plexes of lipopolysaccharride (LPS) and LPS binding Protein. Science
249: 143133, 1990.
in LT overproduction in the lung. Alternatively, several fea-
14. Pugin J, Heumann D, Tomasz A et al. CD14 is a pattern recognition
tures favor a role for AMs in LT production. The cells are receptor. Immunity 1: 50916, 1994.
located on the airway surface, express plasma membrane 15. Hampton RY, Golenbock DT, Penman M et al. Recognition and
IgE receptors, and secrete substantially more LTC4 than do plasma clearance of endotoxin by scavenger receptors. Nature 352:
PMN. Cysteinyl leukotrienes are known to promote mucus 34244, 1991.
hypersecretion, airway inammation, and marked, prolonged 16. Akira S, Takeda K. Toll-like receptor signaling. Nat Rev Immunol 4:
contraction of smooth muscles in airways. 499511, 2004.

140
Macrophages 11
17. Basu S, Fenton MJ. Toll-like receptors: Function and roles in lung 41. Toews GB. Cytokines and pulmonary host defense against microbes.
disease. Am J Physiol Lung Cell Mol Physiol 286: L887lL892, 2004. In: Nelson S, Martin TR (eds). Cytokines in Pulmonary Disease: Infection
18. Cook DN, Pisetsky DS, Schwartz DA. Toll-like receptors in the patho- and Inammation, pp. 117. New York: Marcel Dekker, 2000.
genesis of human disease. Nat Immunol 5: 97579, 2004. 42. Takabayshi K, Corr M, Hayashi T et al. Induction of a homeostatic
19. Decker T. Sepsis: Avoiding its deadly toll. J Clin Invest 113: 138789, circuit in lung tissue by microbial compounds. Immunity 24: 47587,
2004. 2006.
20. Doherty TM, Arditi M. TB, or not TB: That is the question: Does 43. Punturieri T, Alvianani RS, Polack T et al. Specic engagement of TL4
TLR signaling hold the answer? J Clin Invest 114: 1699703, 2004. or TLR3 does not lead to IFN mediated innate immune signal ampli-
21. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive cation and STAT-1 phosphorylation in resident murine alveolar mac-
immune responses. Nat Immunol 5: 98795, 2004. rophages. J Immunol 173: 103342, 2004.
22. Ulevitch RJ. Therapeutics targeting the innate immune system. Nature 44. Onofrio JM, Toews GB, Lipscomb MF et al. Granulocyte alveolar
Rev Immunol 4: 51220, 2004. macrophage interactions in the pulmonary clearance of Staphylococcus
23. Hirshfeld M, Ma Y, Weis JH et al. Cutting repurication of lipopoly- aureus. Am Rev Respir Dis 136: 81823, 1987.
saccharide eliminates signaling through both human and murine Toll- 45. Rehm SR, Gross GN, Pierce AK. Early bacterial clearance from
like receptor 2. J Immunol 166: 57481, 2000. murine lungs: Species-dependent phagocyte response. J Clin Invest 66:
24. Martin TR, Mathison JC, Tobias PS et al. Lipopolysaccharride binding 19499, 1980.
protein enhances the responsiveness of alveolar macrophages to bac- 46. Albelda SM, Smith CW, Ward PA. Adhesion molecules and inam-
terial lipolysaccharide. Implications for cytokine production in normal matory injury. FASEB J 8: 50412, 1994.
and injured lungs. J Clin Invest 90: 220919, 1992. 47. Springer TA. Trac signals for lymphocyte re-circulation and leuko-
25. Malley R, Henneke P, Morse SC et al. Recognition of pneumolysin by cyte immigration: The multi-step paradigm. Cell 76: 30114, 1994.
Toll-like receptor 4 confers resistance to pneumococcal infection. Proc 48. Strieter RM, Kunkel SL, Showell HJ et al. Endothelial cell gene
Natl Acad Sci USA 100: 196671, 2003. expression of a neutrophil chemotactic factor by TNF, LPS, and IL-l.
26. Kurt-Jones EA, Popova L, Kwinn L et al. Pattern recognition receptors Science 243: 146769, 1989.
TLR4 and CD14 mediate response to respiratory syncytial virus. Nat 49. Strieter RM, Phan SH, Showell HJ et al. Monokine-induced neu-
Immunol 1: 398401, 2000. trophil chemotactic factor gene expression in human broblasts. J Biol
27. Knapp S, Wieland CW, vant Veer C et al. Toll-like receptor 2 plays a Chem 264: 1062126, 1989.
role in the early inammatory response to murine pneumococcal pneu- 50. Standiford TJ, Kunkel SL, Basha MA et al. Interleukin-8 gene expres-
monia but does not contribute to antibacterial defense. J Immunol 172: sion by a pulmonary epithelial cell line: A model for cytokine networks
313238, 2004. in the lung. J Clin Invest 86: 194553, 1990.
28. Means TK, Jones BW, Schromm AB et al. Dierential eects of a Toll- 51. Peters-Golden M, Brock TG. Intracellular compartmentalization of
like receptor antagonist on Mycobacterium tuberculosis-induced macro- leukotriene biosynthesis. Am J Respir Crit Care Med 161: 53640, 2000.
phage responses. J Immunol 166: 407482, 2001. 52. Woods J, Evans J, Ethier D et al. 5-lipoxygenase and 5-lipoxygenase
29. Hochrein H, Schlatter B, OKeee M et al. Herpes simplex virus activating protein are localized in the nuclear envelope of activated
type-1 induces IFN-production via Toll-like receptor 9-dependent human leukocytes. J Exp Med 178: 193546, 1993.
and -independent pathways. Proc Natl Acad Sci USA 101: 1141621, 2004. 53. Woods J, Coey M, Brock TG et al. 5-lipoxygenase is located in the
30. Lund J, Sato A, Akira S et al. Toll-like receptor 9 mediated recognition euchromatin of the nucleus in resting human alveolar macrophages and
of Herpes simplex virus-2 by plasmacytoid dendritic cells. J Exp Med translocated to the nuclear envelope upon cell activation. J Clin Invest
198: 51320, 2003. 95: 203540, 1995.
31. Bellocchio S, Moretti S, Perruccio K et al. TLRs govern neutrophil 54. Martin TR, Pistorese B, Chi E et al. Eect of leukotriene B4 in
activity in aspergillosis. J Immunol 173: 740615, 2004. the human lung: Recruitment of neutrophils into the alveolar spaces
32. Diebold SS, Kaisho T, Hemmi H et al. Innate antiviral responses by without a change in protein permeability. J Clin Invest 84: 160919,
means of TLR7-mediated recognition of single-stranded RNA. Science 1989.
303: 152931, 2004. 55. Zlotnik A, Yoshi O. Chemokines: A new classication system and their
33. Heil F, Hemmi H, Hochrein H et al. Species-specic recognition of role in immunity. Immunity 12: 12127, 2000.
single-stranded RNA via Toll-like receptor 7 and 8. Science 303: 1526 56. Christensen PJ, Rolfe MW, Standiford et al. Characterization of the
29, 2004. production of monocyte chemoattractant protein-1 (MCP-1) and
34. Biragyn A, Runi PA, Leifer CA et al. Toll-like receptor 4-dependent interleukin-8 (IL-8) in an allogeneic immune response. J Immunol 151:
activation of dendritic cells by -defensin 2. Science 298: 102529, 120513, 1993.
2002. 57. Liebler JM, Kunkel SL, Burdick MD et al. The production of IL-8 and
35. Frantz S, Kelly RA, Bourcier T. Role of TLR-2 in the activation of MCP-1 by peripheral blood monocytes: Disparate responses to PHA
nuclear factor-B by oxidative stress in cardiac myocytes. J Biol Chem and LPS. J Immunol 152: 24149, 1994.
276: 5197203, 2001. 58. Standiford TJ, Kunkel SL, Lieber JM. The gene expression of MIP-1
36. Park JS, Svetkauskaite D, He Q et al. Involvement of TLR2 and TLR4 from human blood monocytes and alveolar macrophages is inhibited
in cellular activation by high mobility group box 1 protein (HMGB1). by IL-4. Am J Respir Cell Mol Biol 9: 19298, 1993.
J Biol Chem 279: 737077, 2004. 59. Miller MD, Hata S, deWaal Malefyt R, Krangel MS. A novel polypep-
37. Guillot L, Balloy V, McCormack FX, Golenbock DT, Chignard M, tide secreted by activated human T lymphocytes. J Immunol 143:
Si-Tahar M. Cutting edge: The immunostimulatory activity of the 290716, 1989.
lung surfactant protein-A involves Toll-like receptor 4. J Immunol 168: 60. Kasama T, Strieter RM, Standiford TJ et al. Expression and regulation
598992, 2002. of human neutrophil-derived macrophages inammatory protein-1.
38. Smiley ST, King JA, Hancock WW. Fibrinogen stimulates macrophage J Exp Med 178: 6372, 1993.
chemokine secretion through Toll-like receptor 4. J Immunol 167: 61. Paine R III, Rolfe MW, Standiford TJ et al. MCP-1 expression by
288794, 2001. rat type II alveolar epithelial cells in primary culture. J Immunol 150:
39. Okamura Y, Watari M, Jerud ES et al. The extra domain A of bronec- 456170, 1993.
tin activates Toll-like receptor 4. J Biol Chem 276: 1022933, 2001. 62. Standiford TJ, Kunkel SL, Phan SH et al. Alveolar macrophage-
40. Termeer C, Benedix F, Sleeman J et al. Oligosaccharides of hyaluronan derived cytokines induce monocyte chemoattractant protein-1 expres-
activate dendritic cells via Toll-like receptor 4. J Exp Med 195: 99111, sion from human pulmonary type II like epithelial cells. J Biol Chem
2002. 266: 991218, 1991.

141
Asthma and COPD: Basic Mechanisms and Clinical Management

63. Koyama S, Rennard SI, Leikauf GD et al. Bronchial epithelial cells 87. Strieter RM, Polverini PJ, Kunkel SL et al. The functional role of the
release monocyte chemotactic activity in response to smoke and endo- ELR motif in CXC chemokine mediated angiogenesis. J Biol Chem
toxin. J Immunol 147: 97279, 1991. 270: 2734857, 1995.
64. Rolfe MW, Kunkel SL, Standiford TJ et al. Expression and regulation 88. Reynolds HY. Bronchoalveolar lavage. Am Rev Respir Dis 135: 250
of human pulmonary broblast-derived monocyte chemotactic peptide 63, 1987.
(MCP-1). Am J Physiol 263: L53645, 1992. 89. Seatta M, Di Stefano A, Turato G et al. CD8 T lymphocytes in
65. Sousa AR, Lane SJ, Nakhosteen JA et al. Increased expression of the peripheral airways of smokers with chronic obstructive pulmonary
monocyte chemoattractant protein-1 in bronchial tissue from asth- disease. Am J Respir Crit Care Med 157: 82226, 1998.
matic subjects. Am J Respir Cell Mol Biol 10: 14247, 1994. 90. Hogg JC, Chu F, Utokaparch S et al. The nature of small-airway
66. Alan R, York T, Boyars M et al. Increased MCP-1, RANTES and obstruction in chronic obstructive pulmonary disease. N Engl J Med
MIP-1 in bronchoalveolar lavage uid of asthmatic patients. 350: 264553, 2004.
Am J Respir Crit Care Med 153: 1398404, 1996. 91. Turato G, Di Stefano A, Maestrelli P et al. Eect of smoking cessa-
67. Hautamaki RD, Kobayashi DK, Senior RM et al. Macrophage elastase tion on airway inammation in chronic bronchitis. Am J Respir Crit
is required for cigarette smoke-induced emphysema in mice. Science Care Med 152: 166672, 1995.
277: 20024, 1997. 92. Keatings VM, Collins PD, Scott DM et al. Dierences in interleukin-
68. Senior RM, Grin GL, Mecham RP. Chemotactic activity of elastin- 8 and tumor necrosis factor- induced sputum from patients with
derived peptides. J Clin Invest 66: 85962, 1980. chronic obstructive pulmonary disease or asthma. Am J Respir Crit
69. Hunninghake GW, Davidson JM, Rennard S et al. Elastin fragments Care Med 153: 53034, 1996.
attract macrophage precursors to diseased sites in pulmonary emphy- 93. Lang MR, Fiaux GW, Gillooly, M et al. Collagen content of alveolar
sema. Science 212: 92527, 1981. wall tissue in emphysematous and non-emphysematous lungs. Thorax
70. Scott P, Trinchieri G. The role of natural killer cells in host parasite 49: 31926, 1994.
interactions. Curr Opin Immunol 7: 3440, 1995. 94. Damiano VV, Tsang A, Kucich U et al. Immunolocalization of elastase
71. Macatonia SE, Hosken NA, Litton M et al. Dendritic cells produce in human emphysematous lungs. J Clin Invest 78: 48293, 1986.
IL-12 and direct the development of Th1 cells from naive CD4 T 95. Senior RM, Tegner H, Kuhn C et al. The induction of pulmonary
cells. J Immunol 154: 507179, 1995. emphysema induced with human leukocyte elastase. Am Rev Respir
72. Thompson CB. Distinct roles for the costimulatory ligands B7-1 and Dis 116: 46977, 1977.
B7-2 in T helper cell dierentiation. Cell 81: 97982, 1995. 96. Kao RC, Wehner NG, Skubitz KM et al. Proteinase 3. A distinct
73. Toews GB, Vial WC, Dunn MM et al. The accessory cell function of human polymorphonuclear leukocyte proteinase that produces
human AMs in specic T cell proliferation. J Immunol 132: 18186, emphysema in hamsters. J Clin Invest 82: 1693, 1988.
1984. 97. Wang Z, Zheng T, Zhu Z et al. Interferon gamma induction of pul-
74. Lipscomb MF, Lyons CR, Nunez G et al. Human AMs: HLA-DR- monary emphysema in the adult murine lung. J Exp Med 192: 1587
positive macrophages that are poor stimulators of a primary mixed leu- 600, 2000.
kocyte reaction. J Immunol 136: 497504, 1986. 98. Zheng T, Zhu Z, Wang Z et al. Inducible targeting of IL-13 to the
75. Lyons CR, Ball EJ, Toews GB et al. Inability of human AMs to stimu- adult lung causes matrix metalloproteinases and cathepsin-dependent
late resting T cells correlates with decreased antigen-specic T cell- emphysema. J Clin Invest 106: 108193, 2000.
macrophage binding. J Immunol 137: 117380, 1986. 99. Finlay GA, ODriscoll LR, Russel KJ et al. Matrix metalloproteinase
76. Chelen CJ, Fang Y, Freeman GJ et al. Human AMs present antigen expression and production by alveolar macrophages in emphysema.
ineectively due to defective expression of B7 costimulatory cell sur- Am J Respir Crit Care Med 156: 24047, 1997.
face molecule. J Clin Invest 95: 141521, 1995. 100. DArmiento J, Dalal SS, Okada Y et al. Collagenase expression in the
77. Toews GB. Pulmonary dendritic cells: Sentinels of lung-associated lung of transgenic mice causes pulmonary emphysema. Cell 71: 955
lymphoid tissues. Am J Respir Cell Mol Bio1 4: 2045, 1991. 61, 1992.
78. Trinchieri G, Gerosa F. Immunoregulation by interleukin-12. 101. Hantamaki RD, Kobayashi DC, Senior RM et al. Macrophage
J Leukocyte Biol 59: 50511, 1996. elastase required for cigarette smoke induced emphysema in mice.
79. Seder RA, Mosmann TA. Dierentiation of eector phenotypes of Science 277: 20024, 1997.
CD4 and CD8 T cells. In: Paul WE (ed.), Fundamental Immunology, 102. Woodru PG, Koth LL, Yang YH et al. A distinctive alveolar macro-
pp. 879908. Philadelphia. New York: Lippencott. Raven, 1999. phage activation state induced by cigarette smoking. Am J Respir Crit
80. Hauser C, Snapper CM, OHara J et al. T helper cells grown Care Med 172: 138392, 2005.
with hapten-modied cultured Langerhans cells produce interleukin-4 103. Kasahara Y, Tuder RM, Cool CD et al. Endothelial cell death and
and stimulate IgE production by B cells. Eur J Immunol 10: 24551, decreased expression of vascular endothelial growth factor and vascu-
1989. lar endothelial growth factor receptor 2 in emphysema. Am J Respir
81. Bluestone JA. New Perspective of CD28-B7-mediated T cell costimu- Crit Care Med 163: 73744, 2001.
lation. Immunity 2: 55559, 1995. 104. Hu B, Sonstein J, Christensen PJ et al. Decient in vitro and in vivo
82. Ding L, Shevach EM. Activation of CD4 T cells by delivery of the phagocytosis of apoptotic T cells by resident murine alveolar macro-
B7 costimulatory signal on bystander antigen-presenting cells (trans- phages. J Immunol 165: 212433, 2000.
costimulation). Eur J Immunol 24: 85966, 1994. 105. Fadok VA, Bratton DL, Konowal A et al. Macrophages that have
83. Mornex JF, Martinet Y, Yamauchi K et al. Spontaneous expression ingested apoptotic cells in vitro inhibit proinammatory cytokine
of the c-sis gene and release of a platelet-derived growth factor-like production through autocrine/paracrine mechanisms involving TGF-
molecule by human alveolar macrophages. J Clin Invest 78: 6166, , PGE2, and PAF. J Clin Invest 101: 80998, 1998.
1986. 106. Aubus P, Cosso B, Godard P et al. Decreased suppressor cell activity
84. Assoian RK, Fleudelys GE, Stevensen HC et al. Expression and secre- of AMs in bronchial asthma. Am Rev Respir Dis 130: 87578, 1984.
tion of type beta transforming growth factor by activated human mac- 107. Spiteri MA, Knight RA, Jeremy JY et al. Alveolar macrophage-induced
rophages. Cell 53: 28593, 1988. suppression of T cell hyperresponsiveness in asthma is reversed follow-
85. Rose R, Raines EW, Bowen-Pope DF. The biology of platelet-derived ing allergen exposure in vitro. Eur Respir J 7: 143138, 1994.
growth factor. Cell 46: 15569, 1986. 108. Fischer HG, Frosch S, Reske K et al. Granulocyte-macrophage col-
86. Moore BB, Coey MJ, Christensen PJ et al. GM-CSF regulates ony-stimulating factor activates macrophages derived from bone mar-
bleomycin-induced pulmonary brosis via a prostaglandin-dependent row cultures to synthesis of MHC class II molecules and to augment
mechanism. J Immunol 165: 403239, 2000. antigen presentation function. J Immunol 141: 388288, 1988.

142
Macrophages 11
109. Van der Heijden FL, Van Neerven RJI, Van Katwijk et al. Serum-
biopsies from atopic and non-atopic (Intrinsic) asthmatics. J Immunol
IgE-facilitated allergen presentation in atopic disease. J Immunol 150:
163: 632129, 1999.
364350, 1993.
113. Gonzalo JA et al. The coordinated action of CC chemokines in the
110. Racioppi L, Ronchese F, Matis LA et al. Peptide-major histocom-
lung orchestrates allergic inammation and airway hyperresponsive-
patibility complex class II complexes with mixed agonist/antagonist
ness. J Exp Med 188: 15767, 1998.
properties provide evidence for ligand-related dierences in T cell
114. Elsner J et al. The CC chemokine antagonist Met-RANTES inhibits
receptor-dependent intracellular signaling. J Exp Med 177: 104760,
eosinophil eector functions through the chemokine receptors CCR1
1993.
and CCR3. Eur J Immunol 27: 289298, 1997.
111. Ding L, Shevach EM. Activation of CD4 T cells by delivery of the
115. Fels AO, Pawlowski NA, Cramer EB et al. Human AMs produce
B7 costimulatory signal on bystander antigen-presenting cells (trans-
leukotriene B4. Proc Natl Acad Sci USA 79: 786670, 1982.
costimulation). Eur J Immunol 24: 85966, 1994.
116. Tyner JW, Uchida O, Kajiwara N et al. CCL5-CCR5 interaction
112. Ying S et al. Eosinophil chemotactic chemokines (eotaxin, eotaxin-
provides antiapoptotic signals for macrophage survival during viral
2, RANTES, monocyte chemoattractant protein-3 (MCP-3), and
infection. Nature Med 11: 118087, 2005.
MCP-4), and C-C chemokines receptor 3 expression in bronchial

143
Eosinophils
12 CHAPTER

Eosinophil inltration of the airways is a charac- 0.4 109/l under normal conditions [2]. Some Dagmar Simon1 and
teristic feature of obstructive pulmonary diseases
in particular of bronchial asthma. But eosinophils
laboratories list higher upper values, in particular
in children (as high as 0.75 109/l). Eosinophils
Hans-Uwe Simon2
have also been connected with chronic obstruc- circulate only a few hours in the peripheral blood 1
Department of Dermatology,
tive pulmonary disease (COPD) in at least a before they enter the tissues, where they can sur- Inselspital, University of Bern, Bern,
subgroup of patients. Initially, the function of vive for at least 2 weeks [3, 4]. Switzerland
eosinophils was almost exclusively related to their The cytoplasmic granules are composed 2
Department of Pharmacology,
destructive activity mediated by the toxic eects of four distinct populations, which can be University of Bern, Bern, Switzerland
of the eosinophil granule proteins. In the last identied under electron microscopy: primary
two decades research work has made it clear that and secondary granules, small granules and
eosinophils also play important roles in immu- lipid bodies [5]. The primary granules contain
nomodulation as well as in repair and remodeling galectin-10, the Charcot-Leyden crystal protein,
processes. To describe the role of eosinophils in which binds lysophospholipases [6]. The cyto-
bronchial asthma or other eosinophilic diseases, toxic cationic proteins are stored in the second-
we will rst review their functional properties and ary granules. These granules are formed by a core
the regulation of eosinophil numbers. We will containing major basic protein (MBP) and a
then focus on the potential role of eosinophils matrix composed of eosinophil cationic protein
in the pathogenesis of bronchial asthma and (ECP), eosinophil peroxidase (EPO), and eosi-
COPD. However, as the reader will realize our nophil derived neurotoxin (EDN) [7]. Mature
current knowledge on the physiologic and patho- eosinophils also house small granules stor-
genic role of eosinophils is still not complete. ing proteins, such as arylsulphatase B and acid
phosphatase. The lipid bodies contain arachi-
donic acid, the basic component for eicosanoid
production [8]. In addition, both the secondary
MORPHOLOGY AND LOCATION granules and the lipid bodies store a number of
cytokines, chemokines and growth factors [5].
In the absence of eosinophil-specic sur-
Among the leukocytes, eosinophils attract atten- face markers [9], MBP and ECP are widely
tion because of their characteristic appearance used as targets for immunohistochemical stud-
revealing a bilobed nucleus with highly condensed ies. Some investigations revealed that eosinophil
chromatin and a multitude of coarse granules. granule proteins are present not only in the
Eosinophils can easily be identied because of cells, but also in the extracellular spaces suggest-
their strong anity to the acidic dye eosin in blood ing eosinophil degranulation [10]. In addition,
and tissues even when they are paraformaldehyde receptors, which are almost specically expressed
xed and paran embedded. This characteristic by eosinophils, for example, CC chemokine
inspired Paul Ehrlich to name these granular leu- receptor (CCR)3 and interleukin (IL)-5 receptor
kocytes eosinophils after discovering them in 1879 alpha, or the transcription factor GATA-1 can be
[1]. Eosinophils are released from the bone mar- used as markers to identify eosinophils. Recent
row into the peripheral blood, where they represent research using ow cytometry and microarray
15% of the leukocytes with an upper limit of assay techniques demonstrated that eosinophils

145
Asthma and COPD: Basic Mechanisms and Clinical Management

express typical surface molecule patterns consisting of CCR3, Eosinophil


Siglec-8 (Siglec-F), FIRE (F4/80like receptor), CD62 lig-
and, and paired Ig-like receptor A/B (Pir-A/B) at dierent
stages of dierentiation and activation [11].
Under physiologic conditions, eosinophils are found in
the bone marrow, the lymphatic organs such as spleen, lymph
nodes, and thymus, and throughout the gastrointestinal tract
except the esophagus [12]. Recent studies reported the essen-
tial role of eosinophils in mammary gland branch formation
[13] and maturation of the pubertal uterus in mice [14]. In
Granule proteins Cytokines
the gastrointestinal tract, numbers of eosinophils depend MBP IL-1
on the location as well as on the exposure to pathogens in ECP IL-3
asymptomatic individuals [15]. Under normal conditions, EDN IL-5
the baseline levels of eosinophils are controlled by eotaxin-1 EPO IL-6
[16]. Moreover, in preinvolutional human thymi, eosinophils IL-8
[17] and eosinophil precursors [18] have been identied, the IL-10
Respiratory burst IL-12
latter suggesting dierentiation of eosinophils in the thymus. O2 IL-13
Eosinophils have been associated with major histocompat- Leukotrienes IFN-
ibility complex (MHC) class I-restricted selection/deletion LTC4 TGF-
in the thymus, pointing to an immunomodulatory function LTD4 TGF-
under non-pathological conditions [19]. LTE4 CM-CSF
TNF-
Antigen presentation RANTES
HLA-DR Eotaxin

EOSINOPHIL FUNCTIONS FIG. 12.1 Eosinophil effector molecules.

The view of eosinophil function has profoundly changed in


the last two decades. Eosinophils have always been regarded
as destructive eector cells that release toxic granule pro- cationic nature, it aects the charge of surface membranes
teins. The primary function of eosinophils has been related resulting in disturbed permeability, disruption, and injury of
to the protection against helminth parasites [20]. The obser- cell membranes [23]. At low concentrations, MBP stimu-
vation that eosinophils surround parasites in the tissue as lates mediator production by other inammatory cells
well as a number of in vitro and animal experiments revealed (discussed later). ECP and EDN have been identied as
a direct role for eosinophils in killing parasites. Later it ribonucleases [24] and possess antiviral activity [25]. ECP
became clear that eosinophils also play a role in the patho- damages target cell membranes through the formation of
genesis of helminth infections by causing tissue damage pores or transmembrane channels [26]. Besides their ribonu-
and organ dysfunction [20]. Results of more recent research clease activities, both EDN and ECP are potent neurotoxins
indicate that eosinophils play an important role in repair [27]. Eosinophils store abundant amounts of ECP and may
and remodeling processes after tissue damage as well as in release it upon repetitive stimulation with the same agonist,
immunomodulation [21]. Interestingly, eosinophils produce implying that mature eosinophils do not require signicant
and release a broad spectrum of cytokines, chemokines, and de novo ECP synthesis for secretion [28]. EDN was demon-
leukotrienes as well as express cell surface receptors for these strated to induce recruitment and cytokine release by den-
mediators, suggesting autocrine and paracrine mechanisms dritic cells [29]. EPO is involved in the generation of toxic
in regulating eosinophil functions [21]. The main eector oxygen species as part of the respiratory burst.
molecules of eosinophils are summarized in Fig. 12.1. Upon activation, eosinophils specically release their
Activation of eosinophils is required for their migra- granule proteins by exocytosis or degranulation [30]. In
tion into the sites of inammation in the tissue, for the classical exocytosis, single secretory granules are extruded to
production and release of their granule proteins and lipid the cell exterior, whereas in compound exocytosis fusion of
mediators, as well as for the generation of reactive oxygen intracellular granules precedes their release through a sin-
species [5]. Hematopoietins, such as IL-3, IL-5 and granu- gle fusion pore to eectively target surfaces as it has been
locyte-macrophage colony-stimulating factor (GM-CSF), shown for helminth infection [31]. Piecemeal degranula-
increase functional responses of eosinophils to various tion has been identied as mechanism by which cytoplas-
agonists including lipid mediators, complement factors, or mic secretory vesicles containing cytoplasmic crystalloid
chemokines. This eect of hematopoietins is called priming. granules with core components are released from eosi-
nophils. In addition, granule deposition in the tissue may
occur after cytolysis of the eosinophil [30]. A prerequisite
Granule protein production and release for exocytosis is the docking of the vesicles/granules to the
cell membrane mediated by membrane-associated proteins
MBP is highly cytotoxic [22] and plays an essential role forming soluble NSF attachment protein (SNAP) receptors
in host defense against helminth infection. Because of its (SNAREs) [32].

146
Eosinophils 12
Respiratory burst for leukotriene generation [42]. Because of their content
of leukotriene C4 synthetase, eosinophils are an important
The superoxide radical generation after activation of the source of cysteinyl leukotrienes, predominatly LTC4 and its
NADPH oxidase complex presents a powerful weapon by active metabolites LTD4 and LTE4 [8]. Leukotrienes pos-
which eosinophils dispose of pathogens; and this mecha- sess proinammatory activities. They may act intracellularly
nism is even more powerful than that in neutrophils [33]. as intracrine signaling molecules, for example, in regulating
Activated NADPH oxidase catalyzes O2 to O2, which IL-4 release [8]. Leukotrienes may amplify the inamma-
enters further redox pathways to generate hydrogen peroxide tory cascade, for instance by acting as chemotactic factors or
(H2O2) in the presence of superoxide dismutase, or hydroxyl by triggering the release of cytotoxic proteins.
and nitrogen dioxide radicals, after combining with nitric
oxide. The granular protein EPO oxidizes bromide, nitrite,
and thiocyanate in the presence of H2O2, which have the Antigen presentation
potency to disrupt cell membranes or cause intracellular oxi-
dant stress [34]. In vitro experiments have demonstrated that eosinophils are
able to process antigens and express costimulatory molecules
and MHC class II molecules, and therefore may function as
Cytokine production and release antigen-presenting cells in stimulating T-cell responses [44].
Whereas blood eosinophils from normal and eosinophilic
Eosinophils are able to produce, store, and secrete a wide donors do not express MHC class II molecules, HLA-
spectrum of cytokines and chemokines and thus may func- DR synthesis occurs following stimulation with specic
tion as immunomodulatory cells. Although eosinophils are cytokines including IL-3, IL-4, GM-CSF or IFN- [44,
terminally dierentiated cells, their capacity to generate 45]. During transmigration into the tissues, HLA expres-
cytokines can be quite intriguing. Eosinophils from nor- sion is further increased [46]. By expressing costimulatory
mal individuals constitutively express IL-4 and IL-10 with molecules, such as CD80 and CD86, eosinophils provide
further upregulation in response to inammatory signals additional secondary signals for lymphocytes [44]. However,
[35]. Both the cytokines as well as IL-2, IL-5, and IL-13 the role of eosinophils as antigen-presenting cells to nave
are stored in the crystalloid core. Cytokines and chemok- T-cells is minor compared to dendritic cells [47].
ines that may activate cytokine production by eosinophils
are IL-3, IL-5, GM-CSF, interferon (IFN)-, tumor
necrosis factor (TNF)-, and complement factor C5a [5,
36]. In response to stimulation, eosinophils may produce REGULATION OF PRODUCTION,
proinammatory cytokines, which may overlap the typical
spectrum of both T helper 1 and T helper 2 cytokines, reg-
MIGRATION, AND SURVIVAL
ulatory cytokines, and chemokines: IL-1, IL-3, IL-5, IL-6,
IL-8, IL-12, IL-13, IFN-, tumor growth factor (TGF)-, Prerequisite for eosinophil inltration and function in the
TGF-, GM-CSF, TNF-, macrophage inammatory pro- tissues is a precise regulation of their production, migration,
tein (MIP)-1, regulated upon activation, normal T-cell and accumulation as well as activation (Fig. 12.2).
expressed and secreted (RANTES) and eotaxin [4, 5, 37
40]. Recently, eosinophils have been shown to be involved
in innate immune responses by expressing Toll-like recep- Production
tors [41]. By generating IL-10 and TGF-, eosinophils are
suggested to modulate T regulatory cell function and sup- In the bone marrow, eosinophils are generated from
press inammatory events [21]. CD34 hematopoietic progenitor cells under regulation
of critical transcription factors especially GATA-1 and
the eosinophilopoietins IL-5, GM-CSF, and IL-3 [4].
Lipid mediator generation Eosinophils share this progenitor with basophils [48]. IL-5
specically regulates the production rate and dierentiation
In eosinophils, eicosanoid derivatives of arachidonic acid of the eosinophil lineage [49]. The importance of IL-5 is
are generated by both cyclooxygenase and lipoxygenase evident from the studies of IL-5 decient mice, which are
pathways, which have been localized to the sites of the peri- unable to develop eosinophilia upon allergen sensitization
nuclear membranes and the cytoplasmic lipid bodies [8]. and challenge [50]. In contrast, IL-5 transgenic mice exhibit
Platelet-activating factor (PAF) as well as agonists of CCR3 extensive eosinophil production and tissue eosinophilia
can initiate the de novo formation of lipid bodies [8]. To [51]. Anti-IL-5 therapy of asthma patients was shown to
synthesize leukotrienes (LT), eosinophils need to be both lead to an inhibition of eosinophil maturation in the bone
primed (e.g. by IL-3, IL-5, GM-CSF) and activated (e.g. by marrow and a decrease of eosinophil progenitors in the
C5a, PAF) [42, 43]. To reach the threshold needed to phos- bronchial mucosa [52]. Besides IL-5, IL-3 and GM-CSF
phorylate cytosolic phospholipase A2 (PLA2), a concurrent have also been shown to increase eosinophil production
stimulation of G-protein-coupled receptor and cytokine [53]. Whereas the release of eosinophils into the peripheral
receptor resulting in an activation of the mitogen-activated blood circulation requires IL-5 [54]; the migration into the
protein kinase (MAPK) cascade is required [42]. The acti- tissues under physiologic conditions is independent of IL-5
vation of PLA2, which also requires calcium, is essential and mainly under the control of eotaxin-1 as shown for the

147
Asthma and COPD: Basic Mechanisms and Clinical Management

(VCAM)-1 and intercellular adhesion molecule (ICAM)-1,


respectively [36]. IL-4 was shown to induce the expression
of the adhesion molecule VCAM-1, whereas IFN- stimu-
lates ICAM-1 expression [60]. The expression of VLA-4 on
eosinophils, which signicantly contributes to eosinophil
inammation in allergic responses, can be upregulated by
eotaxin [61]. After transmigration through the blood vessel,
eosinophils enter the extracellular matrix where they bind to
matrix proteins such as bronectin. This binding is mediated
by integrins and selectins on the eosinophils. Eosinophils
migrate into the tissue along chemokine gradients. Their
movement is characterized by adhesion/deadhesion to extra-
cellular matrix proteins [36].
Eotaxin and RANTES are important chemokines
for eosinophils and largely contribute to the movement of
eosinophils from the peripheral blood to the sites of inam-
mation [62]. The principal receptor involved in eosinophil
attraction is CCR3 [56]. In addition to eotaxin, RANTES
and monocyte chemoattractant protein (MCP)-1, 2, 3, and
4 may also bind to CCR3 [6365]. IL-5 alone has weak
chemotactic activity, but increases the eect of eotaxin by
enhancing CCR3-mediated responses [65]. Priming of
eosinophils leads to an upregulation of additional chem-
okine receptors including CCR1, the receptor for MIP-1a,
RANTES, and MCP-3 [66].
Complement factors such as the anaphylatoxins C3a
and C5a [67] as well as PAF [68] have also been implicated
in eosinophil recruitment. Moreover, leukotrienes (LTB4)
FIG. 12.2 Mechanisms and key players (bold) leading to eosinophilic and prostaglandins (PGD2) were found to induce eosinophil
inflammation in asthma. chemotaxis [69, 70], whereas lipoxin A4 blocks eosinophil
tracking [71]. All these chemoattractants, C5a, PAF,
LTB4, and PGD2 can be produced and released by mac-
gastrointestinal tract [55], the mammary glands [13], the rophages that were shown to play a role in recruiting eosi-
uterus [14], and the thymus [16]. The receptor for the CC nophils to the lung and peritoneal cavity during helminth
chemokine eotaxin is CCR3, which is expressed on both infection [11]. The T helper 2 cytokine IL-13 was shown to
CD34 progenitor cells and mature eosinophils [56, 57] promote eosinophilia by increasing local IL-5 and/or eotaxin
and can be upregulated during allergic inammation [57]. expression [72].

Migration into tissues Survival


Priming of eosinophils circulating in the peripheral blood is Tissue eosinophilia is regulated not only by recruitment of
a prerequisite for their transmigration through the endothe- eosinophils from the bone marrow but also by eosinophil sur-
lial cell layer, which has been demonstrated for eosinophils vival. Delayed apoptosis as a pathogenic mechanism has been
of asthmatic patients in contrast to healthy individuals [58] shown in several eosinophilic diseases including asthma [73,
and in response to allergen challenge [59]. Migration com- 74]. However, resolution of inammation is associated with
prises a series of sequential events including rolling, adhe- disappearance of eosinophils [75]. Apoptotic eosinophils
sion, and transmigration through the vascular endothelium. are taken up by macrophages [75] and epithelial cells [76].
In the initiation and regulation of these processes a number In addition, eosinophils may be cleared by transepithelial
of cytokines and chemokines are involved. migration [77]. In particular IL-5, but also IL-3 and GM-
The initial steps of eosinophil recruitment are adhe- CSF delay apoptosis and prolong eosinophil survival [78, 3].
sion mediated by interaction of the P-selectin-glycopro- Recently, it has been shown that leptin may also block eosi-
tein ligand (PSGL)-1 on eosinophils with P-selectin on nophil apoptosis [79].
endothelial cells and rolling, a process in which PAF and The signaling events important for cytokine-medi-
eotaxin are involved [5]. While L-selectin is constantly ated antiapoptosis include activation of tyrosine kinases and
expressed on eosinophils, P- and E-selectins on endothe- signal transducers and activators of transcription (STAT)
lial cells are upregulated by the inammatory cytokines proteins resulting in the transcriptional activation of mem-
IL-1 and TNF- [36]. The rm attachment of eosi- bers of the Bcl-2 and inhibitor of apoptosis proteins (IAP)
nophils to the endothelium is mediated by eosinophil sur- families [8082]. IL-5 upregulates the antiapoptotic pro-
face very late activation antigen (VLA)-4 (integrin 41) teins Mcl-1 and Bcl-xL, and inhibits translocation of
and CD11b/CD18 binding to vascular adhesion molecule proapoptotic Bax, resulting in an inhibition of the caspase

148
Eosinophils 12
cascade and thus preventing apoptotic cell death [83, 84]. Eosinophil
Moreover, following IL-5 stimulation, normal blood eosi-
nophils express the antiapoptotic molecules survivin and
cIAP-2 [82]. Although TNF- and TNF-related apoptosis
inducing ligand (TRAIL) are death receptor ligands, they
do not induce eosinophil apoptosis under normal condi-
tions [85]. On the contrary, both TNF- and TRAIL were
shown to prolong eosinophil survival [86, 87]. Stimulation
of CD137, another member of the TNF-family, which Tissue Remodeling Immunoregulation
is expressed on eosinophils of allergic patients, by a spe- destruction FGF-2 T helper 2 cytokines
cic antibody together with IL-5 or GM-CSF blocks the MBP NGF TNF-
inhibitory eect of the survival factors on eosinophil apop- ECP VEGF Leukotrienes
EDN TGF- Antigen presentation
tosis [88]. Eosinophil death can be induced by interaction IL-4
EPO
of the surface death receptors CD95 (Fas) and its ligand, IL-11
Respiratory burst
CD95L (FasL), which is expressed by activated T-cells [89]. IL-13
Therefore, T-cells, known as main producers of eosinophil IL-17
survival factors, may also limit eosinophil expansion within
inammatory sites [90]. FIG. 12.3 Possible roles of eosinophils in the pathogenesis of asthma.

eector function, causing desquamation and destruction


ROLE IN LUNG DISEASES of the epithelium, leads to airway damage and lung dys-
function [21, 95]. MBP has been localized to the sites of
epithelial damage and has been detected in the sputum
Asthma of asthmatic patients [40]. Eosinophils from asthmatic
patients contain, and in response to IL-5 and GM-CSF
Eosinophilia in the blood and/or tissues is a feature of multi- release, signicantly higher amounts of EDN compared
ple disorders. Although the initial cause and the organs vary, to healthy controls [96]. Increased ECP levels in blood,
there are only the following two major pathways that medi- sputum, and bronchoalveolar lavage uid of patients with
ate eosinophilia: (1) a cytokine-mediated increased dier- bronchial asthma have been reported to correlate with air-
entiation and survival of eosinophils (extrinsic eosinophilic way obstruction and disease activity [97]. Polymorphisms
disorders) and (2) a mutation-mediated clonal expansion of in the ECP gene are associated with the development of
eosinophils (intrinsic eosinophilic disorders) [2]. The most asthma [98].
common cause of eosinophilia is the increased generation Nitrotyrosine, a product of EPO, has been observed
of IL-5-producing T-cells, as is observed in allergic diseases in the airways as well as in lung parenchyma of asthmatic
including bronchial asthma (Fig. 12.2). patients [99, 100]. Eosinophils from patients with various
There are a great number of reports in the literature types of asthma were reported to excessively produce reac-
demonstrating the correlation between asthma severity and tive oxygen species [5]. These mechanisms may contribute
levels of eosinophils and their products in blood, sputum, to the damage of resident cells in the lungs. Patients with
bronchoalveolar lavage uid, or bronchial biopsies [91]. bronchial asthma have elevated levels of nitric oxide (NO)
Eosinophils found in the airways of asthmatics are activated in their expired air [101], which is thought to reect the
[92]. Since clinical trials investigating monoclonal anti-IL- extent of eosinophilic inammation of the lower airways
5 antibody treatment in bronchial asthma provided rather [102]. Expired NO is used as diagnostic tool in monitoring
disappointing results, a vehement controversy on the role of asthma [102]. Viral and non-viral infections are known to
eosinophils in the pathogenesis of asthma started. Recent induce asthma exacerbations, in which an increase in blood
studies in genetically targeted mice showed that eosinophils and sputum eosinophil numbers often precedes deteriora-
are crucial for airway hyperresponsiveness and mucus accu- tion of symptoms and lung function [103]. In response to
mulation [93] as well as for airway remodeling [94]. respiratory syncytial virus infection, epithelial cells of the
The dierent activities of eosinophils that contribute airways express chemokines leading to the recruitment and
to the pathogenesis of bronchial asthma can be categorized degranulation of eosinophils [104]. EDN and ECP are
as tissue destruction, remodeling, and immunoregulation RNAses and were shown to function as anti-viral agents
(Fig. 12.3). During asthmatic inammation, eosinophils [25]. However, granule proteins released during viral infec-
interact with leukocytes including lymphocytes, mast cells, tion may also lead to bronchial hyperreactivity and asthma
dendritic cells, macrophages and neutrophils, as well as with exacerbation in susceptible individuals [25, 40].
resident tissue cells, such as epithelial cells, endothelial cells, It should be noted that basic proteins are believed
smooth muscle, broblasts, and nerve cells [21]. to have additional function. For instance, MBP has been
described to induce histamine release from mast cells and
Tissue destruction basophils [105], eosinophil degranulation and cytokine (IL-8)
The release of eosinophil granule proteins, such as MBP, production [106], as well as to increase neutrophil [107, 108]
EPO, ECP, and EDN, has suggested that the main eosinophil and platelet functions [109]. In addition, MBP and EPO

149
Asthma and COPD: Basic Mechanisms and Clinical Management

were reported to increase bronchial hyperresponsiveness by expressing VEGF receptors [124]. In a murine model of
binding to the muscarinic M2 receptors of bronchial smooth asthma, the administration of anti-VEGF receptor anti-
muscles resulting in a vagal overstimulation and bronchocon- bodies reduced eosinophil inltration [125]. The media-
striction [110]. tors, expressed by eosinophils during inammation, can also
activate smooth muscle cells. For example, eotaxin-1 pro-
duced by eosinophils themselves can bind to CCR3, which
Remodeling is highly expressed on airway smooth muscle cells in asth-
Eosinophils are involved in airway remodeling character- matic lungs [126]. Eosinophils have the capacity to gener-
ized by structural changes of the lung including epithelial ate IL-25, which has been detected in bronchial biopsies of
hypertrophy, subepithelial deposition of extracellular matrix patients with asthma, and is suggested to have remodeling
proteins in the lamina reticularis, mucus gland hypertrophy, functions by stimulating airway smooth muscle cells to
airway smooth muscle hypertrophy, and vascular changes express extracellular matrix components [114].
[111]. The matrix deposition (collagen, brin) in the lungs
observed upon prolonged allergen challenge was signi-
cantly increased in wild-type mice compared to eosinophil- Immunoregulation
decient [94]. Many of the factors implicated in remodeling The inammation in asthma is characterized by T helper
are expressed by eosinophils and have been found to be 2 immune responses leading to increased production of
increased in asthmatic airway including broblast growth cytokines such as IL-4, IL-5, IL-9, and IL-13. Since eosi-
factor (FGF)-2, IL-4, IL-11, IL-13, IL-17, nerve growth nophils have been demonstrated to express these cytokines,
factor (NGF), and vascular endothelial growth factor they are suggested to be involved in sustaining a local T
(VEGF) [112]. In patients receiving anti-IL-5 antibody helper 2 immune microenvironment [39]. This view is sup-
therapy, a reduced deposition of extracellular matrix proteins ported by the observation that in eosinophil-decient mice
(tenascin, lumican, procollagen III) beneath the bronchial the production of T helper 2 cytokines is reduced in the
basement membrane has been observed, again suggesting lungs after allergen challenge [93]. By producing cytokines
an essential role of eosinophils in airway remodeling [113]. associated with acute inammatory responses such as TNF-
This decrease of extracellular matrix proteins was accompa- , eosinophils may enhance the inammation in asthma
nied by a signicant reduction of TGF- expression [113]. [36]. Eosinophils are an important source of the cysteinyl
TGF- by autocrineparacrine actions may stimulate eosi- leukotrienes [127]. In bronchial mucosa biopsies of asth-
nophils to generate IL-11, another cytokine with brogenic matics, eosinophils overexpress LTC4 synthetase [128].
potential, which has been found to be signicantly increased LTC4 and its derivatives LTD4 and LTE4 were shown
in the airways of asthmatics compared to healthy controls not only to increase mucus secretion [129] and bronchoc-
[114]. Furthermore, eosinophils express IL-6, whose func- onstriction [130], but also eosinophil survival [131]. In
tion has been related to tissue brosis [114]. Since eosi- asthmatic patients, antigen challenge induces HLA-DR
nophil granule proteins interact with several resident cell expression on recruited eosinophils, which enables them to
types in the lungs, they are suggested to play additional function as antigen-presenting cells in stimulating T-cell
roles in tissue remodeling. MPB interacting with IL-1 responses [132, 133]. Moreover, eosinophils provide cos-
and TGF- stimulates lung broblasts [115], while EPO timulatory signals for lymphocytes by expressing CD40,
products aect endothelial cells [116]. By secreting metal- for which an increased surface expression has been found in
loproteinases, especially matrix metalloproteinase (MMP)- atopic patients [134]. Endobronchial eosinophils are able to
9, eosinophils may directly aect airway remodeling [117]. process inhaled antigens, trac to the regional lymph nodes
MMP-9 has been found to be increased in severe persistent where they stimulate CD4 T-cell responses [135]. Those
asthma and following allergen challenge [118]. That eosi- lymph node eosinophils phenotypically resemble dendritic
nophils play a role in the late-phase reaction and remod- cells and express MHC class II, CD80, and CD86 [136].
eling processes [112] is consistent with the observation that
eosinophils are abundant in the submucosa of lung tissues
from patients with slow-onset fatal asthma [119]. Chronic obstructive pulmonary disease
Eosinophils synthesize proangiogenic mediators
such as VEGF and FGF-2 and thus are able to promote In the pathogenesis of COPD, neutrophils, macrophages,
angiogenesis, which presents an additional important and CD8 T-cells are considered as key eector cells [137,
mechanism in chronic inammation and tissue remodeling 138]. The presence and role of eosinophils in COPD is con-
[120]. In bronchial biopsies of asthmatics, eosinophils are troversial [138]. For instance, airway eosinophilia has been
positively stained for VEGF and FGF-2 [121]. VEGF is reported occurring during acute exacerbations of COPD
released upon stimulation with GM-CSF and IL-5 [122]. [139] and has also been observed in patients with stable
Furthermore, eosinophils can promote angiogenesis directly disease [140]. However, in one study, eosinophil numbers in
by secreting extracellular matrix-degrading enzymes such as induced sputum taken during a stable disease period were
MMP-9 and heparanase [123] or indirectly by IL-8 secre- compared with those during an acute exacerbation in the
tion enhancing MMP-2 and MMP-9 mRNA expression same patients, and no dierences have been observed [141].
by endothelial cells [120]. In contrast; VEGF produced Eosinophilia in COPD has been linked to smoking [142] as
by inammatory and structural cells such as macrophages, well as to an asthmatic component [143]. Moreover, airway
neutrophils, epithelial cells, broblasts, and smooth muscle obstruction was observed to correlate with an increase in
cells, induces migration and mediator release by eosinophils the number of activated eosinophils in the airways [144].

150
Eosinophils 12
Interestingly, eosinophilia in COPD does not seem to Leukotriene antagonists
depend on IL-5 [145], but is linked to an upregulation of
RANTES [146] as well as eotaxin and CCR3 [147]. The Leukotriene antagonists are well-established substances in
production of RANTES may be induced by TNF-, which the treatment of asthma that inhibit the eect of cysteinyl
is increased in acute exacerbations [148]. Elevated levels of leukotrienes, which are generated in high amounts by eosi-
IL-6 and IL-8 were found in the sputum during exacerba- nophils, mast cells, basophils, macrophages, and monocytes
tions of COPD correlating with the number of inamma- during asthmatic inammation [156]. A second way to
tory cells in particular eosinophils [141]. IL-8, although interfere with leukotrienes is the inhibition of their genera-
usually associated with neutrophil chemotaxis, has been tion by blocking the 5-lipoxygenase pathway [157].
suggested to recruit and activate eosinophils in COPD
[139, 149].
The principal mechanisms by which eosinophils Interferons
contribute to airway inammation have been suggested
as being similar to those in asthma [148]. In patients with The treatment with low-dose IFN- was shown to be eec-
COPD, higher numbers of eosinophils have been found in tive in the treatment of patients with severe corticosteroid
bronchial biopsies compared to healthy controls accompa- resistant asthma with or without ChurgStrauss syndrome.
nied by increased eosinophil and ECP levels in bronchoal- The establishment of a correct T helper 1/T helper 2 bal-
veolar lavage uid and induced sputum [150]. In addition ance as well as the induction of IL-10 are regarded as possi-
to ECP, EPO levels are raised in the sputum in patients ble mechanisms [158]. IFN- therapy was shown to reduce
with COPD [151]. Moreover, several studies reported blood eosinophil numbers in patients with severe steroid-
eosinophil inltration of the airway walls associated with dependent asthma. However, physiological asthma param-
increased amounts of ECP in bronchoalveolar lavage uid eters, such as FEV1 and peak expiratory ow rates, did not
and induced sputum during exacerbations of COPD [138, improve upon treatment with IFN- over 90 days [159].
148, 152]. Since COPD exacerbations may be associated
with viral infections, eosinophils are suggested to have a role Anti-IL-5 monoclonal antibodies
in antiviral host defense, for example, by releasing ECP and
EDN acting as ribonucleases [138]. The rst study using a monoclonal antibody to IL-5 investi-
Eosinophilic airway inammation is suggested to gated the eect and safety of a single dose of mepolizumab in
contribute to airow obstruction and symptoms in patients patients with mild bronchial asthma [160]. Although peripheral
with COPD [143]. The patients with sputum eosinophilia blood and sputum eosinophil numbers signicantly decreased,
are more likely to respond to corticosteroid therapy [143]. this treatment had no eect either on airway hyperresponsive-
Therefore, sputum eosinophilia may be used as predictive ness before and after allergen challenge or on the allergen-
marker in identifying patients who will benet from corti- induced late asthmatic response. A second study, in which the
costeroid therapy [153]. same anti-IL-5 antibody was given over 3 months, demon-
strated that the expression of activation markers and T helper
2-cytokine receptors on eosinophils as well as the number of
circulating T-cells and their capacity for generating cytokines
ANTI-EOSINOPHILIC THERAPIES were not aected [161]. Another antibody (SCH55700) was
tested in patients with severe asthma. A single dose of this
anti-IL-5 antibody resulted in a short improvement of baseline
Since eosinophils exhibit a multifunctional role in the patho- FEV1 and decrease of blood eosinophil numbers, but failed to
genesis of asthma, therapeutic approaches aiming to reduce improve asthma symptoms signicantly [162]. Investigating
their numbers and/or activity seem promising. There are sev- eosinophil numbers at dierent sites revealed that anti-IL-5
eral potential mechanisms for how to do so: reduce eosinophil therapy produced only a partial decrease of 52% in the bone
production, stop eosinophil migration into inamed tissues, marrow, and of 55% in the airways, but a 100% decrease in
activate eosinophil apoptosis, prevent eosinophil priming and the peripheral blood [163]. The anti-IL-5 antibody seemed
activation, stop generation and release of mediators, and/or to exclusively reduce mature eosinophils in the bone marrow
antagonize them. Here, we shortly discuss established as well as and in the peripheral blood, whereas CD34/IL-5 receptor
new therapeutic options which specically attack eosinophils. progenitor cells were not reduced [52]. However, the observed
decrease of eosinophil progenitor cells in the bronchial mucosa
suggested that local eosinophilopoiesis was impaired upon
anti-IL-5 therapy. The reason why a single anti-IL-5 treatment
Immunosuppressive therapy failed to improve asthma might be the redundancy of dier-
ent cells and cytokines contributing to eosinophilia and asthma
Corticosteroids are widely used in the treatment of bron-
symptoms.
chial asthma and COPD. Although corticosteroids increase
the rate of eosinophil apoptosis [154], their main eect on
eosinophils appears to be due to inhibition of cytokine and Anti-TNF-
chemokine production by leukocytes, such as T-cells and
resident airway cells [155]. The same mechanism is true for The proinammatory cytokine TNF- plays a role in leu-
other immunosuppressive drugs such as cyclosporine. kocyte recruitment and activation. Although anti-TNF-

151
Asthma and COPD: Basic Mechanisms and Clinical Management

therapy was shown to improve asthma symptoms, it did not References


aect the number of sputum eosinophils [164].
1. Ehrlich P. Beitrge zur Kenntnis der granulierenden Bindegewebszellen
und der eosinophilen Leukozyten. Arch Anat Physiol: 16669, 1879.
2. Simon D, Simon HU. Eosinophilic disorders. J Allergy Clin Immunol
IL-12 119: 1291300, 2007.
3. Simon HU, Youse S, Schranz C, Schapowal A, Bachert C, Blaser K.
IL-12 is a macrophage-derived cytokine that modulates Direct demonstration of delayed eosinophil apoptosis as a mechanism
T-cell responses and therefore might reduce eosinophilic causing tissue eosinophilia. J Immunol 158: 39028, 1997.
inammation. Recombinant human IL-12, administered to 4. Rothenberg ME, Hogan SP. The eosinophil. Annu Rev Immunol 24:
patients with mild asthma in three increasing dosages over 14774, 2006.
3 weeks, resulted in a decrease of eosinophil numbers in the 5. Kariyawasam HH, Robinson DS. The eosinophil: The cell and its
peripheral blood and in the sputum after allergen challenge weapons, the cytokines, its locations. Semin Respir Crit Care Med 27:
as well as in an improvement of the allergen induced airway 11727, 2006.
6. Ackerman SJ, Liu L, Kwatia MA, Savage MP, Leonidas DD,
hyperresponsiveness [165]. However, severe adverse events
Swaminathan GJ, Acharya KR. Charcot-Leyden crystal protein
leading to withdrawal were seen in 4 out of 19 patients. (galectin-10) is not a dual function galectin with lysophospholipase
activity but binds a lysophospholipase inhibitor in a novel structural
function. J Biol Chem 277: 1485968, 2002.
Selectin antagonists 7. Gleich GJ, Adolphson CR. The eosinophilic leukocyte: Structure and
function. Adv Immunol 39: 177253, 1986.
Because recruitment of inammatory cells from the periph- 8. Bandeira-Melo C, Bozza PT, Weller PF. The cellular biology of eosi-
eral blood into the airways is a characteristic feature of nophil eicosanoid formation and function. J Allergy Clin Immunol 109:
asthma, targeting selectins or their ligands is a promising 393400, 2002.
approach in the treatment of asthma [166]. Bimosiamose, 9. Prussin C, Metcalfe DD. IgE, mast cells, basophils and eosinophils.
J Allergy Clin Immunol 111: S48694, 2003.
a pan-selectin antagonist, was studied in patients with mild
10. Leiferman KM, Ackerman SJ, Sampson HA, Haugen HS, Venencie PY,
allergic asthma revealing an inhibitory eect on late-phase Gleich GJ. Dermal deposition of eosinophil-granule major basic pro-
responses but not on airway hyperresponsiveness or exhaled tein in atopic dermatitis: Comparison with onchocerciasis. N Engl J
NO after allergen challenge [167]. Med 313: 28285, 1985.
11. Voehringer D, van Rooijen N, Locksley M. Eosinophils develop in dis-
tinct stages and are recruited to peripheral sites by alternatively acti-
CCR3 antagonists vated macrophages. J Leukoc Biol 81: 143444, 2007.
12. Kato M, Kephart GM, Talley NJ, Wagner JM, Sarr MG, Bonno M,
To block the principal chemokine receptor of eosinophils, McGovern TW, Gleich GJ. Eosinophil inltration and degranulation
the eect of a low-molecular weight CCR3 antagonist has in normal human tissues. Anat Rec 252: 41825, 1998.
13. Gouon-Evans V, Rothenberg ME, Pollard JW. Postnatal mammary
been investigated in a mouse asthma model. This revealed
gland development requires macrophages and eosinophils. Development
promising results as it markedly reduced eosinophil inltra- 127: 226982, 2000.
tion of the airways followed by a normalization of airway 14. Gouon-Evans V, Pollard JW. Eotaxin is required for eosinophil hom-
hyperresponsiveness, reduced mucus production and pre- ing into the stroma of the pubertal and cycling uterus. Endocrinology
vention of airway remodeling [168]. 142: 451521, 2001.
15. Furuta G. Emerging questions regarding eosinophils role in the esophago-
gastrointestinal tract. Curr Opin Gastroenterol 22: 65863, 2006.
Integrin inhibitors 16. Matthews AN, Friend DS, Zimmermann N, Sara MN, Luster AD,
Pearlman E, Wert SE, Rothenberg ME. Eotaxin is required for the base-
Since intergrins are involved in migration of inammatory line level of tissue eosinophils. Proc Natl Acad Sci USA 95: 627378, 1988.
cells including eosinophils from the blood vessels into the 17. Mller E. Localization of eosinophils in the thymus by the peroxidase
reaction. Histochemistry 52: 27379, 1977.
tissues, antagonizing integrins oers another promising
18. Lee I, Yu E, Good RA, Ikehara S. Presence of eosinophilic precursors
treatment option for asthma or allergic diseases. In vitro in the human thymus: Evidence for intra-thymic dierentiation of cells
and in vivo experiments showed that a newly developed in eosinophilic lineage. Pathol Int 45: 65562, 1995.
small molecule VLA-4 (41 integrin) inhibitor may pre- 19. Throsby M, Herbelin A, Pleau JM, Dardenne M. CD11c eosinophils
vent VLA-4/VCAM interactions resulting in reduced eosi- in the murine thymus: Developmental regulation and recruitment upon
nophil migration and inltration in the tissue [169]. MHC class I-restricted thymocyte deletion. J Immunol 165: 196575,
2000.
20. Klion AD, Nutman TB. The role of eosinophils in host defense against
helminth parasites. J Allergy Clin Immunol 113: 3037, 2004.
21. Jacobsen EA, Taranova AG, Lee NA, Lee JJ. Eosinophils: Singularly
ACKNOWLEDGMENTS destructive eector cells or purveyors of immunoregulation. J Allergy
Clin Immunol 119: 131320, 2007.
22. Gleich GJ, Frigas E, Loegering DA, Wassom DL, Steinmuller D.
Work in the laboratory of Hans-Uwe Simon is supported Cytotoxic properties of the eosinophil major basic protein. J Immunol
by grants from the Swiss National Science Foundation 123: 292527, 1979.
(grant #310000-107526), the Stanley Thomas Johnson 23. Kroegel C, Costabel U, Matthys H. Mechanism of membrane damage
Foundation, Bern, and the OPO-Foundation, Zurich. mediated by eosinophil major basic protein. Lancet 1: 138081, 1987.

152
Eosinophils 12
24. Gleich GJ, Loegering DA, Bell MP, Checkel JL, Ackerman SJ, 45. Lucey DR, Nicholson-Weller A, Weller PF. Mature human eosinophils
McKean DJ. Biochemical and functional similarities between human have the capacity to express HLA-DR. Proc Natl Acad Sci USA 86:
eosinophil-derived neurotoxin and eosinophil cationic protein: Homology 134851, 1989.
with ribonucleases. Proc Natl Acad Sci USA 83: 314650, 1986. 46. Yamamoto H, Sedgwick J, Vrtis RF, Busse WW. The eect of transen-
25. Rosenberg HF, Domachowske JB. Eosinophils, eosinophil ribonucle- dothelial migration on eosinophil function. Am J Respir Cell Mol Biol
ases, and their role in host defence against respiratory virus pathogens. 23: 37988, 2000.
J Leukoc Biol 70: 69198, 2001. 47. van Rijt LS, Vos N, Hijdra D, de Vries VC, Hoogsteden HC,
26. Young JD, Peterson CG, Venge P, Cohn ZA. Mechanism of membrane Lambrecht BN. Airway eosinophils accumulate in the mediastinal
damage mediated by human eosinophil cationic protein. Nature 321: lymph nodes but lack antigen-presenting potential for nave T cells.
61316, 1986. J Immunol 171: 337278, 2003.
27. Barker RL, Loegering DA, Ten RM, Hamann KJ, Pease LR, Gleich 48. Denburg JA, Telizyn S, Messner H, Lim B, Jamal N, Ackerman SJ,
GJ. Eosinophil cationic protein cDNA. Comparison with other toxic Gleich GJ, Bienenstock J. Heterogeneity of human peripheral blood
cationic proteins and ribonucleases. J Immunol 143: 95255, 1989. eosinophil-type colonies: Evidence for a common basophileosinophil
28. Simon HU, Weber M, Becker E, Zilberman Y, Blaser K, Levi-Schaer F. progenitor. Blood 66: 31218, 1985.
Eosinophils maintain their capacity to signal and release eosinophil 49. Sanderson CJ. Interleukin-5, eosinophils, and disease. Blood 79: 31019,
cationic protein upon repetitive stimulation with the same agonist. 1992.
J Immunol 165: 406975, 2000. 50. Foster P, Hogan P, Ramsay AJ, Matthaei KI, Young IG. Interleukin-5
29. Yang D, Rosenberg HF, Chen Q, Dyer KD, Kurosaka K, Oppenheim JJ. deciency abolishes eosinophilia, airway hyperreactivity and lung dam-
Eosinophil-derived neurotoxin (EDN), an antimicrobial protein with age in a mouse asthma model. J Exp Med 83: 195201, 1996.
chemotactic activities for dendritic cells. Blood 102: 3396403, 2003. 51. Dent LA, Strath M, Mellor AL, Sanderson CJ. Eosinophilia in trans-
30. Logan MR, Odemuyiwa SO, Moqbel R. Understanding exocytosis in genic mice expressing interleukin-5. J Exp Med 172: 142531, 1990.
immune and inammatory cells: The molecular basis of mediator secre- 52. Menzies-Gow A, Flood-Page P, Sehmi R, Burman J, Hamid Q,
tion. J Allergy Clin Immunol 111: 92332, 2003. Robinson DS, Kay AB, Denburg J. Anti-IL-5 (mepolizumab) therapy
31. Scepek S, Moqbel R, Lindau M. Compound exocytosis and cumulative induces bone marrow eosinophil progenitors in bronchial mucosa of
degranulation by eosinophils and its role in parasite killing. Parasitol atopic asthmatics. J Allergy Clin Immunol 111: 71419, 2003.
Today 10: 27678, 1994. 53. Nishinakamura R, Miyajima A, Mee PJ, Tybulewicz VL, Murray R.
32. Fasshauer D, Sutton RB, Brunger AT, Jahn R. Conserved structural Hematopoiesis in mice lacking the entire granulocyte-macrophage
features of the synaptic fusion complex: SNARE proteins reclassied colony-stimulating factor/interleukin3/interleukin-5 functions. Blood
as Q- and R-SNAREs. Proc Natl Acad Sci USA 95: 1578186, 1998. 88: 245864, 1996.
33. Shult PA, Graziano FM, Wallow IH, Busse WW. Comparison of 54. Palframan RT, Collins PD, Severs NJ, Rothery S, Williams TJ,
superoxide generation and luminol-dependent chemiluminescence with Rankin SM. Mechanisms of acute eosinophil mobilization from the bone
eosinophils and neutrophils from normal individuals. J Lab Clin Med marrow stimulated by interleukin-5: The role of specic adhesion mol-
106: 63845, 1985. ecules and phosphatidylinositol 3-kinase. J Exp Med 188: 162132, 1998.
34. Wang J, Slungaard A. Role of eosinophil peroxidase in host defense 55. Mishra A, Hogan SP, Lee JJ, Foster PS, Rothenberg ME. Fundamental
and disease pathology. Arch Biochem Biophys 445: 25660, 2006. signals that regulate eosinophil homing to the gastrointestinal tract.
35. Nakajima H, Gleich GJ, Kita H. Constitutive production of IL-4 and J Clin Invest 103: 171927, 1999.
IL-10 and stimulation of IL-8 by normal peripheral blood eosinophils. 56. Ponath PD, Qin S, Post TW, Wang J, Wu L, Gerard NP, Newman W,
J Immunol 156: 485966, 1996. Gerard C, Mackay CR. Molecular cloning and characterization of a
36. Lampinen M, Carlson M, Hakansson LD, Venge P. Cytokine-regulated human eotaxin receptor expressed selectively on eosinophils. J Exp Med
accumulation of eosinophils in inammatory disease. Allergy 59: 793 183: 243748, 1996.
805, 2004. 57. Sehmi R, Dorman S, Baatjes A, Watson R, Foley R, Ying S,
37. Youse S, Hemmann S, Weber M, Hlzer C, Hartung K, Blaser K, Robinson DS, Kay AB, OByrne PM, Denburg JA. Allergen-induced
Simon HU. IL-8 is expressed by human peripheral blood eosinophils. uctuation in CC chemokine receptor 3 expression on bone marrow
Evidence for increased secretion in asthma. J Immunol 154: 548190, CD34 cells from asthmatic subjects: Signicance for mobilization
1995. of haemopoietic progenitor cells in allergic inammation. Immunology
38. Lamkhioued B, Gounni AS, Aldebert D, Delaporte E, Prin L, Capron A, 109: 53646, 2003.
Capron M. Synthesis of type 1 (IFN-gamma) and type 2 (IL-4, IL-5, 58. Bruijnzeel PL. Eosinophil tissue mobilization in allergic disorders. Ann
and IL-10) cytokines by human eosinophils. Ann NY Acad Sci 796: NY Acad Sci 725: 25967, 1994.
2038, 1996. 59. Luijk B, Lindemans CA, Kanters D, van der Heijde R, Bertics P,
39. Schmid-Grendelmeier P, Altznauer F, Fischer B, Bizer C, Straumann A, Lammers JW, Bates ME, Koenderman L. Gradual increase in prim-
Menz G, Blaser K, Wthrich B, Simon HU. Eosinophils express func- ing of human eosinophils during extravasation from peripheral blood
tional IL-13 in eosinophilic inammatory diseases. J Immunol 169: to the airways in response to allergen challenge. J Allergy Clin Immunol
102127, 2002. 115: 9971003, 2005.
40. Gleich GJ. Mechanisms of eosinophil-associated inammation. 60. Spoelstra FM, Postma DS, Hovenga H, Noordhoek JA, Kauman HF.
J Allergy Clin Immunol 105: 65163, 2000. Interferon-gamma and interleukin-4 dierentially regulate ICAM-1
41. Nagase H, Okugawa S, Ota Y, Yamaguchi M, Tomizawa H, and VCAM-1 expression on human lung broblasts. Eur Respir J 14:
Matsushima K, Ohta K, Yamamoto K, Hirai K. Expression and func- 75966, 1999.
tion of Toll-like receptors in eosinophils: Activation by Toll-like recep- 61. Rosenberg HF, Phipps S, Foster PS. Eosinophil tracking in allergy
tor 7 ligand. J Immunol 171: 397782, 2003. and asthma. J Allergy Clin Immunol 119: 130310, 2007.
42. Thomet OAR, Wiesmann UN, Blaser K, Simon HU. Dierential inhi- 62. Elsner J, Kapp A. Regulation and modulation of eosinophil eector
bition of inammatory eector functions by petasin, isopetasin and functions. Allergy 54: 1526, 1999.
neopetasin in human eosinophils. Clin Exp Allergy 31: 131020, 2001. 63. Homey B, Zlotnik A. Chemokines in allergy. Curr Opin Immunol 11:
43. Takafuji S, Bischo SC, De Weck AL, Dahinden CA. IL-3 and IL- 62634, 1999.
5 prime normal human eosinophils to produce leukotriene C4 in 64. Nagase H, Yamaguchi M, Jibidi S, Yamada H, Ohta K, Kawasaki H,
response to soluble agonists. J Immunol 147: 385561, 1991. Yoshie O, Yamamoto K, Morita Y, Hirai K. Eosinophil chemotaxis by
44. Shi HZ. Eosinophil functions as antigen presenting cells. J Leukoc Biol chemokines: A study by a simple photometric assay. Allergy 54: 94450,
76: 52027, 2004. 1999.

153
Asthma and COPD: Basic Mechanisms and Clinical Management

65. Shahabuddin S, Ponath P, Schleimer RP. Migration of eosinophils delayed eosinophil apoptosis mediated by granulocyte-macrophage
across endothelial cell monolayers: Interactions among IL-5, endothelial- colony-stimulating factor and interleukin-5. Blood 92: 77883, 1998.
activating cytokines, and C-C chemokines. J Immunol 164: 384754, 2000. 85. Simon HU. Molecules involved in the regulation of eosinophil
66. Phillips RM, Stubbs VE, Henson MR, Williams TJ, Pease JE, Sabroe apoptosis. Chem Immunol Allergy 91: 4958, 2006.
I. Variations in eosinophil chemokine responses: An investigation of 86. Tsukahara K, Nakao A, Hiraguri M, Miike S, Mamura M, Saito Y,
CCR1 and CCR3 function, expression in atopy, and identication of a Iwamoto I. Tumor necrosis factor-alpha mediates antiapoptotic sig-
functional CCR1 promoter. J Immunol 170: 6190201, 2003. nals partially via p38MAP kinase activation in human eosinophils.
67. DiScipio RG, Daern PJ, Jagels MA, Broide DH, Sriramarao P. Int Arch Allergy Immunol 120: S5459, 1999.
A comparison of C3a and C5a-mediated stable adhesion of rolling 87. Robertson NM, Zangrilli JG, Steplewski A, Hastie A, Lindemeyer RG,
eosinophils in postcapillary venules and transendothelial migration in Planeta MA, Smith MK, Innocent N, Musani A, Pascual R, Peters S,
vitro and in vivo. J Immunol 162: 112736, 1999. Litwack G. Dierential expression of TRAIL and TRAIL receptors
68. Warringa RA, Mengelers HJ, Kuijper PH, Raaijmakers JA, Bruijnzel PL, in allergic asthmatics following segmental antigen challenge: Evidence
Koenderman L. In vivo priming of platelet-activating factor-induced for a role of TRAIL in eosinophil survival. J Immunol 169: 598696,
eosinophil chemotaxis in allergic asthmatic individuals. Blood 79: 2002.
183641, 1992. 88. Heinisch IV, Bizer C, Volgger W, Simon HU. Functional CD137
69. Tager AM, Dufour JH, Goodarzi K, Bercury SD, von Adrian UH, receptors are expressed by eosinophils from patients with IgE-
Luster AD. BLTR mediates leukotriene (4)-induced chemotaxis and mediated allergic responses but not by eosinophils from patients with
adhesion and plays a dominant role in eosinophil accumulation in a non-IgE-mediated eosinophilic disorders. J Allergy Clin Immunol 108:
murine model of peritonitis. J Exp Med 192: 43946, 2000. 2128, 2001.
70. Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori Y, 89. Hebestreit H, Youse S, Balatti I, Weber M, Crameri R, Simon D,
Ichimasa M, Sugamura K, Nakamura M, Takano S, Nagata K. Hartung K, Schapowal A, Blaser K, Simon HU. Expression and func-
Prostaglandin D2 selectively induces chemotaxis in T-helper type 2 cells, tion of the Fas receptor on human blood and tissue eosinophils. Eur J
eosinophils, and basophils via seven-transmembrane receptor CRTH2. Immunol 26: 177580, 1996.
J Exp Med 193: 25562, 2001. 90. Simon HU. Eosinophil apoptosis pathophysiologic and therapeutic
71. Bandeira-Melo C, Bozza PT, Dias BL, Cordeiro RS, Jose PJ, implications. Allergy 55: 91015, 2000.
Martins MA, Serhan CN. Lipoxin (LX) A4 and aspirin-triggered 91. Wardlaw AJ, Brightling C, Green R, Woltmann G, Pavord I.
15-epi-LXA4 block allergen-induced eosinophil tracking. J Immunol Eosinophils in asthma and other allergic diseases. Br Med Bull 56:
164: 226771, 2000. 9851003, 2000.
72. Pope SM, Brandt EB, Mishra A, Hogan SP, Zimmermann N, 92. Bousquet J, Chanez P, Lacoste JY, Barneon G, Ghavanian N, Enander I,
Matthei KI, Foster PS, Rothenberg ME. IL-13 induces eosinophil Venge P, Ahlstedt S, Simony-Lafontaine J, Godard P. Eosinophilic
recruitment into the lung by an IL-15- and eotaxin-dependent mecha- inammation in asthma. N Engl J Med 323: 103339, 1990.
nism. J Allergy Clin Immunol 108: 594601, 2001. 93. Lee JJ, Dimina D, Macias MP, Ochkur SI, McGarry MP, ONeill KR,
73. Simon HU, Blaser K. Inhibition of programmed eosinophil death: A key Protheroe C, Pero R, Nguyen T, Cormier SA, Lenkiewicz E, Colbert D,
pathogenic event for eosinophilia?. Immunology Today 16: 5355, 1995. Rinaldi L, Ackerman SJ, Irvin CG, Lee NA. Dening a link with
74. Kankaanranta H, Lindsay MA, Giembycz MA, Zhang X, Moilanan E, asthma in mice congenitally decient in eosinophils. Science 305:
Barnes PJ. Delayed eosinophil apoptosis in asthma. J Allergy Clin 177376, 2004.
Immunol 106: 7783, 2000. 94. Humbles AA, Lloyd CM, McMillan SJ, Friend DS, Xanthou G,
75. Woolley KL, Gibson PG, Carty K, Wilson AJ, Twaddell SH, McKenna EE, Ghiran S, Gerard NP, Yu C, Orkin SH, Gerard C.
Woolley MJ. Eosinophil apoptosis and the resolution of airway inam- A critical role for eosinophils in allergic airways remodelling. Science
mation in asthma. Am J Respir Crit Care Med 154: 23743, 1996. 305: 177679, 2004.
76. Sexton DW, Blaylock MG, Walsh GM. Human alveolar epithelial cells 95. Frigas E, Motojima S, Gleich GJ. The eosinophilic injury to the
engulf apoptotic eosinophils by means of integrin- and phosphatidyl- mucosa of the airways in the pathogenesis of bronchial asthma. Eur
serine receptor-dependent mechanisms: A process upregulated by dex- Respir J 13: S12335, 1991.
amethasone. J Allergy Clin immunol 108: 96269, 2001. 96. Sedgwick JB, Vrtis RF, Jansen KJ, Kita H, Bartemes K, Busse WW.
77. Uller L, Andersson M, Grei L, Persson CG, Erjeflt JS. Occurrence Peripheral blood eosinophils from patients with allergic asthma con-
of apoptosis, secondary necrosis, and cytolysis in eosinophil nasal tain increased intracellular eosinophil-derived neurotoxin. J Allergy
polyps. Am J Respir Crit Care Med 170: 74247, 2004. Clin Immunol 114: 56874, 2004.
78. Yamaguchi Y, Suda T, Ohta S, Tominaga K, Miura Y, Kasahara T. 97. Venge P, Bystrm J, Carlson M, Hakansson L, Karawacjzyk M,
Analysis of the survival of mature eosinophils: Interleukin-5 prevents Peterson C, Seveus L, Trulson A. Eosinophil cationic protein (ECP):
apoptosis in human eosinophils. Blood 78: 254247, 1991. Molecular and biological properties and the use of ECP as a marker
79. Conus S, Bruno A, Simon HU. Leptin is an eosinophil survival factor. of eosinophil activation in disease. Clin Exp Allergy 29: 117286,
J Allergy Clin Immunol 116: 122834, 2005. 1999.
80. Youse S, Hoessli DC, Blaser K, Mills GB, Simon HU. Requirement 98. Munthe-Kaas MC, Gerritsen J, Carlsen KH, Undlien D, Egeland T,
of Lyn and Syk tyrosine kinases for the prevention of apoptosis by Skinningsrud B, Torres T, Carlsen KL. Eosinophil cationic protein
cytokines in human eosinophils. J Exp Med 183: 140714, 1996. (ECP) polymorphisms and association with asthma, s-ECP levels
81. Simon HU, Youse S, Dibbert B, Levi-Schaer F, Blaser K. Anti- and related phenotypes. Allergy 62: 42936, 2007.
apoptotic signals of granulocyte-macrophage colony-stimulating factor 99. Saleh D, Ernst P, Lim S, Barnes PJ, Giaid A. Increased formation of
are transduced via Jak2 tyrosine kinase in eosinophils. Eur J Immunol the potent oxidant peroxynitrite in the airways of asthmatic patients
27: 353639, 1997. is associated with induction of nitric oxide synthase: Eect of inhaled
82. Vassina EM, Youse S, Simon D, Zwicky C, Conus S, Simon HU. glucocorticoid. FASEB J 12: 92937, 1998.
cIAP-2 and survivin contribute to cytokine-mediated delayed eosi- 100. Kaminsky DA, Mitchell J, Carroll N, James A, Soultanakis R, Janssen
nophil apoptosis. Eur J Immunol 36: 197584, 2006. Y. Nitrotyrosine formation in the airways and lung parenchyma of
83. Huang HM, Huang CJ, Yen JJ. Mcl-1 is a common target of stem cell patients with asthma. J Allergy Clin Immunol 104: 74754, 1999.
factor and interleukin-5 for apoptosis in prevention activity via MEK/ 101. Alving K, Weitzberg E, Lundberg JM. Increased amount of nitric
MAPK and PI-3K/Akt pathways. Blood 96: 176471, 2000. oxide in exhaled air of asthmatics. Eur Respir J 6: 136870, 1993.
84. Dibbert B, Daigle I, Braun D, Schranz C, Weber M, Blaser K, 102. Turner S. The role of exhaled nitric oxide in the diagnosis, manage-
Zangemeister-Wittke U, Akbar AN, Simon HU. Role for Bcl-xL in ment and treatment of asthma. Mini Rev Med Chem 7: 53942, 2007.

154
Eosinophils 12
103. Green RH, Brightling CE, McKenna S, Hargadon B, Parker D, colony-stimulating factor and interleukin-5. Am J Respir Cell Mol Biol
Bradding P, Wardlaw AJ, Pavord ID. Asthma exacerbations and spu- 17: 7077, 1997.
tum eosinophil counts: A randomised controlled trial. Lancet 360: 123. Temkin V, Aingorn H, Puxeddu I, Goldshmidt O, Zcharia E,
171521, 2002. Gleich GJ, Vlodavski I, Levi-Schaer F. Eosinophil major basic
104. Harrison AM, Bonville CA, Rosenberg HF, Domachowske JB. protein: First identied natural heparanase-inhibiting protein. J Allergy
Respiratory syncytial viral-induced chemokine expression in the lower Clin Immunol 113: 7039, 2004.
airways: Eosinophil recruitment and degranulation. Am J Respir Crit 124. Freisitzer C, Kaneider NC, Sturn DH, Mosheimer BA, Khler CM,
Care Med 159: 191824, 1999. Wiedermann CJ. Expression and function of the vascular endothelial
105. ODonnell MC, Ackerman SJ, Gleich GJ, Thomas LL. Activation of growth factor receptor FLT-1 in human eosinophils. Am J Respir Cell
basophil and mast cell histamine release by eosinophil granule major Mol Biol 30: 72935, 2004.
basic protein. J Exp Med 157: 198191, 1983. 125. Lee YC, Kwak YG, Song CH. Contribution of vascular endothelial
106. Kita H, Abu-Ghazaleh RI, Sur S, Gleich GJ. Eosinophil major basic growth factor to airway hyperresponsiveness and inammation in a
protein induces degranulation and IL-8 production by human eosi- murine model of toluene diisocyanate-induced asthma. J Immunol
nophils. J Immunol 154: 474958, 1995. 168: 3595600, 2002.
107. Moy JN, Gleich GJ, Thomas LL. Noncytotoxic activation of neu- 126. Joubert P, Hamid Q. Role of airway smooth muscle in airway remod-
trophils by eosinophil granule major basic protein. Eect on super- elling. J Allergy Clin Immunol 116: 71316, 2005.
oxide anion generation and lysosomal enzyme release. J Immunol 145: 127. Weller PF, Lee CW, Foster DW, Corey EJ, Austen KF, Lewis RA.
262632, 1990. Generation and metabolism of 5-lipoxygenase pathway leukotrienes
108. Page SM, Gleich GJ, Roebuck KA, Thomas LL. Stimulation of neu- by human eosinophils: Predominant production of leukotriene C4.
trophil interleukin-8 production by eosinophil granule major basic Proc Natl Acad Sci USA 80: 762630, 1983.
protein. Am J Respir Cell Mol Biol 21: 23037, 1999. 128. Cowburn AS, Sladek K, Soja J, Adamek L, Nizankowska E,
109. Rohrbach MS, Wheatley CL, Slifman NR, Gleich GJ. Activation Szczeklik A, Lam BK, Penrose JF, Austen KF, Holgate ST,
of platelets by eosinophil granule proteins. J Exp Med 172: 127174, Sampson AP. Overexpression of leukotriene C4 synthase in bronchial
1990. biopsies from patients with aspirin-tolerant asthma. J Clin Invest 101:
110. Jacoby DB, Gleich GJ, Fryer AD. Human eosinophil major basic pro- 83446, 1998.
tein is an endogenous allosteric antagonist at the inhibitory muscarine 129. Marom Z, Shelhamer JH, Bach MK, Morton DR, Kaliner M. Slow-
M2 receptor. J Clin Invest 91: 131418, 1993. reacting substances, leukotrienes C4 and D4, increase the release of
111. Pascual RM, Peters SP. Airway remodelling contributes to the pro- mucus from human airways in vitro. Am Rev Respir Dis 126: 44951,
gressive loss of lung function in asthma: An overview. J Allergy Clin 1982.
Immunol 116: 47786, 2005. 130. Adelroth E, Morris MM, Hargreave FE, OByrne PM. Airway respon-
112. Kay AB, Phipps S, Robinson DS. A role for eosinophils in airway siveness to leukotrienes C4 and D4 and to methacholine in patients
remodelling in asthma. Trends Immunol 25: 47782, 2004. with asthma and normal controls. N Engl J Med 315: 48084, 1986.
113. Flood-Page P, Menzies Gow A, Phipps S, Ying S, Wangoo A, 131. Lee E, Robertson T, Smith J, Kilfeather S. Leukitriene receptor
Ludwig MS, Barnes N, Robinson S, Kay AB. Anti-IL-5 treatment antagonists and synthesis inhibitors reverse survival in eosinophils of
reduces deposition of ECM proteins in the bronchial subepithelial asthmatic individuals. Am J Respir Crit Care Med 161: 188186, 2000.
basement membrane of mild atopic asthmatics. J Clin Invest 112: 132. Sedgwick JB, Calhoun WJ, Vrtis RF, Bates ME, McAllister PK, Busse
102936, 2003. WW. Comparison of airway and blood eosinophil function after in
114. Foley SC, Prefontaine D, Hamid Q. Role of eosinophils in airway vivo allergen challenge. J Immunol 149: 371018, 1992.
remodeling. J Allergy Clin Immunol 119: 156366, 2007. 133. Mengelers HJ, Maikoe T, Brinkman L, Hooibrink B, Lammers JW,
115. Rochester CL, Ackerman SJ, Zheng T, Elias JA. Eosinophil-broblast Koenderman L. Immunophenotyping of eosinophils recovered from
interactions. Granule major basic protein interacts with IL-1 and blood and BAL of allergic asthmatics. Am J Respir Crit Care Med 149:
transforming growth factor-beta in the stimulation of lung broblast 34551, 1994.
IL-6-type cytokine production. J Immunol 156: 444956, 1996. 134. Ohkawara Y, Lim KG, Xing Z, Glibetic M, Nakano K, Dolovich
116. Wang JG, Mahmud SA, Thompson JA, Geng JG, Key NS, Slungaard A. J, Croitoru K, Weller PF, Jordana M. CD40 expression by human
The principal eosinophil peroxidase product, HOSNC, is a uniquely peripheral blood eosinophils. J Clin Invest 97: 176166, 1996.
potent phagocyte oxidant inducer of endothelial cell tissue factor 135. Shi HZ, Humbles A, Gerard C, Jin Z, Weller PF. Lymph node traf-
activity: A potential mechanism for thrombosis in eosinophilic inam- cking and antigen presentation by endobronchial eosinophils. J Clin
matory states. Blood 107: 55865, 2006. Invest 105: 94553, 2000.
117. Wiehler S, Cuvelier SL, Chakrabarti S, Patel KD. p38 MAP kinase 136. Duez C, Dakhama A, Tomkinson A, Marquillies P, Balhorn A,
regulates rapid matrix metalloproteinase-9 releases from eosinophils. Tonnel AB, Bratton DL, Gelfand EW. Migration and accumulation
Biochem Biophys Res Commun 315: 46367, 2004. of eosinophils toward regional lymph nodes after airway allergen
118. Mattos W, Lim S, Russell R, Jatakanon A, Chung KF, Barnes PJ. challenge. J Allergy Clin Immunol 114: 82025, 2004.
Matrix metalloproteinase-9 expression in asthma: Eect of asthma 137. Watt AP, Schock BC, Ennis M. Neutrophils and eosinophils: Clinical
severity, allergen challenge, and inhaled corticosteroids. Chest 122: implications of their appearance, presence and disappearance in asthma
154352, 2002. and COPD. Curr Drug Targets Inamm Allergy 4: 41523, 2005.
119. Sur S, Crotty TB, Kephart GM, Hyma BA, Colby TV, Reed CE, 138. Tetley TD. Inammatory cells and obstructive pulmonary disease.
Hunt LW, Gleich GJ. Sudden-onset fatal asthma. A distinct entity Curr Drug Targets Inamm Allergy 4: 60718, 2005.
with few eosinophils and relatively more neutrophils in the airway 139. Yamamoto C, Yoneda T, Yoshikawa M, Fu A, Tokuyama T,
submucosa?. Am Rev Respir Dis 148: 71319, 1993. Tsukaguchi K, Narita N. Airway inammation in COPD assessed by
120. Puxeddu I, Ribatti D, Crivellato E, Levi-Schaer F. Mast cells and sputum levels of interleukin-8. Chest 112: 50510, 1997.
eosinophils: A novel link between inammation and angiogenesis in 140. Balzano G, Stefanelli F, Iorio C, De Felice A, Melillo EM, Martucci
allergic diseases. J Allergy Clin Immunol 116: 53136, 2005. M, Melillo G. Eosinophilic inammation in stable chronic obstruc-
121. Hoshino M, Takahashi M, Takai Y, Sim J, Aoike N. Inhaled corticos- tive pulmonary disease. Relationship with neutrophils and airway
teroids decrease vascularity of the bronchial mucosa in patients with function. Am J Respir Crit Care Med 160: 148692, 1999.
asthma. Clin Exp Allergy 31: 72230, 2001. 141. Bhowmik A, Seemungal TAR, Sapsford RJ, Wedzicha JA. Relation
122. Horiuchu T, Weller PF. Expression of vascular endothelial growth fac- of sputum inammatory markers to symptoms and lung function
tor by human eosinophils: Upregulation by granulocyte macrophage changes in COPD exacerbations. Thorax 55: 11420, 2000.

155
Asthma and COPD: Basic Mechanisms and Clinical Management

142. Lams BE, Sousa AR, Rees PJ, Lee TH. Immunopathology of the small- 156. Steinke JW, Culp JA. Leukotriene synthesis inhibitors versus antago-
airway submucosa in smokers with and without chronic obstructive pul- nists: The pros and cons. Curr Allergy Asthma Rep 7: 12633, 2007.
monary disease. Am J Respir Crit Care Med 158: 151823, 1998. 157. Berger W, De Chandt MT, Cairns CB. Zileuton: Clinical implica-
143. Brightling CE, Monteiro W, Ward R, Parker D, Morgan MD, tions of 5-lipoxygenase inhibition in severe airway disease. Int J Clin
Wardlaw AJ, Pavord ID. Sputum eosinophilia and short-term Pract 61: 66376, 2007.
response to prednisone in chronic obstructive pulmonary disease: A 158. Simon HU, Seelbach H, Ehmann R, Schmitz M. Clinical and immu-
randomised controlled trial. Lancet 356: 148085, 2000. nological eects of low-dose IFN-alpha treatment in patients with
144. Lams BE, Sousa AR, Rees PJ, Lee TH. Subepithelial immunopathol- corticosteroid-resistant asthma. Allergy 58: 125055, 2003.
ogy of the large airways in smokers with and without chronic obstruc- 159. Boguniewicz M, Schneider LC, Milgrom H, Newell D, Kelly N,
tive pulmonary disease. Eur Respir J 15: 51216, 2000. Tam P, Izu AE, Jae HS, Bucalo LR, Leung DY. Treatment of ster-
145. Saetta M, Di Stefano A, Maestrelli P, Turato G, Mapp CE, oid-dependent asthma with recombinant interferon-gamma. Clin Exp
Pieno M, Zanguochi G, Del Prete G, Fabbri LM. Airway eosinophilia Allergy 23: 78590, 1993.
and expression of interleukin-5 protein in asthma and in exacerbations 160. Leckie MJ, ten Brinke A, Khan J, Diamant Z, OConnor BJ,
of chronic bronchitis. Clin Exp Allergy 26: 76674, 1996. Walls CM, Mathur AK, Cowley HC, Chung KF, Djukanovic R,
146. Zhu J, Qiu YS, Majumdar S, Gamble E, Matin D, Turato G, Hansel T, Holgate ST, Sterk PJ, Barnes PJ. Eects of an interleukin-
Fabbri LM, Bernes N, Saetta M, Jeery PK. Exacerbations of bron- 5 blocking monoclonal antibody on eosinophils, airway hyper-
chitis: Bronchial eosinophilia and gene expression for interleukin-4, responsiveness, and the late asthmatic response. Lancet 356: 214448,
interleukin-5, and eosinophil chemoattractants. Am J Respir Crit Care 2000.
Med 164: 10916, 2001. 161. Buttner C, Lun A, Splettstoesser T, Kunkel G, Renz H. Monoclonal
147. Bocchino V, Bertorelli G, Bertrand CP, Ponath PD, Newman W, anti-interleukin-5 treatment suppresses eosinophil but not T-cell
Franco C, Marruchella A, Merlini S, Del Donno M, Zhuo X, Olivieri functions. Eur Respir J 21: 799803, 2003.
D. Eotaxin and CCR3 are upregulated in exacerbations of chronic 162. Kips JC, OConnor BJ, Langley SJ, Woodcock A, Kerstjens HA,
bronchitis. Allergy 57: 1722, 2002. Postma DS, Danzig M, Cuss F, Pauwels RA. Eect of SCH55700,
148. Papi A, Luppi F, Franco F, Fabbri LM. Pathophysiology of exacerba- a humanized anti-human interleukin-5 antibody, in severe persistent
tions in chronic obstructive pulmonary disease. Proc Am Thorac Soc 3: asthma: A pilot study. Am J Respir Crit Care Med 167: 165559, 2003.
24551, 2006. 163. Flood-Page PT, Menzies-Gow AN, Kay AB, Robinson DS.
149. Riise GC, Ahlstedt S, Larsson S, Enander I, Jones I, Larsson P, Eosinophils role remains uncertain as anti-IL-5 only partially depletes
Andersson B. Bronchoal inammation in chronic bronchitis assessed numbers in asthmatic airway. Am J Respir Crit Care Med 167: 199204,
by measurement of cell products in bronchial lavage uids. Thorax 50: 2003.
36065, 1995. 164. Berry M, Brightling C, Pavord I, Wardlaw AJ. TNF-a in asthma. Curr
150. Rutgers SR, Timens W, Kaufmann HF, van der Mark TW, Koeter Opin Pharmacol 7: 27982, 2007.
GH, Postma DS. Comparison of induced sputum with bronchial 165. Bryan SA, OConnor BJ, Matti S, Leckie MJ, Kanabar V, Khan J,
wash, bronchoalveolar lavage and bronchial biopsies in COPD. Eur Warrington SJ, Renzetti L, Rames A, Bock JA, Boyce MJ, Hansel TT,
Respir J 15: 10915, 2000. Holgate ST, Barnes PJ. Eects of recombinant human interleukin-12
151. Keatings VM, Barnes PJ. Granulocyte activation markers in induced on eosinophils, airway hyper-responsiveness, and the late asthmatic
sputum: Comparison between chronic obstructive pulmonary disease, response. Lancet 356: 211416, 2114153, 2000.
asthma and normal subjects. Am J Respir Crit Care Med 155: 44953, 166. Romano SJ. Selectin antagonists: Therapeutic potential in asthma and
1997. COPD. Treat Respir Med 4: 8594, 2005.
152. ODonnell R, Breen D, Wilson S, Djukanovic R. Inammatory cells 167. Beeh KM, Beier J, Meyer M, Buhl R, Zahlten R, Wol G. Bimosiamose,
in the airways in COPD. Thorax 61: 44854, 2006. an inhaled small-molecule pan-selectin antagonist, attenuates late asth-
153. Pizzichini E, Pizzichini MM, Gibson P, Parameswaran K, Gleich GJ, matic reactions following allergen challenge in mild asthmatics: A ran-
Berman L, Dolovich J, Hargreave FE. Sputum eosinophilia predicts domized, double-blind, placebo-controlled clinical cross-over-trial. Pulm
benet from prednisone in smokers with chronic obstructive bronchi- Pharmacol Ther 19: 23341, 2006.
tis. Am J Respir Crit Care Med 158: 151117, 1998. 168. Wegmann M, Gggel R, Sel S, Erb KJ, Kalkbrenner F, Renz H,
154. Walsh GM, Sexton DW, Blaylock MG. Corticosteroids, eosinophils Garn H. Eects of a low-molecular-weight CCR-3 antagonist on
and bronchial epithelial cells: New insights into the resolution of chronic experimental asthma. Am J Respir Cell Mol Biol 36: 6167,
inammation in asthma. J Endocrinol 178: 3743, 2003. 2007.
155. Bentley AM, Hamis Q, Robinson DS, Schotman E, Meng Q, 169. Okigami H, Takeshita K, Tajimi M, Komura H, Albers M, Lehmann
Assou B, Kay AB, Durham SR. Prednisone treatment in asthma. TE, Rlle T, Bacon KB. Inhibition of eosinophilia in vivo by a small
Reduction in the numbers of eosinophils, T cells, tryptase-only posi- molecule inhibitor of very late antigen (VLA)-4. Eur J Pharmacol 559:
tive mast cells, and modulation of IL-4, IL-5, and interferon-gamma 2029, 2007.
cytokine gene expression within the bronchial mucosa. Am J Crit Care
Med 153: 55156, 1996.

156
The Lymphocyte in Asthma and COPD
13 CHAPTER

INTRODUCTION eosinophil-rich inammation to bronchocon-


James G. Martin and
striction and tissue remodeling. T-cell cytokines
recruit, activate, and enhance the survival of a Manuel G. Cosio
Asthma and chronic obstructive pulmonary range of inammatory cells that are the more
disease (COPD) are chronic inammatory Meakins-Christie Laboratories and the
direct mediators of the pathologic processes.
diseases of the airways that share a high prev- Respiratory Division, Department of
The participation of the T-cell in COPD is rel-
alence and, when severe, impose a substan- atively less well understood but involves CD8 Medicine, McGill University, Montreal,
tial burden on aected persons and society at T-cells to a greater extent. Both diseases are Quebec, Canada
large. Although controversy has surrounded the now known to have features mediated by an
extent to which these conditions may be related, adaptive immune response to antigens, attribut-
distinct patterns of airway inammation have able to inhaled soluble antigens in asthma, and
been described in the two conditions and evi- to self-antigens either newly revealed or modi-
dence for the participation of dierent T-cell ed by tissue injury secondary to tobacco smoke
subsets in the pathogenesis of the two diseases in COPD. Why the mucosal immune system of
is emerging. There is, however, some evidence the respiratory tract responds in such dierent
of convergence of the inammatory processes manners to the antigenic stimuli, and results in
in advanced COPD and in severe asthma but, the distinct disease phenotypes is still not very
for the most part, there are clear and obvious clear.
dierences. In particular, although the chronic In this chapter we will review the evi-
inammatory inltrate in both diseases results dence for the role of T-cells in the pathobiol-
in airway obstruction, obstruction is usually ogy of these two major causes of morbidity and
reversible in asthma, and is largely irreversible mortality. We will also discuss the additional
and progressive in COPD. These key dier- roles for certain T-cell subsets, such as the
ences in the pathophysiology of the two dis- potential eect of T-cells in the maintenance
eases are due to the type and consequences of of airway epithelial integrity, that extend the
the inammation. Inammation is largely lim- range of functions beyond the classical adaptive
ited to the airways in asthma whereas both the immune functions most commonly thought of
airways and parenchyma are aected in COPD in association with the T-cell.
and in such a way that irreversible brosis of the
small airways and parenchymal destruction with
emphysema may result. Both diseases when
fully established show evidence of an adaptive
immune response with an excess of activated
T-cells, but of dierent phenotypes, secreting
HISTORICAL NOTE
type 2 cytokines in asthma and type 1 cytokines
in COPD. Descriptions of the immunopathol- Early preoccupations with the role of lym-
ogy of asthma as well as data from animal mod- phocytes in asthma revolved around the role
els leave little doubt about the central place of of B-cells in IgE production. It was observed
the CD4 T-cell in virtually all of the key fea- as early as 1975 that human asthmatic subjects
tures of asthma, ranging from the characteristic appeared to have a relative lymphopenia [1] and

157
Asthma and COPD: Basic Mechanisms and Clinical Management

a defective T-cell response to stimulation by mitogens such


as phytohemagglutinin and concanavalin A [2]. B-cells LYMPHOCYTES IN THE RESPIRATORY TRACT
from atopic subjects were shown to spontaneously synthe-
size IgE in vitro and T-cells from the aected individuals The lymphocytes in the respiratory tract are present in the
were much less inhibitory of this synthesis compared to T- form of localized bronchus-associated lymphoid tissue
cells from unaected persons [3]. Specic immunotherapy (BALT) and the nasal-associated lymphoid tissue (NALT)
led to a correction of the reduced inhibitory T-cell subset that are organized lymphoid aggregates, comprising
[4]. Subsequently elevated CD4 to CD8 ratios in the T- and B-lymphocytes that may mediate adaptive immune
blood of asthmatic subjects was also interpreted as suggest- responses to inhaled antigens. BALT is present at bron-
ing a deciency of suppressor T-cell function in asthmatics chial bifurcations and may be responsible for the synthesis
[5]. Dierences in the ratio of CD4 to CD8 T-cells were and secretion of immunoglobulins in response to antigenic
noted following challenge of allergic asthmatic subjects as stimulation. Humoral immune responses elicited by BALT
a function of whether the subjects demonstrated isolated are primarily IgA secretion. The BALT is in many respects
early responses to challenge or had dual airway responses, analogous to mucosal lymphoid aggregates in the intestine
with both early and late bronchoconstriction. The hypoth- that has important functions in regulating intestinal mucosal
esis proposed was that CD8 T-cells were recruited from immunity (reviewed in [16]). The epithelial cells overlying
the blood to the airways and prevented the development of the BALT lack cilia and appear to be equivalent to M-cells
the late airway response in single responders [6]. Activation in the intestinal epithelium. The M-cells are derived from
of CD4 T-cells (surface expression of IL-2R, VLA-1, basal cells in the respiratory epithelium and, as in the intes-
and HLA-DR) was also noted during acute asthma exac- tine overlying Peyers patches, have the capacity to selec-
erbations [7]. HLA-DR expression on cells in bronchial tively phagocytose and pinocytose particles and molecules
biopsies of stable asthmatics was correlated with airway in their immediate environment. Lymphocytes are also dis-
hyperresponsiveness [8]. tributed within the epithelium and more diusely through
A more obvious link of T-cells to airway inam- the airway wall, invading all of the tissues in the setting of
mation emerged as evidence of the synthesis of high airway inammation.
molecular weight chemotactic factors for eosinophils was
obtained by injecting concanavalin A into the skin of a
guinea pig [9]. However, without doubt, the single most
important step forward in understanding the place of
T-cells in allergic airway inammation occurred with the
INCITING STIMULI
description of Th1 and Th2 type CD4 T-cells in 1986 by
Mosmann and colleagues [10]. For the subsequent 20 years Both asthma and COPD are diseases that involve inam-
research on the role of the T-cell in asthma has focused sin- mation mediated by the respiratory mucosal immune sys-
gularly on the exploration of this paradigm. Indeed much tem. In asthma aeroallergens are the best characterized
of the inammatory process in allergic asthma is explained inducers of disease although other causes such as strong
by this paradigm although added complexity has emerged irritants (often mediating damage through oxidant injury)
as new subsets, such as regulatory T-cells, Th17 cells, and viral illness are also of importance. These same stimuli
T-cells, and iNKT cells that have been described, and have may trigger asthma attacks also. The place of the adaptive
begun to be integrated into the pathogenetic schema of immune system in the pathogenesis of asthma caused by
inammatory airway disease. nonallergic mechanisms is not clear. It is possible that air-
Early observations on airway inammation in way hyperresponsiveness may be induced as a result of air-
subjects with COPD revealed typical innate immune way remodeling from airway insults, bypassing the need for
inammation. Young cigarette smokers were noted to a major participation of adaptive immunity in the process.
have increased cellularity in the bronchoalveolar lav- Model systems, based in recent years on murine and rodent
age (BAL) uid that was attributable to macrophages models, have been heavily exploited to unravel the complex
but few neutrophils were present [11], while older smok- immunological reactions triggered by allergen. The literature
ers had increased numbers of both neutrophils and alveo- is replete with references to murine asthma, for example,
lar macrophages [12]. From these data originated the but it must be acknowledged that models based on a small
concept that the destruction of lung tissue resulted from number of allergen challenges model only a limited part of
the release of potent proteinases from these cells and the disease. Growing literature on the role of viruses, such
the proteinaseanti-proteinase balance became the para- as respiratory syncytial virus and rhinovirus, will comple-
digm for the pathogenesis of COPD for almost 40 years. ment the results from allergen challenge to provide a more
The nature and mechanisms of inammation were recon- complete picture of the triggers and inducers of asthma.
sidered following the description by Finkelstein et al. of a Likewise, the T-cell contribution to other forms of injury
prominent T-cell inltration in the lungs of patients with such as oxidant injury has received little attention. To date
COPD, the degree of which was strongly related to the the mechanisms for activation of T-cells in asthma have
extent of emphysema [13]. These ndings suggested new dealt almost exclusively with allergic mechanisms.
mechanisms for the pathogenesis of COPD through T- The inammatory reaction to cigarette smoke is cen-
cell involvement and prompted us to suggest that COPD tral to COPD and the global initiative workshop summary
might be an autoimmune disease triggered by cigarette for chronic obstructive lung disease (GOLD) denition of
smoking [14, 15]. COPD states that the airow limitation is usually progressive

158
The Lymphocyte in Asthma and COPD 13
and associated with an abnormal inammatory response of the mitochondrial damage, DNA strand breaks, structural/func-
lungs to noxious particles or gases(GOLD Report update 2007, tional modication of proteins, and ultimately cell death.
www.GOLDCOPD.com downloaded May 2008) [17]. In Oxidative modication of proteins has been implicated in
both disease processes there is an important contribution of the immune mechanisms of rheumatoid arthritis, multiple
the innate immune response to the pathobiology and close sclerosis, and arteriosclerosis [32]. Induction of organ-specic
links to lymphocyte function. Of particular importance is autoimmune disease following tissue trauma, inammation,
the dendritic cell, but macrophages and other cells may also and viral or bacterial infections has been frequently reported
play a role in conditioning the T-cell response. Cigarette and likely occurs via tissue damage that results in the availa-
smoke and other pollutants (ozone, NO2, diesel particles) bility of previously hidden antigens or by molecular mimicry
produce an acute innate immune reaction in the lung as do with viral or bacterial products [33]. Infections also provide
the triggers of asthma. The epithelium is exposed to all of abundant cytokines that may aect the expression of co-
these triggers over its surface and through damage, stress, or stimulatory molecules, an additional stimulus important in
stimulation via receptors, such as Toll receptors, is impor- the perpetration of the immune response.
tant in initiating the response. The present evidence suggests Airway or pulmonary parenchymal damage from ciga-
that, by sending danger signals in response to cigarette rette smoke could easily follow the same mechanisms. The
smoke, the epithelium is responsible for the initiation and tissue injury and potential modication of self-proteins could
possibly maintenance of the innate immune response seen generate antigens (e.g. release of cryptic antigens, modied
in smokers. Over 2000 dierent xenobiotic compounds proteins, necrotic cells, apoptotic cells, etc.) and the associated
have been identied in cigarette smoke, and it has been esti- innate immune inammation associated with smoking, could
mated that there are 1014 free radicals in each pu of ciga- provide the necessary soluble mediators and co-stimulatory
rette smoke [18]. Not surprisingly, it has been shown that signals for the initiation or perpetuation of an adaptive
cigarette smoke is cytotoxic to epithelial cells and that the immune response. That this is the case has been recently
extent of injury produced is directly related to the concen- shown by Lee and coworkers who identied elastin as an
tration of smoke to which the cells were exposed [19]. antigen responsible for the T-cell response in COPD [31].
The sustained innate immune response found in the However in a disease like COPD in which multiple cells and
lungs of cigarette smokers (neutrophils, macrophages, eosi- tissues seem to be potentially injured, other cell products from
nophils, mast cells, T-cells, dendritic cells, and perhaps necrotic, stressed, or apoptotic cells might also act as antigenic
natural killer (NK) cells), and their products (cytokines, oxy- determinants. Whether a component of autoimmunity com-
gen radicals, proteinases) are capable of producing matrix plicates asthma is not known but it would not be altogether
and cellular damage. Studies in mice have shown that after surprising since cellular damage and oxidative stress is also a
24 h of cigarette smoke exposure, measurable increases in feature of the disease. This has been recently shown in smok-
desmosine, a marker of elastin breakdown and hydroxypro- ers with COPD in which autoantibodies were found to bind
line, a marker of collagen breakdown, are found in BAL and at least three dierent autoantigens [34].
their levels are correlated with the number of neutrophils in
the BAL [20, 21]. Evidence of connective tissue breakdown
in human smokers also exists. Increased plasma and urine
levels of elastin-derived peptides and desmosine [22, 23]
have been found in COPD patients when compared with MECHANISMS OF T-CELL ACTIVATION AND
nonsmokers [24, 25]. However, the levels of elastin break- DIFFERENTIATION: MODEL SYSTEMS
down products are also elevated in smokers without COPD
[24, 2629]. Smokers with rapid decline in lung function,
likely to develop COPD, were found to excrete 36% more Dendritic cells, CD4, and CD8 T-cells
desmosine than low decliners [30].
However the proteaseanti-protease imbalance Adaptive immune responses are initiated by the dense net-
paradigm by itself cannot explain why only some smok- work of dendritic cells in and under the epithelium [35].
ers develop COPD. It might be closer to reality to consider These cells have projections that appear to sample the air-
the connective tissue breakdown as one of the many con- way luminal contents and can take up antigen for presen-
sequences of the innate inammatory reaction triggered by tation to T-cells (Fig. 13.1). This presentation occurs in
smoking. Furthermore, these breakdown products or peptides draining lymph nodes following maturation of the dendritic
once revealed to the immune system may be antigenic and cells under the inuence of cytokines such as granulocyte-
be presented to T-cells, eventually triggering, in some smok- macrophage colony stimulating factor (GM-CSF). T-cells
ers, a T-cell adaptive immune response against lung antigens sample the dendritic cell surface for antigens in a more or
(autoimmunity), as has been recently shown [31]. There is less stochastic process. High levels of antigen on the cell
extensive literature showing that infectious and environ- surface shorten the time required for the T-cell to form
mental agents have the potential to alter self-proteins that stable contacts with the dendritic cell [36]. In addition to
may then be recognized as antigens by the adaptive immune dendritic cellT-cell contact in the lymph nodes it has also
system. Among the important protein modiers present in been observed that T-cells come into contact with den-
smokers are free radicals/oxidative stress and xenobiotic dritic cells in the airway wall [37]. Indeed following aller-
agents which abound in cigarette smoke. Nitric oxide (NO) gen challenge there is a marked increase in the number of
per se or following its conversion to peroxynitrite by super- dendritic cells in contact with T-cells. This interaction may
oxide or other reactive oxygen species (ROS) may produce short-circuit the longer process that is otherwise required

159
Asthma and COPD: Basic Mechanisms and Clinical Management

Ag
Ag
Antigen uptake
and processing

Epithelium
Lymph node FIG. 13.1 Dendritic cells sample antigen material
in the airway lumen through the cell processes
Afferent
lymphatic that interdigitate between the epithelial cells. The
T-Cell dendritic cell then migrate to the draining lymph
Dendritic cell nodes and are sampled by T-cells that respond
to the antigenic peptide on the surface of the
Maturation dendritic cell. Maturation of the dendritic cell in
response to the conditioning by cytokines such
GM-CSF as granulocyte-macrophage colony stimulating
TARC T-Cell
MDC CCR4 factor (GM-CSF) enhances its antigen-presenting
ability. T-cells with antigen specificity return to
T-Cell Efferent the mucosal surface following their egress from
lymphatic the lymph node through the efferent lymphatic.
T-Cell CCR4 Chemokines such as TARC and MDC favor
movement of Th2 cells into the tissues. These
chemokines are produced by epithelial cells and
dendritic cells, when appropriately stimulated.

and presumably is responsible for the rapid T-cell mediated


response that follows allergen challenge in vivo. Dendritic cell Dendritic cellT-Cell
Following engagement with the dendritic cell the interactions
T-cell is activated, secretes IL-2, and expresses the IL-2
receptor. Clonal expansion takes place and the T-cell phe-
notype is subsequently established in response to a variety of
inuences; cytokines, lipid mediators, and toll-like receptor
OX40 L T-Cell
(TLR) ligands all play a part in the latter phenomenon. The
OX40
presentation of exogenous antigen in association with major MHC II
Ag
histocompatibility complex (MHC) class II molecules leads TCR
to activation of CD4 T-cells with high-anity antigen- CD 80/86

specic T-cell receptors (Fig. 13.2). This is the predominant CD 28

pathway activated by inhaled soluble antigens that com-


monly trigger allergic airway responses. Antigen produced
IgE
intracellularly, such as may result from the infection or FcR
injury of pulmonary cells by virus or other agents like ciga-
rette smoke, is processed by dierent intracellular pathways
and presentation occurs in conjunction with MHC class I Cytokines
molecules. CD8 T-cell activation results. The processing
FIG. 13.2 The dendritic cell interacts with the T-cell through major
and presentation of soluble exogenous antigen to CD8 histocompatibility molecules that harbor the antigenic peptide and
T-cells in an MHC class I restricted manner is called cross the T-cell receptor. Various co-stimulatory molecules are required to
presentation. Soluble antigens destined for cross presenta- allow effector functions to be established. OX40 and OX40L interactions
tion are taken up into early endosomes whereas those des- are important for Th2 differentiation. CD28CD80/86 interactions are
tined for classical MHC class II restricted presentation are important for both Th1 and Th2 development. The binding of IgE to the
taken up in lysosomes [38]. Heat shock proteins are released FcR1 on the dendritic cells facilitates Th2 cell differentiation.
from cells undergoing lytic death, but not from cells dying
of apoptosis [39] and necrotic cell lysates and also stressed
cells have been reported to express cell surface HSP mole- are not random but mediated by the upregulation of the
cules [40] that could activate dendritic cells directly through chemokine receptor CCR5 on the CD8 T-cells, which
receptors including CD91 [41] and TLR-4. Cell-associated directs these cells to sites of antigen-specic dendritic cell
proteins [42, 43] and particulate antigens [44] are much CD4 interaction where the cognate chemokines CCL3
more eective at class MHC I restricted CD8 T-cells and CCL4 are produced [45].
priming than are simple soluble molecules. Cross presenta-
tion is also essential for proper stimulation and functional
development of CD8 T-cell eector and memory function Th1 and Th2 cells
since these depend on the encounter of both CD4 and
CD8 T-cells with the same dendritic cell and the release of The cytokine prole secreted by the CD4 T-cell is of
stimulatory factors from the CD4 T-cell. These encounters critical importance in directing the inammatory response,

160
The Lymphocyte in Asthma and COPD 13
Factors favoring Th2 Basophil
differentiation

Eosinophil

Dendritic cell
TLSP
Mast cell

EDN
(TLR2 Ligand)
OX40 L
5-LO FIG. 13.3 The factors that favor the
NK cell development of Th2 responses are several.
Co-stimulatory molecules such as OX40 and
cys LTs OX40L, upregulation of OX40 by TSLP produced
cys LTs by basophils and structural tissue cells such as
IL-4 epithelial cells, IgE bound to the dendritic cell
IL-10 NKT cell surface, TLR2 ligation by EDN, low lever TLR4
TLR2 Ligand IL-13 stimulation, cysteinyl leukotriene synthesis by
T-Cell dendritic cells and their subsequent interaction
with the Cys-LT1R on the T-cell and Th2 cytokine
TLR4 Ligand production by a variety of cells such as mast
(weak stimulation) Th2 cytokines cells, eosinophils, basophils and NKT cells all
favor Th2 cell differentiation.

whether typical of allergic inammation with a Th2 cytokine basophils may be triggered by allergen to release TSLP in
predominance or delayed type hypersensitivity typical of lymph nodes and thereby promote Th2 cell dierentiation
Th1-driven inammation. The factors that determine the in this site [51].
Th1 and Th2 type responses are several. The dendritic cell Signals that aect the T-cells are also important in
has an important role through the secretion of IL-12 in directing the T-cell dierentiation. It has been recognized
the case of the induction of Th1 responses and IL-4 and for some time that IL-4 is important in creating a milieu
IL-10 in the case of Th2 and T-regulatory cell responses. that favors the development of Th2 responses. Mast cells and
Respiratory mucosal dendritic cells preferentially trigger NK cells, as well as T-cells themselves, are sources of this key
Th2 responses, but after they have matured under the inu- cytokine. Indeed mast cells express most of the cytokines that
ence of cytokines such as GM-CSF they express high lev- control T-cell dierentiation pathways and may potentially
els of MHC II, B7, and IL-12 and produce Th1 responses regulate T-cell fate decisions (reviewed in [52]). Furthermore,
[46]. Concomitant pro-inammatory stimuli condition the signals to the dendritic cell as exemplied by TSLP and sig-
dendritic cell and its subsequent interaction with the T-cell nals to the T-cell such as IL-4 synergize to produce, in this
(Fig. 13.3). Signals mediated via the TLRs are among the instance, a Th2 response [50]. It has long been known that
important stimuli of this sort. Endotoxin-poor antigen trig- the dierent T-cell phenotypes exert counter-regulatory
gers Th2 responses whereas a strong concomitant stimula- eects, IFN- inhibits Th2 cell development whereas IL-10
tion of dendritic cells with lipopolysaccharide, for example, inhibits Th1 cell development. Cysteinyl leukotrienes which
may convert the T-cell response into a Th1 response [47]. are an important feature of allergic reactions are produced
In contrast a TLR-2 agonist results in a Th2 biased T-cell by the dendritic cells and appear to favors Th2 dierentia-
response [48]. Recently it has been demonstrated that eosi- tion through an autocrine action that modulates IL-10 and
nophils, through eosinophil-derived neurotoxin (EDN), IL-12 production by the dendritic cell [53]. Cys-LTs favors
are involved in the process of T-cell dierentiation [49]. IL-10 production whereas inhibition of the Cys-LT1 recep-
EDN, a member of the RNase A superfamily, appears to tor favors IL-12 production and IFN- synthesis by T-cells.
be an endogenous ligand for TLR2. Cytokines also serve Other essential factors for Th1 dierentiation are the
to condition the dendritic cell responses. Thymic stromal lym- transcription factors T-bet and IRF1. T-bet not only induces
phopoietin (TSLP) induces OX40 ligand on dendritic cells Th1 dierentiation of nave T-cells, but also reprograms
that subsequently evokes Th2 cytokine production by CD4 polarized Th2 cells to produce the Th1 cytokine IFN- and
T-cells. These cells produce TNF- and do not express IL-10, negatively regulates the Th2-specic transcription factor
diering from those Th2 cells that produce IL-10 as a part GATA-3 thus promoting the Th1 polarization. A deciency
of their cytokine prole [50]. A problem with this paradigm of T-bet is associated with an exaggerated asthmatic pheno-
for driving Th2 responses was that TSLP appeared to be type in mice [54].
produced in peripheral tissues but not in the lymph nodes. Th1 and Th2 subsets are functionally separated based
This issue has now been resolved by the demonstration that on their cytokine proles but certain types of chemokine

161
Asthma and COPD: Basic Mechanisms and Clinical Management

receptors are selectively expressed on either Th1 or Th2 exposed to long-term cigarette smoke, indicating an impor-
cells and appear to have an important place in determin- tant role of these cells in the development of emphysema
ing the patterns of accumulation of T-cell subsets as a func- [68]. Proof of the concept that CD4 T-cells, and autoim-
tion of lung pathology. Th1 cells express CCR5, CXCR3, munity, may play an important role in COPD has been
and CXCR6 [55] whereas Th2 cells express CCR3, CCR4, provided by demonstrating the induction of emphysema in
and CCR8 [56]. TSLP acts on dendritic cells resulting in nave rats by the adoptive transfer of vascular endothelial-
the synthesis of two chemokines, thymus and activation- sensitized CD4 T-cells from antigen-induced emphysema
regulated chemokine (TARC) and macrophage-derived animals [69].
chemokine (MDC) that serve as chemoattractants for Th2
cells through actions on CCR4. TARC is also expressed in
other sites, such as the bronchial epithelium, and may con- Tc1 and Tc2 cells
tribute to the selective chemoattraction of Th2 cells to the
airways in asthma [57] whereas interferon inducible protein In addition to their cytotoxic properties CD8 T-cells
10 (IP-10/CXCL10) may contribute to CD8 and CD4 also show distinct subsets [Tc1 (Fig. 13.4) and Tc2] with
Th1 cell recruitment in COPD [58]. The expression of the cytokine proles resembling the Th1 and Th2 patterns with
chemokine receptors CXCR3, CCR5, and CXCR6 cor- IL-2 and IFN- production and IL-4, IL-5, and IL-10
related with disease severity; moreover, the dendritic cells production respectively [70]. These subsets termed Tc1 and
produced the respective ligands for these receptors, and the Tc2 evoke similar degrees of delayed-type hypersensitivity
levels again correlated with disease severity in COPD [55]. reactions but the latter cells also cause a somewhat greater
The elucidation of the roles of CD4 and CD8 degree of eosinophilia [70].
T-cell subsets in asthma has depended heavily on the use
of small animal experimental models and either adoptive
transfer or antibody depletion techniques. It is impossible
to review the extensive literature in detail. Dependence of Th17 cells
allergic airway responses on CD4 T-cells has been dem-
onstrated by the technique of adoptive transfer. T-cells The Th17 cells produce large quantities of pro-inammatory
isolated from the drainage lymph nodes of the site of sen- cytokines, most typically IL-17, and seem to be important
sitization to allergen and administered to nave recipients in antibacterial responses and autoimmune diseases. Bacteria
that undergo allergen challenge result in typical late aller- prime human dendritic cells to promote IL-17 production
gic responses [59] and airway hyperresponsiveness [60, 61]. in memory Th cells through the actions of muramyldipep-
Conversely, CD4 T-cell depletion also abolishes airway tide (MDP) on the innate pathogen recognition recep-
responses to allergen challenge [62]. It is now clear that tor nucleotide oligomerization domain 2 (NOD2). MDP
mixed T-cell responses may occur in response to exposure enhanced TLR agonist induction of IL-23 and IL-1, pro-
to soluble antigen. Adoptive transfer of antigen-sensitized moting IL-17 expression in T-cells [71]. In mice, Th17
CD8 T-cells to nave recipient animals that are then cells dierentiate from nave CD4 T-cells in response
allergen-challenged results in delayed airway narrowing and to transforming growth factor- (TGF-) and IL-6 and
typical TR2-type cytokine expression [63]. CD8 T-cells also require IL-23 for their maintenance or expansion.
are also responsible for virally induced airway hyperrespon- IL-23 signaling requires IL-12Rb1, a receptor compo-
siveness in the mouse and the accompanying IL-5-driven nent shared with IL-12. Dierences in signaling involving
eosinophilia [64]. In these situations the CD4 and CD8 IFN--induced transcription factor appear to determine
cells share some common properties such as the secretion whether CD4 Th1 or Th17 cells are produced [72]. In a
of eosinophilia-promoting cytokines. CD4 T-cells have murine model, IL-17 is required during antigen sensitiza-
also been implicated in airway remodeling in the allergic tion to develop an allergic phenotype, as shown in IL-17R-
rat. Again using adoptive transfer, it was shown that the decient mice [73]. The increase in IL-17 expression with
ovalbumin-sensitized and challenged rat developed epi- allergen challenge is inhibitory of the allergic response.
thelial remodeling and airway smooth muscle remodeling Neutralization of IL-17 augments allergic responses
through hyperplasia and reduction in apoptosis [65]. The whereas exogenous IL-17 reduces eosinophil recruitment
capacity of ovalbumin stimulated CD4 T-cells to cause and bronchial hyperreactivity. Eotaxin (CCL11) and TARC
hyperplasia of isolated airway myocytes was demonstrated are downregulated by IL-17 as are IL-5 and IL-13 produc-
in culture, and was contact dependent. tion in regional lymph nodes. IL-4 plays an inhibitory role
The study of the roles of CD4 and CD8 T-cells in the magnitude of IL-17 expression. IL-17 is also respon-
in COPD lags years behind that of asthma. Until recently sible for a neutrophilic component to the bronchial inam-
the evidence of the role of these cells in the immunity of mation induced by allergen challenge of sensitized mice
COPD was mainly circumstantial. Recently it has been [74]. Th17 cells have not yet been reported in COPD but
shown in mice exposed to cigarette smoke that the develop- it would be surprising if they did not play a role in smokers
ment of pathological and functional emphysema is depend- since they have a crucial role in the induction of autoim-
ent on the presence of CD4 and CD8 T-cells [66, 67] mune tissue injury. Furthermore IL-6 whose expression is
and only the animals with emphysema showed signicant elevated in smokers has the potential to prevent the TGF-
expression of Th1 cytokines. In another study it was shown mediated dierentiation of T-cells into regulatory T-cells
that mice depleted of CD8 T-cells had a blunted inam- (Tregs) by inhibiting the expression of FOXP3 and induc-
matory response and did not develop emphysema when ing the expression of IL-17 [75].

162
The Lymphocyte in Asthma and COPD 13
Dendritic cell Elastin fibers

Degraded by MMPS
release cryptic
antigens
MHC I

IL-12 Ag IFN-

TCR

TC1
PAS? FIG. 13.4 Oxidative stress, matrix
metalloproteinase release by inflammatory
cells leads to degradation of the matrix
with release of cryptic antigens from
elastin fibers. These fibers elicit a cytotoxic
Granzyme Perforin Alveolar epithelial cell
T-cell response which induces apoptosis
apoptosis
of alveolar epithelial cells by mechanisms
that involve perforin, granzyme and the
FASFASL pathways.

T-regulatory cells T-cells


There is growing evidence that a signicant aspect of the Important T-cell subsets that do not share the usual T-cell
inammatory reaction in the airways is caused by a failure receptor, such as the TCR-bearing cells and NK T-cells,
of suppression of the response. Active regulation appears to have substantial importance for pulmonary defense and air-
be important to peripheral tolerance to allergens in nonal- way function. T-cells expressing the T-cell receptor have
lergic individuals. In this regard Tregs are receiving sub- a particular association with mucosal epithelial surfaces,
stantial attention. A rapidly growing number of studies but what they do is still largely enigmatic [1]. In contrast
addresses the role of these cells in allergic processes and in to T-cells that recognize peptide antigens the T-cells
models of asthma. Tolerance to allergens is associated with recognize small nonpeptidic molecules and in some cases
the synthesis of IL-10 and TGF- by FOXP3 expressing nonclassical MHC molecules such as CD1. T-cells are
CD4CD25 T-cells. The precise mechanisms by which important in defense against intracellular organisms such as
T-cell inhibition is produced are still not well worked out. Listeria, Pneumocystis carinii and Mycobacterium tuberculosis
It appears that CD4CD25 Tregs are capable of inhib- [79]. However their functions extend beyond their innate
iting allergic responses even if they are harvested from immune defense properties. These cells produce large quan-
IL-10-decient mice and adoptively transferred into wild- tities of IFN- and have been shown to potently downreg-
type mice. The suppression of the Th2 responses is medi- ulate IgE responses to sensitization [80, 81]. As such these
ated by IL-10 producing CD4 T-cells that are activated cells are important mediators of allergen tolerance. They
by the transferred Tregs [76]. Intratracheal administration also synthesize trophic factors such as keratinocyte growth
of CD4CD25 T-cells inhibits allergic airway responses factor (KGF) that is important in epithelial cell regenera-
(airway hyperresponsiveness and inammation) through tion [82], at least in the small intestine and likely also in
an IL-10-dependent mechanism acting through TGF- the airways. The administration of KGF is able to protect
expression. The T-cells required IL-10 treatment prior to the epithelial barrier function in face of allergen chal-
transfer and even when unable to synthesize IL-10 them- lenge. IFN- production by CD8 T-cells is potently
selves, they were still able to inhibit the allergic response inhibitory of late allergic airway responses in the rat [83].
[77]. The inammatory process that follows allergen chal- However, in the mouse there are both pro-inammatory
lenge in a murine model of allergic asthma and the ensu- and anti-inammatory properties associated with T-cells.
ing airway hyperresponsiveness can be attenuated by Treg In OVA-sensitized and challenged mice, T-cells express-
cells that are recruited into the airway mucosa [78]. These ing V1 enhance airways hyperresponsiveness [84], whereas
cells appear after the initial inammation that is triggered cells expressing V4 strongly suppress airways hyperrespon-
by allergen inhalation and suggests that these cells may siveness [85]. The airways hyperresponsiveness-regulatory
normally serve to limit the extent of the allergen-induced T-cells had only minor eects on airway inammation
asthmatic reaction. in these experiments in contrast to rodent models where

163
Asthma and COPD: Basic Mechanisms and Clinical Management

they also inhibit Th2 inammation. Cigarette smoking there is an abrogation of the allergic responses and airway
induces an increase of T-cells both in human [15] and in hyperresponsiveness [102, 103]. However the mechanism
mice [66] exposed to cigarette smoke, but their function in appears to reect an attenuated Th2 response and reduced
smokers is not clear. expression of important cytokines such as IL-13. These nd-
ings are perhaps explained by the eects of cationic proteins,
such as EDN on immune responses, as reviewed above. The
iNKT-cells adoptive transfer of genetically engineered T-cells over-
expressing IL-10 is inhibitory of allergic airway responses
iNKT cells express CD3 and an invariant T-cell receptor and airways hyperresponsiveness [104], consistent with the
-chain (reviewed in [86]). They respond to glycolipid anti- emerging concept of this cytokine as an inhibitory cytokine
gens that are presented by CD1d, and produce large quanti- when produced by T-cells in the airway. The induction of
ties of IFN- and IL-4 [87]. The role of iNKT cells in host airway hyperresponsiveness by repeated ozone exposures has
defense in the lung has largely been inferred from experi- been recently demonstrated to require both IL-17 and NKT
ments in knockout mice that lack iNKT cells, which show cells [105], expanding the range of pertinent stimuli that
increased susceptibility to a wide range of bacterial and viral activate airway T-cells.
pathogens and parasites. It has recently been proposed that There is evidence that CD8 T-cells in the lungs of
iNKT cells also have a role in asthma pathogenesis: experi- COPD patients express IFN- [106] which would enhance
ments with iNKT-decient mice show reduced responses to the inammatory reaction in the lung in addition to that
sensitization and challenge that can be largely reconstituted caused by their cytotoxicity. Transgenic overexpression of
by the adoptive transfer of iNKT cells [87]. In a murine IFN- causes emphysema in the murine lung and IFN-
model, when stimulated by -galactosylceramide iNKT -decient mice are protected against cigarette smoking-
cells produce IL-13 and mediate airway hyperresponsive- induced increases in alveolar cell apoptosis. Transgenic
ness and eosinophilic inammation, providing an additional overexpression of IL-13, a critical cytokine in asthma,
innate immune mechanism for an asthma phenotype [88]. causes emphysema with enhanced lung volumes and com-
pliance, mucus metaplasia, and inammation, in the adult
murine lung. Matrix metalloproteinase (MMP)-2, -9, -12,
-13, and -14 and cathepsins B, S, L, H, and K were induced
by IL-13 in this setting. In addition, treatment with MMP
T-CELL CYTOKINES IN MODELS OF or cysteine proteinase antagonists signicantly decreased the
ASTHMA AND COPD emphysema and inammation, but not the mucus in these
animals. These studies demonstrate that IL-13 is a potent
stimulator of MMP and cathepsin-based proteolytic path-
The eects of the administration of cytokines to animals have ways in the lung. They also demonstrate that IL-13 causes
been examined and provide some clues as to mechanisms of emphysema via an MMP- and a cathepsin-dependent
the links between the T-cells and airway responses. IL-2 mechanism(s) and highlight common mechanisms that
administered to nave rats leads to airway hyperresponsive- may underlie COPD and asthma [107]. Transgenic over-
ness to inhaled methacholine [89]. In high doses, it causes expression of IL-18 also results in murine emphysema, and
peribronchial edema and an eosinophilic airway inam- appears to result from the combined eects of both IL-13
mation. Low doses of IL-2 augment allergic responses, in and IFN- overproduction in the lung [108].
terms of both the magnitude of bronchoconstriction and
the degree of eosinophilic inammation [90]. The increase
in late response is attributable to an increase in sensitivity
to leukotrienes rather than by enhanced synthesis [91, 92].
IL-4 is important in the sensitization phase to allergen in T-CELLS IN THE PATHOGENESIS OF
murine models and is necessary for allergen-induced airway
hyperresponsiveness. However neutralization of its actions at
ASTHMA: HUMAN STUDIES
the time of challenge does not abrogate hyperresponsiveness
[93]. In contrast, IL-13 is of importance to airway hyper- T-cells are frequent in the airway wall. Most intraepithelial
responsiveness at the time of challenge to allergen [9496], T-cells express CD8, whereas CD4 T-cells are more fre-
and its eects depend on intact signaling pathways for IL-13 quently found in the lamina propria. Both subsets mainly
in the epithelium [97]. How the epithelium transduces the have an eector- and/or memory-cell phenotype, as dened
eects of IL-13 is as yet unknown. Interestingly, the asthma by their expression of CD45RO (reviewed in [86]). In
phenotype caused by T-bet deciency in mice is also attrib- asthma the number of T-lymphocytes in the airway wall
utable to IL-13, both airway hyperresponsiveness and air- increases substantially. In agreement with the paradigm for
way remodeling were ameliorated by neutralization of IL-13 T-cell subsets dened on murine T-cell cytokine proles,
[98]. IL-5 has been shown in some models but not in others a predominance of Th2 expressing CD4 T-cells has also
to mediate airway hyperresponsiveness [93, 99101]. Recent been observed in the BAL cells from mild atopic asthmatics
studies help to clarify the issue of the controversial roles of [109]. Furthermore, Th2 cytokine expressing cells increase
IL-5 and eosinophilia in allergic asthma. When eosinophilia in the BAL uid of asthmatic subjects following allergen
is absent, as in IL-5 and eotaxin-decient mice, or in mice challenge [110]. In fact IL-4 and IL-5 mRNA express-
genetically engineered so as to lack eosinophils specically, ing T-cells can be detected even in sputum samples from

164
The Lymphocyte in Asthma and COPD 13
asthmatic subjects [111]. Consistent with various cellular of T-cell populations in the respiratory mucosa are also
and animal models there is also an upregulation of GATA- required before any denite statement about such a strategy
3, the principal transcription factor for Th2 cytokines in can be recommended.
asthmatic airway tissues [112]. Lung lavage following seg- iNKT cells have been recently implicated in airway
mental allergen challenge of the airways of asthmatic sub- pathophysiology. The extent to which ndings from murine
jects has demonstrated that the CD4, CD8, and T-cells models can be extrapolated to human asthma is unclear.
all contribute to the Th2 response through the expression of Elevated numbers of CD4 iNKT cells have recently been
IL-5 and IL-13 [113]. reported in BAL from patients with asthma [123], but other
Bronchial biopsies from asthmatics are frequently studies have failed to conrm these ndings and have con-
positive for IL-5 mRNA expression that is related to the cluded that these cells are relatively uncommon, representing
degree of eosinophilia and is consistent with a key role for less than 2% of the T-cells in the airways in either asthmatic
this Th2 cytokine is causing eosinophilia in asthma [114]. or COPD patients [124]. Therefore it is not clear how perti-
In certain forms of occupational asthma involving low nent current ndings are to asthma in general. It is also not
molecular weight sensitizers eosinophilia appears to be known what the appropriate stimulus for activation of iNKT
associated with IL-5 production by CD8 T-cells [115]. cells is or how these cells may be activated by aeroallergens.
Anti-IL-5 antibody administration markedly reduces eosi- The nal characteristic of asthma, namely remodeling
nophils in the sputum of asthmatics [116], and although has been linked to T-cell function in animals and human
tissue eosinophilia is aected also it is more resistant to the T-cells will also trigger proliferation of airway smooth
neutralizing antibody [117]. It has been dicult to demon- muscle cells [125]. Recent studies describe T-cells among
strate therapeutic benets of anti-IL-5 treatments on surro- smooth muscle bundles in asthma [126], but their pheno-
gates for asthma such as airway hyperresponsiveness or late type and possible eects on airway smooth muscle structure
allergic airway responses. However, favorable eects on tis- or function are as yet unknown.
sue matrix protein deposition were observed, likely through
downstream eects on TGF-, which is synthesized by
eosinophils in asthma. Although it is widely believed that
eosinophils are the major site of synthesis of cysteinyl leu-
kotrienes in asthma there is no formal proof that they are T-CELLS IN THE PATHOGENESIS OF
indeed responsible for the cysteinyl leukotriene synthesis
associated with late allergic responses.
COPD: HUMAN STUDIES
Other Th2 cytokines, such as IL-4 and IL-13, are
also expressed in the airway mucosa of asthmatic subjects, We now recognize that antigens, specically elastin or its
as is the Th1 cytokine IFN- and these cytokines are pre- by-products (probably among others) derived from the
dominantly expressed in T-cells [118]. Clinical trials of a injury of the lung by the innate inammatory response, are
soluble IL-4 receptor have demonstrated only minor thera- also an important trigger for the development of COPD.
peutic eects [119]. A subsequent trial utilizing Pitrakinra, a These antigens are taken up by dendritic cells not in the
recombinant mutant form of the wild-type human IL-4 that larger airways, like in asthma, but deeper inside the lung,
targets the eects of both IL-4 and IL-13, demonstrated small airways, and parenchyma, where the lung tissue suf-
that the late allergic airway response could be attenuated by fers the most injury by cigarette smoke. There is evidence
inhalation of the biological [120]. There is increasing rec- in the literature that cigarette smoking is associated with
ognition of alternative mechanisms of inammation medi- an expansion in the dendritic cell population in the lower
ated by T-cells. The presence of a neutrophil-rich inltrate respiratory tract [127] and with a marked increase in the
in some asthmatics is not well explained by the classical number of mature cells in the lung parenchyma [128]. This
Th2 paradigm. IFN- is often associated with more severe is an indication that the lung response to cigarette-smoke
asthma and the neutrophil chemoattractant CXCL8 (IL-8) exposure follows the established immune response design,
is also found. The family of cytokines that includes IL-17 including innate immunity and readiness for an adaptive
may help shed light on these alternative patterns of inam- immune response, if necessary. There is a similarity between
mation. IL-17 itself may also be a contributor to neutrophilic asthma and COPD in that they are both initiated by the
airway inammation. IL-17 mRNA can be found in asth- presentation of soluble antigenic products to dendritic cells.
matic sputum where it correlates with CXCL8 expression However, very dissimilar inammatory patterns result from
[121] and in bronchoalveolar lavage cells from asthmatic the dendritic cellT-cell interaction. In asthma, the den-
subjects ranging in severity from mild to severe [122]. dritic cell triggers a Th2 T-cell response, while in COPD it
Regulatory T-cells are being increasingly recognized results in a Th1 and a cytotoxic CD8 T-cell response that is
to participate in inammatory diseases and may suppress maintained and even progresses after the challenge by ciga-
inammation in asthma. It is a plausible hypothesis that rette smoke ceases. These dierences emphasize the impor-
defective inhibitory inuences are as important in the gen- tance of the dendritic cell, and its interaction with the antigen
esis of inammation as pro-inammatory pathways. Indeed and the inammatory milieu in determining the fate of the
induction of regulatory T-cells by an appropriate education resulting inammation and disease.
of the mucosal immune systems in early life is now pro- The study that initiated the present interest in the
posed as an alternative hygiene hypothesis. Probiotics have T-cell as a possible important cell in the pathogenesis of
been explored for their benecial eects on eczema but COPD was carried out by Finkelstein et al. in 1995 [13].
clinical trials in asthma are lacking and data on modication These authors used immunohistochemistry and morphometry

165
Asthma and COPD: Basic Mechanisms and Clinical Management

to identify the inammatory cells inltrating the alveolar wall quantied and found an increased number of apoptotic cells
and to dene the extent of emphysema in smokers and non- in the lungs of smokers with COPD which correlated with
smokers undergoing lung resection. Their ndings were sur- the numbers of CD8 cytolytic T-cells in the alveolar wall
prising at the time, as they reported that the most prominent [15]. This and other reports, showing an increased number
inammatory cell in the lung parenchyma of smokers was the of structural lung cells undergoing apoptosis in emphy-
CD3 T-lymphocyte that increased from a mean of 1546 sematous lungs [136] support the idea that CD8 T-cells
cells/mm3 in nonsmokers up to 10,000 cells/mm3 in smok- are inducing apoptosis of endothelial and epithelial cells in
ers. Furthermore, a clear correlation between the number of emphysema.
CD3 T-cells and the extent of emphysema was found, cer- The predominance of CD8 T-cells over CD4
tainly suggestive of the protagonism of the T-lymphocytes T-cells in the lungs of patients with COPD, resembles
in the pathogenesis of emphysema in smokers. Abundant the inammatory reaction seen in many viral diseases
but variable numbers of T-cells (CD3) together with other like Epstein-Barr and LCMV virus in which the num-
inammatory cells were also found in the small airways of the bers of CD8 T-cells are much greater than the CD4.
same patient population [129, 130]. Interestingly the degree Cell-associated proteins [42, 43] and particulate antigens
of airway reactivity, measured prior to surgery, correlated [44] usually presented with HSP and apoptotic cells pre-
with the load of T-cells in the airways in smokers with cen- sented by phagosomes, are much more eective at MHC
trilobular emphysema (CLE), but not in panlobular emphy- class I restricted CD8 T-cells priming than are simple
sema (PLE) or nonsmokers. Because similar total numbers soluble molecules and it can be speculated that these anti-
of CD3 T-cells were present in the two forms of emphy- genic forms mimic the tissue debris associated with cell
sema, we speculated that the T-cells in CLE were behaving injury or viral infections [40] stimulating a larger CD8
dierently, possibly indicating a mixed phenotype of Th1 and proliferation.
Th2 subsets. In support of this possibility is the nding that The relatively mild inltration with CD8 and

Th2 type cytokines IL-4 and IL-5 mRNA, typically found in CD4 T-cells found in patients with mild to moderate
asthma, are abundantly expressed by inammatory cells in the disease increases markedly in the lungs [135] and airways
wall of large airways in some smokers with chronic bronchitis of severely diseased patients. All inammatory cells, except
and COPD [131] and it is possible that clones of T-cells in B-lymphocytes, were found to be increased in the lungs of
smokers with CLE may express a Th2 cytokine prole that patients with severe emphysema, even though these patients
might induce airway reactivity in these cases. had not smoked more than the control subjects. By far,
Following the report by Finkelstein et al. [130] pro- the more numerous cells were the CD4 and the CD8
posing a role for the T-cell in COPD, several authors iden- T-cells but neutrophils, macrophages, and even eosinophils
tied the CD8 T-cell as the predominant lymphocyte were also increased. These studies are of importance show-
in the airways of smokers with COPD [132, 133]. Saetta ing that, in COPD, inammation with an abundance of
showed that the only signicant dierence in the inam- T-lymphocytes and other inammatory cells continues late
matory cell inltrate in asymptomatic smokers and smokers into the disease process.
with COPD was the increase in CD8 T-cells in the small An important nding in one of these studies was
airways of patients with COPD. Furthermore, the number that the average duration of smoking cessation in the
of CD8 T-cells were negatively correlated with the degree severe COPD subjects was 9.2 years [137] indicating that
of airow obstruction, as measured by the FEV1, again sug- the adaptive immune system continues to be involved after
gesting a possible role for these cells in the pathogenesis of smoking cessation, a phenomenon typically found in other
the disease. CD4 T-cells are also found, albeit in smaller autoimmune diseases. In favor of this possibility is the recent
numbers, in the airways of smokers with COPD and these nding of oligoclonality of T-cells in the lungs of patients
cells express IFN- and activated signal transducer and with COPD, a nding supportive of the idea that these
activator of transcription 4 (STAT 4), a transcription factor cells are accumulating in the lung secondary to antigenic
that is essential for activation and commitment of the Th1 stimulation. If correct, COPD may be considered to have an
lineage. The number of T-cells expressing activated STAT autoimmune component to its pathogenesis that is triggered
4 correlated with the degree of airow obstruction [134]. by smoking, as previously suggested [14, 15, 138].
These ndings go along with the previously mentioned Supporting the idea of COPD as an autoimmune dis-
expression of CXCR3 and CCR5 by CD4 and CD8 ease is the key paper by Lee and colleagues characterizing
T-cells and the ligand IP-10 and MIG in lung tissue and antielastin antibodies and T-helper type Th1 type responses
strongly support the idea that COPD is mediated by an in human subjects, which correlate with emphysema sever-
active Th1 immune reaction in the lung comprising both ity [31]. In response to elastin peptides, but not collagen,
CD8 and CD4 T-cells. T-cells from smokers with COPD produced IFN- and
Majo while studying lungs from nonsmokers and IL-10 but no IL-13 and these responses were decreased
smokers obtained at surgery found that the CD8 T-cells by blocking MHC class II molecules. A humoral response
are also the predominant T-cells inltrating the alveo- was also found in these patients that showed circulating
lar wall in smokers with COPD [15, 133], although antibodies against elastin but not collagen, ndings that
CD4 T-cells are also increased [15, 135]. Furthermore, implicate not only the T-cell but the B-cell in the autoim-
the number of both CD8 and CD4 increased with the mune reaction in COPD. T-regulatory cells have been well
amount smoked in smokers with COPD, but not in healthy described in COPD and they may play an important role
smokers, indicating that the CD4 T-cell are also involved in the development of COPD in smokers. Lee [31] and
in the inammatory process in COPD. Majo et al. also Barcelo [139] have shown a lower number of Tregs in the

166
The Lymphocyte in Asthma and COPD 13
lungs of smokers with COPD when compared with non- B-cells have been shown to also play an important
smokers. Furthermore smokers with normal lung function role in COPD. Lymphoid follicles containing monoclonal
had signicantly higher Tregs in the lung than nonsmok- B-cells, dendritic cells, and T-cells, predominantly CD4,
ers, suggesting an enhanced protective role by these T-cells, have been recently described in the airways [143] and
possibly controlling the development of autoimmunity parenchyma [144] of patients with COPD and also in mice
and disease in smokers with normal lung function. It has with cigarette smoke-induced emphysema. These follicles
been shown that CD8 expressing T-cells have immune function as inducible secondary lymphoid tissue for immune
regulatory properties and might play a role in the develop- responses, where antigen presentation can be accomplished
ment of COPD in some smokers. Pons described that, as without lymphatic node migration. This could explain why
seen with Tregs, smokers with normal lung function have dendritic cells found in the lungs of COPD patients express
increased numbers of CD8 T-cells in the lungs while markers of maturation, such as CD80 and CD86 [145] but
smokers with COPD do not [140]. These ndings sug- not CCR7 the homing receptor for lymph nodes [146]. It
gest a possible dysregulation of tolerance in smokers with has been suggested that lymphoid follicles may develop
COPD. in relation to microbial colonization and infection occur-
ring in the later stages of COPD. However no bacterial or
viral products were seen in the follicles suggesting that the
B-cells in the follicles, which are monoclonal, proliferate in
B-CELLS IN ASTHMA AND COPD response to specic lung antigens. The recent demonstra-
tion of IgG autoantibodies with avidity for epithelial and
endothelial cells along with the deposition of antigenanti-
About one-third of adult patients with asthma are classied body immune complexes and complement in the lungs of
as nonatopic and they often suer from more severe disease. patients with COPD further support this interpretation.
The popular term intrinsic asthma became unpopular fol- Altogether these ndings fulll the conventional criteria
lowing the observation that IgE was elevated compared to that dene the presence of antibody responses against self-
control subjects in all asthmatics, irrespective of the skin test antigens as autoimmunity [34]. Furthermore since the B-
reactivity [141] which tends to wane with age. The systemic cells were producing IgG antibodies the participation of the
reaction to sensitization with the expression of specic IgE CD4 T-cells primed for the same antigens as the B-cell is
reects an important B-cell contribution to this disease essential and suggest that a complex immunological process
and there has been renewed interest in the contribution of involving CD4 and CD8 T-cells along with the deposition
IgE to asthma since the introduction of antibody treatment of immune complexes and complement are involved in the
directed against IgE. Some further insights into the poten- mechanism of lung destruction in COPD.
tial role of IgE have been obtained through recent observa-
tions examining IgE synthesis locally in airway tissues. IgE
is characterized by its heavy-chain and it is produced after
heavy-chain switching in B-cells from IgM, IgG, or IgA to
IgE. The switch to IgE is initiated by the cytokines IL-4
or IL-13, produced principally by TH2 cells, which drive
CONCLUSIONS
germline gene transcription. Local IgE synthesis was rst
shown to occur within the nasal mucosa of patients with The role of the T-cells in mediating the dening characteris-
allergic rhinitis [142]. More recently immunopathological tics of asthma is still unclear. Airway inammation is associ-
comparisons of bronchial biopsies from atopic and nonat- ated with ndings that are quite consistent with the ndings
opic patients with asthma have demonstrated expression from animal models. Th2 type inammation follows aller-
of germline gene transcripts and expression of the high gen challenge as expected. However, in certain subsets of
anity IgE receptor, Fc RI mRNA. Evidence acquired by asthma, such as severe asthma, there is often a neutrophilic
the examination of bronchial biopsies from atopic and non- inammation and expression of cytokines and chemokines
atopic patients with asthma and appropriate controls sug- that are not associated with allergen-driven Th2 models.
gested elevated synthesis of mature IgE in the asthmatic Airway hyperresponsiveness continues to defy elucidation
bronchial mucosa by local B-cells. It was argued that local and it likely has multiple causes. In acute murine models
IgE synthesis may well account for most, if not all, biologi- IL-13 is the best single cytokine candidate to explain aller-
cally signicant IgE production, and circulating IgE may gen-induced changes in airway responsiveness. However, it
reect spillover of IgE from synthesis at mucosal sites. It is is likely that complex cascades are involved in such reactions
therefore quite possible that synthesis of bronchial mucosal in human asthma and that airway hyperresponsiveness is
IgE contributes to both atopic and nonatopic asthma, mak- the expression of these diverse pathways. While the under-
ing distinctions based on skin reactivity unnecessary and standing of the mechanisms of COPD has lagged behind
uninformative, except where it is necessary to document the exploration of the inammatory basis for asthma it is
systemic sensitization as may be required in cases of occu- now clear that an antigen-driven adaptive immune reac-
pational asthma. These observations also raise questions tion is also crucial in the production of the disease. It should
about current criteria for the restriction of monoclonal anti- be clearly understood that new emphasis on the adaptive
IgE antibodies, such as omalizumab, to patients that are immune response involving T-and B-cells does not detract
more obviously atopic. Clearly further studies are required from the importance of the innate immune inammation,
in this area to clarify the situation. neutrophils, macrophages, T-cells but rather completes

167
Asthma and COPD: Basic Mechanisms and Clinical Management

the picture. These cells are responsible for the initial tis- 15. Majo J, Ghezzo H, Cosio MG. Lymphocyte population and apoptosis
sue injury and the production of antigens that result in the in the lungs of smokers and their relation to emphysema. Eur Respir J
subsequent adaptive immune response. The autoimmune 17: 94653, 2001.
16. Bienenstock J, McDermott MR. Bronchus- and nasal-associated lym-
paradigm in COPD could explain why only a proportion
phoid tissues. Immunol Rev 2005 206: 2231, 2005.
of smokers develop COPD. Immunological tolerance based 17. Pauwels RA, Buist AS, Ma P, Jenkins CR, Hurd SS. Global strategy for
on Tregs is not an all or none phenomenon but may show the diagnosis, management, and prevention of chronic obstructive pul-
various degrees of ecacy, so that disease may occur in the monary disease: National Heart, Lung, and Blood Institute and World
susceptible subject [147]. Dierent degrees of tolerance to Health Organization Global Initiative for Chronic Obstructive Lung
self-antigens could explain the variable T-cell inamma- Disease (GOLD): Executive summary. Respir Care 46: 798825, 2001.
tion, wide range in function, and disease severity in response 18. Rahman I, Smith CA, Lawson MF, Harrison DJ, Macnee W. Induction
to similar degrees of cigarette exposure in smokers. Auto- of gamma-glutamylcysteine synthetase by cigarette smoke is associated
immunity in COPD could also contribute to the widely rec- with AP-1 in human alveolar epithelial cells. FEBS Lett 396: 2125, 1996.
ognized systemic manifestations of this disease. Hopefully 19. Sun W, Wu R, Last JA. Eects of exposure to environmental tobacco
smoke on a human tracheobronchial epithelial cell line. Toxicology 100:
the increased understanding of the mechanisms of these
16374, 1995.
diseases will provide improved treatment options in time. 20. Churg A, Zay K, Shay S, Xie C, Shapiro SD, Hendricks R, Wright JL.
Acute cigarette smoke-induced connective tissue breakdown requires
both neutrophils and macrophage metalloelastase in mice. Am J Respir
References Cell Mol Biol 27: 36874, 2002.
21. Dhami R, Gilks B, Xie C, Zay K, Wright JL, Churg A. Acute ciga-
1. Gupta S, Frenkel R, Rosenstein M, Grieco MH. Lymphocyte subpop- rette smoke-induced connective tissue breakdown is mediated by neu-
ulations, serum IgE and total eosinophil counts in patients with bron- trophils and prevented by alpha1-antitrypsin. Am J Respir Cell Mol Biol
chial asthma. Clin Exp Immunol 22: 43845, 1975. 22: 24452, 2000.
2. Strannegard IL, Lindholm L, Strannegard O. T lymphocytes in atopic 22. Harel S, Jano A, Yu SY, Hurewitz A, Bergofsky EH. Desmosine
children. Int Arch Allergy Appl Immunol 50: 68492, 1976. radioimmunoassay for measuring elastin degradation in vivo. Am Rev
3. Saxon A, Morrow C, Stevens RH. Subpopulations of circulating B cells Respir Dis 122: 76973, 1980.
and regulatory T cells involved in in vitro immunoglobulin E produc- 23. Stone PJ, Gottlieb DJ, OConnor GT, Ciccolella DE, Breuer R, Bryan-
tion in atopic patients with elevted serum immunoglobulin E. J Clin Rhad J, Shaw HA, Franzblau C, Snider GL. Elastin and collagen
Invest 65: 145768, 1980. degradation products in urine of smokers with and without chronic
4. Canonica GW, Mingari MC, Melioli G, Colombatti M, Moretta L. obstructive pulmonary disease. Am J Respir Crit Care Med 151: 95259,
Imbalances of T cell subpopulations in patients with atopic diseases 1995.
and eect of specic immunotherapy. J Immunol 123: 266972, 1979. 24. Dillon TJ, Walsh RL, Scicchitano R, Eckert B, Cleary EG, McLennan G.
5. Kus J, Tse KS, Vedal S, Chan-Yeung M. Lymphocyte sub-populations in Plasma elastin-derived peptide levels in normal adults, children, and
patients with allergic and non-allergic asthma. Clin Allergy 15: 52329, emphysematous subjects. Physiologic and computed tomographic scan
1985. correlates. Am Rev Respir Dis 146: 114348, 1992.
6. Gonzalez MC, Diaz P, Galleguillos FR, Ancic P, Cromwell O, Kay 25. Schriver EE, Davidson JM, Sutclie MC, Swindell BB, Bernard GR.
AB. Allergen-induced recruitment of bronchoalveolar helper (OKT4) Comparison of elastin peptide concentrations in body uids from
and suppressor (OKT8) T-cells in asthma. Relative increases in OKT8 healthy volunteers, smokers, and patients with chronic obstructive pul-
cells in single early responders compared with those in late- phase monary disease. Am Rev Respir Dis 145: 76266, 1992.
responders. Am Rev Respir Dis 136: 6004, 1987. 26. Betsuyaku T, Nishimura M, Yoshioka A, Takeyabu K, Miyamoto K,
7. Corrigan CJ, Hartnell A, Kay AB. T-lymphocyte activation in acute Kawakami Y. Elastin-derived peptides and neutrophil elastase in bron-
severe asthma. The Lancet 1: 112932, 1988. choalveolar lavage uid. Am J Respir Crit Care Med 154: 72024, 1996.
8. Poulter LW, Power C, Burke C. The relationship between bronchial 27. Davies SF, Oord KP, Brown MG, Campe H, Niewoehner D. Urine
immunopathology and hyperresponsiveness in asthma. Eur Respir J 3: desmosine is unrelated to cigarette smoking or to spirometric function.
79299, 1990. Am Rev Respir Dis 128: 47375, 1983.
9. Hirashima M, Tashiro K, Sakata K, Hirashima M. Isolation of an eosi- 28. Pelham F, Wewers M, Crystal R, Buist AS, Jano A. Urinary excretion of
nophil chemotactic lymphokine as a natural mediator for eosinophil desmosine (elastin cross-links) in subjects with PiZZ alpha-1-antitrypsin
chemotaxis from concanavalin A-induced skin reaction sites in guinea- deciency, a phenotype associated with hereditary predisposition to pul-
pigs. Clin Exp Immunol 57: 21119, 1984. monary emphysema. Am Rev Respir Dis 132: 82123, 1985.
10. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coman RL. 29. Viglio S, Iadarola P, Lupi A, Trisolini R, Tinelli C, Balbi B, Grassi V,
Two types of murine helper T cell clone. Denition according to pro- Worlitzsch D, Doring G, Meloni F, Meyer KC, Dowson L, Hill SL,
les of lymphokine activities and secreted proteins.. J Immunol 136: Stockley RA, Luisetti M. MEKC of desmosine and isodesmosine
234857, 1986. in urine of chronic destructive lung disease patients. Eur Respir J 15:
11. Hunninghake GW, Crystal RG. Cigarette smoking and lung destruc- 103945, 2000.
tion. Accumulation of neutrophils in the lungs of cigarette smokers. 30. Gottlieb DJ, Stone PJ, Sparrow D, Gale ME, Weiss ST, Snider GL,
Am Rev Respir Dis 128: 83338, 1983. OConnor GT. Urinary desmosine excretion in smokers with and with-
12. Martin TR, Raghu G, Maunder RJ, Springmeyer SC. The eects of out rapid decline of lung function: The Normative Aging Study. Am J
chronic bronchitis and chronic air-ow obstruction on lung cell popu- Respir Crit Care Med 154: 129095, 1996.
lations recovered by bronchoalveolar lavage. Am Rev Respir Dis 132: 31. Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S,
25460, 1985. Green L, Hacken-Bitar J, Huh J, Bakaeen F, Coxson HO, Cogswell S,
13. Finkelstein R, Fraser RS, Ghezzo H, Cosio MG. Alveolar inamma- Storness-Bliss C, Corry DB, Kheradmand F. Antielastin autoimmu-
tion and its relation to emphysema in smokers. Am J Respir Crit Care nity in tobacco smoking-induced emphysema. Nat Med 13: 56769,
Med 152: 166672, 1995. 2007.
14. Cosio MG, Majo J, Cosio MG. Inammation of the airways and lung 32. Rose N, Afanasyeva M. Autoimmunity: Busting the atherosclerotic
parenchyma in COPD: Role of T cells. Chest 121: 160S65S, 2002. plaque. Nat Med 9: 64142, 2003.

168
The Lymphocyte in Asthma and COPD 13
33. Steinman L. State of the art. Four easy pieces: Interconnections between an inammatory T helper type 2 cell response through OX40 ligand. J
tissue injury, intermediary metabolism, autoimmunity, and chronic Exp Med 202: 121323, 2005.
degeneration. Proc Am Thorac Soc 3: 48486, 2006. 51. Sokol CL, Barton GM, Farr AG, Medzhitov R. A mechanism for the
34. Feghali-Bostwick CA, Gadgil AS, Otterbein LE, Pilewski JM, initiation of allergen-induced T helper type 2 responses. Nat Immunol,
Stoner MW, Csizmadia E, Zhang Y, Sciurba FC, Duncan SR. 2007.
Autoantibodies in patients with chronic obstructive pulmonary disease. 52. Sayed BA, Brown MA. Mast cells as modulators of T-cell responses.
Am J Respir Crit Care Med 177: 15663, 2008. Immunol Rev 217: 5364, 2007.
35. Lambrecht BN. Dendritic cells and the regulation of the allergic 53. Machida I, Matsuse H, Kondo Y, Kawano T, Saeki S, Tomari S, Obase Y,
immune response. Allergy 60: 27182, 2005. Fukushima C, Kohno S. Cysteinyl leukotrienes regulate dendritic cell
36. Henrickson SE, Mempel TR, Mazo IB, Liu B, Artyomov MN, Zheng functions in a murine model of asthma. J Immunol 172: 183338, 2004.
H, Peixoto A, Flynn MP, Senman B, Junt T, Wong HC, Chakraborty 54. Finotto S, Neurath MF, Glickman JN, Qin S, Lehr HA, Green FH,
AK, von Andrian UH. T cell sensing of antigen dose governs interac- Ackerman K, Haley K, Galle PR, Szabo SJ, Drazen JM, De Sanctis GT,
tive behavior with dendritic cells and sets a threshold for T cell activa- Glimcher LH. Development of spontaneous airway changes consistent
tion. Nat Immunol 9: 28291, 2008. with human asthma in mice lacking T-bet. Science 295: 33638, 2002.
37. Huh JC, Strickland DH, Jahnsen FL, Turner DJ, Thomas JA, Napoli S, 55. Freeman CM, Curtis JL, Chensue SW. CC chemokine receptor 5 and
Tobagus I, Stumbles PA, Sly PD, Holt PG. Bidirectional interactions CXC chemokine receptor 6 expression by lung CD8 cells correlates
between antigen-bearing respiratory tract dendritic cells (DCs) and T with chronic obstructive pulmonary disease severity. Am J Pathol 171:
cells precede the late phase reaction in experimental asthma: DC acti- 76776, 2007.
vation occurs in the airway mucosa but not in the lung parenchyma. 56. Bonecchi R, Bianchi G, Bordignon PP, DAmbrosio D, Lang R,
J Exp Med 198: 1930, 2003. Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A, Sinigaglia F.
38. Burgdorf S, Kurts C. Endocytosis mechanisms and the cell biology of Dierential expression of chemokine receptors and chemotactic
antigen presentation. Curr Opin Immunol 20: 8995, 2008. responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med 187:
39. Srivastava PK, Menoret A, Basu S, Binder RJ, McQuade KL. Heat 12934, 1998.
shock proteins come of age: Primitive functions acquire new roles in an 57. Sekiya T, Miyamasu M, Imanishi M, Yamada H, Nakajima T, Yamaguchi
adaptive world. Immunity 8: 65765, 1998. M, Fujisawa T, Pawankar R, Sano Y, Ohta K, Ishii A, Morita Y,
40. Srivastava P. Interaction of heat shock proteins with peptides and anti- Yamamoto K, Matsushima K, Yoshie O, Hirai K. Inducible expression
gen presenting cells: Chaperoning of the innate and adaptive immune of a Th2-type CC chemokine thymus- and activation-regulated chem-
responses. Ann Rev Immunol 20: 395425, 2002. okine by human bronchial epithelial cells. J Immunol 165: 220513,
41. Porcelli SA, Modlin RL. The CD1 system: Antigen-presenting mol- 2000.
ecules for T cell recognition of lipids and glycolipids. Ann Rev Immunol 58. Grumelli S, Corry DB, Song LZ, Song L, Green L, Huh J, Hacken J,
17: 297329, 1999. Espada R, Bag R, Lewis DE, Kheradmand F. An immune basis for
42. Bachmann MF, Kundig TM, Freer G, Li Y, Kang CY, Bishop DH, lung parenchymal destruction in chronic obstructive pulmonary disease
Hengartner H, Zinkernagel RM. Induction of protective cytotoxic T and emphysema. PLoS Med 1: e8, 2004.
cells with viral proteins. Eur J Immunol 24: 222836, 1994. 59. Watanabe A, Mishima H, Renzi PM, Xu LJ, Hamid Q, Martin JG.
43. Carbone FR, Bevan MJ. Class I-restricted processing and presentation Transfer of allergic airway responses with antigen-primed CD4 but not
of exogenous cell-associated antigen in vivo. J Exp Med 171: 37787, CD8 T cells in Brown Norway rats. J Clin Invest 96: 130310, 1995.
1990. 60. Haczku A, Macary P, Huang TJ, Tsukagoshi H, Barnes PJ, Kay AB,
44. Kovacsovics-Bankowski M, Clark K, Benacerraf B, Rock KL. Ecient Kemeny DM, Chung KF, Moqbel R. Adoptive transfer of allergen-
major histocompatibility complex class I presentation of exogenous specic CD4 T cells induces airway inammation and hyperrespon-
antigen upon phagocytosis by macrophages. Proc Natl Acad Sci USA 90: siveness in brown-Norway rats. Immunology 91: 17685, 1997.
494246, 1993. 61. Mishima H, Hojo M, Watanabe A, Hamid QA, Martin JG. CD4 T
45. Castellino F, Huang AY, tan-Bonnet G, Stoll S, Scheinecker C, cells can induce airway hyperresponsiveness to allergen challenge in the
Germain RN. Chemokines enhance immunity by guiding naive CD8 brown norway rat. Am J Respir Crit Care Med 158: 186370, 1998.
T cells to sites of CD4 T celldendritic cell interaction. Nature 440: 62. Gavett SH, Chen X, Finkelman F, Wills-Karp M. Depletion of murine
89095, 2006. CD4 T lymphocytes prevents antigen-induced airway hyperreactivity
46. Stumbles PA, Thomas JA, Pimm CL, Lee PT, Venaille TJ, Proksch S, and pulmonary eosinophilia. Am J Respir Cell Mol Biol 10: 58793, 1994.
Holt PG. Resting respiratory tract dendritic cells preferentially 63. Isogai S, Taha R, Tamaoka M, Yoshizawa Y, Hamid Q, Martin JG.
stimulate T helper cell type 2 (Th2) responses and require obligatory CD8() alpha beta T cells can mediate late airway responses and air-
cytokine signals for induction of Th1 immunity. J Exp Med 188: 2019 way eosinophilia in rats. J Allergy Clin Immunol 114: 134552, 2004.
31, 1998. 64. Schwarze J, Cieslewicz G, Joetham A, Ikemura T, Hamelmann E,
47. Eisenbarth SC, Piggott DA, Huleatt JW, Visintin I, Herrick CA, Gelfand EW. CD8 T cells are essential in the development of respira-
Bottomly K. Lipopolysaccharide-enhanced, toll-like receptor 4-dependent tory syncytial virus-induced lung eosinophilia and airway hyperrespon-
T helper cell type 2 responses to inhaled antigen. J Exp Med 196: 164551, siveness. J Immunol 162: 420711, 1999.
2002. 65. Ramos-Barbon D, Presley JF, Hamid QA, Fixman ED, Martin JG.
48. Dillon S, Agrawal A, van DT, Landreth G, McCauley L, Koh A, Antigen-specic CD4 T cells drive airway smooth muscle remod-
Maliszewski C, Akira S, Pulendran B. A Toll-like receptor 2 ligand eling in experimental asthma. J Clin Invest 115: 158089, 2005.
stimulates Th2 responses in vivo, via induction of extracellular signal- 66. Guerassimov A, Hoshino Y, Takubo Y, Turcotte A, Yamamoto M,
regulated kinase mitogen-activated protein kinase and c-Fos in den- Ghezzo H, Triantallopoulos A, Whittaker K, Hoidal JR, Cosio MG.
dritic cells. J Immunol 172: 473343, 2004. The development of emphysema in cigarette smoke-exposed mice is
49. Yang D, Chen Q, Su SB, Zhang P, Kurosaka K, Caspi RR, Michalek strain dependent. Am J Respir Crit Care Med 170: 97480, 2004.
SM, Rosenberg HF, Zhang N, Oppenheim JJ. Eosinophil-derived neu- 67. Takubo Y, Guerassimov A, Ghezzo H, Triantallopoulos A, Bates JH,
rotoxin acts as an alarmin to activate the TLR2-MyD88 signal path- Hoidal JR, Cosio MG. Alpha1-antitrypsin determines the pattern of
way in dendritic cells and enhances Th2 immune responses. J Exp Med emphysema and function in tobacco smoke-exposed mice: Parallels
205: 7990, 2008. with human disease. Am J Respir Crit Care Med 166: 1596603, 2002.
50. Ito T, Wang YH, Duramad O, Hori T, Delespesse GJ, Watanabe N, 68. Maeno T, Houghton AM, Quintero PA, Grumelli S, Owen CA,
Qin FX, Yao Z, Cao W, Liu YJ. TSLP-activated dendritic cells induce Shapiro SD. CD8 T Cells are required for inammation and

169
Asthma and COPD: Basic Mechanisms and Clinical Management

destruction in cigarette smoke-induced emphysema in mice. J Immunol 86. Holt PG, Strickland DH, Wikstrom ME, Jahnsen FL. Regulation of
178: 809096, 2007. immunological homeostasis in the respiratory tract. Nat Rev Immunol
69. Taraseviciene-Stewart L, Scerbavicius R, Choe KH, Moore M, 8: 14252, 2008.
Sullivan A, Nicolls MR, Fontenot AP, Tuder RM, Voelkel NF. An ani- 87. Akbari O, Stock P, Meyer E, Kronenberg M, Sidobre S, Nakayama T,
mal model of autoimmune emphysema. Am J Respir Crit Care Med 171: Taniguchi M, Grusby MJ, Dekruy RH, Umetsu DT. Essential role of
73442, 2005. NKT cells producing IL-4 and IL-13 in the development of allergen-
70. Li L, Sad S, Kagi D, Mosmann TR. CD8Tc1 and Tc2 cells secrete dis- induced airway hyperreactivity. Nat Med 9: 58288, 2003.
tinct cytokine patterns in vitro and in vivo but induce similar inam- 88. Meyer EH, Goya S, Akbari O, Berry GJ, Savage PB, Kronenberg
matory reactions. J Immunol 158: 415261, 1997. M, Nakayama T, Dekruy RH, Umetsu DT. Glycolipid activation of
71. van Beelen AJ, Zelinkova Z, Taanman-Kueter EW, Muller FJ, invariant T cell receptor NK T cells is sucient to induce airway
Hommes DW, Zaat SA, Kapsenberg ML, de Jong EC. Stimulation hyperreactivity independent of conventional CD4 T cells. Proc Natl
of the intracellular bacterial sensor NOD2 programs dendritic cells Acad Sci USA 103: 278287, 2006.
to promote interleukin-17 production in human memory T cells. 89. Renzi PM, Sapienza S, Du T, Wang NS, Martin JG. Lymphokine-
Immunity 27: 66069, 2007. induced airway hyperresponsiveness in the rat. Am Rev Respir Dis
72. Kano S, Sato K, Morishita Y, Vollstedt S, Kim S, Bishop K, Honda K, 143: 37579, 1991.
Kubo M, Taniguchi T. The contribution of transcription factor IRF1 90. Renzi PM, Sapienza S, Waserman S, Du T, Olivenstein R, Wang NS,
to the interferon-gamma-interleukin 12 signaling axis and TH1 versus Martin JG. Eect of interleukin-2 on the airway response to antigen
TH17 dierentiation of CD4 T cells. Nat Immunol 9: 3441, 2008. in the rat. Am Rev Respir Dis 146: 16369, 1992.
73. Schnyder-Candrian S, Togbe D, Couillin I, Mercier I, Brombacher F, 91. Nag S, Lamkhioued B, Renzi PM. Interleukin-2-induced increased
Quesniaux V, Fossiez F, Ryel B, Schnyder B. Interleukin-17 is a nega- airway responsiveness and lung Th2 cytokine expression occur after
tive regulator of established allergic asthma. J Exp Med 203: 271525, antigen challenge through the leukotriene pathway. Am J Respir Crit
2006. Care Med 165: 154045, 2002.
74. Hellings PW, Kasran A, Liu Z, Vandekerckhove P, Wuyts A, 92. Renzi PM, Xu L, Yang XX, Powell WS, Martin JG. IL-2 enhances
Overbergh L, Mathieu C, Ceuppens JL. Interleukin-17 orchestrates allergic airway responses in rats by increased inammation but not
the granulocyte inux into airways after allergen inhalation in a mouse through increased synthesis of cysteinyl leukotrienes. J Allergy Clin
model of allergic asthma. Am J Respir Cell Mol Biol 28: 4250, 2003. Immunol 104: 14552, 1999.
75. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, 93. Corry DB, Folkesson HG, Warnock ML, Erle DJ, Matthay MA,
Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the WienerKronish JP, Locksley RM. Interleukin 4, but not interleukin
generation of pathogenic eector TH17 and regulatory T cells. Nature 5 or eosinophils, is required in a murine model of acute airway hyper-
441: 23538, 2006. reactivity. J Exp Med 183: 10917, 1996.
76. Kearley J, Barker JE, Robinson DS, Lloyd CM. Resolution of air- 94. Eum SY, Maghni K, Tolloczko B, Eidelman DH, Martin JG. IL-13
way inammation and hyperreactivity after in vivo transfer of may mediate allergen-induced hyperresponsiveness independently
CD4CD25 regulatory T cells is interleukin 10 dependent. J Exp of IL-5 or eotaxin by eects on airway smooth muscle. Am J Physiol
Med 202: 153947, 2005. Lung Cell Mol Physiol 288: L57684, 2005.
77. Joetham A, Takeda K, Taube C, Miyahara N, Matsubara S, Koya T, 95. Walter DM, McIntire JJ, Berry G, McKenzie AN, Donaldson DD,
Rha YH, Dakhama A, Gelfand EW. Naturally occurring lung Dekruy RH, Umetsu DT. Critical role for IL-13 in the development
CD4()CD25() T cell regulation of airway allergic responses depends of allergen-induced airway hyperreactivity. J Immunol 167: 466875,
on IL-10 induction of TGF-beta. J Immunol 178: 143342, 2007. 2001.
78. Strickland DH, Stumbles PA, Zosky GR, Subrata LS, Thomas JA, 96. Wills-Karp M, Luyimbazi J, Xu X, Schoeld B, Neben TY, Karp CL,
Turner DJ, Sly PD, Holt PG. Reversal of airway hyperresponsiveness Donaldson DD. Interleukin-13: Central mediator of allergic asthma.
by induction of airway mucosal CD4CD25 regulatory T cells. J Exp Science 282: 225861, 1998.
Med 203: 264960, 2006. 97. Kuperman DA, Huang X, Koth LL, Chang GH, Dolganov GM, Zhu Z,
79. Boismenu R, Havran WL. Gammadelta T cells in host defense and Elias JA, Sheppard D, Erle DJ. Direct eects of interleukin-13 on
epithelial cell biology. Clin Immunol Immunopathol 86: 12133, 1998. epithelial cells cause airway hyperreactivity and mucus overproduction
80. McMenamin C, Pimm C, McKersey M, Holt PG. Regulation of IgE in asthma. Nat Med 8: 88589, 2002.
responses to inhaled antigen in mice by antigen-specic gamma delta 98. Finotto S, Hausding M, Doganci A, Maxeiner JH, Lehr HA, Luft C,
T cells. Science 265: 186971, 1994. Galle PR, Glimcher LH. Asthmatic changes in mice lacking T-bet
81. McMenamin C, McKersey M, Kuhnlein P, Hunig T, Holt PG. are mediated by IL-13. Int Immunol 17: 9931007, 2005.
Gamma delta T cells down-regulate primary IgE responses in rats to 99. Eum SY, Maghni K, Hamid G, Eidelman DH, Campbell H, Isogai S,
inhaled soluble protein antigens. J Immunol 154: 439094, 1995. Martin JG. Inhibition of allergic airways inammation and airway
82. Chen YP, Chou K, Fuchs E, Havran WL, Boismenu R. Protection of hyperresponsiveness in mice by dexamethasone: Role of eosinophils,
the intestinal mucosa by intraepithelial gamma delta T cells. Proc Natl IL-5, eotaxin, and IL-13. J Allergy Clin Immunol 111: 104961, 2003.
Acad Sci USA 99: 1433843, 2002. 100. Mould AW, Ramsay AJ, Matthaei KI, Young IG, Rothenberg ME,
83. Isogai S, Athiviraham A, Fraser RS, Taha R, Hamid Q , Martin JG. Foster PS. The eect of IL-5 and eotaxin expression in the lung on
Interferon-gamma-dependent inhibition of late allergic airway responses eosinophil tracking and degranulation and the induction of bron-
and eosinophilia by CD8 gammadelta T cells. Immunology 122: 23038, chial hyperreactivity. J Immunol 164: 214250, 2000.
2007. 101. Nag SS, Xu LJ, Hamid Q, Renzi PM. The eects of IL-5 on airway
84. Hahn YS, Taube C, Jin N, Takeda K, Park JW, Wands JM, Aydintug physiology and inammation in rats. J Allergy Clin Immunol 111:
MK, Roark CL, Lahn M, OBrien RL, Gelfand EW, Born WK. 55866, 2003.
V gamma 4 gamma delta T cells regulate airway hyperreactivity to 102. Lee JJ, Dimina D, Macias MP, Ochkur SI, McGarry MP, ONeill KR,
methacholine in ovalbumin-sensitized and challenged mice. J Immunol Protheroe C, Pero R, Nguyen T, Cormier SA, Lenkiewicz E, Colbert D,
171: 317078, 2003. Rinaldi L, Ackerman SJ, Irvin CG, Lee NA. Dening a link with
85. Lahn M, Kanehiro A, Takeda K, Terry J, Hahn YS, Aydintug MK, asthma in mice congenitally decient in eosinophils. Science 305:
Konowal A, Ikuta K, OBrien RL, Gelfand EW, Born WK. MHC 177376, 2004.
class I-dependent Vgamma4 pulmonary T cells regulate alpha beta 103. Mattes J, Yang M, Mahalingam S, Kuehr J, Webb DC, Simson L,
T cell-independent airway responsiveness. Proc Natl Acad Sci USA 99: Hogan SP, Koskinen A, McKenzie AN, Dent LA, Rothenberg
885055, 2002. ME, Matthaei KI, Young IG, Foster PS. Intrinsic defect in T-cell

170
The Lymphocyte in Asthma and COPD 13
production of interleukin (IL)-13 in the absence of both IL-5 and and TH1-type (interleukin 2 and interferon gamma) cytokines in
eotaxin precludes the development of eosinophilia and airways hyper- bronchoalveolar lavage and bronchial biopsies from atopic asthmatic
reactivity in experimental asthma. J Exp Med 195: 143344, 2002. and normal control subjects. Am J Respir Cell Mol Biol 12: 47787,
104. Oh JW, Seroogy CM, Meyer EH, Akbari O, Berry G, Fathman CG, 1995.
Dekruy RH, Umetsu DT. CD4 T-helper cells engineered to produce 119. Borish LC, Nelson HS, Lanz MJ, Claussen L, Whitmore JB,
IL-10 prevent allergen-induced airway hyperreactivity and inamma- Agosti JM, Garrison L. Interleukin-4 receptor in moderate atopic
tion. J Allergy Clin Immunol 110: 46068, 2002. asthma-A phase I/II randomized, placebo-controlled trial. Am J
105. Pichavant M, Goya S, Meyer EH, Johnston RA, Kim HY, Respir Crit Care Med 160: 181623, 1999.
Matangkasombut P, Zhu M, Iwakura Y, Savage PB, Dekruy RH, 120. Wenzel S, Wilbraham D, Fuller R, Getz EB, Longphre M. Eect
Shore SA, Umetsu DT. Ozone exposure in a mouse model induces of an interleukin-4 variant on late phase asthmatic response to aller-
airway hyperreactivity that requires the presence of natural killer T gen challenge in asthmatic patients: Results of two phase 2a studies.
cells and IL-17. J Exp Med 205: 38593, 2008. Lancet 370: 142231, 2007.
106. Saetta M, Mariani M, Panina-Bordignon P, Turato G, Buonsanti C, 121. Bullens DM, Truyen E, Coteur L, Dilissen E, Hellings PW,
Baraldo S et al. Increased expression of the chemokine receptor Dupont LJ, Ceuppens JL. IL-17 mRNA in sputum of asthmatic
CXCR3 and its ligand CXCL10 in peripheral airways of smokers patients: Linking T cell driven inammation and granulocytic inux?
with chronic obstructive pulmonary disease. Am J Respir Crit Care Respir Res 7: 135, 2006.
Med 165: 14049, 2002. 122. Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Page N, Olivenstein R,
107. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, Riese RJ Jr., Elias J, Chakir J. IL-17 is increased in asthmatic airways and induces
Chapman HA Jr., Shapiro SD, Elias JA. Inducible targeting of IL- human bronchial broblasts to produce cytokines. J Allergy Clin
13 to the adult lung causes matrix metalloproteinase- and cathepsin- Immunol 108: 43038, 2001.
dependent emphysema. J Clin Invest 106: 108193, 2000. 123. Akbari O, Faul JL, Hoyte EG, Berry GJ, Wahlstrom J, Kronenberg M,
108. Hoshino T, Kato S, Oka N, Imaoka H, Kinoshita T, Takei S, Kitasato Y, Dekruy RH, Umetsu DT. CD4 invariant T-cell-receptor natural
Kawayama T, Imaizumi T, Yamada K, Young HA, Aizawa H. killer T cells in bronchial asthma. N Engl J Med 354: 111729, 2006.
Pulmonary inammation and emphysema: Role of the cytokines IL- 124. Vijayanand P, Seumois G, Pickard C, Powell RM, Angco G, Sammut D,
18 and IL-13. Am J Respir Crit Care Med 176: 4962, 2007. Gadola SD, Friedmann PS, Djukanovic R. Invariant natural killer T cells
109. Robinson DS, Hamid Q, Ying S, Tsicopoulos A, Barkans J, in asthma and chronic obstructive pulmonary disease. N Engl J Med 356:
Bentley AM, Corrigan C, Durham SR, Kay AB. Predominant TH2- 141022, 2007.
like bronchoalveolar T-lymphocyte population in atopic asthma. 125. Lazaar AL, Albelda SM, Pilewski JM, Brennan B, Pure E,
N Engl J Med 326: 298304, 1992. Panettieri RA Jr., T lymphocytes adhere to airway smooth muscle
110. Robinson D, Hamid Q, Bentley A, Ying S, Kay AB, Durham SR. cells via integrins and CD44 and induce smooth muscle cell DNA
Activation of CD4 T cells, increased TH2-type cytokine mRNA synthesis. J Exp Med 180: 80716, 1994.
expression, and eosinophil recruitment in bronchoalveolar lavage after 126. Begueret H, Berger P, Vernejoux JM, Dubuisson L, Marthan R,
allergen inhalation challenge in patients with atopic asthma. J Allergy Tunon-de-Lara JM. Inammation of bronchial smooth muscle in
Clin Immunol 92: 31324, 1993. allergic asthma. Thorax 62: 815, 2007.
111. Olivenstein R, Taha R, Minshall EM, Hamid QA. IL-4 and IL-5 127. Casolaro MA, Bernaudin JF, Saltini C, Ferrans VJ, Crystal RG.
mRNA expression in induced sputum of asthmatic subjects: Comparison Accumulation of Langerhans cells on the epithelial surface of the
with bronchial wash. J Allergy Clin Immunol 103: 23845, 1999. lower respiratory tract in normal subjects in association with cigarette
112. Nakamura Y, Ghaar O, Olivenstein R, Taha RA, Soussi-Gounni A, smoking. Am Rev Respir Dis 137: 40611, 1988.
Zhang DH, Ray A, Hamid Q. Gene expression of the GATA-3 tran- 128. Soler P, Moreau A, Basset F, Hance AJ. Cigarette smoking-induced
scription factor is increased in atopic asthma. J Allergy Clin Immunol changes in the number and dierentiated state of pulmonary den-
103: 21522, 1999. dritic cells/Langerhans cells. Am Rev Respir Dis 139: 111217, 1989.
113. Krug N, Erpenbeck VJ, Balke K, Petschallies J, Tschernig T, 129. Bosken CH, Hards J, Gatter K, Hogg JC. Characterization of the
Hohlfeld JM, Fabel H. Cytokine prole of bronchoalveolar lavage- inammatory reaction in the peripheral airways of cigarette smokers
derived CD4(), CD8(), and gammadelta T cells in people with using immunocytochemistry. Am Rev Respir Dis 145: 91117, 1992.
asthma after segmental allergen challenge. Am J Respir Cell Mol Biol 25: 130. Finkelstein R, Ma HD, Ghezzo H, Whittaker K, Fraser RS,
12531, 2001. Cosio MG. Morphometry of small airways in smokers and its rela-
114. Hamid Q , Azzawi M, Ying S, Moqbel R, Wardlaw AJ, Corrigan CJ, tionship to emphysema type and hyperresponsiveness. Am J Respir
Bradley B, Durham SR, Collins JV, Jeery PK. Expression of mRNA Crit Care Med 152: 26776, 1995.
for interleukin-5 in mucosal bronchial biopsies from asthma. J Clin 131. Zhu J, Majumdar S, Qiu Y, Ansari T, Oliva A, Kips JC, Pauwels RA,
Invest 87: 154146, 1991. De RV, Jeery PK. Interleukin-4 and interleukin-5 gene expression
115. Maestrelli P, Del Prete GF, De CM, DElios MM, Saetta M, Di SA, and inammation in the mucus-secreting glands and subepithelial
Mapp CE, Romagnani S, Fabbri LM. CD8 T-cell clones producing tissue of smokers with chronic bronchitis. Lack of relationship with
interleukin-5 and interferon-gamma in bronchial mucosa of patients CD8() cells. Am J Respir Crit Care Med 164: 222028, 2001.
with asthma induced by toluene diisocyanate. Scand J Work Environ 132. OShaughnessy TC, Ansari TW, Barnes NC, Jeery PK. Inammation
Health 20: 37681, 1994. in bronchial biopsies of subjects with chronic bronchitis: Inverse rela-
116. Leckie MJ, ten Brinke A, Khan J, Diamant Z, OConnor BJ, Walls CM, tionship of CD8 T lymphocytes with FEV1. Am J Respir Crit Care
Mathur AK, Cowley HC, Chung KF, Djukanovic R, Hansel TT, Med 155: 85257, 1997.
Holgate ST, Sterk PJ, Barnes PJ. Eects of an interleukin-5 blocking 133. Saetta M, Di SA, Turato G, Facchini FM, Corbino L, Mapp CE,
monoclonal antibody on eosinophils, airway hyper-responsiveness, Maestrelli P, Ciaccia A, Fabbri LM. CD8 T-lymphocytes in periph-
and the late asthmatic response. Lancet 356: 214448, 2000. eral airways of smokers with chronic obstructive pulmonary disease.
117. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Am J Respir Crit Care Med 157: 82226, 1998.
Ludwig MS, Barnes N, Robinson D, Kay AB. Anti-IL-5 treatment 134. Di SA, Caramori G, Capelli A, Gnemmi I, Ricciardolo FL, Oates T,
reduces deposition of ECM proteins in the bronchial subepithelial Donner CF, Chung KF, Barnes PJ, Adcock IM. STAT4 activation in
basement membrane of mild atopic asthmatics. J Clin Invest 112: smokers and patients with chronic obstructive pulmonary disease. Eur
102936, 2003. Respir J 24: 7885, 2004.
118. Ying S, Durham SR, Corrigan CJ, Hamid Q, Kay AB. Phenotype of 135. Retamales I, Elliott WM, Meshi B, Coxson HO, Pare PD, Sciurba
cells expressing mRNA for TH2-type (interleukin 4 and interleukin 5) FC, Rogers RM, Hayashi S, Hogg JC. Amplication of inammation

171
Asthma and COPD: Basic Mechanisms and Clinical Management

in emphysema and its association with latent adenoviral infection. Am of epsilon germline gene transcripts, IL-4, and IL-13 in allergic nasal
J Respir Crit Care Med 164: 46973, 2001. mucosa after ex vivo allergen exposure. J Allergy Clin Immunol 106:
136. Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF. 4652, 2000.
Endothelial cell death and decreased expression of vascular endothe- 143. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
lial growth factor and vascular endothelial growth factor receptor 2 in Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The
emphysema. Am J Respir Crit Care Med 163: 73744, 2001. nature of small-airway obstruction in chronic obstructive pulmonary
137. Shapiro SD. End-stage chronic obstructive pulmonary disease: The disease. N Engl J Med 350: 264553, 2004.
cigarette is burned out but inammation rages on. Am J Respir Crit 144. van der Strate BW, Postma DS, Brandsma CA, Melgert BN,
Care Med 164: 33940, 2001. Luinge MA, Geerlings M, Hylkema MN, van den BA, Timens W,
138. Agusti A, Macnee W, Donaldson K, Cosio M. Hypothesis: Does Kerstjens HA. Cigarette smoke-induced emphysema: A role for the B
COPD have an autoimmune component? Thorax 58: 83234, 2003. cell? Am J Respir Crit Care Med 173: 75158, 2006.
139. Barcelo B, Pons J, Ferrer JM, Sauleda J, Fuster A, Agusti AG. 145. Demedts IK, Bracke KR, Van PG, Testelmans D, Verleden GM,
Phenotypic characterisation of T-lymphocytes in COPD: Abnormal Vermassen FE, Joos GF, Brusselle GG. Accumulation of dendritic
CD4CD25 regulatory T-lymphocyte response to tobacco smoking. cells and increased CCL20 levels in the airways of patients with
Eur Respir J 31: 55562, 2008. chronic obstructive pulmonary disease. Am J Respir Crit Care Med
140. Pons J, Sauleda J, Ferrer JM, Barcelo B, Fuster A, Regueiro V, 175: 9981005, 2007.
Julia MR, Agusti AG. Blunted gamma delta T-lymphocyte response 146. Bratke K, Klug M, Bier A, Julius P, Kuepper M, Virchow JC,
in chronic obstructive pulmonary disease. Eur Respir J 25: 44146, 2005. Lommatzsch M. Function-associated surface molecules on airway
141. Burrows B, Martinez FD, Halonen M, Barbee RA, Cline MG. dendritic cells in cigarette smokers. Am J Respir Cell Mol Biol, 2008.
Association of asthma with serum IgE levels and skin-test reactivity 147. Brent L. The 50th anniversary of the discovery of immunologic toler-
to allergens. N Engl J Med 320: 27177, 1989. ance. N Engl J Med 349: 138183, 2003.
142. Cameron L, Hamid Q, Wright E, Nakamura Y, Christodoulopoulos P,
Muro S, Frenkiel S, Lavigne F, Durham S, Gould H. Local synthesis

172
The Neutrophil and Its Special Role in
Chronic Obstructive Pulmonary Disease
14CHAPTER

INTRODUCTION and CT quantication of emphysema over the Elizabeth Sapey1 and


subsequent 4 years. Gas transfer (the most direct Robert A. Stockley2
physiological measure of emphysema) has been
1
There is a substantial body of evidence to support shown to be inversely proportional to the lev- Department of Medicine, Birmingham
the hypothesis that the neutrophil is the primary els of neutrophil-associated markers, such as University, Birmingham, UK
eector cell in chronic obstructive pulmonary myeloperoxidase and human neutrophil lipoca- 2
Lung Investigation Unit, University
disease (COPD) and that neutrophil proteinases, lin, in patients with COPD [13]. Both clinical Hospital Birmingham, NHS Foundation
especially neutrophil elastase (NE), are responsi- and subclinical emphysema (noted on HRCT) Trust, Edgbaston, Birmingham, UK
ble for the main pathological features seen. are associated with an increase in NE and other
Studies have shown that patients with neutrophil proteins in BALF [14, 15] and in
COPD have increased numbers of neutrophils established emphysema, severity is propor-
in sputum [1, 2] and bronchoalveolar lavage tional to NE immunoreactivity in tissue [16]
uid (BALF) [3] compared with asympto- and enzyme activity in BALF [17]. Finally, both
matic smokers, and that the percentage of neu- neutrophil counts and NE concentration appear
trophils in BALF is higher in patients with the to decline with smoking cessation [18] which is
greatest degree of airow obstruction [4, 5]. consistent with the benets of this intervention.
Furthermore, in a recent study of bronchial COPD, while primarily a lung disease, is
biopsies, small airways intraepithelial neutrophil associated with increased co-morbidity includ-
counts were greater in patients with COPD ing cardiovascular disease and systemic pathol-
compared with both smoking and nonsmoking ogy such as muscle wasting and dysfunction.
controls, and correlated with airway obstruc- It has been hypothesized that persistent low
tion [6]. Increased neutrophil counts have been grade inammation may drive the co-morbidity
found in the bronchial walls and in the BALF and the systemic eects noted with this disease
samples from patients during exacerbations of [19]. The systemic manifestations of COPD are
COPD [7, 8], with increased neutrophil seques- important, as they are not only associated with
tration in the pulmonary microcirculation and increased morbidity, but are also predictive of
such episodes relate to subsequent progression disease outcome, especially body mass index
of airow obstruction [9]. Resolution of the (BMI) which forms part of the BODE index
neutrophilic inammation occurs approximately (body mass, airow obstruction, dyspnea, and
5 days after appropriate treatment of the exac- exercise capacity) [20]. There is controversy con-
erbation, which coincides with clinical recovery cerning whether increased and sustained pul-
[10]. In addition, a recent study by Donaldson monary inammation causes increased systemic
et al. [11] reported that patients with higher inammation. However C-reactive protein
numbers of neutrophils in sputum had a faster (CRP) (a generic marker of systemic inamma-
decline in FEV1 compared with those with tion) is raised in patients with COPD compared
lower neutrophil counts, losing approximately to healthy controls [21] irrespective of cigarette
1% more than predicted each year. smoke exposure and ischemic heart disease (an
Furthermore Parr et al. [12] demonstrated independent cause of a raised CRP). It is unclear
that baseline markers of neutrophilic inamma- whether these systemic changes are incidental or
tion relate to subsequent decline of lung function driven by pulmonary inammation. In a recent

173
Asthma and COPD: Basic Mechanisms and Clinical Management

study of 16 patients with moderate to severe COPD, a rela- to excessive tissue destruction [23] Tables 14.1 and 14.2
tionship was present between absolute sputum neutrophil provides an overview of neutrophil proteinases and anti-
counts, CRP, and BMI less than 21 (see Fig. 14.1). These proteinases in COPD.
data indicate an association between the neutrophil load in This chapter provides an overview of the role of the
the lungs and systemic manifestations of the disease, again neutrophil in COPD, starting with neutrophil maturation
highlighting the central role of this cell in many aspects of and structure. Cytokines and chemoattractants which are
COPD. important in neutrophil activation and recruitment will
The neutrophil was rst implicated in the pathogen- be discussed and neutrophil migration into lung and neu-
esis and progression of COPD in 1963, where it was noted trophil apoptosis will be outlined, with particular reference
that severe emphysema in early adulthood was linked with a to COPD. The evidence for the actions of neutrophil pro-
deciency of alpha 1 antitrypsin (1-AT), the serum inhibi- teinases in the pathogenesis of COPD will be reviewed,
tor of the proteolytic enzyme NE [22]. Since then, the highlighting in vitro and in vivo work. Finally, the role that
Proteinase/ Anti-proteinase theory has dominated research neutrophils may play in asthma, likely through similar path-
in COPD. This states that in health, proteinase activity is ogenic mechanisms is discussed briey.
controlled by anti-proteinases, such as 1-AT and secre-
tory leukocyte proteinase inhibitor (SLPI), thereby limit-
ing tissue damage. In COPD, an imbalance is believed to
exist, either from the increased activity of proteinases or a NEUTROPHIL STRUCTURE, FUNCTION,
real or functional deciency in anti-proteinases, which leads AND APOPTOSIS

Neutrophils have a characteristic multilobed nucleus and


33
abundant storage granules in their cytoplasm. The mature
31 neutrophil has three chemically distinct granule types, which
29
27
BMI (kg/m2)

25
TABLE 14.1 Overview of neutrophil serine proteinases.
23
21 Neutrophil elastase Cathepsin G Proteinase 3
19
17
Size 218 amino acids 235 amino acids 222 amino
acids
15
1.5 1 0.5 0 0.5 1 1.5
Mass (kDa) 39 28.5 29
Absolute sputum neutrophil counts (106/ml)
Substrate Val Xaa  Ala Aromatic acids Same as NE,
FIG. 14.1 The relationship between BMI and absolute sputum neutrophil specificity Xaa in P1 position accepts basic
counts in patients with moderate to severe stable COPD and chronic amino acids in
bronchitis. The relationship between absolute sputum neutrophil counts P1 position
and BMI in 12 patients with stable moderate to severe COPD according
to GOLD criteria and chronic bronchitis. Neutrophil counts were obtained Elastolytic 20% potency 40% potency
from spontaneous sputum samples produced over 4 h from waking; activity of NE of NE
r 0.8, p 0.01.

TABLE 14.2 Endogenous inhibitors of neutrophil proteinases.

Inhibitor Source Target proteinase Inhibition

Alpha 1antitrypsin Produced in the liver NE, CG, PR3 Irreversible


Found in serum

Alpha 1 antichymotrypsin Liver and macrophages CG Reversible

Alpha 2 macroglobulin Liver and macrophages NE, CG, PR3, MMP-8, MMP-9 Irreversible

Secretory leukoproteinase inhibitor Mucosa NE, CG Reversible

Elafin Mucosa NE, PR3 Reversible

Monocyte-neutrophil elastase inhibitor Neutrophils/monocytes NE, CG, PR3 Irreversible

Tissue inhibitors of Many cell types including epithelial MMP-8 and MMP-9 Irreversible
metalloproteinases cells, fibroblasts, neutrophils, and
monocytes

174
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
appear at dierent stages of maturation (see Table 14.3). overwhelmed [27]. This mechanism prevents cell necrosis
The neutrophil dierentiates and matures within bone and the release of the remaining cellular content of protein-
marrow, developing from a bipotential progenitor cell, the ase and other mediators.
granulocyte-macrophage colony forming unit. In the rst Apoptosis is regulated cell death, which allows the
developmental stage, the cell divides and dierentiates elimination of unwanted or damaged cells. At the present
from myeloblasts to promyelocytes. During this stage the time, three caspase-dependent apoptosis pathways have been
azurophilic or primary granules are produced. These gran- described. The rst pathway is triggered in response to extra-
ules contain myeloperoxidase (MPO), antibacterial proteins cellular signals (the receptor-mediated extrinsic pathway),
(such as defensins, lysozyme, and azurocidin), and three mediated by the binding of tumor necrosis factor related
serine proteinases, NE, cathepsin G (CG), and proteinase 3 proteins (such as Fas ligand) to death receptors on the cell
(PR3). The proteinases are produced as preproenzymes, with surface. This results in the formation of the death inducing
gene expression stopping at the metamyelocyte stage [24] signaling complex (DISC), which activates a series of cas-
and are activated by a lysosomal cysteine proteinase, dipep- pases, leading to DNA fragmentation by DNAse [2832].
tidyl peptidase [25]. The specic or secondary granules are A second pathway, the mitochondrial intrinsic pathway,
formed as the cell enters the myelocytic stage. These gran- responds to stress signals by the release of cytochrome c from
ules contain lysozyme, lactoferrin, collagenase, and various mitochondria. Cytochrome c, apoptotic protease activating
membrane receptors. The small storage granules are formed factor-1 (Apaf-1), and caspase-9 activate caspase-3 leading
last, at the metamyelocyte stage, and contain gelatinase and to apoptosis [3335]. Finally, in the endoplasmic reticulum
cathepsin B and D. At maturation (which takes approxi- pathway, caspase-12 is activated in response to stress signals
mately 2 weeks) each neutrophil contains a full complement such as hypoxia [36, 37].
of proteins which provide the mechanisms of cell migration, Clearance following apoptosis has no inammatory
opsonophagocytosis, and a formidable arsenal against path- sequelae. However, if phagocytosis fails, apoptotic cells undergo
ogens. However, the neutrophil proteinases (especially NE) secondary necrosis with cell rupture, which is inammatory in
also have the capacity to be intensely destructive, degrad- nature and increases both the proinammatory proteins and
ing structural lung proteins (including elastin, collagen, and proteinase burden in the lung [38]. Figure 14.2 describes the
gelatin) and appear to be involved in the posttranslational appearance of neutrophils during induced lung inammation,
processing of enzymes, cytokines, and receptors [23]. Fully showing both activated and apoptotic cells, apoptotic cells
mature neutrophils leave the bone marrow in a nonactivated undergoing secondary necrosis, and neutrophils containing
state and have a half-life of 410 h before marginating phagosomes with neutrophilic cell remnants.
and entering tissue pools [26]. Once in tissue, neutrophils The increased numbers of neutrophils seen in the
are usually removed by apoptosis leading to their recogni- lungs of patients with COPD may accumulate because of
tion and phagocytosis by macrophages in the main and by an increased inux from peripheral blood or because of
other neutrophils when the macrophage clearance system is prolonged neutrophil survival. Exposure to cigarette smoke
TABLE 14.3 Enzymes and other constituents of human neutrophil granules.

Granules

Constituents Azurophil Specific Small storage

Antimicrobial Myeloperoxidase Lysozyme


Lysozyme Lactoferrin
Definsins
Bacterial permeability-
increasing protein (BPI)

Neutral proteinases Elastase Collagenase Gelatinase


Cathepsin G Complement activator Plasminogen activator
Proteinase 3 Phospholipase A2

Acid hydrolases Cathepsin D Cathepsin D


-d-Glucuronidase -Mannosidase -d-Glucuronidase -Mannosidase
Phospholipase A2

Cytoplasmic membrane CR3, CR4


receptors fMLP receptors
Laminin receptors

Others Chondroitin-4-sulphate Cytochrome b558 Cytochrome b558


Monocyte Chemotactic factor
Histaminase
Vitamin B12 binding protein

The enzymes and other constituents of human neutrophil granules.


fMLP: N-formylmethionyl-leucyl-phenylalanine

175
Asthma and COPD: Basic Mechanisms and Clinical Management

healthy controls despite patients with COPD having a sig-


nicantly higher percentage of neutrophils in the samples.
The authors suggested that the increased neutrophilic pres-
ence in COPD was due to an increased inux rather than
a reduction in apoptosis. Yoshikawa et al. [46] also found
higher numbers of neutrophils in induced sputum from
patients with COPD compared with healthy controls, but
described a reduced neutrophilic chemotactic response to
IL-8 and N-formyl methionyl-leucyl phenylalanine (fMLP)
in the COPD patients which worsened with increasing dis-
ease severity. In this study, the authors postulated that an
increased inux could not explain the higher numbers of
cells seen in COPD, and that increased survival should be
considered. This issue has yet to be resolved.

PROMIGRATORY STIMULI

Neutrophils migrate into the lung in response to soluble


mediators. Promigratory stimuli can be classied as nonchem-
otactic cytokines, chemotactic cytokines, or chemoattractants.
Cytokines comprise families of molecules includ-
ing interleukins, lymphokines, monokines, growth factors,
interferons, and chemokines. Two of the most important
proadhesive cytokines in COPD are tumor necrosis factor
FIG. 14.2 Neutrophils during an LPS-induced lung inflammationElectron
alpha (TNF-) and interleukin-1 (IL-1).
micrographs displaying neutrophils during an LPS-induced lung
inflammation. (A) Highly activated neutrophils are shown (N) (characterized
TNF- is produced by activated monocytes and
by phagosomes and/or cytoplasmatic protrusions), lying amongst macrophages (but also epithelium, endothelium, and prob-
apoptotic neutrophils (black arrow) and cell debris (black arrowhead). ably smooth muscle cells) and has been implicated in the
(B) Secondary necrosis (characterized by membrane rupture of cells with pathogenesis of COPD [47, 48]. Increased levels of TNF-
an otherwise apoptotic morphology) of neutrophils is shown. Neutrophils have been measured in serum, sputum [49], and in bron-
containing large phagosomes (asterisks) enclosing neutrophilic cell choalveolar lavage samples from patients with COPD, and
remnants, such as apoptotic nuclei and neutrophil granulae, are shown in in smokers demonstrating a dose-dependent relationship
images (CE). Used with kind permission from Rydell-Tormanen et al., Respir with cigarette exposure, [50] with further increases during
Res 7: 143, 2006. exacerbations [51]. TNF- has also been associated with
the systemic manifestations of COPD, including a low
BMI [52, 53] (perhaps via leptin, [54]) and abnormal rest-
ing energy expenditure [55]. TNF- overexpression (due
appears to stimulate neutrophil dierentiation and matu- to genetic polymorphism) has been linked to early COPD
ration, causing peripheral leukocytosis, [39, 40] which has development or rapid progression [56]. In support of this,
been found to correlate with the severity of airow obstruc- mouse models with an inducible TNF- gene construct
tion [41, 42]. This suggests that a peripheral neutrophilia have shown that overexpression of TNF- is associated
facilitates an increased neutrophil transmigration into the with the development of emphysema associated with a gen-
lung, leading to a higher proteinase burden with subsequent eral increase in lung inammation [57] probably by induc-
tissue damage. Platz and colleagues [43] described a reduc- ing MMP production [58]. Conversely TNF- receptor
tion of peripheral neutrophil apoptosis (based on Annexin knock-out mice demonstrate reduced smoking or elastase-
V-PE binding and nuclear morphology) during exacerba- induced emphysema in comparison with the wild type
tions of COPD compared with healthy controls, which [59, 60] suggesting a central role for this cytokine.
returned to control levels upon resolution of symptoms. IL-1 is also produced by macrophages (although
This increased-cell longevity would facilitate increased neu- neutrophils and epithelial cells produce the cytokine) and
trophil egression into lung, again predisposing to enhanced increased levels have been found in sputum of patients with
lung damage. Once in the lungs, it has been suggested that stable COPD, which increase further during exacerbations
reduced neutrophil apoptosis may account for the high [61]. Furthermore, IL-1 production is enhanced by cells
numbers seen in the lung and the increased inammation cultured from smokers with COPD following cigarette
described in COPD although recent studies present con- smoke exposure compared with controls [62] and there is
icting results [44]. Rytila et al. [45] found no dierences in some evidence that overexpression of IL-1 (caused by pol-
the proportion of apoptotic neutrophils in induced sputum, ymorphisms such as the 511 SNP with a cytosine/thymine
or in the in vitro anti-apoptotic activity detected in the spu- transition) may increase susceptibility to COPD [63, 64].
tum uid phase in patients with COPD, healthy smokers or In a study of patients with severe COPD, data from our

176
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
(A) 4 (B) 0.7
3 0.9

IL-1 in plasma (pM log)


Sputum IL-1 (pM log)

2 1.1
1
1.3
0
1.5
1
1.7
2
3 1.9

4 2.1
15 17 19 21 23 25 27 29 31 20 25 30 35 40 45 50 55
BMI (kg/m2) FEV1 percent predicted

FIG. 14.3 The relationship between IL-1 and disease severity in COPD. Patients had moderate to severe COPD (as defined by GOLD) and chronic bronchitis.
Each data point represents the average of 11 measurements of IL-1 and a single BMI (A) or (B) FEV1 for one patient over 1 month. Spontaneous sputum
samples were collected over a 4 h period from waking. The regression lines are drawn and the correlation co-efficient (r) and significance (p) are as follows:
IL-1 and BMI r 0.7, p 0.01; IL-1 and FEV1 r 0.56, p 0.02.

group have shown a negative correlation between plasma epithelial and endothelial cells, leading to neutrophil adhe-
IL-1 and FEV1 and sputum sol IL-1 and BMI, clearly sion, chemotaxis, and degranulation.
linking IL-1 with COPD disease severity (see Fig. 14.3). LTB4 is mainly produced by monocytes, alveolar mac-
Furthermore, animal studies using inducible IL-1 systems rophages, and activated neutrophils and its production is
and complex knock-out mice have described a neutrophilic upregulated by a number of inammatory mediators includ-
inltrate, distal airspace enlargement, increased thickness of ing C5a, IL-1, TNF-, granulocyte-macrophage colony
the conducting airways, and enhanced mucin production stimulating factor (GM-CSF), PAF, NE, and even LTB4
when IL-1 was overexpressed [65]. itself [74, 75]. LTB4 enhances neutrophil aggregation and
TNF- and IL-1 are not directly chemotactic, but chemotaxis via two neutrophil surface receptors. A low an-
they act by increasing expression of integrins on the neu- ity receptor induces degranulation and increases oxidative
trophil surface and increasing endothelial expression of metabolism whereas a high anity receptor induces aggre-
selectins and intercellular adhesion molecule 1 (ICAM-1), gation, chemokinesis, and adhesion via the integrin Mac-1
aiding capture and rm adhesion of neutrophils to the vas- [76]. LTB4 may also activate endothelial cell monolayers
cular endothelium (see later). They also increase the pro- in vitro enhancing neutrophil emigration [77]. LTB4 con-
duction of directly chemotactic mediators from endothelial centrations are elevated in sputum [78] and exhaled breath
cells to enhance the migration process. condensate [79] from patients with COPD. Concentrations
Neutrophils possess at least ve receptors for chemo- correlate with the degree of airway neutrophilia [80] and
tactic stimuli and a number of chemotactic mediators have increase further during bacterial exacerbations [10] return-
been implicated in the pathogenesis of COPD, especially ing to baseline once bacteria have been successfully eradi-
the chemokine CXCL8 and the neutrophil chemoattract- cated [81]. In addition, LTB4 concentrations also correlate
ant LTB4. with the subsequent decline in gas transfer implicating neu-
CXCL8 is primarily produced by leukocytes (mono- trophilic inltration [12].
cytes, T-cells, neutrophils, and natural killer cells) and airway In vitro chemotaxis studies, using functional anti-
epithelial cells. Production is not constitutive but is induced bodies against CXCL8 and an LTB4 receptor antagonist,
by proinammatory cytokines, such as IL-1 and TNF- have demonstrated that 30% of the chemotactic activity of
[66], bacteria and bacterial products, [67, 68] viruses, such sputum can be accounted for by CXCL8, and up to 50%
as adenovirus and rhinovirus [69, 70], and oxidants, for by LTB4 [82, 83]. Furthermore, Woolhouse and colleagues
example, from cigarette smoke [71]. Once secreted, CXCL8 [82] found that LTB4 concentrations correlated with the
binds to CXC receptors (1 and 2) on leukocytes result- overall chemotactic activity of sputum, suggesting that
ing in activation of protein kinase B and GTPases, which LTB4 in particular may be central to the neutrophilic inux
lead to enhanced neutrophil adherence to endothelial cells seen in COPD, at least in the stable state.
(by increasing expression of 2-integrins) and directed cell It is unclear whether other inammatory mediators
migration. CXCL8 also activates Ras and eventually are important in the pathogenesis of COPD. At least ve
mitogen-activated protein kinases and extracellular signal- other chemokines mediate neutrophil responses in humans;
related kinases in neutrophils, causing degranulation. three forms of growth related oncogenes (GRO) (, ,
In COPD, sputum CXCL8 correlates with levels of and ), epithelial cell derived neutrophil activating pep-
neutrophil activation markers, such as MPO and NE [72], tide (ENA)-78, and neutrophil-activated peptide (NAP)-2.
and relates to airow obstruction, [73] oxygen saturation, GRO is detectable in bronchial secretions [84] and both
cigarette exposure [49], and progression of emphysema GRO and ENA-78 have been measured in BALF [85].
quantied by CT scan [12]. It has been suggested that oxi- One study has suggested that chemotaxis toward GRO
dative stress (caused by cigarette smoke and bacterial and and NAP2 is increased in COPD compared with healthy
viral infections) induces CXCL8 production in both airway controls and smokers, [86] but their contribution to the

177
Asthma and COPD: Basic Mechanisms and Clinical Management

pathogenesis of COPD is unknown. A number of other along the vessel wall is a normal feature of circulating neu-
inammatory mediators including C5a, fragments of brin, trophils and is due to reversible binding of transmembrane
elastin and collagen, 1-AT polymers, and bacterial products glycoprotein adhesive molecules called selectins, which are
such as fMLP, are also neutrophil chemoattractants found found both on neutrophils and endothelial cells [91].
in the lung, but their importance in neutrophil recruitment Leukocyte Selectin (L-Selectin) is constitutively
in COPD has yet to be established. The numbers of fMLP expressed on projecting microvilli on the surface of neu-
receptors is elevated in both healthy smokers and subjects trophils and binds an endothelial ligand which is believed to
with COPD who smoked, but not nonsmoking patients be a sialomucin oligosaccharide (potentially a fucosylated var-
with COPD [87] suggesting that fMLP may play a role in iant of CD34). L-Selectin-induced neutrophil capture can be
the recruitment of neutrophils leading to the development transient in noninamed tissue (stick and release), however,
of disease and its progression whilst smoking and especially during both nonpathogenic and pathogenic rolling interac-
during bacterial exacerbations (where bacterial formyl pep- tions, L-Selectin, once bound, is shed from neutrophils [92].
tides may be most important). Both L-Selectin binding and shedding is enhanced in the
The individual importance of each proinammatory presence of inammatory products such as TNF-, CXCL8,
or promigratory protein in COPD has yet to be unraveled, fMLP, and LPS [93]. Once bound and cleaved, L-Selectin
certainly many of these proteins appear to have similar molecules cannot be replaced and low expression has been
actions and a degree of overlap or redundancy is likely to associated with neutrophil apoptosis [94].
exist in the inammatory cascade. However, there is increas- In the presence of inammation, at least two further
ing interest in identifying the key inammatory mediators endothelial bound selectins are expressed,
in COPD as potential targets for therapeutic interventions. Platelet Selectin (P-Selectin) and Endothelial Selectin
To date, trials which have aimed to block a specic media- (E-Selectin): P-Selectin is stored intracellularly in Weibel-
tor have not proved helpful [88] including a recent trial of Palade bodies in endothelial cells [95] and can be mobi-
anti-TNF- [89]. It is likely that a greater understanding of lized rapidly in response to various inammatory mediators
each mediator, its naturally occurring antagonists and their including oxygen-free radicals, components of the comple-
relation to COPD is required to develop and clarify more ment cascade, and many cytokines [96]. Its neutrophilic
successful treatment strategies. However, Gompertz et al. counterligand is P-Selectin glycoprotein ligand-1 (PSGL1)
were able to show in a short phase-2 study that antagonism which is uniformly expressed on the surface of neutrophils.
of ve lipoxygenase activating proteins reduced LTB4 in the P-SelectinPSGL1 interactions occur after L-Selectinlig-
airway secretions and subsequent markers of neutrophilic and interactions, and have greater longevity. In the presence
inammation [90]. Although not dramatically eective, this of other adhesion molecules, P-Selectin/PSGL1 binding
may provide a model for future studies. slows neutrophil rolling velocities and eventually causes cell
tethering to the endothelium but in the absence of other
adhesive events, binding is also transient [97].
E-Selectin is not stored, and peak expression is seen
NEUTROPHIL MIGRATION 46 h after endothelial exposure to inammatory mediators
[98]. It binds to E-Selectin ligand 1 [99] and it is thought
to maintain neutrophil tethering after P-Selectin has been
Neutrophils are present at both the bronchial and alveolar downregulated. E-Selectin expression is increased in both
level in COPD, and therefore it is likely that neutrophil serum and BAL in patients with COPD and relates to lung
migration occurs from the bronchial and pulmonary circu- function [100] suggesting a common causality.
lation. Most of our understanding of neutrophil migration
has been derived from in vitro studies using systemic ves-
sels in the mesentery and dermis. Studies using samples of Firm adhesion
lung tissue suggests that while migration in the bronchial The next step in neutrophil migration is the transition from
circulation occurs in a similar fashion to migration in other reversible rolling to rm adhesion with the endothelium.
tissues, migration in the pulmonary capillaries may depend This is achieved by the sequential activation of neutrophil
on distinct mechanisms. receptors called integrins [101, 102].
The integrins are heterodimeric transmembrane
glycoproteins that comprise an and a subunit, which
Neutrophil migration in the together form an extracellular binding site. Integrins are
found on many haematopoietic cells, with diering and
bronchial circulation subunits. The two most important integrins in neutrophils
share a 2 subunit (CD18), and are called macrophage anti-
Initiation of migration gen 1 (MAC-1; CD11b/CD18) and lymphocyte-associated
In the bronchial circulation, neutrophils migrate from ves- function antigen 1 (LFA-1; CD11a/CD18). A third CD18
sel to tissue in a step-like process, dictated by the sequential integrin, p150,95 can also promote neutrophil tracking,
activation of adhesive proteins and their ligands on neu- but MAC-1 appears to be the most important integrin in
trophils and endothelial cells. models of neutrophil migration [103, 104].
Migration begins with the capture of neutrophils MAC-1 is stored in secretory granules [105, 106]
from owing blood which causes the cell to roll along the and is rapidly mobilized to the cell surface after exposure to
endothelial surface. Tethering and rolling of the neutrophil inammatory stimuli (including fMLP, TNF- and LPS).

178
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
Inammatory stimuli also promote transcription and trans- models have demonstrated that neutrophils can migrate
lation of the MAC-1 gene via a G-protein, Rho, [107] fur- up and down chemical gradients, responding to one sig-
ther increasing its surface expression [96]. Some MAC-1 is nal, migrating to its concentration peak and then migrat-
expressed constitutively on the neutrophil, but these proteins ing up a novel, more distant chemoattractant gradient, from
are incapable of binding ligands unless activated by intra or endothelium to tissue.
extracellular signals, where conformational changes occur Neutrophils can ignore a high concentration source
exposing a requisite binding epitope [108]. Neutrophils (due to receptor saturation, desensitization, and/or receptor
from patients with COPD have increased baseline surface sequestration) and preferentially respond to novel chemoat-
expression of MAC-1, which increases further following tractants [122]. Evaluations of chemotaxis by the under-
activation, [109] suggesting that this adhesion molecule agarose method has suggested that neutrophils are able to
may play a role in neutrophil migration in this disease. migrate in the direction of the vector sum of two or more
MAC-1 has a high anity for ICAM-1, an immu- diering signals [123]. If two sources of the same agonist
noglobulin-like protein that is expressed in low numbers on are used, migration toward the second attractant is poor,
the endothelial cell surface, but is rapidly induced in inam- [122] so two distinct agonists are required for precise tar-
mation [110]. Vascular cell adhesion molecule-1 (VCAM- geting. These ndings may account for the observation that
1) is also an immunoglobulin-like molecule expressed by activated cells characteristically secrete multiple induced
endothelial cells which it binds selectively to 41-integrin chemoattractants concurrently. Cells can regain their prior
[called very late antigen-4 (VLA-4)] on neutrophils [111]. sensitivity, but this process takes time, requiring recycling of
MAC-1/ICAM-1 interactions cause enhanced expression receptors. For example neutrophils preincubated with low
of both ICAM-1 and VCAM-1 on endothelial cells, sug- chemotactic levels of LTB4 showed a signicant reduction
gesting that both may be important in inammatory driven in chemotaxis to LTB4 which improved after 10 min [122].
neutrophil migration [112] and levels of ICAM-1 are raised Neutrophil movement itself is achieved by the for-
signicantly in COPD in contrast to asthma or healthy mation of locally protruding actin-rich pseudopods while
controls and correlate with overall pulmonary neutrophil retracting other regions of the cell body [124]. In order to
inltration [113]. achieve this, directed neutrophil chemotaxis begins with cell
polarization. Extracellular chemoattractant gradients causes
Migration the localized accumulation of PtdIns(3,4,5)P3 on the side
of the cell facing the highest chemoattractant concentra-
The nal step of neutrophil recruitment from the bron-
tion. PtdIns(3,4,5)P3 signaling guides the localized polym-
chial circulation to the lungs is transendothelial migration.
erization of F-actin (lamentous actin), which in turn leads
This occurs preferentially at tricellular junctions [114] and
to pseudopod extension, which confers cellular motility. The
depends upon activation of platelet endothelial cell adhe-
persistent presence of PtdIns(3,4,5)P3 at a particular site on
sion molecule (PECAM1) [115] which is distributed evenly
the plasma membrane causes cells to acquire an elongated
around the neutrophil and at intercellular junctions of
shape in which one F-actin enriched pseudopod becomes
endothelial cells. PECAM1 is thought to act as a homing
the leading edge of the migrating cell, whereas retraction
beacon that directs migration toward cellular junctions and
of pseudopods at the rear and sides of the cell is mediated
blocking PECAM1 on either neutrophils or endothelial cells
by cortical myosin II [125]. Although there may be many
using antibodies does not prevent adhesion but does prevent
proximal signal pathways by which polarization is regulated,
migration through the basement membrane both in vitro
the key event appears to be activation of the RhoGTPases,
and in vivo [116].
Rho and Rac via integrins [126]. The 41 integrin forms a
Once through the endothelial cell layer, leukocytes
complex with paxillin and GIT1 which inhibits Rac acti-
bind to matrix components such as collagen and laminin
vation at the sides and rear of the cell, but this inhibitory
via 1 integrins, with VLA-6 and -9 being perhaps the most
complex is impeded at the leading edge of the cell by phos-
important in allowing neutrophils to move through venule
phorylation of the 4 integrin cytoplasmic tail, which allows
basement membrane and lung tissue [117119]. Endothelial/
Rac activation [127]. At the front of the cell Rac activation
neutrophil PECAM1 interactions lead to increased neu-
leads to the F-actin polymerization while at the rear of the
trophil surface expression of VLA-6 (61) and VLA-6 facil-
itates passage thorough the basement membrane and beyond. cell Rho activation leads to assembly of myosin, with both
To support this, neutrophils from PECAM1 knock-out mice GTPases working in a co-operative manner to establish and
do not have the associated rise in VLA-6 which is seen in the stabilize cell polarity [128].
wild type [120]. Migration is accompanied by release of neu- Recent studies have elucidated how the neutrophil
trophil proteinases especially NE, [121] which may facilitate steers from one direction to another. Pseudopods are made
passage by matrix degradation, exposing laminin for VLA-6 in spatially restricted sites by splitting of the leading edge of
binding. the cell, and the generation of these protrusions appears to
be random in both their direction and timing. In a study
where cells were exposed to a promigratory stimulus, which
Direction of migration and neutrophil movement was then relocated once an accurate trajectory was estab-
Neutrophils migrating within the lung encounter multiple lished, pseudopod generation occurred randomly, but pseu-
chemoattractant signals in complex spatial and temporal dopods that extended toward the stimulus were more likely
patterns as endothelial, epithelial cells, and immune cells to be retained, while those which extended in inaccurate
respond to infection or injury. Individual chemoattractants directions were retracted (see Fig. 14.4). Therefore it appears
can vary in their ability to aect neutrophils and in vitro that neutrophils migrate up chemoattractant gradients by

179
Asthma and COPD: Basic Mechanisms and Clinical Management

that migrating cells can change their behavior depending


on the matrix and stimuli they are exposed to. For exam-
ple, in a noncross-linked collagen 3D matrix, cells migrate
along bers, but can squeeze between bers in the presence
of anti-proteinases, therefore migration is not inhibited but
redirected. On the other hand in a dense cross-linked col-
lagen 3D matrix, cells require proteinases to migrate and
migration is inhibited in the presence of anti-proteinases.
Cell-derived matrices are cross-linked but have gaps and
migrating cells appear to preferentially seek out these gaps
to migrate, which may or may not therefore require protei-
nases, depending on the size of the gap [142]. If neutrophil
migration in vivo is partially proteinase dependent, in vitro
FIG. 14.4 Neutrophil migration by pseudopod generation toward a migration may not require degradation of loose extracel-
promigratory stimulus. (A) A polarized (elongated) neutrophil migrating lular matrix by proteinases to allow cell passage, but instead
toward IL-8 in a Zigmond chamber. The direction of the chemoattractant the generation/activation of inammatory chemokines and
gradient is shown by the large arrow. (B) A representation of pseudopods cytokines or modulation of adhesion molecules (enhancing
generated over the time course. Here, pseudopod generation was noted expression, increasing their activation, or exposing binding
across the breadth of the leading edge of the cell in a random fashion. sites [117]) by proteinases may be sucient, and this may
(C) Pseudopods generated toward the stimulus were more likely to be more easily decreased or prevented by inhibitors, explain-
be retained (the sustained pseudopods over the time course is shown ing the apparent experimental contradictions.
schematically). (D) Pseudopods that extended in inaccurate directions
were more likely to be retracted (the retracted pseudpods over the time
course are shown schematically). It appears that neutrophils migrate up
chemoattractant gradients by choosing the best aligned of competing
randomly generated pseudopods.
Neutrophil migration in the
pulmonary circulation
The majority of neutrophils appear to enter the lung from
choosing the best aligned of competing randomly generated the pulmonary capillary network rather than the postcap-
pseudopods [129]. illary venules [143, 144]. Neutrophil emigration into the
lung from the pulmonary circulation is less well understood
Transmigration through extracellular matrix than from the bronchial circulation, however there seem to
Neutrophil proteinases are released during migration be important dierences in mechanism, probably due to the
through extracellular matrix [121] but it has been dicult size of capillary networks. 4060% of pulmonary capillary
to ascertain whether proteinases are necessary for neu- segments are narrower than a spherical neutrophil, which
trophil migration. Chemotaxis through articial substrates on average is 78 m in diameter [145, 146]), so neutrophil
in response to fMLP can be inhibited by 50% by 1-AT rolling is unlikely because of size constraints. Neutrophils
[130] and CG antibodies; synthetic inhibitors of CG and entering the pulmonary capillary network have to undergo
1-antichymotrypsin (1-ACT) also reduce neutrophil a shape change, from a sphere to an oblong, to allow pas-
migration [131]. Furthermore, fMLP-stimulated migration sage through these narrow vessels. This slows their transit
across an articial basement membrane is also reduced by time, and indeed, radiolabeled neutrophils from patients
inhibitors of both NE and MMP-9 [132]. However, in vitro with stable COPD have been shown to have a slower tran-
studies of endothelial monolayers and basement membrane sit time in the pulmonary circulation compared with red
matrices have shown consistently that proteinase inhibitors blood cells [147]. When activated, neutrophils become less
are ineective at stopping neutrophil migration [133, 134] deformable due to actin polymerization, and this slows their
although degradation of basement membrane components progression through the capillary network further [148].
is reduced [135]. In animal studies, neutrophils from mice In such circumstances there is no need for transient adhe-
whose genes for NE and CG had been knocked out sion via the selectins to initiate neutrophil rolling along the
showed normal migration both in vitro and in vivo when endothelium, as the leukocytes are already in close con-
exposed to LPS although pathogen clearance was impaired tact with the endothelium, and indeed there is evidence to
[136] and mice decient in gelatinase B had normal neu- suggest that some migration can occur without adhesion
trophilic migration into the lungs [137]. However, animal molecules [149, 150].
studies of cigarette smoke inhalation suggest that neutrophil In the bronchial circulation, adhesion and migration
inux into the lung is reduced in the presence of proteinase appear to be primarily dependent upon ICAM-1 interactions,
inhibitors [138, 139]. however, in the pulmonary circulation both CD18-dependent
Therefore the exact mechanisms remain unresolved, and -independent adhesion pathways have been described
but the conicting results may well reect the study model and the path utilized appears to be stimulus-specic. For
used, including the density of the ECM studied and in example, in animal models, bacteria such as Streptococcus
particular the presence or absence of cross-linked collagen pneumoniae and Staphylococcus aureus and hydrochloric acid
(compare, e.g. [140, 141]). The majority of recent stud- have induced CD18-independent neutrophil migration,
ies have used tumor cells, but there is evidence to suggest while human studies have demonstrated CD18-independent

180
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
migration toward host-derived chemoattractants such as adhesion molecule expression, and enhancement of the res-
LTB4, CXCL8, and sputum [150,151]. In contrast, IL-1, piratory burst [162, 163]. However, none of these options
phorbol myristate acetate, and gram-negative bacterial stimuli have been studied in COPD. Whatever the dierences in
including LPS elicit migration via pathways predominantly neutrophils, they cannot reect environmental factors (such
mediated by CD18 [152155] CD18-dependent migra- as prolonged cigarette smoke exposure) as only a proportion
tion may not only be stimulus-driven but also inammatory of smokers develop signicant airow obstruction indicating
mediator driven. Rabbits produce both CXCL8 and TNF- a genetic susceptibility [23]. In susceptible individuals, envi-
during a bacterial pneumonia but CXCL8 and TNF- pro- ronmental exposure may lead to increased neutrophil pool-
duction during a gram-negative pneumonia are 2- and 10- ing in the lung, increased degranulation and hence increased
fold greater respectively than seen during a gram-positive tissue damage; although it remains unclear whether current
pneumonia [156]. It may be the varying concentrations of observations reect a predisposing factor for COPD or a
these proinammatory stimuli which elicit a CD18-depend- consequence of the disease.
ent response, perhaps by inducing nuclear factor- [152]
rather than the bacterial insult itself. Animal models have
suggested that selectins are required where CD18-dependent
migration occurs, but not for CD18-independent migration.
For example, L-Selectin knock-out mice had signicantly THE EFFECTS OF NEUTROPHIL
less neutrophil recruitment into the lungs in response to LPS PROTEINASES IN COPD
(a CD18-dependent stimulus) while recruitment in response
to Streptococcus pneumoniae (a CD18-independent stimu-
lus) was unaected [157]. The actions of neutrophil elastase
A signicant physiological role of NE is bacterial killing.
Migration in COPD This is achieved when opsonized bacteria are ingested in
phagosomes which fuse with lysozymes containing protein-
Current evidence suggests that the increase in neutrophils ases and oxidants. NE is also intensely destructive and this is
in the lungs is not due to delayed apoptosis and it has been believed to be the primary cause of lung damage seen both
hypothesized that neutrophils in COPD reect an enhanced in emphysema and in chronic bronchitis. NE was the rst
chemotactic response. Certainly, neutrophils from patients of the serine proteinases to be shown to produce emphy-
with chronic bronchitis and emphysema demonstrate sema in animal models. Intratracheal instillation of puried
enhanced migration toward common chemoattractants in vitro NE-induced emphysema in dogs and hamsters [164166]
compared with healthy controls [158]. and intratracheal instillation of other elastases have given
Unfortunately, studies examining the expression similar results in various animal modelsPapain (plant
of adhesion molecules have produced conicting results. elastase) in rats, [167] neutrophil lysates in dogs [168], and
Noguera [109] measured MAC-1, LFA-1, and L-Selectin porcine pancreatic elastase in rats and hamsters [169, 170].
expression on neutrophils from controls and patients with In fact the development of emphysema is specic to elastase
COPD, prior and poststimulation with TNF-. Neutrophils activity with emphysema severity relating proportionally to
from patients with COPD had enhanced expression of the elastolytic potency of the elastase used [169, 170] and
these adhesion molecules compared with controls, and dif- in these models emphysema can be prevented by specic
ferences were even more pronounced following stimulation elastase inhibitors [171, 172].
with chemoattractants. Woolhouse et al. [159] demonstrated NE acts upon a wide range of proteins and can degrade
upregulation of CD11b (a component of MAC-1) on neu- elastin, bronectin, and collagen, [173, 174] and can also
trophils from smokers with COPD compared with controls. decrease the activity of immunoglobulins and activate com-
However, Gonzalez [160] found no dierences between lev- ponents of the complement cascade [175, 176]. NE may also
els of adhesion molecules in smokers with and without air- aect wound healing, by its actions on transforming growth
ow obstruction and Yoshikawa et al. [161] found reduced factor and the epithelins [177,178]. During activation,
chemotaxis toward sputum sol in patients with COPD azurophil granule proteinases (including NE) are expressed
compared with healthy controls. on the neutrophil membrane [179] and in vitro, over 95%
The increased neutrophil migration and degranula- remains associated with the cell by a charge-dependent mech-
tion seen in COPD has led to a belief that neutrophils anism, while less than 5% is released into the liquid mileu or
may be dierent from those in health. The dierences may directly onto tissue. Thus NE most commonly causes damage
be accounted for by genetic polymorphisms and be appar- by close contact between cells and matrix [180, 181]. Recent
ent during maturation in bone marrow. Alternatively neu- elegant studies have shown that NE polarizes toward the
trophils could be primed following their release into the leading edge of the neutrophil as it migrates. Some is then
circulation, perhaps by inammatory cytokines, so that they left behind as the cell moves on where it may cause collateral
are more responsive, with an increased ability to degranu- damage for instance to the connective tissue [182].
late compared with those of healthy individuals. This prim- Free NE activity has been detected in secretions of
ing may also occur during transmigration and studies have patients with COPD [72] and this is felt to be fundamen-
conrmed dierences between neutrophils prior to and fol- tal in the development of the condition in vivo. Free NE
lowing migration even in healthy controls with increased may accumulate from degranulating neutrophils, or (in con-
expression of proteinases on the cell surface, increased trast with apoptotic cells) may be freely released during cell

181
Asthma and COPD: Basic Mechanisms and Clinical Management

necrosis [183]. Also the process of phagocytosis may cause lung. This inammatory response is greater in patients with
the release of signicant quantities of proteinases into the 1-AT deciency who have higher levels of LTB4 and
media (sloppy eating), especially during frustrated phago- elastase providing an amplication to neutrophilic recruitment
cytosis, when cells attempt to ingest large particles [184]. and subsequent increase in the potential for tissue destruction
Free NE can also be released from activated macrophages, compared with patients with normal anti-proteinase function
which scavenge the proteinase from apoptotic neutrophils thereby explaining the more severe and rapidly progressive
via endocytosis and subsequently release it during the rst disease of 1-AT deciency patients [78].
24 h of their own inammatory response [185]. This is The most important risk factor for COPD is ciga-
important, as although cell-associated proteinases have par- rette smoke exposure and the relationships between smoke
tial resistance to native inhibitors such as 1-AT, [180] free inhalation and NE have been studied. In animal models
NE is more readily inactivated by both serum- and tissue- NE knock-out mice are partially protected (45%) against
based inhibitors [186] if sucient quantities are present, the development of emphysema, [202] however, these mod-
which means that in health, free NE should be completely els are limited, as CG and PR3 persist, and CG has been
inactivated at a short distance from the activated cell. shown to cause secretory cell metaplasia [166] whereas PR3
Indeed, it has been shown that the concentration of causes both emphysema and secretory cell metaplasia [203].
free NE released from neutrophils falls exponentially away Furthermore, animal models have suggested that both syn-
from the cell [187]. In healthy subjects, the serum concen- thetic [171] and natural NE inhibitors [172, 204, 205] can
tration of 1-AT is 30Mol, which is at least two orders of limit emphysema when delivered simultaneously with the
magnitude lower than NE concentrations in the neutrophil elastase insult. Most animal models suggest that neutrophil
granule, and inhibits NE on a one to one molar basis. inux is greatest during the early stages of lung damage
Therefore following degranulation NE cannot be completely with macrophage inux, and their metalloproteinases accu-
inhibited until it has diused far enough to reduce its con- mulating at a later stage [206, 207]. However, even when
centration to 30Mol. This phenomenon is called quantum given in established disease, NE inhibitors still limit inam-
proteolysis [187] and was clearly demonstrated in a series mation and connective tissue breakdown [138].
of experiments using serum from patients with normal or COPD is characterized by periods of stability of symp-
decient 1-AT where an area of obligate enzyme activity toms punctuated with exacerbations [which are intermittent
existed even with serum from healthy subjects but was far worsening of symptoms and (most probably) of the inam-
greater using serum from patients with 1-AT deciency matory load [208]]. NE activity relates to sputum purulence,
[188]. Membrane bound NE is less susceptible to anti- which is due to MPO which can be graded visually [10].
proteinases [180] and the combination of increased free and During bacterial exacerbations of COPD, sputum purulence,
membrane bound NE present in COPD may be sucient neutrophil inux, NE activity, and tissue degradation prod-
to overcome local inhibitors, causing the tissue destruction ucts increase [10, 209, 210] and therefore it is likely that pro-
which is characteristic of the disease. teinase-induced tissue damage also increases. More frequent
NE interacts with matrix proteins and cells, and this exacerbations are associated with a faster decline in lung func-
aects not only its own activity, but also the ecacy of its tion as well as increased morbidity and mortality and it may
inhibitors and other proteinases. For example, although free be that the increases in NE load and activity during these
NE is irreversibly inhibited by 1-AT, elastin bound NE is periods causes progressive lung damage which is reected in
poorly inhibited by 1-AT, while the inhibitory eect of SLPI markers of disease severity [208]. Table 14.4 summarizes the
is unaected [189]. Adhesion to goblet cells appears to alter actions of NE in COPD.
the neutrophil membrane, enhancing the release of mem-
brane bound NE into the intercellular space [190] potentially
causing mucus secretion. Furthermore, animal models have Matrix metalloproteinases
demonstrated that NE can induce secretory cell metaplasia
which is prevented by NE-specic inhibitors [190192]. It There have been signicant advances in our understanding
has been suggested that mucous gland epidermal growth fac- of the role of matrix metalloproteinases (MMPs) in COPD.
tor receptor interacts with NE as part of a signaling cascade Both neutrophils and macrophages produce large amounts
[193]. In human studies, there is a clear relationship between of MMPs and their inhibitors, the tissue inhibitors of metal-
the amount of mucus production and the concentration of loproteinases (TIMPs). The MMPs are proteolytic enzymes
active NE in the lung secretions. that are secreted as proenzymes (activated by other MMPs
NE can damage the respiratory epithelium in vitro and NE) and remain bound to cell membranes. MMPs
[194], reduce ciliary beating [195], and trigger a state of not only degrade matrix proteins, but also inactivate anti-
oxidative stress in cells [196] all of which are abrogated by proteinases such as 1-AT and 1-ACT, activate enzymes
NE inhibitors. NE can also induce apoptosis of epithelial involved in the clotting cascade, and interact with cytokines
cells [197] and detachment of bronchial epithelial cells and adhesion molecules, so their potential role in the patho-
from the extracellular matrix [198] and both PR3 and NE genesis of COPD is complex and wide ranging.
induce detachment and apoptosis of endothelial cells [199] Over 24 mammalian MMPs have been described,
which has also been implicated in the pathogenesis of and although they are primarily grouped according to
COPD [200]. the proteins they degrade (collagenases, gelatinases, etc.),
NE stimulates the release of LTB4 by macrophages most can degrade many substrates and together they can
[201] and may cause release of CXCL8 from bronchial epi- degrade all extracellular substrates. MMPs are inhibited by
thelial cells, which enhances neutrophil migration into the 2-macroglobulin and by the four TIMPs described to date

182
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
TABLE 14.4 The actions of neutrophil elastase in COPD COPD [137]. In a further study, MMP-9 knock-out mice
displayed greater neutrophil inux than the wild type, per-
Bacterial killing Intracellular: engulfed organisms in
haps because MMP-9 can degrade neutrophil chemoattract-
phagosome
ants [191]. However smoke-exposed guinea pigs displayed a
Extracellular: targeting and cleaving bacterial reduction in the severity of emphysema and MMP-9 activ-
virulence factors in released granule proteins ity in BALF after introduction of a broad spectrum MMP
inhibitor, although this may also have reected inhibition
Degradation ECM components
of other metalloproteinases [219]. Genetic polymorphisms
Cystatin C of MMP-9 have, however, been identied which cause
TIMPs enhanced protein expression, and in a Japanese study, poly-
T-Lymphocyte surface antigen morphism 1562C/T has been associated with an increased
risk of smoking-induced emphysema [220] although this
Activation MMP-2, MMP-3, MMP-9, Cathepsin B has yet to be replicated in other populations [221].
MMP-8 has been less studied in COPD compared
Modification of Enhances epithelial secretion of CXCL8
with MMP-9, however one study described both raised con-
inflammatory Enhances macrophage secretion of LTB4 centrations and increased activity of MMP-8 in the induced
mediators
Inhibits cellular response to TNFRII prolonging sputum of patients with COPD compared with smokers
half-life of TNF without evidence of COPD. Furthermore both the levels
Increases SLPI expression but reduces
and activity of MMP-8 correlated inversely with FEV1 and
secretion of SLPI
positively with sputum neutrophil counts [222]. There are
currently no animal studies to elucidate the importance of
Increases elafin expression MMP-8 in COPD.
Increases alpha 1 antitrypsin expression by
monocytes and alveolar macrophages

Cell migration NE/alpha 1 antitrypsin complexes are


chemotactic for neutrophils
INTERACTIONS BETWEEN
Modification of ICAM-1 expression enhancing
MMPS AND NE IN COPD
adhesion
It is likely that the serine proteinases and MMPs act syn-
Cell apoptosis Increases epithelial and endothelial cell
ergistically in lung disease. NE degrades TIMPs [223]
apoptosis
facilitating MMP activity, and can activate several MMPs
Cell function Disruption and detachment of epithelial cells converting proenzymes, including MMP-9, to the active
form [224]. Conversely MMP-12, for instance, inactivates
Reduces ciliary beating of columnar
1-AT thereby enhancing NE activity [225]. The major-
epithelium
ity of animal knock out models of emphysema support a
Enhances oxidative stress multifaceted pathogenic process in the disease, as inhibi-
Increases mucin MUC5AC protein content tion of serine, cysteine, or MMPs all show partial protec-
Increases bacterial adherence and tion from the development of emphysema [138, 191]. The
colonization noticeable exception is MMP-12 and there have been sev-
eral conicting animal studies comparing MMP-12 activity,
This table is not exhaustive and references are not included for reasons of space. For macrophage and neutrophil inux, and emphysema. MMP-
an excellent review of the cell signaling by neutrophil proteinases see Ref. [211]. 12 knock-out mice (Line 129 mice) exposed to cigarette
smoke did not develop emphysema and lung inammation
was reduced, although macrophage recruitment to the lungs
[212]. The main MMPs secreted by neutrophils are MMP-9 was normal in response to monocyte chemoattractant pro-
(Gelatinase B), which degrades collagen, elastin and gelatin; tein-1 (MCP-1). All elastolytic activities in the wild-type
and MMP-8 (neutrophil collagenase) which degrades col- mice were derived from MMP-12 and emphysema sever-
lagen types IIII. ity was increased following instillation of MCP-1 suggest-
MMP-9 is increased in lung tissue, BALF, and plasma ing a very specic mechanistic pathway [226]. In a study
taken from patients with COPD, [213, 214] and levels are of Sprague-Dawley rats, neutrophil numbers increased in
negatively correlated with airow obstruction and relate to the rst month following smoke exposure, but breakdown
the number of sputum neutrophils [215, 216]. MMP-9 not of connective tissue and the development of emphysema
only acts as a proteinase, but also modies cellular func- occurred later and related to a subsequent increase in mac-
tions by regulating cytokines and matrix bound growth fac- rophages [206]. Furthermore, neutrophil depletion (with a
tors and therefore may have a role in lung remodeling after signicant reduction in NE activity) did not protect against
the inammatory insult resolves [217, 218]. MMP-9 de- smoke-induced emphysema, while macrophage depletion
cient mice exposed to intratracheal LPS showed no dier- (with normal neutrophil activity) did. These results have
ences in histological tissue damage, neutrophil migration, questioned the importance of the neutrophil in the patho-
or inltration compared to the wild-type mouse, suggest- genesis of COPD (at least in rodents) and have suggested
ing a limited role for this MMP in the pathogenesis of that the macrophage was the only eector cell.

183
Asthma and COPD: Basic Mechanisms and Clinical Management

However, in a further study BALF from cigarette seen following cigarette smoke in mouse models, probably
smoke-exposed mice showed an acute inux of neutrophils by enhancing neutrophil migration into the lung [227].
with evidence of both elastin and collagen degradation in a In order to understand the relationship between mac-
dose-dependent manner at 6 and 24 h. This had resolved by rophages and neutrophils in COPD, MMP-12 knock-out
48 h and was reduced by neutrophil depletion or the admin- mice were studied further. Cigarette smoke exposed MMP-
istration of the serine proteinase inhibitor 1-AT [207] 12 knock-out mice did not display the early neutrophilic
although macrophage numbers were not aected. Other inltrate or the release of desmosine and hydroxyproline
animal models have also found an early rise in neutrophil (matrix breakdown products) that is characteristic of wild-
number and activity, and a corresponding rise in matrix type mice, although low levels of macrophage inltration
breakdown products after exposure to cigarette smoke [138]. did occur. When the MMP-12 knock-out mice under-
In guinea pigs, NE inhibitors reduced TNF- and emphy- went intratracheal instillation of normal macrophages, a
sema by approximately 30%, but only if given at the start of neutrophil inux was seen and the use of an MMP inhibi-
exposure and when inhibitors were given after 4 months of tor also prevented a neutrophilic inltrate and subsequent
exposure, these eects were not seen. matrix breakdown [211]. Following these observations,
TNF- receptor knock-out mice were also protected studies demonstrated that cigarette smoke-induced pro-
from the acute neutrophil inltrate and connective tissue duction of TNF- from alveolar macrophages in wild-type
breakdown seen following cigarette smoke exposure in wild- mice, but not in MMP-12 knock-out mice while levels of
type mice [211] and mice that produce lower levels of TNF- TNF- mRNA were the same in both groups. It was sur-
following an inammatory insult (Line 129 mice) also mised that MMP-12 processes TNF- after secretion and
appeared to have less neutrophils and less emphysema com- it is likely, therefore, that MMP-12 is needed to activate
pared with the wild-type mice [197, 211]. In fact TNF- TNF- which in turn initiates neutrophil recruitment, lead-
appears to be central to about 70% of the tissue destruction ing to degranulation and tissue damage [228]. Figure 14.5

Degradation of
extracellular matrix

1 Release of chemotactic
peptides

2 MMP-12 5 NE

Macrophage Neutrophil
4 Release of chemotactic
3 TNF- cytokines, increased levels
of E-Selectin

6 Activation of proMMP-12 to MMP 12.


Inactivation of TIMP
7 MMP-12 inactivates 1 anti-trypsin

8 Macrophages scavenge free NE

FIG. 14.5 The proinflammatory actions of MMP-12 and the interactions between neutrophils and macrophages
1 Cigarette smoke causes the release of chemotactic peptides which recruit macrophages and neutrophils to airways.
2 Activated macrophages secrete MMP-12 which activates epithelial cells and causes degradation of extracellular matrix components.
3 MMP-12 facilitates the release of active TNF-a which activates epithelial cells further.
4 Activated epithelial cells release chemotactic cytokines and increase the expression of proadhesive proteins such as E-Selectin.
5 Neutrophils migrate to areas of activated epithelium and release free NE during frustrated phagocytosis or sloppy eating. NE promotes epithelial cell
apoptosis and increases expression of proinflammatory mediators such as LTB4 and CXCL8.
6 NE proteolytically converts proMMP-12 to active MMP-12 and proteolytically inactivates TIMP, both of which enhance MMP-12 activity.
7 MMP-12 proteolytic inactivation of a1-AT causes the further release of promigratory proteins and allows increased NE activity.
8 Macrophages scavenge free NE which is then reused to further degrade extracellular matrix components.

184
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
summarizes the proinammatory properties of MMP-12 Whatever the stimulus, once present, neutrophils have
and macrophage/neutrophil interactions. the potential to cause the pathological features seen by the
When interpreting results from in vitro work and ani- release of proteinases and reactive oxygen species, as described
mal models, it is crucial to consider that there may be impor- previously. In particular there is destruction of the epithelial cell
tant variations in the pathogenesis of COPD between cell layer [194], which is often a feature of the airways in asthma
types and diering species, and indeed, conicting results are [249]. Indeed recent studies have conrmed the importance of
common. However it seems likely that neutrophil inltration proteinase function in both the acute inammatory response
is an early event and that both neutrophil numbers and elas- and the subsequent mucous cell metaplasia that accompanies
tolytic activity relate well to lung damage in the early stages postinfectious murine models of asthma, [24] although these
of disease. Macrophages seem central to neutrophil recruit- ndings have yet to be conrmed in humans. Further work is
ment, probably via activation of TNF- by metalloprotein- required, but current evidence seems to support the premise
ases, and may be needed to sustain the inammatory process that while eosinophils may be dominant in the early stages of
and hence development of emphysema. disease in atopic individuals, neutrophils form a key feature of
more severe, nonatopic asthma [250].

THE NEUTROPHIL IN ASTHMA


CONCLUSIONS
Although current concepts of asthma pathogenesis focus
primarily on CD4 T cells and eosinophilic inammation, There is strong evidence supporting the belief that the neu-
there are robust data suggesting that the neutrophil may also trophil is central to the pathogenesis of COPD. The neu-
be important in the pathophysiology of asthma. Absolute trophil is the only cell that has been shown to be able to
neutrophil counts appear to be raised in a subset of patients cause all of the pathological changes of the disease includ-
with asthma, and indeed, in one study up to 60% of patients ing emphysema, mucus hypersecretion, epithelial destruc-
studied had non-eosinophilic airway inammation present in tion, and reduced ciliary beating. In vitro models, animal
induced sputum samples [229]. Measures of chronic asthma models, and human studies have led to conicting results
severity, such as FEV1, correlate with the degree of airway regarding the importance of various components of the
neutrophilia in stable disease [230233]. Furthermore, neu- inammatory response. However, based upon current evi-
trophilic inammation has been clearly documented in acute dence it appears likely that the neutrophil and in particular,
exacerbations of asthma, in status asthmaticus, and in noc- NE is associated with at least the initial lung damage seen
turnal asthma [234236] and has been reported as the pre- in COPD although macrophages appear necessary either to
dominant cell type in fatal asthma [237, 238]. These ndings initiate the acute neutrophilic response (via TNF-) and/or
cannot be explained by patients smoking status, co-morbid- sustain the subsequent inammatory response causing ini-
ity, or the presence of acute infection alone, suggesting that tiation and progression of emphysema. Studies so far have
neutrophils may well have an important role in this disease not provided a means to study the eects of other complex
[239]. Finally, although eosinophilic inammation is sensi- processes, such as the recurrent exacerbations in COPD, or
tive to corticosteroid treatment, neutrophils respond poorly the eect of smoking cessation and to clarify our under-
to this treatment modality, and may be the underlying cause standing of the overlapping actions of proteinases. Detailed
of steroid resistant cases of asthma [240, 241]. studies of cells and chemoattractants in man will remain
It has been proposed that neutrophils enter the air- crucial in dissecting the mechanisms. Although studied in
ways in response to infection. In murine models, pulmonary much less detail, it is likely that neutrophils also play a role
infection with mycoplasma or Sendai virus is associated with in asthma, with the involvement of similar pathways.
a neutrophilic inltrate. This leads to a chronic (and in most
cases a permanent) airway hyperresponsiveness that per-
sists long after the infection has resolved [242, 243]. In vivo References
there is serologic and polymerase chain reaction derived evi-
dence associating chronic asthma with airway infection with 1. Rutgers SR, Timens W, Kaufmann HF, van der Mark TW, Koeter GH,
both viruses and atypical bacteria (which have in turn been Postma DS. Comparison of induced sputum with bronchial wash, bron-
associated with increased neutrophil counts) [244, 245]. choalveolar lavage and bronchial biopsies in COPD. Eur Respir J 15:
However, even if infection initiates neutrophil inux into 10915, 2000.
the lungs, treatment of the infection has not been associated 2. Stanescu D, Sanna A, Veriter C, Kostianev S, Calcagni PG, Fabbri LM
with a reduction of symptoms, suggesting that on-going et al. Airways obstruction, chronic expectoration and rapid decline in
infection may not be sustaining the neutrophilic features of FEV1 in smokers are associated with increased levels of sputum neu-
trophils. Thorax 51: 26771, 1996.
the disease process.
3. Martin TR, Raghu G, Maunder RJ, Springmeyer SC. The eects of
Asthmatic epithelial cells constitutively hypersecrete chronic bronchitis and chronic air ow obstruction on lung cell popula-
both IL-8 and IL-17 (both of which are chemotactic for tions recovered by bronchoalveolar lavage. Am Rev Respir Dis 132: 260,
neutrophils), [246, 247] and it has been suggested that these 1985.
cells are abnormal in asthma, both when quiescent and active, 4. Thompson AB, Daughton D, Robbins RA et al. Intraluminal airway
driving inammation by the release of proinammatory inammation in chronic bronchitis: Characterisation and correlation
mediators [248]. This theory has yet to be fully explored. with clinical parameters. Am Rev Respir Dis 140: 152737, 1989.

185
Asthma and COPD: Basic Mechanisms and Clinical Management

5. Lacoste JY, Bousquet J, Chanez P et al. Eosinophilic and neutrophilic development of acute experimental arthritis. J Clin Invest 109: 36371,
inammation in asthma, chronic bronchitis and chronic obstructive 2002.
pulmonary disease. J Allergy Clin Immunol 92: 53748, 1993. 26. Burnett D. Neutrophils. In: Stockley RA (ed.), Pulmonary Defenses,
6. Pilette C, Colinet B, Kiss R, Andre S, Kaltner H, Gabius HJ, Delos M, pp. 11326. WileyEurope Chichester, 1997.
Vaerman JP, Decramer M, Sibelle Y. Increased galectin 3 expression and 27. Rydell-Tormanen K, Uller L, Erjefalt JS. Neutrophil cannibalism a
intra-epithelial neutrophils in small airways in severe chronic obstruc- back up when the macrophage clearance system is insucient. Respir
tive pulmonary disease. Eur Respir J 45, 2007, epub ahead of print. Res 14(7): 143, 2006.
7. Balbi B, Bason C, Balleari E et al. Increased bronchoalveolar granu- 28. Muzio M, Stockwell BR, Stennicke HR, Salvesen GS, Dixit VM. An
locytes and granulocyte/macrophage colony stimulating factor during induced proximity model for caspase-8 activation. J Biol Chem 273:
exacerbations of chronic bronchitis. Eur Respir J 10: 84650, 1997. 292226, 1998.
8. Tsoumakidou M, Tzanakis N, Chrysofakis G, Kyriakou D, Siafakas 29. Muzio M, Chinnaiyan AM, Kischkel FC, ORourke K, Shevchenko A,
NM. Changes in sputum T-lymphocyte subpopulations at the onset of Ni J, Scadi C, Bretz JD, Zhang M, Gentz R, Mann M, Krammer
severe exacerbations of chronic obstructive pulmonary disease. Respir PH, Peter ME, Dixit VM. FLICE, a novel FADD-homologous ICE/
Med 99(5): 57279, 2005. CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death
9. Selby C, Drost E, Lannan S et al. Neutrophil retention in the lungs inducing signaling complex. Cell 85: 81727, 1996.
of patients with chronic obstructive pulmonary disease. Am Rev Respir 30. Scadi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ,
Dis 143: 135964, 1991. Debatin KM, Krammer PH, Peter ME. Two CD95 (APO-1/Fas) sig-
10. Gompertz S, OBrien C, Bayley D, Hill SL, Stockley RA. Changes in naling pathways. EMBO J 17: 167587, 1998.
bronchial inammation during acute exacerbations of chronic bronchi- 31. Hirata H, Takahashi A, Kobayashi S, Yonehara S, Sawai H, Okazaki T,
tis. Eur Respir J 17(6): 111219, 2001. Yamamoto K, Sasada M. Caspases are activated in a branched protease
11. Donaldson GC, Seemungal TA, Patel IS, Bhowmik A, Wilkinson TM, cascade and control distinct downstream processes in Fas-induced
Hurst JR, Maccallum PK, Wedzicha JA. Airway and systemic inam- apoptosis. J Exp Med 187: 587600, 1998.
mation and decline in lung function in patients with COPD. Chest 32. Tang D, Kidd VJ. Cleavage of DFF-45/ICAD by multiple caspases is
128(4): 19952004, 2005. essential for its function during apoptosis. J Biol Chem 273: 2854952,
12. Parr DG, White AJ, Bayley DL, Guest PJ, Stockley RA. Inammation 1998.
in sputum relates to progression of disease in COPD: A prospective 33. Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of apoptotic
study. Respir Res 7: 136, 2006. program in cell-free extracts: Requirement for dATP and cytochrome
13. Ekberg-Jansson A, Andersson B, Bake B, Boijsen M, Enanden I, c. Cell 86: 14757, 1996.
Rosengren A et al. Neutrophil associated activation markers in healthy 34. Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human
smokers relates to a fall in DLCO and to emphysematous change on protein homologous to C. elegans CED-4, participates in cytochrome
high resolution CT. Respir Med 95(5): 36373, 2001. c-dependent activation of caspase-3. Cell 90: 40513, 1997.
14. Yoshioka A, Betsuyaku T, Nishimura M, Miyamoto K, Kondo T, 35. Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, Newmeyer
Kawakami Y. Excessive neutrophil elastase in bronchoalveolar lavage uid DD, Wang HG, Reed JC, Nicholson DW, Alnemri ES, Green DR,
in subclinical emphysema. Am J Respir Crit Care Med 152: 212732, 2005. Martin SJ. Ordering the cytochrome c-initiated caspase cascade:
15. Betsuyaku T, Nishimura M, Takeyabu K, Tanino M, Venge P, Xu S Hierarchical activation of caspases-2, -3, -6, -7, -8, and -10 in a cas-
et al. Neutrophil granule proteins in bronchoalveolar lavage uid from pase-9-dependent manner. J Cell Biol 144: 28192, 1999.
patients with subclinical emphysema. Am J Respir Crit Care Med 159: 36. Rao RV, Hermel E, Castro-Obregon S, del Rio G, Ellerby LM, Ellerby
198591, 1999. HM, Bredesen DE. Coupling endoplasmic reticulum stress to the cell
16. Ge YM, Zhu YJ, Luo WC, Gong YH, Zhang XQ. Damaging role death program. Mechanism of caspase activation. J Biol Chem 276:
of neutrophil elastase in the elastic ber and basement membrane in 3386974, 2001.
human emphysematous lung. Chin Med J 103(7): 58894, 1990. 37. Szegezdi E, Fitzgerald U, Samali A. Caspase-12 and ER-stress-
17. Fujita J, Nelson NL, Daughton DM, Dobry CA, Spurzem JR, Irino S mediated apoptosis: The story so far. Ann NY Acad Sci 1010: 18694, 2003.
et al. Evaluation of elastase and antielastase balance in patients with 38. Rydell-Tormanen K, Uller L, Erjefait JS. Direct evidence of secondary
chronic bronchitis and pulmonary emphysema. Am Rev Respir Dis 142: necrosis of neutrophils during intense lung inammation. Eur Respir J
5762, 1990. 28(2): 26874, 2006.
18. Rennard SI, Daughton D, Fujita J, Oehlerking MB et al. Short term 39. Corre F, Lellouch J, Schwartz D. Smoking and leucocyte counts: results
smoking reduction is associated with reduction in measures of lower of an epidemiological study. Lancet 2: 63234, 1971.
respiratory tract inammation in heavy smokers. Eur Respir J 3(7): 40. Van Eeden SF, Hogg JC. The response of human bone marrow to
75259, 1990. chronic cigarette smoking. Eur Respir J 15: 91521, 2000.
19. Sin DD, Anthonisen NR, Soriano JB, Agusti AG. Mortality in COPD: 41. Yeung MC, Buncio AD. Leukocyte count, smoking and lung function.
Role of co-morbidities. Eur Respir J 28(6): 124557, 2006. Am J Med 76: 3137, 1984.
20. Celli BR, Cote CG, Marin JM, Caanova C, Montes de Oca M, 42. Sparrow D, Glynn RJ, Cohen M, Weiss ST. The relationship of the
Mendez RA, Pinto-plata V, Cabral HJ. The body mass index, airow peripheral leukocyte count and cigarette smoking to pulmonary func-
obstruction, dyspnoea and exercise capacity index in chronic obstruc- tion among adult young men. Chest 86: 38386, 1984.
tive pulmonary disease. N Engl J Med 350(10): 100512, 2004. 43. Pletz MW, Ioanas M, de Roux A, Burkhardt O, Lode H. Reduced
21. Pinto-Plata VM, Mullerova H, Toso JF, Feudjo-Tepie M, Soriano JB, spontaneous apoptosis in peripheral blood neutrophils during exacer-
Vessey RS, Celli BR. C-Reactive protein in patients with COPD, con- bations of COPD. Eur Respir J 23(4): 53237, 2004.
trol smokers and non-smokers. Thorax. 61(1): 2328, 2006. 44. Uller L, Persson CG, Erjefalt JS. Resolution of airway disease: Removal
22. Laurell CBaSE. The electrophoretic alpha-globulin pattern of serum in of inammatory cells through apoptosis, egression or both. Trends
alpha antitrypsin deciency. Scand J Clin Invest 15: 13240, 1963. Pharmscol Sci 27(9): 46166, 2006.
23. Stockley RA. Neutrophils and the pathogenesis of COPD. Chest 121: 45. Rytila P, Plataki M, Bucchieri F, Uddin M, Nong G, Kinnula VL.
151S55S, 2002. Djukanovic. Airway neutrophilia in COPD is not associated with
24. Fouret P, du Bois RM, Bernaudin JF, Takahashi H, Ferrans VJ, Crystal increased neutrophil survival. Eur Respir J 28: 116369, 2006.
RG. Expression of the neutrophil elastase gene during human bone 46. Yoshikawa T, Dent G, Ward J, Angco G, Nong G, Nomura N,
marrow cell dierentiation. J Exp Med 169: 83345, 1989. Hirata K, Djukanovic R. Impaired neutrophil chemotaxis in chronic
25. Adkison AM, Raptis SZ, Kelley DG, Pham CTN. Dipeptidyl pepti- obstructive pulmonary disease. Am J Respir crit Care Med 175(5): 473
dase I activates neutrophil-derived serine proteases and regulates the 79, 2007.

186
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
47. Churg A, Dai J, Tai H, Xie C, Wright JL. Tumour necrosis factor alpha 67. DiMango E, Zar HJ, Bryan R, Prince A. Diverse Pseudomonas aeru-
is central to acute cigarette smoke induced inammation and connec- ginosa gene products stimulate respiratory epithelial cells to produce
tive tissue breakdown. Am J Respir Crit Care Med 166: 84954, 2002. interleukin-8. J Clin Invest 96: 220410, 1995.
48. Sakao S, Tatsumi K, Igari H, Watanebe R, Shino Y, Shirasawa H, 68. Khair OA, Davies RJ, Devalia JL. Bacterial-induced release of inam-
Kuriyama T. Association of tumour necrosis factor alpha gene pro- matory mediators by bronchial epithelial cells. Eur Respir J 9(9): 1913
moter polymorphism with low attenuation areas on high resolution 22, 1996.
CT in patients with COPD. Chest 122: 41620, 2002. 69. Alcorn MJ, Booth JL, Coggeshall KM, Metcalf JP. Adenovirus type 7
49. Hacievliyagil SS, Gunen H, Mutlu LC, Karabulut AB, Temel I. induces interleukin-8 production via activation of extracellular regu-
Association between cytokines in induced sputum and severity of lated kinase. J Virol 75: 645059, 2001.
chronic obstructive pulmonary disease. Respir Med 100(5): 84654, 70. Johnston SL, Papi A, Monick MM, Hunninghake GW. Rhinoviruses
2006. induce interleukin 8 mRNA and protein production in human mono-
50. Kuschner WG, DAlessandro A, Wong H, Blanc PD. Dose dependent cytes. J Infect Dis 175: 32329, 1997.
cigarette smoking- related inammatory responses in healthy adults. 71. Deforge LE, Preston AM, Takeuchi E, Kenney J, Boxer LA, Remick
Eur Respir J 9: 198994, 1996. DG. Regulation of interleukin 8 gene expression by oxidant stress.
51. Aaron SD, Angel JB, Lunau M et al. Granulocyte inammatory J Biol Chem 268: 2556876, 1993.
markers and airway infection during acute exacerbations of chronic 72. Hill AT, Bayley D, Stockley RA. The interrelationship of sputum
obstructive pulmonary disease. Am J Respir Crit Care Med 163: 34955, inammatory markers in patients with chronic bronchitis. Am J Respir
2001. Crit Care Med 160: 89398, 1999.
52. Di Francia M, Barbier D, Mege JL, Orehek J. Tumor necrosis factor 73. Yamamoto C, Yoneda T, Yoshikawa M, Fu A, Tokuyama T,
alpha levels and weight loss in chronic obstructive pulmonary disease. Tsukaguchi K et al. Airway inammation in COPD assessed by spu-
Am J Respir Crit Care Med 150: 145355, 1994. tum levels of Interleukin-8. Chest 112(2): 50510, 1997.
53. de Godoy I, Donahoe M, Calhoun WJ, Mancino J, Rogers RM. Elevated 74. Borgeat PE, Krump E, Palmanteir R, Picard S, Dallaire N, McDonald P
TNF-alpha production by peripheral blood monocytes of weight-losing et al. The synthesis of leukotrienes by the human neutrophil. In:
COPD patients. Am J Respir Crit Care Med 153: 63337, 1996. Holgate S (ed.), SRS-A to leukotrienes; the dawning of a new treatment,
54. Calikoglu M, Sahin G, Unlu A, Ozturk C, Tamer L, Ercan B, Kanik pp. 6984. Blackwell Oxford, 2003.
A, Atik U. Lepitn and TNF levels in patients with chronic obstruc- 75. Crooks SW, Stockley RA. Leukotriene B4. Int J Biochem Cell Biol 30:
tive pulmonary disease and their relationship to nutritional parameters. 17378, 1998.
Respiration 71(1): 4550, 2004. 76. Tonnesen MG, Anderson DC, Springer TA, Knedler A, Avdi N,
55. Nguyen LT, Bedu M, Caillaud D, Beaufrere B, Beaujon G, Vasson M, Henson PM. Adherence of neutrophils to cultured human microvas-
Coudert J, Ritz P. Increased resting energy expenditure is related to cular endothelial cells. Stimulation by chemotactic peptides and lipid
plasma TNF alpha concentration in stable COPD patients. Clin Nutr mediators and dependence upon the Mac-1, LFA-1, p150,95 glyco-
18(5): 26974, 1999. protein family. J Clin Invest 83: 637, 1989.
56. Brogger J, Steen VM, Eiken HG, Gulsvik A, Bakke P. Genetic associa- 77. Nohgawa M, Sasada M, Maeda A, Asagoe K, Harakawa N, Takano K
tion between COPD polymorphisms in TNF, ADRB2 and EPHX1. et al. Leukotriene B4 activated endothelial cells promote transendothe-
Eur Respir J 27(4): 68288, 2006. lial neutrophil migration. J Leuk Biol 62: 2039, 1997.
57. Vuillemenot BR, Rodriguez JF, Hoyle GW. Lymphoid tissue and 78. Hill AT, Bayley D, Campbell EJ, Hill SL, Stockley RA. Airways
emphysema in the lungs of transgenic mice inducibly expressing inammation in chronic bronchitis; the eects of smoking and alpha1-
TNF. Am J Respir Crit Care Med 30: 43848, 2004. antitrypsin deciency. Eur Respir J 15: 88690, 2000.
58. Wright JL, Tai H, Wang R, Wang X, Churg A. Cigaretter smoke 79. Montuschi P, Kharitonov S, Ciabattoni G, Barnes PJ. Exhaled leukot-
up-regulates pulmonary vascular matrix metalloproteinases via TNF alpha rienes and prostaglandins in COPD. Thorax 58(7): 58588, 2003.
signaling. Am J Physiol Lung Cell Mol Physiol 292(1): L12533, 2007. 80. Bhowmik A, Seemungal TAR, Sapsford RJ. Relation of sputum
59. Churg A, Wang RD, Tai H, Wang X, Xie C, Wright JL. Tumour inammatory markers to symptoms and lung function changes in
necrosis factor alpha drives 70% of cigarette smoke-induced emphy- COPD. Thorax 55: 11420, 2000.
sema in the mouse. Am J Respir Crit Care Med 170: 49298, 2004. 81. White AJ, Gompertz S, Bayley DL, Hill SL, OBrien C, Unsal I et al.
60. Lucey EC, Keane J, Kuang PP, Snider GL, Goldstein RH. Severity of Resolution of bronchial inammation is related to bacterial eradica-
elastase-induced emphysema is decreased in TNF and IL-1 receptor tion following treatment of exacerbations of chronic bronchitis. Thorax
decient mice. Lab Invest 82: 7985, 2002. 58(8): 68085, 2003.
61. Chung KF. Cytokines in chronic obstructive pulmonary disease. Eur 82. Woolhouse IS, Bayley DL, Stockley RA. Sputum chemotactic activ-
Respir J 34: 50s59s, 2001. ity in chronic obstructive pulmonary disease: Eect of 1-anti-trypsin
62. Rusznak C, Mills PR, Devalia JL, Sapsford RJ, Davies RJ, Lozewicz deciency and the role of leukotriene B4 and interleukin 8. Thorax. 57:
S. Eect of cigarette smoking on the permeability and IL-1beta and 70914, 2002.
sICAM-1 release from cultured human bronchial epithelial cells of 83. Beech KM, Kornmann O, Buhl R et al. Neutrophil chemotactic activ-
never smokers, smokers and patients with chronic obstructive pulmo- ity of sputum from patients with COPD: Role of interleukin 8 and
nary disease. Am J Respir Cell Mol Biol 23(4): 53036, 2000. Leukotriene B4. Chest 123: 124047, 2003.
63. Hegab EA, Sakamoto T, Saitoh W et al. Polymorphisms of IL-4, 84. Traves SL, Culpitt SV, Russell RE, Barnes PJ, Donnelly LE. Increased
IL-13 and ADRB2 genes in COPD. Chest 126: 183239, 2004. levels of the chemokine GROalpha and MCP-1 in sputum samples
64. Asada M, Yamaya M, Ebihara S, Yasufa H, Tomita N, Kubo H, Sasaki H, from patients with COPD. Thorax 57: 59095, 2002.
Hegab A, Sakamoto T, Sekizawa K. Interleukin-1 gene polymrphisms 85. Morrison D, Strieter RM, Donnelly SC, Burdick MD, Kunkel SL,
associated with COPD. Chest 128: 107273, 2005. MacNee W. Neutrophil chemokines in bronchoalveolar lavage uid
65. Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K. and leukocyte-conditioned medium from smokers and non-smokers.
Interleukin-1beta causes pulmonary inammation, emyphysema and Eur Respir J 12: 106772, 1998.
airway remodelling in the adult murine lung. Am J Respir Cell Mol Biol 86. Traves SL, Smith SJ, Barnes PJ, Dinelly LE. Specic CXC but not CC
32(4): 31118, 2005. chemokines cause elevated monocyte migration in COPD. A role for
66. Matsushima K, Morishita K, Yoshimura T, Lavu S, Kobayashi Y, Lew W CXCR2. J Leuk Biol 76(2): 44150, 2004.
et al. Molecular cloning of a human monocyte-derived neutrophil chem- 87. Matheson M, Rynell AC, McClean M, Berend N. Cigarette smoking
otactic factor (MDNCF) and the induction of MDNCF mRNA by increases neutrophil formyl methionyl leucyl phenylalanine receptor
interleukin-1 and tumour necrosis factor. J Exp Med 169: 148590, 1988. numbers. Chest 123(5): 164246, 2003.

187
Asthma and COPD: Basic Mechanisms and Clinical Management

88. de Boer W. Perspectives for cytokine antagonist therapy in COPD. 111. Davenpeck KL, Steeber DA, Tedder TF, Bochner BS. Rat neutrophils
DDT 10(2): 93106, 2005. express 4 and 1 integrins and bind to vascular adhesion molecule 1
89. Rennard SI, Fogarty C, Kelsen S, Long W, Ramsdell J, Allison J, and mucosal adressin cell adhesion molecule. Blood 91: 234146, 1988.
Mahler D, Saadeh C, Siler T, Snell P, Korenblat P, Smith W, Kaye 112. Clayton A, Evans RA, Pettit E, Hallett M, Williams JD, Steadman R.
M, Mandel M, Andrews C, Prabhu R, Donohue JF, Watt R, Lo KH, Cellular activation through the ligation of intercellular adhesion mol-
Schlenker-Herceg R, Barnathan ES, Murray J. The safety and ecacy ecule1. J Cell Sci 111: 44353, 1998.
of iniximab in moderate to severe chronic obstructive pulmonary 113. Qu Q , Zhoa M, Gu G. ICAM-1 and HLA-DR expression in bron-
disease. Am J Respir Crit Care Med 175(9): 92634, 2007. chial epithelial cells from patients with chronic obstructive pulmonary
90. Gompertz S, Stockley RA. A randomised controlled trial of a disease. Zhonghua Jie He He Hu Xi Za Zhi 21(7): 41114, 1998.
Leukotriene synthesis inhibitor in patients with COPD. Chest 122: 114. Burns MJ, Walker DC, Brown ES, Thurmon LT, Bowden RA,
28994, 2002. Keese CR et al. Neutrophil transendothelial migration is independ-
91. Wagner JG, Roth RA. Neutrophil migration mechanisms with an ent of tight junctions and occurs preferentially at tricellular corners.
emphasis on pulmonary vasculature. Pharmacol Rev 52(3): 34974, 2000. J Immunol 159: 2893903, 1997.
92. Walchek B, Kahn J, Fisher JM, Wang BB, Fisk RS, Payan DG et al. 115. Newman PJ. The biology of PECAM-1. J Clin Invest 99: 3, 1997.
Neutrophil rolling altered by inhibition of L-Selectin shedding in 116. Muller WA. The role of PECAM1 in leukocyte emigration. Studies
vitro. Nature 380: 72023, 1996. in vitro and in vivo. J Leuk Biol 66: 698704, 1995.
93. Drost EM, MacNee W. Potential role of IL8, platelet activating fac- 117. Yadav R, Larbi KY, Young RE, Nourshargh S. Migration of leuko-
tor and TNF- in the sequestration of neutrophils in the lung; eects cytes through the vessel wall and beyond. Thromb Haemost 90(4):
on neutrophil deformability, adhesion receptor expression and chemo- 598606, 2002.
taxis. Eur J Immunol 32: 393403, 2002. 118. Shang XZ, Issekutz AC. 2 (CD18) and 1 (CD29) Integrin mecha-
94. Matsuba KT, Van Eeden SF, Bicknell SG, Walker BAM, Hayashi S, nisms in migration of human polymorphonuclear leucocytes and
Hogg JC. Apoptosis in circulating PMNs: Increasing susceptibility in monocytes through lung broblast barriers. Shared and distinct
L-selectin decient PMN. Am J Physiol 272: H2852hH2858, 1997. mechanisms. Immunol 92: 52735, 1997.
95. Malik AR, Lo SK. Vascular endothelial adhesion molecules and tissue 119. Shang XZ, Yednock T, Issekutz AC. 9 Integrin is expressed on
inammation. Pharmacol Rev 48: 21329, 1996. human neutrophils and contributes to neutrophil migration through
96. Spertini O, Luscinskas FW, Munro JM, Grin JD, Gimbrone MAJ, human lung and synovial broblast barriers. J Leuk Biol 66: 80916,
Tedder TF. Leukocyte adhesion molecule-1 (LAM1, L-selectin) 1999.
interacts with an inducible endothelial cell ligand to support leuko- 120. Thompson RD, Noble KE, Larbi KY, Dewar A, Duncan GS, Mak
cyte adhesion. J Immunol 147: 256578, 1991. TW et al. Platelet-endothelial cell adhesion molecule-1 (PECAM-
97. Alon R, Chen S, Puri KD, Finger EB, Springer TA. The kinetics of 1) decient mice demonstrate a transient and cytokine-specic role
L-selectin tethers and the mechanisms of selectin mediated rolling. for PECAM1 in leukocyte migration through perivascular basement
J Cell Biol 138: 116980, 1997. membrane. Blood 97(6): 185460, 2001.
98. Klein CL, Bittenger F, Kohler H, Wagner M, Otto M, Hermanns I 121. Wright DC, Gallin JL. Secretory responses of human neutrophils:
et al. Comparative studies on vascular endothelium in vitro. Eects of Exocytosis of specic (secondary) granules by human neutrophils
cytokines on the expression of E-selectin. Pathobiology 65: 8392, 1995. during adherence in vitro and during exudation in vivo. J Immunol
99. Steegmaier M, Levinovitz A, Isenmann S et al. The E-selectin ligand 123: 25894, 1979.
ESL-1 is a variant of a receptor for broblast growth factor. Nature 122. Foxman EF, Campbell JJ, Butcher EC. Multistep navigation and the
373: 61520, 1995. combinatorial control of leukocyte chemotaxis. J Cell Boil 139: 1349
100. Rise GC, Larsen S, Lofdahl CG, Andersson BA. Circulating cell 59, 1997.
adhesion molecules in bronchial lavage and serum in COPD patients 123. Foxman EF, Kunkel EJ, Butcher EC. Integrating conicting chemo-
with chronic bronchitis. Eur Respir J 7(9): 167377, 1994. tactic signals: the role of memory in leukocyte navigation. J Cell Boil
101. Crockett-Torabi A, Fantone JC. The Selectins: Insights into selectin 147(3): 57787, 1999.
induced intracellular signalling in leukocytes. Immunol Res 14: 237 124. Devreotes P, Janetopoulos C. Eukaryote chemotaxis: Distinctions
51, 1995. between directional sensing and polarisation. J Biol Chem: 2044548,
102. Williams MA, Solomkin JS. Integrin mediated signalling in human 1988.
neutrophil functioning. J Leuk Biol 65: 72536, 1999. 125. Charest PG, Firtel RA. Big roles for small GTPases in the control of
103. Diamond MS, Staunton DE, de Fourgerolles AR et al. ICAM-1 directed cell movement. Biochem J 401: 37790, 2007.
(CD-54): a counter receptor for MAC-1 (CD11b/CD18). J Cell Biol 126. Huttunlocher A. Cell polarisation mechanisms during directed cell
111: 312939, 1990. migration. Nature Cell Biol 7(4): 33637, 2005.
104. Rainger GE, Buckley C, Simmons DL, Nash GB. Cross-talk between 127. Nishiya N, Kiosses WB, Han J, Ginsberg GH. An 4
cell adhesion molecules regulates the migration velocity of neu- integrinpaxillinArf-GAP complex restricts Rac activation to the
trophils. Curr Biol 17: 31625, 1997. leading edge of migrating cells. Nature Cell Biol 7: 34345, 2005.
105. Borregaard N, Cowland JB. Granules of the human neutrophilic poly- 128. Ohta Y, Hartwig JH, Stossel TP. A Rho and ROCK regulated GAP
morphonuclear leukocyte. Blood 89: 350321, 1997. for Rac binds lamin A to control actin remodelling. Nature Cell Biol
106. Sengelov H. Secretory vesicles of human neutrophils. Eur J Haem 28: 80314, 2006.
57(Suppl 58): 624, 1996. 129. Andrew N, Insall RH. Chemotaxis in shallow gradients is mediated
107. Laudanna C, Campbell JJ, Butcher EC. Role of Rho in chemoat- independently of PtdIns 3-kinase by biased choices between random
tractant activated leukocyte adhesion through integrins. Science 271: protrusions. Nature Cell Biol 9(2): 193200, 2007.
98183, 1996. 130. Stockley RA, Shaw J, Aord SC, Morrison HM, Burnett D. Eect of
108. Michishita M, Videm V, Arnaout MA. A novel divalent cation-bind- alpha one proteinase inhibitor on neutrophil chemotaxis. Am J Respir
ing site in the A domain of the beta 2 integrin CR3 (CD11b/CD18) Cell Mol Biol 2: 16370, 1990.
is essential for ligand binding. Cell 72: 85767, 2003. 131. Lomas DA, Stone SR, Llewellyn-Jones C, Keogan MT, Wang ZM,
109. Noguera A, Batle S, Miralles C, Iglesias J, Busquets X, MacNee W Rubin H et al. The control of neutrophil chemotaxis by inhibitors of
et al. Enhanced neutrophil response in chronic obstructive pulmonary cathepsin G and chymotrypsin. J Biol Chem 270: 43723443, 1995.
disease. Thorax 56: 43237, 2001. 132. Delclaux C, Delacourt C, DOrtho MP, Boyer V, Lafuma C, Harf A.
110. Hashimoto M, Shingu M, Ezaki I, Nobunaga M, Minamihara M, Role of gelatinase B and elastase in human polymorphonuclear neu-
Kato K et al. Production of soluble ICAM1 from human endothelial trophil migration across basement membrane. Am J Respir Cell Mol
cells induced by IL-1 and TNF. Inammation 18: 16373, 1994. Biol 14: 28895, 1996.

188
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
133. Allport JR, Ding HT, Ager A, Steeber DA, Tedder TF, Luscinskas pulmonary and systemic microcirculation of rabbits. J Immunol 144:
FW. L-selectin shedding does not regulate human neutrophil attach- 232733, 1990.
ment, rolling, or transmigration across human vascular endothelium 153. Ramamoorthy C, Sasaki SS, Su DL, Sharar SR, Harlan JM, Winn
in vitro. J Immunol 158: 436572, 1997. RK. CD18 adhesion blockade decreases bacterial clearance and neu-
134. Mackeral AJ, Cottell DC, Russell KJ, Fitxgerald MX, OConner CM. trophil recruitment after intrapulmonary E Coli but not after Staph
Migration of neutrophils across human pulmonary endothelial cells is Aureus. J Leuk Biol 61: 16772, 1997.
not blocked by matrix metalloproteinase or serine proteinase inhibi- 154. Qin L, Quinlan WM, Doyle NA, Graham L, Sligh JE, Takei F et al.
tors. Am J Respir Cell Mol Biol 20: 120919, 1999. The role of CD11/CD18 and ICAM1 in acute pseudomonas aerugi-
135. Steadman R, St John PL, Evans RA, Thomas GL, Davies M, Heck nosa induced pneumonia. J Immunol 157: 501621, 1996.
LW et al. Human neutrophils do not degrade major basement mem- 155. Hellewell PG, Young SK, Henson PM, Worthen GS. Disparate role
brane components during chemotactic migration. Int J Biochem Cell of the 2 integrin CD18 in the local accumulation of neutrophils in
Biol 29: 9931004, 1997. pulmonary and cutaneous inammation in rabbit. Am J Respir Cell
136. Tkalcevic J, Novelli M, Phylactides M, Iredale JP, Segal AW, Roes J. Mol Biol 10: 39198, 1994.
Impaired immunity and enhanced resistance to endotoxin in the 156. Shoburg DS, Quinlan WM, Hebert CA, Ashkenazi A, Doerschuk
absence of neutrophil elastase and cathepsin G. Immunity 12(201): CM. Cytokine production in response to stimuli that elicit either
210, 2000. CD18 dependent or CD18 independent neutrophil emigration. Am J
137. Betsuyaku T, Shipley JM, Liu Z, Senior RM. Neutrophil emigration Respir Crit Care Med 147: A1088, 1994.
in the lungs, peritoneum, and skin does not require gelatinase B. Am J 157. Doyle NA, Bhagwan SD, Meek BB, Kutkoshi GJ, Steeber DA,
Respir Cell Mol Biol 20: 13039, 1999. Tedder TF et al. Neutrophil margination, sequestration and emigra-
138. Wright JL, Farmer SG, Churg A. Synthetic serine elastase inhibi- tion in the lungs of L-selectin decient mice. J Clin Invest 99: 52633,
tor reduces cigarette smoke induced emphysema in guinea pigs. Am J 1997.
Respir Crit Care Med 166: 95460, 2002. 158. Burnett D, Chamba A, Hill SL, Stockley RA. Neutrophils from sub-
139. Delacourt C, Herigault S, Delclaux C, Poncin A, Levame M, Harf jects with chronic obstructive disease show enhanced chemotaxis and
A et al. Protection against acute lung injury by intravenous or intrat- extracellular proteolysis. Lancet 2: 104346, 1987.
racheal pretreatment with EPI-HNE-4; a new potent neutrophil 159. Woolhouse IS, Bayley DL, Lalor P, Adams DH, Stockley RA.
elastase inhibitor. Am J Respir Cell Mol Biol 26: 29097, 2002. Endothelial interactions of neutrophils under ow in chronic obstruc-
140. Chun TH, Sabeh F, Ota I, Murphy H, McDonagh KT, Holmbeck K tive pulmonary disease. Eur Respir J 25(4): 61217, 2005.
et al. MTI-MMP dependent neovessel formation with the controls of 160. Gonalez S, Hards J, van Eeden S, Hogg JC. The expression of adhe-
a 3 dimension extra cellular matrix. J Cell Biol 167: 75767, 2004. sion molecules in cigarette smoke induced airways obstruction. Eur
141. Sabeh F, Ota I, Holmbeck K, Birkedal-Hansen H et al. Tumor cell Respir J 9: 19952001, 1996.
trac through the extra cellular matrix is controlled by the mem- 161. Yoshikawi T, Dent G, Ward J, Angco G, nong G, Nomura N,
brance anchored collagenases MTI-MMP. J Cell Biol 167: 76981, Hirata K, Djukanovic R. Impaired neutrophil chemotaxis in chronic
2004. obstructive pulmonary disease. Am J Respir Crit Care Med 175(5):
142. Even-Ram S, Yamada K. Cell migration in a 3D matrix. Curr Opin 47379, 2007.
Cell Biol 17: 52432, 2005. 162. Yadav R, Larbi KY, Young RE, Nourshargh S. Migration of leuko-
143. Doerschuk CM, Allard MF, Hogg JC. Neutrophil kinetics in rab- cytes through the vessel wall and beyond. Thromb Haemost 90(4):
bits during infusion of zymosan-activated plasma. J Appl Physiol 67: 598606, 2002.
8895, 1989. 163. Dangereld J, Larbi KY, Huang MT, Dewar A, Nourshargh S.
144. Downey GP, Worthen GS, Hyde DM. Neutrophil sequestration and PECAM-1 (CD!) homophillc interaction up-regulates 61 on
migration in localised pulmonary inammation. capillary localisation transmigrated neutrophils in vivo and plays a functional role in the
and migration across the interalveolar septum. Am Rev Respir Dis ability of 6 integrins to mediate leukocyte migration through
147(1): 16876, 1993. the perivascular basement membrane. J Exp Med 196(9): 120111,
145. Hogg JC. Neutrophil kinetics and lung injury. Physiolog Rev 67(1249): 2002.
1295, 1987. 164. Jano A, Sloan B, Weinbaum G, Damiano V, Sandhaus RA, Elias J
146. Doerschuck CM, Beyers N, Coxson HO, Wiggs B, Hogg JC. et al. Experimental emphysema induced with puried human neu-
Comparison of neutrophil and capillary diameters and their relation trophil elastase; tissue localisation of the instilled protease. Am Rev
to neutrophil sequestration in the lung. J Appl Physiol 74: 304045, Respir Dis 115(3): 46178, 1977.
1993. 165. Senior RM, Tegner H, Kuhn C, Ohlsson K, Starcher BC,
147. Selby C, Drost E, Gillooly M et al. Neutrophil sequestration in lungs Pierce JA. The induction of pulmonary emphysema with human leu-
removed at surgery: The eect of microscopic emphysema. Am J Respir kocyte elastase. Am Rev Respir Dis 116(3): 46975, 1977.
Crit Care Med 149: 152633, 1994. 166. Lucey EC, Stone PJ, Breuer R, Christensen TG, Calore JD,
148. Erzurum SC, Downey GP, Doherty DE, Schwabb B, Elson EL, Catanse A et al. Eect of combined human neutrophil cathepsin G
Worthen GS. Mechanisms of lipopolysaccharide retention. J Immunol and elastase on induction of secretory cell metaplasia and emphy-
149: 15462, 1992. sema in hamsters, with in vitro observations on elastolysis by these
149. Doerschuk CM. The role of CD18 mediated adhesion in neutrophil enzymes. Am Rev Respir Dis 132: 36266, 1985.
sequestration induced by infusion of activated plasma in rabbits. Am J 167. Gross P, Pzer EH, Tolker B, Babyok MA, Kaschak M. Experimental
Respir Cell Mol Biol 7: 14048, 1992. emphysema; its production with papain in normal and silicoctic rats.
150. Morland CM, Morland BJ, Darbyshire PJ, Stockley RA. Migration of Arch Environ Health 11: 5058, 1964.
CD18 decient neutrophils in vitro; evidence for a CD18 independ- 168. Marco V, Mass B, Meranze DR, Weinbaum G, Kimbel P. Induction
ent pathway induced by IL8. Biochem Biophys Acta 1500(1): 7076, of experiemental emphysema in dogs using leukocyte homogenates.
2000. Am Rev Respir Dis 104: 59598, 1971.
151. Mackerel AJ, Russel KJ, Brady CS, FitzGerald MX, OConnor CM. 169. Blackwood CE, Hosannah Y, Perman E, Keller S, Mandl L.
Interleukin 8 and leukotriene B4 but not formylmethionyl leucylphe- Elasteolytic titre of inducing enzyme as determinant of the response.
nylalanine stimulate CD 18 independent migration of neutrophils Proc Soc Exp Biol Med 144: 45054, 1973.
across human pulmonary endothelial cells in vitro. Am J Respir Cell 170. Snider GL, Hayes JA, Franzblau C, Kagan HM, Stone PJ, Korthy A.
Mol Biol 23: 15461, 2000. Relationship between elasteolytic activity and experimental emphy-
152. Doerschuk CM, Winn RK, Coxson HO, Harlan JM. CD18 depend- sema inducing properties of papain preparations. Am Rev Respir Dis
ent and independent mechanisms of neutrophil emigration in 110: 25457, 1974.

189
Asthma and COPD: Basic Mechanisms and Clinical Management

171. Lucey EC, Stone PJ, Powers JC, Snider GL. Amelioration of human 190. Takeyama K, Agusti C, Ueki IF, Lausier J, Cardell LO, Nadel JA.
neutrophil elastase-induced emphysema in hamsters by pretreatment Neutrophil-dependent goblet cell degranulation; role of membrane
with an oligopeptide chlormethyl ketone. Eur Respir J 2: 42127, bound elastase and adhesion molecules. Am J Physiol 19: L294302,
1989. 1998.
172. Rudolphus A, Kramps JA, Mauve I, Dijkman JH. Intratracheally 191. Lanone S, Zheng T, Zhu Z, Liu W, Lee CG, Ma B et al. Overlapping
instilled antileukoprotease and alpha 1 proteinase inhibitor; eect and enzyme specic contributions of matrix metalloproteinases-9
on human neutrophil elastase-induced experimental emphysema and and -12 in IL-13 induced inammation and remodelling. J Clin Invest
pulmonary localisation. Histochem J 26: 81727, 1994. 110(463): 474, 2002.
173. Beith JG. Elastases: catalytic and biological properties. In: Mecham 192. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, Riese RJJ et al. Inducible
RP (ed.), Biology of extracellular matrix: Regulation of Matrix targeting of IL-13 to the adult lung causes matrix metalloproteinase
Accumulation, pp. 217320. Orlando FL: Academic, 1986. and cathepsin dependent emphysema. J Clin Invest 106: 144546, 2000.
174. Kaenah W, Buttle DJ, Burnett D, Hollander AP. Cleavage of native 193. Shim JJ, Dabbagh K, Ueki IF, Dao-Pick T, Takeyama K, Tam DCW
type 1 collgane by human neutrophil elastase. Biochem J 330: 897902, et al. IL-13 induces mucin production by stimulating epidermal
1998. growth factor receptors and by activating neutrophils. Am J Physiol
175. Niederman MS, Merrill WW, Polomski LM, Reynolds HY, Gee JB. Lung Cell Mol Physiol 280: L13440, 2001.
Inuence of IgA and elastase on trachea cell bacterial adherence. Am 194. Smallman LA, Hill SL, Stockley RA. Reduction of ciliary beat fre-
Rev Respir Dis 133: 25560, 1986. quency in vitro by sputum from patients with bronchiectasis; a serine
176. Vogt W. Cleavage of the fth component of complement and genera- proteinase eect. Thorax 39: 66367, 1984.
tion of a functionally active C5b6-like complex by human leukocyte 195. Amitani R, Wilson R, Rutman A, Read R, Ward C, Burnett D et al.
elastase. Immunolbiology 201: 47077, 2000. Eect of human neutrophil elastase and pseudomonas aeruginosa
177. Fick RBJ, Robbins RA, Squier SU, Schoderbeck WE, Russ WD. proteinases on human respiratory epithelium. Am J Respir Cell Mol
Complement activation in cystic brosis respiratory uids: In vivo Biol 4: 2632, 1991.
and in vitro generation of a C5a and chemotactic activity. Paediatr Res 196. Aoshiba K, Yasuda K, Yasui S, Tamaoki J, Nagai A. Serine protein-
20: 125868, 1986. ases increase oxidative stress in lung cells. Am J Physiol Lung Cell Mol
178. Ashcroft GS, Lei K, Jin W et al. Secretory leukocyte protease inhibi- Physiol 281: L55664, 2001.
tor mediates non redundant functions necessary for normal wound 197. Nakajoh M, Fukushima T, Suzuki K, Yamaya M, Nakayama K,
healing. Nat Med 6: 114753, 2000. Sekizawa K et al. Retinoic acid inhibits elastase-induced injury in
179. Owen CA, Campbell MA, Sannes PL, Boukedes SS, Campbell EJ. human lung epithelial cell lines. Am J Respir Cell Mol Biol 28: 296
Cell-surface bound elastase and cathespin G on human neutrophils; a 304, 2002.
novel, non-oxidative mechanism by which neutrophils focus and pre- 198. Rickard K, Rennard S. Neutrophil elastase causes detachment of
serve catalytic activity of serine proteinases. J Cell Biol 131: 77589, bronchial epithelial cells from extracellular matrix. Am Rev Respir Dis
1995. 139: 406, 1989.
180. Owen CA, Campbell MA, Boukedes SS, Campbell EJ. Cytokines 199. Ballieux BE, Hiemstra PS, Klar-Mohamad N, Hagen EC, Van Der
regulate membrane-bound leukocyte elastase on neutrophils; a novel Woude FJ, Daha MR. Detachment and cytolysis of human endothe-
mechanism for eector activity. Am J Physiol Lung Cell Mol Physiol lial cells by proteinase 3. Eur J Immunol 24: 321115, 1994.
272: L385lL393, 1997. 200. Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y,
181. Campbell EJ, Campbell MA, Owen CA. Bioactive proteinase three on Salvemini D et al. Oxidative stress and apoptosis interact and cause
the cell surface of human neutrophils: Quantication, catalyic activity emphysema due to vascular endothelial growth factor receptor block-
and susceptibility to inhibition. J Immunol 165: 336674, 2000. ade. Am J Respir Cell Mol Biol 29(1): 8897, 2003.
182. Cepinskas G, Sandig M, Kvietys PR. PAF induced elastase dependent 201. Hubbard RC, Fells G, Gadek J, Pacholok S, Humes J, Crystal RG.
neutrophil transendothelial migration is associated with the mobilisa- Neutrophil accumulation in the lung in a1-antitrypsin deciency;
tion of elastase to the neutrophil surface and localised to the migrat- spontaneous release of leukotriene B4 by alveolar macrophages. J Clin
ing front. J Cell Sci 112(Pt 12): 193745, 1999. Invest 88: 89197, 1991.
183. Fadok VA, Bratton DL, Guthrie L, Henson PM. Dierential eects 202. Shapiro SD. Animal models for COPD. Chest 117: 223S27S,
of apoptotic versus lysed cells on macrophage production of cytokines 2000.
Role of proteinases. J Immunol 166: 684754, 2001. 203. Kao RC, Wehner NG, Skubitz KM, Gray BH, Hoidal JR. A dis-
184. Ohlsson K, Linder C, Lundberg E, Axelsson L. Release of cytokines tinct human polymorphonuclear leukocyte proteinase that produces
and proteases from human peripheral blood mononuclear and poly- emphysema in hamsters. J Clin Invest 82: 196373, 1988.
morphonuclear cells following phagocytosis and LPS stimulation. 204. Stone PJ, Lucey EC, Virca GD, Christensen TG, Breuer R, Snider
Scand J Clin Lab Invest 56: 46170, 1996. GL. Alpha-1 protease inhibitor moderates human neutrophil elastase-
185. Russell REK, Thorley A, Culpitt SV, Dodd S, Donnelly LE, induced emphysema and secretory cell metaplasia in hamsters. Eur
Dermattos C et al. Alveolar macrophage-mediated elastolysis; roles of Respir J 3: 67378, 1990.
matrix metalloproteinases, cysteine and serine proteases. Am J Physiol 205. Lucey EC, Stone PJ, Ciccolella DE, Breuer R, Christensen TG,
Lung Cell Mol Physiol 283: L86773, 2002. Thompson RC et al. Recombinant human secretory leukocyte-
186. Weitz JL, Huang AJ, Landman SL, Nicholson SC, Silverstein SC. protease inhibitor; in vitro properties and amelioration of human
Elastase-mediated brinogenolysis by chemoattractant stimulated neutrophil elastase induced emphysema and secretory cell metaplasia.
neutrophils occurs in the presence of physiologic concentrations of J Lab Clin Med 115: 22432, 1990.
antiproteinases. J Exp Med 166: 183650, 1987. 206. Ofulue A, Ko M, Abboud R. Time course of neutrophil and macro-
187. Liou TG, Campbell EJ. Quantum proteolysis resulting from release of phage elastinolytic activities in cigarette induced emphysema. Am J
single granules by human neutrophils; a novel, nonoxidative mecha- Physiol Lung Cell Mol Physiol 275: L113444, 1998.
nism of extracellular proteolytic activity. J Immunol 157: 262431, 1996. 207. Zay K, Loo S, Xie C, Devine DV, Wright JL, Churg A. Role of neu-
188. Campbell EJ, Campbell MA, Boukedes SS, Owen CA. Quantum trophils and a1 antitrypsin in coal and silica-induced connective tissue
proteolysis by neutrophils; implications for pulmonary emphysema in breakdown. Am J Physiol Lung Cell Mol Physiol 276: L26979, 1999.
a1antitrypsin deciency. J Clin Invest 104: 33744, 1999. 208. Sapey E, Stockley RA. COPD exacerbations: Aetiology. Thorax 61(3):
189. Rice WG, Weiss SJ. Regulation of proteolysis at the neutrophilsub- 25058, 2006.
strate interface by secretory leukoprotease inhibitor. Science 249: 178 209. Dowson LJ, Guest PJ, Stockley RA. Longitudinal changes in
81, 1990. physiological, radiological and health status measurements in

190
The Neutrophil and Its Special Role in Chronic Obstructive Pulmonary Disease 14
alpha(1)-antitrypsin deciency and factors associated with decline. 229. Gibson PG, Simpson JL, Saltos N. Heterogeneity of airway inam-
Am J Respir Crit Care Med 164: 18059, 2001. mation in persistent asthma. Chest 119: 132936, 2001.
210. Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA. 230. Wenzel SE, Szeer SJ, Leung DYM et al. Bronchoscopic evaluation
Relationship between exacerbation frequency and lung function decline of severe asthma. Am J Respir Crit Care Med 156: 73743, 1997.
in chronic obstructive pulmonary disease. Thorax 57: 84752, 2002. 231. Jatakanon A, Uasuf C, Maziak W et al. Neutrophilic inammation
211. Churg A, Dai J, Tai H, Xie C, Wright JL. Tumour necrosis factor-a is in severe persistent asthma. Am J Respir Crit Care Med 160: 153237,
central to acute cigarette smoke-induced inammation and connec- 1999.
tive tissue breakdown. Am J Respir Crit Care Med 166: 84954, 2002. 232. Louis R, Lau LCK, Bron AO et al. The relationship between airway
212. Baker AH, Edwards DR, Murphy G. Metalloproteinase inhibitors: inammation and asthma severity. Am J Respir Crit Care Med 161:
Biological actions and therapeutic opportunities. J Cell Sci 115: 3719 916, 2000.
27, 2002. 233. Woodru PG, Khashayar R, Lazarus SC et al. Relationship between
213. Segura-Valdez L, Pardo A, Gaxiola M, Uhal BD, Beceril C, Selman airway inammation; hyperresponsiveness and obstruction in asthma.
M. Upregulation of gelatinases A and B, Collagenases 1 and 2 and J Allergy Clin Immunol 108: 75358, 2001.
increased parenchymal cell death in COPD. Chest 117: 68494, 2000. 234. Ordonez CL, Shaughnessy TE, Matthay MA, Fahy JV. Increased
214. Mao JT, Tashkin DP, Belloni PN, Baratelli F, Roth MD. All-trans neutrophil numbers and IL8 levels in secretions in acute severe
retinoic acid modulates the balance of matrix metalloproteinase-9 and asthma: clinical and biological signicance. Am J Respir Crit Care Med
tissue inhibitor of metalloproteinase-1 in patients with emphysema. 161: 118590, 2000.
Chest 124(5): 172432, 2003. 235. Martin RJ, Cicutto LC, Smith HR, Ballard RD, Szeer SJ. Airways
215. Beeh KM, Kornmann O, Buhl R. Sputum matrix metalloprotein- inammation in nocturnal asthma. Am Rev Respir Dis 143: 35157,
ase-9, tissue inhibitor of metalloproteinase-1 and their molar ratio in 1991.
patients with COPD, idiopathic pulmonary brosis and healthy con- 236. Carroll N, Carello S, Cooke C, James A. Airway structure and
trols. Respir Med 97(6): 63439, 2003. inammatory cells in fatal attacks of asthma. Eur Respir J 19: 70915,
216. Vignola AM, Bonanno A, Mirabella A, Riccobono L, Mirabella F, 1996.
Prota M et al. Increased levels of elastase and alpha-1 antitrypsin in 237. Sur S, Crotty TB, Kephart GM, Hyma BA, Colby TV, Reed CE,
sputum of asthmatic patients. Am J Respir Crit Care Med 157: 50511, Hunt LW, Gleich GJ. Sudden onset fatal asthma: distinct entity with
1998. few eosinophils and relatively more neutrophils in the airway submo-
217. Atkinson JJ, Senior RM. Matrix metalloproteinase-9 in lung remod- cosa? Am Rev Respir Dis 148: 71219, 1993.
elling. Am J Respir Cell Mol Biol 28(1): 1224, 2003. 238. Woodru PG, Fahy JV. A role for neutrophils in asthma? Am J Med
218. Li H, Cui D, Tong X, Ma N, Cui X, Lu L et al. The role of matrix 112: 498500, 2002.
metalloproteinases in extracellular matrix remodelling in chronic 239. Keatings VM, Jatakanon A, Worsdell YM, Barnes PJ. Eects of
obstructive pulmonary disease rat models. Zhonghua Nei Ke Za Zhi inhaled and oral glucocorticoids on inammatory indices in asthma
41(6): 39398, 2002. and COPD. Am J Respir Crit Care Med 155: 54248, 1997.
219. Selman M, Cisneros-Lira J, Gaxiola M, Ramirez R, Kudlacz EM, 240. Tillie-Leblond I, Gosset P, Tonnel AB. Inammatory events in severe
Mitchell PG et al. Matrix metalloproteinases inhibition attenuates acute asthma. Allergy 60: 2329, 2005.
tobacco smoke-induced emphysema in guinea pigs. Chest 123: 1633 241. Martin RJ, Chu HW, Honour JM, Harbeck RJ. Airway inammation
41, 2003. and bronchial hyper-responsiveness after mycoplasma pneumonia infec-
220. Minematsu N, Nakamura H, Tateno H, Nakajima T, Yagaguchi K. tion in a murine model. Am J Respir Crit Care Med 24: 57782, 2001.
Genetic polymorphisms in matrix metalloproteinase-9 and pulmo- 242. Castleman WL, Sorkness RL, Lemanske RF, McAllister PK. Viral
nary emphysema. Biochem Biophys Res Commun 289(1): 11619, 2001. bronchiolitis during early life induces increased numbers of bron-
221. Wallace AM, Sandford AJ. Genetic polymorphisms of matrix metal- chiolar mast cells and airway hyper-responsiveness. Am J Pathol 137:
loproteinases: Functional importance in the development of chronic 82131, 1990.
obstructive pulmonary disease? Am J Pharmacogenom 2(3): 16775, 243. ten Brinke A, van Dissel JT, Sterk PJ et al. Persistent airow limita-
2002. tion in adult-onset non-atopic asthma is associated with serological
222. Vernooy JH, Lindeman JH, Jacobs JA, Hanemaaijer R, Wouters EF. evidence of Chylamydia pneumoniae infection. J Allergy Clin Immunol
Increased activity of matrix metalloproteinase-8 and matrix metal- 107: 44954, 2001.
loproteinase-9 in induced sputum from patients with COPD. Chest 244. Black PN, Scicchitano R, Jenkins CR et al. Serological evidence of infec-
126(6): 180210, 2004. tion with Chylamydia pneumoniae is related to the severity of asthma. Eur
223. Itoh Y, Nagase H. Preferential inactivation of tissue inhibitor of met- Respir J 15: 25459, 2000.
alloproteinase-1 that is bound to the precursor of metalloproteinase- 245. Kraft M, Cassell GH, Henson JE et al. Detection of Mycoplasma
9 (progelatinase B) by human neutrophil elastase. J Biol Chem 270: pneumoniae in the airways of adults with chronic asthma. Am J Respir
1651821, 1995. Crit Care Med 158: 9981001, 1998.
224. Ferry G, Lonchamp M, Pennel L, de Nanteuil G, Canet E, Tucker 246. Bayram H, Devalia JL, Khair OA et al. Comparison of ciliary activity
GC. Activation of MMP-9 by neutrophil elastase in an in vivo model and inammatory mediator release from bronchial epithelial cels of
of acute lung injury. FEBS 402: 11115, 1997. non atopic nonasthmatic subjects and atopic asthmatic patients and
225. Desrochers PE, Je rey JJ, Weiss SJ. Interstitial collagenase (matrix the eects of diesel exhaust particles in vitro. J Allergy Clin Immunol
metalloproteinase-1) expresses serpinase activity. J Clin Invest 87(6): 102: 77182, 1998.
225865, 1991. 247. Linden A. Role of interleukin 17 and the neutrophil in asthma. Int
226. Hautamaki RD, Kobayashi DM, Senior RM, Shapiro SD. Arch Allergy Immunol 126: 17984, 2001.
Requirement for macrophage elastase for acute cigarette smoke- 248. Akk AM, Simmons PM, Chan HW, Agapov E, Holtzman MJ,
induced emphysema in mice. Science 277: 20024, 1997. Grayson MH, Pham CTN. Dipeptidyl peptidase I- dependent neu-
227. Churg A, Wang RD, Tai H, Wang X, Xie C, Wright JL. Tumour trophil recruitment modulates the inammatory response to Sendai
necrosis factor-alpha drives 70% of cigarette smoke induced emphy- virus infection. J Immunol 180: 353542, 2008.
sema in the mouse. Am J Respir Crit Care Med 170(5): 49298, 2004. 249. Je rey PK, Wardlow AJ, Nelson FC, Collins JV, Kay AB. Bronchial
228. Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, Wright JL. biopsies in ssthma: An ultrastructural quantitative study and correla-
Macrophage metalloelastase mediates acute cigarette smoke induced tion with hyperactivity. Am Rev Respir Dis 140: 174553, 1989.
inammation via tumor necrosis factor-alpha release. Am J Respir Crit 250. Kamath AV, Pavord ID, Ruparelia PR, Chilvers ER. Is the neutrophil
Care Med 167(8): 108389, 2003. the key eector cell in severe asthma? Thorax 60: 52930, 2005.

191
Fibroblasts
15 CHAPTER

INTRODUCTION FIBROBLAST FUNCTION Lynne A. Murray1, Darryl


A. Knight2 and Geoffrey
Fibroblasts are typically spindle-shaped cells Fibroblasts play a myriad of important roles in J. Laurent3
with an oval at nucleus found in the intersti- normal tissue function. In the lung, they coor- 1
Manager of Pharmocology Promedior,
tial spaces of organs. In the lung, they reside dinate organogenesis and budding of the lung
Inc, Malvern, PA, USA
in highly complex multicellular environments, from the foregut, through intimate bi-directional 2
Department of Pharmacology and
usually closely apposed to the epithelium or communication with adjacent epithelial cells.
endothelium. They are the primary source of They are also key cells in the production and Therapeutics, University of British
extracellular matrix (ECM) proteins, which, in homeostasis of the ECM. In the lung, the great- Columbia, Vancouver, BC, Canada
3
addition to providing a scaold for cells, play est number of broblasts are found in the sub- UCLMS, Center for Respiratory
key roles in determining cell phenotype and epithelial layer of the conducting airways and Research, University St/Rayne Institute,
function. In these contexts, broblasts contrib- the interstitium of the lung parenchyma. Here London, UK
ute to injury responses in both the initiation they are in a prime location to interact with the
and the resolution phases. epithelial and endothelial cells. These interactions
In chronic lung diseases including asthma, are likely important in disease settings and will be
chronic obstructive pulmonary disease (COPD), further discussed in this chapter. Fibroblasts are
and idiopathic pulmonary brosis (IPF), there metabolically active cells capable of synthesiz-
are changes in the number and phenotype of ing, secreting, and degrading ECM components
broblasts. These changes play a critical role in including collagens, proteoglycans, tenascin, lam-
the loss of normal tissue architecture and func- inin, and bronectin. These cells continually syn-
tion associated with these diseases. Although thesize ECM proteins although the amount they
considerable research eort has focused on secrete is tightly regulated. For example, up to
modulating leukocyte function and inamma- 90% of all procollagen molecules are degraded
tion, relatively few studies have investigated the intracellularly prior to secretion, depending on
eects of existing or novel therapies on brob- tissue and age. Further, broblasts generate matrix
last function. Moreover, there have not been metalloproteinases (MMPs) and their inhibitors,
sucient eorts to develop new approaches to tissue inhibitor of metalloproteinases (TIMPs),
regulate broblast function in these diseases. thus controlling tissue architecture and matrix
This chapter explores the current knowledge turnover rates.
about the role of broblasts in lung homeosta- MMPs exert proteolytic activities on vari-
sis and pathological disorders associated with ous proteins including many ECM components
chronic remodeling. A better understanding of and are thus central to ECM formation [1].
the phenotypes of disease-associated brob- They have been shown to be elevated in asthma
lasts may highlight pathways specic to disease and COPD (reviewed in [1]); as well as in IPF
pathologies, giving rise to targeted therapeutics. where MMP1, 2, and 9 were colocalized to the
Finally, we review the potential of opportunities epithelium surrounding brotic lesions, whereas
arising for better therapeutic intervention strat- increased TIMP2 was also observed suggest-
egies targeting broblasts that will either halt or ing that the MMP activity may be inhibited
potentially reverse brosis. and that the brotic region not degraded [2].

193
Asthma and COPD: Basic Mechanisms and Clinical Management

Another function of MMPs is to activate growth factors in vitro studies by using broblasts isolated from IPF lungs.
and chemokines, thus potentially promoting the brotic Fibroblasts isolated from brotic environments are pheno-
and inammatory milieu [35]. typically dierent from non-brotic broblasts [1719].
Furthermore, broblasts from a pro-brotic environment
exhibit both altered responsiveness to growth factors and
enhanced chemokine receptor expression, which has also been
MYOFIBROBLASTS observed in murine models of pulmonary remodeling [20].
Taken together, these studies suggest a distinct heterogeneity
in broblast function and phenotype in the brotic lung.
Myobroblasts express -smooth muscle actin (-SMA) The progression and the severity of many lung dis-
and have contractile and secretory properties that are central eases, notably IPF, are tightly associated with regions of
to controlling tissue architecture [6]. They express a panel broblast accumulation and proliferation; the extent of these
of markers that have been correlated with the site of origin. regions, termed broblastic foci, has become a reliable indi-
For example, myobroblasts found in the peripheral and cator of survival. The increased number of (myo)broblasts
subpleural regions of brosis express -SMA, vimentin, and seen in these diseases implies that they are hyperprolifera-
desmin, whereas cells found in other regions of the lung do tive and/or resistant to apoptosis. However, whether they
not express desmin [7]. In vitro, broblast-to-myobroblast proliferate faster than normal broblasts is still controver-
trans-dierentiation can be induced by transforming growth sial [17, 21, 22]. We and other investigators have reported
factor-1 (TGF-1), and it has been hypothesized that that broblasts derived from IPF lungs proliferate faster
TGF-1 found locally at sites of brosis trans-dierentiate than the cells derived from normal lung tissue [17]. In con-
resident broblasts into myobroblasts [8, 9]. trast, few others have also shown that the growth rate of
It has been proposed that the contractile properties
IPF broblasts was signicantly slower than that of normal
of myobroblasts are central to wound healing by limiting
broblasts. However, the specic techniques used to isolate
the amount of exposed wound area [10]. However, the sus-
primary broblasts dier between laboratories; therefore
tained presence of contractile myobroblasts in the inter-
direct comparisons may not be suitable.
stitium of the lung may cause a retraction of parenchymal
Moreover, discrepancies may be due to the site in the
tissue mediating alveolar collapse and resulting in the char-
lung from which broblasts are harvested, since the mag-
acteristic honeycombing seen in the lungs of IPF patients,
nitude of inammation and brosis are heterogeneous in
or add to the increase in alveolar size, which is character-
distribution. Thus, areas of active brosis may yield hyper-
istic of COPD [11]. There is also a signicant increase in
proliferative broblasts, compared to the areas of established
myobroblast numbers in the airways of asthmatic patients
brosis where cells may be hypoproliferative.
following allergen challenge [12], which correlate with sub-
To begin to address this diversity, recent studies have
epithelial collagen deposition [13].
used microarray technologies to prole global gene expression
In normal wound healing, myobroblasts sequentially
in pulmonary brosis in man and mouse models. These stud-
perpetuate and then dampen inammation via the secre-
ies have showed that the expression of almost 500 genes are
tion of chemokines, cytokines, arachidonic acid metabolites,
increased more than twofold in brotic lungs, including many
and protease inhibitors [14]. When activated, they express
genes related to cytoskeletal reorganization, ECM, cellular
cell surface adhesion molecules allowing specic interac-
metabolism and protein biosynthesis, signaling, proliferation,
tions with immune and inammatory cells, including lym-
and survival [8]. Studies examining human lung broblast
phocytes, mast cells, and neutrophils. If these processes
global gene expression in response to TGF-1 have shown
become dysregulated, brosis may ensue with catastrophic
almost 150 genes upregulated, representing several functional
consequences for lung function.
categories described earlier. These included 80 genes that
Given the importance of myobroblasts to wound
were not previously known to be TGF-1-responsive [8].
healing and brosis, dening both the origin and the mech-
There was excellent concordance between gene expression in
anisms leading to their clearance will greatly add to our
human and experimental models, giving us some condence
understanding of the role of this cell in brotic diseases.
in the value of our eorts to model human disease.
At sites of normal wound healing, once sucient ECM
has been deposited and remodeled, broblasts and myo-
broblasts undergo apoptosis [15, 16]. This serves to limit
the excessive deposition of ECM and also dampen the pro-
inammatory and pro-brotic milieu. However, for reasons
still unclear, myobroblasts persist in brotic conditions.
ORIGIN OF FIBROBLASTS AND
MYOFIBROBLASTS: PLASTICITY OF
RESIDENT FIBROBLASTS
FIBROBLASTS AND FIBROSIS As remodeling of the lung is associated with the accumula-
tion of broblasts and myobroblasts, understanding the der-
Most insight into the potential role of broblasts at driv- ivation of these cells is critical to our understanding of disease
ing pulmonary remodeling, as well as phenotypic dif- processes. Current thinking suggests that there may be mul-
ferences in broblasts found in brotic regions versus tiple pathways through which broblasts and myobroblasts
those located in normal tissue, has been garnered from are derived [14]. These include proliferation and plasticity of

194
Fibroblasts 15
resident cells, such as broblasts and epithelial cells. Further, undergo cytoskeletal changes as well as altered expression
emerging data highlight a signicant role of circulating, bone of surface molecules. For example, a downregulation of
marrow-derived cells at remodeling lung architecture [23]. E-cadherin and zona occludens 1 (ZO-1) with a concomi-
tant upregulation of vimentin as well as the ED-A bronec-
tin is needed for migration and transition to a mesenchymal
phenotype [36, 37]. The majority of the work evaluating
FACTORS INVOLVED IN FIBROBLAST EMT has been performed in vitro; however, the full extent
MYOFIBROBLAST DIFFERENTIATION of this pathogenic pathway in vivo is currently being evalu-
ated. In animal models of kidney brosis, it has been esti-
mated that up to 20% of the broblasts found in the brotic
As has been described, broblasts can be induced to dif- lesions were derived from the epithelium through EMT
ferentiate into myobroblasts. It is becoming increasingly [33]. The idea of EMT promoting the brosis observed in
accepted that the broblastmyobroblast transition begins asthma and IPF is rapidly beginning to evolve [32]. It is still
with the appearance of the protomyobroblasts that are not known whether EMT contributes to the excess ECM
hyperproliferative and migratory but do not synthesize deposition. Future work correlating the timecourse of EMT
signicant amounts of ECM proteins [24]. Under specic induction with disease staging will also be very insightful.
conditions, the protomyobroblast evolves and may result This pathogenic process may provide novel therapeutic tar-
in a transition into a dierentiated myobroblast, charac- gets such that inhibiting or reversing EMT may provide
terized by the presence of organized stress bers containing clinical benet to patients with brosis-associated diseases.
-SMA. Myobroblasts can, according to the experimental
or clinical situation, express other smooth muscle cell con-
tractile proteins, such as myosin heavy chains or desmin.
Generally, these cells are thought to be hypoproliferative but CIRCULATING PROGENITOR
responsible for secretion of the bulk of collagens I and III.
Although the processes involved in the appearance of
MESENCHYMAL CELLS
protomyobroblasts are at present not well explored, the tran-
sition from the protomyobroblast to a dierentiated myo- Along with the epithelium, recent studies have also high-
broblast has been related to the production of TGF-1 by lighted a role for bone marrow-derived circulating cells, or
inammatory cells, and possibly by broblasts themselves. brocytes, in promoting lung brosis by dierentiating into
The action of TGF-1 also depends on the local presence of broblasts or myobroblasts [23, 38].
the cellular bronectin ED-A splice variant. Thus, myobrob- Fibrocytes appear to be pleiotropic in function and a
last dierentiation is regulated by both a cell product and the variety of extracellular and intracellular markers have been
ECM. Moreover, it is becoming more accepted that mechani- used to characterize them, including CD45, indicating
cal factors play an important role in both transitions through haematopoietic origin; CD34 or CD13; and various chem-
either TGF- generation [25] or TGF- activation [26]. okine receptors which have been demonstrated to modu-
late migration in vitro and in vivo [3943]. However, the
underlying features of brocytes are that they are derived
from the bone marrow and are positive for type I collagen.
EPITHELIAL CELLS: EPITHELIAL Moreover, it has recently been postulated that an over-
MESENCHYMAL TRANSITION exuberant recruitment of these cells to sites of pulmonary
injury contributes to the aberrant deposition of collagen,
which ultimately induces pathologic brosis [4448].
Another potential pool of broblasts may arise by a proc- Fibrocytes are pleuripotent in their ability to dieren-
ess called epithelialmesenchymal transition (EMT). EMT tiate into other cell lineages, as has been demonstrated with
is a dynamic process by which epithelial cells undergo phe- brocyte-derived adipocytes [49]. Furthermore, these cells
notypic transition to fully dierentiated and motile mes- are extremely plastic in vitro, making both the derivation
enchymal cells, such as broblasts and myobroblasts [27, and the characterization of these cells unclear. Exposure of
28]. This process occurs normally during early fetal devel- brocytes to TGF-1 in vitro results in the cells transition-
opment where there is seamless plasticity between epithelial ing into a myobroblast phenotype and producing bronec-
and mesenchymal cells. The dierentiation between airway tin and type III collagen [48]. Using an adoptive transfer
epithelial cells of one type and another, for example type I model of bone marrow cells from green uorescent protein
pneumocytes transitioning into goblet cells, has been previ- (GFP) transgenic mice into recipient mice challenged with
ously described [2932]. However, the switching of an epi- intratracheal bleomycin (to initiate lung injury), recruited
thelial cell into a phenotype that moves beyond the original GFP brocytes were shown to dierentiate into brob-
cells embryonic lineage has recently been hypothesized as a lasts while resident lung broblasts dierentiated into
driving factor in brosis [3335]. myobroblasts [39]. These data would indicate that the
Epithelial cells exposed to TGF-1, alone or in brocytes may serve to replenish the pool of broblasts
combination with other growth factors such as epider- from which myobroblasts are derived.
mal growth factor (EGF), begin the process of EMT by There is increasing evidence that brocytes may also
the increased expression of MMPs that enable basement have a pathogenic role in asthma [41, 48]. A correlation
membrane degradation and cell detachment. The cells also in the number of brocytes in the basement membrane of

195
Asthma and COPD: Basic Mechanisms and Clinical Management

asthmatics and the extent of subepithelial brosis has been TGF-1 is upregulated in the lungs of IPF patients
recently described [50]. Increased numbers of these cells and asthmatics [5156] (reviewed in [57]). Interestingly,
have been reported in the airways of asthmatics and aller- expression of TGF-1 is nearly absent in the bronchial epi-
gen challenge further increased cell recruitment to the sub- thelial cells but is highly expressed in inammatory cells
epithelial region [48]. Interestingly, CD34 colocalized with beneath the basement membrane where subepithelial bro-
-SMA, suggesting that CD34 brocytes trac to sites sis predominates [58]. Transient over-expression of TGF-1
of active remodeling and dierentiate into myobroblast- or pulmonary delivery of this cytokine to mouse lungs
like cells [48]. Further studies are necessary to delineate the induces a pronounced interstitial brosis mediated by aber-
fate of brocytes and the total impact of this pathway to rant ECM generation and deposition, as well as the presence
other diseases such as COPD. of myobroblasts [59]. Further, over-expression of TGF-1
in the lungs of mice also induces a brotic response through
modulation of apoptotic and inammatory pathways, as well
as aberrant expression of MMP12 [60, 61]. Epithelial cells
FIBROBLAST ACTIVATION and eosinophils also produce large amounts of TGF-2,
especially following allergen challenge [6265]. However,
Fibroblasts are activated by numerous signals such as mechan- the role in remodeling and brosis is not as well studied as
ical forces imposed during bronchoconstriction, ECM interac- that of TGF-1, although it has been shown to induce epi-
tions, and hypoxia. Furthermore, a large number of mediators, thelial cell mucus production [63] and exert pro-brogenic
produced by various cell types, and proteases of the coagula- eects [66].
tion cascade are known to promote broblast proliferation, TGF-1 regulates numerous biological activities, such
collagen synthesis, migration, and dierentiation. as proliferation, apoptosis, and dierentiation via Smad-
dependant and Smad-independent mechanisms [67, 68]
(Fig. 15.1). TGF- is produced in an inactive form tethered
Transforming growth factor to a latency-associated peptide (LAP) by covalent bonding.
Activation of, and signaling through, its receptors requires
TGF- is one of the most potent pro-brotic mediators exposure of the active site of the ligand either through con-
in vitro and a strong candidate as a central player in remod- formational change (as induced by integrin-mediated acti-
eling diseases including asthma and IPF. The emergence of vation) or through cleavage of the LAP.
therapeutics directed against the TGF- pathway will shed Using a transgenic mouse model of Smad-3 de-
light on the role of this growth factor in diseases. Although ciency, TGF-/Smad-3 signaling was shown to be required
antagonism of TGF- by a number of strategies ameliorates for alveolar integrity and ECM homeostasis, and this path-
experimental brosis, inhibition of this growth factor family way is involved in pathogenic mechanisms mediating tissue
as a valid therapeutic target requires rigorous interpretation, destruction and brogenesis [69]. TGF- signaling pathway
since it plays an important role as an inhibitor of immune involves the phosphorylation of downstream Smad proteins,
responses and normal cell dierentiation. comprising the receptor-regulated (R)-Smad (Smad 2, 3), the

Smad-Dependent TGF Signaling

P
TGFRII II I Cytoplasm
R-Smad

ALK 1 ALK 5
Pathway pathway

P P
R-Smad R-Smad
Smad 1 Co-Smad
Smad 2
Smad 5 Smad 4 Smad 3
Smad 8 FIG. 15.1 TGF- signaling pathways. TGF-
is believed to be a major signaling pathway
I-Smad
regulating fibroblast functions. All three
TGF- isoforms bind to the TGF-RII which
Smad 6
P then phosphorylates the TGFRI chain. This
Smad 7
Co-Smad R-Smad activates the kinase activity of TGF-RI, which
then activates Smad proteins. These, in turn,
bind Smad 4, translocate to the nucleus, and
Nucleus modulate gene expression. Inhibitory Smads,
P
Co-Smad R-Smad including Smad 6 and 7, can inhbibit Smad
DNA
Binding
signaling. TGF- can also signal through a
partner
P
variety of Smad independent pathways.
DNA
Co-Smad R-Smad Activation of these pathways may occur
Binding through the kinase activity of either TGF-RI
partner Target gene or TGF-RII.

196
Fibroblasts 15
co-Smad, Smad 4). These activated Smad complexes translo- diseases where brosis is often a feature. These include acute
cate to the nucleus, bind to specic consensus sequences on respiratory distress syndrome (ARDS), and the interstitial
target DNA to upregulate the transcription of many genes. lung diseases including IPF [8993]. In one of these stud-
Regulation of this pathway occurs through the synthesis of ies, thrombin was shown to represent a large proportion of
the inhibitory Smad-7. Although Smad signaling is gener- the broblast proliferation capacity of bronchoalveolar lav-
ally considered the predominant pathway, Smad-independ- age (BAL) uid in patients with lung brosis, suggesting
ent signaling through P38, Akt/PI-3 K, and extracellular that it might be a major player [91]. Animal models have
signal-regulated kinase (ERK) pathways is also observed. strengthened the connection between these proteases and
These pathways generally lead to dierent cellular responses the occurrence of brosis. For example, increased thrombin
[70, 71]. In this regard, while Smad signaling generally arises is found in the lungs of mice challenged with bleomycin
through activation of the type I TGF- receptor, P38 signal- and pharmacological inhibition of thrombin signicantly
ing occurs through activation of the type II receptor, imply- reduced the collagen deposition [90, 94].
ing dierent mechanisms of activation and regulation [70]. It is now clear that proteases of the coagulation cas-
cade, including factor VIIa, factor Xa, and thrombin, exert
IL-13 and IL-4 pro-inammatory and pro-brotic eects via the pro-
Interleukin (IL)-13 and IL-4 are pleiotropic, Th2-associated tease-activated receptors (PAR). The PAR comprise four
cytokines,with numerous distinct and overlapping func- members, PAR1 to PAR4 and between them the coagula-
tions. They share overlapping but not redundant roles due tion proteases of the extrinsic pathway can target all four
to the shared IL13R1 receptor subunit. IL-13 is elevated receptors. In terms of inuencing broblast function, PAR1,
in the lungs of IPF patients and is associated with brotic the high anity thrombin receptor, is the major receptor
pathologies and aberrant remodeling at various tissue sites by which thrombin and factor Xa exert their potent pro-
[72, 73]. The pro-brotic role for IL-13 in asthma and pul- brotic eects. This receptor has emerged as a promising
monary brosis has been recently reviewed [74, 75]. There new target to prevent brosis in the setting of both IPF and
are dierences in the downstream events following IL-4 or ARDS, based on studies demonstrating that thrombin inhi-
IL-13 signaling. IL-13 activates epithelial cells and goblet bition partially blocks experimental brosis [95] and that
cells causing mucous production, goblet cell hyperplasia and mice decient for PAR1 are protected from lung inam-
EMT [76, 77]. Various animal models of pulmonary brosis mation, pulmonary edema, and lung collagen accumulation
have indicated a more pro-brotic role for IL-13 than IL-4. following bleomycin injury [96]. Recently, the role of PAR
Indeed it has been hypothesized that IL-4 is involved in the has been linked to epithelial cell activation of TGF-. It
initiation of brosis whereas IL-13 is central to the mainte- has been known for some time that mice decient for the
nance of the brotic response, reviewed in Chapter 8 [78]. 6 subunit are protected from pulmonary brosis due to a
Both IL-13 and IL-4 induce pro-brotic responses in defective ability to activate TGF- [97] but recent stud-
broblasts, responses that are comparable to those induced ies suggest that activation of PAR1 leads to the activation
by TGF-1 [79, 80]. Both IL-4 and IL-13 induce broblast of latent TGF- via this v6-dependent mechanism [94].
proliferation [81]. Interestingly, IL-4 and IL-13 induced Together these data suggest thrombin may be upstream to
greater broblast proliferation from patients with mild the activation of TGF-.
asthma compared to responses elicited on broblasts derived The PAR have also been implicated in airways dis-
from severe asthma patients [82]. This suggests an altered eases (reviewed in [98100]). The mechanism for this eect
broblast phenotype which is dependent on disease stag- is still being explored but it may be due to pro-inammatory
ing. IL-13 and IL-4 have also been demonstrated to play eects of PAR activation. For example, PAR-2-decient
pro-brotic roles on airway epithelial cells by promoting mice had decreased eotaxin/CCL11 inux along with
epithelial cell proliferation and also by inducing mitogenic reduced eosinophilia following antigen challenge in an
TGF-2 production from these cells [83]. Interestingly, allergen sensitization and challenge model of asthma [101,
IL-13 and not IL-4 promotes the dierentiation of brob- 102]. In relation to COPD, Churg and coworkers recently
lasts to myobroblasts [81, 82]. The other mechanism by suggested that 1-antitrypsin inhibition of TNF- and
which IL-13 can mediate brosis is via the second IL-13 MMP12 production in animals exposed to cigarette smoke
receptor, IL-13R2 [84]. This receptor was, until very may in part be due to inhibition of serine proteases activat-
recently, thought to be a decoy receptor as it has a very short ing PAR1 on macrophages [103].
cytoplasmic tail and is frequently shed from the surface of
cells [85, 86]. However IL-13 binds to IL-13R2 with a
much higher anity than IL-13R1 [87, 88]. Recent data
suggests that IL-13R2 has signaling capabilities resulting
in TGF-1 secretion from macrophages [84]. CLINICAL TARGETING OF FIBROSIS
IN LUNG PATHOLOGIES
Thrombin and coagulation Currently there are no therapeutic molecules in use in clini-
pathway-associated proteases cal respiratory medicine that specically target broblasts
or broblast function. Further, myobroblasts are likely to
Thrombin and several other proteases of the coagulation be a key to the chronicity of IPF and other related diseases
pathway have been implicated in a number of pulmonary and therefore strategies that modulate the functions of

197
Asthma and COPD: Basic Mechanisms and Clinical Management

these cells may have a signicant impact on the treatment 16. Darby I, Skalli O, Gabbiani G. Alpha-smooth muscle actin is tran-
of brotic remodeling. siently expressed by myobroblasts during experimental wound heal-
Data emerging suggests that targeting-specic media- ing. Lab Invest 63(1): 2129, 1990.
17. Moodley YP et al. Fibroblasts isolated from normal lungs and those with
tors during asthma, such as IL-13, may impact disease
idiopathic pulmonary brosis dier in interleukin-6/gp130-mediated
through modulating broblast function. Moreover, increased cell signaling and proliferation. Am J Pathol 163(1): 34554, 2003.
pathogenic functions ascribed to broblasts indicate that this 18. Scadi AK et al. Oncostatin M stimulates proliferation, induces col-
cell type may be attractive to target therapeutically. Aberrant lagen production and inhibits apoptosis of human lung broblasts.
activation of broblasts during brosis results in excessive Br J Pharmacol 136(5): 793801, 2002.
collagen deposition, which ultimately results in a loss of tis- 19. Keerthisingam CB et al. Cyclooxygenase-2 deciency results in a loss
sue function. As more is learnt about broblasts and cells of the anti-proliferative response to transforming growth factor-beta in
capable of generating excess ECM, specic cell markers human brotic lung broblasts and promotes bleomycin-induced pul-
may be identied that could allow for a selective targeting monary brosis in mice. Am J Pathol 158(4): 141122, 2001.
with minimal detriment to normal tissue. Therefore, target- 20. Hogaboam CM et al. Dierential monocyte chemoattractant protein-1
and chemokine receptor 2 expression by murine lung broblasts
ing broblasts directly may result in a decrease in ECM syn-
derived from Th1- and Th2-type pulmonary granuloma models.
thesis and a potential benecial clinical outcome, but unlike J Immunol 163(4): 2193201, 1999.
immunosuppressants and non-specic anti-inammatory 21. Jacoby DB, Gleich GJ, Fryer AD. Human eosinophil major basic pro-
agents, novel therapeutics would leave cellular defense mech- tein is an endogenous allosteric antagonist at the inhibitory muscarinic
anisms necessary for host defense in the lung intact. M2 receptor. J Clin Invest 91(4): 131418, 1993.
22. Ramos C et al. Fibroblasts from idiopathic pulmonary brosis and
normal lungs dier in growth rate, apoptosis, and tissue inhibitor of
metalloproteinases expression. Am J Respir Cell Mol Biol 24(5): 59198,
References 2001.
23. Quan TE, Cowper SE, Bucala R. The role of circulating brocytes in
1. Demedts IK et al. Matrix metalloproteinases in asthma and COPD. brosis. Curr Rheumatol Rep 8(2): 14550, 2006.
Curr Opin Pharmacol 5(3): 25763, 2005. 24. Desmouliere A, Darby IA, Gabbiani G. Normal and pathologic soft
2. Fukuda Y et al. Localization of matrix metalloproteinases-1, -2, and -9 tissue remodeling: Role of the myobroblast, with special emphasis on
and tissue inhibitor of metalloproteinase-2 in interstitial lung diseases. liver and kidney brosis. Lab Invest 83(12): 1689707, 2003.
Lab Invest 78(6): 68798, 1998. 25. Lindahl GE et al. Activation of broblast procollagen alpha 1(I)
3. Li Q et al. Matrilysin shedding of syndecan-1 regulates chemokine transcription by mechanical strain is transforming growth factor-
mobilization and transepithelial eux of neutrophils in acute lung beta-dependent and involves increased binding of CCAAT-binding fac-
injury. Cell 111(5): 63546, 2002. tor (CBF/NF-Y) at the proximal promoter. J Biol Chem 277(8): 615361,
4. Ludbrook SB et al. The integrin alphavbeta3 is a receptor for the 2002.
latency-associated peptides of transforming growth factors beta1 and 26. Sheppard D. Transforming growth factor beta: A central modulator of
beta3. Biochem J 369(Pt 2): 31118, 2003. pulmonary and airway inammation and brosis. Proc Am Thorac Soc
5. Sheppard D. Roles of alphav integrins in vascular biology and pulmo- 3(5): 41317, 2006.
nary pathology. Curr Opin Cell Biol 16(5): 55257, 2004. 27. Zavadil J, Bottinger EP. TGF-beta and epithelial-to-mesenchymal
6. Ohta K et al. Immunohistochemical identication and characterization transitions. Oncogene 24(37): 576474, 2005.
of smooth muscle-like cells in idiopathic pulmonary brosis. Am J Respir 28. Zavadil J et al. Integration of TGF-beta/Smad and Jagged1/Notch
Crit Care Med 152(5 Pt 1): 165965, 1995. signalling in epithelial-to-mesenchymal transition. EMBO J 23(5):
7. Zhang K et al. Myobroblasts and their role in lung collagen gene 115565, 2004.
expression during pulmonary brosis. A combined immunohistochem- 29. Borok Z et al. Modulation of t1alpha expression with alveolar epithelial
ical and in situ hybridization study. Am J Pathol 145(1): 11425, 1994. cell phenotype in vitro. Am J Physiol 275(1 Pt 1): L155L164, 1998.
8. Chambers RC et al. Global expression proling of broblast responses 30. Danto SI et al. Reversible transdierentiation of alveolar epithelial
to transforming growth factor-beta1 reveals the induction of inhibitor cells. Am J Respir Cell Mol Biol 12(5): 497502, 1995.
of dierentiation-1 and provides evidence of smooth muscle cell phe- 31. Torday JS, Torres E, Rehan VK. The role of broblast transdieren-
notypic switching. Am J Pathol 162(2): 53346, 2003. tiation in lung epithelial cell proliferation, dierentiation, and repair in
9. Desmouliere A et al. Transforming growth factor-beta 1 induces alpha- vitro. Pediatr Pathol Mol Med 22(3): 189207, 2003.
smooth muscle actin expression in granulation tissue myobroblasts 32. Kim KK et al. Alveolar epithelial cell mesenchymal transition develops
and in quiescent and growing cultured broblasts. J Cell Biol 122(1): in vivo during pulmonary brosis and is regulated by the extracellular
10311, 1993. matrix. Proc Natl Acad Sci USA 103(35): 1318085, 2006.
10. Gabbiani G, Badonnel MC. Contractile events during inammation. 33. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its
Agents Actions 6(13): 27780, 1976. implications for brosis. J Clin Invest 112(12): 177684, 2003.
11. Dacic S, Yousem SA. Histologic classication of idiopathic chronic 34. Selgas R, Jimenez-Heernan J, Lopez-Cabrera M. On the epithelial
interstitial pneumonias. Am J Respir Cell Mol Biol 29(3 Suppl): S59, mesenchymal transition of mesothelial cells. Kidney Int 66(2): 86667,
2003. 2004.
12. Gizycki MJ et al. Myobroblast involvement in the allergen-induced 35. Valcourt U et al. TGF-beta and the Smad signaling pathway support
late response in mild atopic asthma. Am J Respir Cell Mol Biol 16(6): transcriptomic reprogramming during epithelialmesenchymal cell
66473, 1997. transition. Mol Biol Cell 16(4): 19872002, 2005.
13. Brewster CE et al. Myobroblasts and subepithelial brosis in bron- 36. Grunert S, Jechlinger M, Beug H. Diverse cellular and molecular
chial asthma. Am J Respir Cell Mol Biol 3(5): 50711, 1990. mechanisms contribute to epithelial plasticity and metastasis. Nat Rev
14. Hinz B et al. The myobroblast: One function, multiple origins. Am J Mol Cell Biol 4(8): 65765, 2003.
Pathol 170(6): 180716, 2007. 37. Iwano M et al. Evidence that broblasts derive from epithelium during
15. Desmouliere A et al. Apoptosis mediates the decrease in cellularity tissue brosis. J Clin Invest 110(3): 34150, 2002.
during the transition between granulation tissue and scar. Am J Pathol 38. Quan TE et al. Circulating brocytes: Collagen-secreting cells of the
146(1): 5666, 1995. peripheral blood. Int J Biochem Cell Biol 36(4): 598606, 2004.

198
Fibroblasts 15
39. Hashimoto N et al. Bone marrow-derived progenitor cells in pulmo- 65. Thompson HG et al. Epithelial-derived TGF-beta2 modulates basal
nary brosis. J Clin Invest 113(2): 24352, 2004. and wound-healing subepithelial matrix homeostasis. Am J Physiol Lung
40. Moore BB et al. The role of CCL12 in the recruitment of brocytes Cell Mol Physiol 291(6): L127785, 2006.
and lung brosis. Am J Respir Cell Mol Biol 35(2): 17581, 2006. 66. Puddicombe SM et al. Involvement of the epidermal growth factor
41. Phillips RJ et al. Circulating brocytes trac to the lungs in response receptor in epithelial repair in asthma. FASEB J 14(10): 136274, 2000.
to CXCL12 and mediate brosis. J Clin Invest 114(3): 43846, 2004. 67. Pinkas J, Teicher BA. TGF-beta in cancer and as a therapeutic target.
42. Moore BB et al. The role of CCL12 in the recruitment of brocytes Biochem Pharmacol 72(5): 52329, 2006.
and lung brosis. Am J Respir Cell Mol Biol, 2006. 68. Sanders YY, Kumbla P, Hagood JS. Enhanced myobroblastic dier-
43. Moore BB et al. CCR2-mediated recruitment of brocytes to the alve- entiation and survival in Thy-1(-) lung broblasts. Am J Respir Cell Mol
olar space after brotic injury. Am J Pathol 166(3): 67584, 2005. Biol, 2006.
44. Abe R et al. Peripheral blood brocytes: Dierentiation pathway and 69. Bonniaud P et al. Smad3 null mice develop airspace enlargement and
migration to wound sites. J Immunol 166(12): 755662, 2001. are resistant to TGF-beta-mediated pulmonary brosis. J Immunol
45. Chesney J, Bucala R. Peripheral blood brocytes: Mesenchymal pre- 173(3): 2099108, 2004.
cursor cells and the pathogenesis of brosis. Curr Rheumatol Rep 2(6): 70. Shi Y, Massague J. Mechanisms of TGF-beta signaling from cell
5015, 2000. membrane to the nucleus. Cell 113(6): 685700, 2003.
46. Chesney J et al. Regulated production of type I collagen and inam- 71. Scadi AK et al. Regulation of human lung broblast phenotype and
matory cytokines by peripheral blood brocytes. J Immunol 160(1): function by vitronectin and vitronectin integrins. J Cell Sci 114(Pt 19):
41925, 1998. 350716, 2001.
47. Bucala R et al. Circulating brocytes dene a new leukocyte subpopu- 72. Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev
lation that mediates tissue repair. Mol Med 1(1): 7181, 1994. Immunol 4(8): 58394, 2004.
48. Schmidt M et al. Identication of circulating brocytes as precursors of 73. Hancock A et al. Production of interleukin 13 by alveolar macrophages
bronchial myobroblasts in asthma. J Immunol 171(1): 38089, 2003. from normal and brotic lung. Am J Respir Cell Mol Biol 18(1): 6065,
49. Hong KM et al. Characterization of human brocytes as circulating 1998.
adipocyte progenitors and the formation of human adipose tissue in 74. Wills-Karp M. Interleukin-13 in asthma pathogenesis. Immunol Rev
SCID mice. FASEB J 19(14): 202931, 2005. 202: 17590, 2004.
50. Nihlberg K et al. Tissue brocytes in patients with mild asthma: 75. Finkelman FD et al. Interleukin-4- and interleukin-13-mediated host
A possible link to thickness of reticular basement membrane? Respir protection against intestinal nematode parasites. Immunol Rev 201:
Res 7: 50, 2006. 13955, 2004.
51. Coker RK et al. Diverse cellular TGF-beta 1 and TGF-beta 3 gene 76. Yasuo M et al. Relationship between calcium-activated chloride chan-
expression in normal human and murine lung. Eur Respir J 9(12): nel 1 and MUC5AC in goblet cell hyperplasia induced by interleukin-
25017, 1996. 13 in human bronchial epithelial cells. Respiration 73(3): 34759, 2006.
52. Khalil N et al. TGF-beta 1, but not TGF-beta 2 or TGF-beta 3, is dif- 77. Atherton HC, Jones G, Danahay H. IL-13-induced changes in the
ferentially present in epithelial cells of advanced pulmonary brosis: An goblet cell density of human bronchial epithelial cell cultures: MAP
immunohistochemical study. Am J Respir Cell Mol Biol 14(2): 13138, kinase and phosphatidylinositol 3-kinase regulation. Am J Physiol Lung
1996. Cell Mol Physiol 285(3): L73039, 2003.
53. Khalil N et al. Increased production and immunohistochemical locali- 78. Murray LA et al. Fibroblasts and the Extracellular Matrix, in Allergy
zation of transforming growth factor-beta in idiopathic pulmonary and Allergic Diseases. In: Kay AB, Kaplan AP, Bousquet J, Holt PG,
brosis. Am J Respir Cell Mol Biol 5(2): 15562, 1991. (eds). 2nd edition. Blackwell Publishing Limited, 412435, 2008.
54. Yoshida K, Gage FH. Cooperative regulation of nerve growth fac- 79. Oriente A et al. Interleukin-13 modulates collagen homeostasis in human
tor synthesis and secretion in broblasts and astrocytes by broblast skin and keloid broblasts. J Pharmacol Exp Ther 292(3): 98894, 2000.
growth factor and other cytokines. Brain Res 569(1): 1425, 1992. 80. Hashimoto S et al. IL-4 and IL-13 induce myobroblastic pheno-
55. Okumura KK et al. Cortisol and TGF-beta inhibit secretion of type of human lung broblasts through c-Jun NH2-terminal kinase-
platelet-activating factor-acetylhydrolase in a monocyte-macrophage dependent pathway. J Allergy Clin Immunol 107(6): 10018, 2001.
model system [corrected]. Mol Hum Reprod 3(11): 92732, 1997. 81. Saito A et al. Potential action of IL-4 and IL-13 as brogenic factors
56. Vignola AM et al. Transforming growth factor-beta expression in on lung broblasts in vitro. Int Arch Allergy Immunol 132(2): 16876,
mucosal biopsies in asthma and chronic bronchitis. Am J Respir Crit 2003.
Care Med 156(2 Pt 1): 59199, 1997. 82. Kraft M et al. IL-4, IL-13, and dexamethasone augment broblast pro-
57. Howell JE, McAnulty RJ. TGF-beta: Its role in asthma and therapeu- liferation in asthma. J Allergy Clin Immunol 107(4): 6026, 2001.
tic potential. Curr Drug Targets 7(5): 54765, 2006. 83. Richter A et al. The contribution of interleukin (IL)-4 and IL-13 to
58. Magnan A et al. Altered compartmentalization of transforming growth the epithelial-mesenchymal trophic unit in asthma. Am J Respir Cell
factor-beta in asthmatic airways. Clin Exp Allergy 27(4): 38995, 1997. Mol Biol 25(3): 38591, 2001.
59. Sime PJ et al. Adenovector-mediated gene transfer of active transform- 84. Fichtner-Feigl S et al. IL-13 signaling through the IL-13alpha2 recep-
ing growth factor-beta1 induces prolonged severe brosis in rat lung. tor is involved in induction of TGF-beta1 production and brosis. Nat
J Clin Invest 100(4): 76876, 1997. Med 12(1): 99106, 2006.
60. Kang HR et al. TGF-beta 1 stimulates pulmonary brosis and inam- 85. Gauchat JF et al. A novel 4-kb interleukin-13 receptor alpha mRNA
mation via a bax-dependent, bid-activated pathway that involves matrix expressed in human B, T, and endothelial cells encoding an alternate
metalloproteinase-12. J Biol Chem, 2007. type-II interleukin-4/interleukin-13 receptor. Eur J Immunol 27(4):
61. Lee CG et al. Early growth response gene 1-mediated apoptosis is 97178, 1997.
essential for transforming growth factor {beta}1-induced pulmonary 86. Orchansky PL et al. An interleukin (IL)-13 receptor lacking the cyto-
brosis. J Exp Med 200(3): 37789, 2004. plasmic domain fails to transduce IL-13-induced signals and inhibits
62. Torrego A et al. Expression and activation of TGF-beta isoforms in acute responses to IL-4. J Biol Chem 272(36): 2294047, 1997.
allergen-induced remodelling in asthma. Thorax 62(4): 30713, 2007. 87. Rahaman SO et al. IL-13R(alpha)2, a decoy receptor for IL-13 acts
63. Chu HW et al. Transforming growth factor-beta2 induces bronchial epi- as an inhibitor of IL-4-dependent signal transduction in glioblastoma
thelial mucin expression in asthma. Am J Pathol 165(4): 1097106, 2004. cells. Cancer Res 62(4): 11039, 2002.
64. Tschumperlin DJ et al. Mechanical stress triggers selective release of 88. Wu AH, Low WC. Molecular cloning and identication of the human
brotic mediators from bronchial epithelium. Am J Respir Cell Mol Biol interleukin 13 alpha 2 receptor (IL-13Ra2) promoter. Neuro-oncol 5(3):
28(2): 14249, 2003. 17987, 2003.

199
Asthma and COPD: Basic Mechanisms and Clinical Management

89. Ludwicka-Bradley A, Bogatkevich G, Silver RM. Thrombin-mediated 96. Howell DC et al. Absence of proteinase-activated receptor-1 signaling
cellular events in pulmonary brosis associated with systemic sclerosis aords protection from bleomycin-induced lung inammation and
(scleroderma). Clin Exp Rheumatol 22(3 Suppl 33): S3846, 2004. brosis. Am J Pathol 166(5): 135365, 2005.
90. Howell DC, Laurent GJ, Chambers RC. Role of thrombin and its 97. Munger JS et al. The integrin alpha v beta 6 binds and activates latent
major cellular receptor, protease-activated receptor-1, in pulmonary TGF beta 1: A mechanism for regulating pulmonary inammation
brosis. Biochem Soc Trans 30(2): 21116, 2002. and brosis. Cell 96(3): 31928, 1999.
91. Hernandez-Rodriguez NA et al. Role of thrombin in pulmonary bro- 98. Lan RS, Stewart GA, Henry PJ. Role of protease-activated receptors
sis. Lancet 346(8982): 107173, 1995. in airway function: A target for therapeutic intervention? Pharmacol
92. Ohba T et al. Scleroderma bronchoalveolar lavage uid contains Ther 95(3): 23957, 2002.
thrombin, a mediator of human lung broblast proliferation via induc- 99. Moatt JD, Page CP, Laurent GJ. Shooting for PARs in lung dis-
tion of platelet-derived growth factor alpha-receptor. Am J Respir Cell eases. Curr Opin Pharmacol 4(3): 22129, 2004.
Mol Biol 10(4): 40512, 1994. 100. Laurent GJ. No bit PARt for PAR-1. Am J Respir Cell Mol Biol 33(3):
93. Kimura M et al. The signicance of cathepsins, thrombin and ami- 21315, 2005.
nopeptidase in diuse interstitial lung diseases. J Med Invest 52(12): 101. Takizawa T et al. Abrogation of bronchial eosinophilic inammation
93100, 2005. and attenuated eotaxin content in protease-activated receptor 2-decient
94. Jenkins RG et al. Ligation of protease-activated receptor 1 enhances mice. J Pharmacol Sci 98(1): 99102, 2005.
alpha(v)beta6 integrin-dependent TGF-beta activation and promotes 102. Schmidlin F et al. Protease-activated receptor 2 mediates eosinophil
acute lung injury. J Clin Invest 116(6): 160614, 2006. inltration and hyperreactivity in allergic inammation of the airway.
95. Howell DC et al. Direct thrombin inhibition reduces lung collagen, J Immunol 169(9): 531521, 2002.
accumulation, and connective tissue growth factor mRNA levels in 103. Churg A et al. Alpha1-antitrypsin suppresses TNF-alpha and MMP-
bleomycin-induced pulmonary brosis. Am J Pathol 159(4): 138395, 12 production by cigarette smoke-stimulated macrophages. Am J
2001. Respir Cell Mol Biol 37(2): 14451, 2007.

200
Epithelial Cells
16
CHAPTER

INTRODUCTION born at early gestational age that are exposed to Scott H. Randell1,
variable treatment regimes, including diering
preparations of exogenous surfactant [3], con-
Kimberlie Burns2 and
Respiratory tract epithelial cells constitute a ventional versus high-frequency oscillatory ven- Richard C. Boucher2
front-line physical barrier between the organism tilation [4], nitric oxide [5], and inhaled versus 1
Departments of Cell and Molecular
and the environment. They serve vital functions systemic glucocorticoids [6]. The spectrum of Physiology and Medicine, The University
in host physiology and mucosal defense and lung disease resulting from premature birth has
of North Carolina at Chapel Hill, NC, USA
are central to the pathogenesis of asthma and also evolved and now presents a histopathologic 2
Department of Medicine,
chronic obstructive pulmonary disease (COPD). picture characterized by diuse developmental
abnormalities in lung structure [7]. Wheezing is The University of North Carolina
This chapter concisely addresses epithelial cell
development, structure, cell lineages, integrated more common after premature birth, especially at Chapel Hill, NC, USA
function in mucus clearance, and presents an in susceptible populations and in the presence
overview of their role in immune responses rel- of chorioamnionitis [8]. Lung function may
evant to asthma and COPD. normalize somewhat during later childhood
and despite a higher prevalence of respiratory
symptoms, airway hyperactivity was not present
in one cohort of adults born prematurely [9, 10].
LUNG DEVELOPMENT AND Time will tell if those born prematurely are pre-
EPITHELIAL CELL DIFFERENTIATION disposed to COPD, especially if they smoke.
Prenatal airway and lung development
sets the stage for the transition to air breathing
The tracheo-bronchial and lung epithelium are at birth, including the essential function of lung
endoderm-derived, originating as a foregut evagi- liquid absorption, which depends on presence of
nation at 3.5 to 7 weeks of gestation in humans epithelial ion channels [11]. Available evidence
[1]. Complex, reciprocal molecular interactions also indicates a period of dynamic postnatal
between epithelium and the underlying mesen- adaptation and maturation of the epithelium. For
chyme govern development, resulting in elabo- example, in both animals and humans, mucous
rate branching morphogenesis that forms the secretory cells are induced in late gestation and
airways and lungs [2]. There is a centrifugal pat- are highly abundant in the neonatal period but
tern of cyto-dierentiation and structural devel- normally regress by adulthood [1217]. Although
opment that culminates in the normally formed a few studies exist [16, 17], there is a relative pau-
bronchial tree and lungs at end gestation, includ- city of quantitative data on airway epithelial cell
ing maturation of the pulmonary surfactant sys- kinetics and dierentiation during the late pre-
tem. Detailed discussion of molecular regulation natal and early postnatal stages of normal human
of lung epithelial development and abnormalities airway development. Environmental exposures
surrounding premature birth is beyond the scope and infections during infancy and early child-
of this chapter. hood mold airway and lung structure and func-
Survival of premature birth and its compli- tion and likely have a lasting inuence on the
cations has increased dramatically in recent years, ultimate development of disease. There is intense
creating a growing population of individuals interest and controversy regarding the hygiene

201
Asthma and COPD: Basic Mechanisms and Clinical Management

hypothesis, which proposes that inadequate or inappropriate (A)


Ciliated Secretory
early postnatal stimulation skews the immune system, pro-
moting the development of allergy, asthma, and autoimmunity
[18, 19]. Environmental exposures and infections occur in the
context of a dynamic airway epithelial proliferation and cyto-
dierentiation program during which responsiveness to vari-
ous stimuli (pollution, second-hand tobacco smoke, infection)
is likely altered. However, the basic cellular composition of the
developing and maturing postnatal human airway epithelium Basal
is still inadequately described. Mechanisms regulating changes
in epithelial proliferation and maturation, dierential sensitiv- (B)
ity to relevant stimuli and the impact of any dierences on
interactions with the developing immune system during this
potentially critical period are still not well understood.

STRUCTURE OF THE EPITHELIUM AND


TROPHIC UNITS (C)
Nonciliated bronchiolar

In the normal adult human, epithelial cell populations vary


systematically as a function of airway level. Portions of the
nasal passages and the trachea to approximately the 610th
generation bronchi are lined by a tall columnar pseudostrati-
ed mucociliary epithelium consisting principally of basal,
TII
intermediate, ciliated, and mucous secretory (goblet) cells
(Fig. 16.1). In this region, submucosal glands are abundant.
Proceeding distally, cartilage support and glands diminish
and become absent in the bronchioles, which are initially
also lined by a columnar pseudostratied epithelium. In TI
AM
terminal and respiratory bronchioles, the epithelium transi-
tions to a simple columnar and eventually cuboidal epithe-
lium, where Clara cells replace mucous secretory cells [20].
Individual pulmonary neuroendocrine cells (PNECs) and
highly innervated collections of PNECs (neuroendocrine
FIG. 16.1 Lung epithelial cell morphology. (A) Normal human bronchus,
bodies) are distributed within the pseudostratied, columnar, (B) bronchiole, and (C) terminal bronchiole and alveoli. One micrometer-
and cuboidal epithelial zones and extend into the proximal thick plastic sections, Richardsons stain, original magnification 400X,
alveolar region [21]. PNECs have been implicated in lung AM alveolar macrophage, TI and TII Type I and II alveolar epithelial
development, in supporting epithelial stem cell niches, and cells, respectively.
possibly as peripheral chemosensors that alter postnatal
growth and adaptation [22]. Finally, the alveolar region epi-
thelium is composed mainly of thin type I and cuboidal type epithelilal squamous metaplasia and sub-epithelial bro-
II alveolar epithelial cells with critical roles to create the thin sis that is characteristic of COPD [26]. Sub-epithelial
diusion barrier and produce surfactant, respectively. Just as remodeling, including basal lamina thickening, airway wall
the relative abundance of surface cell types changes along brosis, increased vascularity, and smooth muscle hypertro-
the respiratory tract axis, each segment is characteristically phy/hyperplasia, similarly contribute to the pathophysiology
vascularized, innervated, and variably populated by mesen- of asthma [27, 28]. However, the detailed cellular compo-
chymal and bone marrow-derived cells. sition and molecular mechanisms of communication within
The epithelium, its underlying matrix, submucosal cells, epithelial-mesenchymal trophic units at each level in the
and tissue structures constitute the epithelial-mesenchymal human airway in health and disease are not well understood.
trophic unit [23]. Reciprocal cellular communication within Airway epithelial cell renewal during the steady state and
the trophic unit underlies lung development and likely after injury occurs within the context of the trophic unit.
regulates steady-state cell behavior and repair after injury,
for example by the activation of TGF- [24]. Studies in a
primate model suggest that neonatal environmental pollut- LUNG EPITHELIAL STEM AND
ant and allergen exposure alter the epithelial-mesenchymal
trophic unit, resulting in a permanent compromise of air-
PROGENITOR CELLS
way growth and development [25]. Pathologic epithelial-
mesenchymal interactions occurring via IL-1 and TGF- In thoroughly studied epithelia such as the epidermis and
have been recently implicated in the development of airway intestine, temporal and spatial patterns of cell renewal,

202
Epithelial Cells 16
and the regulation of progenitorprogeny relationships are
becoming well understood [29, 30]. Stem cells reside in
specic niches and proliferation, migration, and cell dier-
entiation are highly orchestrated within and between the
epithelium and mesenchyme. Stem cell niches in the airway
and lung epithelium are less understood than in many other
organs, but there has been recent progress. The subject of
lung epithelial stem cells has been recently reviewed [31].
It is important to note that most studies of cell lineages
have been performed in rodent models and strict transla-
tion to humans remains unknown. Briey, metabolic pulse
labeling with DNA precursors has long shown that both
basal and columnar secretory cell types of the normal adult
pseudostratied airway epithelium divide, albeit relatively
infrequently [32, 33]. However, repair of epithelial dam-
age is brisk. Many cell types rearrange their cytoskeleton
and migrate to cover denuded basal lamina. Subpopulations
of cells proliferate and ultimately re-dierentiate [3436].
There has been controversy, but it appears that while cili-
ated cells actively participate in repair by migration and de-
dierentiation [37], they do not enter the pool of dividing
cells [38]. Subsets of cells within both the basal and colum-
nar cell populations can reconstitute a full mucociliary epi-
thelium in an in vivo transplantation model [39, 40]. Thus,
many cell types can serve as progenitors during repair of
injury. This high degree of plasticity in the airway epi-
thelium is likely driven by the necessity for rapid epithelial
repair to prevent brotic ingrowth and airway obliteration.
Classically, stem cells are dened by their capacity to
both self-renew and dierentiate, whereas progenitor cells FIG. 16.2 Cell hierarchy in the pseudostratified airway epithelium. In this
have limited self-renewal capacity. The applicability of clas- model, a subset of basal cells gives rise to transiently amplifying cells with
sical denitions from well-known model systems to the basal and intermediate cell morphologies that generate differentiated
highly plastic airway epithelium is questionable, but there cells (straight black arrows). The circular arrows represent growth capacity.
The red arrows indicate plasticity-differentiated cells are capable of
does appear to be a hierarchy of stemness within the pool
dedifferentiation (backwards arrows), redifferentiaton, and phenotypic
of identied progenitor cells. Colony formation on plastic conversion during repair of injury. Reproduced, with permission, from
dishes in vitro [41], clonal growth [42], lineage tracing [43], Randell SH. Airway epithelial stem cells and the pathophysiology of chronic
and DNA label retention [44] suggest that although many obstructive pulmonary disease. Proc Am Thorac Soc 8: 71825, 2006.
cells can contribute to repair of injury, basal cells likely rep-
resent a stem cell compartment in the adult pseudostratied
epithelium (Fig. 16.2).
The simple columnar to cuboidal bronchiolar epi- the broncho-alveolar junction zone [50, 51]. Here, specic
thelium of rodents is composed of Clara, ciliated, PNEC, Clara-like cells co-express SCGB1a1, SP-C, SP-A, CD-
and brush cells. Secretoglobin (SCGB) 1a1, also known as 34 and Sca-1, and proliferate in response to naphthalene
CC10 or CCSP, commonly serves as a marker for Clara or bleomycin injury [51]. The cells reportedly grow clonally
cells. However, SCGB1a1 is expressed in a spectrum of cells, in vitro, dierentiate into both Clara and distal lung type
present in multiple airway levels, with varying dierentiation I and type II epithelial cells, and generate adenocarcino-
potentials and behaviors. Following oxidant gas exposure of mas when expressing an active K-ras oncogene in vivo [51].
rodents which damages ciliated cells, surviving bronchiolar Epithelial cell lineages in the bronchioles and proximal
Clara cells proliferate to restore the epithelium [45, 46]. alveolar region are illustrated in Fig. 16.3. It is important to
When the toxin naphthalene is administered to mice, almost conrm, in humans, the dual airway and alveolar potential
all Clara cells are killed due to selective metabolic activation of the putative broncho-alveolar stem cells found in mice.
in this cell type. Ciliated cells shed their cilia and migrate It has been known for many years that alveolar type II
to cover the denuded bronchiolar basal lamina, but do not cells proliferate to repair injuries that damage the large, thin,
re-enter the cell cycle [38], whereas surviving Clara cells and putatively fragile type I alveolar epithelial cells [52].
proliferate [47]. A population of naphthalene-resistant Palisades of reactive, cuboidal type II cells are frequently
Clara cells resides within, or close to, PNEC clusters and present when there is chronic distal lung injury. Although
DNA label retention studies suggest that this unit consti- there is evidence for a spectrum of proliferative potential
tutes a stem cell niche [48]. Following naphthalene injury, amongst the type II cell population in the distal alveolar
PNECs also proliferate, but they are apparently a dis- epithelium [53], epithelial stem cell hierarchies in the distal
tinct lineage system not requiring, nor generating, Clara alveoli are still poorly understood. The importance of eec-
cells [49]. A second stem cell niche has been proposed in tive epithelial repair in the alveolus is underscored by the

203
Asthma and COPD: Basic Mechanisms and Clinical Management

FIG. 16.3 Cell hierarchy in the bronchiolar


epithelium. The circular arrows represent
growth capacity and differentiation potential.
The pulmonary neuroendocrine cell (PNEC)
lineage is apparently separate from other
epithelial cell types in the adult. Variant Clara
cells present in association with PNEC bodies
serve as progenitor cells of the bronchiolar
epithelium. Variant Clara cells found near the
bronchiole-alveolar junction may generate
both bronchiolar and alveolar epithelium.
Reproduced, with permission, from Randell
SH. Airway epithelial stem cells and the
pathophysiology of chronic obstructive
pulmonary disease. Proc Am Thorac Soc 8:
71825, 2006.

recent discovery that mutations in telomerase reverse tran- and COPD remains unknown. The metastasis of lung can-
scriptase or the RNA component of the telomerase complex cer cells is thought to involve a process similar to EMT that
are a cause of idiopathic pulmonary brosis [54, 55]. In these is regulated by TGF- and the extracellular matrix [63].
cases, decient chromosome maintenance due to telomerase Due to the promise of regenerative medicine for
deciency likely impairs epithelial regeneration, resulting chronic lung disease after neonatal respiratory disorders or
in aberrant epithelialmesenchymal interactions that ulti- for more typical adult forms of pulmonary emphysema, there
mately cause progressive brosis. Altered airway epithelial is intense interest in lung stem cells. Furthermore, the impor-
mesenchymal interactions, whether due to decient epithelial tant issue of lung cancer initiating cells and stem cells within
repair or not, may similarly result in airway sub-epithelial lung cancers is highly related to stem cell biology in the nor-
brosis and remodeling in asthma and COPD. The relation- mal lung. Recent advances point towards additional stud-
ship between epithelial repair capacity and the development ies needed to characterize lung stem cells and their niches,
of pulmonary emphysema has not yet been dened. and to elucidate steady state and repairing cell lineages. The
Two somewhat controversial topics have entered the identication of unique gene and protein markers indica-
lung epithelial stem cell dialogue. The rst is whether circu- tive of cell position in the lung epithelial stem cell hierarchy
lating cells become epithelial cells by undergoing a process will enable generation of novel transgenic animals that will
called trans-dierentiation. In the original report it was foster more extensive and precise lineage mapping, which is
claimed that, following lethal doses of ionizing radiation and claimed to be the gold standard by some stem cell authori-
bone marrow transplantation, circulating cells of bone mar- ties [64]. Furthermore, such markers may enable purica-
row origin reconstituted a substantial percentage of airway tion and functional testing of candidate stem and progenitor
and alveolar epithelial cells [56]. Subsequent studies suggest cell populations from both animals and humans. A greater
that apparent replacement of the epithelium from circulat- understanding of the molecular pathways regulating nor-
ing cells may be due to fusion events between myeloid cells mal cell behavior and disease phenotypes underlies rational
and epithelial cells and that transdierentiation is unlikely approaches to cellular therapy for lung diseases, whether
[57]. A recent analysis of cord blood stem cell conversion it involves augmenting epithelial repair by stimulating
to epithelium is illustrative, and suggests that if transdier- growth or by inhibiting growth as in the case of epithelial
entiation occurs, circulating cells may generate only a very hyperplasia/metaplasia and lung cancer.
small percentage of the epithelium [58]. The growing con-
sensus is that most epithelial replacement occurs via lung
tissue-specic resident or so-called somatic, stem cells. The
presence of regional-specic stem cells as the target cells for
transformation and cancer initiation may underlie the het-
INTEGRATED EPITHELIAL FUNCTION
erogeneity of lung cancer types [59]. A second area of recent
interest is epithelial to mesenchymal transition (EMT), A continuous epithelial layer, composed of characteristic cell
which is a normal developmental process, for example in types along the respiratory tract axis, forms a physical bar-
kidney development, where epithelial cells loose polarity rier between the outside environment and the underlying
and gain features of mesenchymal cell types. Recent studies matrix and circulatory system. The airways deliver warmed
suggest that lung epithelial cells exposed to TGF- in vitro and humidied inhaled air to the alveoli while protecting
may convert into myobroblast-like cells and that EMT gas exchange structures from potentially harmful airborne
occurs in lung brosis models in animals and in human idi- chemicals, particles, and pathogens. The epithelium pro-
opathic pulmonary brosis [60, 61] (reviewed in Ref. [62]). vides both general and regionally specialized functions that
The signicance of EMT for airway remodeling in asthma are key to normal physiology and host defense. The airway

204
Epithelial Cells 16
epithelium is a principal generator of nitric oxide [65] and
coordinates uid and mucous secretion to foster eective MUCINS AND MUCUS
mucociliary transport and productive cough [66]. The dis-
tal lung alveolar epithelium plays a key role in lung uid Mucus is composed of mucins, proteins, lipids, ions and
balance, the prevention and resolution of alveolar edema water present on mucosal surfaces. Mucins are highly gly-
after injury [67] and is the source of pulmonary surfactant. cosylated proteins that can be released from mucous secre-
Furthermore, the epithelium in both the conducting and tory cell granules and/or by shedding from the surface of
respiratory zones plays a key role during innate and adap- many cell types. Intermolecular disulde bonds within
tive immune responses. the principal gel-forming lung mucins, MUC5AC and
MUC5B, enable multimerization and extensive interdigi-
tation creating a visco-elastic network capable of trapping
a wide variety of inhaled particles [80]. Increased mucous
MUCOCILIARY AND MUCUS CLEARANCE secretory cell number and increased mucin gene expression
and glycoprotein production characterize both asthma and
COPD. Regulations of airway epithelial secretory cell dif-
A layer of uid and mucus normally ows over conducting ferentiation, mucin gene and glycoprotein expression and
airway surfaces toward the pharynx. This cleansing action the secretory pathways that mediate mucin release have
is a critical, physiologically regulated, protective mecha- been reviewed [8183]. In addition to the surface epithe-
nism of the airways and lungs, whose failure causes recur- lium, submucosal glands in the cartilaginous airways are a
rent infections characteristic of the genetic diseases cystic signicant source of uid, mucins, and other proteins such
brosis and primary ciliary dyskinesia [68]. Eective mucus as anti-microbial factors [8486]. Gland hypertrophy is a
clearance results from coordinated secretion of mucins and pathologic hallmark of both asthma and COPD and likely
other proteins along with sucient water for hydration, contributes to mucin hypersecretion [27, 28].
which is locally regulated by airway epithelial ion trans-
port. Adequately hydrated mucus is propelled by beating
cilia and, when necessary, by cough. The combination of
mucociliary and cough clearance equals mucus clearance.
As measured by movement of inhaled and deposited radio-
CILIATED CELLS
active tracer particles, mucus clearance declines in propor-
tion to airow obstruction in acute exacerbations of asthma, Ciliated cells have long and branched microvilli plus approx-
and returns toward normal after eective therapy [6971]. imately 200 cilia per cell that occupy a large percentage of
Several studies indicate that mucus clearance is decreased the normal airway epithelial luminal surface (Fig. 16.4). Cilia
by tobacco smoking and in COPD [7275]. The accumula- in the trachea and bronchi are denser and longer than in the
tion of inammatory mucoid secretions in the small airways bronchioles, but cilia average approximately 250 nm in diam-
in COPD, which likely reects both increased local mucin eter and 6 m in length. The shaft, or axoneme, of motile
production and decreased clearance [76], is correlated with cilia consists of nine outer microtubule doublets, a central
the degree of physiologic impairment [77]. Drug therapies microtubule pair, inner and outer dynein arms, nexin links,
such as -adrenergic agonists, cholinergic antagonists and and radial spokes. The axoneme enters the ciliated cell and is
corticosteroids used to treat asthma and COPD may have anchored by a cytoplasmic basal body similar to the centriole
benecial eects on mucus transport, but consistent positive of mitotic cells. Greater than 200 unique proteins constitute
eects on mucus clearance have been dicult to substan- the cilia and basal body [87]. In normal airways, cilia beat
tiate [78, 79]. Additional specic therapies directed toward at 1020 Hz in a coordinated manner that generates direc-
balancing mucin production and ion and water transport to tional, metachronal waves to assist in the propulsion of uid,
improve mucus clearance will likely be useful. mucus and trapped particles towards the pharynx.

FIG. 16.4 Ciliated cell ultrastructure.


Conventional transmission electron
microscopy of a normal human nasal
biopsy. (A) Longitudinal section across the
apical border of a ciliated cell showing
a clear area near the cell surface with
cilia tips enmeshed in viscous material.
Original magnification 12,000 X. (B) Cross
sections of cilia axonemes, illustrating the
9 2 arrangement of microtubules in
motile cilia. ODA: outer dynein arm; IDA:
inner dynein arm; RS: radial spoke, original
magnification 50,000 X.

205
Asthma and COPD: Basic Mechanisms and Clinical Management

is thought to coordinately inhibit ENaC. ATP can also be


ION AND WATER TRANSPORT AND INTEGRATED metabolized to adenosine, which will activate A2B adenosine
FUNCTION FOR EFFECTIVE MUCUS CLEARANCE receptors, generating cAMP and activating protein kinase
C to ultimately open CFTR. Active CFTR is also reported
Mucous secretory and ciliated cells distinctly contribute to to directly inhibit ENaC. Apical membrane Cl secre-
mucociliary clearance which, combined with cough, consti- tion through CFTR and/or CaCC increases airway sur-
tutes mucus clearance, a key airway defense mechanism [88]. face hydration whereas Na entry through ENaC decreases
Collectively, the system is organized and physiologically airway surface hydration. Water follows its osmotic gradi-
regulated to balance mucin production with ion and water ent transcellularly through aquaporins and, a small part, via
transport to maintain surface hydration at levels sucient to para-cellular pathways. In the distal lung, similar processes,
foster mucus transportability. Airway surface liquid is nor- mainly involving Na absorption through ENaC but also
mally present in two layers, an overlying viscous mucus layer involving CFTR, are thought to regulate normal alveolar
and a periciliary layer (PCL) in which cilia can freely beat. uid balance and are important in the resolution of pulmo-
A slip layer provides lubrication at the interface of these nary edema after acute lung injury [67].
two layers [66]. The mucus layer and the PCL can be visu- Endogenous mediators active in regulation of airway
alized after special xation techniques of tissues or in cul- and lung surface liquid have additional roles, which may
ture preparations of live, dierentiated airway epithelial cells pose conundrums relevant to potential therapies for asthma
[8991] (Fig. 16.5). By balancing Cl secretion and Na and COPD. For example, although ATP promotes airway
absorption at the epithelial luminal membrane, the airway surface hydration via activation of epithelial P2Y2 purin-
epithelium regulates PCL volume to fully hydrate the PCL ergic receptors, ATP is also a danger signal involved in
and maintain its depth at the height of the outstretched cilia regulation of the immune system [98]. Excess airway ATP
[90, 92, 93]. There are two principal apical membrane chan- may contribute to an asthma-like phenotype via pro inam-
nels, the cystic brosis transmembrane conductance regulator matory eects, including the recruitment and activation of
(CFTR) and the Ca2-activated Cl channel (CaCC), that dendritic cells (DCs) [99]. Similarly, adenosine activation of
mediate Cl transport. Na transport occurs through the epithelial A2B receptors is important to activate CFTR and
epithelial Na channel (ENaC). CFTR, CaCC, and ENaC hydrate the epithelial surface, but high adenosine levels in
activity is locally and coordinately regulated by changes in adenosine deaminase decient mice promotes inammation,
the PCL concentration of nucleotides and nucleosides and inhaled adenosine causes broncho-constriction in asth-
[9497]. In this system, ATP released by cells activates matic humans [100]. Pharmacologic therapies directed at
P2Y2 purinergic receptors, which stimulates IP3 formation, reducing adenosine or antagonizing adenosine receptors may
causing Ca2 release that activates CaCC. Decreased PIP2 have the unwanted eect of creating a cystic brosis-like

FIG. 16.5 Respiratory tract ion and water


transport and mucus clearance. (A) The airway
epithelium coordinates fluid and mucous
secretion to foster effective mucus clearance.
It is still uncertain how much alveolar lining
fluid leaves the distal lung via the airways (gray
arrow). (B) Ion channels in the apical plasma
membrane, regulated by local concentrations of
nucleotides and nucleosides, maintain adequate
airway surface hydration. (C) Airway surface
liquid consists of two layers, a clear periciliary
layer and overlying mucus. Perflurocarbon-
OsO4 fixed, well-differentiated human bronchial
epithelial cell culture, thin plastic section, original
magnification 500X, Richardsons stain. (A) and
(B) are adapted, with permission, from Ref. [68].

206
Epithelial Cells 16
phenotype [97]. Furthermore, although elevated adeno- stimulus. Preformed substrates such as arachidonic acid can
sine produces whole animal pro-inammatory eects, be metabolized and released and/or gene expression/stability
adenosine acting via A2A receptors on macrophages is anti- can be modied to change patterns of epithelial cell mediator
inammatory [101]. The pleiotropic eects of media- production. Receptor engagement and downstream signaling
tors such as ATP and adenosine are likely concentration-, may induce pro-inammatory cytokines and chemokines
compartment-, receptor-, and cell type-specic and must be that selectively attract neutrophils, monocytes, eosinophils,
considered when proposing therapies based on manipulation and basophils to ostensibly eliminate pathogens. The process
of their levels or receptors. is dynamic, and repeat or chronic exposure to substances that
activate innate epithelial cell immune system receptors will
modulate epithelial cell behavior upon subsequent exposure
[108]. The epithelium thus plays a vital innate immune role
LUNG EPITHELIAL FUNCTION IN IMMUNITY to perceive and respond to danger, with mechanisms in place
to modulate and orchestrate inammation depending on the
type, degree, and chronicity of stimulation. Functional poly-
The mammalian immune system is broadly divided into morphisms in genes involved in epithelial innate immunity
innate and adaptive branches. Innate immunity is the are likely important modiers related to the development of
rst line of defense that is constantly on guard and requires lung disease.
no prior exposure to harmful substances or pathogens to
induce a protective response. Adaptive immunity takes time
to develop via specic, antigen triggered cellular interactions
between DCs, T-, and B-cells and has profound memory. ROLE IN ADAPTIVE IMMUNITY
The two branches are highly intertwined. For example, the
cell and chemical milieu produced by the innate immune
system is critical for the development of adaptive immunity, Beyond regulating luminal dwell time and permeability of
and conversely, products of adaptive immune cells can alter allergens and pathogens, the airway and lung epithelium
the function of cells involved in innate immunity. The air- play key regulatory roles in adaptive immunity. Cytokines,
way and lung epithelium is integral to innate and adaptive chemokines, and other molecules produced by the epithe-
immunity and is both an eector and target in the altered lium at steady state, and especially after innate immune
immune function characteristic of asthma and COPD. stimulation, modulate adaptive immunity by attracting DCs,
T-, and B-cells and altering their function (reviewed in Ref.
[109]). In turn, the epithelium responds to products of
adaptive immune cells, establishing complex feedback loops
ROLE IN INNATE IMMUNITY that ultimately determine tissue inammation and lesion
development. The epithelium also plays a functional role by
facilitating the transport of secretory immunoglobulin.
The airway and lung epithelium is a selective physical barrier The conducting airway epithelium and alveolar inter-
that coordinates removal of potentially harmful substances by stitium contain resident DCs that sample the luminal micro-
mucus clearance. It also plays important roles in host defense environment and present antigens to T-cells locally, or after
by secreting a wide spectrum of protective, antimicrobial, migrating to regional lymph nodes. The numbers of DCs in
homeostatic, and immune regulatory factors including anti- the airways and lungs, and their functional state, are altered
oxidants, defensins, collectins, complement components, by numerous chemokines, cytokines, and other molecules
lactoferrin, lysozyme, and protease inhibitors (reviewed in involved in DC attraction and activation that are secreted
Refs [102, 103]). Normally protective innate immune func- by epithelial cells [110]. Chronic inammatory diseases are
tions can be enhanced when pathogen/microbial associated regulated by T-cell activation and polarization and recent
patterns or other chemical danger signals are recognized discoveries have added new T-cell subsets to the landscape.
by innate immune system receptors expressed by epithelial Organ parenchymal cells participate in the dierentiation
cells. The receptors include transmembrane molecules such of Th1, Th2, Th17, and T-regulatory (Treg) cells from nave
as the toll-like receptors (TLRs) [104], protease-activated T-cells and create feedback loops that regulate and sustain
receptors (PARs) [105] and purinergic receptors [98] as local responses. For example, epithelial IL-6 production
well as cytosolic proteins including the nucleotide-binding, positively reinforces Th17 cell polarization [111]. Conversely,
leucine-rich repeat receptors (NLRs) [106] and the retin- T-cell products aect the epithelium, such as the well-known
oic acid-inducible gene I-like helicases (RLHs) [107]. The induction of mucin by the Th2 cytokine IL-13 [112, 113].
substances detected by these multiple classes of receptors Th17 cell production of IL-22 and its action on the epithe-
include endogenous homeostatic factors such as ATP as well lium appears to play an important role in epithelial repair and
as molecules derived from microbes and pathogens includ- mucosal defense against gram-negative bacteria [114]. The
ing bacterial lipopolysaccharide and viral RNA. airway and lung are populated by immunoglobulin secret-
Receptor engagement induces downstream signaling ing plasma cells. B-cell stimulation and class switching is
that modies cellular behavior in many ways to eliminate the strongly inuenced by other adaptive immune cells but there
perceived danger. Airway epithelial ion channels can be acti- is mounting evidence that epithelium-derived factors partici-
vated to induce uid secretion accompanied by mucin release pate in regulation of B-cell class switching and immunoglob-
and increased cilia beating to ush away the oending ulin production [109]. The epithelium plays an integral

207
Asthma and COPD: Basic Mechanisms and Clinical Management

role in adaptive immunity by transporting secretory immu- 17. Lamb D, Reid L. Acid glycoproteins produced by the mucous cells of
noglobulin from the lamina propria where it is produced into the bronchial submucosal glands in the fetus and child: A histochemi-
the lumen, a process that may be impaired in asthma and cal autoradiographic study. Br J Dis Chest 66: 24853, 1972.
18. Racila DM, Kline JN. Perspectives in asthma: Molecular use of micro-
COPD [115].
bial products in asthma prevention and treatment. J Allergy Clin Immunol
To summarize, complex reciprocal interactions between 116: 12025, 2005.
the epithelium and the adaptive immune system regulate the 19. Schaub B, Lauener R, von Mutius E. The many faces of the hygiene
integrated response to allergens and infectious agents and hypothesis. J Allergy Clin Immunol 117: 96977, 2006.
play important roles in asthma and COPD. 20. Mercer RR, Russell ML, Roggli VL, Crapo JD. Cell number and
distribution in human and rat airways. Am J Respir Cell Mol Biol 10:
61324, 1994.
21. De Proost I, Pintelon I, Brouns I, Kroese AB, Riccardi D, Kemp PJ,
References Timmermans JP, Adriaensen D. Functional Live Cell Imaging of the
Pulmonary Neuroepithelial Body Microenvironment. Am J Respir Cell
1. Wert SE. Normal and abnormal structural development of the lung. In: Mol Biol 2008 doi:10. 1165/rcmb. 2008-0011OC.
Polin RA, Fox WW, Abman SH (eds). Fetal and Neonatal Physiology, 22. Cutz E, Yeger H, Pan J. Pulmonary neuroendocrine cell system in
pp. 78394. Philadelphia: Saunders, 2004. pediatric lung disease-recent advances. Pediatr Dev Pathol 10: 41935,
2. van Tuyl M, Post M. Molecular mechanisms of lung development and 2007.
lung branching morphogenesis. In: Polin RA, Fox WW, Abman SH 23. Evans MJ, Van Winkle LS, Fanucchi MV, Plopper CG. The attenu-
(eds). Fetal and Neonatal Physiology, pp. 81221. Philadelphia: Saunders, ated broblast sheath of the respiratory tract epithelial-mesenchymal
2004. trophic unit. Am J Respir Cell Mol Biol 21: 65557, 1999.
3. Pster RH, Soll RF, Wiswell T. Protein containing synthetic surfactant 24. Araya J, Cambier S, Morris A, Finkbeiner W, Nishimura SL. Integrin-
versus animal derived surfactant extract for the prevention and treat- mediated transforming growth factor-beta activation regulates home-
ment of respiratory distress syndrome. Cochrane Database Syst Rev, ostasis of the pulmonary epithelial-mesenchymal trophic unit. Am J
2007, CD006069. Pathol 169: 40515, 2006.
4. Henderson-Smart DJ, Cools F, Bhuta T, O ringa M. Elective high 25. Plopper CG, Smiley-Jewell SM, Miller LA, Fanucchi MV, Evans MJ,
frequency oscillatory ventilation versus conventional ventilation for Buckpitt AR et al. Asthma/allergic airways disease: Does postnatal
acute pulmonary dysfunction in preterm infants. Cochrane Database Syst exposure to environmental toxicants promote airway pathobiology?
Rev, 2007, CD000104. Toxicol Pathol 35: 97110, 2007.
5. Barrington KJ, Finer NN. Inhaled nitric oxide for respiratory failure in 26. Araya J, Cambier S, Markovics JA, Wolters P, Jablons D, Hill A et al.
preterm infants. Cochrane Database Syst Rev, 2007, CD000509. Squamous metaplasia amplies pathologic epithelial-mesenchymal
6. Shah SS, Ohlsson A, Halliday H, Shah VS. Inhaled versus systemic interactions in COPD patients. J Clin Invest 117: 355162, 2007.
corticosteroids for the treatment of chronic lung disease in ventilated 27. Holgate ST. Epithelium dysfunction in asthma. J Allergy Clin Immunol
very low birth weight preterm infants. Cochrane Database Syst Rev, 2007, 120: 123344, 2007.
CD002057. 28. Jeery PK. Remodeling and inammation of bronchi in asthma and
7. Baraldi E, Filippone M. Chronic lung disease after premature birth. chronic obstructive pulmonary disease. Proc Am Thorac Soc 1: 17683,
N Engl J Med 357: 194655, 2007. 2004.
8. Kumar R, Yu Y, Story RE, Pongracic JA, Gupta R, Pearson C et al. 29. Fuchs E. Skin stem cells: Rising to the surface. J Cell Biol 180: 27384,
Prematurity, chorioamnionitis, and the development of recurrent 2008.
wheezing: A prospective birth cohort study. J Allergy Clin Immunol 121: 30. Barker N, Clevers H. Tracking down the stem cells of the intestine:
87884, 2008. Strategies to identify adult stem cells. Gastroenterology 133: 175560,
9. Narang I, Baraldi E, Silverman M, Bush A. Airway function measure- 2007.
ments and the long-term follow-up of survivors of preterm birth with 31. Kotton DN, Fine A. Lung stem cells. Cell Tissue Res 331: 14556,
and without chronic lung disease. Pediatr Pulmonol 41: 497508, 2006. 2008.
10. Narang I, Rosenthal M, Cremonesini D, Silverman M, Bush A. 32. Donnelly GM, Haack DG, Heird CS. Tracheal epithelium: Cell kinet-
Longitudinal evaluation of airway function 21 years after preterm ics and dierentiation in normal rat tissue. Cell Tissue Kinet 15: 11930,
birth. Am J Respir Crit Care Med 178: 7480, 2008. 1982.
11. Barker PM, Southern KW. Regulation of liquid secretion and absorption 33. Breuer R, Zajicek G, Christensen TG, Lucey EC, Snider GL. Cell
by the fetal and neonatal lung. In: Polin RA, Fox WW, Abman SH kinetics of normal adult hamster bronchial epithelium in the steady
(eds). Fetal and Neonatal Physiology, pp. 82234. Philadelphia: Saunders, state. Am J Respir Cell Mol Biol 2: 5158, 1990.
2004. 34. Desai LP, Aryal AM, Ceacareanu B, Hassid A, Waters CM. RhoA and
12. Randell SH, Shimizu T, Bakewell W, Ramaekers FC, Nettesheim P. Rac1 are both required for ecient wound closure of airway epithelial
Phenotypic marker expression during fetal and neonatal dierentia- cells. Am J Physiol 287: L113444, 2004.
tion of rat tracheal epithelial cells. Am J Respir Cell Mol Biol 8: 54655, 35. Shimizu T, Nishihara M, Kawaguchi S, Sakakura Y. Expression of
2006. phenotypic markers during regeneration of rat tracheal epithelium fol-
13. Otani EM, Newkirk C, McDowell EM. Development of hamster tra- lowing mechanical injury. Am J Respir Cell Mol Biol 11: 8594, 1994.
cheal epithelium: IV. Cell proliferation and cytodierentiation in the 36. Erjefalt JS, Persson CG. Airway epithelial repair: Breathtakingly quick
neonate. Anat Rec 214: 18392, 1986. and multipotentially pathogenic. Thorax 52: 101012, 1997.
14. Leigh MW, Gambling TM, Carson JL, Collier AM, Wood RE, Boat TF. 37. Park KS, Wells JM, Zorn AM, Wert SE, Laubach VE, Fernandez LG
Postnatal development of tracheal surface epithelium and submucosal et al. Transdierentiation of ciliated cells during repair of the respira-
glands in the ferret. Exp Lung Res 10: 15369, 1986. tory epithelium. Am J Respir Cell Mol Biol 34: 15157, 2006.
15. Van Winkle LS, Fanucchi MV, Miller LA, Baker GL, Gershwin LJ, 38. Rawlins EL, Ostrowski LE, Randell SH, Hogan BLM. Lung develop-
Schelegle ES et al. Epithelial cell distribution and abundance in rhe- ment and repair: Contribution of the ciliated lineage. Proc Natl Acad Sci
sus monkey airways during postnatal growth and development. J Appl USA 104: 41017, 2007.
Physiol 97: 235563, 2004. 39. Liu JY, Nettesheim P, Randell SH. Growth and dierentiation of tra-
16. Gaillard DA, Lallement AV, Petit AF, Puchelle ES. In vivo ciliogenesis cheal epithelial progenitor cells. Am J Physiol Lung Cell Mol Physiol 266:
in human fetal tracheal epithelium. Am J Anat 185: 41528, 1989. L296307, 1994.

208
Epithelial Cells 16
40. Avril-Delplanque A, Casal I, Castillon N, Hinnrasky J, Puchelle E, 61. Willis BC, Liebler JM, Luby-Phelps K, Nicholson AG, Crandall ED,
Peault B. Aquaporin-3 expression in human fetal airway epithelial pro- du Bois RM et al. Induction of epithelial-mesenchymal transition in
genitor cells. Stem Cells 23: 9921001, 2005. alveolar epithelial cells by transforming growth factor-beta1: Potential
41. Randell SH, Comment CE, Ramaekers FCS, Nettesheim P. Properties role in idiopathic pulmonary brosis. Am J Pathol 166: 132132, 2005.
of rat tracheal epithelial cells separated based on expression of cell sur- 62. Willis BC, Borok Z. TGF-beta-induced EMT: Mechanisms and
face alpha-galactosyl end groups. Am J Respir Cell Mol Biol 4: 54454, implications for brotic lung disease. Am J Physiol Lung Cell Mol Physiol
1991. 293: L52534, 2007.
42. Schoch KG, Lori A, Burns KA, Eldred T, Olsen JC, Randell SH. 63. Shintani Y, Maeda M, Chaika N, Johnson KR, Wheelock MJ.
A subset of mouse tracheal epithelial basal cells generates large colo- Collagen I promotes epithelial-to-mesenchymal transition in lung can-
nies in vitro. Am J Physiol 286: L63142, 2004. cer cells via transforming growth factor-beta signaling. Am J Respir Cell
43. Engelhardt JF, Schlossberg H, Yankaskas JR, Dudus L. Progenitor cells Mol Biol 38: 95104, 2008.
of the adult human airway involved in submucosal gland development. 64. Nystul TG, Spradling AC. Breaking out of the mold: Diversity within
Development 121: 203146, 1995. adult stem cells and their niches. Curr Opin Genet Dev 16: 46368,
44. Borthwick DW, Shahbazian M, Todd KQ, Dorin JR, Randell SH. 2006.
Evidence for stem-cell niches in the tracheal epithelium. Am J Respir 65. Bove PF, van der Vilet A. Nitric oxide and reactive nitrogen species in
Cell Mol Biol 24: 66270, 2001. airway epithelial signaling and inammation. Free Radic Biol Med 41:
45. Evans MJ, Johnson LV, Stephens RJ, Freeman G. Renewal of the ter- 51527, 2006.
minal bronchiolar epithelium in the rat following exposure to NO2 or 66. Randell SH, Boucher RC, for the University of North Carolina Virtual
O3. Lab Invest 35: 24657, 1976. Lung Group. Eective mucus clearance is essential for respiratory
46. Evans MJ, Shami SG, Cabral-Anderson LJ, Dekker NP. Role of non- health. Am J Respir Cell Mol Biol 35:208, 2006.
ciliated cells in renewal of the bronchial epithelium of rats exposed to 67. Berthiaume Y, Matthay MA. Alveolar edema uid clearance and acute
NO2. Am J Pathol 123: 12633, 1986. lung injury. Respir Physiol Neurobiol 159: 35059, 2007.
47. Van Winkle LS, Buckpitt AR, Nishio SJ, Isaac JM, Plopper CG. 68. Livraghi A, Randell SH. Cystic brosis and other respiratory diseases
Cellular response in naphthalene-induced Clara cell injury and bron- of impaired mucus clearance. Toxicol Pathol 35: 11629, 2007.
chiolar epithelial repair in mice. Am J Physiol 269: L80018, 1995. 69. Messina MS, ORiordan TG, Smaldone GC. Changes in mucocili-
48. Hong KU, Reynolds SD, Giangreco A, Hurley CM, Stripp BR. Clara ary clearance during acute exacerbations of asthma. Am Rev Respir Dis
cell secretory protein-expressing cells of the airway neuroepithelial 143: 99397, 1991.
body microenvironment include a label-retaining subset and are critical 70. ORiordan TG, Zwang J, Smaldone GC. Mucociliary clearance in
for epithelial renewal after progenitor cell depletion. Am J Respir Cell adult asthma. Am Rev Respir Dis 146: 598603, 1992.
Mol Biol 24: 67181, 2001. 71. Shah RV, Amin M, Sangwan S, Smaldone GC. Steroid eects on
49. Reynolds SD, Hong KU, Giangreco A, Mango GW, Guron C, mucociliary clearance in outpatient asthma. J Aerosol Med 19: 20820,
Morimoto Y et al. Conditional Clara cell ablation reveals a self-renewing 2006.
progenitor function of pulmonary neuroendocrine cells. Am J Physiol 72. Goodman RM, Yergin BM, Landa JF, Golinvaux MH, Sackner MA.
278: L125663, 2000. Relationship of smoking history and pulmonary function tests to tra-
50. Giangreco A, Reynolds SD, Stripp BR. Terminal bronchioles harbor a cheal mucous velocity in nonsmokers, young smokers, ex-smokers, and
unique airway stem cell population that localizes to the bronchoalveo- patients with chronic bronchitis. Am Rev Respir Dis 117: 20514, 1978.
lar duct junction. Am J Pathol 161: 17382, 2002. 73. Vastag E, Matthys H, Koehler D, Gronbeck L, Daikeler G.
51. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S Mucociliary clearance and airways obstruction in smokers, ex-smokers
et al. Identication of bronchoalveolar stem cells in normal lung and and normal subjects who never smoked. Eur J Respir Dis 66(Suppl.
lung cancer. Cell 121: 82335, 2005. 139): 93100, 1985.
52. Adamson IY, Bowden DH. The type 2 cell as progenitor of alveolar 74. Camner P, Mossberg B, Philipson K. Tracheobronchial clearance and
epithelial regeneration. A cytodynamic study in mice after exposure to chronic obstructive lung disease. Scand J Respir Dis 54: 27281, 1973.
oxygen. Lab Invest 30: 3542, 1974. 75. Smaldone GC, Foster WM, ORiordan TG, Messina MS, Perry RJ,
53. Reddy R, Buckley S, Doerken M, Barsky L, Weinberg K, Anderson KD Langenback EG. Regional impairment of mucociliary clearance in
et al. Isolation of a putative progenitor subpopulation of alveolar epi- chronic obstructive pulmonary disease. Chest 103: 139096, 1993.
thelial type 2 cells. Am J Physiol 286: L65867, 2004. 76. Hogg JC. Pathophysiology of airow limitation in chronic obstructive
54. Tsakiri KD, Cronkhite JT, Kuan PJ, Xing C, Raghu G, Weissler JC pulmonary disease. Lancet 364: 70921, 2004.
et al. Adult-onset pulmonary brosis caused by mutations in telomer- 77. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L et al.
ase. Proc Natl Acad Sci USA 104: 755257, 2007. The nature of small-airway obstruction in chronic obstructive pulmo-
55. Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C nary disease. N Engl J Med 350: 264553, 2004.
et al. Telomerase mutations in families with idiopathic pulmonary 78. Restrepo RD. Inhaled adrenergics and anticholinergics in obstructive
brosis. N Engl J Med 356: 131726, 2007. lung disease: Do they enhance mucociliary clearance? Respir Care 52:
56. Krause DS, Theise ND, Collector MI, Henegariu O, Hwang S, 115973, 2007.
Gardner R et al. Multi-organ, multi-lineage engraftment by a single 79. Bennett WD. Eect of beta-adrenergic agonists on mucociliary clear-
bone marrow-derived stem cell. Cell 105: 36977, 2001. ance. J Allergy Clin Immunol 110(6 Suppl.): S29197, 2002.
57. Kotton DN, Fabian AJ, Mulligan RC. Failure of bone marrow to recon- 80. Sheehan JK, Kesimer M, Pickles R. Innate immunity and mucus struc-
stitute lung epithelium. Am J Respir Cell Mol Biol 33: 32834, 2005. ture and function. Novartis Found Symp 279: 15566, 2006.
58. Sueblinvong V, Loi R, Eisenhauer PL, Bernstein IM, Suratt BT, 81. Rose MC, Voynow JA. Respiratory tract mucin genes and mucin glyc-
Spees JL et al. Derivation of lung epithelium from human cord blood- oproteins in health and disease. Physiol Rev 86: 24578, 2006.
derived mesenchymal stem cells. Am J Respir Crit Care Med 177: 82. Thornton DJ, Sheehan JK. From mucins to mucus: Toward a more
70111, 2008. coherent understanding of this essential barrier. Proc Am Thorac Soc 1:
59. Giangreco A, Groot KR, Janes SM. Lung cancer and lung stem cells: 5461, 2004.
Strange bedfellows? Am J Respir Crit Care Med 175: 54753, 2007. 83. Williams OW, Sharafkhaneh A, Kim V, Dickey BF, Evans CM.
60. Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Airway mucus: From production to secretion. Am J Respir Cell Mol Biol
Brumwell AN et al. Alveolar epithelial cell mesenchymal transition 34: 52736, 2006.
develops in vivo during pulmonary brosis and is regulated by the 84. Inglis SK, Wilson SM. Cystic brosis and airway submucosal glands.
extracellular matrix. Proc Natl Acad Sci USA 103: 1318085, 2006. Pediatr Pulmonol 40: 27984, 2005.

209
Asthma and COPD: Basic Mechanisms and Clinical Management

85. Wine JJ, Joo NS. Submucosal glands and airway defense. Proc Am 100. Mohsenin A, Blackburn MR. Adenosine signaling in asthma and
Thorac Soc 1: 4753, 2004. chronic obstructive pulmonary disease. Curr Opin Pulm Med 12:
86. Ballard ST, Spadafora D. Fluid secretion by submucosal glands of the 5459, 2006.
tracheobronchial airways. Respir Physiol Neurobiol 159: 27177, 2007. 101. Mohsenin A, Mi T, Xia Y, Kellems RE, Chen JF, Blackburn MR.
87. Fliegauf M, Omran H. Novel tools to unravel molecular mechanisms Genetic removal of the A2A adenosine receptor enhances pulmo-
in cilia-related disorders. Trends Genet 22: 24145, 2006. nary inammation, mucin production, and angiogenesis in adenosine
88. Knowles MR, Boucher RC. Mucus clearance as a primary innate deaminase-decient mice. Am J Physiol Lung Cell Mol Physiol 293:
defense mechanism for mammalian airways (Perspective). J Clin L753L761, 2007.
Invest 109: 57177, 2002. 102. Martin TR, Frevert CW. Innate immunity in the lungs. Proc Am
89. Sims DE, Westfall JA, Kiorpes AL, Horne MM. Preservation of tra- Thorac Soc 2: 40311, 2005.
cheal mucus by nonaqueous xative. Biotech Histochem 66: 17380, 1991. 103. Zaas AK, Schwartz DA. Innate immunity and the lung: Defense at
90. Matsui H, Grubb BR, Tarran R, Randell SH, Gatzy JT, Davis CW the interface between host and environment. Trends Cardiovasc Med
et al. Evidence for periciliary liquid layer depletion, not abnormal ion 15: 195202, 2005.
composition, in the pathogenesis of cystic brosis airways disease. Cell 104. Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate
95: 100515, 1998. immunity. Cell 124: 783801, 2006.
91. Tarran R, Boucher RC. Thin-lm measurements of airway surface 105. Shpacovitch V, Feld M, Hollenberg MD, Luger TA, Steinho, M.
liquid volume/composition and mucus transport rates in vitro. Meth Role of protease-activated receptors in inammatory responses, innate
Mol Med 70: 47992, 2002. and adaptive immunity. J Leukoc Biol 83: 130922, 2008.
92. Tarran R, Button B, Boucher RC. Regulation of normal and cystic 106. Wilmanski JM, Petnicki-Ocwieja T, Kobayashi KS. NLR proteins:
brosis airway surface liquid volume by phasic shear stress. Annu Rev Integral members of innate immunity and mediators of inammatory
Physiol 68: 54361, 2005. diseases. J Leuk Biol 83: 1330, 2008.
93. Tarran R, Grubb BR, Gatzy JT, Davis CW, Boucher RC. The relative 107. Takeuchi O, Akira S. MDA5/RIG-I and virus recognition. Curr Opin
roles of passive surface forces and active ion transport in the modu- Immunol 20: 1722, 2008.
lation of airway surface liquid volume and composition. J Gen Physiol 108. Wu Q , Lu Z, Verghese MW, Randell SH. Airway epithelial cell tol-
118: 22336, 2001. erance to Pseudomonas aeruginosa. Respir Res 6: 26, 2005.
94. Lazarowski ER, Tarran R, Grubb BR, van Heusden CA, Okada S, 109. Kato A, Schleimer RP. Beyond inammation: Airway epithelial
Boucher RC. Nucleotide release provides a mechanism for airway sur- cells are at the interface of innate and adaptive immunity. Curr Opin
face liquid homeostasis. J Biol Chem 279: 3685564, 2004. Immunol 19: 71120, 2007.
95. Tarran R. Regulation of airway surface liquid volume and mucus trans- 110. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells:
port by active ion transport. Proc Am Thorac Soc 1: 4246, 2004. Linking innate and adaptive immunity in asthma. Nat Rev Immunol
96. Tarran R, Trout L, Donaldson SH, Boucher RC. Soluble mediators, 8: 193204, 2008.
not cilia, determine airway surface liquid volume in normal and cystic 111. Schmidt-Weber CB, Akdis M, Akdis CA. TH17 cells in the big pic-
brosis supercial airway epithelia. J Gen Physiol 127: 591604, 2006. ture of immunology. J Allergy Clin Immunol 120: 24754, 2007.
97. Rollins BM, Burn M, Coakley RD, Chambers LA, Hirsh AJ, Clunes 112. Cohn L, Homer RJ, MacLeod H, Mohrs M, Brombacher F,
MT, Lethem MI, Donaldson SH, Tarran R. A2B Adenosine Receptors Bottomly K. Th2-induced airway mucus production is dependent on
Regulate the Mucus Clearance Component of the Lungs Innate IL4Ralpha, but not on eosinophils. J Immunol 162: 617883, 1999.
Defense System. Am J Respir Cell Mol Biol 2008. doi:10.1165/rcmb. 113. Gavett SH, OHearn DJ, Karp CL, Patel EA, Schoeld BH,
2007-0450OC. Finkelman FD et al. Interleukin-4 receptor blockade prevents airway
98. Bours MJ, Swennen EL, Di Virgilio F, Cronstein BN, Dagnelie PC. responses induced by antigen challenge in mice. Am J Physiol Lung
Adenosine 5-triphosphate and adenosine as endogenous signaling mol- Cell Mol Physiol 272: L253L261, 1997.
ecules in immunity and inammation. Pharmacol Ther 112: 358404, 114. Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, Pociask DA et al.
2006. IL-22 mediates mucosal host defense against Gram-negative bacte-
99. Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA, rial pneumonia. Nat Med 14: 27581, 2008.
Muskens F et al. Extracellular ATP triggers and maintains asthmatic 115. Pilette C, Durham SR, Vaerman JP, Sibille Y. Mucosal immunity in
airway inammation by activating dendritic cells. Nat Med 13: 91319, asthma and chronic obstructive pulmonary disease: A role for immu-
2007. noglobulin A? Proc Am Thorac Soc 1: 12535, 2004.

210
Airway Mucus Hypersecretion in
Asthma and COPD: Not the Same?
17 CHAPTER

Duncan F. Rogers
INTRODUCTION reects lack of asthma control, leading in turn to
accelerated loss of lung function and increased Section of Airway Disease,
mortality [7]. National Heart & Lung Institute,
Patients with asthma or chronic obstructive The association of airway mucus hyperse- Imperial College, London, UK
pulmonary disease (COPD) invariably exhibit cretion with asthma and COPD is not neces-
characteristics of airway mucus hypersecretion, sarily cause and eect. The excess mucus may
comprising sputum production [1, 2], excessive merely be a result of the inammatory proc-
mucus in the airway lumen [3] (Figs. 17.1 and esses causing the disease. Consequently, treating
17.2), goblet cell hyperplasia [36] and submu- the mucus problem will not inevitably treat the
cosal gland hypertrophy [3, 4] (Fig. 17.3). The disease, especially if the changes that have led
pathophysiological sequelae of mucus hyperse- to increased mucus production are irreversible.
cretion are airway obstruction, airow limitation, Nevertheless, as discussed above, mucus hyper-
ventilation-perfusion mismatch, and impair- secretion does contribute to clinical symptoms
ment of gas exchange. In addition, compro- in certain groups of patients with asthma and
mised mucociliary function, with reduced mucus COPD. This suggests that it is important to
clearance, can encourage bacterial colonization develop drugs that address the airway mucus
leading to repeated chest infections and exac- problem in these patients. The rst issue per-
erbations, particularly in COPD. In COPD, a taining to the rational development of drugs to
city-wide epidemiological study in Copenhagen treat airway mucus hypersecretion is to under-
showed that a number of clinical parameters, stand the nature of the underlying disease proc-
including deterioration in lung function, risk esses in asthma and COPD and how these
of hospitalization and death, were increased in relate to the mucus hypersecretory phenotype
patients with COPD who had chronic mucus in each condition. Secondly, it is important to
hypersecretion compared with patients with know which aspect(s) of mucus hypersecretion
either no sputum production or with only a should be targeted.
small amount of intermittent sputum produc- The present chapter addresses the above
tion [7] (Fig. 17.4). In addition, patients who issues by discussion of similarities and dier-
had chronic airway mucus hypersecretion and ences in the pulmonary inammatory prole
were also susceptible to chest infections showed between asthma and COPD and how these may
increased mortality and morbidity compared relate to the generation of the airway mucus
with patients with mucus hypersecretion but hypersecretory state in the two conditions. To
without infections (Fig. 17.4). In contrast, there set these discussions in context, the chapter
is much less information on the contribution of begins with consideration of sputum, airway
mucus to pathophysiology and clinical symp- mucus, and mucins, followed by an outline of
toms in asthma. However, excess mucus may not the characteristics of mucus hypersecretion in
only obstruct the airway lumen, but also contrib- asthma and COPD, emphasizing dierences
ute to the development of airway hyperrespon- between the two conditions and the theoreti-
siveness [8]. The latter possibilities led to the cal requirements for drugs aimed at eectively
suggestion that chronic mucus hypersecretion inhibiting airway mucus hypersecretion.

211
(A) (B)

M
M

(C) (D)

M
E

FIG. 17.1 Mucus obstruction of the airways in asthma and COPD. (A) Mucus plugging in asthma. Complete occlusion by mucus plugs (M) of an
intrapulmonary bronchus (arrow), cut in longitudinal section, in a patient who died of an acute-severe asthma attack. (B) Bronchoconstriction and luminal
mucus in fatal asthma. Intrapulmonary airway (transverse section) of a patient who died of an acute-severe asthma attack showing airway epithelium (arrow)
thrown into folds by smooth muscle contraction, and occlusion by mucus (M) of remaining luminal space. This relatively small amount of mucus would not be
expected to significantly reduce airflow in the relaxed, non-constricted airway. (C) Airway mucus tethering in asthma. Transverse section through a bronchiole
of a patient with asthma showing incomplete release, or continuity, of secreted mucin from airway epithelial goblet cells (arrows). Similar appearances of
mucus tethering are not observed in the airways in COPD. (D) Mucopurulent secretions (M) in an intrapulmonary bronchus in a patient with COPD.

(A) (C)
25
Airway level FIG. 17.2 Relative amounts of airway luminal
M
E ** mucus in asthma and COPD. (A) Lungs from patients
20 Central * dying either of causes other than lung disease
Distal * (controls, C), or from chronic asthma (Chr. A),
acute-severe asthma (A-S. A), emphysema (Emph.),
15 or chronic bronchitis (CB), were cut in histological
MOR

section and stained with hematoxylineosin and


Image elasticaGoldner stains to visualize mucin (M). The
analysis 10 stained sections were viewed by light microscopy
and intrapulmonary airways assessed for amount
(B) * * of luminal mucus. E: bronchial epithelium. (B)
* Airways of interest were subjected to computer-
5
based image analysis. The irregular perimeter of
AM the bronchial epithelium was digitally converted
AB to a circle, from which the area of mucus (AM) was
0
C Chr. A A-S. A Emph. CB expressed as a ratio of the area of the bronchus (AB),
(n 4) (n 5) (n 3) (n 7) (n 6) to give a mucus-occupying ratio, MOR (AM/AB), as
shown in panel C. *p  0.01, **p  0.01 compared
Patient group with controls. Redrawn from data in Refs [3, 4].
212
Airway Mucus Hypersecretion in Asthma and COPD: Not the Same? 17
with infection and inammation. Inammation leads to
SPUTUM mucus hypersecretion, ciliary dysfunction, and changes in the
composition and biophysical properties of airway secretions
Coughing or hawking to bring up phlegm is a sign of [12]. Inammatory cells, particularly neutrophils, which are
respiratory disease [2, 9]. It indicates airway mucus hyperse- recruited to the airway to combat infection, disappear from
cretion, coupled with impaired mucus clearance and mucus the airway either through programmed cell death (apopto-
retention, and is a feature of many patients with asthma [10] sis) or by necrosis. Necrotic neutrophils release proinam-
or COPD [11]. The expectorated excessive secretions are matory mediators that damage the epithelium and recruit
referred to as sputum. The characteristics of sputum change more inammatory cells. They also release DNA [13] and
lamentous actin (F-actin) from the cytoskeleton. DNA and
F-actin copolymerize to form a second rigid network within
(A) (C) airway secretions [14, 15]. Neutrophil-derived myeloperoxi-
30 dase imparts a characteristic green color to inamed airway
* * secretions, which are termed mucopurulent (17.1). Excessive
** infection turns the secretions dark yellow, green, or brown,
and these are termed purulent.
20
Gland (%)

Histology
(B)
AIRWAY MUCUS
10
Airway mucus is a complex dilute aqueous solution of lipids,
G glycoconjugates, and proteins. It comprises salts, enzymes
and anti-enzymes, oxidants and antioxidants, exogenous
Image analysis bacterial products, endogenous antibacterial secretions, cell-
0
C Emph. CB Chr. A A-S. A derived mediators and proteins, plasma-derived mediators
and proteins, and cell debris such as DNA. Airway mucus
FIG. 17.3 Relative size of airway submucosal glands in asthma and COPD. is considered to form a liquid bi-layer whereby an upper
(A) Lungs from patients dying either of causes other than lung disease gel layer oats above a lower, more watery sol, or pericili-
(controls, C), or from chronic asthma (Chr. A), acute-severe asthma (A-S. A), ary liquid, layer [16] (Fig. 17.5). There may be a thin layer
emphysema (Emph.), or chronic bronchitis (CB), were cut in histological
of surfactant lying in between, and separating, the gel and
section and stained with hematoxylineosin and elasticaGoldner stains
to visualize the submucosal glands. The stained sections were viewed by
sol layers [17]. The functions of the sol layer are debated,
light microscopy and intrapulmonary airways assessed for size of glands. but are presumed to include lubrication of the beat-
(B) Airways of interest were subjected to computer-based image analysis, ing cilia. The surfactant layer might facilitate spreading of
and the area proportion of glands (G) expressed as a percentage of the wall the gel layer over the epithelial surface. The gel layer traps
area, as shown in panel C. *p  0.01, **p  0.01 compared with controls. particles and is moved on the tips of the beating cilia.
Redrawn from data in Refs [3, 4]. The inhaled particles are trapped in the sticky gel layer

(A) Lung function (B) Hospitalization


25 5
Risk of hospitalization

Men Women Men Women


Excess decline in

20
FEV1 (ml/year)

4
15
10 3
5 2
0
1
5
No M M CMH No M M CMH No M M CMH No M M CMH

(C) Mortality FIG. 17.4 Clinical impact of airway mucus


(D) Infection  mortality
hypersecretion in COPD. The Copenhagen Heart
500 4 Study [7] showed that patients with chronic mucus
(relative risk: log)

hypersecretion (CMH), as defined by long-term phlegm


COPD death

(relative risk)
COPD death

50 production, had an accelerated decline in lung


CMH 3
function (A), increased risk of hospitalization (B),
increased risk of death (C), and a further increased
5 2 risk of death when linked with pulmonary infection
CMH
(D), when compared with subjects without mucus
1 1 production (CMH) or patients with some sputum
production but without infection. No M: no sputum
80 60 40 20 CMH CMH CMH (mucus) production; M: non-chronic sputum (mucus)
FEV1 (% predicted) infection production. Redrawn after data in Ref. [7].

213
Asthma and COPD: Basic Mechanisms and Clinical Management

and are removed from the airways by mucociliary clear- absorb energy when moving. Elasticity is a solid-like prop-
ance. When the mucus reaches the throat, it is either erty and is the capacity to store the energy used to move
swallowed and delivered to the gastrointestinal tract for or deform it. Viscoelasticity confers a number of properties
degradation or, if excessive, as in respiratory disease, it is upon the mucus that allow eective interaction with cilia.
coughed out [18]. These properties have been variously described in terms of
Respiratory tract mucus requires the correct combi- spinnability, adhesiveness, and wettability [21]. An impor-
nation of viscosity and elasticity for optimal eciency of tant characteristic of mucus is that it is non-Newtonian:
ciliary interaction [19, 20]. Viscosity is a liquid-like char- its viscosity decreases as the applied force increases [22].
acteristic and is the resistance to ow and the capacity to Consequently, the ratio of stress to rate of strain is non-
linear, with the result that the more forcefully the cilia beat,
the more easily the mucus moves. Viscoelasticity is con-
Mucociliary ferred on the mucus primarily by high molecular weight
transport
mucous glycoproteins, termed mucins.

Surfactant
Gel
Sol } Mucus
Epithelium

Ciliated
RESPIRATORY TRACT MUCINS
Plasma Goblet Basal
cell
exudation cell cell
In health, mucins comprise up to 2% by weight of the air-
way mucus [23]. In the airways, mucins are produced by
goblet cells in the epithelium [24] and sero-mucous glands
Submucosal gland
in the submucosa [25], which in respiratory diseases such
as asthma and COPD may increase in number (hyperpla-
sia) and size (hypertrophy) respectively (Fig. 17.5). Mucins
are long, thread-like, complex glycoconjugates. They con-
Asthma sist of a linear peptide backbone (termed apomucin), which
Inflammation is encoded by specic mucin (MUC) genes (see below), to
COPD
which hundreds of carbohydrate side-chains are O-linked,
but also with additional N-linked glycans [26]. The glyco-
sylation pattern is complex and extremely diverse [27], and is
associated with complimentary motifs on bacterial cell walls,
thereby facilitating broad-spectrum bacterial attachment
and subsequent clearance [28, 29]. Within the main pro-
tein core are variable numbers of tandemly repeated serine-
and/or threonine-rich regions which are unique in size and
sequence for each mucin [18], and represent sites for mucin
glycosylation. These complex glycoproteins are polydis-
perse, linear polymers that can be fragmented by reduction
to give monomers termed reduced subunits [3033].
There are at least two structurally and functionally distinct
classes of mucin, namely the membrane-associated mucins
and the secreted (gel-forming or non-gel-forming) mucins
(Table 17.1). Membrane-tethered mucins, which have a
hydrophobic domain that anchors the mucin in the plasma
FIG. 17.5 Airway mucus secretion and hypersecretion. Upper panel:
membrane, contribute to the composition of the cell surface
In healthy airways, mucus forms a bi-layer over the epithelium, with [18]. Secretory mucins are stored intracellularly in secretory
surfactant (dotted line) separating the gel and sol layers. Mucins secreted granules and are released at the apical surface of the cell in
by goblet cells and submucosal glands confer viscoelasticity on the mucus
which facilitates mucociliary clearance of inhaled particles and irritants.
Mucus hydration is regulated by salt (and, hence water) flux across the
TABLE 17.1 Human mucin genes expressed in the airways and lung.
epithelium: the glands also secrete water. Plasma proteins exuded from the
tracheobronchial microvasculature bathe the submucosa and contribute
Classification of gene product Genes
to the formation of the mucus. The above processes are under the control
of nerves and regulatory mediators. Lower panel: Airway inflammation
Membrane associated MUC1, MUC4, MUC11, MUC13,
(in asthma and COPD) induces changes associated with a mucus
MUC20
hypersecretory phenotype, including increased plasma exudation (more
prominent in asthma than COPD), goblet cell hyperplasia, via differentiation Secreted, cysteine rich (gel MUC2, MUC5AC, MUC5B, MUC19
from basal cells, and associated increased mucus synthesis and secretion,
forming)
and submucosal gland hypertrophy (with associated increased
mucus production), leading to increased luminal mucus (and airway Secreted, cysteine poor MUC7
obstruction).

214
Airway Mucus Hypersecretion in Asthma and COPD: Not the Same? 17
response to stimuli. It would appear that mucus production
is such a fundamental homeostatic process that virtually all
MUCIN GENES AND GENE PRODUCTS
acute interventions examined trigger airway mucin secre-
tion (Table 17.2). In addition, many of these same mediators Twenty human mucin (MUC) genes have so far been
when administered more chronically not only induce mucin identied. Of these, only nine, namely MUC1, MUC2,
secretion but also upregulate mucin gene expression, with MUC4, MUC5AC, MUC5B, MUC7, MUC11, MUC13,
concomitant increases in mucin synthesis and goblet cell and MUC20 are expressed in the human respiratory tract
hyperplasia (Table 17.2). [18] (Table 17.1). Of these, only MUC2, MUC5AC, and
MUC5B, the classic gel-forming mucins, are found in air-
way secretions. However, only MUC5AC and MUC5B
glycoproteins, localized adjacent to each other on chro-
TABLE 17.2 Inducers of airway mucus secretion, goblet cell hyperplasia mosome 11p15.5, are considered the major gel-forming
and mucin (MUC) gene expression/mucin synthesis. mucins in both normal respiratory tract secretions as well as
in airway secretions from patients with respiratory diseases
Stimulation Secretion Hyperplasia MUC [3439]. Small amounts of MUC2 may, however, be found
Cytokines in secretions from irritated airways (see below).
Advances in biochemistry, molecular biology, and
IL-1  NP NP biophysics mean that the structure and functions of the air-
IL-6  NP Yes way MUC gene products are becoming better characterized
IL-9 NP NP Yes [23, 26]. The predicted sequences of the MUC1, MUC 4,
MUC11, MUC13, and MUC20 mucins suggest they are
IL-13 (IL-4)  Yes Yes
membrane bound, with an extracellular mucin domain and
TNF-  Yes* Yes* a hydrophobic membrane-spanning domain (Table 17.1).
In contrast, MUC2, MUC5AC, MUC5B, MUC6, and
Gases
MUC7 gene products are secreted mucins (Table 17.1).
Cigarette smoke, ozone  Yes Yes The technology for studying the contribution to physiology
Nitric oxide / NP NP and pathophysiology of the individual MUC gene prod-
ucts lags well behind that of investigation of gene expres-
Reactive oxygen 0/ NP NP
sion [18]. MUC1, MUC2, and MUC8 genes are expressed
species
in both the epithelium and submucosal glands, whereas
Inflammatory mediators MUC4, MUC5AC, and MUC13 are expressed primarily
in the epithelium. In contrast, MUC5B and MUC7 genes
Bradykinin  NP NP
are expressed primarily in the glands. Use of currently avail-
Cysteinyl leukotrienes  NP NP able antibodies conrms that the MUC5AC gene product
Endothelin 0/ NP NP is a goblet cell mucin, whilst MUC5B predominates in
Histamine  NP NP
the glands, albeit that some MUC5AC and MUC7 is also
usually present [23]. Interestingly, MUC4 mucin localizes
PAF  Yes* Yes* to the ciliated cells. The mucin content of secretions from
Prostaglandins 0/ NP NP patients with hypersecretory respiratory diseases may dier
Proteinases  Yes NP from normal (see below).
Purine nucleotides  NP NP

Neuronal pathways
MUC5AC
Cholinergic nerves  NP NP
Cholinoceptor agonists  Yes NP MUC5AC mucin, initially isolated as a tracheobronchial
Nicotine  Yes NP mucin [40], is found in airway secretions pooled from
healthy individuals [35, 37]. Increased levels of MUC5AC
Tachykininergic nerves  NP NP
protein have also been shown to be present in the airways of
Substance P  NP NP patients with asthma [5], which suggests that this mucin may
Neurokinin A  NP NP contribute to the pathophysiology of asthma. MUC5AC is
the main mucin produced by the goblet cells in the tracheo-
Miscellaneous bronchial surface epithelium. However, MUC5AC can be
EGF (TNF-) NP Yes Yes found highly expressed not only in human bronchial epithe-
lium, but also in bronchial submucosal glands, nasal mucosa,
Sensitization followed  Yes Yes gastric epithelium, endocervix epithelium, and submucosal
by challenge glands. This mucin has been found to be highly oligomer-
Notes: : highly potent; : marked effect; : lesser effect; 0: minimal effect; EGF: ized, which makes it an ideal gel-forming molecule. The
epidermal growth factor; IL: interleukin; NP: effect not published. expression of many genes, such as MUC5AC, in airway epi-
*
Effect only observed with PAF (platelet activating factor) and TNF- (tumor necrosis thelial cells is regulated by various neurohumoral factors and
factor-) in combination. inammatory mediators (Table 17.2).

215
Asthma and COPD: Basic Mechanisms and Clinical Management

MUC5B (A)

MUC5B mucins are also a major component of tenacious Chronic


mucus plug from the lungs of a patient who died in sta- bronchitis
tus asthmaticus [38, 41] and in sputum from patients with
chronic bronchitis [39], which suggests that MUC5B is
a major component of lung mucus from patients with
obstructive lung diseases [42]. MUC5B mucin exists as dif-
ferently charged glycoforms (termed the low-charge and Small
high-charge glycoforms) and is secreted primarily by the airways Emphysema
disease
mucous cells in the bronchial submucosal glands [34, 38, 43,
44]. However, it has been shown that MUC5B mucins are
also synthesized by goblet cells [39], and are expressed in
the tracheal and bronchial glands, salivary glands, endocer-
(B) (C)
vix, gall bladder, and pancreas. MUC5B is unique in that it
does not appear to be polymorphic.
From the above, it appears that, in healthy individu- CB
als, MUC5B is mainly expressed in the airway submucosal CB
glands, which are restricted to the more proximal, cartilagi-
nous airways. In contrast, MUC5AC expression is generally SAD E
restricted to goblet cells in the upper and lower respiratory SAD E
tracts [45, 46]. Thus, the composition of normal mucus can
be altered depending on the relative contribution to the
secretions of these dierent cellular sources [47]. In respira- FIG. 17.6 The pathophysiological components of COPD. (A) COPD
tory diseases associated with airway mucus hypersecretion, comprises three interlinked conditions, namely chronic bronchitis
such as asthma and COPD, further changes in the compo- (CB: long-term airway mucus hypersecretion), small airways disease
sition of the mucus, and in the mucus secretory phenotype (SAD: also known as chronic bronchiolitis), and emphysema (E: alveolar
destruction). The relative contribution to airway obstruction in any one
in general, are observed, as discussed below.
patient of each component is invariably unclear. (B) Patient in whom
mucus hypersecretion predominates (may be identifiable by excessive
sputum production). (C) Patient in whom mucus hypersecretion
contributes proportionally less to airflow limitation than SAD and E.

AIRWAY MUCUS HYPERSECRETORY


PHENOTYPE IN COPD AIRWAY MUCUS HYPERSECRETORY
PHENOTYPE IN ASTHMA
COPD comprises three overlapping conditions, namely
chronic bronchitis (airway mucus hypersecretion), chronic Asthma is a chronic inammatory condition of the airways
bronchiolitis (small airways disease), and emphysema (airspace characterized by variable airow limitation that is at least
enlargement due to alveolar destruction) [10] (Fig. 17.6). The partially reversible, either spontaneously or with treatment
following discussion considers the bronchitic component of [55, 56]. It has specic clinical and pathophysiological
COPD. The airways of patients with COPD contain excessive features [57], including mucus obstruction of the airways
amounts of mucus [6], which is markedly increased above [8]. The latter is particularly evident in a proportion of
that in control subjects [3, 48]. The excessive luminal mucus is patients who die in status asthmaticus, where many airways
associated with increased amounts of mucus-secreting tissue. are occluded by mucus plugs [5860]. The plugs are highly
Goblet cell hyperplasia is a cardinal feature of chronic bron- viscous and comprise plasma proteins, DNA, cells, prote-
chitis [6], with increased numbers of goblet cells in the air- oglycans [61], and mucins [41, 58, 61]. Incomplete plugs
ways of cigarette smokers either with chronic bronchitis and are found in the airways of asthmatics who have died from
chronic airow limitation [49] or with or without productive causes other than asthma [62], which indicates that plug
cough [50]. Submucosal gland hypertrophy also characterizes formation is a chronic, progressive process. There is also
chronic bronchitis [3, 6, 51, 52], and the amount of gland cor- more mucus in the central and peripheral airways of both
relates with amount of luminal mucus [3]. chronic and severe asthmatics compared with control sub-
The number of ciliated cells and the length of indi- jects [4]. Analysis of asthmatic sputum indicates that the
vidual cilia is decreased in patients with chronic bronchitis mucus comprises DNA, lactoferrin, eosinophil cationic pro-
[53]. Ciliary aberrations include compound cilia, cilia with tein, and plasma proteins such as albumin and brinogen
an abnormal axoneme or intracytoplasmic microtubule dou- [6365], as well as mucins [5, 64, 66]. The increased amount
blets, and cilia enclosed within periciliary sheaths [54]. These of lumenal mucus reects an increase in amount of airway
abnormalities coupled with mucous hypersecretion are pre- secretory tissue, due to both goblet cell hyperplasia [4, 5]
sumably associated with reduced mucus clearance and airway and submucosal gland hypertrophy [58], although the latter
mucus obstruction in the bronchitic component of COPD. is not characteristic of all patients with asthma [4].

216
Airway Mucus Hypersecretion in Asthma and COPD: Not the Same? 17
Airway epithelial fragility, with epithelial shedding chronic bronchitis [80]. Conversely, cough is not so eec-
in extreme cases, is a signicant feature of asthma [67]. tive in COPD patients with impaired lung elastic recoil.
Shedding includes loss of ciliated cells, with presumably a
concomitant reduction in mucus clearing capacity. As in
COPD above, abnormalities in airway ciliated cells and cilia Asthma
have been described in asthma. The ciliated cells themselves
may be damaged, with loss of cilia, vacuolization of the endo- Airway mucociliary clearance is well documented as being
plasmic reticulum and mitochondria, and microtubule dam- impaired in asthma [72]. Clearance is impaired even in
age [6870]. These abnormalities could be caused by some patients in remission [81] and in those with mild stable
of the inammatory mediators generated in the airways of disease [82]. Mucus clearance is proportionally reduced in
asthmatic patients, for example, eosinophil major basic pro- symptomatic asthmatics [83] and during exacerbations [84].
tein [71]. A variety of other ciliostatic and ciliotoxic com- In addition, the normal slowing of mucus clearance during
pounds are also present in asthmatic airway secretions [72]. sleep is more pronounced in asthmatic patients [85, 86],
In summary of this section, the combination of an and this could be a contributory factor in nocturnal asthma.
increased amount of mucus-secreting tissue, with associ- The mechanisms underlying the reduced mucus clearance
ated mucus hypersecretion, the production of viscid mucus, in asthma are not clearly dened, although airway inam-
and abnormal ciliary function leads to reduced mucus clear- mation is considered to be a major contributor [72].
ance and the development of airway mucus obstruction
in asthma.

MECHANISMS OF DEVELOPMENT OF AIRWAY


MUCOCILIARY CLEARANCE IN GOBLET CELL HYPERPLASIA
ASTHMA AND COPD
Airway goblet cell hyperplasia is a predominant feature of
Clearance of mucus from the airways is impaired in patients asthma and COPD (see above), and is an often-used end
with a variety of respiratory diseases, including asthma point in animal models of respiratory disease [87]. The cel-
and COPD [73]. However, it should be noted that there lular composition of the airway epithelium can alter both by
are often discrepancies in results between studies that are cell division and by dierentiation of one cell into another
invariably due to dierences in methodology [74, 75], but [88]. There are at least eight cell types in the airway epithe-
may also be due to observations made at dierent stages of lium of the conducting airways. Of these, the basal, serous,
disease. and Clara cells are considered progenitor cells with the
capacity to undergo division followed by dierentiation into
mature ciliated or goblet cells. In specic experimental
COPD conditions, for example, exposure to cigarette smoke, goblet
cell division contributes in part to the hyperplasia. However,
Mucus clearance is generally considered to be impaired in dierentiation of non-granulated airway epithelial cells is a
patients with COPD [73]. However, the validity of these major route for production of new goblet cells [8890]. In
studies is dependent upon patient selection and the exclu- experimental animals, production of goblet cells is usually at
sion of patients with asthma. For example, patients clas- the expense of the progenitor cells, most notably serous and
sied as having obstructive chronic bronchitis and with Clara cells, which decrease in number as goblet cell numbers
a bronchial reversibility of less than 15% had slower lung increase. Serous-like cells and Clara cells are found in macro-
mucus clearance than patients with reversibility greater than scopically normal bronchioles in human lung [91]. Whether
15% and who, therefore, were likely to be asthmatic [76]. there is a reduction in number in respiratory disease is not
Nevertheless, mucus clearance is signicantly reduced in reported, but merits investigation. Reduction in the relative
heavy smokers [77] and in patients with chronic bronchitis proportion of serous and Clara cells has pathophysiological
[78]. Lung mucus clearance diers between patients with signicance because they produce a number of antiinam-
chronic airway obstruction, with or without emphysema matory, immunomodulatory, and antibacterial molecules vital
[79]. Both groups of patients were smokers or ex-smokers to host defence [92, 93]. For example, serous cells produce
and had productive cough, but lung elastic recoil pressure lysozyme, lactoferrin, the secretory component of IgA, per-
was reduced in the emphysema group. Mucus clearance oxidases, and at least two protease inhibitors. Clara cells pro-
from central airways was similar. In contrast, clearance from duce Clara cell 10-kDa protein, also known as uteroglobulin,
the peripheral lung was faster in the emphysema group than Clara cell 55-kDa protein, Clara cell tryptase, -galactoside-
in the patients without emphysema. Importantly, forced binding lectin, possibly a specic phospholipase, and sur-
expirations and cough markedly increased peripheral clear- factant proteins A, B, and D. Thus, in respiratory diseases
ance in the non-emphysema group but not in the emphy- associated with airway mucus hypersecretion it seems that not
sema group. Comparable ndings in a subsequent study only is there goblet cell hyperplasia, with associated mucus
led to the suggestion that cough compensates relatively hypersecretion, but also a reduction in serous and Clara cells,
eectively for decreased mucus clearance in patients with with concomitant potential for impaired host defence.

217
Asthma and COPD: Basic Mechanisms and Clinical Management

DIFFERENCES IN MUCUS HYPERSECRETORY many common features, there are specic characteristics
unique to each condition [9496]. These dierences in turn
PHENOTYPE BETWEEN ASTHMA AND COPD may contribute to dierences in pathophysiology of airway
mucus hypersecretion between the two conditions.
In order to develop appropriate models of airway mucus Asthma is invariably an allergic disease that aects
obstruction and devise drugs to aid mucus clearance, it is the airways, rather than the lung parenchyma, and is char-
necessary to understand the similarities and dierences in acterized by Th2-lymphocyte orchestration of pulmonary
the features of mucus obstruction for dierent hypersecre- eosinophilia. The reticular layer beneath the basement
tory conditions. There are a number of dierences in the membrane is markedly thickened and the airway epithelium
pathophysiology of airway mucus hypersecretion between is fragile, features not usually associated with COPD. The
asthma and COPD (Fig. 17.7). bronchial inammatory inltrate comprises activated T-cells
(predominantly CD4 cells) and eosinophils. Neutrophils
are generally sparse in stable disease. In contrast, COPD is
Airway inflammation in asthma and COPD currently perceived as predominantly a neutrophilic disor-
der governed largely by macrophages and epithelial cells. It
Any dierences in airway mucus hypersecretory pheno- is associated primarily with cigarette smoking. Three con-
type are presumably related, either directly (genetic pre- ditions comprise COPD, namely mucous hypersecretion,
disposition) or indirectly (environmental exposure), or bronchiolitis, and emphysema. The latter two features are
a combination of the two, to dierences in the airway not associated with asthma. In addition, and in contrast to
inammatory phenotype. Thus, although the underlying asthma, CD8 T-lymphocytes predominate and pulmonary
pulmonary inammation of asthma and COPD shares eosinophilia is generally associated with exacerbations.

Asthma
Normal lcgf MUC5B  MUC5AC  MUC2
MUC5B  MUC5AC

Mucous Plasma
cells exudation
Goblet Ciliated
cell cell

Serous Inflammation
cells

COPD
lcgf MUC5B  MUC5AC  MUC2
Bacterium

Inflammation

FIG. 17.7 Putative differences in the airway mucus hypersecretory phenotype between asthma and COPD. Compared with normal, in asthma, there is
airway inflammation (predominantly eosinophils and Th2 lymphocytes), an increased amount of luminal mucus with an increased content of MUC5AC and
MUC5B mucins, with the probability of an increased ratio of the low-charge glycoform (lcgf ) of MUC5B to MUC5AC, the appearance of small amounts of
MUC2 in the secretions, epithelial fragility with loss of ciliated cells, marked goblet cell hyperplasia, submucosal gland hypertrophy (although, in contrast
to COPD see below without a marked increase in mucous cell to serous cell ratio), tethering of mucus to goblet cells, and plasma exudation. In COPD,
there is airway inflammation (predominantly macrophages and neutrophils), increased luminal mucus, increased amounts of MUC5AC and MUC5B mucins,
an increased ratio of lcgf MUC5B to MUC5AC above that in asthma, small amounts of MUC2, goblet cell hyperplasia, submucosal gland hypertrophy (with an
increased proportion of mucous to serous cells), and a susceptibility to bacterial infection. Many of these differences require further investigation in greater
number of subjects.

218
Airway Mucus Hypersecretion in Asthma and COPD: Not the Same? 17
Both asthma and COPD have a characteristic port- It is noteworthy that it is COPD, a disease in which patients
folio of inammatory mediators and enzymes, many of are prone to infection [11], that has the proportional reduc-
which dier between the two conditions [97, 98]. At a sim- tion in serous cells, rather than asthma, a condition in which
plied level, histamine, interleukin (IL)-4, and eotaxin are patients are not so notably prone to chest infection.
associated with asthma, whilst IL-8, neutrophil elastase, and
matrix metalloproteinases are associated with COPD. Thus,
there are specic dierences between asthma and COPD Goblet cells in asthma and COPD
in their airway inammation and remodeling. These dier-
ences may in turn exert dierent inuences on the devel- In contrast to normal airways, goblet cells in the airways
opment of airway mucus obstruction in the two conditions from patients with COPD contain not only MUC5AC
(Fig. 17.7). but also MUC5B [39, 104] and MUC2 [23, 105]. This
distribution is dierent to that in the airways of patients
with asthma, where MUC5AC and MUC5B show a simi-
Airway mucus in asthma and COPD lar histological pattern to normal controls [106, 107]. It
is noteworthy that although MUC2 is located in goblet
Airway mucus in asthma is more viscous than in COPD, cells in irritated airways, and MUC2 mRNA is found in
with the airways of asthmatic patients tending to develop, the airways of smokers [48], MUC2 mucin is either not
and subsequently become blocked by, gelatinous mucus found in airway secretions from normal subjects or patients
plugs [8]. Whether or not mucus in asthma has an intrinsic with chronic bronchitis [35], or is found only in very small
biochemical abnormality is unclear. In general terms, spu- amounts in asthma and COPD [47, 108]. The signicance
tum from patients with asthma is more viscous than that of the above combined observations is unclear, but suggests
from patients with chronic bronchitis [65, 99, 100]. Mucus that there are dierences in goblet cell phenotype between
plugs in asthma dier from airway mucus gels in chronic asthma and COPD.
bronchitis in that they are stabilized by non-covalent inter-
actions between extremely large mucins assembled from
conventional sized subunits [41]. This suggests an intrinsic Submucosal glands in asthma and COPD
abnormality in the mucus due to a defect in assembly of the
mucin molecules, and could account for the increased vis- A notable dierence between asthma and COPD is in the
cosity of the mucus plugs in asthma. Plug formation may bronchial submucosal glands [109]. In asthma, although
also be due, at least in part, to increased airway plasma hypertrophied, the glands are morphologically normal with
exudation in asthma compared with COPD [101, 102]. In an even distribution of mucous and serous cells. In contrast,
addition, in direct contrast to COPD, exocytosed mucins in in chronic bronchitis, gland hypertrophy is characterized
asthma are not released fully from the goblet cells, leading by a markedly increased number of mucous cells relative
to tethering of luminal mucins to the airway epithelium to serous cells, particularly in severe bronchitis. The reduc-
[103]. This tethering may also contribute to plug formation. tion in number of gland serous cells may have clinical sig-
One explanation of mucus tethering is that neutrophil pro- nicance. The serous cells are a rich source of antibacterial
teases, the predominant inammatory cell in COPD [10], enzymes such as lysozyme and lactoferrin [25]. Thus, the
cleave goblet cell-attached mucins. In asthma, the inam- airway mucus layer in COPD patients may have a reduced
matory cell prole, predominantly airway eosinophilia [57], antibacterial capacity compared with that in asthma. This
does not generate the appropriate proteases to facilitate reduction, coupled with the change in MUC5B glycoforms
mucin release. in COPD (see above), could further explain, at least in part,
the much higher incidence of bacterial chest infections in
COPD compared with asthma.
Mucin species in asthma and COPD
Dierent mucin species, or at least dierent proportions of Ciliated cells in asthma and COPD
these species, appear to be present in respiratory tract secre-
tions in COPD and asthma. MUC5AC and a low-charge It is not clear whether or not there are dierences in the
glycoform of MUC5B are the major mucin species in air- airway ciliary abnormalities between COPD and asthma
way secretions from patients with COPD and asthma [35 (see above). However, epithelial fragility and shedding are
37, 39, 47]. There is signicantly more of the low-charge features of asthma rather than COPD [95], which suggests
glycoform of MUC5B in the respiratory diseases than in that there may be greater loss of, and damage to, the ciliated
normal control secretions [47]. An interesting dierence cells in asthma compared with COPD.
between the disease conditions is that there is a propor- From the above, it may be seen that there are theo-
tional increase in the MUC5B mucin over the MUC5AC retical and actual dierences in the nature of airway mucus
mucin in airway secretions from patients with COPD com- obstruction between COPD and asthma. How these relate
pared with secretions from patients with asthma [37]. The to pathophysiology and clinical symptoms in the two con-
above data require conrmation in more samples. The sig- ditions is, for the most part, unclear. However, these dissim-
nicance of the change in MUC5B glycoforms between the ilarities indicate that dierent treatments are required for
dierent diseases is unclear. However, it may relate to dif- eective treatment of airway mucus obstruction in dierent
ferences in propensity of bacterial colonization of the lungs. respiratory diseases.

219
Asthma and COPD: Basic Mechanisms and Clinical Management

inhaled insult. However, abnormal production of mucus


POTENTIAL THERAPIES FOR AIRWAY MUCUS can contribute to respiratory disease. Airway obstruction
HYPERSECRETION by mucus is a common feature of a number of severe res-
piratory conditions, including asthma and COPD. These
Current and potential therapies for asthma and COPD are diseases share pulmonary inammation and remodeling as
discussed elsewhere in this volume. However, there are a a pathophysiological characteristic, although with diering
number of avenues for specic treatment of airway mucus proles of inammatory cells and mediators. One aspect of
hypersecretion that are worthy of brief mention. Some the remodeling is airway mucus hypersecretion, associated
of the mechanisms for development of airway goblet cell with the shared features of goblet cell hyperplasia and sub-
hyperplasia and the associated mucus hypersecretory phe- mucosal gland hypertrophy. They also each have a number
notype in asthma and COPD are becoming clearer [8, 110]. of unique features that characterize their airway mucus
Many regulatory and inammatory mediators and enzymes obstruction. For example, plasma exudation, mucus plug for-
increase mucus secretion and induce MUC gene expression, mation, and mucus tethering are features of asthma, whereas
mucin synthesis, and goblet cell hyperplasia in experimental submucosal gland hypertrophy with a disproportionate
systems (Table 17.2). These mediators are intermediates in increase in the ratio of mucous to serous cells is a signicant
a cascade of pathophysiological events leading from initiat- feature in COPD. Understanding of the relative importance
ing factors (such as allergen exposure in asthma or cigarette of the dierences and similarities in the pathophysiology
smoking in COPD) setting up a chronic inammatory/ of the dierent mucus hypersecretory phenotypes between
repair response, which in turn leads to mucus hypersecre- these two respiratory diseases should lead to rational devel-
tion and associated airway obstruction and clinical symp- opment of pharmacotherapeutic interventions. At present,
toms (Figs 17.517.7). A small number of key molecules there is considerable interest in inhibitors of EGF receptor
may be involved in translating the actions of the dier- tyrosine kinases, MEK/ERK and CLCA channels to sup-
ent inammatory mediators into airway mucus hyper- press goblet cell hyperplasia, and in molecules to selectively
secretion, namely epidermal growth factor (EGF) and interfere with mucin exocytosis (such as MARCKS-related
its receptor tyrosine kinase signaling pathway [111], the peptide and retargeted clostridial endopeptidases) and
mitogen-activated kinase kinase and extracellular signal- thereby inhibit airway mucus hypersecretion.
regulated kinase (MEK/ERK) pathway [112], calcium-
activated chloride (CLCA) channels [113, 114], and the
retinoic acid receptor (RAR)- signaling pathway [115]. A References
wide variety of small molecule antagonists and inhibitors of
these pathways are currently in pharmacotherapeutic devel- 1. Medical Research Council. Denition and classication of chronic bron-
opment [12]. In addition, inhibition of the process of mucin chitis for clinical and epidemiological purposes. Lancet i: 77579, 1965.
exocytosis, with concomitant inhibition of secretion and, 2. Openshaw PJ, Turner-Warwick M. Observations on sputum production
thereby, mucus hypersecretion, is currently receiving atten- in patients with variable airow obstruction; implications for the diag-
tion from two dierent directions. Firstly, MARCKS protein nosis of asthma and chronic bronchitis. Respir Med 83: 2531, 1989.
is a chaperone molecule essential for movement of newly 3. Aikawa T, Shimura S, Sasaki H, Takishima T, Yaegashi H, Takahashi T.
synthesized mucin-containing granules to the apical surface Morphometric analysis of intraluminal mucus in airways in chronic
of mucin-secreting cells [116]. Inhibition of MARCKS obstructive pulmonary disease. Am Rev Respir Dis 140: 47782, 1989.
by a MARCKS-related peptide inhibits mucin secretion 4. Aikawa T, Shimura S, Sasaki H, Ebina M, Takishima T. Marked gob-
let cell hyperplasia with mucus accumulation in the airways of patients
in vitro in human airway epithelial secretory cells [116] and
who died of severe acute asthma attack. Chest 101: 91621, 1992.
mucus hypersecretion in a mouse model of asthma [117]. 5. Ordonez CL, Khashayar R, Wong HH, Ferrando R, Wu R, Hyde
Secondly, SNARE complex proteins are essential for the DM, Hotchkiss JA, Zhang Y, Novikov A, Dolganov G, Fahy JV. Mild
docking of mucin granules to the internal apical surface and moderate asthma is associated with airway goblet cell hyperplasia
of mucin-secreting cells. We have developed a fully recom- and abnormalities in mucin gene expression. Am J Respir Crit Care Med
binant fusion protein comprising the translocation domain 163: 51723, 2001.
and endopeptidase of Clostridium botulinum, which nor- 6. Reid L. Pathology of chronic bronchitis. Lancet i: 27578, 1954.
mally enters nerves and cleaves SNARE proteins mediat- 7. Vestbo J. Epidemiological studies in mucus hypersecretion. Novartis
ing neurotransmission [118], with EGF to target EGF Found Symp 248: 312, 2002.
receptors on airway mucus-secreting cells. This molecule 8. Rogers DF. Airway mucus hypersecretion in asthma: An undervalued
pathology?. Curr Opin Pharmacol 4: 24150, 2004.
now cleaves SNARE proteins linking mucin granules with
9. Turner-Warwick M, Openshaw P. Sputum in asthma. Postgrad Med J
mucous cell apical membranes, and thereby inhibits mucin 63(Suppl 1): 7982, 1987.
secretion from human respiratory epithelial cells [119] and 10. Global Initiative for Asthma. Global strategy for asthma management
mucus hypersecretion in a rat model of COPD [120]. and prevention. www.ginasthma.org, 2006.
11. Global Initiative for Chronic Obstructive Lung Disease. Global strat-
egy for the diagnosis, management, and prevention of chronic obstruc-
tive pulmonary disease. www.goldcopd.org, 2006.
CONCLUSIONS 12. Rogers DF, Barnes PJ. Treatment of airway mucus hypersecretion. Ann
Med 38: 11625, 2006.
13. Lethem MI, James SL, Marriott C, Burke JF. The origin of DNA asso-
Production of airway mucus is a vital homeostatic mecha- ciated with mucus glycoproteins in cystic brosis sputum. Eur Respir J
nism that protects the respiratory tract from a barrage of 3: 1923, 1990.

220
Airway Mucus Hypersecretion in Asthma and COPD: Not the Same? 17
14. Shah SA, Santago P, Rubin BK. Quantication of biopolymer lament 38. Thornton DJ, Howard M, Khan N, Sheehan JK. Identication of
structure. Ultramicroscopy 104: 24454, 2005. two glycoforms of the MUC5B mucin in human respiratory mucus.
15. Tomkiewicz RP, Kishioka C, Freeman J, Rubin BK. DNA and actin Evidence for a cysteine-rich sequence repeated within the molecule.
lament ultrastructure in cystic brosis sputum. In: Baum G (ed.), J Biol Chem 272: 956166, 1997.
Cila, Mucus and Mucociliary Interactions, pp. 33341. New York: Marcel 39. Wickstrom C, Davies JR, Eriksen GV, Veerman EC, Carlstedt I.
Dekker, 1998. MUC5B is a major gel-forming, oligomeric mucin from human sali-
16. Knowles MR, Boucher RC. Mucus clearance as a primary innate vary gland, respiratory tract and endocervix: Identication of glyco-
defense mechanism for mammalian airways. J Clin Invest 109: 57177, forms and C-terminal cleavage. Biochem J 334: 68593, 1998.
2002. 40. Guyonnet DV, Audie JP, Debailleul V, Laine A, Buisine MP, Galiegue-
17. Morgenroth K, Bolz J. Morphological features of the interaction Zouitina S, Pigny P, Degand P, Aubert JP, Porchet N. Characterization
between mucus and surfactant on the bronchial mucosa. Respiration 47: of the human mucin gene MUC5AC: A consensus cysteine-rich
22531, 1985. domain for 11p15 mucin genes?. Biochem J 305: 21119, 1995.
18. Rose MC, Voynow JA. Respiratory tract mucin genes and mucin glyco- 41. Sheehan JK, Richardson PS, Fung DC, Howard M, Thornton DJ.
proteins in health and disease. Physiol Rev 86: 24578, 2006. Analysis of respiratory mucus glycoproteins in asthma: A detailed
19. King M. Experimental models for studying mucociliary clearance. Eur study from a patient who died in status asthmaticus. Am J Respir Cell
Respir J 11: 22228, 1998. Mol Biol 13: 74856, 1995.
20. King M. Physiology of mucus clearance. Paediatr Respir Rev 7(Suppl 1): 42. Rose MC, Nickola TJ, Voynow JA. Airway mucus obstruction: Mucin
S212S214, 2006. glycoproteins, MUC gene regulation and goblet cell hyperplasia. Am J
21. Houtmeyers E, Gosselink R, Gayan-Ramirez G, Decramer M. Respir Cell Mol Biol 25: 53337, 2001.
Regulation of mucociliary clearance in health and disease. Eur Respir J 43. Desseyn JL, Buisine MP, Porchet N, Aubert JP, Laine A. Genomic
13: 117788, 1999. organization of the human mucin gene MUC5B. cDNA and genomic
22. Sleigh MA, Blake JR, Liron N. The propulsion of mucus by cilia. Am sequences upstream of the large central exon. J Biol Chem 273: 30157
Rev Respir Dis 137: 72641, 1988. 64, 1998.
23. Davies JR, Herrmann A, Russell W, Svitacheva N, Wickstrm C, 44. Sharma P, Dudus L, Nielsen PA, Clausen H, Yankaskas JR,
Carlstedt I. Respiratory tract mucins: Structure and expression patterns. Hollingsworth MA, Engelhardt JF. MUC5B and MUC7 are dif-
Mucus Hypersecretion in Respiratory Disease, pp. 7688. Chichester: John ferentially expressed in mucous and serous cells of submucosal glands
Wiley & Sons, 2002. in human bronchial airways. Am J Respir Cell Mol Biol 19: 3037,
24. Rogers DF. Airway goblet cell hyperplasia in asthma: Hypersecretory 1998.
and anti-inammatory?. Clin Exp Allergy 32: 112427, 2002. 45. Audie JP, Janin A, Porchet N, Copin MC, Gosselin B, Aubert JP.
25. Finkbeiner WE. Physiology and pathology of tracheobronchial glands. Expression of human mucin genes in respiratory, digestive, and repro-
Respir Physiol 118: 7783, 1999. ductive tracts ascertained by in situ hybridization. J Histochem Cytochem
26. Thornton DJ, Rousseau K, McGuckin MA. Structure and function of 41: 147985, 1993.
the polymeric mucins in airways mucus. Annu Rev Physiol, 70: 459 46. Reid CJ, Gould S, Harris A. Developmental expression of mucin genes in
486, 2008. the human respiratory tract. Am J Respir Cell Mol Biol 17: 59298, 1997.
27. Hanisch FG. O-glycosylation of the mucin type. Biol Chem 382: 47. Kirkham S, Sheehan JK, Knight D, Richardson PS, Thornton DJ.
14349, 2001. Heterogeneity of airways mucus: Variations in the amounts and
28. Dell A, Morris HR. Glycoprotein structure determination by mass glycoforms of the major oligomeric mucins MUC5AC and MUC5B.
spectrometry. Science 291: 235156, 2001. Biochem J 361: 53746, 2002.
29. Moniaux N, Escande F, Porchet N, Aubert JP, Batra SK. Structural 48. Steiger D, Fahy J, Boushey H, Finkbeiner WE, Basbaum C. Use of
organization and classication of the human mucin genes. Front Biosci 6: mucin antibodies and cDNA probes to quantify hypersecretion in vivo
D1192206, 2001. in human airways. Am J Respir Cell Mol Biol 10: 53845, 1994.
30. Sheehan JK, Thornton DJ, Somerville M, Carlstedt I. Mucin structure. 49. Saetta M, Turato G, Baraldo S, Zanin A, Braccioni F, Mapp CE,
The structure and heterogeneity of respiratory mucus glycoproteins. Am Maestrelli P, Cavallesco G, Papi A, Fabbri LM. Goblet cell hyperplasia
Rev Respir Dis 144: S49, 1991. and epithelial inammation in peripheral airways of smokers with both
31. Thornton DJ, Davies JR, Kraayenbrink M, Richardson PS, Sheehan JK, symptoms of chronic bronchitis and chronic airow limitation. Am J
Carlstedt I. Mucus glycoproteins from normal human tracheobron- Respir Crit Care Med 161: 101621, 2000.
chial secretion. Biochem J 265: 17986, 1990. 50. Mullen JB, Wright JL, Wiggs BR, Pare PD, Hogg JC. Structure of
32. Thornton DJ, Devine PL, Hanski C, Howard M, Sheehan JK. central airways in current smokers and ex-smokers with and with-
Identication of two major populations of mucins in respiratory secre- out mucus hypersecretion: Relationship to lung function. Thorax 42:
tions. Am J Respir Crit Care Med 150: 82332, 1994. 84348, 1987.
33. Thornton DJ, Sheehan JK, Carlstedt I. Heterogeneity of mucus glyco- 51. Reid L. Measurement of the bronchial mucous gland layer: A diagnos-
proteins from cystic brotic sputum. Are there dierent families of tic yardstick in chronic bronchitis. Thorax 15: 13241, 1960.
mucins?. Biochem J 276: 67782, 1991. 52. Restrepo G, Heard BE. The size of the bronchial glands in chronic
34. Hovenberg HW, Davies JR, Carlstedt I. Dierent mucins are pro- bronchitis. J Pathol Bacteriol 85: 30510, 1963.
duced by the surface epithelium and the submucosa in human trachea: 53. Wanner A. Clinical aspects of mucociliary transport. Am Rev Respir
Identication of MUC5AC as a major mucin from the goblet cells. Dis 116: 73125, 1977.
Biochem J 318: 31924, 1996. 54. McDowell EM, Barrett LA, Harris CC, Trump BF. Abnormal cilia
35. Hovenberg HW, Davies JR, Herrmann A, Linden CJ, Carlstedt I. in human bronchial epithelium. Arch Pathol Lab Med 100: 42936,
MUC5AC, but not MUC2, is a prominent mucin in respiratory secre- 1976.
tions. Glycoconj J 13: 83947, 1996. 55. American Thoracic Society. Standards for the diagnosis and care of
36. Sheehan JK, Howard M, Richardson PS, Longwill T, Thornton DJ. patients with chronic obstructive pulmonary disease (COPD) and
Physical characterization of a low-charge glycoform of the MUC5B asthma. Am Rev Respir Dis 136: 22544, 1987.
mucin comprising the gel-phase of an asthmatic respiratory mucous 56. British Thoracic Society. The British guidelines on asthma manage-
plug. Biochem J 338: 50713, 1999. ment. Thorax 52(Suppl 1): S121, 1997.
37. Thornton DJ, Carlstedt I, Howard M, Devine PL, Price MR, Sheehan 57. Eapen SS, Busse WW. Asthma. Clin Allergy Immunol 16: 32553, 2002.
JK. Respiratory mucins: Identication of core proteins and glycoforms. 58. Dunnill MS. The pathology of asthma with special reference to changes
Biochem J 316: 96775, 1996. in the bronchial mucosa. J Clin Pathol 13: 2733, 1960.

221
Asthma and COPD: Basic Mechanisms and Clinical Management

59. Houston JC, De Navasquez S, Trounce JR. A clinical and pathological tracheobronchial clearance rate over three days in chronic bronchitis.
study of fatal cases of status asthmaticus. Thorax 8: 20713, 1953. Eur Respir J 8: 188693, 1995.
60. Saetta M, Di Stefano A, Rosina C, Thiene G, Fabbri LM. Quantitative 81. Pavia D, Bateman JR, Sheahan NF, Agnew JE, Clarke SW.
structural analysis of peripheral airways and arteries in sudden fatal Tracheobronchial mucociliary clearance in asthma: Impairment dur-
asthma. Am Rev Respir Dis 143: 13843, 1991. ing remission. Thorax 40: 17175, 1985.
61. Bhaskar KR, OSullivan DD, Coles SJ, Kozakewich H, Vawter GP, 82. Bateman JR, Pavia D, Sheahan NF, Agnew JE, Clarke SW. Impaired
Reid LM. Characterization of airway mucus from a fatal case of status tracheobronchial clearance in patients with mild stable asthma. Thorax
asthmaticus. Pediatr Pulmonol 5: 17682, 1988. 38: 46367, 1983.
62. Dunnill MS. The morphology of the airways in bronchial asthma. 83. Foster WM, Langenback EG, Bergofsky EH. Lung mucociliary func-
In: Stein M (ed.), New Directions in Asthma, pp. 21321. Park Ridge: tion in man: Interdependence of bronchial and tracheal mucus trans-
American College of Physicians, 1975. port velocities with lung clearance in bronchial asthma and healthy
63. Fahy JV, Liu J, Wong H, Boushey HA. Cellular and biochemical anal- subjects. Ann Occup Hyg 26: 22744, 1982.
ysis of induced sputum from asthmatic and from healthy subjects. Am 84. Messina MS, ORiordan TG, Smaldone GC. Changes in mucociliary
Rev Respir Dis 147: 112631, 1993. clearance during acute exacerbations of asthma. Am Rev Respir Dis
64. Fahy JV, Steiger DJ, Liu J, Basbaum CB, Finkbeiner WE, Boushey 143: 99397, 1991.
HA. Markers of mucus secretion and DNA levels in induced sputum 85. Bateman JR, Pavia D, Clarke SW. The retention of lung secretions
from asthmatic and from healthy subjects. Am Rev Respir Dis 147: during the night in normal subjects. Clin Sci Mol Med Suppl 55: 523
113237, 1993. 27, 1978.
65. Lopez-Vidriero MT, Reid L. Chemical markers of mucous and serum 86. Pavia D, Lopez-Vidriero MT, Clarke SW. Mediators and mucocili-
glycoproteins and their relation to viscosity in mucoid and purulent ary clearance in asthma. Bull Eur Physiopathol Respir 23(Suppl 10):
sputum from various hypersecretory diseases. Am Rev Respir Dis 117: 89s94s, 1987.
46577, 1978. 87. Rogers DF. In vivo preclinical test models for studying airway mucus
66. Lopez-Vidriero MT, Reid L. Bronchial mucus in health and disease. secretion. Pulm Pharmacol Ther 10: 12128, 1997.
Br Med Bull 34: 6374, 1978. 88. Ayers MM, Jeery PK. Proliferation and dierentiation in mamma-
67. Bousquet J, Jeery PK, Busse WW, Johnson M, Vignola AM. Asthma. lian airway epithelium. Eur Respir J 1: 5880, 1988.
From bronchoconstriction to airways inammation and remodeling. 89. Nadel JA, Burgel PR. The role of epidermal growth factor in mucus
Am J Respir Crit Care Med 161: 172045, 2000. production. Curr Opin Pharmacol 1: 25458, 2001.
68. Beasley R, Roche WR, Roberts JA, Holgate ST. Cellular events in the 90. Rogers DF. Airway goblet cells: Responsive and adaptable front-line
bronchi in mild asthma and after bronchial provocation. Am Rev Respir defenders. Eur Respir J 7: 1690706, 1994.
Dis 139: 80617, 1989. 91. Rogers AV, Dewar A, Corrin B, Jeery PK. Identication of serous-like
69. Carson JL, Collier AM, Fernald GW, Hu SC. Microtubular disconti- cells in the surface epithelium of human bronchioles. Eur Respir J 6:
nuities as acquired ciliary defects in airway epithelium of patients with 498504, 1993.
chronic respiratory diseases. Ultrastruct Pathol 18: 32732, 1994. 92. Basbaum CB, Jany B, Finkbeiner WE. The serous cell. Annu Rev
70. Laitinen LA, Heino M, Laitinen A, Kava T, Haahtela T. Damage of Physiol 52: 97113, 1990.
the airway epithelium and bronchial reactivity in patients with asthma. 93. Singh G, Katyal SL. Clara cell proteins. Ann NY Acad Sci 923: 4358,
Am Rev Respir Dis 131: 599606, 1985. 2000.
71. Gleich GJ, Loegering DA, Frigas E, Filley WV. The eosinophil granule 94. Djukanovic R. Airway inammation in asthma and its consequences:
major basic protein: Biological activities and relationship to bronchial Implications for treatment in children and adults. J Allergy Clin
asthma. Monogr Allergy 18: 27783, 1983. Immunol 109: S539S548, 2002.
72. Del Donno M, Bittesnich D, Chetta A, Olivieri D, Lopez-Vidriero 95. Jeery PK. Dierences and similarities between chronic obstructive
MT. The eect of inammation on mucociliary clearance in asthma: pulmonary disease and asthma. Clin Exp Allergy 29(Suppl 2): 1426,
An overview. Chest 118: 114249, 2000. 1999.
73. Wanner A, Salathe M, ORiordan TG. Mucociliary clearance in the 96. Saetta M, Turato G, Maestrelli P, Mapp CE, Fabbri LM. Cellular and
airways. Am J Respir Crit Care Med 154: 1868902, 1996. structural bases of chronic obstructive pulmonary disease. Am J Respir
74. Clarke SW, Pavia D. Lung mucus production and mucociliary Crit Care Med 163: 13049, 2001.
clearance: Methods of assessment. Br J Clin Pharmacol 9: 53746, 97. Barnes PJ. New treatments for COPD. Nat Rev Drug Discov 1:
1980. 43746, 2002.
75. Pavia D, Sutton PP, Agnew JE, Lopez-Vidriero MT, Newman SP, 98. Barnes PJ, Chung KF, Page CP. Inammatory mediators of asthma:
Clarke SW. Measurement of bronchial mucociliary clearance. Eur J An update. Pharmacol Rev 50: 51596, 1998.
Respir Dis Suppl 127: 4156, 1983. 99. Charman J, Reid L. Sputum viscosity in chronic bronchitis,
76. Moretti M, Lopez-Vidriero MT, Pavia D, Clarke SW. Relationship bronchiectasis, asthma and cystic brosis. Biorheology 9: 18599,
between bronchial reversibility and tracheobronchial clearance in 1972.
patients with chronic bronchitis. Thorax 52: 17680, 1997. 100. Shimura S, Sasaki T, Sasaki H, Takishima T, Umeya K. Viscoelastic
77. Goodman RM, Yergin BM, Landa JF, Golivanux MH, Sackner MA. properties of bronchorrhoea sputum in bronchial asthmatics.
Relationship of smoking history and pulmonary function tests to Biorheology 25: 17379, 1988.
tracheal mucous velocity in nonsmokers, young smokers, ex-smokers, and 101. Rogers DF. Physiology of airway mucus secretion and pathophysiol-
patients with chronic bronchitis. Am Rev Respir Dis 117: 20514, 1978. ogy of hypersecretion. Respir Care 52: 113446, 2007.
78. Agnew JE, Little F, Pavia D, Clarke SW. Mucus clearance from the 102. Rogers DF, Evans TW. Plasma exudation and oedema in asthma.
airways in chronic bronchitis Smokers and ex-smokers. Bull Eur Br Med Bull 48: 12034, 1992.
Physiopathol Respir 18: 47384, 1982. 103. Shimura S, Andoh Y, Haraguchi M, Shirato K. Continuity of airway
79. van der Schans CP, Piers DA, Beekhuis H, Koeter GH, van der Mark goblet cells and intraluminal mucus in the airways of patients with
TW, Postma DS. Eect of forced expirations on mucus clearance in bronchial asthma. Eur Respir J 9: 1395401, 1996.
patients with chronic airow obstruction: Eect of lung recoil pressure. 104. Chen Y, Zhao YH, Di YP, Wu R. Characterization of human mucin
Thorax 45: 62327, 1990. 5B gene expression in airway epithelium and the genomic clone of
80. Ericsson CH, Svartengren K, Svartengren M, Mossberg B, Philipson K, the amino-terminal and 5'-anking region. Am J Respir Cell Mol Biol
Blomquist M, Camner P. Repeatability of airway deposition and 25: 54253, 2001.

222
Airway Mucus Hypersecretion in Asthma and COPD: Not the Same? 17
105. Davies JR, Carlstedt I. Respiratory tract mucins. In: Salathe M and mucus production in bronchial epithelium of patients with
(ed.), Cilia and Mucus: From Development to Respiratory Defense, asthma. J Allergy Clin Immunol 109: 24650, 2002.
pp. 16778. New York: Marcel Dekker Inc., 2001. 114. Zhou Y, Shapiro M, Dong Q, Louahed J, Weiss C, Wan S, Chen Q,
106. Groneberg DA, Eynott PR, Lim S, Oates T, Wu R, Carlstedt I, Dragwa C, Savio D, Huang M, Fuller C, Tomer Y, Nicolaides NC,
Roberts P, McCann B, Nicholson AG, Harrison BD, Chung KF. McLane M, Levitt R. A calcium-activated chloride channel blocker
Expression of respiratory mucins in fatal status asthmaticus and mild inhibits goblet cell metaplasia and mucus overproduction. Mucus
asthma. Histopathology 40: 36773, 2002. Hypersecretion in Respiratory Disease, pp. 15065. Chichester: Wiley,
107. Groneberg DA, Eynott PR, Oates T, Lim S, Wu R, Carlstedt I, 2002.
Nicholson AG, Chung KF. Expression of MUC5AC and MUC5B 115. Donnelly LE, Rogers DF. Antiproteases and retinoids for treatment
mucins in normal and cystic brosis lung. Respir Med 96: 8186, of chronic obstructive pulmonary disease. Expert Opin Ther Patents 13:
2002. 134572, 2003.
108. Davies JR, Svitacheva N, Lannefors L, Kornfalt R, Carlstedt I. 116. Li Y, Martin LD, Spizz G, Adler KB. MARCKS protein is a key
Identication of MUC5B, MUC5AC and small amounts of MUC2 molecule regulating mucin secretion by human airway epithelial cells
mucins in cystic brosis airway secretions. Biochem J 344(Pt 2): in vitro. J Biol Chem 276: 4098290, 2001.
32130, 1999. 117. Singer M, Martin LD, Vargaftig BB, Park J, Gruber AD, Li Y,
109. Glynn AA, Michaels L. Bronchial biopsy in chronic bronchitis and Adler KB. A MARCKS-related peptide blocks mucus hypersecretion
asthma. Thorax 15: 14253, 1960. in a mouse model of asthma. Nat Med 10: 19396, 2004.
110. Rogers DF. The role of airway secretions in COPD: Pathophysiology, 118. Foster KA. A new wrinkle on pain relief: Re-engineering clostridial
epidemiology and pharmacotherapeutic options. COPD 2: 34153, neurotoxins for analgesics. Drug Discov Today 10: 56369, 2005.
2005. 119. Foster KA, Adams EJ, Durose L, Cruttwell CJ, Marks E, Shone CC,
111. Burgel PR, Nadel JA. Roles of epidermal growth factor receptor acti- Chaddock JA, Cox CL, Heaton C, Sutton JM, Wayne J, Alexander
vation in epithelial cell repair and mucin production in airway epithe- FC, Rogers DF. Re-engineering the target specicity of clostridial
lium. Thorax 59: 99296, 2004. neurotoxins a route to novel therapeutics. Neurotox Res 9: 1017,
112. Hewson CA, Edbrooke MR, Johnston SL. PMA induces the 2006.
MUC5AC respiratory mucin in human bronchial epithelial cells, via 120. Adams EJ, Foster K, Barnes PJ, Rogers DF. Inhibition of airway
PKC, EGF/TGF-alpha, Ras/Raf, MEK, ERK and Sp1-dependent mucin secretion by a retargeted clostridial endopeptidase in a rat
mechanisms. J Mol Biol 344: 68395, 2004. model of COPD. Am J Respir Crit Care Med, 175, (abstracts issue):
113. Toda M, Tulic MK, Levitt RC, Hamid Q. A calcium-activated A995, 2008.
chloride channel (HCLCA1) is strongly related to IL-9 expression

223
Airway Smooth Muscle
18CHAPTER

Yassine Amrani1,
Calcium responses induced by Omar Tliba2, Vera P.
INTRODUCTION Krymskaya2, Michael W.
bronchoconstrictor agents
Sims2 and Reynold A.
Airway smooth muscle (ASM) functions as Using the uorescent dye Fura-2, studies per- Panettieri Jr.2
the primary eector cell that regulates bron- formed on ASM cells have shown that agonist- 1
chomotor tone. In asthma and chronic obstructive induced elevation of the cytosolic free calcium Department of Infection, Immunity
pulmonary disease (COPD), bronchoconstriction ([Ca2]i) concentration is biphasic. The initial and Inflammation, University of
evoked by smooth muscle shortening promotes rapid and transient phase of [Ca2]i elevation Leicester, UK
airway obstruction, a hallmark of asthma and results from the depletion of inositol-1,4,5 tri- 2
Pulmonary, Allergy and Critical
COPD. Recent evidence also suggests that ASM sphosphates (IP3)-sensitive calcium stores; see Care Division, Airways Biology
may undergo hypertrophy and/or hyperplasia and the review by Amrani and Panettieri [1]. This Initiative, University of Pennsylvania,
modulate inammatory responses by secreting transient increase in intracellular calcium initi- Philadelphia, PA, USA
chemokines and cytokines. This chapter reviews ates the contraction process through activation
current studies focusing on excitationcontraction of a calciumcalmodulin-dependent myosin
coupling, signaling pathways modulating ASM light chain kinase and subsequent phosphoryla-
growth, and cytokine-induced eects on ASM tion of the 20 kDa myosin light chain, the cen-
synthetic responses. tral regulatory mechanism of ASM contraction;
see the review by Giembycz and Raeburn [2].
The subsequent sustained elevation of intracel-
lular [Ca2]i is regulated by the activation of
REGULATION OF CALCIUM a calcium inux across the plasma membrane
since depletion of extracellular calcium or the
SIGNALING IN ASM use of calcium channel inhibitors prevents this
sustained phase [3, 4]. The role of the calcium
Because increases in cytosolic calcium directly inux in ASM cells after agonist stimulation is
regulate the initiation and the development of not completely understood, but evidence sug-
ASM contraction, changes in calcium homeos- gests that calcium entry plays an important role
tasis may promote bronchial hyperresponsiveness in maintaining the plateau phase of ASM con-
in asthma. Evidence suggests that various medi- traction via a PKC-dependent mechanism [2].
ators and cytokines important in the pathogen-
esis of asthma, which augment agonist-induced
contractile function, also directly alter calcium Pathways regulating [Ca2]i
signaling. This section reviews (1) the critical influx
calcium-dependent mechanisms involved in
the regulation of ASM contraction, and (2) the Although agonist-mediated increases in [Ca2]i
potential molecular mechanisms that regulate have been extensively characterized, much less is
calcium signaling. known about the cellular mechanisms linking the

225
Asthma and COPD: Basic Mechanisms and Clinical Management

transient and the sustained phase. In 1986, Putney proposed the ROCCs SOCCs
concept of store-dependent calcium entry (also called the capa-
citative model) where depletion of intracellular stores directly Calcium channels
regulates calcium entry at the plasma membrane via store-
operated calcium channels (SOCC). This calcium inux con-
G-protein PLC
tributes to the relling of the internal stores [5]. The existence

of capacitative calcium entry in excitable cells, such as smooth
muscle cells, was demonstrated using drugs, such as thapsi- IP3
gargin, that cause depletion of sarco-endoplasmic reticulum Ca2
Internal
(SER) by directly inhibiting the activity of the SER-associated stores Ca2
calcium-ATPases (SERCA) [6]. Calmodulin
SERCA
In human ASM cells, although thapsigargin-sensitive
MLCK PKC
SERCA2a and 2b isoforms exist, SERCA2b is the predomi-
nant isoform. In these cells, depletion of intracellular cal- Thapsigargin
cium stores in response to thapsigargin activates a calcium MLC-P
inux with a magnitude that is dependent upon the dura-
tion of stimulation with thapsigargin [1]. These data sug- Initial phase Sustained phase
gest that pathways that activate calcium inux in ASM are
linked to the lling state of SERCA2b-associated internal
calcium stores. Release of calcium from SER also regulates
calcium-dependent chloride and nonselective cation chan- Contraction
nels, leading to membrane depolarization and opening of
voltage-dependent channels; see the review by Janssen [7].
Pretreatment of human ASM cells with thapsigargin also
FIG. 18.1 Sources of calcium involved in the regulation of agonist-
abrogates the calcium responses induced by bradykinin, his- induced ASM contraction. Contractile agonists activate G-protein-coupled
tamine, or carbachol, suggesting that thapsigargin-sensitive receptors that stimulate PLC and evoke contraction via the mobilization of
calcium stores involve, at least in part, those activated by the calcium from IP3-sensitive internal stores. The increase in [Ca2]i activates
contractile agonists [3]. calcium-/calmodulin-sensitive myosin light chain kinase (MLCK), with
By mobilizing the same calcium stores, it is plau- subsequent phosphorylation of the 20-kDa myosin light chain (MLC)
sible that both thapsigargin and agonists activate similar and initiation of the cross-bridge cycling between actin and myosin. The
SOCC-dependent calcium inux pathways in ASM. This is sustained phase of contraction is thought to involve calcium entry through
supported by the fact that the amplitude of the sustained both ROCCs and SOCCs. In contrast to ROCCs that remain open as long as
increase in [Ca2]i is dependent on the amplitude of the the contractile agonist is present, SOCCs are triggered by the filling state
of sarco-endoplasmic reticulum calcium ATPases (SERCA)-associated
initial transient phase [3]. Whether additional mechanisms
internal stores.
are involved, such as concomitant activation of receptor-
operated calcium channels (ROCC) previously described in
ASM [3, 4] or involvement of a released soluble mediator
controlling calcium inux [8], remains unknown.
Collectively, these data show that the thapsigargin- demonstrate that tumor necrosis factor (TNF)- and IL-1
sensitive intracellular calcium stores in ASM not only serve enhance bovine smooth muscle contractility to acetylcholine
as a source of calcium to initiate the transient response and other contractile agonists by involving an increased mobi-
to agonists, but also participate in the regulation of cal- lization of intracellular [Ca2]i [10]. Accordingly, proinam-
cium inux in ASM cells as proposed by the capacitative matory cytokines may prime ASM cells for a nonspecic
model. Figure 18.1 summarizes the potential sources of increase in calcium responsiveness, an eect that appears not
calcium mobilized by contractile agonists to regulate ASM to involve a change in the receptor anity for its ligand [11,
contraction. 12]. The fact that TNF- potentiates increases in calcium
induced by thapsigargin, which depletes SER calcium stores,
suggests a possible modulatory eect of TNF- on SER-
Calcium homeostasis in ASM: A possible associated regulatory proteins such as SERCA. In addition,
target for proinflammatory agents TNF- and IL-1 also augment agonist-evoked phosphoi-
nositide turnover, suggesting that cytokines may modulate
receptor-coupled phospholipase C (PLC) activity; see the
Amplification of contractile receptor-coupled review by Amrani et al. [11]. In support of this hypothesis,
calcium signaling TNF- also enhanced calcium signals in response to NaF
The observation that cultured ASM cells derived from [1], an agent that directly activates G-proteins in ASM cells,
hyperresponsive Fisher rats have an enhanced calcium sig- and upregulated expression of Gq and Gi proteins in human
nal to serotonin suggests the existence of altered calcium ASM cells [13].
metabolism in a model of airway hyperresponsiveness [9]. Together, these data suggest that TNF- can induce
The underlying mechanism is unknown, but evidence shows a hyperresponsive phenotype by enhancing signaling
that proinammatory factors may play an important role in pathways downstream from G-protein-coupled receptor
regulating calcium metabolism in ASM. Recent studies (GPCR) activation.

226
Airway Smooth Muscle 18
TABLE 18.1 Potential modulators of calcium metabolism in ASM cells.

Factors Effect Species References

Modulation of receptor- IL-1, TNF- Ca2 transient, IP3 turnover Human [11, 12]
coupled signal transduction 2
Acrolein Ca transient and Rat [14]
frequency oscillations
Major basic protein Ca2 transient and plateau Bovine [15]
Mechanical strain IP3 turnover Bovine [16]
2
PLA2 Ca signals Bovine [17]
Allergen challenge Ca2 signals, IP3 Rat [9]
5-phosphatase
Chronic hypoxia Ca2 frequency oscillations, Rat [18]
receptor affinity for agonist

Modulation of receptor Fenoterol Histamine H1 receptor Bovine [19]


density IL-1 Bradykinin B2 receptor Human [12]
TNF- Muscarinic receptor Human [1,13]

() increase; () decrease; (IP3) inositol 1,4,5 trisphosphate.

Other reports have also described that proinamma- fenoterol may lead to the worsening of asthma by modulat-
tory stimuli modulate GPCR/PLC signaling pathways in ing, at least in part, bronchial hyperresponsiveness; see the
cultured ASM cells (Table 18.1). Pretreatment of bovine review by Beasley et al. [20]. Figure 18.2 summarizes the
ASM cells with either eosinophil-derived polycationic pro- potential mechanisms involved in the modulation of ago-
teins or myelin basic protein increased bradykinin-induced nist-evoked calcium signaling in response to various stimuli.
transients as well as sustained elevations of [Ca2]i [15].
Incubation with either the aldehyde pollutant acrolein or
the proinammatory enzyme PLA2 signicantly increased Increase in the calcium sensitivity of the
the intensity as well as the frequency of calcium transients contractile apparatus
in response to acetylcholine [14, 17]. Finally, other experi- Increase in the sensitivity of myolaments to calcium also
mental conditions, such as chronic hypoxia or mechanical represents a mechanism by which the contractile function
strain, also modulate calcium signaling induced by agonists of ASM can be enhanced. Abnormal calcium sensitivity has
in ASM cells. Belouchi et al. [18] reported that the ampli- been described in ASM derived from allergen-sensitized
tude of calcium transients in response to low concentrations animals or from passively sensitized tissues; see the review
of acetylcholine was signicantly higher in freshly iso- by Schmidt and Rabe [21]. The role played by proinam-
lated tracheal smooth muscle cells from hypoxic rats than matory agents in the impairment of calcium sensitivity has
in those obtained from normoxic animals. Investigators been suggested in vitro. Nakatani et al. [22], by simultane-
also showed that mechanical strain augmented carbachol- ously measuring [Ca2]i and isometric tensions in response
induced IP3 turnover through the regulation of G-protein to acetylcholine, showed that brief exposure to TNF-
and/or PLC activities [16]. enhanced the calcium sensitivity of contractile elements in
bovine tracheal smooth muscle. Surprisingly, TNF- did
Alteration in the density of contractile receptors not aect the calcium signals induced by acetylcholine. In
Exposure of cultured ASM to various proinammatory guinea pig tracheal smooth muscle, short-term treatment
mediators also modulates the density of contractile agonist with TNF- also increased calcium sensitivity of the con-
receptors. ASM cells exposed to TNF- have a dramatic tractile apparatus [23].
decrease in muscarinic receptor density [3, 13]. In contrast, Together, these studies suggest that, in addition to
levels of the bradykinin B2 receptor are rapidly increased the modulation of receptor-coupled signal transduction,
in human bronchial smooth muscle exposed to IL-1 via a increase in calcium sensitization of contractile elements rep-
prostanoid-dependent regulation of gene transcription [12]. resents another downstream target potentially modulated by
Surprisingly, fenoterol, a 2-agonist, as well as other cAMP- proinammatory agents.
elevating agents, signicantly upregulates the expression Because ASM is an essential eector cell modulat-
of histamine H1 receptor in bovine ASM cells, an eect ing bronchoconstriction, changes in ASM properties can
that involves both increased gene expression and mRNA be regarded as a potential mechanism contributing to the
stability [19]. This increase in H1 receptor expression was increased ASM contractility associated with asthma. A
associated with an increase in ASM responsiveness to his- variety of stimuli present in asthmatic airways may induce
tamine in contraction studies. This may be important since hyperresponsiveness by modulating calcium signaling in

227
Asthma and COPD: Basic Mechanisms and Clinical Management

? Allergen challenge

2-agonist
Inflammatory mediators

Pollutant
Chronic hypoxia
Abnormal mechanical strain
Receptor



G-protein PLC

IP3

Internal stores Ca2


SERCA2b

Amplification of
Ca2 signals

FIG. 18.2 Potential modulators of contractile receptor-coupled calcium signaling in ASM. A variety of stimuli may induce a hyperresponsive phenotype,
which modulates G-protein-coupled receptor activation in response to agonists. (?) indicates the possible role played by inflammatory mediators in allergen-
induced changes in calcium responsiveness to contractile agonist. The potential intracellular targets underlying the increase in calcium responsiveness
induced by these various stimuli are shown with a bold star. IP3: inositol-1,3,5 trisphosphate; PLC: phospholipase C; [Ca2]i: intracellular calcium.

Normal Asthma/COPD

Acute Chronic

Epithelium

Features
Basement
membrane T-cells
Mast cells
Mediators Cellcell interaction via
CD40, ICAM-1, CD44

Enhanced signaling
PI3K, NF-IB pathways
Smooth muscle

Increased Ca2 signals


Increased Ca2 signals Increased cell number
Increased cytokine/chemokine release

Modulation of ASM function

FIG. 18.3 Factors affecting ASM function in acute and chronic state of asthma/COPD. During the acute inflammation, a variety of mediators, such as
cytokines, can modulate ASM contractile function by enhancing calcium signaling to agonists. These mediators regulate the recruitment and activation
of eosinophils and T-lymphocytes in the airway mucosa, a characteristic histopathological feature of the chronic disease. The persistence of airway
inflammation via the production of cytokines and chemokines by both inflammatory as well as structural cells has the potential to directly stimulate ASM
proliferation or indirectly as a result of T-cell-ASM interaction mediated by cell surface expression of various CAM proteins such as ICAM-1, CD40, and CD44.
Enhanced activation of PI3K or the transcription factor NF-B in ASM may also stimulate mitogenic and synthetic functions.

228
Airway Smooth Muscle 18
response to contractile agonists as summarized in Fig. 18.3. These PLCs are the critical regulatory enzymes in the acti-
Whether this amplication of calcium signaling contributes vation of the PI pathway. The family of PLC contains
to the increased contractility in asthmatic patients remains src-homology SH2 and SH3 domains and is regulated by
to be investigated. tyrosine phosphorylation. In ASM cells, some growth factors
which activate receptors with intrinsic tyrosine kinases have
been identied. Platelet-derived growth factor (PDGF) and
epidermal growth factor (EGF) in human ASM cells [2931]
ASM CELL PROLIFERATION and IGF-1 in bovine and rabbit ASM cells [3234] have
been shown to induce myocyte proliferation. However, the
role of PLC-1 activation in modulating ASM cell growth
Increased ASM mass is a characteristic histopathological nd- remains unknown.
ing in the airways of chronic severe asthmatics and of COPD Other PLC isoforms are controlled by G-proteins
patients. Although increased ASM mass is, in part, due to and/or calcium. Although the role of PLC activation in
ASM cell proliferation, the mechanisms that regulate ASM mediating G-protein-dependent cell growth is complex, G-
cell growth remain unclear. Many studies have characterized protein activation appears critically important in transduc-
the stimulation of ASM growth in response to mitogenic ing contractile agonist-induced cell growth. G-proteins are
agents such as polypeptide growth factors, inammatory composed of three distinct subunits, , , and , the latter
mediators, and cytokines. The observation that contractile ago- two existing as a tightly associated complex [35]. Although
nists induce smooth muscle cell proliferation may be a critical -subunits were considered the functional components
link between the chronic stimulation of muscle contraction important in downstream signaling events, recent evidence
and the myocyte proliferation [24]. Although the specic cel- suggests that -subunits also play a critical role in modu-
lular and the molecular mechanisms by which agonists induce lating cell function [36].
cell proliferation remain to be elucidated, similarities exist Advances in single-cell microinjection techniques in
between signal transduction processes activated by these agents combination with the development of neutralizing antibod-
and those of known growth factors, which can also stimulate ies to specic G subunits have enabled investigators to
smooth muscle contraction. The complex interaction between characterize the role of G-protein activation in cell prolifer-
signaling pathways that induce myocyte proliferation and ation. Using these techniques, studies with 3T3 broblasts
those that inhibit cell growth by stimulation of apoptosis may have determined that, while both thrombin and bradyki-
promote airway remodeling as seen in the bronchi of patients nin required Gq activation to mobilize cytosolic calcium, to
with asthma, bronchiolitis obliterans, or chronic bronchitis. generate IP3 and to induce mitogenesis, thrombin, but not
Smooth muscle cell proliferation is stimulated by bradykinin, induces cell growth by stimulating Gi2 [37].
mitogens that fall into two broad categories: These studies determined that a single mitogen may require
1. those that activate receptors with intrinsic tyrosine kinase functional coupling to distinct subtypes of G-proteins in
activity (RTK); order to stimulate cell growth. Collectively, these data also
provide a mechanism to explain why some, but not all, ago-
2. those that mediate their eects through receptors nists induce cell proliferation while mobilizing comparable
coupled to heterotrimeric GTP binding proteins levels of cytosolic calcium.
(G-proteins) and activate nonreceptor-linked tyrosine Recently, the role of PLC activation and IP3 in medi-
kinases found in the cytoplasm. ating contractile agonist-induced ASM cell growth has
Although both pathways increase cytosolic calcium been explored. Several contractile agonists, which mediate
through activation of PLC, dierent PLC isoenzymes appear their eects through GPCRs, induce ASM cell prolifera-
to be involved. Activated PLC hydrolyzes phosphatidylinosi- tion. Studies have determined that histamine [38] and sero-
tol bisphosphate (PIP2) to inositol trisphosphate (IP3) and tonin induce canine and porcine ASM cell proliferation.
diacylglycerol (DAG). These second messengers activate other Endothelin-1, leukotriene D4 and U-46619, a thrombox-
cytosolic tyrosine kinases as well as serine and threonine ane A2 mimetic, induce rabbit ASM cell growth [39], and
kinases (protein kinase C, G, and N) that have pleiotropic thrombin induces mitogenesis in human ASM cells [30].
eects including the activation of proto-oncogenes, which Although the mechanisms that mediate these eects are
are a family of cellular genes (c-onc), that control normal cel- unknown, agonist-induced cell growth probably is modu-
lular growth and dierentiation. Recent reviews attempted to lated by activation of G-proteins in a manner similar to that
summarize current progress in our understanding of cellular described in vascular smooth muscle (VSM).
mechanisms leading to smooth muscle cell proliferation [25 Using human ASM cells, Panettieri et al. [30] exam-
28]. This section will focus on the role of PLC activation and ined whether contractile agonist-induced human ASM cell
the phosphatidylinositol 3-kinase (PI3K) signaling pathway growth was dependent on PLC activation and IP3 forma-
in smooth muscle cell mitogenesis. tion. These investigators examined the relative eects of
bradykinin and thrombin on myocyte proliferation and PI
turnover. Thrombin, but not bradykinin, stimulated ASM
PLC activation cell proliferation despite a vefold greater increase in [3H]-
inositol phosphate formation in cells treated with bradykinin
Receptors with intrinsic tyrosine kinase activity and those as compared with those treated with thrombin. Inhibition of
coupled to G-proteins both activate specic PLC isoforms. PLC activation with U-73122 had no eect on thrombin- or

229
Asthma and COPD: Basic Mechanisms and Clinical Management

EGF-induced myocyte proliferation. In addition, pertus- role in PI3K-mediated signaling of immune responses [53].
sis toxin completely inhibited thrombin-induced ASM cell Class IA isoforms are mainly activated by receptor and non-
growth but had no eect on PI turnover induced by either receptor tyrosine kinases while Class IB p110 is activated
thrombin or bradykinin [30]. Taken together, these studies by G subunits of GPCRs [54]. Class II isoforms are
suggest that thrombin induced human ASM cell growth mainly associated with the phospholipid membranes and are
by activation of a pathway that was pertussis toxin-sensitive present in the endoplasmic reticulum and Golgi apparatus
and independent of PLC activation or PI turnover. [55]. Class III isoforms, structurally related to yeast vesicu-
Compared to RTK-dependent growth factors, con- lar sorting protein Vps34p [56], are mammalian homologs
tractile agonists, with the exception of thrombin and that use only membrane phosphatidylinositol as a substrate
sphingosine-1-phosphate, appear to be less eective human and generate phosphatidylinositol-3-monophosphate.
ASM mitogens [40]. In cultured human ASM cells, PI3K activation by multiple inputs, such as growth
100 mol/l histamine or serotonin induces two- to three- factors, insulin, cytokines, cellcell, and cellmatrix adhe-
fold increases in [3H]-thymidine incorporation as compared sion, provided by both receptor and nonreceptor tyrosine
with that obtained from unstimulated cells. EGF, serum, kinases leads to activation of serinethreonine kinase Akt
or phorbol esters, which directly activate protein kinase (Fig. 18.4). Downstream of PI3K the tumor suppressor
C, induce 2030 fold increases in [3H]-thymidine incor- complex tuberous sclerosis complex (TSC)1/TSC2 is sub-
poration [41]. In rabbit ASM cells, endothelin-1 induces ject to direct inhibitory phosphorylation by Akt [5759].
cell proliferation by activating phospholipase A2, and by TSC2 forms a complex with tumor suppressor TSC1 and
generating thromboxane A2 and LTD4 [39, 42]. In human regulates mTOR/S6K1 signaling [60] by directly control-
ASM cells, however, endothelin-1, thromboxane A2 and ling the activity of the small GTPase Rheb via the GTPase
LTD4 appear to have little eect on ASM cell proliferation activating protein (GAP) domain of TSC2 [61]. Rheb
despite these agonists inducing increases in cytosolic cal- directly binds to Raptor [62, 63] and controls the activ-
cium [30, 43, 44]. ity of the mTOR/Raptor complex (mTORC1), which, in
Clearly, interspecies variability exists with regard to turn, directly phosphorylates and activates S6K1 (Fig. 18.4).
contractile agonist-induced cell proliferation. These models,
however, may prove useful in dissecting downstream signal-
ing events that modulate the dierential eects of contrac-
tile agonists on ASM cell proliferation.
Growth factors

PI3K signaling pathway


RTK

TSC1
PI3K PDK Akt TSC2
The PI3K signaling pathway is a highly conserved signal
transduction network regulating a variety of cellular func-
Rheb
tions including cell proliferation, dierentiation, trans-
formation, cell motility, and apoptosis [45, 46]. PI3Ks are
? mTOR
a subfamily of lipid kinases that catalyze the addition of mTOR Raptor GL
phosphate molecules specically to the 3-position of the Rictor GL
inositol ring of phosphoinositides [47]. Tumor suppres-
4E-BP1 S6K1
sor PTEN (phosphatases and tensin homolog) negatively
regulates PI3K signaling by specically dephosphorylating eIF-4E
the 3
-OH position of the inositol ring of the lipid second
messengers phosphatidylinositol-3,4,5-trisphosphate. [48 S6 S6 S6
Protein synthesis
50], PI3K lipid products are not substrates for the PI-spe-
cic PLC enzymes that cleave inositol phospholipids into
membrane-bound DAG and soluble inositol phosphates. Cell growth and proliferation
The 3-phosphoinositides function as second messengers FIG. 18.4 The PI3K signaling pathway is critical for regulating cell growth
and activate downstream eector molecules such as PDK1, and proliferation. Growth-factor (GF)-induced activation of PI3K initiates
p70s6 kinase (S6K1), protein kinase C, and Akt [47, 51]. a signaling cascade involving sequential activation of phosphoinositide-
The ability of PI3K to regulate diverse functions may be dependent kinase (PDK) and Akt/PKB. Akt/PKB-dependent phosphorylation
due to the existence of multiple isoforms that have specic of TSC2 results in dissociation of TSC1/TSC2 membrane complex and
substrate specicities and that reside in unique cytoplasmic activation of Rheb. Next, Rheb activates mTOR complex 1 (mTORC1) which
locations within the cell [52]. consists of mTOR, Raptor, and GL. This is followed by activation of S6K1
PI3K isoforms can be divided into three classes with subsequent phosphorylation of ribosomal protein S6. In parallel,
based on their structure and substrate specicity. Class IA mTOR-dependent phosphorylation of the binding protein 4E releases
from the inhibitory complex eukaryotic initiation factor 4E (eIF-4E), which
PI3Ks are cytoplasmic heterodimers composed of a 110-
initiates protein synthesis, resulting in cell growth and proliferation.
kDa (p110, , or ) catalytic subunit and an 85-kDa (p85, Rapamycin-insensitive complex mTORC2, which consists of mTOR,
p55, or p50) adaptor protein. Catalytic subunits p110 Rictor, and GL, regulates Akt/PKB phosphorylation; however, the precise
and p110 are ubiquitously expressed in mammalian cells. mechanism of mTORC2 regulation by Rheb and TSC1/TSC2 remains to be
Catalytic subunit p110 is expressed predominantly in lym- elucidated. 4E-BP1: eIF-4E binding protein; Rheb: Ras homolog enriched in
phocytes and lymphoid tissues and, therefore, may play a brain; RTK: receptor tyrosine kinase.

230
Airway Smooth Muscle 18
mTOR is a part of both complexes: the rapamycin-sensitive A recent study demonstrated that GPCR activation
mTOR/Raptor (TORC1) phosphorylating S6K1, and the by inammatory and contractile agents can synergize with
rapamycin-insensitive mTOR/Rictor (TORC2) phospho- RTK activation to augment human ASM growth. In EGF-
rylating Akt [64] and regulating Rac1 activation [65]. The stimulated cells, GPCR-mediated potentiation does not
PI3K-TSC1/TSC2-Rheb-mTOR-S6K1 signaling pathway appear mechanistically linked to increased EGFR (epider-
is highly conserved across species, and controls cell growth mal growth factor receptor) or p42/p44 MAPK activation
and proliferation [45, 46, 61]. but is associated with sustained activation of S6K1 for sev-
Studies demonstrate that PI3K signaling cascade eral hours after the initial early phase of activation. These
plays a critical role in regulating human airway and pulmo- ndings not only provide insight into mechanisms by which
nary arterial VSM cell proliferation [66, 67]. Wortmannin inammation contributes to ASM hyperplasia/hypertrophy
and LY294002, two potent inhibitors of PI3K, inhibit in diseases such as asthma and COPD, but also suggest a
DNA synthesis in bovine ASM, porcine, and rat VSM cells general mechanism by which GPCRs and RTKs interact to
stimulated with PDGF, basic broblast growth factor, angi- promote cell growth [78].
otensin II, or serum [6871]. Stimulation of 1 adrenergic PDGF, EGF, and thrombin, which transduce their
receptors with noradrenaline activated mitogenesis, Ras, signals by activating distinct pathways through RTKs and
MAPK, and PI3K in human VSM cells in a wortman- GPCR, induce PI3K activity in human ASM cells [67, 74,
nin-sensitive manner [72]. In rat thoracic aorta VSM cells, 79, 80]. Importantly, cross talk between GPCR and RTK
wortmannin completely blocked angiotensin II-induced signaling pathways occurs at the PI3K levels and promotes a
Ras activation but had no eect on mitogen-activated pro- synergy among growth factors and contractile agonists [78,
tein kinase (MAPK) activation or protein synthesis [73]. 79, 81]. Src protein tyrosine kinase integrates GPCR- and
Thrombin, which induces human ASM cell growth by RTK-induced signaling pathways that ultimately regulate
activating a receptor presumably coupled to both Gi and cell proliferation, migration, dierentiation, and gene tran-
Gq proteins [30], requires PI3K activation to mediate its scription [82, 83]. Src kinases have a well-established role in
growth eects [74]. In bovine ASM, the mitogenic eects the regulation of cell proliferation. The Src tyrosine kinase
of PDGF or endothelin-1 (ET-1) have been attributed to is the rst tyrosine kinase to be identied and linked to
their ability to stimulate PI3K or S6K1. These data sug- the regulation of cell proliferation [82, 83]. Because more
gest that in both airway and VSM cells PI3K is involved in than one mechanism is involved in Src activation, this
mitogenic signaling induced by numerous agents. enzyme is critical in transducing signals, which originate
In order to determine whether PI3K activation is through GPCRs and RTKs and result in integrated cellular
necessary or sucient to stimulate human ASM DNA syn- responses, such as DNA synthesis and migration. The cel-
thesis, Krymskaya et al. [67] used transfected cells with a lular functions regulated by Src were identied by using pre-
chimeric model of Class IA PI3K in which the inter-SH2 dominantly overexpressed Src and its mutants in established
region of p85 regulatory subunit was covalently linked to its immortalized cell lines and in vitro assays. Using selective
binding site at the p110 N-terminal region of the catalytic cell-permeable Src inhibitors PP1 and PP2, Src inhibition
subunit [75]. Transient expression of constitutively active of DNA synthesis was demonstrated in guinea pig ASM
p110* was sucient to induce DNA synthesis in ASM cells [84] and rat aortic smooth muscle cells [85]. In our study
in the absence of mitogens [67]. Interestingly, in human using pharmacological and molecular approaches, we dem-
ASM cells the level of p110*-induced DNA synthesis was onstrate that Src is necessary and sucient for human ASM
markedly lower than that induced by EGF, thrombin, or DNA synthesis. Interestingly, PP2 had dierential eects
serum; and thus, although PI3K activation is sucient to on EGF- versus PDGF- and thrombin-induced DNA
induce DNA synthesis in ASM cells, other signaling path- synthesis: IC50 for EGF was 5.5 M, which is a magni-
ways that act in parallel or that are more eective induc- tude higher compared to the IC50 for PDGF and thrombin,
ers of PI3K may play a role in modulating mitogen-induced which was 0.3 M for both agonists. The high sensitiv-
DNA synthesis in these cells. ity of thrombin-induced DNA synthesis to Src inhibition
In numerous cell types, PI3K has been shown to be is potentially due to a critical role of Src in modulating
an important mediator of S6K1 activation in response to mitogenic signaling from activated GPCRs to its down-
serum and growth factors [76]. S6K1 is a critical enzyme stream targets [86, 87]. We also found that the dierential
for mitogen-induced cell cycle progression through the G1 Src regulation of cell proliferation correlated with dieren-
phase and translational control of mRNA transcripts that tial activation of Src-associated PI3K. Thus, upon stimula-
contain a polypyrimidine tract at their transcriptional start tion of human ASM cells with PDGF or thrombin, PI3K
site [77]. activity was associated with Src but not in cells stimulated
EGF and thrombin signicantly stimulate S6K1, and with EGF. Since PI3K activation is critical for cell prolif-
wortmannin, LY294002, and rapamycin completely block eration, the lack of Src-associated PI3K activation may
this activation in ASM cells [74]. Moreover, transient expres- explain the insensitivity of EGF-induced ASM DNA syn-
sion of constitutively active p110* PI3K activates S6K1 in thesis to blocking Src. Importantly, our previously published
the absence of stimulation with mitogens, while overexpres- study identied the signaling pathway of EGF-dependent
sion of a dominant-negative p85 PI3K abolished EGF- PI3K activation in HASM cells, which involved activation
and thrombin-induced S6K1 activation [74]. Thus, EGF of the ErbB2 receptor of the EGF receptor family [88].
and thrombin induce activation of S6K1 in human ASM Collectively, these data suggest a dierential role of Src in
cells and mitogen-induced activation of S6K1 appears PI3K growth promoting signaling stimulated by EGF, PDGF,
dependent. and thrombin.

231
Asthma and COPD: Basic Mechanisms and Clinical Management

eotaxin-1/CC chemokine ligand 11 [103]. Extracellular


ASM: AN IMMUNOMODULATORY CELL matrix (ECM) proteins seem to play an important role in
the regulation of eotaxin induced by dierent interleukins,
Asthma is a disease characterized, in part, by reversible air- IL-13, IL-1, or TNF- in ASM cells cultured from sub-
ow obstruction, hyperresponsiveness, and inammation. jects with asthma compared with healthy donors [104].
COPD, which includes chronic bronchitis, emphysema, and Interestingly, increased expression of eotaxin is observed
bronchiectasis, is characterized by predominantly irrevers- during the co-culture of ASM with mast cells [105], sug-
ible airow obstruction associated with neutrophilic airway gesting that cellcell interaction is important to consider in
inammation. Traditional concepts concerning airway inam- the regulation of causes of airway inammation.
mation have focused on tracking leukocytes and on the In asthma and COPD, -adrenergic agonists, by
eects of inammatory mediators, cytokines, and chemokines elevating [cAMP]i, promote bronchodilation. Studies have
secreted by these cells. ASM, the major eector cell responsi- investigated whether cAMP pathways also modulate chem-
ble for bronchomotor tone, has been viewed as a passive tissue okine secretion by ASM cells. In TNF--stimulated ASM
responding to neurohumoral control and inammatory medi- cells, eotaxin and RANTES expression was potently and
ators. New evidence, however, suggests that ASM may secrete eectively inhibited by isoproterenol, PGE2, dibutyl-cAMP,
cytokines and chemokines and express cell adhesion mol- or phosphodiesterase inhibitors, rolipram and cilomilast [91,
ecules (CAMs) that are important in modulating submucosal 106]. TNF--induced IL-8 secretion was inhibited by the
airway inammation. The cellular and molecular mechanisms combination of cAMP-mobilizing agents and corticoster-
that regulate the immunomodulatory functions of ASM may oids [107]. Similarly, sphingosine-1-phosphate, which acti-
oer new and important therapeutic targets in treating these vates a Gs-protein-coupled receptor and increases [cAMP]i,
common lung diseases. abrogated TNF--induced RANTES secretion in ASM
cells [108].
Current evidence suggests that chemokine secretion
Chemokine and cytokine release induced by inammatory mediators is inhibited by dexam-
by ASM cells ethasone in human ASM cells. Cytokine-induced secretion
of RANTES [9193], MCP [93], eotaxin [106], and GM-
The various cell types that inltrate the inamed submu- CSF [109] was abrogated with corticosteroids. In most of
cosa present the potential for many important cellcell these studies, corticosteroid and cAMP-mobilizing agents
interactions. Eosinophils, macrophages, neutrophils, and also acted additively to inhibit chemokine secretion.
lymphocytes initiate and perpetuate airway inammation IL-6, a pleiotropic cytokine, induces smooth muscle
by producing proinammatory mediators. Evidence also cell hyperplasia [110], but also modulates B- and T-cell pro-
suggests that exposure of ASM to cytokines or growth fac- liferation and immunoglobulin secretion. IL-6 secretion by
tors alters contractility and calcium homeostasis [1] and ASM cells is inducible by multiple stimuli, including IL-1,
induces SMC hypertrophy and hyperplasia; see the review TNF-, TGF-, and sphingosine-1-phosphate, a recently
by Lazaar et al. [89]. described mediator in asthma [91, 108, 111, 112]. Although
Studies now show that ASM cells secrete a number of corticosteroids are potent inhibitors of IL-6 secretion [112],
cytokines and chemoattractants. Studies of bronchial biopsies agents that elevate [cAMP]i actually increase IL-6 secretion
in mild asthmatics reveal constitutive staining for RANTES, [91]. This suggests an intriguing role for IL-6 in regulat-
a CC chemokine, in ASM [90]; in vitro, RANTES secretion ing airway hyperreactivity and is in agreement with murine
is induced by TNF- and IFN- [9193]. Other chemok- models in which IL-6 overexpression promotes airway
ines that are secreted by ASM cells include eotaxin, an eosi- hyporesponsiveness [113].
nophil chemoattractant, and monocyte chemotactic proteins Finally, additional cytokines that are secreted by human
(MCP)-1, MCP-2, MCP-3 [9395]. ASM cells include IL-1 and other IL-6 family cytokines,
Similarly, the CXC chemokine IL-8 is also secreted such as leukemia inhibitory factor and IL-11 [111, 114,
by ASM in response to TNF-, IL-1, and bradykinin, a 115]. ASM cells may also play a role in promoting both the
contractile agonist [96, 97]. IL-8 has been described to recruitment and the survival of eosinophils by secretion of
regulate both proliferation and migratory function of ASM GM-CSF and IL-5 [109, 114, 116].
cells [98]. Understanding the factors that stimulate IL-8
production may be of therapeutic benet for the treatment
of asthma. Studies found that IL-8 is induced by several Receptors involved in cell adhesion
factors including tryptase, the most abundant mast cell
product [99] as well as IL-17/IL-1 via both transcrip- CAMs mediate leukocyteendothelial cell interactions
tion factors, AP-1 and NF-B [100]. Others found that the during the process of cell recruitment and homing [117]
antimicrobial peptide LL-37 also induces the production (Chapter 21). The expression and activation of a cascade of
of IL-8 [101] providing a novel mechanism through which CAMs that include selectins, integrins, and members of the
inammatory cell products could participate in the patho- immunoglobulin superfamily, as well as the local produc-
genesis of asthma. Additional studies showed that IL-4 also tion of chemoattractants, leads to leukocyte adhesion and
stimulates eotaxin/CCL-11 expression in ASM cells, in transmigration into lymph nodes and sites of inammation
part via the activation of STAT6 transcription factor [102] involving nonlymphoid tissues.
while Rahman et al. found that (p38, p42/p44 ERK, JNK) In addition to mediating leukocyte extravasation and
signaling pathways play a critical role in IL-17A-induced transendothelial migration, CAMs promote submucosal or

232
Airway Smooth Muscle 18
subendothelial contact with cellular and ECM components. dexamethasone, whereas IL-1-induced cyclooxygenase-2
New evidence suggests that CAMs mediate inammatory expression was abrogated [119121]. In contrast, cytokine-
cellstromal cell interactions that may contribute to air- induced CAM expression is sensitive to cAMP-mobilizing
way inammation. ASM cells express intercellular adhesion agents [122].
molecule (ICAM)-1 and vascular cell adhesion molecule
(VCAM)-1, which are inducible by a wide range of inam-
matory mediators. Contractile agonists, such as bradykinin Receptors involved in leukocyte
and histamine, in contrast, have little eect on ASM CAM activation and immune modulation
expression [118]. ASM cells also constitutively express CD44,
the primary receptor of the matrix protein hyaluronan [118]. CAMs can function as accessory molecules for leukocyte
Activated T-lymphocytes adhere via LFA-1 and VLA-4 activation [117, 123, 124]. Whether CAMs expressed on
to cytokine-induced ICAM-1 and VCAM-1 on cultured smooth muscle serve this function remains controversial.
human ASM cells. Moreover, an integrin-independent com- ASM cells do express major histocompatibility complex
ponent of lymphocytesmooth muscle cell adhesion appeared (MHC) class II and CD40 following stimulation with
to be mediated by CD44hyaluronan interactions [118]. IFN- [125, 126]. Recent studies also suggest that human
Current hypotheses suggest that steroids may directly ASM cells express low levels of CD80 (B7.1) and CD86
inhibit gene expression, such as CAMs, by altering gene pro- (B7.2) [127]. The physiological relevance of these ndings
moter activity and/or by abrogating critical signaling events remains unknown since ASM cells cannot present alloanti-
such as NF-B or AP-1 activation that then modulate gene gen to CD4 T-cells, despite the expression of MHC class II
expression as shown in Fig. 18.5. Interestingly, dexametha- and costimulatory molecules [125].
sone had no eect on TNF-- or IL-1-induced NF-B Functionally, however, adhesion of stimulated CD4 T-
activation in human ASM cells [119]. Further, cytokine- cells can induce smooth muscle cell DNA synthesis [118].
induced ICAM-1 expression in ASM cells, which is com- This appears to require direct cellcell contact and cannot be
pletely dependent on NF-B activation, was not aected by mimicked by treatment of the cells with T-cell conditioned

Albuterol,
PGE2
Salmeterol Steroid
Formeterol TNF-, IL-1

Gs

Adenylyl cyclase p38, ERK1/2, JNK


NF-B
Cilomast cAMP
PDE ?
rolipram ?
AP-1
5
-AMP A-kinase GR

Nucleus
CRE Gene GRE

Steroid-insensitive genes Steroid-sensitive genes


ICAM-1 IL-6, IL-8
RANTES, eotaxin, GM-CSF
COX2

FIG. 18.5 Putative signaling pathways that regulate cytokine-induced synthetic responses in ASM cells. [cAMP]i-mobilizing agents inhibit cytokine-induced
chemokine and CAM expression. IL-6 secretion, however, is augmented by agents that increase [cAMP]i. Cytokine-induced NF-B activation in human
ASM is corticosteroid-insensitive. A-kinase: cAMP-dependent protein kinase A; CRE: cAMP response element; ERK: extracellular signal-regulated kinase; GR:
glucocorticoid receptor; GRE: glucocorticoid response element; Gs: guanine-nucleotide binding protein; JNK: jun kinase; NF-B: nuclear factor B; PDE:
phosphodiesterase.

233
Asthma and COPD: Basic Mechanisms and Clinical Management

medium. In addition, ligation of CD40 on ASM increases ICAM-1, IL-6, IL-8, GM-CSF, RANTES, MCP-1,
intracellular calcium as well as IL-6 secretion, while cross- GRO-, and NAP-2 [141].
linking VCAM-1 cells activates PI3K and augments growth Whether ASM is a therapeutic target for steroids in
factor-induced ASM cell proliferation [126, 128]. These asthma is yet unclear. Several in vitro studies in cultured
studies highlight the fact that direct interactions between ASM cells conrmed that glucocorticoids (GCs) are eec-
leukocytes and smooth muscle cells via immune receptors, tive in suppressing induction of inammatory genes includ-
such as CD40, or adhesion receptors, such as ICAM-1 or ing IL-6, eotaxin, RANTES, GM-CSF, ICAM-1, and
VCAM-1, contribute to the modulation of the local milieu CD38 8 to name just a few [106, 119, 142145]. Further,
resulting in smooth muscle cell activation. in situ hybridization and immunohistochemistry studies
Increased amounts of nitric oxide (NO) have been revealed the in vivo expression of glucocorticoid receptor
detected in patients with asthma; see the review by Gaston (GR) in ASM of both normal and asthmatic subjects [146].
et al. [129] and Chapter 32. NO appears to have a selective- We previously showed that cultured ASM cells express both
suppressive eect on the Th1 subset of T-helper cells, sug- GR isoforms, GR-, and under certain inammatory con-
gesting that increased levels of NO may therefore lead to ditions, GR- [143]. Interestingly, Roth et al. [147] showed
the predominantly Th2-type response associated with GC treatment induced GR nuclear translocation from
asthma. NO synthase has been demonstrated in ASM cells cytosol to the nucleus in bronchial smooth muscle cell lines
where it results in an inhibition of ASM cell proliferation from healthy individuals or subjects with asthma and those
[130, 131]. Thus the role of ASM-derived NO needs to be with emphysema. Similar results were obtained in cultured
further dened, as it may have both benecial and deleteri- ASM obtained from Roth et al. in 2002 [148, 149]. The
ous eects in the airway. activation of GR by GC through its binding to a gluco-
ASM cells produce large amounts of PGE2, and to a corticoid response element (GRE) DNA sequence has also
lesser extent other prostanoids, following stimulation with been demonstrated in cultured ASM cells [143, 147, 149].
proinammatory cytokines [120]. Although PGE2 is a Interestingly, although steroid eects on human ASM
potent bronchodilator, it also has signicant immunological cells have been investigated [150], their modulatory eects
eects. For example, PGE2 can decrease expression of CD23 on gene expression are complex and poorly characterized.
(FcRII), which has been shown to be expressed on human First, the eects of steroids are gene specic. For example,
ASM cells [132], and synergize with IL-4 to induce IgE reports showed that dexamethasone eectively inhibits
synthesis [133]. Release of PGE2 by ASM cells may also be cytokine-induced IL-6, RANTES or eotaxin expression,
important in modulating the inammatory milieu by inhib- while the same steroid has little eect on cytokine-induced
iting smooth muscle cell secretion of chemokines [134]. ICAM-1 expression [106, 119, 142]. Second, steroid-
suppressive eects are time dependent since dexamethasone
partially abrogates cytokine-mediated ICAM-1 expression
Therapeutic modulation of inflammatory at early time points, but has no eect at later time points
gene expression [119]. Third, steroid inhibitory action is also stimulus spe-
cic. While dexamethasone signicantly inhibits IL-1-
Issa and colleagues uncovered a novel mechanism by which induced GM-GSF secretion, it has only a partial eect on
steroids suppress cytokine-induced inammatory genes in GM-GSF secretion induced by thrombin [144].
human ASM cells [135]. MPK-1 was found to be essen- Recently, Miller et al. [151], using an animal model
tial in mediating the inhibitory action of dexamethasone of asthma, showed that GC treatment signicantly reduced
on growth-related oncogene protein (GRO)- induced by allergen-induced increases in peribronchial collagen depo-
IL-1 and TNF-, thus conrming the role of MAPK in sition and levels of total lung collagen but failed to reduce
inammatory gene regulation. Activation of MAPK path- allergen-induced increases in the thickness of the peribron-
ways by IL-1 and TNF- seem to be important in the reg- chial smooth muscle. In line with this, Roth et al. [147]
ulation of thymic stromal lymphopoietin, a novel factor that showed that GC failed to inhibit the proliferative response
promotes dendritic cell-mediated T-helper (Th)-2 inamma- in bronchial ASM from patients with asthma. Further in
tory responses [136]. Other steroid-sensitive genes include vitro studies showed that the anti-proliferative and anti-
TLR2, TLR3, and TLR4 reported to be induced by TNF- migratory eects of the GC were impaired by contact of
in ASM cells [137]. CD38 was among the steroid-sensitive ASM with collagen. GC also failed to inhibit ASM cells
genes as dexamethasone completely abrogated its expres- cultured on collagen, but not on laminin [148], suggesting
sion and increased enzymatic activity following treatment that in the collagen-rich microenvironment of the inamed
with TNF- [138]. The observation that IFN can act as an and brotic asthmatic airway, ASM may contribute to ster-
eective inhibitor on some, but not all, TNF--inducible oid resistance. In addition, we also recently showed that
inammatory genes [139] suggests that a better understand- steroid-suppressive eects are dramatically impaired in
ing of the IFN-dependent pathways could provide new ASM cells treated with multiple cytokines [143]. Indeed,
insight into the design of more potent anti-inammatory the specic combination of TNF- with IFNs, but not with
drugs. Macrolides, such as erythromycin and azithromycin, IL-1 or IL-13, impairs the actions of multiple classes of
have been recently shown to suppress IL-17-induced pro- GCs, uticasone, dexamethasone, and budesonide [143] by
duction of IL-8 and 8-isoprostane in ASM cells while a mechanism involving the upregulation of GR- isoform.
immunosuppressive agents had no eect [140]. Targeting Recently, Tliba and colleagues [152] found for the rst time
IKK was recently found to be an alternative option for the that an increased level of IRF-1 by pro-asthmatic cytokines
suppression of a number of inammatory genes including is insensitive to GCs and can dramatically inuence the

234
Airway Smooth Muscle 18
anti-inammatory action of steroids via mechanisms that via the activation and the recruitment of inammatory cells.
remain to be determined. This study supports the hypothesis Further, cytokine-induced alterations in calcium homeostasis
that inammation is a key modulator of steroid responsive- render the ASM hyperresponsive to contractile agonists. In
ness in structural airway cells. chronic severe asthma and COPD, ASM hypertrophy and
Collectively, the fact that ASM cells can secrete and hyperplasia may potentially render the asthmatic airway irre-
express dierent immunomodulatory proteins, combined versibly obstructed.
with the functional expression of GR, give ASM cells a Further elucidation of the cellular and the molecular
particular importance as cellular targets for inhaled GC. mechanisms that regulate ASM function in asthma and
However, further in vivo studies are needed to conrm the COPD will oer new therapeutic targets in the treatment
aforementioned in vitro observations to determine the role of asthma, chronic bronchitis, and emphysema.
of ASM in modulating the anti-inammatory eects of
GC, notably in steroid-resistant patients.
While ASM has long been established as a key eec- References
tor cell in the pathogenesis of asthma, its role in COPD has
only recently been appreciated. Several studies have demon-
1. Amrani Y, Panettieri RA Jr. Cytokines induce airway smooth muscle
strated increased ASM mass in the small airways of smokers cell hyperresponsiveness to contractile agonists. Thorax 53: 71316, 1998.
with airow obstruction compared to smokers with normal 2. Giembycz MA, Raeburn D. Current concepts on mechanisms of force
lung function [153156]. In contrast to asthma, however, generation and maintenance in airway smooth muscle. Pulm Pharmacol
where large, central airways are the predominant site of 5: 27997, 1992.
ASM hypertrophy and hyperplasia, ASM hypertrophy in 3. Amrani Y, Magnier C, Wuytack F, Enouf J, Bronner C. Ca2 increase
COPD is uniquely observed in small airways with a diam- and Ca2 inux in human tracheal smooth muscle cells: Role of Ca2
eter of approximately 2 mm [155, 157, 158]. The extent of pools controlled by sarco-endoplasmic reticulum Ca2-ATPase 2 iso-
ASM hypertrophy observed in small airways correlates with forms. Br J Pharmacol 115: 120410, 1995.
the severity of airow obstruction [155, 156], though associ- 4. Murray RK, Kotliko MI. Receptor-activated calcium inux in human
airway smooth muscle cells. J Physiol (Lond) 435: 12344, 1991.
ated increases in matrix deposition in the walls of these air-
5. Putney JW. Excitement about calcium signaling in inexcitable cells.
ways may also contribute to airow obstruction by limiting Science 262, 67678, 1993.
the extent to which ASM relaxation results in increased air- 6. Gibson A, McFadzean I, Wallace P, Wayman CP. Capacitative Ca2
way luminal diameter [159]. Airway inammation appears entry and the regulation of smooth muscle tone. Trends Pharmacol Sci
to be one important cause of ASM hypertrophy in COPD, 19: 26669, 1998.
as activated T-lymphocytes have been demonstrated to bind 7. Janssen LJ. Calcium handling in airway smooth muscle: Mechanisms
VCAM-1 and CD44, inducing DNA synthesis [118]. and therapeutic implications. Can Respir J 5: 49198, 1998.
While ASM has historically been viewed as a tar- 8. Randriamampita C, Tsien RY. Emptying of intracellular Ca2 stores
get tissue, regulating bronchomotor tone and responding to releases a novel small messenger that stimulates Ca2 inux. Nature 364:
neurohumoral control and inammatory mediators, emerg- 80914, 1994.
9. Tao FC, Tolloczko B, Eidelman DH, Martin JG. Enhanced Ca(2)
ing evidence suggests that ASM may also play an important
mobilization in airway smooth muscle contributes to airway hyperre-
immunomodulatory role in the small airway inammation sponsiveness in an inbred strain of rat. Am J Respir Crit Care Med 160:
characteristic of COPD [160, 161]. ASM cells are capable 44653, 1999.
of expressing numerous proinammatory and chemotactic 10. Reynolds AM, Holmes MD, Scicchitano R. Cytokines enhance airway
cytokines and chemokines germane to COPD, in addition smooth muscle contractility in response to acetylcholine and neurokinin
to several relevant growth factors and proteases, as outlined A. Respirology 5: 15360, 2000.
above. Further, the pattern of ASM gene expression depends 11. Amrani Y, Chen H, Panettieri RA Jr.. Activation of tumor necrosis
highly on the inammatory environment [93, 96, 97, 162, factor receptor 1 in airway smooth muscle: A potential pathway that
163], underscoring the importance of interactions between modulates bronchial hyper-responsiveness in asthma? Respir Res 1:
ASM and the nearby epithelium. A great deal of work remains 4953, 2000.
12. Schmidlin F, Scherrer D, Daeer L, Bertrand C, Landry Y, Gies JP.
to be done to elucidate the role of ASM in the pathogenesis
Interleukin-1 induces bradykinin B2 receptor gene expression through
of COPD, but all available evidence indicates that it is likely a prostanoid cyclic AMP dependent pathway in human bronchial
an important component of the pathologic processes leading smooth muscle cells. Mol Pharmacol 53: 100915, 1998.
to airway remodeling and irreversible airow obstruction. 13. Hotta K, Emala CW, Hirshman CA. TNF- upregulates Gi and
Gq protein expression and function in human airway smooth muscle
cells. Am J Physiol (Lung Cell Mol Physiol) 276/20: L40511, 1999.
14. Roux E, Hyvelin JM, Savineau JP, Marthan R. Calcium signaling in air-
SUMMARY way smooth muscle cells is altered by in vitro exposure to the aldehyde
acrolein. Am J Respir Cell Mol Biol 19: 43744, 1998.
15. Wylam ME, Gungor N, Mitchell RW, Umans JG. Eosinophils, major
New evidence suggests that ASM, the major eector cell reg- basic protein, and polycationic peptides augment bovine airway myo-
cyte Ca2 mobilization. Am J Physiol. (Lung Cell Mol Physiol) 274:
ulating bronchomotor tone, also plays an important role as an
L9971005, 1998.
immunomodulatory cell. ASM, exposed to a variety of medi- 16. An SS, Hai CM. Mechanical strain modulates maximal phosphatidyli-
ators and cytokines, can undergo phenotypic changes that nositol turnover in airway smooth muscle. Am J Physiol (Lung Cell Mol
may be important in the pathogenesis of asthma and COPD. Physiol) 277: L96874, 1999.
ASM, which secretes chemokines and cytokines, may partici- 17. Takata Y, Nishimura Y, Maeda H, Yokoyama M. Phospholipase A2
pate in or even perpetuate the mucosal inammatory changes augments contraction and intracellular calcium mobilization through

235
Asthma and COPD: Basic Mechanisms and Clinical Management

thromboxane A2 in bovine tracheal smooth muscle. Eur Respir J 14: induces c-fos expression. Am J Physiol (Lung Cell Mol Physiol) 259/3:
396404, 1999. L36571, 1990.
18. Belouchi NE, Roux E, Savineau JP, Marthan R. Eect of chronic 39. Noveral JP, Grunstein MM. Role and mechanism of thromboxane-
hypoxia on calcium signalling in airway smooth muscle cells. Eur induced proliferation of cultured airway smooth muscle cells. Am J
Respir J 14: 7479, 1999. Physiol (Lung Cell Mol Physiol) 263/7: L55561, 1992.
19. Mak JC, Roel AF, Katsunuma T, Elzinga CR, Zaagsma J, Barnes PJ. 40. Panettieri RA Jr.. Airways smooth muscle cell growth and proliferation.
Up-regulation of airway smooth muscle histamine H(1) receptor mRNA, In: Raeburn D, Giembycz MA (eds). Airways Smooth Muscle: Development
protein, and function by beta(2)-adrenoceptor activation. Mol Pharmacol and Regulation of Contractility, pp. 4168. Basel, Switzerland: Birkhauser
57: 85764, 2000. Verlag, 1994.
20. Beasley R, Pearce N, Crane J, Burgess C. Beta-agonists: What is the 41. Panettieri RA Jr., Cohen MD, Bilgen G. Airway smooth muscle cell
evidence that their use increases the risk of asthma morbidity and mor- proliferation is inhibited by microinjection of the catalytic subunit of
tality? J Allergy Clin Immunol 104: S18S30, 1999. cAMP dependent kinase. Am Rev Respir Dis 147: A252, 1993.
21. Schmidt D, Rabe KF. Immune mechanisms of smooth muscle hyper- 42. Noveral JP, Rosenberg SM, Anbar RA, Pawlowski NA, Grunstein MM.
reactivity in asthma. Allergy Clin Immunol 105: 67382, 2000. Role of endothelin-1 in regulating proliferation of cultured rabbit
22. Nakatani Y, Nishimura Y, Nishiumua T, Maeda H, Yokoyama M. Tumor airway smooth muscle cells. Am J Physiol (Lung Cell Mol Physiol) 263/7:
necrosis factor- augments contraction and cytosolic Ca2 through L31724, 1992.
phospholipase A2 in bovine tracheal smooth muscle. Eur J Pharm 392: 43. Panettieri RA Jr., Murray RK, DePalo LR, Yadvish PA, Kotliko MI.
17582, 2000. A human airway smooth muscle cell line that retains physiological
23. Parris JR, Cobban HJ, Littlejohn AF, MacEwan DJ, Nixon GF. Tumor responsiveness. Am J Physiol: Cell Physiol 256/25: C32935, 1989.
necrosis factor- activates a calcium sensitization pathway in guinea-pig 44. Panettieri RA Jr., Tan EML, Ciocca V, Luttmann MA, Leonard TB,
bronchial smooth muscle. J Physiol 518: 56169, 1999. Hay DWP. Eects of LTD4 on human airways smooth muscle cell
24. Panettieri RA Jr., Kotliko MI. Cellular and molecular mechanisms proliferation, matrix expression, and contraction in vitro: Dierential
regulating airway smooth muscle cell physiology and pharmacology. sensitivity to cysteinyl leukotriene receptor antagonists. Am J Respir
In: Fishman AP, Elias JA, Fishman JA, Grippi MA, Kaiser LR, Senior Cell Mol Biol 19: 45361, 1998.
RM (eds). Pulmonary Diseases and Disorders, 3rd edn, pp. 10717. New 45. Engelman JA, Ji L, Cantley LC. The evolution of phosphatidylinositol
York: McGraw-Hill, 1998. 3-kinases as regulators of growth and metabolism. Nat Rev Genet 7:
25. Condlie aM, Cadwallader KA, Walker TR, Rintoul RC, Cowburn 60619, 2006.
AS, Chilvers ER. Phosphoinositide 3-kinase: A critical signaling event 46. Krymskaya VP. Targeting phosphatidylinositol 3-kinase pathway in
in pulmonary cells. Respir Res 1: 2429, 2000. airway smooth muscle: Rationale and promise. BioDrugs 21: 8595, 2007.
26. Hershenson MB, Abe MK. Mitogen-activated signaling in airway 47. Rameh LE, Cantley LC. The role of phosphoinositide 3-kinase lipid
smooth muscle. A central role for Ras. Am J Respir Cell Mol Biol 21: products in cell function. J Biol Chem 274: 834750, 1999.
65154, 1999. 48. Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C,
27. Page K, Hershenson MB. Mitogen-activated signaling and cell cycle Rodgers L, McCombie R, Bigner SH, Giovanella BC, Ittmann M,
regulation in airway smooth muscle. Front Biosci 5: d25867, 2000. Tycko B, Hibshoosh H, Wigler MH, Parsons R. PTEN, a putative
28. Thomas CF Jr., Limper AH. Phosphatidylinositol kinase regulation of protein tyrosine phosphatase gene mutated in human brain, breast, and
airway smooth muscle cell proliferation. Am J Respir Cell Mol Biol 23: prostate cancer. Science 275: 194347, 1997.
42930, 2000. 49. Maehama T, Taylor GS, Dixon JE. PTEN and myotubularin:
29. Hirst SJ, Barnes PJ, Twort CHC. Quantifying proliferation of cultured Novel phosphoinositide phosphatases. Annu Rev Biochem 70: 24779,
human and rabbit airway smooth muscle cells in response to serum 2001.
and platelet-derived growth factor. Am J Respir Cell Mol Biol 7: 57481, 50. Steck PA, Pershouse MA, Jasser SA, Yung WKA, Lin HK, Ligon
1992. AH, Langford LA, Baumgard ML, Hattier T, Davis T, Frye C, Hu R,
30. Panettieri RA Jr., Hall IP, Maki CS, Murray RK. -Thrombin increases Swedlund B, Teng DHR, Tavtigian SV. Identication of a candidate
cytosolic calcium and induces human airway smooth muscle cell prolif- tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is
eration. Am J Respir Cell Mol Biol 13: 20516, 1995. mutated in multiple advanced cancers. Nat Genet 15: 35662, 1997.
31. Stewart AG, Grigoriadis G, Harris T. Mitogenic actions of endothelin-1 51. Toker A. Protein kinases as mediators of phosphoinositide 3-kinase
and epidermal growth factor in cultured airway smooth muscle. Clin signaling. Mol Pharmacol 57: 65258, 2000.
Exp Pharmacol Physiol 21: 27785, 1994. 52. Vanhaesebroeck B, Watereld MD. Signaling by distinct classes of
32. Cohen P, Noveral JP, Bhala A, Nunn SE, Herrick DJ, Grunstein MM. phosphoinositide 3-kinases. Exp Cell Res 253: 23954, 1999.
Leukotriene D4 facilitates airway smooth muscle cell proliferation 53. Chantry D, Vojtek A, Kashishian A, Holtzman D, Wood C, Gray PW,
via modulation of the IGF axis. Am J Physiol (Lung Cell Mol Physiol) Cooper JA, Hoekstra MF. p110, a novel phosphatidylinositol 3-kinase
269/13: L15157, 1995. catalytic subunit that associates with p85 and is expressed predomi-
33. Kelleher MD, Abe MK, Chao T-SO, Jain M, Green JM, Solway J, nantly in leukocytes. J Biol Chem 272: 1923641, 1997.
Rosner MR, Hershenson MB. Role of MAP kinase activation in 54. Stoyanov B, Volonia S, Hanck T, Rubio I, Loubtchenkov M, Malek D,
bovine tracheal smooth muscle mitogenesis. Am J Physiol (Lung Cell Stoyanova S, Vanhaesebroeck B, Dhand R, Nurnberg B, Gierschik P,
Mol Physiol) 268/12: L894901, 1995. Seedorf K, Hsuan JJ, Watereld MD, Wetzker R. Cloning and charac-
34. Noveral JP, Bhala A, Hintz RL, Grunstein MM, Cohen P. The insulin- terization of a G protein-activated human phosphoinositide 3-kinase.
like growth factor axis in airway smooth muscle cells. Am J Physiol (Lung Science 269: 69093, 1995.
Cell Mol Physiol) 267/11: L76165, 1994. 55. Domin J, Gaidarov I, Smith MEK, Keen JH, Watereld MD. The class
35. Hepler JR, Gilman AG. G proteins. Trends Biochem Sci 17: 38387, II phosphoinositide 3-kinase PI3K-C2 is concentrated in the trans-
1992. Golgi network and present in clathrin-coated vesicles. J Biol Chem 275:
36. Sternweis PC. The active role of in signal transduction. Curr Opin 1194350, 2000.
Cell Biol 6: 198203, 1994. 56. Volinia S, Dhand R, Vanhaesebroeck B, MacDougall L, Stein R,
37. Lamorte VJ, Harootunian AT, Spiegel AM, Tsien RY, Feransisco Zvelebil MJ, Domin J, Panaretou C, Watereld MD. A human phos-
JR. Mediation of growth factor induced DNA synthesis and calcium phatidylinositol 3-kinase complex related to the yeast Vps34p-Vps15p
mobilization by Gq and Gi2. J Cell Biol 121: 9199, 1993. protein sorting system. EMBO J 14: 333948, 1995.
38. Panettieri RA Jr., Yadvish PA, Kelly AM, Rubinstein NA, Kotliko 57. Dan HC, Sun M, Yang L, Feldman RI, Sui XM, Yeung RS, Halley
MI. Histamine stimulates proliferation of airway smooth muscle and DJJ, Nicosia SV, Pledger WJ, Cheng JQ. PI3K/AKT pathway regulates

236
Airway Smooth Muscle 18
TSC tumor suppressor complex by phosphorylation of tuberin. J Biol 76. Chung J, Grammer TC, Lemon KP, Kazlauskas A, Blenis J. PDGF-
Chem 277: 3536470, 2002. and insulin-dependent pp70S6k activation mediated by phosphatidyli-
58. Manning BD, Tee AR, Longdon MN, Blenis J, Cantley LC. nositol-3-OH kinase. Nature 370: 7175, 1994.
Identication of the tuberous sclerosis complex-2 tumor suppressor 77. Jeeries HBJ, Fumagalli S, Dennis PB, Reinhard C, Pearson RB,
gene product tuberin as a target of the phosphoinositide 3-kinase/Akt Thomas G. Rapamycin suppresses 5
TOP mRNA translation through
pathway. Mol Cell 10: 15162, 2002. inhibition of p70s6k. EMBO J 16: 3693704, 1997.
59. Potter CJ, Pedraza LG, Xu T. Akt regulates growth by directly phos- 78. Krymskaya VP, Orsini MJ, Eszterhas AJ, Brodbeck KC, Benovic JL,
phorylating Tsc2. Nature Cell Biol 4: 65865, 2002. Panettieri RA Jr., Penn RB. Mechanisms of proliferation synergy by
60. Goncharova EA, Goncharov DA, Eszterhas A, Hunter DS, Glassberg receptor tyrosine kinase and G protein-coupled receptor activation in
MK, Yeung RS, Walker CL, Noonan D, Kwiatkowski DJ, Chou MM, human airway smooth muscle. Am J Respir Cell Mol Biol 23: 54654,
Panettieri Jr RA, Krymskaya VP. Tuberin regulates p70s6 kinase acti- 2000.
vation and ribosomal protein S6 phosphorylation. A role for the TSC2 79. Billington CK, Kong KC, Bhattacharyya R, Wedegaertner PB, Panettieri
tumor suppressor gene in pulmonary lymphangioleiomyomatosis RA, Chan TO, Penn RB. Cooperative regulation of p70S6 kinase by
(LAM). J Bio chem 277: 3095867, 2002. receptor tyrosine kinases and G protein-coupled receptors augments air-
61. Goncharova EA, Krymskaya VP. Pulmonary lymphangioleiomyomatosis way smooth muscle growth. Biochemistry 44: 14595605, 2005.
(LAM): Progress and current challenges. J Cell Biochem 103: 36982, 80. Krymskaya VP, Goncharova EA, Ammit AJ, Lim PN, Goncharov DA,
2008. Eszterhas A, Panettieri RA Jr. Src is necessary and sucient for human
62. Long X, Ortiz-Vega S, Lin Y, Avruch J. Rheb binding to mammalian airway smooth muscle cell proliferation and migration. FASEB J 19:
target of rapamycin (mTOR) is regulated by amino acid suciency. 42830, 2005.
J Biol Chem 280: 2343336, 2005. 81. Billington CK, Penn RB. Signaling and regulation of G protein-coupled
63. Long X, Lin Y, Ortiz-Vega S, Yonezawa K, Avruch J. Rheb binds and receptors in airway smooth muscle. Respir Res 4: 123, 2003.
regulates the mTOR kinase. Curr Biol 15: 70213, 2005. 82. Brown MT, Cooper JA. Regulation, substrates and functions of src.
64. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation Biochim Biophys Acta 1287: 12149, 1996.
and regulation of Akt/PKB by the rictor-mTOR complex. Science 307: 83. Thomas SM, Brugge JS. Cellular functions regulated by Src family
1098101, 2005. kinases. Annu Rev Cell Dev Biol 13: 513609, 1997.
65. Jacinto E, Loewith R, Schmidt A, Lin S, Ruegg MA, Hall A, Hall MN. 84. Tsang F, Hwa Choo H, Dawe GS, Fred Wong WS. Inhibitors of the
Mammalian TOR complex 2 controls the actin cytoskeleton and is tyrosine kinase signaling cascade attenuated thrombin-induced guinea
rapamycin insensitive. Nature Cell Biol 6: 112228, 2004. pig airway smooth muscle cell proliferation. Biochem Biophys Res
66. Goncharova EA, Ammit AJ, Irani C, Carroll RG, Eszterhas AJ, Commun 293: 7278, 2002.
Panettieri RA Jr., Krymskaya VP. Phosphatidylinositol 3-kinase is 85. Sayeski PP, Ali MS. The critical role of c-Src and the Shc/Grb2/ERK2
required for proliferation and migration of human pulmonary vas- signaling pathway in angiotensin II-dependent VSMC proliferation.
cular smooth muscle cells. Am J Physiol (Lung Cell Mol Physiol) 283: Exp Cell Res 287: 33949, 2003.
L354L363, 2002. 86. Luttrell LM, Ferguson SSG, Daaka Y, Miller WE, Maudsley S, Della
67. Krymskaya VP, Ammit AJ, Homan RK, Eszterhas AJ, Panettieri RA Jr.. Rocca GJ, Lin FT, Kawakatsu H, Owada K, Luttrell DK, Caron MG,
Activation of class IA phosphatidylinositol 3-kinase stimulates DNA Lefkowitz RJ. -Arrestin-dependent formation of 2 adrenergic receptor-
synthesis in human airway smooth muscle cells. Am J Physiol (Lung Cell Src protein kinase complexes. Science 283: 65561, 1999.
Mol Physiol) 280: L1009L1018, 2001. 87. Marinissen MJ, Gutkind JS. G-protein-coupled receptors and signal-
68. Bacqueville D, Casagrande F, Perret B, Chap H, Darbon JM, Breton- ing networks: Emerging paradigms. Trends Pharmacol Sci 22: 36876,
Douillon M. Phosphatidylinositol 3-kinase inhibitors block aortic 2001.
smooth muscle cell proliferation in mid-late G1 phase: Eect on cyclin- 88. Krymskaya VP, Homan R, Eszterhas A, Kane S, Ciocca V,
dependent kinase 2 and the inhibitory protein p27KIP1. Biochem Biophys Panettieri RA Jr. EGF activates ErbB-2 and stimulates phosphatidyli-
Res Commun 244: 63036, 1998. nositol 3-kinase in human airway smooth muscle cells. Am J Physiol
69. Saward L, Zahradka P. Angiotensin II activates phosphatidylinositol (Lung Cell Mol Physiol) 276/20: L24655, 1999.
3-kinase in vascular smooth muscle cells. Circ Res 81: 24957, 1997. 89. Lazaar AL, Amrani Y, Panettieri RA Jr.. The role of inammation in
70. Scott PH, Belham CM, Al-Hadh J, Chilvers ER, Peacock AJ, Gould the regulation of airway smooth muscle cell function and growth. In:
GW, Plevin R. A regulatory role for cAMP in phosphatidylinositol Busse W, Holgate S (eds). Asthma and Rhinitis, 2nd edn, pp. 140213.
3-kinase/p70 ribosomal S6 kinase-mediated DNA synthesis in platelet- Oxford, UK: Blackwell Science, Ltd., 2000.
derived-growth-factor-stimulated bovine airway smooth-muscle cells. 90. Berkman N, Krishnan VL, Gilbey T, Newton R, OConnor B, Barnes
Biochem J 318: 96571, 1996. PJ, Chung KF. Expression of RANTES mRNA and protein in airways
71. Weiss RH, Apostolids A. Dissociation of phosphatidylinositol 3-kinase of patients with mild asthma. Am J Respir Crit Care Med 154: 180411,
activity and mitogenic inhibition in vascular smooth muscle cells. Cell 1996.
Signal 7: 11322, 1995. 91. Ammit AJ, Homan RK, Amrani Y, Lazaar AL, Hay DWP,
72. Hu ZW, Shi XY, Homan BB. 1 Adrenergic receptors activate phos- Torphy TJ, Penn RB, Panettieri RA Jr. TNF-induced secretion
phatidylinositol 3-kinase in human vascular muscle cells. J Biol Chem of RANTES and IL-6 from human airway smooth muscle cells:
271: 897782, 1996. Modulation by cAMP. Am J Respir Cell Mol Biol 23: 794802, 2000.
73. Takahashi T, Kawahara Y, Okuda M, Ueno H, Takeshita A, Yokoyama 92. John M, Hirst SJ, Jose PJ, Robichaud A, Berkman N, Witt C, Twort
M. Angiotensin II stimulates mitogen-activated protein kinases and CHC, Barnes PJ, Chung KF. Human airway smooth muscle cells
protein synthesis by a Ras-independent pathway in vascular smooth express and release RANTES in response to T helper 1 cytokines.
muscle cells. J Biol Chem 272: 1601822, 1997. Regulation by T helper 2 cytokines and corticosteroids. J Immunol 158:
74. Krymskaya VP, Penn RB, Orsini MJ, Scott PH, Plevin RJ, Walker 184147, 1997.
TR, Eszterhas AJ, Amrani Y, Chilvers ER, Panettieri RA Jr. 93. Pype JL, Dupont LJ, Menten P, Van Coillie E, Opdenakker G, Van
Phosphatidylinositol 3-kinase mediates mitogen-induced human air- Damme J, Chung KF, Demedts MG, Verleden GM. Expression of
ways smooth muscle cell proliferation. Am J Physiol (Lung Cell Mol monocyte chemotactic protein (MCP)-1, MCP-2, and MCP-3 by
Physiol) 277/21: L65L78, 1999. human airway smooth-muscle cells. Modulation by corticosteroids and
75. Hu Q, Klippel A, Muslin AJ, Fantl WJ, Williams LT. Ras-dependent T-helper 2 cytokines. Am J Respir Cell Mol Biol 21: 52836, 1999.
induction of cellular response by constitutively active phosphatidyli- 94. Chung KF, Patel HJ, Fadlon EJ, Rousell J, Haddad E-B, Jose PJ,
nositol-3 kinase. Science 268: 1002, 1995. Mitchell J, Belvisi M. Induction of eotaxin expression and release from

237
Asthma and COPD: Basic Mechanisms and Clinical Management

human airway smooth muscle cells by IL-1 and TNF: Eects of IL-6-type cytokines. Am J Physiol (Lung Cell Mol Physiol) 273/17:
IL-10 and corticosteroids. Br J Pharmacol 127: 114550, 1999. L64855, 1997.
95. Ghaar O, Hamid Q, Renzi PM, Allahkverdi Z, Molet S, Hogg JC, 112. McKay S, Hirst SJ, Bertrand-de Haas M, de Jonste JC, Hoogsteden HC,
Shore SA, Luster AD, Lamkhioued B. Constitutive and cytokine- Saxena PR, Sharma HS. Tumor necrosis factor- enhances mRNA
stimulated expression of eotaxin by human airway smooth muscle expression and secretion of interleukin-6 in cultured human airway
cells. Am J Respir Crit Care Med 159: 193342, 1999. smooth muscle cells. Am J Respir Cell Mol Biol 23: 10311, 2000.
96. John M, Au B-T, Jose PJ, Lim S, Saunders M, Barnes PJ, Mitchell JA, 113. DiCosmo BF, Geba GP, Picarella D, Elias JA, Rankin JA, Stripp BR,
Belvisi MG, Chung KF. Expression and release of interleukin-8 by Whitsett JA, Flavell RA. Airway epithelial cell expression of inter-
human airway smooth muscle cells: Inhibition by Th-2 cytokines and leukin-6 in transgenic mice. Uncoupling of airway inammation and
corticosteroids. Am J Respir Cell Mol Biol 18: 8490, 1998. bronchial hyperreactivity. J Clin Invest 94: 202835, 1994.
97. Pang L, Knox AJ. Bradykinin stimulates IL-8 production in cultured 114. Hakonarson H, Maskeri N, Carter C, Chuang S, Grunstein MM.
human airway smooth muscle cells: Role of cyclooxygenase products. Autocrine interaction between IL-5 and IL-1 mediates altered
J Immunol 161: 250915, 1998. responsiveness of atopic asthmatic sensitized airway smooth muscle.
98. Govindaraju V, Michoud MC, Al-Chalabi M, Ferraro P, Powell J Clin Invest 104: 65767, 1999.
WS, Martin JG. Interleukin-8: Novel roles in human airway smooth 115. Knight DA, Lydell CP, Zhou D, Weir TD, Schellenberg RR, Bai TR.
muscle cell contraction and migration. Am J Physiol: Cell Physiol 291: Leukemia inhibitory factor (LIF) and LIF receptor in human lung:
C95765, 2006. Distribution and regulation of LIF release. Am J Respir Cell Mol Biol
99. Mullan CS, Riley M, Clarke D, Tatler A, Sutclie A, Knox AJ, Pang L. 20: 83441, 1999.
Beta-tryptase regulates IL-8 expression in airway smooth muscle 116. Hallsworth MP, Soh CPC, Twort CHC, Lee TH, Hirst SJ. Cultured
cells by a PAR-2-independent mechanism. Am J Respir Cell Mol Biol human airway smooth muscle cells stimulated by interleukin-1
38(5):6008, 2008. enhance eosinophil survival. Am J Respir Cell Mol Biol 19: 91019, 1998.
100. Dragon S, Rahman MS, Yang J, Unruh H, Halayko AJ, Gounni AS. 117. Springer TA. Adhesion receptors of the immune system. Nature 346:
IL-17 enhances IL-1beta-mediated CXCL-8 release from human 42534, 1990.
airway smooth muscle cells. Am J Physiol (Lung Cell Mol Physiol) 292: 118. Lazaar AL, Albelda SM, Pilewski JM, Brennan B, Pur E, Panettieri
L102329, 2007. RA Jr.. T lymphocytes adhere to airway smooth muscle cells via
101. Zuyderduyn S, Ninaber DK, Hiemstra PS, Rabe KF. The antimicro- integrins and CD44 and induce smooth muscle cell DNA synthesis.
bial peptide LL-37 enhances IL-8 release by human airway smooth J Exp Med 180: 80716, 1994.
muscle cells. J Allergy Clin Immunol 117: 132835, 2006. 119. Amrani Y, Lazaar AL, Panettieri RA Jr. Up-regulation of ICAM-
102. Odaka M, Matsukura S, Kuga H, Kokubu F, Kasama T, Kurokawa M, 1 by cytokines in human tracheal smooth muscle cells involves an
Kawaguchi M, Ieki K, Suzuki S, Watanabe S, Homma T, Takeuchi H, NF-B-dependent signaling pathway that is only partially sensitive
Nohtomi K, Schleimer RP, Adachi M. Dierential regulation of to dexamethasone. J Immunol 163: 212834, 1999.
chemokine expression by Th1 and Th2 cytokines and mechanisms of 120. Belvisi MG, Saunders MA, Haddad E-B, Hirst SJ, Yacoub MH,
eotaxin/CCL-11 expression in human airway smooth muscle cells. Barnes PJ, Mitchell JA. Induction of cyclo-oxygenase-2 by cytokines
Int Arch Allergy Immunol 143(Suppl 1): 8488, 2007. in human cultured airway smooth muscle cells: Novel inammatory
103. Rahman MS, Yamasaki A, Yang J, Shan L, Halayko AJ, Gounni AS. role of this cell type. Br J Pharmacol 120: 91016, 1997.
IL-17A induces eotaxin-1/CC chemokine ligand 11 expression in 121. Pang L, Knox AJ. Eect of interleukin-1, tumour necrosis factor-
human airway smooth muscle cells: Role of MAPK (Erk1/2, JNK, and interferon- on the induction of cyclo-oxygenase-2 in cultured
and p38) pathways. J Immunol 177: 406471, 2006. human airway smooth muscle cells. Br J Pharmacol 121: 57987, 1997.
104. Chan V, Burgess JK, Rato JC, OConnor B J, Greenough A, Lee 122. Panettieri RA Jr., Lazaar AL, Pur E, Albelda SM. Activation of
TH, Hirst SJ. Extracellular matrix regulates enhanced eotaxin expres- cAMP-dependent pathways in human airway smooth muscle cells
sion in asthmatic airway smooth muscle cells. Am J Respir Crit Care inhibits TNF--induced ICAM-1 and VCAM-1 expression and T
Med 174: 37985, 2006. lymphocyte adhesion. J Immunol 154: 235865, 1995.
105. Liu L, Yang J, Huang Y. Human airway smooth muscle cells express 123. Dustin ML, Springer TA. Role of lymphocyte adhesion receptors
eotaxin in response to signaling following mast cell contact. Respiration in transient interactions and cell locomotion. Annu Rev Immunol 9:
73: 22735, 2006. 2766, 1991.
106. Pang L, Knox AJ. Regulation of TNF--induced eotaxin release from 124. van Seventer GA, Newman W, Shimuzu Y, Nutman TB, Tanaka
cultured human airway smooth muscle cells by 2-agonists and corti- Y, Horgan KJ, Gopal TV, Ennis E, OSullivan D, Grey H. Analysis
costeroids. FASEB J 115: 26169, 2001. of T cell stimulation by superantigen plus major histocompat-
107. Pang L, Knox AJ. Synergistic inhibition by 2-agonists and corticos- ibility complex class II molecules or by CD3 monoclonal antibody:
teroids on tumor necrosis factor--induced interleukin-8 release from Costimulation by puried adhesion ligands VCAM-1, ICAM-1, but
cultured human airway smooth-muscle cells. Am J Respir Cell Mol Biol not ELAM-1. J Exp Med 174: 90113, 1991.
23: 7985, 2000. 125. Lazaar AL, Reitz HE, Panettieri RA Jr., Peters SP, Pur E. Antigen
108. Ammit AJ, Hastie AT, Edsall LC, Homan RK, Amrani Y, receptor-stimulated peripheral blood and bronchoalveolar lavage-derived
Krymskaya VP, Kane SA, Peters SP, Penn RB, Spiegel S, Panettieri T cells induce MHC class II and ICAM-1 expression on human airway
RA Jr.. Sphingosine 1-phosphate modulates human airway smooth smooth muscle. Am J Respir Cell Mol Biol 16: 3845, 1997.
muscle cell functions that promote inammation and airway remod- 126. Lazaar AL, Amrani Y, Hsu J, Panettieri RA Jr., Fanslow WC, Albelda
eling in asthma. FASEB J 15: 121214, 2001. SM, Pur E. CD40-mediated signal transduction in human airway
109. Saunders MA, Mitchell JA, Seldon PM, Yacoub MH, Barnes PJ, smooth muscle. J Immunol 161: 312027, 1998.
Giembycz MA, Belvisi MG. Release of granulocyte-macrophage 127. Hakonarson H, Kim C, Whelan R, Campbell D, Grunstein MM. Bi-
colony stimulating factor by human cultured airway smooth muscle directional activation between human airway smooth muscle cells and
cells: Suppression by dexamethasone. Br J Pharmacol 120: 54546, T lymphocytes: Role in induction of altered airway responsiveness.
1997. J Immunol 166: 293303, 2001.
110. De S, Zelazny ET, Souhrada JF, Souhrada M. IL-1 and IL-6 induce 128. Lazaar AL, Krymskaya VP, Das SK. VCAM-1 activates phosphati-
hyperplasia and hypertrophy of cultured guinea pig airway smooth dylinositol 3-kinase and induces p120(Cbl) phosphorylation in
muscle cells. J Appl Physiol 78: 155563, 1995. human airway smooth muscle cells. J Immunol 166: 15561, 2001.
111. Elias JA, Wu Y, Zheng T, Panettieri RA Jr. Cytokine- and virus- 129. Gaston B, Drazen JM, Loscalzo J, Stamler JS. The biology of nitrogen
stimulated airway smooth muscle cells produce IL-11 and other oxides in the airways. Am J Respir Crit Care Med 149: 53851, 1994.

238
Airway Smooth Muscle 18
130. Hamad AM, Johnson SR, Knox AJ. Antiproliferative eects of NO induction of cyclo-oxygenase-2: Inhibition by dexamethasone and
and ANP in cultured human airway smooth muscle. Am J Physiol 277: uticasone propionate. Br J Pharmacol 126: 131524, 1999.
L91018, 1999. 146. Adcock IM, Gilbey T, Gelder CM, Chung KF, Barnes PJ.
131. Patel HJ, Belvisi MG, Donnelly LE, Yacoub MH, Chung KF, Glucocorticoid receptor localization in normal and asthmatic lung.
Mitchell JA. Constitutive expressions of type I NOS in human airway Am J Respir Crit Care Med 154: 77182, 1996.
smooth muscle cells: Evidence for an antiproliferative role. FASEB J 147. Roth M, Johnson PR, Borger P, Bihl MP, Rudiger JJ, King GG, Ge
13: 181016, 1999. Q, Hostettler K, Burgess JK, Black JL, Tamm M. Dysfunctional inter-
132. Hakonarson H, Grunstein MM. Autologously up-regulated Fc recep- action of C/EBPalpha and the glucocorticoid receptor in asthmatic
tor expression and action in airway smooth muscle mediates its altered bronchial smooth-muscle cells. N Engl J Med 351: 56074, 2004.
responsiveness in the atopic asthmatic sensitized state. Proc Natl Acad 148. Bonacci JV, Schuliga M, Harris T, Stewart AG. Collagen impairs glu-
Sci USA 95: 525762, 1998. cocorticoid actions in airway smooth muscle through integrin signal-
133. Roper RL, Conrad DH, Brown DM, Warner GL, Phipps RP. ling. Br J Pharmacol 149: 36573, 2006.
Prostaglandin E2 promotes IL-4-induced IgE and IgG1 synthesis. J 149. Roth M, Johnson PR, Rudiger JJ, King GG, Ge Q, Burgess JK,
Immunol 145: 264451, 1990. Anderson G, Tamm M, Black JL. Interaction between glucocorticoids
134. Lazzeri N, Belvisi MG, Patel HJ, Yacoub MH, Fan Chung K, and beta2 agonists on bronchial airway smooth muscle cells through
Mitchell JA. Eects of prostaglandin E(2) and cAMP elevating drugs synchronised cellular signalling. Lancet 360: 129399, 2002.
on GM-CSF release by cultured human airway smooth muscle cells. 150. Hirst SJ, Lee TH. Airway smooth muscle as a target of glucocorticoid
Relevance to asthma therapy. Am J Respir Cell Mol Biol 24: 4448, action in the treatment of asthma. Am J Respir Crit Care Med 158:
2001. S2016, 1998.
135. Issa R, Xie S, Khorasani N, Sukkar M, Adcock IM, Lee KY, Chung 151. Miller M, Cho JY, McElwain K, McElwain S, Shim JY, Manni M,
KF. Corticosteroid inhibition of growth-related oncogene protein- Baek JS, Broide DH. Corticosteroids prevent myobroblast accumu-
alpha via mitogen-activated kinase phosphatase-1 in airway smooth lation and airway remodeling in mice. Am J Physiol (Lung Cell Mol
muscle cells. J Immunol 178: 736675, 2007. Physiol) 290: L16269, 2006.
136. Zhang K, Shan L, Rahman MS, Unruh H, Halayko AJ, Gounni AS. 152. Tliba O, Damera G, Banerjee A, Gu S, Baidouri H, Keslacy S,
Constitutive and inducible thymic stromal lymphopoietin expres- Amrani Y. Cytokines induce an early steroid resistance in airway
sion in human airway smooth muscle cells: Role in chronic obstruc- smooth muscle cells: novel role of interferon regulatory factor-1. Am J
tive pulmonary disease. Am J Physiol (Lung Cell Mol Physiol) 293: Respir Cell Mol Biol 38(4): 46372, 2008.
L37582, 2007. 153. Bosken CH, Wiggs BR, Pare PD, Hogg JC. Small airway dimensions
137. Sukkar MB, Xie S, Khorasani NM, Kon OM, Stanbridge R, Issa R, in smokers with obstruction to airow. Am Rev Respir Dis 142: 563
Chung KF. Toll-like receptor 2, 3, and 4 expression and function in 70, 1990.
human airway smooth muscle. J Allergy Clin Immunol 118: 64148, 154. Cosio MG, Hale KA, Niewoehner DE. Morphologic and morpho-
2006. metric eects of prolonged cigarette smoking on the small airways.
138. Kang BN, Tirumurugaan KG, Deshpande DA, Amrani Y, Panettieri Am Rev Respir Dis 122: 265321, 1980.
RA, Walseth TF, Kannan MS. Transcriptional regulation of CD38 155. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
expression by tumor necrosis factor-alpha in human airway smooth Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The
muscle cells: Role of NF-kappaB and sensitivity to glucocorticoids. nature of small-airway obstruction in chronic obstructive pulmonary
FASEB J 20: 10002, 2006. disease. N Engl J Med 350: 264553, 2004.
139. Keslacy S, Tliba O, Baidouri H, Amrani Y. Inhibition of TNF- 156. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L, Mapp
inducible inammatory genes by IFN is associated with altered NF- CE, Maestrelli P, Ciaccia A, Fabbri LM. CD8 T-lymphocytes in
B transactivation and enhanced HDAC activity. Mol Pharmacol 71: peripheral airways of smokers with chronic obstructive pulmonary
60918, 2007. disease. Am J Respir Crit Care Med 157: 82226, 1998.
140. Vanaudenaerde BM, Wuyts WA, Geudens N, Dupont LJ, Schoofs 157. Kuwano K, Bosken CH, Pare PD, Bai TR, Wiggs BR, Hogg JC.
K, Smeets S, Van Raemdonck DE, Verleden GM. Macrolides inhibit Small airways dimensions in asthma and in chronic obstructive pul-
IL17-induced IL8 and 8-isoprostane release from human airway monary disease. Am Rev Respir Dis 148: 122025, 1993.
smooth muscle cells. Am J Transplant 7: 7682, 2007. 158. Tiddens HA, Pare PD, Hogg JC, Hop WC, Lambert R, de Jongste
141. Catley MC, Sukkar MB, Chung KF, Jaee B, Liao SM, Coyle AJ, JC. Cartilaginous airway dimensions and airow obstruction in
Haddad el B, Barnes PJ, Newton R. Validation of the anti-inammatory human lungs. Am J Respir Crit Care Med 152: 26066, 1995.
properties of small-molecule IkappaB kinase (IKK)-2 inhibitors by 159. Cosio M, Ghezzo H, Hogg JC, Corbin R, Loveland M, Dosman J,
comparison with adenoviral-mediated delivery of dominant-negative Macklem PT. The relations between structural changes in small air-
IKK1 and IKK2 in human airways smooth muscle. Mol Pharmacol 70: ways and pulmonary-function tests. N Engl J Med 298: 127781, 1978.
697705, 2006. 160. Chung KF. The role of airway smooth muscle in the pathogenesis of
142. Ammit AJ, Lazaar AL, Irani C, ONeill GM, Gordon ND, Amrani Y, airway wall remodeling in chronic obstructive pulmonary disease. Proc
Penn RB, Panettieri RA Jr. Tumor necrosis factor--induced secretion Am Thoracic Soc 2: 34754, discussion 37172, 2005.
of RANTES and interleukin-6 from human airway smooth muscle 161. Panettieri RA Jr. Airway smooth muscle: An immunomodulatory cell.
cells: Modulation by glucocorticoids and -agonists. Am J Respir Cell J Allergy Clin Immunol 110: S26974, 2002.
Mol Biol 26: 46574, 2002. 162. Hardaker EL, Bacon AM, Carlson K, Roshak AK, Foley JJ, Schmidt
143. Tliba O, Cidlowski JA, Amrani Y. CD38 expression is insensitive to DB, Buckley PT, Comegys M, Panettieri JRA, Sarau HM, Belmonte
steroid action in cells treated with tumor necrosis factor-alpha and KE. Regulation of TNF-- and IFN--induced CXCL10 expression:
interferon-gamma by a mechanism involving the up-regulation of Participation of the airway smooth muscle in the pulmonary inam-
the glucocorticoid receptor beta isoform. Mol Pharmacol 69: 58896, matory response in chronic obstructive pulmonary disease. FASEB J
2006. 18: 19193, 2004.
144. Tran T, Fernandes DJ, Schuliga M, Harris T, Landells L, Stewart AG. 163. Jarai G, Sukkar M, Garrett S, Duroudier N, Westwick J, Adcock I,
Stimulus-dependent glucocorticoid-resistance of GM-CSF produc- Chung KF. Eects of interleukin-1beta, interleukin-13 and trans-
tion in human cultured airway smooth muscle. Br J Pharmacol 145: forming growth factor-beta on gene expression in human airway
12331, 2005. smooth muscle using gene microarrays. Eur J Pharmacol 497: 25565,
145. Vlahos R, Stewart AG. Interleukin-1alpha and tumour necrosis 2004.
factor-alpha modulate airway smooth muscle DNA synthesis by

239
Tracheobronchial Circulation
19CHAPTER

INTRODUCTION along the airways and anastomoses have been


demonstrated at pre-, post-, and capillary-levels Adam Wanner1 and
[2], venous blood from the intraparenchymal Gabor Horvath2
Since the blood circulation typically participates bronchial vasculature mainly drains through 1
Division of Pulmonary and Critical Care
in inammatory processes at the tissue level, the post-capillary pulmonary vessels [3] to the left
Medicine, University of Miami Miller
vasculature of the tracheobronchial tree can be heart [4]. Venous blood from the trachea and
expected to undergo structural and functional major bronchi drains through the vena azygos School of Medicine, Miami, FL, USA
2
changes in asthma and chronic obstructive pul- and superior vena cava to the right heart. Department of Pulmonology,
monary disease (COPD), conditions that are Under physiological conditions, total Semmelweiss University, Budapest,
associated with airway inammation. As will bronchial blood ow comprises 0.51% of car- Hungary
be shown in this chapter, there are distinct dif- diac output. The major part of blood ow is dis-
ferences in the vascular abnormalities between tributed to the subepithelial tissues where the
asthma and COPD. The purpose of this chap- microvasculature comprises 1020% of tissue
ter is to review the pathophysiological role of volume [5]. Subepithelial blood ow (Qaw) has
the tracheobronchial circulation in asthma and been reported to range between 30 and 95 ml
COPD and the vascular eects of pharmaco- min1 100 g wet tissue1 in dierent species
logic interventions. The in vivo measurement including in human [6, 7]. Its main function
of tracheobronchial blood ow in man usually presumably is to nourish the epithelium that has
captures subepithelial blood ow in the con- one of the highest metabolic rates in the body.
ducting airways, also termed airway blood ow In healthy human adults, Qaw in the tracheo-
(Qaw); therefore, this review will focus on Qaw bronchial tree to a 200 m depth from the epi-
in asthma and COPD, with a brief discussion thelial surface amounts to between 25 and 40 l
of the normal airway circulation as background. min1 ml1, where ml reects the anatomical
dead space lined by the epithelium [810].
Qaw is inuenced by a variety of factors
(Table 19.1). Although adrenergic and cholin-
NORMAL AIRWAY CIRCULATION ergic nerves have been demonstrated in the
airway wall [11, 12], physiological and pharma-
cological studies suggest that the main nervous
The airway circulation, which derives its blood control of the airway circulation is by the sym-
from the systemic circulation, is the principal pathetic nervous system [11]. In dierent ani-
vascular supply to the airway wall (Fig. 19.1) mal models, sympathetic nerve stimulation [13],
[1]. Bronchial arteries usually arise from the close-arterial injection of adrenergic agonists
aorta or intercostal arteries and form a peri- [14], and inhalation of selective -adrenergic
bronchial plexus surrounding the bronchial wall. agonists [15] have been shown to induce vaso-
Branches penetrate the muscular layer to form constriction. Selective 2-adrenergic agonists
a subepithelial plexus. The two interconnected increase bronchial blood ow in sheep [6, 8], a
plexuses follow airways as far as the terminal nding subsequently conrmed in humans [16]
bronchiole. Although bronchial capillaries anas- (Table 19.1). Parasympathetic nerve stimulation
tomose freely with the pulmonary circulation and intravascular administration of cholinergic

241
Asthma and COPD: Basic Mechanisms and Clinical Management

FIG. 19.1 Section of sheep bronchus


(glutaraldehyde-fixed at 100 mmHg aortic
pressure, paraffin embedded, HE-stained,
200). A: arterioles; C: capillaries; V: venule;
SG: submucosal gland; N: autonomic neuron.
Section kindly provided by A. Mariassy.

TABLE 19.1 Vasoactive substances that potentially could participate in and calcitonin gene-related peptide (CGRP) released from
asthma-and COPD associated airway inflammation. unmyelinated sensory aerents induce vasodilation [11].
Nitrergic mechanisms have recently been reported to regu-
Vasodilators Vasoconstrictors late bronchovascular tone and partially mediate -adrenergic
-adrenergic agonists -adrenergic agonists
vasodilator responses [15, 18, 19]. Finally, physicochemical
stresses including hydrostatic pressure, temperature, humid-
Cholinergic agonists NPY ity, deviations from normal osmolarity, hypoxia, and acid
base relations have all been reported to inuence airway
VIP, PHI, PHM LTC4, LTD4 blood ow [20]. These factors may be of relevance in asthma
NO TNF- and COPD where hydrostatic airway and pleural pressures
are altered by the abnormal lung mechanics; there may be
SP, NKA, CGRP ET-1 disturbances in gas exchange; there is airway inamma-
tion; and there may be hemodynamic changes with elevated
Histamine, bradykinin, PAF GCS (indirect)
pulmonary arterial, right atrial, and left atrial pressures due
5-HT PGE1, FGF2, PGD2 to secondary pulmonary hypertension or cardiovascular
comorbidity.
VEGF

VIP: vasoactive intestinal polypeptide; PHI: polypeptide histidine isoleucine; PHM:


polypeptide histidine methionine; NO: nitric oxide; SP: substance P; NKA: neurokinin
A; CGRP: calcitonin gene related peptide; PAF: platelet-activating factor; 5-HT: AIRWAY CIRCULATION IN ASTHMA
serotonine; PG: prostaglandin; VEGF: vascular endothelial growth factor; NPY:
neuropeptide Y; LT: leukotriene; TNF-: tumor necrosis factor-; ET-1: endothelin-1;
Allergic and non-allergic inammation is considered a major
GCS: glucocorticosteroids.
factor in the vascular changes that have been associated with
asthma [82]. The inammatory mechanisms include the
agonists cause vasodilatation in several animal models complex actions of inammatory cells and mediators, neuro-
[11], whereas aerosolized acetylcholine has only minimal transmitters, and neuropeptides on vascular endothelial and
eects [17]. Nonadrenergic, noncholinergic mechanisms smooth muscle cells. The main vascular manifestations are
have also been described. Vasoactive intestinal polypeptide hyperemia, hyperpermeability, and edema formation.
(VIP), polypeptide histidine isoleucine (PHI), or polypep-
tide histidine methionine (PHM) released by parasym-
pathetic nerves are vasodilators, whereas neuropeptide Y Structure
(NPY) released by sympathetic nerves is a vasoconstric-
tor in the tracheobronchial circulation [13, 14]. Sensory It has been recognized for many years that asthmatics have
neuropeptides, as substance P (SP), neurokinin A (NKA), an increased cross-sectional submucosal vascular area, an

242
Tracheobronchial Circulation 19
100 (A) Methoxamine/healthy
*p  0.05 versus all 15
Methoxamine/asthmatic
80 *
Qaw (l min1 ml)

10
60

Qaw (l* min1* ml1)


5
40

0
20

0 5 *
HNS HES CS COPD COPD A A
p.ICS p.ICS
10 *
FIG. 19.2 Airway blood flow (Qaw) in healthy non-smokers (HNS), healthy
*
ex-smokers (HES), current smokers (CS), COPD, COPD after long-term
treatment with an inhaled corticosteroid (COPD p.ICS), asthma (A), and 15
asthma after long-term treatment with an inhaled corticosteroid (A p.ICS). 0.0 0.4 0.8 1.2 1.6 2.0 2.4
Mean values with SE in brackets. Adapted from Refs [9] and [29]. Drug dose (mg)

(B)
*
increased size and number of vessels in their airway wall, 15 *
and intimal thickening [2124]. This can be considered the
vascular component of airway wall remodeling. The stimu- *
10
lus for new vessel formation in asthma is unclear, although Qaw (l* min1* ml1) *
various endothelial growth factors, inammatory cytokines,
and other putative angiogenic factors have been proposed 5
[2527]. It appears that the structural changes are already
present during childhood and may even be present in atopic 0
children without clinical asthma [28].

5

Function
10 Albuterol/healthy
Airway blood flow Albuterol/asthmatic
Allergic and non-allergic airway inammation has been 15
shown to be associated with an increase in Qaw (Fig. 19.2) 0.0 0.2 0.4 0.6 0.8 1.0 1.2
[6, 14]. This is also the case in subjects with stable asthma Drug dose (mg)
[810, 30]. An increase in Qaw is generally thought to result
from dilatation of resistance arteries. Numerous inamma- FIG. 19.3 Effects of inhaled methoxamine and albuterol on airway blood
tory mediators have a vasodilator eect [8]. Histamine has flow (Qaw) in 11 healthy (A) and 11 asthmatic (B) subjects. Mean values SE.
a triphasic eect characterized by an initial vasodilation, *p  0.05 versus baseline. Reproduced from Ref. [8].
followed by vasoconstriction and then a long-lasting vasodi-
lation [8]. Sensory neuropeptides released from aerent
nerves are also strong vasodilators. Reex bronchial vasodi-
lation is largely mediated by cholinergic and noncholinergic Microvascular hyperpermeability
parasympathetic vagal pathways [31]. Not all hemodynami- Asthma-associated leakage of macromolecules from the
cally active inammatory mediators are vasodilators. For microvasculature occurs through endothelial pores and
example, endothelin-1, a potent vasoconstrictor is increased vesicles or through the formation of intercellular gaps in
in the airway of asthmatics [32]. Its release may be induced post-capillary venules. Inammatory mediators and sensory
by pro-inammatory cytokines such as tumor necrosis autonomic nerve stimulation have been shown to increase
factor- [33]. However, the net eect of inammatory medi- microvascular permeability by producing intercellular gaps
ators is vasodilation. [11, 27] and induce interstitial edema formation. In addition,
While baseline Qaw is increased in asthma, vascular plasma components can collect in the airway lumen by pass-
reactivity is markedly deranged, with blunted 2-adrenergic ing through paracellular gaps between epithelial cells thereby
vasodilator responsiveness and enhanced -adrenergic vaso- contributing to excessive airway secretions. Vasodilation and
constrictor responsiveness (Fig. 19.3) [8, 9, 30, 34]. The former hyperperfusion-related microvascular congestion have been
observation suggests abnormal endothelium-dependent shown to potentiate microvascular hyperpemeability [35]. In
vasodilation in the airway circulation. this regard, blood ow is related to edema formation.

243
Asthma and COPD: Basic Mechanisms and Clinical Management

AIRWAY CIRCULATION IN COPD

Structure
Early reports on the structure of the airway circulation in
COPD suggested that intrapulmonary bronchial artery
branches are obliterated or narrowed in emphysematous
regions of the lungs and that the bronchial venous system
is greatly expanded [36, 37]. These ndings have not been
supported by recent studies that showed increased airway
wall vascularity in stable COPD with a slight increase in
the number of vessels in small airways, but to a much lesser
degree than in asthma, where there is an increased vascular-
ity throughout the bronchial tree [29, 38]. Thus, in contrast FIG. 19.4 Airway blood flow reactivity to inhaled albuterol (180 g) in
to asthma, angiogenesis in the airway wall does not appear healthy non-smokers (HNS), healthy ex-smokers (HES), current smokers
to be a typical feature of COPD. (CS), COPD, COPD after long-term treatment with an inhaled corticosteroid
(COPD p.ICS) and after washout of the medication (COPD p.w/0), asthma
However, in patients with COPD accompanied by
(A), and asthma after long-term treatment with an inhaled corticosteroid
bronchiectasis, there is an extensive remodeling of the air- (A p.ICS) and after the washout of the medication (A p. w/o). Vertical axis:
way circulation with a marked proliferation and enlarge- change in airway blood flow. Mean values with SE in brackets. Adapted
ment of the bronchial arteries and numerous pre-capillary from Refs [29] and [42].
anastomoses between the pulmonary and bronchial circula-
tion [3941]. This vascular remodeling is a likely cause of
hemoptysis encountered by patients with bronchiectasis. it is conceivable that the blunted albuterol responsiveness
It is also likely that the primary stimuli for the develop- of Qaw reects an abnormality in endothelium-dependent
ment of bronchiectasis and airway vascular hypertrophy relaxation. To what extent the defective vascular reactiv-
are suppurative airway infections that typically are seen in ity contributes to the development of COPD is unknown.
these patients. In support of this assumption, chronic lung It could be speculated that the blunted vasodilator response
abscesses also have been shown to lead to a localized hyper- in the airways reects systemic vascular disease that often
trophy and hyperplasia of the airway circulation [41]. co-exists with COPD [45, 46].

Function Cigarette Smoking

We found no signicant dierence in Qaw among ex-smok- Acute effects


ers with COPD (51  3 l min1 ml1 anatomical dead- In several extrapulmonary vascular beds, smoking a cigarette
space), healthy ex-smokers (HES) (41  3 l min1 ml1), has been shown to decrease blood ow transiently [4749].
healthy current smokers (46  2 l min1 ml1) and healthy The airway circulation is the rst and only systemic vascular
lifetime non-smokers (41  4 l min1 ml1), although the bed to be exposed to cigarette smoke directly and is exposed
values tended to be higher in COPD patients (Fig. 19.2) to the highest concentration of cigarette smoke constituents.
[29]. Similarly, Paredi et al. [38] reported normal Qaw val- Their eects on airway blood ow therefore may be dier-
ues in COPD. These ndings are consistent with the mor- ent from their systemic vascular eects. In anesthetized
phological observations that do not support the presence of pigs, cigarette smoke increases airway blood ow transiently
signicant new vessel formation in stable COPD in con- during the duration of inhalation, an eect that resembles
trast to asthma. However, airway blood ow is increased inhalation challenge with NO [50, 51]. This could be inter-
in patients with COPD and bronchiectasis, reecting the preted as indicating a vasodilator eect of NO contained in
above described hyperplasia of the airway circulation in cigarette smoke. Inasmuch as cigarette smoke is an irritant,
bronchiectasis [37]. more prolonged vascular eects could be expected in con-
Albuterol-induced vasodilation is blunted in ex-smokers scious humans. Furthermore, cigarette smoke contains nico-
with COPD (Fig. 19.4) [29], similarly to patients with tine, a sympathomimetic that has variable hemodynamic
asthma [8, 9]. Albuterol-induced vasodilation has been actions in dierent vascular beds [52, 53].
used as an index of endothelium-dependent vasodilation We found that Qaw increased by a mean of 81% at
and endothelial function, especially in current smokers [43]. 5 min after smoking a cigarette and returned to baseline by
Since the COPD patients were ex-smokers and the asth- 30 min in healthy smokers [54], whereas nicotine admin-
matics lifetime non-smokers, it cannot be assumed that istered by nasal spray (systemic action) or oral inhalation
the abnormal albuterol responsiveness was due to endothe- (local and systemic action) had no eect on Qaw, suggesting
lial dysfunction. However, inasmuch as oxidative stress has a pharmacologic or irritant eect of other cigarette smoke
been closely associated with endothelial dysfunction [43, constituents. Thus, smoking a cigarette seems to cause
44] and endogenously generated oxidants have been impli- transient vasodilation in the airway and vasoconstriction
cated in the pathogenesis of both COPD and asthma, in extrapulmonary systemic vascular beds. Whether or not

244
Tracheobronchial Circulation 19
repetitive vasodilator responses to cigarette smoking have a when blood ow was increased [61], whereas inhaled his-
role in the pathogenesis of COPD is not known. tamine [62] and antigen challenge-induced [63] bronchoc-
onstriction were prolonged when bronchial blood ow was
Long-term effects decreased. Therefore one might speculate that the inam-
While healthy current and ex-smokers have a near-normal matory increase in airway blood ow would enhance the
baseline Qaw as mentioned above, they have an abnormal clearance of locally released inammatory substances (e.g.
vasodilator response to albuterol, presumably as a manifesta- spasmogens) and decrease the magnitude and duration
tion of endothelial dysfunction as shown by Mendes et al. [29] of the eect of inhaled bronchoactive drugs. In addition,
(Fig. 19.4). There was a partial recovery of albuterol respon- increased airway blood ow could favor the distribution
siveness in healthy ex-smokers while albuterol responsiveness of systemically administered drugs to the airways and the
remained blunted in the ex-smokers with COPD. The partial accumulation of inammatory cells in the airway wall. Thus,
reversibility of albuterol responsiveness after smoking cessa- the inammatory increase in airway blood ow appears to
tion in healthy ex-smokers but not in ex-smokers with COPD have clinically benecial as well as undesirable eects on
suggests that the abnormal vascular reactivity is sustained in inammation and its drug treatment.
COPD, possibly due to ongoing airway inammation.
It may well be that the abnormal albuterol responsive-
ness in smokers reects systemic endothelial dysfunction Heat and water exchange
and that there is a correlation between systemic endothe-
Exercise-induced bronchoconstriction is a common condi-
lial dysfunction and albuterol-induced vascular relaxation
tion in asthmatics. Reactive hyperemia in response to airway
in the airway among healthy smokers. This remains to be
cooling and increased airway liquid and interstitial osmolar-
investigated.
ity caused by water loss have been proposed to have a criti-
cal role [63, 64]. The former may narrow the airway while
the latter may promote mediator release from inammatory
cells. However, while hyperventilation with frigid air, a sub-
FUNCTIONAL CONSEQUENCES stitute for exercise-related hyperventilation, has been shown
to cause a marked increase in Qaw in healthy subjects [81],
this may not be the case in asthmatics, who have a blunted
The vascular changes described above could participate in vasodilator responsiveness. This remains to be studied.
the physiological and clinical manifestations of asthma and
COPD; however, this is still a matter of debate.
Mucociliary clearance
Mucosal thickness
The role of airway blood ow in supporting the mucociliary
As vessels are known to occupy a signicant portion of the apparatus is incompletely understood. Mucociliary clear-
inner airway wall, it is possible that the vascular compo- ance was impaired in the immediate postoperative period
nent of inammation increases mucosal thickness enough after lung transplantation [65], and clearance of inhaled
to increase airow resistance. Airway vascular engorgement, particles was signicantly impaired when bronchial blood
submucosal edema, and luminal uid accumulation have all ow was stopped in sheep [66]. The eect of increased air-
been proposed to contribute to the excessive airway narrow- way blood ow on mucociliary clearance in asthma has not
ing and enhanced airway responsiveness in asthma. Several been examined.
animal and human studies have examined this experimen-
tally; the results have been conicting with respect to the
eects of vascular congestion on airway caliber [5559].
The observation that rapid infusion of intravenous uids PHARMACOLOGIC INTERVENTIONS
causes airway obstruction in humans supports this theory
[57]. However, another study has not been able to conrm
this nding [58, 59]. There is less controversy on the eect In asthma and COPD, the dearth of physiological data on
of airway edema on airway caliber: edema of the inner the pathogenetic role of the airway circulation precludes the
wall internal to the smooth muscle contributes to airway formulation of meaningful therapeutic recommendations.
hyperresponsiveness as the same degree of muscle shorten- An exception may be the reversal of microvascular hyper-
ing causes greater luminal narrowing than in the normal permeability, an intervention that is likely to increase air-
airway [60]. way patency and decrease airow resistance. Although such
an eect is dicult to demonstrate experimentally, anti-
inammatory agents, especially inhaled glucocorticoster-
Drug, mediator, and cell transport oids can be expected to reduce airway edema by inhibiting
microvascular hyperpermeability based on observation in
Inammation-associated blood ow, permeability, and extrapulmonary vascular beds [36, 37, 67]. In contrast, the
interstitial barrier changes could alter transport functions physiological and clinical consequences of the increased
of the airway vasculature as suggested by previous studies. Qaw and the blunted vasodilator responsiveness (e.g. to
Desmopressin uptake by the nasal mucosa was enhanced 2-a-drenergic agonists) of the airway circulation in asthma

245
Asthma and COPD: Basic Mechanisms and Clinical Management

and COPD are not known. These vascular changes could 80


contribute to the pathogenesis of the disease or constitute a

Qaw (l * min1 * ml1)


vascular adaptation that may counteract the eects of airway 70
inammation on airway function. Likewise, the blunted vasodi- *
* *
lator response in the airways of patients with COPD may be 60
contributing to or mitigate the airway disease of COPD. *
*
It therefore might be more useful to focus on the 50 * * *
eects of adrenergic agonists and glucocorticosteroids on * *
Qaw because these agents remain the mainstay of pharma- 40 *
cotherapy in obstructive lung disease.
30
Adrenergic agonists 0 30 60 90 120
Time (min)
Inhaled methoxamine, an -adrenergic agonist, causes
a dose-dependent transient decrease in Qaw, with an FIG. 19.5 Airway blood flow (Qaw) before and after the inhalation of
enhanced responsiveness in asthmatics compared to healthy fluticasone (), beclomethasone (o) and budesonide () in asthmatic
subjects (n 10). Data are presented as mean SD. * p  0.05 versus
subjects (Fig. 19.3) [8]. Higher doses of methoxamine cause
baseline value. Reproduced with from Ref. [10].
bronchoconstriction in asthmatics. However, the vasocon-
strictor eect is already seen at methoxamine doses that do
not cause bronchoconstriction or have systemic side eects.
Thus combining an inhaled 2-adrenergic agonist or gluco- of the acute vasoconstrictor action of the drug, presumably
corticosteroid with a low dose of an -adrenergic agonist through a genomic action (Fig. 19.4) [42]. A similar eect is
might increase residence of the former drugs in the airway seen with oral montelukast, suggesting that Qaw is a marker
tissue and enhance and prolong their biological eects. of airway inammation in asthma [69]. The observed res-
The vasodilator eect of inhaled albuterol, a 2- toration of a nearly normal Qaw by long-term glucocorti-
adrenergic agonist, is blunted in asthmatics and patients with costeroid treatment may reect a reversal of inammatory
COPD (Fig. 19.3) [8, 9, 30, 34]. In as much as vasodilation vasodilation and/or an eect on vascular remodeling. For
may be an undesirable side eect of inhaled 2-adrenergics instance, inhaled beclomethasone (2001500 mg daily) has
when administered for bronchodilation, the vascular hypore- been reported to decrease the subepithelial area occupied by
sponsiveness to this drug in asthma and COPD could be vessels in asthmatics [70]. The underlying mechanism is not
considered a therapeutic advantage. Unfortunately, inhaled known, but glucocorticosteroids have been reported to reduce
glucocorticosteroids that are typically co-administered with vascular endothelial growth factor expression in pulmonary
2-adrenergic agonists restore vasodilator responsiveness to vascular smooth muscle cells [71].
albuterol [42]. This eect is seen after long-term glucocorti-
costeroid treatment and after pretreatment with a single dose Restoration of 2-adrenergic vasodilation
of a glucocorticosteroid, suggesting that both genomic and In patients with asthma and COPD in whom inhaled
non-genomic glucocorticosteroid actions are involved [34]. albuterol fails to increase Qaw, inhaled glucocorticosteroid
therapy restores normal albuterol responsiveness (Fig. 19.4)
Glucocorticosteroids [29, 42]. Inasmuch as 2-adrenergic vasodilation is mediated
by the endothelium, these conditions may be associated with
There is increasing evidence that glucocorticosteroids have a defect in endothelial function, and glucocorticosteroids may
profound eects on the airway vasculature. Among other turn out to have a role in the treatment of endothelial dys-
actions, such as reduction of microvascular hyperpermeabil- function. Given the prevalence of cardiovascular disease in
ity, glucocorticosteroids regulate Qaw. COPD and its relation to endothelial dysfunction [45, 46],
there is a need to examine the eects of glucocorticosteroids
on endothelial function in the extrapulmonary systemic cir-
Acute vasoconstriction
culation as well. Data base studies indeed have suggested that
Inhaled glucocorticosteroids decrease Qaw rapidly and inhaled glucocorticoteroids have a benecial eect on cardio-
transiently by a non-genomic action that is mediated by the vascular morbidity and mortality in COPD [7274].
noradrenergic nervous system as the eect can be blocked
with an oral 1-adrenoceptor blocking agent. Thus a sin-
gle dose of inhaled uticasone (880 and 1760 mg) causes Glucocorticosteroid2-adrenergic
a decrease in Qaw within 30 min, with a return toward interaction
baseline by 90 min in asthmatics and healthy subjects [68].
Other inhaled glucocorticosteroids such as bleclomethasone These stress hormones have been shown to potentiate each
and budesonide have similar eects (Fig. 19.5) [10]. others eect in target tissues including the airway. It has been
known for some time that through a genomic action, gluco-
Reversal of increased Qaw in asthma corticosteroids can upregulate the synthesis and G-protein
Treatment with inhaled uticasone (440 mg daily) for 2 weeks coupling of 2-adrenergic receptors, thereby enhancing the
reverses the asthma-associated increase of Qaw independent tissue responsiveness to 2-adrenergic agonists [75, 76].

246
Tracheobronchial Circulation 19
In addition, glucocorticosteroids have acute, non- in the pig: Evidence for non-adrenergic mechanisms involving neu-
genomic actions in the airway that can potentiate the eects ropeptide Y. Acta Physiol Scand 155(2): 193204, 1995.
of 2-adrenergic agonists as shown above. Inhaled 2- 14. Laitinen LA, Laitinen MA, Widdicombe JG. Dose-related eects of
pharmacological mediators on tracheal vascular resistance in dogs. Br J
adrenergic agonists are cleared from the airway tissue by local
Pharmacol 92: 7039, 1987.
metabolism and vascular clearance. Glucocorticosteroids 15. Barker JA, Chediak AD, Baier HJ et al. Tracheal mucosal blood ow
inhibit within minutes the uptake of organic cations includ- responses to autonomic agonists. J Appl Physiol 65(2): 82934, 1998.
ing adrenergic agonists into non-neuronal cells containing 16. Onorato DJ, Demirozu MC, Breitenbcher A et al. Airway mucosal
the agonists metabolizing enzymes [7780]. This could blood ow in humans. Response to adrenergic agonists. Am J Respir
increase the concentration of adrenergic agonists at adren- Crit Care Med 149(5): 113237, 1994.
ergic receptor sites in airway tissue, thereby potentiating 17. Charan NB, Carvalho P, Johnson SR et al. Eect of aerosolized
the agonists eects on airway and airway vascular smooth acetylcholine on bronchial blood ow. J Appl Physiol 85(2): 43236,
muscle. The latter eect has now been demonstrated in vivo 1998.
in patients with asthma in whom pretreatment with a sin- 18. Carvalho P, Johnson SR, Charan NB. Non-cAMP-mediated bronchial
arterial vasodilation in response to inhaled -agonists. J Appl Physiol
gle glucocorticosteroid dose potentiated the vasodilator
84(1): 21521, 1998.
response to albuterol within minutes [34]. In addition, the 19. Carvalho P, Thompson WH, Charan NB. Comparative eects of
acute vasoconstrictor action of inhaled glucocorticosteroids -receptor stimulation and nitrergic inhibition on bronchovascular tone.
could reduce the vascular clearance thereby further enhanc- J Appl Physiol 88: 168589, 2000.
ing the biological eect of inhaled 2-adrenergic agonists. 20. Wanner A. Circulation of the airway mucosa. J Appl Physiol 67(3):
Although these eects thus far have not been studied with 91725, 1989.
long-acting 2-adrenergic agonists, the current practice 21. Erjefalt I, Persson CGA. Eects of adrenaline and terbutaline on medi-
of treating obstructive lung disease with long-acting 2- ator-increased vascular permeability in the cat trachea. Br J Pharmacol
adrenergic agonist-glucocorticosteroid combination formu- 77: 399, 1982.
lations is supported by the above shown rapid interaction 22. Kuwano K, Bosken CH, Par PD. Small airways dimensions in asthma
and in chronic obstructive pulmonary disease. Am Rev Respir Dis 148:
between the two classes of drugs.
122025, 1993.
23. Green FHY, Butt JC, James AL, Carroll NG. Abnormalities of the
bronchial arteries in asthma. Chest 130: 102533, 2006.
References 24. Li X, Wilson JW. Increased vascularity of the bronchial mucosa in mild
asthma. Am J Respir Crit Care Med 156(1): 22933, 1997.
1. Bernard SL, Glenny RW, Polissar NL et al. Distribution of pulmo- 25. Bousquet J, Jeery PK, Busse WW et al. Asthma. From bronchocon-
nary and bronchial blood supply to airways measured by uorescent striction to airways inammation and remodeling. Am J Respir Crit
microspheres. J Appl Physiol 80(2): 43036, 1996. Care Med 161: 172045, 2000.
2. Charan NB, Carvalho PG. Anatomy of the normal bronchial circu- 26. Simcock DE, Kanabar V, Clarke GW, OConnor BJ, Lee TH, Hirst
latory system in humans and animals. In: Butler J (ed.), The bronchial SJ. Proangiogenic activity in bronchoalveolar lavage uid from patients
circulation, pp. 4577. New York: Marcel Dekker, 1992. with asthma. Am J Respir Crit Care Med 176: 14653, 2007.
3. Baile EM, Par PD, Ernest D . Distribution of blood ow and 27. Kanazawa H, Nomura S, Asai K. Roles of angiopoetin-1 and angiopoetin-
neutrophil kinetics in bronchial vasculature of sheep. J Appl Physiol 2 on airway vascular permeability in asthmatic patients. Chest 131: 1035
82(5): 146671, 1997. 41, 2007.
4. Wagner EM, Mitzner W, Brown RH. Site of functional bronchopul- 28. Barbato A, Turato G, Baraldo S, Bazzan E, Calabrese F, Panizzolo C,
monary anastomoses in sheep. Anat Rec 254: 36066, 1999. Zanin MA, Zuin R, Maestrelli P, Fabbri LM, Saetta M. Epithelial
5. Mariassy AT, Gazeroglu H, Wanner A. Morphometry of the subepi- damage and angiogenesis in the airways of children with asthma. Am J
thelial circulation in sheep airways. Am Rev Respir Dis 143: 16266, Respir Crit Care Med 174: 97581, 2006.
1991. 29. Mendes ES, Campos M, Wanner A. Airway blood ow reactivity in
6. Wanner A, Chediak AD, Csete ME. Airway mucosal blood ow: healthy smokers and in ex-smokers with or without COPD. Chest 129:
Response to autonomic and inammatory stimuli. Eur Respir J 89398, 2006.
12: 618s623s, 1990. 30. Paredi P, Kharitonov SA, Barnes PJ. Correlation of exhaled breath tem-
7. Baile EM, Pare PD. Methods of measuring bronchial blood ow. In: perature with bronchial blood ow in asthma. Respir Res 6: 1521, 2005.
Butler J (ed.), The Bronchial Circulation, pp. 10196. New York: Marcel 31. Pisarri TE, Zimmerman MP, Adrian TE et al. Bronchial vasodilator
Dekker, 1992. pathways in the vagus nerve of dogs. J Appl Physiol 86(1): 10513, 1999.
8. Brieva J, Wanner A. Adrenergic airway vascular smooth muscle respon- 32. Mattoli S, Soloperto M, Marini M, Fasoli A. Levels of endothelin in the
siveness in healthy and asthmatic subjects. J Appl Physiol 90: 66569, bronchoalveolar lavage uid of patients with symptomatic asthma and
2001. reversible airow obstruction. J Allergy Clin Immunol 88(3 Pt 1): 37684,
9. Kumar SD, Emery MJ, Atkins ND et al. Airway mucosal blood ow in 1991.
bronchial asthma. Am J Respir Crit Care Med 158: 15356, 1998. 33. Wagner EM. TNF- induced bronchial vasoconstriction. Am J Physiol
10. Mendes ES, Pereira A, Danta I et al. Comparative bronchial vasocon- Heart Circ Physiol 279: H946H951, 2000.
strictive ecacy of inhaled glucocorticosteroids. Eur Respir J 21: 989 34. Mendes ES, Campos MA, Wanner A. Acute eect of a glucocorticos-
93, 2003. teroid on beta-adrenergic airway and airway smooth muscle reactivity
11. Widdicombe JG, Webber SE. Neuroregulation and pharmacology of in patients with mild asthma. J Allergy Clin Immunol 121: 7004, 2008.
the tracheobronchial circulation. In: Butler J (ed.), The bronchial circula- 35. Erjefalt I, Persson CGA. Eects of adrenaline and terbutaline on medi-
tion, pp. 24989. New York: Marcel Dekker, 1992. ator-increased vascular permeability in the cat trachea. Br J Pharmacol
12. Canning BJ, Fischer A. Localization of cholinergic nerves in lower 77: 399408, 1982.
airways of guinea pigs using antisera to choline acetyltransferase. Am J 36. Cudkowicz L, Armstrong JB. The Bronchial arteries in pulmonary
Physiol 272: L73138, 1997. emphysema. Thorax 8: 4658, 1953.
13. Franco-Cereceda A, Matran R, Alving K et al. Sympathetic vascular 37. Liebow AA. The bronchopulmonary venous collateral circulation with
control of the laryngeo-tracheal, bronchial and pulmonary circulation special reference to emphysema. Am J Pathol 29: 25163, 1953.

247
Asthma and COPD: Basic Mechanisms and Clinical Management

38. Paredi P, Ward S, Cramer D, Barnes P, Kharitonov SA. Normal bron- 60. James AL, Par PD, Hogg JC. The mechanics of airway narrowing in
chial blood ow in COPD is unaected by inhaled corticosteroids and asthma. Am Rev Respir Dis 139: 24246, 1989.
correlates with exhaled nitric oxide. Chest 131: 107581, 2007. 61. Olano LS, Titus CR, Shea MS et al. Eect of intranasal histamine on
39. Liebow AA, Hales MR, Lindskog GE. Enlargement of the bronchial nasal mucosal blood ow and the antidiuretic activity of desmopressin.
arteries, and their anastomoses with the pulmonary arteries in bron- J Clin Invest 80(3): 89095, 1987.
chiectasis. Am J Pathol 25: 21131, 1949. 62. Kelly L, Kolbe J, Mitzner W et al. Bronchial blood ow aects recovery from
40. Fishman AP. The clinical signicance of the pulmonary collateral cir- constriction in dog lung periphery. J Appl Physiol 60(6): 195459, 1986.
culation. Circulation 24: 67790, 1961. 63. Csete ME, Chediak AD, Abraham WM et al. Airway blood ow
41. Charan NB, Turk GM, Dhand R. The role of bronchial circulation in modies allergic airway smooth muscle contraction. Am Rev Respir Dis
lung abscess. Am Rev Respir Dis 131: 12124, 1985. 144(1): 5963, 1991.
42. Brieva JL, Danta I, Wanner A. Eect of inhaled an glucocorticosteroid 64. McFadden ER. Hypothesis: Exercise-induced asthma as a vascular
on airway mucosal blood ow in mild asthma. Am J Respir Crit Care phenomenom. Lancet 335: 88083, 1990.
Med 161: 29396, 2000. 65. Paul A, Marelli D, Shennib H et al. Mucociliary function in autotrans-
43. Schindler C et al. Mechanisms of beta-adrenergic receptor-mediated planted, allotransplanted, and sleve resected lungs. J Thorac Cardiovasc
vasodilation. Clin Pharmacol Ther 75: 4959, 2004. Surg 98: 52328, 1989.
44. Zeiher AM et al. Long-term cigarette smoking impairs endothelium- 66. Wagner EM, Foster WM. Importance of airway blood ow on particle
dependent coronary arterial vasodilator function. Circulation 92: clearance from the lung. J Appl Physiol 81(5): 187883, 1996.
1094100, 1995. 67. Hashimoto M, Tanaka H, Shoshaku A. Quantitative analysis of bron-
45. Barr RG, Meisa-Vela S, Austin JHM, Basner RC, Keller BM, chial wall vascularity in the medium and small airways of patients with
Reeves AP, Shimbo D, Stevenson L. Impaired ow-mediated dilation asthma and COPD. Chest 127: 96572, 2005.
is associated with low pulmonary function and emphysema in ex-smokers. 68. Kumar SD, Brieva JL, Danta I et al. Transient eect of inhaled u-
Am J Respir Crit Care Med 176: 12007, 2007. ticasone on airway mucosal blood ow in subjects with and without
46. McAllister DA, Maclay JD, Mills NL, Mair G, Miller J, Anderson D, asthma. Am J Respir Crit Care Med 161: 91821, 2000.
Newby DE, Murchison JT, McNee W. Arteial stiness is independently 69. Mendes ES, Campos MA, Hurtado A, Wanner A. Eect of monte-
associated with emphysema severity in patients with chronic obstructive lukast and uticasone propionate on airway mucosal blood ow in
pulmonary disease. Am J Respir Crit Care Med 176: 120814, 2007. asthma. Am J Respir Crit Care Med 169: 113134, 2004.
47. Czernin J, Waldherr C. Cigarette smoking and coronary blood ow. 70. Orsida BE, Li X, Hickey B et al. Vascularity in asthmatic airways:
Prog Cardiovasc Dis 45: 395404, 2003. Relation to inhaled steroid dose. Thorax 54: 28995, 1999.
48. Yamamoto Y, Nishiyama Y, Monden T, Satoh K, Ohkawa M. A study 71. Nauck M, Roth M, Tamm M et al. Induction of vascular endothelial growth
of the acute eect of smoking on cerebral blood ow using 99mTc- factor by platelet-activating factor and platelet-derived growth factor is down-
ECD SPET. Eur J Nucl Med Mol Imaging 30: 61214, 2003. regulated by corticosteroids. Am J Respir Cell Mol Biol 16: 398406, 1997.
49. Monfrecola G, Riccio G, Savarese C, Posteraro G, Procaccini EM. The 72. Huiart L, Ernst P, Ranouil X, Suissa S. Low-dose inhaled corticoster-
acute eect of smoking on cutaneous microcirculation blood ow in oids and the risk of acute myocardial infarction in COPD. Eur Respir J
habitual smokers and nonsmokers. Dermatology 197: 11518, 1998. 25: 63439, 2005.
50. Alving K, Fornhem C, Weitzberg E, Lundberg JM. Nitric oxide medi- 73. Macie C, Wooldrage K, Manfreda J, Anthonisen NR. Inhaled corti-
ates cigarette smoke-induced vasodilatory responses in the lung. Acta costeroids and mortality in COPD. Chest 130: 64046, 2006.
Physiol Scand 146: 4078, 1992. 74. Man SFP, Sin DD. Eects of corticosteroids on systemic inammation in
51. Alving K, Fornhem C, Lundberg JM. Pulmonary eects of endogene- chronic obstructive pulmonary disease. Proc Am Thor Soc 2: 7882, 2005.
ous and exogeneous nitric oxide in the pig: relation to cigarette smoke 75. Mak JC, Nishikawa M, Barnes PJ. Glucocorticosteroids increase beta-
inhalation. Br J Pharmacol 110: 73946, 1993. 2 adrenergic receptor transcription in human lung. Am J Physiol 268:
52. Benowitz NL. Cigarette smoking and cardiovascular disease: L41l46, 1995.
Pathophysiology and implications for treatment. Prog Cardiovasc Dis 76. Adcock IM, Stevens DA, Barnes PJ. Interactions of glucocorticoids
46: 91111, 2003. and 2- agonists. Eur Respir J 9: 16068, 1996.
53. Vleeming W, Rambali B, Opperhuizen A. The role of nitric oxide in 77. Horvath G, Wanner A. Inhaled corticosteroids: Eects on the airway
cigarette smoking and nicotine addiction. Nicotine Tob Res 4: 34148, vasculature in bronchial asthma. Eur J Respir Dis 27: 17287, 2006.
2002. 78. Horvath G, Sutto Z, Torbati A, Conner GE, Salathe M, Wanner A.
54. Kanwaldeep R, Mendes E, Wanner A. Acute eect of cigarette smoke Norepinephrine transport by the extraneuronal monoamine transporter
and nicotine on airway blood ow and airow in healthy smokers. in human bronchial arterial smooth muscle cells. Am J Physiol Lung Cell
Lung 184: 36368, 2006. Mol Physiol 285: L82937, 2003.
55. Cabanes LR, Weber SN, Matran R et al. Bronchial hyperresponsive- 79. Horvath G, Lieb T, Conner GE, Salath M, Wanner A. Steroid sensi-
ness to methacholine in patients with impaired left ventricular func- tivity of norepinephrine uptake by human bronchial arterial and rabbit
tion. N Engl J Med 320(20): 131748, 1989. aortic smooth muscle cells. Am J Respir Cell Mol Biol 25(4): 5006, 2001.
56. Csete ME, Abraham WM, Wanner A. Vasomotion inuences airow 80. Horvath G, Mendes ES, Schmid N, Schmid A, Conner GE, Salathe M,
in peripheral airways. Am Rev Respir Dis 141(6): 140913, 1990. Wanner A. The eect of corticosteroids on the disposal of long-acting
57. Gilbert IA, Winslow CJ, Lenner KA et al. Vascular volume expansion beta2-agonists by airway smooth muscle cells. J Allergy Clin Immunol
and thermally induced asthma. Eur Respir J 6(2): 18997, 1993. 120: 11039, 2007.
58. Blosser S, Mitzner W, Wagner EM. Eects of increased bronchial 81. Kim HH, LeMerre C, Demirozu CM et al. Eect of hyperventilation
blood ow on airway morphometry, resistance, and reactivity. J Appl on airway mucosal blood ow in normal subjects. Am J Respir Crit Care
Physiol 76(4): 162429, 1994. Med 154(5): 156366, 1996.
59. Tang GJ, Freed AN. The role of submucosal oedema in increased 82. Je rey P. Remodeling and inammation of bronchi in asthma and
peripherial airway resistance by intravenous volume loading in dogs. chronic obstructive pulmonary disease. Proc Am Thor Soc 1: 17683,
Eur Respir J 7: 31117, 1994. 2004.

248
Pulmonary Vessels
20 CHAPTER

Roberto Rodrguez-
Roisin and Joan Albert
CHRONIC OBSTRUCTIVE In patients with end-stage COPD and
pulmonary hypertension or cor pulmonale, post- Barber
PULMONARY DISEASE mortem studies have shown deposition of longi- Servei de Pneumologia, Hospital Clnic,
tudinal muscle, brosis, and elastosis that cause IDIBAPS, Universitat de Barcelona,
intimal enlargement in pulmonary muscular
Introduction arteries [1, 2]. In the arterioles there is a devel-
Barcelona, Spain
opment of a medial coat of circular smooth
Pulmonary vascular circulation abnormalities are muscle, bounded by a new elastic lamina, with
common in chronic obstructive pulmonary disease deposition of longitudinal muscle and brosis of
(COPD). It is likely that, pulmonary hyperten- the intima. As compared with patients with mild
sion associated with COPD is the most frequent COPD and healthy nonsmokers, Santos et al. [3]
form of pulmonary hypertension. Concepts gath- showed substantial enlargement of the intima in
ered in the systemic circulation have contributed lung tissue specimens obtained from lung vol-
to a better understanding of changes occurring in ume reduction surgery. Conversely, medial thick-
pulmonary vessels. This has promoted a change ness was slightly reduced in patients with severe
in the notion of the pathogenesis of pulmonary emphysema when compared with patients with
hypertension from a vasoconstrictive phenom- milder form of COPD.
enon to a cell proliferative disorder. Some of the In patients with mild-to-moderate COPD,
concepts used to explain the pathogenesis of the morphometric studies of pulmonary muscular
most severe forms of pulmonary hypertension arteries have shown enlargement of the intimal
have been extrapolated to COPD, yielding to layer with luminal reduction [47] (Fig. 20.1).
new approaches to its pathobiology. In this sec- Intimal enlargement occurs in muscular arteries
tion, we will review the current knowledge on the of dierent sizes, although it is more pronounced
mechanisms of pulmonary vascular changes asso- in small arteries with an external diameter less
ciated with COPD. than 500 m [5]. In addition, the number of
small pulmonary muscular arteries, with a diam-
Lung structure eter 200 m, is increased [8], indicating mus-
cularization of arterioles. Intimal hyperplasia
Pulmonary vascular remodeling is broadly results from proliferation of poorly dierentiated
dened as an active process of profound struc- smooth muscle cells and deposition of elastic
tural changes reecting an abnormal tissue repair and collagen bers [9]. Changes in the media
response to an acute and/or chronic insult. In of muscular arteries are less conspicuous and the
COPD, the latter process results from profound majority of morphometric studies have failed to
brocellular changes that lead to an enlargement show dierences in the thickness of the muscu-
of the vessel wall an active process that aects lar layer when comparing patients with mild-
preferentially small and pre-capillary arteries of to-moderate COPD with control subjects [5].
the pulmonary vasculature identied at dierent Moreover, the magnitude of this intimal thick-
degrees of COPD severity. ening, not dierent from that shown by mild

249
Asthma and COPD: Basic Mechanisms and Clinical Management

thus 25% of the patients already having pulmonary hyper-


tension at rest. The incidence of resting pulmonary hyper-
tension was greater in the subset of patients who initially
developed pulmonary hypertension during exercise (32%),
as compared with those without exercise-induced pulmo-
nary hypertension (16%), such that increased PAP during
exercise was an independent predictor for the subsequent
development of pulmonary hypertension [13]. These results
indicate that in COPD pulmonary circulation, derange-
ment can be present years before pulmonary hypertension
is apparent at rest. Further, this study suggests that exercise
tests might be of interest to demonstrate pulmonary circula-
tion disturbances, which are consistent with morphometric
studies that show conspicuous structural changes in pulmo-
nary muscular arteries in mild COPD patients [4, 6].
FIG. 20.1 Photomicrograph of a pulmonary muscular artery from a Pulmonary hypertension is a common complica-
patient with COPD showing prominent intimal hyperplasia and lumenal tion of COPD. Its presence is associated with increased
narrowing. Immunostaining with antibody against -smooth muscle actin mortality, more frequent exacerbations, and greater use of
reveals marked proliferation of smooth muscle cells in the intima.
health-care resources. At present, there is no specic and
eective treatment for this condition in COPD. However,
COPD patients, is also present in heavy smokers with nor- recent advances in the understanding of the pathobiology
mal spirometry [6, 7]. of pulmonary hypertensive states, along with the develop-
ment of eective treatments for pulmonary arterial hyper-
tension, have opened a new perspective that might be
Clinical background relevant in COPD. Nevertheless, the actual prevalence
of pulmonary hypertension in COPD remains elusive,
In COPD pulmonary hypertension is considered to be because it has not been assessed systematically using right
present when mean pulmonary artery pressure (PAP) heart catheterization across the wide clinical spectrum of
exceeds the upper normal limit of 20 mmHg [10]. In gen- COPD. Hemodynamic data currently available are limited
eral, the degree of pulmonary hypertension in COPD is to patients with advanced disease.
of low-to-moderate magnitude and rarely exceeds 35 Three studies have provided data in a large series of
40 mmHg. Both right atrial pressure and pulmonary capil- patients. The rst study is of patients with severe emphy-
lary wedge pressure tend to be normal, as well as the cardiac sema screened for lung volume reduction surgery and the
output [10], a pattern at variance with other pulmonary incidence of pulmonary hypertension (PAP  20 mmHg)
hypertensive conditions (i.e. idiopathic pulmonary arterial was very high (91%), although of mild-to-moderate magni-
hypertension, congenital heart disease, thromboembolic dis- tude (2035 mmHg); [13] in 5% of patients, however, PAP
ease) in which PAP can reach extremely high levels, close to values exceeded 35 mmHg. Although there were signicant
those of the systemic circulation, usually in the context of a but modest correlations between PAP and FEV1 and arte-
reduced cardiac output. rial PO2, PAP was more closely related to pulmonary artery
Pulmonary hypertension in COPD evolves over time occlusion pressure, slightly increased in most of the patients,
and its severity correlates with the degree of airow obstruc- suggesting that gas trapping may contribute to this increased
tion and the impairment of pulmonary gas exchange [11, capillary pressure. The second study is a retrospective analysis
12]. However, the rate of progression of pulmonary hyper- of over 1000 COPD patients. The prevalence of severe pul-
tension in COPD is slow and usually PAP is only moder- monary hypertension (PAP  40 mmHg) was 1.1% [14].
ately elevated, even in patients with advanced stages [11]. This cohort of patients with disproportionate pulmonary
Weitzenblum et al. [11] studied the natural history of PAP hypertension had moderate airow limitation, severe hypox-
in a cohort of over 100 COPD patients followed up to 5 emia, hypercapnia, and very low diusing capacity, indicating
years with PAP increases on an average of 0.6 mmHg/year. that these patients may share some characteristics of idio-
Although the rate of increase of PAP was slightly higher pathic pulmonary arterial hypertension. In a third study with
in patients without pulmonary hypertension at the onset of approximately 200 COPD patients who were candidates
the follow-up compared with those who already had pul- for lung volume reduction surgery or lung transplantation,
monary hypertension, the PAP increase was more closely [15] pulmonary hypertension (i.e. PAP  25 mmHg) was
related to the progress of gas exchange disturbances than to observed in 50% of the patients, although of mild severity
the initial PAP values [11]. (2635 mmHg). In 10% of the patients, pulmonary hyperten-
The beginning of pulmonary hypertension in the nat- sion was moderate (3645 mmHg) and, in 4% of the patients,
ural history of COPD has intrigued investigators. Kessler severe (45 mmHg). A cluster analysis identied a subset of
et al. [13] assessed the outcome of pulmonary hemodynam- patients characterized by moderate impairment of airway
ics in patients with moderate COPD without pulmonary function and high PAP values, along with severe hypoxemia,
hypertension at rest and observed 7 years later that PAP supporting the concept of the existence of a cohort of COPD
had increased by 2.6 mmHg (annual rise, 0.4 mmHg/year), patients with disproportionate pulmonary hypertension.

250
Pulmonary Vessels 20
Pathophysiology concept of cor pulmonale. The suggestion has been made that
this term should be abandoned in favor of a more precise
Hypoxic pulmonary vasoconstriction, right ventricular denition based on objective evidence of right ventricular
function, peripheral edema, and exercise are four key factors hypertrophy, enlargement, functional abnormality, or failure.
inuencing the development of pulmonary hypertension. Although peripheral edema may be a sign of venous
Pulmonary vasoconstriction in response to hypoxia congestion due to upstream transmission of right ventricular
reduces perfusion in poorly ventilated or nonventilated lling pressures, in advanced COPD edema is more related
lung units and diverts blood ow to better-ventilated units, to hypercapnia rather than to raised jugular pressures [27,
thereby restoring ventilationperfusion (VA/Q) imbalance 28]. Some patients may present peripheral edema without
toward normal and increasing PaO2. Teleologically, hypoxic hemodynamic signs of right heart failure or PAP changes
vasoconstriction plays a fundamental role in matching pul- [29, 30]. This has lead to reconsideration of the mechanisms
monary blood ow to alveolar ventilation. Failure to main- for peripheral edema formation in COPD [29, 3032].
tain this balance, and the consequent VA/Q mismatching, In COPD, peripheral edema results from a complex
is the major cause for both hypoxemia and hypercapnia in interaction between the hemodynamic changes and the bal-
COPD [16]. Studies using inert gases [17] have shown that ance between edema promoting and protective factors. In
hypoxic pulmonary vasoconstriction release while breathing COPD patients with pulmonary hypertension and coexist-
high oxygen concentration worsens the underlying VA/Q ing chronic respiratory failure, hypoxemia and hypercap-
imbalance in patients with dierent degrees of COPD nia aggravate venous congestion by further activating the
severity [16]. In general terms, the contribution of hypoxic sympathetic nervous system, which is already stimulated
vasoconstriction to correct or restore VA/Q matching tends by right atrial distension. Sympathetic activation decreases
to be greater in patients with less severe COPD [18]; con- renal plasma ow, stimulates the reninangiotensinaldos-
versely, hypoxic vasoconstriction remains less inuential terone system and promotes tubular reabsorption of
in patients with severe structural derangement of pulmo- bicarbonate, sodium, and water. Vasopressin, which also
nary muscular arteries [4]. Furthermore, the magnitude of contributes to edema formation, is released when patients
contraction in isolated pulmonary artery rings induced by become hyponatremic and its plasma levels rise in patients
a hypoxic stimulus is negatively correlated to endothelial with hypoxemia and hypercapnia [31].
function and directly related to the arterial PO2 [19], sug- Atrial natriuretic peptide is released from distended
gesting that the vascular dysfunction is associated with atrial walls and may act as an edema-protective mechanism
an altered response to hypoxia that further worsens gas since it has vasodilator, diuretic, and natriuretic properties.
exchange. Accordingly, the contribution of hypoxic vaso- Nevertheless, these eects are usually insucient to coun-
constriction to gas exchange in COPD should be taken into terbalance the edema-promoting mechanisms. Peripheral
account when administering vasodilators that might poten- edema may develop or worsen during episodes of COPD
tially inhibit such a response since they may also impair gas exacerbation. Weitzenblum et al. [26] identied a sub-
exchange [17]. group of patients with more marked peripheral edema that
In patients with COPD and pulmonary hyperten- was attributed to hemodynamic signs of right heart failure
sion, the PAP is not markedly elevated and the rate of pro- (increase in end-diastolic pressure). Compared with patients
gression of pulmonary hypertension is slow. Therefore, the with normal end-diastolic pressure, patients with right heart
right ventricle has time to adapt to the modest increase failure had more marked increase in PAP, and more severe
in afterload. When PAP is chronically elevated the right hypoxemia and hypercapnia. This suggests that worsening
ventricle dilates and both end-diastolic and end-systolic of pulmonary hypertension during exacerbations contrib-
volumes increase. The stroke volume of right ventricle is utes to edema formation.
usually maintained, whereas the ejection fraction decreases. Physical exercise produces abnormal increases in
Subsequent hypertrophy of the right ventricular wall in PAP, especially in patients who have pulmonary hyperten-
persistent pulmonary hypertension reduces its tension and sion at rest [33]. As noted above, patients that appear to
hence the afterload. The reduction in right ventricular ejec- be more prone to the development of pulmonary hyper-
tion fraction (RVEF) is inversely related to PAP; nonethe- tension may show an abnormal increase in PAP during
less, a decrease in RVEF does not imply that there is a true exercise, years before pulmonary hypertension is apparent
ventricular dysfunction [20]. Measurements of end-systolic at rest [13]. Dierent studies have identied a number of
pressurevolume relationships have shown that a stable mechanisms for exercise-induced pulmonary hypertension
COPD contractility of the right ventricle lies within nor- in COPD, including hypoxic vasoconstriction, reduction
mal limits, irrespective of the PAP values [21, 22]. However, of the capillary bed by emphysema, extramural compres-
during COPD exacerbations, when PAP increases mark- sion by increased alveolar pressure, or impaired release of
edly, the contractility of the right ventricle is reduced in endothelium-derived relaxing factors [3436]. These mech-
patients presenting clinical signs of right heart failure [23, anisms may coexist hence several factors may contribute to
24]. Cardiac output in COPD is usually preserved and the development of exercise-induced pulmonary hyperten-
might rise during exacerbations [25, 26], even when right sion in a given individual. In COPD, PAP during exercise
heart failure is apparent. Therefore, the usual denition of is greater than predicted by the PVR equation, suggesting
heart failure as a reduction in cardiac output does not apply active pulmonary vasoconstriction on exertion [34]. The lat-
to COPD. These questions relating to the nature of right ter may be due to an enhancement of hypoxic vasoconstric-
heart failure in COPD have led to a reassessment of the tion by decreased mixed venous PO2, increased tone of the

251
Asthma and COPD: Basic Mechanisms and Clinical Management

FIG. 20.2 Immunodetection of


endothelial nitric oxide synthase
(eNOS) in pulmonary muscular
arteries from a nonsmoker control
subject, a smoker with normal lung
function, a patient with moderate
COPD, and a patient with severe
emphysema. Whereas endothelial
cells of the nonsmoker subject show
strong immunoreactivity to eNOS, the
intensity of the signal is weak or absent
in pulmonary arteries of the smoker
subject and patients with COPD.

sympathetic nervous system, and/or decreased arterial pH pulmonary circulations. Vascular actions of endothelium are
[37]. During exercise, patients may develop dynamic hyper- mediated through the balanced release of potent vasoactive
ination due to expiratory ow limitation that results in mediators, such as NO, prostacyclin, endothelin-1 (ET-1),
increased alveolar pressure which is transmitted to the capil- and angiotensin. Some of these mediators are also potent
lary wedge pressure [38]. Furthermore, increased ventilation modulators of proliferative and inammatory responses.
during exercise, in the presence of airow limitation, results Endothelial dysfunction has been shown in pulmo-
in signicant swings of intrathoracic pressure that may nary arteries of both end-stage [44] and mild [7] COPD
reduce cardiac output by altering systemic venous return patients. Impairment of endothelial function results from
or by increasing left ventricular afterload [39]. Impairment changes in the expression and release of vasoactive media-
of endothelial release of vasorelaxing agents such as nitric tors. Endothelium-derived NO is a potent endogenous
oxide (NO) may also contribute to an impaired dilator vasodilator with anti-proliferative eects in the vessel walls.
response to increases in ow [40]. However, in a subset of In pulmonary arteries of COPD patients with pulmonary
COPD patients who developed pulmonary hypertension hypertension, the expression of endothelial NO synthase
during exercise, external administration of NO did not (eNOS) is reduced [45]. Interestingly, in smokers, eNOS
block the abnormal increase in PAP, suggesting that the expression is also reduced [46] (Fig. 20.2), likely explaining
defective endogenous NO release plays a marginal role in the endothelial dysfunction present in mild disease states
exercise-induced pulmonary hypertension [41]. Since pul- [7]. Prostacyclin, which is also synthesized by endothelial
monary hypertension may develop at moderate levels of cells, exerts similar actions to NO and we know that the
exercise, it has been suggested that repeated episodes of pul- expression of prostacyclin synthase is reduced in pulmo-
monary hypertension during daily activities, such as stairs nary arteries of patients with severe emphysema [47]. ET-1
climbing or walking, could contribute to the development is a potent vasoconstrictor that also exerts a mitogenic
of right ventricular hypertrophy [42]. eect on arterial smooth muscle cells, its expression being
augmented in pulmonary arteries of patients with COPD
and pulmonary hypertension, [48] but not in patients with
Pathobiology mild-to-moderate disease [46, 49]. These ndings indicate
that endothelial dysfunction, with changes in the expres-
The eld of vascular biology has made a major breakthrough sion and release of endothelium-derived vasoactive media-
over the last decades since the seminal studies identied tors that regulate cell growth, is a common characteristic
the key role of endothelium in the regulation of vascular nding of COPD. It may appear early in the natural his-
homeostasis [43]. Endothelial dysfunction is a common tory of the disease and could contribute to further changes
disturbance in hypertensive states of both the systemic and in vascular structure and function. Hypoxia, inammation,

252
Pulmonary Vessels 20
and cigarette smoke might all contribute to the origin of associated with intimal hyperplasia of pulmonary muscular
pulmonary endothelial damage in COPD. arteries [4]. Further, there is an increased number of CD8
Hypoxia is a potential mechanism that may explain T-lymphocytes inltrating the adventitia of pulmonary arter-
endothelial cell damage and vascular remodeling in COPD. ies in patients with mild-to-moderate COPD [7] (Fig. 20.3).
Hypoxia may downregulate eNOS expression and induce Inammatory cells are a source of cytokines and growth
smooth muscle cell and adventitial broblast prolifera- factors that may target the endothelial cells and contribute
tion [50, 51]. In addition, hypoxia elicits the contraction to the development of structural and functional abnormali-
of pulmonary arteries. However, the role of hypoxia as the ties of the vessel wall. Indeed, the number of inammatory
major etiological factor for pulmonary vascular impairment cells inltrating the pulmonary arteries is directly related to
in COPD is now being re-assessed. First, COPD patients both endothelial dysfunction and to intimal hyperplasia [7].
show a wide variation in the individual response of the pul- Smokers with normal spirometry also exhibit an increased
monary circulation to changes in inspired oxygen concen- number of CD8 T cells with a reduction of the CD4/
tration, [52] and the correlation between PaO2 and PAP CD8 ratio in the arterial adventitia, as compared with non-
remains weak [12]. Second, long-term oxygen therapy can- smokers [7] (Fig. 20.3). This suggests that cigarette smoking
not reverse pulmonary hypertension completely [53]. Third, might induce inammatory changes in pulmonary arteries
structural abnormalities and endothelial dysfunction in pul- before the development of lung functional disturbances.
monary arteries can be observed in nonhypoxemic patients Cigarette smoke products may directly inuence pul-
with mild COPD and also in smokers with normal spirom- monary vessel structure. Smokers with normal spirometry
etry [4, 6, 7]. Therefore, hypoxia does not completely explain show prominent changes in pulmonary arteries, such as
the pulmonary vascular changes in COPD. smooth muscle cell proliferation [9], impairment of endothe-
Inammation may play a role in the pathogenesis lial function [7], reduced expression of eNOS [46], increased
of pulmonary vascular abnormalities associated with expression of vascular endothelial growth factor (VEGF) [3],
COPD, although its precise eects have not been estab- and CD8 T-cell inltrate [7]. Most of these changes cannot
lished yet. Inammatory cell inltration of small airways is be dierentiated from those seen in COPD patients. These
changes are clearly absent in nonsmokers, and have been
replicated in part in a guinea pig model using chronic expo-
sure to cigarette smoke [54]. VEGF protein content in lung
tissue is reduced in severe emphysema such that the expres-
sion of VEGF varies according to the severity of COPD
(Fig. 20.4). In the animal model, cigarette smoke exposure
induces muscularization of pre-capillary vessels and increases
PAP [55]. It is of note that vascular abnormalities are clearly
shown after 2 months of cigarette smoke exposure with-
out any evidence of emphysema [55], suggesting that ciga-
rette smoke-induced vascular abnormalities may precede the
development of pulmonary emphysema [56]. Moreover, rapid
changes in gene expression of VEGF, VEGF receptor-1,
ET1, and inducible NOS [57], mediators that regulate vas-
cular cell growth and vessel contraction, likely involved in the
pathogenesis of pulmonary vascular changes of COPD, are
induced in this animal model.
FIG. 20.3 Small pulmonary muscular artery with several CD8 Cigarette smoking is a well-known risk factor for the
lymphocytes infiltrating the adventitia (arrows). development of systemic vascular disease. Active and passive

FIG. 20.4 Photomicrographs of


pulmonary muscular arteries from a
nonsmoker, a patient with moderate
COPD, and a patient with severe
emphysema, immunostained with
antibody against vascular endothelial
growth factor (VEGF). Positive cells
(brown) were located in the intima.
Patients with moderate COPD showed
the greatest immunoreactivity to VEGF.

253
Asthma and COPD: Basic Mechanisms and Clinical Management

exposure to tobacco smoke produces endothelial dysfunc- Gas exchange pathophysiology


tion in both coronary and systemic arteries [58]. Exposure
of pulmonary artery endothelial cells to cigarette smoke Two studies have investigated the status of pulmonary gas
extract causes the inhibition of both eNOS and prostacy- exchange and the behavior of the pulmonary circulation in
clin synthase at the level of protein content and mRNA patients with stable moderate-to-severe persistent asthma.
expression [59, 60]. Cigarette smoke contains a number Patients with unstable, persistent asthma were characterized
of products that have the potential to produce endothelial by longstanding severe airow limitation (FEV1  40%
injury; among these, the aldehyde acrolein seems to play a predicted).
prominent role since it reduces the expression of prostacyc- One of the studies showed that the VA/Q distribu-
lin synthase in endothelial cells [60]. tions demonstrated a broad unimodal blood ow VA/Q pat-
In summary, several evidences suggest that the ini- tern [62]. Accordingly, very little perfusion was associated
tial event in the natural history of pulmonary hypertension with alveolar units with low VA/Q ratios and intrapulmo-
in COPD could be pulmonary endothelial injury induced nary shunt was conspicuously absent; areas of high VA/Q
by cigarette-smoke products with the subsequent down- regions were also negligible and dead space was within
regulation of eNOS and prostacyclin synthase expression, the normal range. The overall amount of VA/Q imbalance,
and impairment of endothelial function. When the disease as assessed by the dispersion of pulmonary blood ow and
progresses, sustained exposure to hypoxemia and inam- that of alveolar ventilation, was slightly increased. The cor-
mation may induce further pulmonary vascular remodeling, relation between VA/Q markers and the degree of airow
thus amplifying the initial eects of cigarette smoke. obstruction, expressed as either FEV1 or peak expiratory
ow or mid-expiratory airow rates, was very modest.
Another study [63] showed similar VA/Q ndings. As in
the previous study [62], there was no correlation between
the VA/Q mismatching and PaO2, pointing to the vital role
BRONCHIAL ASTHMA played by the extra-pulmonary determinants of pulmonary
gas exchange, such as cardiac output and minute ventila-
tion. In this study [62], pulmonary hemodynamics were
Structural background within normal limits. Upon 100% oxygen breathing, it was
shown that VA/Q disturbances further deteriorated. Since
In patients who died of sudden fatal asthma, there is an FEVl did not change, this VA/Q impairment may suggest,
inammatory response in the peripheral airways character- other things being equal, release of hypoxic pulmonary
ized by lumenal occlusion, resulting from airway narrowing vasoconstriction. Despite the changes in the amount of
due to increased smooth muscle thickness, and, importantly, the dispersion of blood ow distribution, it is of note that
an inammatory inltrate, including both mononuclear there were no changes in PAP, indicating that the VA/
cells and eosinophils together with airway secretions [61]. Q descriptors are more sensitive to hyperoxic mixtures
A salient nding is that the muscular pulmonary arter- changes than conventional measures of pulmonary vascular
ies neighboring inamed and narrowed airways show an pressures.
adventitial inammatory inltrate, mainly consisting of
increased numbers of eosinophils. Increased intimal brous
thickening with bands of longitudinal muscle within the References
intima and also externally to the elastica lamina, character-
istic features of chronic alveolar hypoxia, were not exhibited 1. Wilkinson M, Langhorne CA, Heath D, Barer GR, Howard P.
by these vessels. That both the intimal and medial thickness A pathophysiological study of 10 cases of hypoxic cor pulmonale. Q J Med
of muscular pulmonary arteries was within normal lim- 249: 6585, 1988.
its suggested that the eects of regional hypoxia produced 2. Wright JL, Petty T, Thurlbeck WM. Analysis of the structure of the
by mucus plugging were transient and short enough in muscular pulmonary arteries in patients with pulmonary hypertension
and COPD: National Institutes of Health Nocturnal Oxygen Therapy
duration to fail to induce the structural derangement usu-
Trial. Lung 170: 10924, 1992.
ally shown in patients with chronic hypoxic lung diseases 3. Santos S, Peinado VI, Ramirez J, Morales-Blanhir J, Bastos R, Roca J,
[61]. These pulmonary vasculature ndings are of interest Rodriguez-Roisin R, Barbera JA. Enhanced expression of vascu-
for understanding the behavior of the pulmonary circula- lar endothelial growth factor in pulmonary arteries of smokers and
tion and its interplay with the VA/Q matching, particularly patients with moderate chronic obstructive pulmonary disease. Am J
while breathing 100% oxygen. In addition to an increased Respir Crit Care Med 167: 125056, 2003.
smooth muscle thickness in the peripheral airways, this 4. Barber JA, Riverola A, Roca J, Ramirez J, Wagner PD, Ros D,
study [61] demonstrated an increased thickness of the non- Wiggs BR, Rodriguez-Roisin R. Pulmonary vascular abnormalities
muscular airway wall components, most likely resulting and ventilationperfusion relationships in mild chronic obstructive
from inammatory cell inltrate, plasma exudate, and vas- pulmonary disease. Am J Respir Crit Care Med 149: 42329, 1994.
5. Magee F, Wright JL, Wiggs BR, Par PD, Hogg JC. Pulmonary vas-
cular congestion, a combination of changes possibly occur-
cular structure and function in chronic obstructive pulmonary disease.
ring during an acute eruption of the inammatory cascade. Thorax 43: 18389, 1988.
However, not all the airways were occluded by mucus plug- 6. Peinado VI, Barber JA, Ramirez J, Gomez FP, Roca J, Jover L,
ging, as almost half of the airways were without secretions, Gimferrer JM, Rodriguez-Roisin R. Endothelial dysfunction in pul-
whereas the other half showed some degree of luminal monary arteries of patients with mild COPD. Am J Physiol 274:
occlusion. L908L913, 1998.

254
Pulmonary Vessels 20
7. Peinado VI, Barbera JA, Abate P, Ramirez J, Roca J, Santos S, 27. Campbell EJM, Short DS. The cause of oedema in cor pulmonale.
Rodriguez-Roisin R. Inammatory reaction in pulmonary muscular Lancet I: 118486, 1960.
arteries of patients with mild chronic obstructive pulmonary disease. 28. Baudouin SV. Oedema and cor pulmonale revisited. Thorax 52: 4012,
Am J Respir Crit Care Med 159: 160511, 1999. 1997.
8. Hale KA, Ewing SL, Gosnell BA, Niewoehner DE. Lung disease in 29. MacNee W, Wathen CG, Flenley DC, Muir AD. The eects of con-
long-term cigarette smokers with and without chronic airow obstruc- trolled oxygen therapy on ventricular function in patients with stable
tion. Am Rev Respir Dis 130: 71621, 1984. and decompensated cor pulmonale. Am Rev Respir Dis 137: 128995,
9. Santos S, Peinado VI, Ramirez J, Melgosa T, Roca J, Rodriguez-Roisin R, 1988.
Barber JA. Characterization of pulmonary vascular remodelling in 30. MacNee W, Wathen CG, Hannan WJ, Flenley DC, Muir AL. Eects
smokers and patients with mild COPD. Eur Respir J 19: 63238, 2002. of pirbuterol and sodium nitroprusside on pulmonary haemodynamics
10. Naeije R. Should pulmonary hypertension be treated in chronic in hypoxic cor pulmonale. Br Med J (Clin Res Ed) 287: 116972, 1983.
obstructive pulmonary disease? In: Weir EK, Archer SL, Reeves JT 31. Lee-Chiong TL, Matthay RA. The heart in the stable COPD patient.
(eds). The diagnosis and treatment of pulmonary hypertension, pp. 20939. In: Similowski T, Whitelaw WA, Derenne JP (eds). Clinical manage-
Mount Kisco: Futura Publishing, 1992. ment of chronic obstructive pulmonary disease, pp. 475532. New York:
11. Weitzenblum E, Sautegeau A, Ehrhart M, Mammosser M, Hirth C, Marcel Dekker, Inc, 2002.
Roegel E. Long-term course of pulmonary arterial pressure in chronic 32. Palange P. Renal and hormonal abnormalities in chronic obstructive
obstructive pulmonary disease. Am Rev Respir Dis 130: 99398, 1984. pulmonary disease (COPD). Thorax 53: 98991, 1998.
12. Scharf SM, Iqbal M, Keller C, Criner G, Lee S, Fessler HE. 33. Burrows B, Kettel LJ, Niden AH, Rabinowitz M, Diener CF. Patterns
Hemodynamic characterization of patients with severe emphysema. of cardiovascular dysfunction in chronic obstructive lung disease.
Am J Respir Crit Care Med 166: 31422, 2002. N Engl J Med 286: 91218, 1972.
13. Kessler R, Faller M, Weitzenblum E, Chaouat A, Aykut A, Ducolone A, 34. Agust AGN, Barber JA, Roca J, Wagner PD, Guitart R, Rodriguez-
Ehrhart M, Oswald-Mammosser M. Natural history of pulmonary Roisin R. Hypoxic pulmonary vasoconstriction and gas exchange
hypertension in a series of 131 patients with chronic obstructive lung during exercise in chronic obstructive pulmonary disease. Chest 97:
disease. Am J Respir Crit Care Med 164: 21924, 2001. 26875, 1990.
14. Chaouat A, Bugnet AS, Kadaoui N, Schott R, Enache I, Ducolone A, 35. Wright JL, Lawson L, Par PD, Hooper RO, Peretz DI, Nelems JM,
Ehrhart M, Kessler R, Weitzenblum E. Severe pulmonary hyperten- Schulzer M, Hogg JC. The structure and function of the pulmonary
sion and chronic obstructive pulmonary disease. Am J Respir Crit Care vasculature in mild chronic obstructive pulmonary disease. Am Rev
Med, 2005. Respir Dis 128: 7027, 1983.
15. Thabut G, Dauriat G, Stern JB, Logeart D, Levy A, Marrash-Chahla R, 36. Harris P, Segal N, Bishop JM. The relation between pressure and ow
Mal H. Pulmonary hemodynamics in advanced COPD candidates in the pulmonary circulation in normal subjects and in chronic bron-
for lung volume reduction surgery or lung transplantation. Chest 127: chitis. Cardiovasc Res 2: 7383, 1968.
153136, 2005. 37. Naeije R, Barber JA. Pulmonary hypertension associated with COPD.
16. Barber JA. Chronic obstructive pulmonary disease. In: Roca J, Crit Care 5: 28689, 2001.
Rodriguez-Roisin R, Wagner PD (eds). Pulmonary and Peripheral Gas 38. Butler J, Schrijen F, Henriquez A, Polu JM, Albert RK. Cause of the
Exchange in Health and Disease, pp. 22961. New York: Marcel Dekker, raised wedge pressure on exercise in chronic obstructive pulmonary
Inc, 2000. disease. Am Rev Respir Dis 138: 35054, 1988.
17. Roca J, Wagner PD. Contribution of multiple inert gas elimination tech- 39. Montes de Oca M, Rassulo J, Celli BR. Respiratory muscle and cardi-
nique to pulmonary medicine. 1. Principles and information content of opulmonary function during exercise in very severe COPD. Am J Respir
the multiple inert gas elimination technique. Thorax 49: 81524, 1994. Crit Care Med 154: 128489, 1996.
18. Barber JA, Roger N, Roca J, Rovira I, Higenbottam TW, Rodriguez- 40. Rubanyi GM, Romero JC, Vanhoutte PM. Flow-induced release of
Roisin R. Worsening of pulmonary gas exchange with nitric oxide inhala- endothelium-derived relaxing factor. Am J Physiol 250: H114549,
tion in chronic obstructive pulmonary disease. Lancet 347: 43640, 1996. 1986.
19. Peinado VI, Santos S, Ramirez J, Roca J, Rodriguez-Roisin R, 41. Roger N, Barber JA, Roca J, Rovira I, Gomez FP, Rodriguez-Roisin R.
Barber JA. Response to hypoxia of pulmonary arteries in COPD: An Nitric oxide inhalation during exercise in chronic obstructive pulmo-
in vitro study. Eur Respir J 20: 33238, 2002. nary disease. Am J Respir Crit Care Med 156: 8006, 1997.
20. Weitzenblum E, Chaouat A. Right ventricular function in COPD: Can 42. Weitzenblum E. The pulmonary circulation and the heart in chronic
it be assessed reliably by the measurement of right ventricular ejection lung disease. Monaldi Arch Chest Dis 49: 23134, 1994.
fraction? Chest 113: 56769, 1998. 43. Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in
21. Crottogini AJ, Willshaw P. Calculating the end-systolic pressurevol- the relaxation of arterial smooth muscle by acetylcholine. Nature 288:
ume relation. Circulation 83: 112123, 1991. 37376, 1980.
22. Biernacki W, Flenley DC, Muir AL, MacNee W. Pulmonary hyper- 44. Dinh-Xuan AT, Higenbottam T, Clelland C, Pepke-Zaba J, Cremona G,
tension and right ventricular function in patients with COPD. Chest Butt AY, Large S, Wells FC, Wallwork J. Impairment of endothelium-
94: 116975, 1988. dependent pulmonary-artery relaxation in chronic obstructive pulmo-
23. MacNee W. Pathophysiology of cor pulmonale in chronic obstructive nary disease. N Engl J Med 324: 153947, 1991.
pulmonary disease. Part One. Am J Respir Crit Care Med 150: 83352, 45. Giaid A, Saleh D. Reduced expression of endothelial nitric oxide syn-
1994. thase in the lungs of patients with pulmonary hypertension. N Engl J
24. MacNee W. Pathophysiology of cor pulmonale in chronic obstructive Med 333: 21421, 1995.
pulmonary disease. Part Two. Am J Respir Crit Care Med 150: 115868, 46. Barber JA, Peinado VI, Santos S, Ramirez J, Roca J, Rodriguez-Roisin R.
1994. Reduced expression of endothelial nitric oxide synthase in pulmonary
25. Barber JA, Roca J, Ferrer A, Felez MA, Diaz O, Roger N, Rodriguez- arteries of smokers. Am J Respir Crit Care Med 164: 70913, 2001.
Roisin R. Mechanisms of worsening gas exchange during acute exac- 47. Nana-Sinkam SP, Lee JD, Sotto-Santiago S, Stearman RS, Keith RL,
erbations of chronic obstructive pulmonary disease. Eur Respir J 10: Choudhury Q, Cool C, Parr J, Moore MD, Bull TM, Voelkel NF,
128591, 1997. Geraci MW. Prostacyclin prevents pulmonary endothelial cell
26. Weitzenblum E, Apprill M, Oswald M, Chaouat A, Imbs JL. apoptosis induced by cigarette smoke. Am J Respir Crit Care Med 175:
Pulmonary hemodynamics in patients with chronic obstructive pulmo- 67685, 2007.
nary disease before and during an episode of peripheral edema. Chest 48. Giaid A, Yanagisawa M, Langblen D, Michel RP, Levy R, Shennib H,
105: 137782, 1994. Kimura S, Masaki T, Duguid WP, Path FRC, Stewart DJ. Expression

255
Asthma and COPD: Basic Mechanisms and Clinical Management

of endothelin-1 in the lungs of patients with pulmonary hypertension. 56. Yamato H, Churg A, Wright JL. Guinea pig pulmonary hyperten-
N Engl J Med 328: 173239, 1993. sion caused by cigarette smoke cannot be explained by capillary bed
49. Melgosa M, Peinado VI, Santos S, Morales J, Ramirez J, Roca J, destruction. J Appl Physiol 82: 164453, 1997.
Rodriguez-Roisin R, Barber JA. Expression of endothelial nitric oxide 57. Wright JL, Tai H, Dai J, Churg A. Cigarette smoke induces rapid changes
synthase (eNOS) and endothelin-1 (ET-1) in pulmonary arteries of in gene expression in pulmonary arteries. Lab Invest 82: 139198, 2002.
patients with severe COPD. Eur Respir J 22: 20s, 2003. 58. Celermajer DS, Adams MR, Clarkson P, Robinson J, McCredie R,
50. Rabinovitch M, Gamble W, Nadas AS, Miettinen OS, Reid L. Rat Donald A, Deaneld JE. Passive smoking and impaired endothelium-
pulmonary circulation after chronic hypoxia: Hemodynamic and struc- dependent arterial dilatation in healthy young adults. N Engl J Med
tural features. Am J Physiol 236: H81827, 1979. 334: 15054, 1996.
51. Stenmark KR, Fasules J, Hyde DM, Voelkel NF, Henson J, Tucker A, 59. Su Y, Han W, Giraldo C, Li YD, Block ER. Eect of cigarette smoke
Wilson H, Reeves JT. Severe pulmonary hypertension and arterial extract on nitric oxide synthase in pulmonary artery endothelial cells.
adventitial changes in newborn calves at 4,300 m. J Appl Physiol 62: Am J Respir Cell Mol Biol 19: 81925, 1998.
82130, 1987. 60. Nana-Sinkam SP, Lee JD, Sotto-Santiago S, Stearman RS, Keith RL,
52. Ashutosh K, Mead G, Dunsky M. Early eects of oxygen administra- Choudhury Q, Cool C, Parr J, Moore MD, Bull TM, Voelkel NF,
tion and prognosis in chronic obstructive pulmonary disease and cor Geraci MW. Prostacyclin prevents pulmonary endothelial cell apopto-
pulmonale. Am Rev Respir Dis 127: 399404, 1983. sis induced by cigarette smoke. Am J Respir Crit Care Med 175: 67685,
53. Weitzenblum E, Sautegeau A, Ehrhart M, Mammosser M, Pelletier A. 2007.
Long-term oxygen therapy can reverse the progression of pulmonary 61. Saetta M, Di Stefano A, Rosina C, Thiene G, Fabbri LM. Quantitative
hypertension in patients with chronic obstructive pulmonary disease. structural analysis of peripheral airways and arteries in sudden fatal
Am Rev Respir Dis 131: 49398, 1985. asthma. Am Rev Respir Dis 143: 13843, 1991.
54. Wright JL, Churg A. A model of tobacco smoke-induced airow 62. Ballester E, Roca J, Ramis L, Wagner PD, Rodriguez-Roisin R.
obstruction in the guinea pig. Chest 121: 188S91S, 2002. Pulmonary gas exchange in severe chronic asthma. Response to 100%
55. Wright JL, Churg A. Eect of long-term cigarette smoke exposure on oxygen and salbutamol. Am Rev Respir Dis 141: 55862, 1990.
pulmonary vascular structure and function in the guinea pig. Exp Lung 63. Corte P, Young IH. Ventilationperfusion relationships in symptomatic
Res 17: 9971009, 1991. asthma. Response to oxygen and clemastine. Chest 88: 16775, 1985.

256
Adhesion Molecules in Asthma
and Airway Diseases
21 CHAPTER

All cells express a number of transmembrane lies play critical roles in establishing polarity of Dean Sheppard
proteins that could be characterized as adhe- epithelia and endothelia and in maintaining and
sion receptors. Most of these proteins have been regulating endothelial and epithelial permeabil- Department of Medicine and Lung
thus classied based on their role in facilitating ity barriers. Cadherins are the core proteins of Biology Center, University of California,
attachment of cells to either components of the cellcell junctions termed adherans junctions San Francisco, CA, USA
extracellular matrix or other cells. However, which serve as central organizers of epithelial
many adhesion receptors also transduce biologi- and endothelial cellcell adhesion by homotypic
cal signals that modulate cell behavior that are interactions of cadherin extracellular domains
either loosely connected or unrelated to such [1, 2]. Claudins are critical components of tight
adhesion events. In this chapter, I will review junctions that determine the degree of paracel-
the structures of some of the most important lular permeability of endothelia and epithelia to
families of adhesion receptors, will summarize large macromolecules and even small ions [3].
what is known about the patterns of adhe- These proteins also bind directly to family mem-
sion receptor expression and function on cells bers on adjacent cells, but, in contrast to cadher-
relevant to asthma and other chronic airway ins, claudins can associate with either identical
diseases, and will discuss examples of in vivo family members or nonidentical members in
evidence implicating some of these proteins in tight junctions. Multiple claudin isoforms are
pathways which contribute to phenotypic fea- expressed on airway epithelial cells [4]. Occudin
tures of airway diseases. [5], the product of a single gene with multiple
spice variants, can serve as a central organizer of
other tight junction proteins [6].
Immunoglobulin superfamily members
ADHESION MOLECULE FAMILIES play more complex roles in cell adhesion. Some
family members, such as the endothelial adhe-
sion protein, platelet endothelial cell adhesion
Adhesion molecules can be divided into fami- molecule (PECAM), or the small subfamily of
lies based on their primary amino acid sequence junctional adhesion molecules ( JAMs), also play
similarity. Some adhesion molecule families roles in homotypic cellcell adhesion, but JAMs
principally mediate cellcell adhesion, others have also been reported to bind to members of
are principally involved in cell adhesion to the other adhesion protein families (e.g., integrins)
extracellular matrix, and some are involved in and to play roles in heterotypic cellcell adhe-
both types of adhesion events. Families involved sion (e.g., between endothelial or epithelial cells
in cellcell adhesion include claudins, occludins, and migrating leukocytes). Other immunoglob-
cadherins, and immunoglobulin superfamily ulin superfamily members (e.g., intercellular
members. Claudins, occludins, and cadher- adhesion molecules, ICAMs and vascular adhe-
ins are all monomeric proteins that principally sion molecule, VCAM) are restricted to roles
mediate homotypic cellcell adhesion, usu- in heterotypic cellcell adhesion, principally
ally through direct binding to members of the through their roles as integrin ligands. Selectins,
same family expressed at the lateral margins of another small family of adhesion molecules,
adjacent cells (Table 21.1). These protein fami- also principally mediate heterotypic cellcell

257
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 21.1 Selected adhesion molecules of potential importance in asthma and COPD

Name Structure Binding partners Location Comments

Claudins Multiple membrane spanning Homotypic and heterotypic Tight junctions Regulate ion permeability at
(tetraspanin) bindings to Claudins tight junctions

JAMs Single membrane spanning Ig Homotypic and/or heterotypic Tight junction Mediate leukocyte trans-
family members binding to JAMS. Also bind endothelial migration,
selected integrins endothelial and epithelial
cellcell adhesion

Selectins Single membrane spanning Trans-membrane Platelets, leukocytes, and Mediate leukocyte rolling, can
monomers with lectin domain proteoglycans with sialyl-Lewis endothelial cells initiate intracellular signals.
X motif Potential drug target for
asthma

VCAM1 Single membrane spanning Ig Integrins 41 and 91 Endothelial cells, tumor cells, Counter-receptor for integrins
family member smooth muscle, pericytes 41 and 91. Important for
recruitment of leukocytes

ICAM1 Single membrane spanning Ig 2 Integrins Endothelial and epithelial cells Counter-receptor for 2
family member integrins. Possible drug target
for asthma

41 Integrin Transmembrane heterodimer VCAM1, fibronectin, ADAMs Endothelium, pericytes, Potential drug target for
proteins smooth muscle, lymphocytes, asthma
eosinophils, and mast cells

v6 Integrin Transmembrane heterodimer TGF1 and -3, fibronectin, Epithelial cells Activates TGF-. Negatively
tenascin C regulates macrophage
protease expression and
emphysema

v8 Integrin Transmembrane heterodimer TGF1 and -3, vitronectin Airway epithelial cells, Activates TGF-. Involved in
fibroblasts, dendritic cells, and cross talk between airway
T-cells fibroblasts and epithelial cells

interactions, in that case through interactions with carbohy-


drate components of proteoglycan counter-receptors.
Receptors for components of the extracellular matrix
Y
can also play complex roles in cell biology. Syndecans are
transmembrane proteoglycans that appear to function solely
X Talin
as receptors for components of the extracellular matrix.
However, integrins, initially identied as extracellular Z
matrix receptors, are now known to recognize a wide variety ILK
of spatially restricted proteins as ligands, including cellular Tensin
Actin
counter-receptors, growth factors, and proteases [7]. Adhesion, tissue
Integrins were initially identied based on their ability integrity
to mediate stable adhesion to xed extracellular matrix lig- (conserved from
ands. Integrin-mediated cell adhesion depends on the abil- C.elegans (2)
Cell- and tissue-specific
to mammals (24))
ity of subunit cytoplasmic domains to bind to a group of responses (vertebrates)
adaptor proteins that link integrins to the actin cytoskeleton
(Fig. 21.1). By connecting the relatively rigid extracellular FIG. 21.1 Integrin functions. Integrins evolved to link the actin
cytoskeleton and the extracellular matrix, a function that is highly conserved
matrix to the relatively rigid actin cytoskeleton, integrins
from C. elegans to humans (shown on right). This linkage is accomplished
play important roles in maintaining cohesion of multicellular through formation of a highly conserved multiprotein complex that
organs and organisms. These factors also position integrins includes talin, tensin, and the integrin-linked kinase to the integrin
as essential components of cellular force transducers which subunit cytoplasmic domain. In vertebrates, integrins have evolved to
allow cells to detect and respond to mechanical deforma- recruit a multitude of additional cytoplasmic and transmembrane adaptor
tion. The proteins involved in linking integrins and actin are and signaling proteins to these complexes, allowing these receptors to
highly conserved from Caenorhabditis elegans to humans. modulate a wide variety of cellular behaviors (shown on left).

258
Adhesion Molecules in Asthma and Airway Diseases 21
During the evolutionary transition from invertebrates Chemokine
to vertebrates, there was a rapid increase in the size of the Selectin Chemokine receptor
integrin family, and integrins have been adapted to bind to a
number of additional ligands and to modulate a wide variety of Selectin Integrin
cellular behaviors, including cell proliferation, dierentiation, ligand
Ig family member
and survival [8]. These new functions have been accomplished
by recruitment of cytoplasmic kinases (such as src and focal
adhesion kinase) to the multiprotein complexes assembled on
integrin cytoplasmic domains and by lateral associations with a
number of additional signaling molecules and adaptor proteins
(e.g., tetraspanins, growth factor receptors, urokinase receptor,
u-PAR, and the integrin-associated protein).

ROLES OF ADHESION RECEPTORS ON


LEUKOCYTES AND ENDOTHELIAL CELLS IN FIG. 21.2 Multistep process of leukocyte rolling arrest and trans-
REGULATING AIRWAY INFLAMMATION endothelial migration.Initial slowing of leukocytes from rapidly moving
blood is usually mediated by interactions of selectins on leukocytes
and endothelial cells with cell surface carbohydrate ligands (left). Close
Airway and lung parenchymal inammation are important contact with chemokines then triggers an intracellular signal that changes
features of asthma and chronic obstructive pulmonary dis- leukocyte integrins from a low-affinity to a high-affinity state and mediates
ease (COPD). Inhibition of recruitment of specic popula- stable arrest through interaction with endothelial immunoglobulin family
tions of inammatory cells into the airways has thus been members (center). Dynamic regulation of integrin adhesion then allows
migration of adherent leukocytes across the endothelium and into and
considered as a potential therapeutic intervention in these
through the extravascular space (right).
diseases. Recruitment of inammatory cells from the blood-
stream into tissues is a multistep process [9], as outlined in
Fig. 21.2. Key steps in this process include initial slowing
of rapidly moving cells, a process called rolling [10]. These The next step in leukocyte recruitment, rm adhesion
slowed cells must then undergo rm adhesion to completely to the vessel wall, is mediated principally by the interaction
arrest their movement in the bloodstream and then need to of integrins on leukocytes with immunoglobulin superfamily
resume a dierent form of slow migration to exit the vas- members on endothelial cells. The specic integrins involved,
culature either in induced gaps between adjacent endothe- and the corresponding immunoglobulin family counter-
lial cells or by passing directly through the induced holes receptors, vary among dierent leukocytes. Neutrophils
within individual endothelial cells. Once they have exited were initially thought to arrest their movement by virtue
the bloodstream, leukocytes then migrate through the of adhesion of the integrins L2 and M2 with the
interstitial space where some arrest in the vicinity of nerves endothelial counter-receptor ICAM-1, whereas monocytes,
and airway smooth muscle, others arrest within the airway lymphocytes, and eosinophils [16] utilize 41, which inter-
epithelium, and some traverse the epithelium and enter the acts with endothelial VCAM-1 [17]. However, neutrophils
airspaces. Each of these steps of adhesion and migration also express the integrin 91, which avidly binds VCAM-
involves a suite of adhesion receptors expressed on the leu- 1 [18], and lymphocytes, monocytes, and eosinophils also
kocytes, endothelial cells, or other target extravascular cells. utilize L2 and thus arrest on ICAM-1.
The rst step in this process, leukocyte rolling, is As with the initial rolling step, rm arrest is tightly
principally mediated by interactions of a selectin on leu- regulated through signaling in both leukocytes and endothe-
kocytes (L selectin) with specic carbohydrate ligands on lial cells. On the leukocyte side, integrins in circulating leu-
endothelial cells and selectins on endothelial cells (P and E kocytes are generally maintained in a nonadhesive (inactive)
selectins) with additional carbohydrate ligands on leukocytes conformation. In response to signals generally initiated by the
[11]. This process is principally regulated by the induction binding of chemokines to their cognate G-protein-coupled
of expression of L selectin ligands and P and E selectins receptors (which are presented to integrins on rolling leu-
only on endothelial cells that have been activated by pro- koctyes by virtue of their interactions with proteoglycans on
inammatory cytokines, such as interleukin-1 and TNF-. the luminal surface of endothelial cells), the short cytoplas-
In this process, selectins play a critical role in facilitating mic domains of the integrin- and - subunits are separated
the rolling of neutrophils along the vessel wall. However, by interactions with one or more cytoplasmic binding part-
it is now clear that certain members of the integrin fam- ners, and this change in subunit spacing induces a massive
ily, for example the integrins 41 and 47, can medi- conformational change in the extracellular domains of both
ate rolling even in the absence of input from selectins [12, subunits that result in a high-anity conformation that is
13]. Selectin-mediated rolling may play an important role competent to bind to endothelial ligands. On the endothe-
in asthma, since inhibition of selectins or their ligands has lial side, the same pro-inammatory cytokines that induce
been shown to reduce allergen-induced airway hyperrespon- expression of P and E selectins increase transcription and
siveness in models of allergic asthma [14, 15]. subsequent surface expression of ICAM-1 and VCAM-1.

259
Asthma and COPD: Basic Mechanisms and Clinical Management

4-containing integrins are more widely expressed


ROLES OF INTEGRINS AND THEIR LIGANDS IN than 2-integrins but are also expressed on a number of leu-
LEUKOCYTE MIGRATION AND RETENTION IN kocytes, including lymphocytes, eosinophils, and mast cells.
THE EXTRAVASCULAR SPACE Blockade of 4-integrins has been shown to protect against
induction of airway hyperresponsiveness in models of aller-
gic asthma in mice and sheep [23, 24]. Mice lacking 4 only
Whereas the molecular steps involved in rolling and rm on leukocytes have also been shown to be protected from
adhesion are relatively well characterized, much less is know induction of airway hyperresponsiveness, suggesting that 4-
about how subsequent steps of extravasation and migra- integrins (41 and 47) might be reasonable targets for
tion and retention at specic sites are regulated. These steps treatment of asthma. Although this strategy was conceived as
also require repeated cycles of adhesion and de-adhesion. a way to prevent the recruitment of leukocytes into the air-
Although multiple molecular pathways have been shown to ways, local delivery of blocking antibody into the airways of
modulate these steps in various in vitro systems, the relative allergic sheep prevented the development of airways hyperre-
signicance of most of these for specic leukocyte popula- sponsiveness (AHR) without aecting leukocyte recruitment
tions in vivo remains to be determined. One of the steps [24]. These results suggest that protection from AHR might
that has just begun to receive attention is the retention of be due to inhibition of 4-integrin-mediated signals initi-
leukocytes at specic tissue sites. The potential importance ated on cells that are already present in the airways, rather
of this step in asthma was demonstrated by mice lacking than simply to eects on leukocyte recruitment.
the metalloprotease, MMP2, which were found to have dra- Based on these encouraging pre-clinical studies, both
matically increased susceptibility to fatal respiratory failure humanized antibodies and small molecule antagonists of
in response to airway allergen challenge after prior allergic 4-integrins have been developed as potential drug treat-
sensitization [19]. Mortality in these mice was shown to be ments for asthma. Humanized antibody has been shown to
associated with abnormal retention of recruited leukocytes be dramatically eective for prevention of new brain lesions
in the pulmonary interstitium, with a marked decrease in the in another inammatory disease, multiple sclerosis [25].
subsequent migration of these cells into the airway lumen. However, the development of fatal or disabling progressive
One of the critical steps involved in retention of multifocal leukoencephalopathy in three patients treated
inammatory cells within the airway epithelium is induced with this antibody, presumably as a consequence of activa-
expression of ICAM-1 on airway epithelial cells. Another tion of latent JC virus infection, has dramatically slowed
key step is the induction of the integrin E7, which can be eorts to use system blockade of 4-integrins for treatment
induced on lymphocytes and mast cells by the action of the of asthma [26, 27]. Because of the potential eectiveness of
cytokine transforming growth factor (TGF-), activated local inhibition of these integrins, inhalational therapy with
on epithelial cells at sites of airway injury (see below). E7 4-antagonists is still worthy of consideration.
recognizes an epithelial adhesion receptor, E Cadherin, Selectins have also been targeted in pre-clinical stud-
as its principal ligand [20], and through this interaction ies in mice and sheep. Inhibition of one of the three selectin
increases the residence time of mast cells and lymphocytes family members (L selectin) also inhibited airway inam-
within the epithelium. These leukocytes utilize the E7- mation, allergen-induced late airway narrowing, and AHR
integrin and presumably E Cadherin for retention within in allergic sheep [14]. As with 4-blockade, local delivery
the epithelium. of blocking antibody to L selectin was as eective as sys-
temic administration, suggesting relevant eects of ligation
of selectins on inammatory cells after their recruitment
into the airway wall.
POTENTIAL THERAPEUTIC RELEVANCE OF
LEUKOCYTE CELL ADHESION RECEPTORS
AND THEIR ENDOTHELIAL AND EPITHELIAL ADHESION RECEPTORS ON AIRWAY
COUNTER-RECEPTORS SMOOTH MUSCLE

Because airway inammation is a prominent feature of Airway smooth muscle is clearly an important target tissue
asthma, and because leukocytes are the principal source of in asthma. An increase in smooth muscle mass is a common
cytokines (e.g., interleukin-13) known to play a major role feature in both human asthma and experimental models of
in inducing airway hyperresponsiveness and mucus meta- chronic allergen challenge, and alterations in in vivo smooth
plasia, leukocyte adhesion molecules and their endothe- muscle responsiveness might be an important determinant of
lial and/or epithelial ligands have been extensively studied airway hyperresponsiveness. Airway smooth muscle cells can
as potential therapeutic targets. Nearly two decades ago, express ICAM-1 and VCAM-1 and a number of integrins,
blockade of the 2-integrin ligand, ICAM-1, was shown to including at least 11, 21, 31, 71, 81, 91, and
prevent airway hyperresponsiveness in a murine model of v3. These integrins are localized at intercalated disks that
allergic asthma [21]. Subsequent studies with mice lacking connect adjacent smooth muscle cells and are thus likely to
the integrin 2 have conrmed an important role for this play some role in regulating the strength of smooth muscle
family of integrins in the specic homing of Th2 eector T contraction. Airway smooth muscle cells exhibit markedly dif-
cells to the lungs [22]. ferent proliferative responses and modulate their production of

260
Adhesion Molecules in Asthma and Airway Diseases 21
pro-inammatory cytokines when they are plated on dierent appear to utilize completely dierent mechanisms for TGF-
integrin ligands [28, 29]. Furthermore, integrins in vascular activation. v6 is restricted to epithelial cells and acti-
smooth muscle cells have been shown to modulate contrac- vates TGF- by mechanically deforming the latent complex
tion by regulating the activity of associated L-type voltage- (Annes). This process provides access to the active site in
gated calcium channels [30, 31]. The in vivo signicance of TGF- without releasing free TGF- for diusion to dis-
adhesion receptors on airway smooth muscle cells thus seems tant receptors and thus spatially restricts TGF- activity to
like a fruitful area for future investigation. the surface of v6 expressing epithelial cells [35]. In con-
trast, v8, which is much more widely expressed, presents
the latent complex to transmembrane metalloproteinases
expressed on the same cell, which cleave the latent complex
ADHESION RECEPTORS ON PULMONARY allowing release of diusible active TGF- that could act at
EPITHELIAL CELLS a distance [36].
Current evidence suggests that v6 plays a central
role in activating TGF- on the surface of alveolar epithe-
One potential contributing factor to increased respon- lial cells, where the activated TGF- can exert eects on
siveness to environmental stimuli in asthma and COPD adjacent broblasts, endothelial cells, and alveolar macro-
would be localized increases in epithelial permeability. The phages which make direct physical contacts with alveolar
normal impermeable character of the airway epithelium is epithelial cells. This process has been shown to play impor-
determined by the function of the adhesion molecules that tant roles in regulating pulmonary brosis [35] and several
make up adherens junctions (E Cadherin) and tight junc- models of acute lung injury [38], where genetic deletion or
tions (claudins and occludins). In contrast to responses in antibody blockade of the v6-integrin dramatically pro-
endothelial cells, where permeability can be rapidly (within tects against induction of in vivo pathology. Microarray
minutes) increased in response to a number of physiological studies on the lungs of mice lacking the 6 subunit (and
stimuli (e.g., VEGF, TGF-, and thrombin), changes in epi- thus the v6-integrin) identied the macrophage metallo-
thelial permeability usually occur with a time course of sev- protease, MMP12 as an important target of negative regu-
eral hours and are associated with loss of adhesion molecules lation by this integrin, since MMP12 levels were increased
(especially E Cadherin) from the cell surface. In the setting 200-fold in the lungs of 6 knockout mice [39]. Induction
of chronic inammation, for example with exposure to active of MMP12 leads to the development of age-related emphy-
TGF-, E Cadherin is internalized and epithelial permeabil- sema in these mice and can be prevented by expression
ity increases. However, the in vivo relevance of these events of the integrin or low levels of active TGF- in lung epi-
to chronic airway disease has not been determined. thelium [40]. These ndings suggest that v6-mediated
Airway epithelial cells express several members of the TGF- activation plays an important homeostatic role in
integrin family, including 21, 31, 64 91, v5, preventing the development of emphysema in response to
v6, and v8 [32]. Epithelial injury and inammation can normally inconsequential environmental insults (Fig. 21.3).
dramatically alter the expression of these integrins [33]. The
integrin v6, which is normally expressed at low levels in the
conducting airway epithelium of healthy individuals, is upreg-
ulated in the airways of patients with COPD and asthma [34].

RGD Macrophage
INTEGRIN-MEDIATED TGF- ACTIVATION TGF-R
SMAD4
RGD
At least two of the integrins expressed on airway epithe- R-SMADs
lial cells, v6 [35] and v8 [36], play a role in activating 6
latent complexes of TGF-. TGF- is a pleiotropic cytokine v
MMP12
that plays important roles in regulating tissue brosis, in
induced proliferation of airway smooth muscle, in recruiting
and retaining tissue mast cells and in negatively regulating
adaptive immunity. It is thus clear that TGF- could play
important roles in several basic processes relevant to asthma Epithelium Environmental Emphysema
and COPD. Mice with a knockin mutation of the TGF-1 insults
gene, which uniquely prevents integrin-mediated activation,
have an identical inammatory phenotype as mice com- FIG. 21.3 Homeostatic regulation of macrophage MMP12 expression by
integrin-mediated activation of TGF-. Environmental insults (e.g., cigarette
pletely lacking TGF-1 have [37], suggesting that integrin-
smoke) increase macrophage expression of MMP12 (and other proteases that
mediated activation may be the central in vivo mechanism can degrade the extracellular matrix of the lung. In parallel, the same insults
of TGF-1 activation. Indirect evidence suggests similar activate a pathway through which the epithelial integrin, v6, activates
importance for activation of TGF-3. TGF-. Activated TGF- binds to TGF- receptors on macrophages, activating
v6 and v8 have dierent patterns of cellular a signaling pathway involving transcription factors (receptor SMADs R-
expression. Each binds to the same sequence of the latency- SMADs) that pair with co-SMADs to shut off transcription of the MMP12
associated peptides of TGF-1 and -3, but these two integrins gene, thereby providing a brake for this potentially destructive process.

261
Asthma and COPD: Basic Mechanisms and Clinical Management

INTEGRIN-MEDIATED TGF- ACTIVATION loss of this function could contribute to the development of
emphysema. TGF- activation by the v8-integrin plays
IN THE CONDUCTING AIRWAYS an important role in homeostasis in the airway wall, and
alterations in this function could contribute to airway wall
Both v6 and v8 are expressed on epithelial cells in the remodeling in COPD. The multiple critical roles played by
conducting airways. Under resting conditions, cultured air- adhesion molecules in airway health and disease make these
way epithelial cells express both integrins and secrete both proteins attractive candidates for improvements in treat-
TGF-1 and TGF-2 but do not appear to activate either ment and monitoring of patients with asthma and COPD.
isoform [41]. In response to mechanical wounding, these
cells can be induced to activate TGF-1 (but not TGF-2)
and both integrins appear to contribute to this activation. REFERENCES
The net eect of this autocrine TGF- activation is a slow-
ing of the rate of sheet migration and subsequent wound 1. Takeichi M. Cadherin cell adhesion receptors as a morphogenetic reg-
closure. Interestingly, whereas blockade of v8, like block- ulator. Science 251(5000): 145155, 1991.
ade of TGF-1, accelerates the rate of wound closure, block- 2. Gumbiner BM. Cell adhesion: The molecular basis of tissue architec-
ade of v6 has no net eect on closure rates. This appears ture and morphogenesis. Cell 84(3): 34557, 1996.
to be due to opposing eects of this integrin on activating 3. Anderson JM. Molecular structure of tight junctions and their role in
TGF- (which slows closure rates) and on directly enhanc- epithelial transport. News Physiol Sci 16: 12630, 2001.
ing epithelial migration, as shown by slowing of wound clo- 4. Wang F, Daugherty B, Keise LL, Wei Z, Foley JP, Savani RC, Koval M.
sure when anti-v6 antibody was added to wounded cells Heterogeneity of claudin expression by alveolar epithelial cells. Am J
Respir Cell Mol Biol 29(1): 6270, 2003.
in the presence of inhibitors of TGF- or v8.
5. Furuse M, Hirase T, Itoh M, Nagafuchi A, Yonemura S, Tsukita S,
In contrast to v6, which is restricted in its expres- Tsukita S. Occludin: A novel integral membrane protein localizing at
sion to airway epithelial cells and is highly induced in tight junctions. J Cell Biol 123(6 Pt 2): 177788, 1993.
response to injury, v8 is also expressed on airway brob- 6. Nusrat A, Chen JA, Foley CS, Liang TW, Tom J, Cromwell M, Quan C,
lasts [42]. Recent work has shown that v8 can mediate Mrsny RJ. The coiled-coil domain of occludin can act to organize
autocrine activation of TGF- by airway broblasts and structural and functional elements of the epithelial tight junction. J Biol
that this eect inhibits to production of hepatocyte growth Chem 275(38): 2981622, 2000.
factor by adjacent epithelial cells. Through this process, 7. Hynes RO. Integrins: Versatility, modulation, and signaling in cell
airway broblasts can also induce expression of v6 on adhesion. Cell 69: 1125, 1992.
epithelial cells (which is highly inducible by TGF-) gen- 8. Clark EA, Brugge JS. Integrins and signal transduction pathways: The
road taken. Science 268(5208): 23339, 1995.
erating a positive feedback loop that would be predicted to
9. McEver RP. Leukocyte-endothelial cell interactions. Curr Opin Cell
inhibit airway epithelial cell proliferation and contribute to Biol 4(5): 84049, 1992.
the development of airway wall brosis. 10. Luscinskas FW, Kansas GS, Ding H, Pizcueta P, Schleienbaum BE,
Tedder TF, Gimbrone MA Jr. Monocyte rolling, arrest and spreading
on IL-4-activated vascular endothelium under ow is mediated via
sequential action of L-selectin, beta 1-integrins, and beta 2-integrins.
SUMMARY J Cell Biol 125(6): 141727, 1994.
11. McEver RP. Selectins: Lectins that initiate cell adhesion under ow.
Curr Opin Cell Biol 14(5): 58186, 2002.
Adhesion molecules play critical roles in maintaining the 12. Alon R, Kassner PD, Carr MW, Finger EB, Hemler ME, Springer TA.
structural integrity of all components of the airway wall. In The integrin VLA-4 supports tethering and rolling in ow on VCAM-1.
the epithelium and endothelium, occludins, claudins, cad- J Cell Biol 128(6): 124353, 1995.
herins, and JAMs regulate paracellular permeability. Airway 13. Jones DA, McIntire LV, Smith CW, Picker LJ. A two-step adhesion
injury or inammation, as occurs in patients with asthma cascade for T cell/endothelial cell interactions under ow conditions.
J Clin Invest 94(6): 244350, 1994.
and COPD, can perturb epithelial and endothelial perme-
14. Abraham WM, Ahmed A, Sabater JR, Lauredo IT, Botvinnikova Y,
ability by altering the expression and/or function of these Bjercke RJ, Hu X, Revelle BM, Kogan TP, Scott IL, Dixon RA,
proteins. Selectins, integrins, and their immunoglobulin Yeh ET, Beck PJ. Selectin blockade prevents antigen-induced late
family member counter-receptors, play central roles in the bronchial responses and airway hyperresponsiveness in allergic sheep.
recruitment of leukocytes into the airway wall and in regu- Am J Respir Crit Care Med 159(4 Pt 1): 120514, 1999.
lating the function of leukocytes that have been recruited. 15. Rosen SD, Tsay D, Singer MS, Hemmerich S, Abraham WM.
Promising pre-clinical studies suggest that members of each Therapeutic targeting of endothelial ligands for L-selectin (PNAd) in
of these classes of adhesion molecules are potential drug a sheep model of asthma. Am J Pathol 166(3): 93544, 2005.
targets for treatment of asthma. Integrins also contribute 16. Sriramarao P, von Andrian UH, Butcher EC, Bourdon MA,
to signaling responses of airway cells to input from other Broide DH. L-selectin and very late antigen-4 integrin promote eosi-
nophil rolling at physiological shear rates in vivo. J Immunol 153(9):
receptor families, including growth factor and cytokine
423846, 1994.
receptors, and can modulate survival, growth, and dieren- 17. Elices MJ, Osborn L, Takada Y, Crouse C, Luhowskyj S, Hemler ME,
tiation of each of the cell types present in the airway wall. Lobb RR. VCAM-1 on activated endothelium interacts with the leu-
Two integrins, v6 and v8, are also largely responsible kocyte integrin VLA-4 at a site distinct from the VLA-4/bronectin
for in vivo activation of two of three mammalian isoforms binding site. Cell 60(4): 57784, 1990.
of TGF-. TGF- activation by the v6-integrin is criti- 18. Taooka Y, Chen J, Yednock T, Sheppard D. The integrin alpha9beta1
cal for the maintenance of normal alveolar homeostasis, and mediates adhesion to activated endothelial cells and transendothelial

262
Adhesion Molecules in Asthma and Airway Diseases 21
neutrophil migration through interaction with vascular cell adhesion activation of L-type calcium channels in vascular smooth muscle causes
molecule-1. J Cell Biol 145(2): 41320, 1999. arteriole vasoconstriction. Circ Res 90(4): 47380, 2002.
19. Corry DB, Rishi K, Kanellis J, Kiss A, Song Lz LZ, Xu J, Feng L, 31. Wu X, Mogford JE, Platts SH, Davis GE, Meininger GA, Davis MJ.
Werb Z, Kheradmand F. Decreased allergic lung inammatory cell Modulation of calcium current in arteriolar smooth muscle by alphav
egression and increased susceptibility to asphyxiation in MMP2- beta3 and alpha5 beta1 integrin ligands. J Cell Biol 143(1): 24152,
deciency. Nat Immunol 3(4): 34753, 2002. 1998.
20. Cepek KL, Shaw SK, Parker CM, Russell GJ, Morrow JS, Rimm DL, 32. Sheppard D. Airway epithelial integrins: Why so many? Am J Respir
Brenner MB. Adhesion between epithelial cells and T lymphocytes Cell Mol Biol 19(3): 34951, 1998.
mediated by E-cadherin and the alpha E beta 7 integrin. Nature 33. Sheppard D. Epithelial integrins. Bioessays 18(8): 65560, 1996.
372(6502): 19093, 1994. 34. Weinacker A, Ferrando R, Elliott M, Hogg J, Balmes J, Sheppard D.
21. Wegner CD, Gundel RH, Reilly P, Haynes N, Letts LG, Rothlein R. Distribution of integrins alpha v beta 6 and alpha 9 beta 1 and their
Intercellular adhesion molecule-1 (ICAM-1) in the pathogenesis of known ligands, bronectin and tenascin, in human airways. Am J Respir
asthma. Science 247(4941): 45659, 1990. Cell Mol Biol 12: 54756, 1995.
22. Lee SH, Prince JE, Rais M, Kheradmand F, Shardonofsky F, Lu H, 35. Munger JS, Huang X, Kawakatsu H, Griths MJ, Dalton SL, Wu J,
Beaudet AL, Smith CW, Soong L, Corry DB. Dierential requirement Pittet JF, Kaminski N, Garat C, Matthay MA, Rifkin DB, Sheppard D.
for CD18 in T-helper eector homing. Nat Med 9(10): 128186, 2003. The integrin alpha v beta 6 binds and activates latent TGF beta 1:
23. Abraham WM, Gill A, Ahmed A, Sielczak MW, Lauredo IT, A mechanism for regulating pulmonary inammation and brosis.
Botinnikova Y, Lin KC, Pepinsky B, Leone DR, Lobb RR, Adams SP. Cell 96(3): 31928, 1999.
A small-molecule, tight-binding inhibitor of the integrin alpha(4)beta(1) 36. Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H,
blocks antigen-induced airway responses and inammation in experi- Sheppard D, Broaddus VC, Nishimura SL. The integrin alpha(v)beta8
mental asthma in sheep. Am J Respir Crit Care Med 162(2 Pt 1): 60311, mediates epithelial homeostasis through MT1-MMP-dependent acti-
2000. vation of TGF-beta1. J Cell Biol 157(3): 493507, 2002.
24. Abraham WM, Sielczak MW, Ahmed A, Cortes A, Lauredo IT, 37. Yang Z, Mu Z, Dabovic B, Jurukovski V, Yu D, Sung J, Xiong X,
Kim J, Pepinsky B, Benjamin CD, Leone DR, Lobb RR et al. Alpha Munger JS. Absence of integrin-mediated TGFbeta1 activation in vivo
4-integrins mediate antigen-induced late bronchial responses and recapitulates the phenotype of TGFbeta1-null mice. J Cell Biol 176(6):
prolonged airway hyperresponsiveness in sheep. J Clin Invest 93(2): 78793, 2007.
77687, 1994. 38. Pittet JF, Griths MJ, Geiser T, Kaminski N, Dalton SL, Huang X,
25. Miller DH, Khan OA, Sheremata WA, Blumhardt LD, Rice GP, Brown LA, Gotwals PJ, Koteliansky VE, Matthay MA, Sheppard D.
Libonati MA, Willmer-Hulme AJ, Dalton CM, Miszkiel KA, TGF-beta is a critical mediator of acute lung injury. J Clin Invest 107(12):
OConnor PW. A controlled trial of natalizumab for relapsing multiple 153744, 2001.
sclerosis. N Engl J Med 348(1): 1523, 2003. 39. Kaminski N, Allard JD, Pittet JF, Zuo F, Griths MJ, Morris D,
26. Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoen- Huang X, Sheppard D, Heller RA. Global analysis of gene expres-
cephalopathy complicating treatment with natalizumab and interferon sion in pulmonary brosis reveals distinct programs regulating lung
beta-1a for multiple sclerosis. N Engl J Med 353(4): 36974, 2005. inammation and brosis. Proc Natl Acad Sci USA 97(4): 177883,
27. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. 2000.
Progressive multifocal leukoencephalopathy in a patient treated with 40. Morris DG, Huang X, Kaminski N, Wang Y, Shapiro SD, Dolganov G,
natalizumab. N Engl J Med 353(4): 37581, 2005. Glick A, Sheppard D. Loss of integrin alpha(v)beta6-mediated TGF-
28. Nguyen TT, Ward JP, Hirst SJ. beta1-Integrins mediate enhancement beta activation causes Mmp12-dependent emphysema. Nature 422(6928):
of airway smooth muscle proliferation by collagen and bronectin. 16973, 2003.
Am J Respir Crit Care Med 171(3): 21723, 2005. 41. Neurohr C, Nishimura SL, Sheppard D. Activation of transforming
29. Peng Q, Lai D, Nguyen TT, Chan V, Matsuda T, Hirst SJ. Multiple growth factor-beta by the integrin alphavbeta8 delays epithelial wound
beta 1 integrins mediate enhancement of human airway smooth muscle closure. Am J Respir Cell Mol Biol 35(2): 25259, 2006.
cytokine secretion by bronectin and type I collagen. J Immunol 174(4): 42. Araya J, Cambier S, Morris A, Finkbeiner W, Nishimura SL. Integrin-
225864, 2005. mediated transforming growth factor-beta activation regulates homeos-
30. Waitkus-Edwards KR, Martinez-Lemus LA, Wu X, Trzeciakowski tasis of the pulmonary epithelial-mesenchymal trophic unit. Am J Pathol
JP, Davis MJ, Davis GE, Meininger GA. alpha(4)beta(1) Integrin 169(2): 40515, 2006.

263
Extracellular Matrix
22 CHAPTER

matrix components that comprise the tissue [2,


INTRODUCTION 12]. In the lung they can be largely categorized
Maurice Godfrey
into four groups: the collagens, proteoglycans, Center for Human Molecular Genetics,
Until recently the extracellular matrix was elastic bers, and non-collagenous proteins. Munroe Meyer Institute, University
thought of solely as a static structural support Connective tissue in the lung imparts the of Nebraska Medical Center, Omaha,
network. We now know that the extracellular appropriate mechanical sturdiness and elastic NE, USA
matrix is comprised by a large and varied group resilience that permits the lung to expand and
of dynamic macromolecules and their regula- relax repeatedly. In addition to this mechanical
tory factors [1] which provides structural sup- function, the extracellular matrix is organized to
port and is a physical barrier. However, it also allow for ecient gas exchange (Table 22.1).
elicits cellular responses and its interactions
are involved in development and organ forma-
tion [2]. Disruption of normal extracellular Collagens
matrix during disease processes can lead to an
inammatory response that exacerbate aber- By far the most abundant of the extracellular
rant remodeling of the lung [3, 4]. Much has matrix elements are the collagens. The collagens
also been learned about the role of the extra- are the largest, in number, and most abundant,
cellular matrix in asthma and chronic obstruc- in content, of the extracellular matrix elements.
tive pulmonary disease (COPD) using mouse There are more than 20 dierent collagen types
models [5, 6]. These studies tend to support and even more genes that encode these colla-
observations of altered function in people with gens [1, 2, 13].
polymorphic variants of extracellular matrix Collagens are characterized by a triple
molecules [79]. helical structure whereby three polypeptide
Extracellular matrix molecules are a part chains wind around each other. To form this
of a nely regulated system of development, helical conguration, every third amino acid
maintenance, and repair. In addition to the must be a glycine. Glycine, with its very small
structural macromolecules that are discussed in side chain is the only amino acid that is able
this chapter, there are regulatory molecules that to t in the center of the helix without caus-
are essential components of the extracellular ing any distortion. The primary structure of the
matrix [10, 11]. In this chapter we will review triple helical region of collagens is described by
the extracellular matrix constituents of the res- the general pattern of [Gly-X-Y]n as a repeat-
piratory system. ing triplet. About 20% of the X and Y positions
in this triplet are the imino acids, proline, and
hydroxyproline. The hydroxyl group of hydroxy-
COMPONENTS OF THE proline is essential for hydrogen bonding which
EXTRACELLULAR MATRIX imparts stability to the triple helix. Similarly,
lysine and hydroxylysine residues in the heli-
cal region are important for covalent cross-
The biomechanical properties of the lung links within and between collagen molecules.
are primarily determined by the extracellular Hydroxylysines are also subject to glycosylation.

265
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 22.1 Summary of the major extracellular matrix constituents of not appear to play a signicant role in lung branching [25].
the lung and their localization. Type V collagen has also been localized in the lung. While it
is found near the basement membrane, it is unclear whether
Collagens this minor collagen is part of the basement membrane or
Type I Alveolar, bronchial, vascular walls
solely in the nearby interstitial matrix [26]. The basement
membrane of the lung also contains Type XVIII collagen, a
Type II Bronchial and tracheal cartilage member of the collagen subfamily called MULTIPLEXINs
[27]. These multiplexins have multiple triple helical domains
Type III Co-localizes with Type I collagen and interruptions, hence their name [1]. Cartilage collagens,
Type IV Basement membrane Types II, IX, and XI along with cartilage matrix protein (not
a collagen) are primary components of the trachea [28].
Type V Basement membrane, interstitium Despite the biochemical, immunochemical, ultrastruc-
tural in addition to molecular characterization of collagens
Elastin Alveolar septa, blood vessels, bronchial walls,
in normal lung, their precise function and roles in pulmo-
pleura
nary homeostasis is still not completely elucidated. Studies
Microfibrils Found with elastin, scaffold for elastin continue to determine the cellular inuence of collagens;
deposition their mechanical properties as they relate to lung mechan-
ics, interactions between collagen types and other matrix
Proteoglycans Alveolar walls, basement membrane molecules; kinetics of maturation and turnover; and their
self-assembly. Nevertheless, the extracellular matrix com-
Laminin Basement membrane
ponents of the lung show that they are part of a dynamic
Integrin (receptors for Mesenchymal and epithelial cells structure. When perturbed by environmental assault (eg.
extracellular matrix smoking, industrial polution) or disease (genetic mutations),
molecules) this dynamic balance can be altered and the biomechanical
properties of the lung changed [2].
Fibronectin Basement membrane, interstitium

Elastin
The number of glycosylated residues vary with the dierent
collagen type [14, 15]. Elastin is the main elastic protein in vertebrates and is
Collagen -chains are synthesized as pro--chains responsible for the ability of the lung to recoil following
with non-helical amino and carboxy terminal ends. Cleavage each cycle of expansion and contraction. Thus, like col-
of the propeptides is essential for the proper stacking of lagen, elastin is an integral component of the interstitial
triple helical collagen molecules to form an extracellular matrix of the lung. Given its importance for this elastic
matrix. Amino acid substitutions, especially the invariant resilience, elastin has been localized to the alveoli, pleura,
glycine residues, cause a delay (momentary pause, ie. nano- conducting airways, and vascular tissues [29, 30]. Elastin is
seconds) in formation of the triple helix. This delay leads to secreted as a monomer of so-called tropoelastin, generally
an increase in hydroxylated and glycosylated residues. These, by cells that are of mesenchymal origin. The exquisite abil-
so-called, overmodied collagens are characteristic of a ity of repeated recoil is a unique property of the elastic ber.
number of heritable connective tissue disorders [16]. The ability to undergo repeated rounds of recoil are believed
About 60% of the connective tissue protein mass in to be due to the high hydrophobic amino acid content of
the adult lung is collagen. As expected Type I is the pre- elastin [31]. These hydrophobic amino acids are arranged as
dominant type. The lung also contains a signicant amount to allow cross-links to form that are believed to be critical
of Type III collagen, a major constituent of skin and vas- for its function. Recent studies have also shown the impor-
cular tissues. The Type III bers are thin which allows for tance of optimal iron levels for elastic ber integrity [32]. It
a more compliant tissue. Studies have shown that Type III is important to note that none of these extracellular matrix
collagen is essential for brillogenesis of Type I collagen in elements function in a vacuum. They comprise a character-
normal development [1720]. Types I and III collagen pro- istic architecture and thus a function in an interrelated and
vide the structural framework for the alveolar wall [2]. integrated fashion. Therefore, elastin provides its unique
Type IV collagen is an important constituent of base- function in the context of the collagens, proteoglycans, and
ment membranes [1, 21, 22]. The basement membrane, as microbrils.
visualized by electron microscopy, consists of two layers, the The elastic ber is among the most long-lasting struc-
lamina lucida and lamina densa. In the lung, the basement tures in the body. Studies have shown that humans retain
membrane forms continuously along the bronchial epithelia elastin an entire lifetime [33]. This stability appears to be
from before 12 weeks gestation [23]. In addition to Type IV due to very specic cross-links between elastin mono-
collagen, basement membranes contain other extracellular mers in the mature elastic bers. It is in fact these unique
matrix molecules such as proteoglycans and laminins. The cross-links, formed from lysine residues, that produce novel
Type IV collagen appears to serve as a scaold for binding amino acids, desmosine and isodesmosine, that are markers
of laminin and proteoglycans leading to the formation of for the presence of elastin in tissues.
the basement membrane [24]. During lung branching, the Early structural studies tended to describe elastic
epithelium of the elongating bronchial buds and tubules are bers as having an amorphous appearance. More recent
lined with Type IV collagen. Type III collagen, however, does studies, however, have shown a rather complex structure.
266
Extracellular Matrix 22
Two morphologically and chemically distinct entities have RGD motifs interact with cell surface receptors to mediate
been identied. Elastin, the insoluble polymer composed cell adhesion [49]. These receptors are part of the integrin
of tropoelastin monomers and microbrils. Structure and family, transmembrane proteins that interact with extracel-
function of microbrils are discussed below [34, 35]. lular matrix proteins to anchor cells within the extracellular
Elastin, in its polymerized form, constitutes more than matrix. While the role of elastin in mediating elastic recoil is
90% of the mature elastic ber. The amino acid structure of well established, microbrils too appear to have some elas-
tropoelastin is highly conserved in evolution [36]. In all spe- ticity. In fact, speculation exists that microbrils alone per-
cies studied, tropoelastin has a modular structure of alternat- formed the function of elastic bers prior to the evolution
ing hydrophobic and cross-linking domains. While sequence of tropoelastin. Closely related to brillin-1 is brillin-2.
variation exists, it is the hydrophobicity that is exquisitely Fibrillin-2 shares the domain structure with brillin-1 and
conserved. The conservation appears greater in the cross- in the EGF containing regions are about 80% identical at
linking regions. Unlike the large variety and numerous gene the amino acid level. There are several important dierences
products that encode the collagens, elastin is present as a sin- that may reect diering functional roles. Fibrillin-2 con-
gle gene in a single copy. Interestingly, the various isoforms tains two RGD sites and the domain that is proline rich in
of elastin that are present are produced by alternative splicing brillin-1 is glycine rich in brillin-2 [47, 48, 50].
of a common transcript [3739]. At present, however, the Fibrillin-1 and -2 are dierentially expressed both
functional roles of these isoforms are not understood [40]. temporally and spatially. In most cases, developmental
Mouse studies have shown that animals that lack expression of the brillin genes displays a diphasic pattern.
any elastin expression do not undergo normal alveogen- Thus, expression of brillin-2 occurs earlier in development
esis. Mice with only 50% normal elastin do develop nor- than brillin-1. Studies at the level of mRNA have shown
mal lungs while expression of less than 50% elastin leads FBN2 transcript accumulation prior to tissue dieren-
to abnormal lung development [30]. Signicantly, however, tiation followed by their rapid decrease. FBN1 transcripts
is the fact that injurious stimuli make mice with less than then begin to increase gradually. Studies have shown that
optimal elastin levels more prone to severe lung disease brillin-2 is found preferentially in elastic tissues, such as
[30]. Recent studies have also demonstrated a polymorphic elastic cartilage, tunica media of the aorta, and along the
variant in human elastin causing early onset COPD in a bronchial tree. The two brillins, therefore, may have dif-
family [41]. Severe early onset pulmonary disease has also fering functional roles. It has been hypothesized that bril-
been documented in smokers whose elastin mutation causes lin-2 may have a greater role during early morphogenesis
autosomal dominant cutis laxa [42]. in directing the assembly of elastic bers, while brillin-1 is
mainly responsible for load bearing [5052].
While the brillins are regarded as the major con-
Microfibrils stituents of microbrils, there are additional families of
proteins that are part of the microbrils and the elastic
Elastin-associated microbrils have been classically dened ber. These additional families include the latent trans-
as 1012 nm diameter brils when seen by electron micro- forming growth factor beta binding proteins (LTBPs);
scopy [43]. They were initially identied as structures that microbril-associated glycoproteins (MAGPs) and microbril-
surrounded or were within mature elastic bers. It is now associated proteins (MFAPs); bulins; emilin; Big-H3; and
known that microbrils can also exist without the presence lysyl oxidase. The structural and functional roles of most of
of elastin [44]. Analyses of developing tissues, including the these proteins are not clear. We do know that LTBP-2 is
lung and major blood vessels, have shown that microbrils evolutionarily the closest of the LTBPs to the brillins and
are deposited rst followed by the deposition of elastin [45]. has been isolated from tissue rich in microbrils. MAGP-
Thus, the hypothesis that microbrils act as a scaold for 1 has also been immunolocalized to both elastin-associated
the deposition of elastin to produce mature elastic bers has and naked microbrils. MAGP-1 interacts directly with
emerged. The complete function and, for the matter, con- tropoelastin monomers and may play a primary role in elas-
stituents of the microbrils are presently unknown. tic ber formation (see Ref. [53] for a review).
Ultrastructurally, microbrils display a beads on a Studies in mice have shown that bulin-5 null animals
string structure with a diameter of 8 to 12 nm and are com- have disrupted elastic bers and pulmonary emphysema
posed of several proteins, the most abundant of which is among other phenotypes [54, 55]. Fibulin-5 mutations in
brillin-1 [46]. Fibrillin-1, the product of the FBNI gene, people appear not to be associated with lung abnormalities
is a cysteine rich-glycoprotein with a molecular weight of [56]. The hypothesis that microbrils play a role in TGF-
about 350 kDa. The extracellular domain structure of bril- signaling has lead to uncovering of emphysema in Marfan-
lin-1 is divided into ve distinct regions. The most abundant like mice [57]. In addition, pulmonary abnormalities in sev-
of these domains are calcium binding epidermal growth eral LTBP mouse mutants have also been demonstrated [58,
factor-like (EGF) motifs, that occur some 43 times. Four 59]. The eects of TGF- signaling on lung development
additional EGF motifs that do not bind calcium are also and branching are reviewed in Ref. [60].
present. Interspersed between their calcium binding domains It is apparent that we are just beginning to understand
are seven transforming growth factor 1 binding protein the roles of the extracellular matrix in forming complex tis-
domains each containing eight cysteine residues [47, 48]. sues. The myriad of molecular interactions involved with the
These eight cysteine domains are globular in structure and numerous proteins noted above that are required to build an
interrupt the multiple stretches of the EGF modules that are elastic ber are just being uncovered. The temporal or tissue
believed to form rod-like structures. The middle 8-cysteine specic expression of the various microbrillar components
domains contain an RGD (arginineglycineaspartate) site. is also largely unknown. Mouse studies will be critical for
267
Asthma and COPD: Basic Mechanisms and Clinical Management

the continued progress in elucidating the complex signaling of extracellular matrix molecules is the binding site for
role of the extracellular matrix. integrins [88, 89]. Integrins are expressed early in human
lung development [90, 91]. Animal model manipulation of
some integrins have resulted in reduced bronchial branch-
Proteoglycans ing suggesting a vital role for some integrin dependent
interactions in lung development [86, 88, 92, 93]. Integrins
Proteoglycans comprise a large group of multidomain also function in signaling pathways to mediate migration
core proteins to which glycosaminoglycans are attached. and dierentiation of epithelial cells [9496].
Glycosaminoglycans are unbranched carbohydrate chains
of repeating disaccharide units. Since most are negatively
charged, they bind to other matrix molecules, cell adhesion Fibronectin
molecules, and growth factors [61, 62]. They are integral to
maintain normal pulmonary structure and function. Of the Fibronectin is a widely distributed glycoprotein found in
species of proteoglycans, heparan sulfate and dermatan sul- embryonic tissue that plays a material role in morphogenesis
fate appear to be the most abundant [2, 63]. These proteogly- [97, 98]. Fibronectin plays an important role in cell attach-
cans are also a reservoir for heparin. Chondroitin sulfates, ment [99]. While it has been localized in several regions of
proteoglycans found in cartilagenous tissues, are present in the developing lung, it is primarily seen in regions of airway
bronchioles, while heparan sulfate is the major proteoglycan bifurcation [89]. Fibronectin is co-distributed with collagen
of the gas exchange tissue. Heparin is also found primarily and may be required for normal collagen deposition [100,
in the gas exchange tissues and pleura, but, as expected, not 101]. Its precise role, however, has not yet been elucidated.
the cartilagenous bronchioles [63, 64]. Biochemical studies
over the past quarter century have helped to elucidate the
composition of lung proteoglycans, but their precise func- EXTRACELLULAR MATRIX AND LUNG FUNCTION
tions are still not completely understood.
Heparan sulfate interacts with laminin and appears
to be essential for human development and lung branching The matrix of the normal lung imparts the strength and resil-
[6568]. Studies also demonstrate that proteoglycans are ience for the continuous cycling of inspiration and expiration
important for the function of growth factors. Much of this (Fig 22.1). For proper gas exchange, the components of the
modulation by proteoglycans appears to involve binding and extracellular matrix are distributed in a fashion to reduce the
signaling of TGF and its receptor [6972]. These molecules boundary between erythrocytes and oxygen [26]. When the
may also stimulate production of other matrix constituents or exquisite balance of matrix glycoproteins is disturbed, (by
act as receptors for the extracellular matrix [73, 74]. structural mutations of matrix components or environmen-
Here again mouse targeting of perlecan, a heparan tal damage) the consequences on the lung are manifested
sulfate proteoglycan, have shown that null mice who sur- by pathology. In most cases the assault on the lung leads to
vive embryonic life die perinatally due to respiratory failure brosis. Fibrosis leads to the thickening of the alveolar walls,
[75, 76]. reduction in lung volume, reduced lung elasticity, and fun-
damentally anomalous gas exchange. There are a number
of causes of pulmonary brosis in addition to those due to
Laminin genetic abnormalities of matrix molecules or their modiers
(see below).
Laminins are cross-shaped molecules that comprise several In some cases, pulmonary brosis is a result of some
dierent types and are a part of lung development [1, 77 other primary disease [102]. Examples include infections
79]. Analysis of the developing lung has shown the expres- (viral and fungal) or immune disorders (rheumatoid arthri-
sion of some laminin chains as early as 10 weeks of human tis, scleroderma). As expected, environmental assaults com-
gestation [80]. Laminins are expressed along the basement prise the greatest number of causes of pulmonary brosis.
membrane, but temporal and spatial expression of dierent As one might predict, tobacco smoke causing emphysema is
laminin subunits has been documented [25, 81]. Laminin among the major causes of COPD [103]. Fibrogenic dusts,
expression may play a role in cell dierentiation in the lung such as asbestos, toxins, and chemicals, such as insecticides
as well as other tissues. There is some evidence to suggest and herbicides, are additional causes of brosis in the lung.
that laminin may play a role in alveolar morphogenesis [82]. Iatrogenic causes, such as pharmacotherapy and therapeutic
A lamininheparan sulfate interaction may be essential to irradiation may also lead to pulmonary brosis.
lumen formation and branching morphogenesis [83].

HERITABLE DISORDERS OF CONNECTIVE


Integrins TISSUE
Integrins are cell surface glycoproteins that serve as recep-
tors for the extracellular matrix [8486]. Integrins are Heritable disorders of connective tissue (HDCT) are a
composed of many subtypes all arranged as heterodimers series of disorders caused by mutations in structural or mod-
[85]. These heterodimers can selectively bind several dif- ifying components of the extracellular matrix. Since most
ferent matrix constituents [87]. In fact, the RGD sequence of these gene products are expressed in multiple tissues,

268
Extracellular Matrix 22
pleiotropic manifestations are observed in most HDCT. It Hunter syndrome (MPS II), caused by a deciency in
is important to distinguish HDCT from connective tissue iduronate 2 sulfatase, is characterized by diagnosis prior to
disorders that are autoimmune in nature such as rheuma- age 4, mental retardation, and death before age 15. A mild
toid arthritis and systemic lupus erythematosus. For a com- form has also been described with moderate skeletal and
prehensive review of the HDCT, see Refs [16, 104]. respiratory involvement and survival to adulthood without
Despite the abundance of collagen and elastic bers intellectual impairment. Respiratory complications include
in the lung, the number of HDCT with primary and severe upper airway obstruction, nasal congestion, and thick rhi-
pulmonary disease is rather small. While there are numer- norrhea. As children grow older, pharyngeal hypertrophy,
ous HDCT, for the purposes of this abbreviated review only tongue enlargement, and supraglottic swelling may result in
those disorders caused by defects in genes expressed in the obstructive sleep apnea and death. Upper respiratory infec-
pulmonary system or with pulmonary complications will be tions are common. Abnormalities of the trachea have been
discussed. Osteogenesis imperfecta (OI) comprises several documented. MPS II is an X-linked disorder, thus virtually
disorders of varying severity that are caused by mutations in all eected individuals are male. Pulmonary complications
Type I collagen. In its most severe form, OI Type II, is char- are observed in the majority of individuals with the gamut
acterized by sever bone fragility and stillbirth or neonatal of MPS types.
death. Due to the small chest in these neonates, pulmonary The chondrodysplasias are another group of HDCT
insuciency and failure to ventilate can cause death in the often classied based on radiographic involvement of the
few who are not stillborn. In OI Type III, characterized by long bones. Severe respiratory diculty is present in a
moderate to severe bone fragility, the thorax is often coni- number of these disorders due to a greatly restricted thorax,
cal in shape. Due to the softness of the bones in the chest, which leads to early death. Defects in Type II collagen are
an often lethal, respiratory failure may occur in neonates. well documented in several of these disorders.
Despite these ndings, most individuals with OI, except of The Marfan syndrome (MFS) is a prototypical mem-
course Type II OI, do not have severe pulmonary deciency ber of the HDCT. It is now well known that mutations
even though Type I collagen is abundant in the lung matrix. in the gene encoding brillin-1 cause MFS. Thus, MFS is
The EhlersDanlos syndromes (EDS) are another due to abnormalities of the elastic ber system. Clinically,
series of HDCT. The classic type, previously called Types I MFS is characterized by defects in the cardiovascular, skel-
and II are now known to be caused by mutations in Type etal, and ocular systems. Given the presence of elastic bers
V collagen. The most severe form of EDS is the, so-called, in the lung it is not surprising that pulmonary complica-
vascular type, previously called Type IV EDS. This form of tions also occur. The principal respiratory system abnormal-
EDS is caused by mutations in Type III collagen. ity in MFS is spontaneous pneumothorax, which occurs
Pulmonary complications are rare in EDS. However, in approximately 5% of aected individuals. This indicates
some cases of mediastinal and subcutaneous emphysema that spontaneous pneumothorax is statistically several hun-
and spontaneous pneumothorax have been reported. In dred times more likely in a person with MFS than in the
addition, dilation of the trachea and bronchi has also been general population. In fact, the diagnosis of MFS has often
described in some cases of EDS. been made after an initial event of a spontaneous pneumot-
Pseudoxanthoma elasticum (PXE) is an HDCT horax. Spontaneous pneumothorax may be familial even in
whose molecular pathogenesis has been recently elucidated. the absence of MFS. Apical bullae are also known to occur
PXE is characterized by abnormalities in the skin, eyes, and in MFS and may be a predisposing factor for spontaneous
vasculature. Although the mechanism of the pathogenesis pneumothorax. Emphysema and congenital cystic lung have
remains unclear, the generalized dystrophy of elastic b- also been documented in MFS.
ers does occasionally manifest in the lung. Degenerative Pulmonary function too has been studied in MFS but
changes in the walls of alveoli and miliary mottling of the has been interpreted as essentially normal. In some cases
lungs have been reported. of MFS, severe kyphoscoliosis may be a major contributor
The mucopolysaccharidoses (MPS) are a large group of to pulmonary failure, however, that cannot be attributed to
disorders that are defects in enzymes required in the process- abnormal brillin in the lung, but to the generalized skeletal
ing of glycosaminoglycan molecules (see Proteoglycans). The dysplasia.
large number of dierent enzymatic defects makes a com- Therefore, one can conclude that primary defects in
prehensive review here impossible. Nevertheless, pulmonary extracellular matrix proteins or their modifying enzymes
complications have been observed in several MPS. For exam- can cause an array of pulmonary disease. However, even
ple, Hurler syndrome (MPS I H) is due to defects in -l- when the pulmonary abnormality leads to premature death,
iduronidase. Hurler syndrome is characterized by diagnosis the manifestations in other organ systems are often more
prior to age 2 early corneal clouding, kyphoscoliosis, men- prominent (Table 22.2).
tal retardation, and death by age 10. All individuals with
Hurler syndrome experience severe respiratory problems.
Accumulation of glycosaminoglycans in the oropharyngeal
trachea leads to airway obstruction. Radical pharyngoplasty EXTRACELLULAR MATRIX IN ASTHMA
provides some temporary relief, but obstruction recurs. These
individuals are also prone to repeated upper respiratory infec-
AND COPD
tions. Deformity of the thorax and abnormalities of bronchial
cartilage contributes to reduced chest expansion and decreased Alterations in connective tissue likely play key roles in
vital capacity. Bronchopneumonia is a frequent cause of death. the pathogenesis of both asthma and COPD [105107].

269
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 22.2 Pulmonary manifestations of primary matrix abnormalities. Destruction of elastin is thought to be a major feature in
the development of emphysema. Exposure of the lung to
Matrix Pulmonary enzymes with elastolytic activity results in emphysema in
element Disorder abnormality animals [108]. Histologic examination of the emphyse-
matous lung shows disrupted elastic bers [109]. Urinary
Type I collagen Osteogenesis Pulmonary
excretion of desmosine, a specic marker for degradation
imperfecta (various insufficiency;
of elastin, has been observed in smokers and former smok-
types) Pulmonary
ers with COPD. Smokers without COPD did not excrete
hypertension
desmosine [110, 111]. Because individuals with severe de-
Type III collagen EhlersDanlos Spontaneous ciency of -1-protease inhibitor, an inhibitor of several
(vascular type) pneumothorax; serine proteases, are at markedly increased risk to develop
hemoptysis emphysema [112], these proteases have been thought to
have a major role in the destruction of elastin. Recent stud-
Elastin Dominant cutis laxa Bronchiectasis; ies, however, suggest that the matrix metalloproteases con-
emphysema tribute to the development of emphysema [113, 114].
Fibulin-5 Recessive cutis laxa Emphysema
In addition to tissue destruction, there is evidence
of excess deposition of extracellular matrix (i.e. brosis in
Fibrillin-1 Marfan syndrome Pneumothorax; COPD). In addition to the destruction of elastin in emphy-
pulmonary blebs; sema, there is an increase in deposition of collagen [115
emphysema 117]. Fibrosis of the small airways is also a regular feature of

(A)

Bronchiole

Arter
y

Alveolar spaces

(B)
Epithelium

Basemembrane
type IV collagen
Basementmembrane
type IV collagen

Endothelium

Bronchiole
Dots represent sea
of proteoglycans

Trible helical
Elastic fibers with collagen
microfibrillar scaffold

FIG. 22.1 (A) Photomicrograph of normal H&E stained lung (original magnification 100 ). Various part are labeled in the figure. (B) An artists rendition of
a bronchiole. The outer (epithelium) and inner (endothelium) cellular layers surround the extracellular matrix containing triple helical collagen, elastic fibers
with their associated microfibrils, and proteoglycans.

270
Extracellular Matrix 22
chronic bronchitis [118] and is related to the development 11. Bourbon J, Boucherat O, Chailley-Heu B et al. Control mechanisms
of airow limitation [119, 120]. The airways in COPD are of lung alveolar development and their disorders in bronchopulmonary
of smaller than expected diameter; this may be a direct con- dysplasia. Pediatr Res 57: 38R46R, 2005.
12. Garcia CS, Prota LF, Morales MM et al. Understanding the mecha-
sequence of this brotic process [120].
nisms of lung mechanical stress. Braz J Med Biol Res 39: 697706, 2006.
Extracellular matrix alterations have also been docu- 13. Ricard-Blum S, Ruggiero F. The collagen superfamily: From the extracel-
mented in asthma [121]. For example, the connective tis- lular matrix to the cell membrane. Pathol Biol (Paris) 53: 43042, 2005.
sue in the large airways, include thickening of the lamina 14. Bornstein P, Sage H. Structurally distinct collagen types. Annu Rev
reticularis with increased deposition of collagen types I, III, Biochem 1003: 410039957, 1980.
and bronectin [122]; tenascin [123]; and fragmentation 15. Burgeson RE, Morris NP. The collagen family of proteins. In: Uitto J,
of the airway elastic ber network [124, 125]. The thick- Perejda AJ (eds). Connective Tissue Disease: Molecular Pathology of the
ened lamina reticularis is particularly striking as the epithe- Extracellular Matrix, pp. 328. New York: Marcel Dekker, Inc, 1987.
lium can be seen to rest on a dense nearly acellular layer of 16. McKusicks Heritable Disorders of Connective Tissue, P. Beighton, ed. 5th
apparently homogeneous connective tissue. Whether these edn, pp. 1748. St. Louis: Mosby, 1993.
17. Bienskowski RS. Interstitial collagens. In: Crystal RG, West JB (eds).
changes are altered with therapy of asthma is controversial
The Lung, pp. 38188. New York: Raven Press, 1991.
[126129], but the connective tissue changes may occur 18. Bradley K, McConnell Breul S, Crystal RG. Lung collagen heteroge-
somewhat independently of measures of clinical severity neity. Proc Natl Acad Sci USA 71: 282832, 1974.
[127, 130]. The functional signicance of the altered con- 19. Bradley KH, McConnell SD, Crystal RG. Lung collagen composition
nective tissue in asthma likely leads to compromised lung and synthesis: Characterization and changes with age. J Biol Chem 249:
function [106]. Similar changes in the small airways may 267483, 1974.
account for increased peripheral airway resistance, reduced 20. Hay ED. Cell Biology of the Extracellular Matrix, 2nd edn.
airway compliance [131], and progressive airow limita- New York: Plenum Press, 1991.
tion in asthma [132, 133]. Importantly, growth factors are 21. Maina JN, West JB. Thin and strong! The bioengineering dilemma in
critical for the extracellular matrix remodeling in asthma the structural and functional design of the bloodgas barrier. Physiol
Rev 85: 81144, 2005.
[134, 135].
22. Yurchenco PD, Furthmayr H. Self-assembly of basement membrane
Recognizing the complexity of the extracellular collagen. Biochemistry 23: 183950, 1984.
matrix, it seems likely that many alterations in these struc- 23. Lallemand AV, Ruocco SM, Gaillard DA. Expression and immunohis-
tural molecules will contribute to the pathophysiology of tochemical localization of laminin and type IV collagen in developing
asthma and COPD. It seems likely as well that the many human fetal tracheal glands. Int J Dev Biol 37: 49195, 1993.
genetic variations in connective tissue metabolism, which 24. Leblond CP, Inoue S. Structure, composition, and assembly of base-
are beginning to be characterized, will contribute to the het- ment membrane. Am J Anat 185: 36790, 1989.
erogeneity observed in asthma and COPD. Understanding 25. Virtanen I, Laitinen A, Tani T et al. Dierential expression of laminins
the genetic milieu will undoubtedly help to predict those at and their integrin receptors in developing and adult human lung. Am J
greatest risk [4, 30, 136]. Respir Cell Mol Biol 15: 18496, 1996.
26. Clark JG, Kuhn C3, McDonald JA et al. Lung connective tissue. Int
Rev Connect Tissue Res 331: 10249331, 1983.
27. Saarela J, Rehn M, Oikarinen A et al. The short and long forms of type
XVIII collagen show clear tissue specicities in their expression and
References location in basement membrane zones in humans. Am J Pathol 153:
61126, 1998.
1. Ayad S, Boot-Handford RP, Humphries MJ et al. The Extracellular 28. Kelley J. Collagen. In: Massaro D (ed.), Lung Cell Biology, pp. 82166.
Matrix: Facts Book. . New York: Academic Press, 1998. New York: Marcel Dekker, 1989.
2. Suki B, Ito S, Stamenovic D et al. Biomechanics of the lung paren- 29. Foster JA, Rich CB, Curtiss SW et al. Elastin. In: Massaro D (ed.),
chyma: Critical roles of collagen and mechanical forces. J Appl Physiol Lung Cell Biology, pp. 867905. New york: Marcel Dekker, 1989.
98: 189299, 2005. 30. Shifren A, Mecham RP. The stumbling block in lung repair of emphy-
3. Chapman HA. Disorders of lung matrix remodeling. J Clin Invest 113: sema: Elastic ber assembly. Proc Am Thorac Soc 3: 42833, 2006.
14857, 2004. 31. Sandberg LB, Soskel NT, Leslie JG. Elastin structure, biosynthesis, and
4. Fixman ED, Stewart A, Martin JG. Basic mechanisms of development relation to disease states. N Engl J Med 304: 56679, 1981.
of airway structural changes in asthma. Eur Respir J 29: 37989, 2007. 32. Hill CH, Ashwell CM, Nolin SJ et al. Dietary iron deciency compro-
5. Aszodi A, Legate KR, Nakchbandi I et al. What mouse mutants teach mises normal development of elastic bers in the aorta and lungs of
us about extracellular matrix function. Annu Rev Cell Dev Biol 22: chicks. J Nutr 137: 1895900, 2007.
591621, 2006. 33. Shapiro SD, Endicott SK, Province MA et al. Marked longevity of
6. Warburton D, Gauldie J, Bellusci S et al. Lung development and sus- human lung parenchymal elastic bers deduced from prevalence of
ceptibility to chronic obstructive pulmonary disease. Proc Am Thorac Soc D-aspartate and nuclear weapons-related radiocarbon. J Clin Invest 87:
3: 66872, 2006. 182834, 1991.
7. Hersh CP, DeMeo DL, Lazarus R et al. Genetic association analysis of 34. Brown PL, Mecham L, Tisdale C et al. The cysteine residues in the car-
functional impairment in chronic obstructive pulmonary disease. Am J boxy terminal domain of tropoelastin form an intrachain disulde bond
Respir Crit Care Med 173: 97784, 2006. that stabilizes a loop structure and positively charged pocket. Biochem
8. Joos L, He JQ, Shepherdson MB et al. The role of matrix metallopro- Biophys Res Commun 186: 54955, 1992.
teinase polymorphisms in the rate of decline in lung function. Hum 35. Mecham RP, Heuser J. Three-dimensional organization of extracellular
Mol Genet 11: 56976, 2002. matrix in elastic cartilage as viewed by quick freeze, deep etch electron
9. Kaartinen V, Warburton D. Fibrillin controls TGF-beta activation. Nat microscopy. Connect Tissue Res 24: 8393, 1990.
Genet 33: 33132, 2003. 36. Boyd CD, Christiano AM, Pierce RA et al. Mammalian tropoelastin:
10. Barnes PJ. Mediators of chronic obstructive pulmonary disease. Multiple domains of the protein dene an evolutionarily divergent
Pharmacol Rev 56: 51548, 2004. amino acid sequence. Matrix 11: 23541, 1991.

271
Asthma and COPD: Basic Mechanisms and Clinical Management

37. Heim RA, Pierce RA, Deak SB et al. Alternative splicing of rat tropoe- 60. Roth-Kleiner M, Post M. Similarities and dissimilarities of branching and
lastin mRNA is tissue-specic and developmentally regulated. Matrix septation during lung development. Pediatr Pulmonol 40: 11334, 2005.
11: 35966, 1991. 61. Ruoslahti E. Structure and biology of proteoglycans. Annu Rev Cell
38. Indik Z, Yeh H, Ornstein-Goldstein N et al. Structure of the elastin Biol: 422955, 1988.
gene and alternative splicing elastin mRNA. In: Sandell L, Boyd CD 62. Ruoslahti E. Proteoglycans in cell regulation. J Biol Chem 264: 13369
(eds). Extracellular Matrix Genes, pp. 22150. New York: Academic 72, 1989.
Press, 1990. 63. Radhakrishnamurthy B, Berenson SG. Proteoglycans of the lung. In:
39. Parks WC, Secrist H, Wu LC et al. Developmental regulation of Massaro D (ed.), Lung Cell Biology, pp. 9811010. New York: Marcel
tropoelastin isoforms. J Biol Chem 263: 441623, 1988. Dekker, Inc, 1989.
40. Parks WC, Deak SB. Tropoelastin heterogeneity: Implications for 64. Hance AJ, Crystal RG. The connective tissue of lung. Am Rev Respir
protein function and disease. Am J Respir Cell Mol Biol 2: 399406, 1990. Dis 112: 657711, 1975.
41. Kelleher CM, Silverman EK, Broekelmann T et al. A functional muta- 65. Schuger L, O
Shea KS, Nelson BB et al. Organotypic arrangement
tion in the terminal exon of elastin in severe, early-onset chronic of mouse embryonic lung cells on a basement membrane extract:
obstructive pulmonary disease. Am J Respir Cell Mol Biol 33: 35562, Involvement of laminin. Development 110: 109199, 1990.
2005. 66. Schuger L, Skubitz AP, Gilbride K et al. Laminin and heparan sulfate
42. Urban Z, Gao J, Pope FM et al. Autosomal dominant cutis laxa with proteoglycan mediate epithelial cell polarization in organotypic cultures
severe lung disease: Synthesis and matrix deposition of mutant tropoe- of embryonic lung cells: Evidence implicating involvement of the inner
lastin. J Invest Dermatol 124: 119399, 2005. globular region of laminin beta 1 chain and the heparan sulfate groups
43. Low FN. Microbrils: Fine lamentous components of the tissue of heparan sulfate proteoglycan. Dev Biol 179: 26473, 1996.
space. Anat Rec 142: 13137, 1962. 67. Schuger L, Skubitz AP, OShea KS et al. Identication of laminin
44. Streeten BW, Licari PA, Marucci AA et al. Immunohistochemical domains involved in branching morphogenesis: Eects of anti-laminin
comparison of ocular zonules and the microbrils of elastic tissue. monoclonal antibodies on mouse embryonic lung development. Dev
Invest Ophthalmol Vis Sci 21: 13035, 1981. Biol 146: 53141, 1991.
45. Cleary EG. The microbrillar component of the elastic bers: 68. Thompson SM, Connell MG, Fernig DG et al. Novel phage display
Morphology and biochemistry. In: Uitto J, Perejda AJ (eds). Connective antibodies identify distinct heparan sulfate domains in developing
Tissue Disease: Molecular Pathology of the Extracellular Matrix, pp. 5581. mammalian lung. Pediatr Surg Int 23: 41117, 2007.
New York: Marcel Dekker, 1987. 69. Border WA, Ruoslahti E. Transforming growth factor-beta in disease:
46. Sakai LY, Keene DR, Engvall E. Fibrillin, a new 350-kD glycoprotein, is The dark side of tissue repair. J Clin Invest 90: 17, 1992.
a component of extracellular microbrils. J Cell Biol 103: 2499509, 1986. 70. Hildebrand A, Romaris M, Rasmussen LM et al. Interaction of the
47. Lee B, Godfrey M, Vitale E et al. Linkage of Marfan syndrome and a small interstitial proteoglycans biglycan, decorin and bromodulin with
phenotypically related disorder to two dierent brillin genes. Nature transforming growth factor beta. Biochem J 302: 34, 1994.
352: 33034, 1991. 71. Lopez Casillas F, Wrana JL, Massague J. Betaglycan presents ligand to
48. Pereira L, D
Alessio M, Ramirez F et al. Genomic organization of the the TGF beta signaling receptor. Cell 73: 143544, 1993.
sequence coding for brillin, the defective gene product in Marfan syn- 72. Yamaguchi Y, Mann DM, Ruoslahti E. Negative regulation of trans-
drome. Hum Mol Genet 2: 96168, 1993. forming growth factor-beta by the proteoglycan decorin. Nature 346:
49. Sakamoto H, Broekelmann T, Cheresh DA et al. Cell-type specic rec- 28184, 1990.
ognition of RGD- and non-RGD-containing cell binding domains in 73. Berneld M, Kokenyesi R, Kato M et al. Biology of the syndecans: A
brillin-1. J Biol Chem 271: 491622, 1996. family of transmembrane heparan sulfate proteoglycans. Annu Rev Cell
50. Zhang H, Apfelroth SD, Hu W et al. Structure and expression of Biol 93: 836593, 1992.
brillin-2 a novel microbrillar component preferentially located in 74. Mast BA, Diegelmann RF, Krummel TM et al. Hyaluronic acid mod-
elastic matrices. J Cell Biol 124: 85563, 1994. ulates proliferation, collagen and protein synthesis of cultured fetal
51. Mariencheck MC, Davis EC, Zhang H et al. Fibrillin-1 and brillin- broblasts. Matrix 13: 44146, 1993.
2 show temporal and tissue-specic expression in developing elastic 75. Arikawa-Hirasawa E, Watanabe H, Takami H et al. Perlecan is essen-
tissues. Connect Tissue Res 31: 8797, 1995. tial for cartilage and cephalic development. Nat Genet 23: 35458,
52. Zhang H, Hu W, Ramirez F. Developmental expression of brillin 1999.
genes suggests heterogeneity of extracellular microbrils. J Cell Biol 76. Costell M, Gustafsson E, Aszodi A et al. Perlecan maintains the integ-
129: 116576, 1995. rity of cartilage and some basement membranes. J Cell Biol 147: 1109
53. Robinson PN, Godfrey M. The molecular genetics of Marfan syndrome 22, 1999.
and related microbrillopathies. J Med Genet 37: 925, 2000. 77. Burgeson RE, Chiquet M, Deutzmann R et al. A new nomenclature
54. Nakamura T, Lozano PR, Ikeda Y et al. Fibulin-5/DANCE is essential for the laminins. Matrix Biol 14: 20911, 1994.
for elastogenesis in vivo. Nature 415: 17175, 2002. 78. Nguyen NM, Senior RM. Laminin isoforms and lung development:
55. Yanagisawa H, Davis EC, Starcher BC et al. Fibulin-5 is an elastin- All isoforms are not equal. Dev Biol 294: 27179, 2006.
binding protein essential for elastic bre development in vivo. Nature 79. Timpl R, Rohde H, Robey PG et al. Laminin a glycoprotein from
415: 16871, 2002. basement membranes. J Biol Chem 254: 993337, 1979.
56. Hu Q , Loeys BL, Coucke PJ et al. Fibulin-5 mutations: Mechanisms 80. Lallemand AV, Ruocco SM, Gaillard DA. Synthesis and expression of
of impaired elastic ber formation in recessive cutis laxa. Hum Mol laminin during human foetal lung development. Anat Rec 242: 23341,
Genet 15: 337986, 2006. 1995.
57. Neptune ER, Frischmeyer PA, Arking DE et al. Dysregulation of 81. Uehara Y, Minowa O, Mori C et al. Placental defect and embryonic
TGF-beta activation contributes to pathogenesis in Marfan syndrome. lethality in mice lacking hepatocyte growth factor/scatter factor. Nature
Nat Genet 33: 40711, 2003. 373: 7025, 1995.
58. Colarossi C, Chen Y, Obata H et al. Lung alveolar septation defects in 82. Kouretas D, Karinch AM, Rishi A et al. Conservation analysis of rat
LTBP-3-null mice. Am J Pathol 167: 41928, 2005. and human SP-A gene identies 5
anking sequences of rat SP-A
59. Sterner-Kock A, Thorey IS, Koli K et al. Disruption of the gene encoding that bind rat lung nuclear proteins. Exp Lung Res 19: 485503, 1993.
the latent transforming growth factor-beta binding protein 4 (LTBP- 83. Schuger L, O
Shea S, Rheinheimer J et al. Laminin in lung develop-
4) causes abnormal lung development, cardiomyopathy, and colorectal ment: Eects of anti-laminin antibody in murine lung morphogenesis.
cancer. Genes Dev 16: 226473, 2002. Dev Biol 137: 2632, 1990.

272
Extracellular Matrix 22
84. Albelda SM, Buck CA. Integrins and other cell adhesion molecules. 110. Gottlieb DJ, Stone PJ, Sparrow D et al. Urinary desmosine excre-
FASEB J 4: 286880, 1990. tion in smokers with and without rapid decline of lung function: The
85. Hynes RO. Integrins: Versatility, modulation, and signaling in cell Normative Aging Study. Am J Respir Crit Care Med. 154: 129095,
adhesion. Cell 69: 1125, 1992. 1996.
86. Wu JE, Santoro SA. Dierential expression of integrin alpha subunits 111. Stone PJ, Gottlieb DJ, OConnor GT et al. Elastin and collagen degra-
supports distinct roles during lung branching morphogenesis. Dev dation products in urine of smokers with and without chronic obstruc-
Dyn 206: 16981, 1996. tive pulmonary disease. Am J Respir Crit Care Med 151: 95259, 1995.
87. Glukhova MA, Koteliansky VE. Integrins, cytoskeletal and extracellu- 112. Laurell CB, Eriksson S. The electrophoretic alpha 1-globulin pattern
lar matrix proteins in developing smooth muscle cells of human aorta. of serum in alpha 1-antitrypsin deciency. Scand J Clin Lab Invest 15:
In: Schwartz SM, Mecham RP (eds). The Vascular Smooth Muscle Cell, 13240, 1963.
pp. 3779. San Diego: Academic Press, 1995. 113. DArmiento J, Dalal SS, Okada Y et al. Collagenase expression in the
88. Roman J, Little CW, McDonald JA. Potential role of RGD-binding lungs of transgenic mice causes pulmonary emphysema. Cell 71: 955
integrins in mammalian lung branching morphogenesis. Development 61, 1992.
112: 55158, 1991. 114. Hautamaki RD, Kobayashi DK, Senior RM et al. Requirement for
89. Roman J, McDonald JA. Expression of bronectin, the integrin alpha macrophage elastase for cigarette smoke-induced emphysema in mice.
5, and alpha-smooth muscle actin in heart and lung development. Am Science 277: 20024, 1997.
J Respir Cell Mol Biol 6: 47280, 1992. 115. Finlay GA, ODonnell MD, OConnor CM et al. Elastin and collagen
90. Coraux C, Delplanque A, Hinnrasky J et al. Distribution of integrins remodeling in emphysema. A scanning electron microscopy study. Am
during human fetal lung development. J Histochem Cytochem 46: 803 J Pathol 149: 140515, 1996.
10, 1998. 116. Lang MR, Fiaux GW, Gillooly M et al. Collagen content of alveolar
91. Sheppard D. Functions of pulmonary epithelial integrins: From devel- wall tissue in emphysematous and non-emphysematous lungs. Thorax
opment to disease. Physiol Rev 83: 67386, 2003. 49: 31926, 1994.
92. Kreidberg JA, Donovan MJ, Goldstein SL et al. Alpha 3 beta 117. Pierce JA, Hocott JB, Ebert RV. The collagen and elastin content of
1 integrin has a crucial role in kidney and lung organogenesis. the lung in emphysema. Ann Intern Med 55: 21021, 1961.
Development 122: 353747, 1996. 118. Cosio M, Ghezzo H, Hogg JC et al. The relations between structural
93. Sheppard D. Roles of alphav integrins in vascular biology and pulmo- changes in small airways and pulmonary-function tests. N Engl J Med
nary pathology. Curr Opin Cell Biol 16: 55257, 2004. 298: 127781, 1978.
94. Caniggia I, Liu J, Han R et al. Identication of receptors bind- 119. Finkelstein R, Ma HD, Ghezzo H et al. Morphometry of small air-
ing bronectin and laminin on fetal rat lung cells. Am J Physiol 270: ways in smokers and its relationship to emphysema type and hyper-
L45968, 1996. responsiveness. Am J Respir Crit Care Med 152: 26776, 1995.
95. Clark EA, Brugge JS. Integrins and signal transduction pathways: The 120. Kuwano K, Bosken CH, Pare PD et al. Small airways dimensions in
road taken. Science 268: 23339, 1995. asthma and in chronic obstructive pulmonary disease. Am Rev Respir
96. Sheppard D. Epithelial integrins. Bioessays 18: 65560, 1996. Dis 148: 122025, 1993.
97. Hynes RO, Yamada KM. Fibronectins: Multifunctional modular glyc- 121. Stenmark KR, Davie N, Frid M et al. Role of the adventitia in pulmo-
oproteins. J Cell Biol 95: 36977, 1982. nary vascular remodeling. Physiology (Bethesda) 21: 13445, 2006.
98. Murphy-Ullrich JE, Mosher DF. Fibronectin and disease processes. 122. Roche WR, Beasley R, Williams JH et al. Subepithelial brosis in the
In: Uitto J, Perejda AJ (eds). Connective Tissue Disease: Molecular bronchi of asthmatics. Lancet 1: 52024, 1989.
Pathology of the Extracellular Matrix, pp. 45573. New York: Marcel 123. Laitinen A, Altraja A, Kampe M et al. Tenascin is increased in airway
Dekker, Inc, 1987. basement membrane of asthmatics and decreased by an inhaled ster-
99. Ruoslahti E. Fibronectin and its receptors. Annu Rev Biochem: 57375 oid. Am J Respir Crit Care Med 156: 95158, 1997.
413, 1988. 124. Bousquet J, Lacoste JY, Chanez P et al. Bronchial elastic bers in nor-
100. Furie MB, Frey AB, Rifkin DB. Location of a gelatin-binding region mal subjects and asthmatic patients. Am J Respir Crit Care Med 153:
of human plasma bronectin. J Biol Chem 255: 439194, 1980. 164854, 1996.
101. Hahn LH, Yamada KM. Identication and isolation of a collagen- 125. Mauad T, Xavier AC, Saldiva PH et al. Elastosis and fragmentation
binding fragment of the adhesive glycoprotein bronectin. Proc Natl of bers of the elastic system in fatal asthma. Am J Respir Crit Care
Acad Sci USA 76: 116063, 1979. Med 160: 96875, 1999.
102. Clark JG. The molecular pathology of pulmonary brosis. In: Uitto J, 126. Hoshino M, Nakamura Y, Sim JJ et al. Inhaled corticosteroid reduced
Perejda AJ (eds). Connective Tissue Disease: Molecular Pathology of the lamina reticularis of the basement membrane by modulation of insu-
Extracellular Matrix. New York: Marcel Dekker, Inc, 1987. lin-like growth factor (IGF)-I expression in bronchial asthma. Clin
103. Rennard SI, Togo S, Holz O. Cigarette smoke inhibits alveolar repair: Exp Allergy 28: 56877, 1998.
A mechanism for the development of emphysema. Proc Am Thorac Soc 127. Laitinen LA, Laitinen A, Altraja A et al. Bronchial biopsy ndings
3: 7038, 2006. in intermittent or early asthma. J Allergy Clin Immunol 98: S36,
104. Connective Tissue and its Heritable Disorders Molecular, Genetic, and 1996.
Medical Aspects, B. Steinmann, ed. pp. 1709. New York: Wiley-Liss, 1993. 128. Olivieri D, Chetta A, Del Donno M et al. Eect of short-term treat-
105. Davidson W, Bai TR. Lung structural changes in chronic obstructive ment with low-dose inhaled uticasone propionate on airway inam-
pulmonary diseases. Curr Drug Targets Inamm Allergy 4: 64349, 2005. mation and remodeling in mild asthma: A placebo-controlled study.
106. Pascual RM, Peters SP. Airway remodeling contributes to the pro- Am J Respir Crit Care Med 155: 186471, 1997.
gressive loss of lung function in asthma: An overview. J Allergy Clin 129. Trigg CJ, Manolitsas ND, Wang J et al. Placebo-controlled immun-
Immunol 116: 47786, 2005. opathologic study of four months of inhaled corticosteroids in asthma.
107. Postma DS, Timens W. Remodeling in asthma and chronic obstruc- Am J Respir Crit Care Med 150: 1722, 1994.
tive pulmonary disease. Proc Am Thorac Soc 3: 43439, 2006. 130. Laitinen LA, Altraja A, Karjalainen E-M et al. Early interventions
108. Snider GL, Lucey EC, Stone PJ. Animal models of emphysema. Am in asthma with inhaled corticosteroids. J Allergy Clin Immunol 105:
Rev Respir Dis 133: 14969, 1986. S582585, 2000.
109. Fukuda Y, Masuda Y, Ishizaki M et al. Morphogenesis of abnormal 131. Wagner EM, Liu MC, Weinmann GG et al. Peripheral lung resistance
elastic bers in lungs of patients with panacinar and centriacinar in normal and asthmatic subjects. Am Rev Respir Dis 141: 58488,
emphysema. Hum Pathol 20: 65259, 1989. 1990.

273
Asthma and COPD: Basic Mechanisms and Clinical Management

132. Lange P, Parner J, Vestbo J et al. A 15-year follow-up study of ventila- 135. Burgess JK. Connective tissue growth factor: A role in airway remod-
tory function in adults with asthma. N Engl J Med 339: 1194200, elling in asthma? Clin Exp Pharmacol Physiol 32: 98894, 2005.
1998. 136. Brody JS, Spira A. State of the art. Chronic obstructive pulmonary
133. Peat JK, Woolcock AJ, Cullen K. Rate of decline of lung function in disease, inammation, and lung cancer. Proc Am Thorac Soc 3: 53537,
subjects with asthma. Eur J Respir Dis 70: 17179, 1987. 2006.
134. Boxall C, Holgate ST, Davies DE. The contribution of transforming
growth factor-beta and epidermal growth factor signalling to airway
remodelling in chronic asthma. Eur Respir J 27: 20829, 2006.

274
Inflammatory
4
PART

Mediators and
Pathways
Prostanoids
23 CHAPTER

2. cycloxygenase-2 (COX-2), which is mainly


ARACHIDONIC ACID METABOLISM an inducible form, although also to some
Paul M. OByrne
extent presents constitutively in the CNS, in Firestone Institute for Respiratory
The release of arachidonic acid from cell mem- the juxtaglomerular apparatus of the kidney, Health, St. Josephs Healthcare and
brane phospholipids, through the action of a in the lung, in platelets, and in the placenta Department of Medicine, McMaster
family of phospholipases, can result in the pro- during late gestation. University, Hamilton, ON, Canada
duction of a wide variety of mediators which Both isoforms contribute to the inam-
may be relevant in the pathogenesis of asthma. matory process, but COX-2 is induced during
These lipid mediators have traditionally been acute and chronic inammation, resulting in an
considered in two classes: enhanced formation of prostaglandins. Some
1. mediators which result from the action of the studies have indicated that associations between
cycloxygenases on arachidonic acid, which are COX-2 polymorphisms, prostaglandin produc-
prostaglandins (PG) or thromboxane (Tx) tion, and the development of asthma and atopy
[1], while there is evidence that COX-1 poly-
2. mediators which result from the action of morphisms do not appear to play a substantial
the enzymes 5-, 12-, or 15-lipoxygenase on role in genetic pre-disposition for asthma or
arachidonic acid, which are the leukotrienes asthma severity [2].
(LT) and lipoxins (Lx). Prostaglandins and thromboxane mediate
Platelet-activating factor (PAF) has also their eects through activation of specic recep-
been recognized to be a mediator formed dur- tors, and there is cross-activation of these receptors
ing arachidonic acid metabolism. by the dierent agonists. The receptor designation
The oxidative metabolism of arachidonic has been accepted as the most potent agonist fol-
acid by cycloxygenases produces the cyclic endo- lowed by the term prostanoid. Thus, the throm-
peroxides PGG2 and PGH2 (Fig. 23.1). The boxane receptor is designated the TP receptor,
subsequent action of prostaglandin isomerases and the PGE receptor is the EP receptor. There
produces either PGD2 or PGE2, reductive cleav- are DP, EP, FP, and TP receptors; the EP recep-
age produces PGF2a, while one of two termi- tors are subdivided into EP1EP4 [3].
nal synthetases on the endoperoxide produces
PGI2 and TxA2. Cycloxygenase appears to be
present in most cells; however, the cycloxygen-
ase metabolite(s) released from a particular cell ROLE IN ASTHMA
are quite specic (e.g. TxA2 from platelets, and
PGI2 from endothelial cells). This suggests that
terminal synthetases are cell-specic. All of the cycloxygenase products of arachi-
At least two isoforms of cycloxygenase donic acid metabolism have been prepared by
have been identied: total chemical synthesis and, with the exception
of thromboxane, are readily available for study.
1. cycloxygenase-1 (COX-1), which is Thromboxane has an exceedingly short half-
constitutively present mainly in the gastric life (about 30 s) and studies with thromboxane
mucosa, kidney and platelets have been limited to a few, very circumscribed,

277
Asthma and COPD: Basic Mechanisms and Clinical Management

Cell membrane Phospholipases

Lyso-PAF Arachidonic acid

Cycloxygenase 1 or 2 5-lipoxygenase 15-lipoxygenase

PGG2 5-HPETE 15-HPETE

PGH2
LTA4 Lipoxins
PGD2 TxA2
LTB4
PGE2 LTC4
PGI2
PGF2
LTD4
TxB2
9 11 PGF2
LTE4 FIG. 23.1 The spectrum of eicosanoids produced
6-keto PGF2 as a consequence of arachidonic acid metabolism.

experimental preparations, none of them in the airways. the airways after inhaled ozone; however, a cycloxygenase
Fortunately, several stable thromboxane mimetics have product was released during the inammatory response which
been developed. These are endoperoxides which activate the caused airway hyperresponsiveness. Subsequently, a reputed
thromboxane receptor and mimic the biological actions of combined cycloxygenase and lipoxygenase inhibitor,
thromboxane but have substantially longer half-lives. BW755C, was also demonstrated to prevent the development
Prostaglandins are believed to have a variety of eects of airway hyperresponsiveness after inhaled ozone in dogs [7].
on airway function in asthma. The prostaglandins are most Inhibition of cycloxygenase by indomethacin also prevents
easily considered in two classes. There are stimulatory pros- the development of airway hyperresponsiveness in other spe-
taglandins, such as PGD2, PGF2a, and TxA2, which are cies including that which occurs after C5a des Arg exposure
potent bronchoconstrictors, and inhibitory prostagland- in rabbits [8] and following inhaled allergen in sheep [9].
ins, such as PGE2, which can reduce bronchoconstrictor Cycloxygenase products have been implicated in the
responses and attenuate the release of bronchoconstrictor pathogenesis of allergen-induced early asthmatic as well as
mediators, such as acetylcholine, from airway nerves. late asthmatic responses in human subjects. This has been
Evidence has been obtained in both animal models done by pretreating subjects with several dierent cycloxy-
of airway hyperresponsiveness [4] and in human subjects genase inhibitors. One study reported that pretreatment
with asthma that cycloxygenase metabolites are involved with indomethacin inhibited the late response, without hav-
in causing aspirin-induced bronchoconstriction [5], or ing a major eect on the early response [10]. Another study,
bronchoconstriction after exercise [6]. There is, however, however, where subjects were pretreated with indomethacin,
little convincing evidence that cycloxygenase metabolites could not conrm these observations on allergen-induced
are important in causing the ongoing, persisting airway either early responses or late responses [11]. However, in
hyperresponsiveness that is characteristic of asthma. This is this latter study, indomethacin resulted in a signicant
because several studies have failed to demonstrate any eect inhibition of the development of allergen-induced airway
of cycloxygenase inhibitors on stable airway hyperrespon- hyperresponsiveness, which suggests that a cycloxygenase
siveness in asthmatic subjects. product is involved in the pathogenesis of this response.
The initial studies examining the role of cycloxygenase
metabolites in the pathogenesis of transient airway hyper-
responsiveness after an inammatory stimulus were carried
out using a cycloxygenase inhibitor, indomethacin, in dogs STIMULATORY PROSTAGLANDINS
in whom airway hyperresponsiveness had been induced by
exposure to inhaled ozone [4]. Indomethacin did not alter
baseline airway responsiveness to inhaled acetylcholine, Prostaglandin D2
but did prevent the development of airway hyperrespon-
siveness after inhaled ozone. Despite the absence of airway PGD2 is known to be released from stimulated dispersed
hyperresponsiveness, the magnitude of the inammatory human lung cells in vitro and from the airways of allergic
response, as measured by the numbers of neutrophils in the human subjects which have been stimulated by allergen
airway epithelium, was not altered by indomethacin. This [12]. PGD2 synthase is present in mast cells, T helper 2
suggested that a cycloxygenase product was not respon- (TH2) cells and other leukocytes. PGD2 is rapidly metab-
sible for the chemotaxis of acute inammatory cells into olized (with a half-life of 1.5 min in blood), and the main

278
Prostanoids 23
products that have been detected in vivo are PGJ2 and 9 Thromboxane A2
11 PGF2. PGD2 is a bronchoconstrictor of human airways
and is more potent when inhaled by human subjects than TxA2 is a potent constrictor of smooth muscle. It was
PGF2 [13]. originally described as being released from platelets, but
The biological eects of PGD2 occur through stimu- it is now known to be released from other cells, including
lation of the G-protein-coupled receptors DP1, CRTH2 macrophages and neutrophils. As noted above, the biologi-
(also known as DP2), and TP receptors [14], and in part cal half-life of TxA2 is very short, so it has been implicated
indirectly through a presynaptic action on airway cholin- in disease processes through identication of its more sta-
ergic nerves to release acetylcholine [15]. The bronchoc- ble metabolite thromboxane B2 (TxB2) in biological u-
onstrictor eects of PGD2 are mediated by stimulation of ids; through the use of the stable TxA2 analogs U44069
TP receptors [16]. PGD2 also caused vasodilatation lead- or U46619, which mimic most of the biological eects of
ing to erythema and edema [17], which occurs through TxA2; and through the use of inhibitors of TxA2 synthe-
PGD2 stimulation of the DP1 receptor [18]. PGD2 is also sis and antagonists of the TxA2 receptor. Using these tech-
known to cause chemotaxis of eosinophils [19] and TH2 niques, TxA2 has been implicated in
lymphocytes and modulation of TH2-cytokine produc-
tion, and these eects are mediated by PGD2 stimulation the pathogenesis of airway hyperresponsiveness in dogs
of CRTH2 [20]. CRTH2 is a receptor unrelated to DP1 [29]and primates [30];
and which was originally described as an orphan chem- in the late cutaneous response to intradermal allergen in
oattractant-like receptor expressed by TH2 cells [21] and humans [31];
which is now known to be selectively expressed by TH2 in the late asthmatic response after inhaled allergen in
lymphocytes, eosinophils, and basophils [22]. PGD2 can humans [32];
stimulate the production of the cytokines IL-4, IL-5, and
IL-13 by human TH2 cells in the absence of co-stimulation in airway hyperresponsiveness in asthmatic subjects [33].
[23]. It has been suggested that DP1 and CRTH2 coop- Other studies, however, have demonstrated slight,
erate to promote TH2-dependent allergic responses [14]. but statistically signicant, inhibition of the magnitude of
Activation of DP1 promotes an environment in which the allergen-induced early, but not the late responses after
polarization of TH2 cells can occur, whereas CRTH2 medi- pretreatment with a thromboxane synthetase inhibitor or
ates their recruitment and activation to produce cytokines. receptor agonist [34, 35]. These studies suggest that throm-
Also, stimulation of DP1 by PGD2 has been suggested to boxane may be released following allergen challenge, and
induce regulatory T-cells and to control the extent of airway may account for a portion of the airway narrowing observed
inammation by an IL-10 dependent mechanism [24]. during the early asthmatic response; however, thrombox-
Subthreshold contractile concentrations of PGD2 ane is not important in the airway hyperresponsiveness that
have been demonstrated to increase airway responsive- occurs following allergen inhalation.
ness to inhaled histamine and methacholine in asthmatic
subjects [25]. Thus, PGD2 released in human airways after
allergen inhalation has the potential to both cause acute
broncho-constriction and increase airway hyperresponsive-
ness to other constrictor mediators. However, specic recep-
tor antagonists for PGD2 or inhibitors of its production are
INHIBITORY PROSTAGLANDINS
not yet available to allow a precise evaluation of the impor-
tance of this cycloxygenase metabolite in the spontaneous The dierentiation of the prostaglandins into stimulatory or
asthmatic response. inhibitory mediators is somewhat inappropriate. For exam-
ple, both PGE2 and PGF2 can have dierent eects on the
airways depending on the time after inhalation at which
Prostaglandin PGF2 the response is measured [36]. However, the main action
of PGE2 and PGI2 on airway function is to relax airway
PGF2 may also play a role as a mediator of bronchoconstric- smooth muscle and to antagonize the contractile responses
tion and airway hyperresponsiveness following inhaled aller- of other bronchoconstrictor agonists. In addition PGE2 is
gen in human subjects. PGF2 is a potent bronchoconstrictor extremely potent at inhibiting the release of acetylcholine
in asthmatic airways [26], and when inhaled at subthreshold from airway cholinergic nerves [28].
constrictor concentrations increases airway responsiveness in The evidence that inhibitory prostaglandins play a
dogs and human subjects [27]. PGF2 also causes bronchoc- role in modulating the contractile responses of agonists in
onstriction in human subjects partially through cholinergic- asthmatic subjects comes from studies which have demon-
mediated bronchoconstriction [28]. As with PGD2, there strated that tachyphylaxis (a decreased response to repeated
are no selective PGF2 receptor antagonists available, which stimulation) occurs following repeated challenges with
would allow identication of the specic importance of these inhaled histamine, when challenges are separated by up to
metabolites in mediating theses airway responses. It has been 6 h [37]. Also, repeated exercise challenges in asthmatic
suggested that all contractile prostaglandins act via the TP1 subjects, at intervals up to 4 h, also result in less bronchoc-
receptor. Therefore, dierentiation of the relative impor- onstriction occurring after the second when compared to
tance of the contractile prostaglandins in causing asthmatic the initial exercise challenge [38]. This has been termed
responses may prove to be dicult. exercise refractoriness. This inhibitory eect are abolished

279
Asthma and COPD: Basic Mechanisms and Clinical Management

10 10 Diluent

5 5
PGE2
0 0
PGE2

FEV1 (% change)
FEV1 (% change)

5 5

10 10

15 15

20 Placebo 20


Placebo
25 25

30 30
0 5 10 15 20 25 30 0 1 2 3 4 5 6 7
Time post exercise (min) Time post allergen (h)

FIG. 23.2 The attenuation of exercise-induced bronchoconstriction FIG. 23.3 The attenuation of allergen-induced early and late asthmatic
by pretreatment with inhaled PGE2. Reproduced with permission from responses by pretreatment with inhaled PGE2. Reproduced with permission
Ref. [40]. from Ref. [51].

50 Placebo
*
eosinophils (%)

PGE2 *
Sputum

25

0
50
EG2  cells (%)

*
Sputum

25 *

0
1.0 *
MCC (%)
Sputum

0.5

0.0
FIG. 23.4 The attenuation of allergen-induced
Pre 7 24 increases in sputum eosinophils, EG2 cells (activated
Time post allergen (h) eosinophils) and metachromatic cells (mast cells and
basophils), at 7 h and 24 h after allergen inhalation,
Allergen effect PGE2 effect by pretreatment with inhaled PGE2. Reproduced with
permission from Ref. [51].

by pretreatment with indomethacin [38]. This suggests lation, reduce airway responsiveness to both histamine and
that inhibitory prostaglandins, released as a consequence methacholine [39], and that inhaled PGE2 largely abolishes
of exercise, could modify bronchoconstrictor responses in exercise-induced bronchoconstriction [40] (Fig. 23.2).
asthmatics. This hypothesis is supported by studies which These results are consistent with studies of airway
have demonstrated that pretreatment of asthmatic subjects smooth muscle in vitro, where histamine tachyphylaxis
with oral PGE1, in doses which do not cause bronchodi- occurs through inhibitory prostaglandin release [41] and

280
Prostanoids 23
with studies of dogs in vivo where histamine tachyphylaxis such as inhaled allergen in asthmatic patients. PGD2 also
is inhibited by indomethacin [42]. In addition, histamine promotes the chemotaxis of eosinophils and Th2 cells, as
tachyphylaxis in asthmatic subjects is blocked by pretreat- well as the release of Th2 cytokines through stimulation of
ment with an H2 receptor antagonist in asthmatics, sug- CRTH2. Also, there is evidence that inhibitory prostaglan-
gesting that H2 receptor stimulation is involved with the dins are released by asthmatic airways, which reduces bron-
development of histamine tachyphylaxis [43]. Contraction choconstrictor responses to stimuli such as exercise, and
of asthmatic airways by histamine also reduces airway that this eect is mediated by leukotriene-induced PGE2
responsiveness to acetylcholine and exercise [44]. This lack release. PGE2 also appears to play a role in inhibiting leu-
of specicity suggests that either receptor downregulation or kotriene overproduction in patients with aspirin-intolerant
an alteration of the contractile properties or airway smooth asthma. Finally, PGE2 can also attenuate allergen-induced
muscle is occurring. airway responses and eosinophilic inammation. These
An initial hypothesis to explain exercise refractori- studies suggest that endogenous production of PGE2 does
ness in asthmatics was that histamine is released following have an important inuence on the magnitude of asthmatic
exercise, causing bronchoconstriction, but this also provides responses to stimuli such as exercise or inhaled allergens.
partial protection against subsequent exercise bronchocon-
striction, through PGE2 released by stimulation of histamine
H2 receptors. However, several studies have suggested that References
this hypothesis is incorrect. First, the marked attenuation
of exercise-induced bronchoconstriction by pretreatment 1. Chan IH, Tang NL, Leung TF, Ma SL, Zhang YP, Wong GW et al.
with cysLT1, that is leukotriene (LT)D4-receptor antago- Association of prostaglandin-endoperoxide synthase 2 gene polymorphisms
nists indicates that LTD4, rather than histamine, is the main with asthma and atopy in Chinese children. Allergy 62(7): 8029, 2007.
mediator responsible for exercise-induced bronchoconstric- 2. Shi J, Misso NL, Duy DL, Bradley B, Beard R, Thompson PJ et al.
tion [45]. Second, exercise refractoriness is not prevented by Cyclooxygenase-1 gene polymorphisms in patients with dierent
pretreatment with the H2-receptor antagonists, which eec- asthma phenotypes and atopy. Eur Respir J 26(2): 24956, 2005.
3. Coleman RA, Smith WL, Narumiya S. International Union of
tively prevent histamine tachyphylaxis [46]. Therefore, his-
Pharmacology classication of prostanoid receptors: properties, dis-
tamine-stimulated inhibitory prostaglandin release does not
tribution, and structure of the receptors and their subtypes. Pharmacol
appear to be the cause of exercise refractoriness. Rev 46(2): 20529, 1994.
These studies raise the possibility that exercise refrac- 4. OByrne PM, Walters EH, Aizawa H, Fabbri LM, Holtzman MJ,
toriness is caused by leukotriene-stimulated inhibitory pros- Nadel JA. Indomethacin inhibits the airway hyperresponsiveness but
taglandin release. This hypothesis is supported by evidence not the neutrophil inux induced by ozone in dogs. Am Rev Respir Dis
that there is interdependence between the cycloxygenase 130(2): 22024, 1984.
and lipoxygenase pathways of arachidonate metabolism in 5. OSullivan S, Dahlen B, Roquet A, Larsson L, Dahlen SE, Kumlin M.
causing exercise bronchoconstriction and refractoriness in Urinary 9 alpha, 11 beta-PGF2 as a marker of mast cell activation in
asthmatic subjects. [47] allergic and aspirin-intolerant asthma. Adv Exp Med Biol 433: 15962,
1997.
The release of cysteinyl leukotrienes in the airways is
6. OSullivan S, Roquet A, Dahlen B, Larsen F, Eklund A, Kumlin M
also important in the pathophysiology of aspirin-intolerant et al. Evidence for mast cell activation during exercise-induced bron-
asthma. This is associated with over-expression of the choconstriction. Eur Respir J 12(2): 34550, 1998.
enzyme LTC4 synthetase in the airways of patients with 7. Fabbri LM, Aizawa H, OByrne PM, Bethel RA, Walters EH,
aspirin-intolerant asthma [48]. In addition, pretreatment Holtzman MJ et al. An anti-inammatory drug (BW755C) inhibits
with inhaled PGE2 prevents not only the bronchocon- airway hyperresponsiveness induced by ozone in dogs. J Allergy Clin
striction caused by aspirin, but also the increases in urinary Immunol 76(2 Pt 1): 16266, 1985.
LTE4 associated with it [49]. These results suggest that, in 8. Berend N, Armour CL, Black JL. Indomethacin inhibits the increased
patients with aspirin-intolerant asthma, PGE2 may play airway responsiveness to histamine following inhalation of C5a des
a critical role in inhibiting the overproduction of cystei- Arg in rabbits. Agents Actions 18(56): 46872, 1986.
9. Lanes S, Stevenson JS, Codias E, Hernandez A, Sielczak MW,
nyl leukotrienes, as this inhibiting eect is lost when the
Wanner A et al. Indomethacin and FPL-57231 inhibit antigen-
patients ingest aspirin or other cycloxygenase inhibitors. induced airway hyperresponsiveness in sheep. J Appl Physiol 61(3):
Inhaled PGE2 has also been demonstrated to attenuate 86472, 1986.
allergen-induced bronchoconstrictor responses (Fig. 23.3) 10. Fairfax AJ. Inhibition of the late asthmatic response to house dust mite
and airway hyperresponsiveness [50], and to attenuate by non-steroidal anti-inammatory drugs. Prostaglandins Leukot Med
allergen-induced airway eosinophilic inammation [51] 8(3): 23948, 1982.
(Fig. 23.4). Thus, these ndings suggest that endogenous 11. Kirby JG, Hargreave FE, Cockcroft DW, OByrne PM. Eect of
PGE2, produced in response to inhaled allergen, may play a indomethacin on allergen-induced asthmatic responses. J Appl Physiol
role in regulating the associated airway inammation. 66(2): 57883, 1989.
12. Murray JJ, Tonnel AB, Brash AR, Roberts LJ, Gosset P, Workman R
et al. Release of prostaglandin D2 into human airways during acute
antigen challenge. N Engl J Med 315(13): 8004, 1986.
13. Hardy CC, Robinson C, Tatterseld AE, Holgate ST. The bronchoc-
SUMMARY onstrictor eect of inhaled prostaglandin D2 in normal and asthmatic
men. N Engl J Med 311(4): 20913, 1984.
14. Pettipher R, Hansel TT, Armer R. Antagonism of the prostaglandin
In asthmatic patients, it is likely that PGD2 and TxA2 are D2 receptors DP1 and CRTH2 as an approach to treat allergic dis-
involved in causing acute bronchoconstriction after stimuli eases. Nat Rev Drug Discov 6(4): 31325, 2007.

281
Asthma and COPD: Basic Mechanisms and Clinical Management

15. Tamaoki J, Sekizawa K, Graf PD, Nadel JA. Cholinergic neuromod- after indomethacin pretreatment. Br J Clin Pharmacol 19(4): 45970,
ulation by prostaglandin D2 in canine airway smooth muscle. J Appl 1985.
Physiol 63(4): 1396400, 1987. 33. Fujimura M, Sakamoto S, Saito M, Miyake Y, Matsuda T. Eect of a
16. Coleman RA, Sheldrick RL. Prostanoid-induced contraction of human thromboxane A2 receptor antagonist (AA-2414) on bronchial hyper-
bronchial smooth muscle is mediated by TP-receptors. Br J Pharmacol responsiveness to methacholine in subjects with asthma. J Allergy Clin
96(3): 68892, 1989. Immunol 87(1 Pt 1): 2327, 1991.
17. Flower RJ, Harvey EA, Kingston WP. Inammatory eects of prostag- 34. Manning PJ, Stevens WH, Cockcroft DW, OByrne PM. The role of
landin D2 in rat and human skin. Br J Pharmacol 56(2): 22933, 1976. thromboxane in allergen-induced asthmatic responses. Eur Respir J
18. Giles H, Le P, Bolofo ML, Kelly MG, Robertson AD. The classica- 4(6): 66772, 1991.
tion of prostaglandin DP-receptors in platelets and vasculature using 35. Beasley RC, Featherstone RL, Church MK, Raerty P, Varley JG,
BW A868C, a novel, selective and potent competitive antagonist. Br J Harris A et al. Eect of a thromboxane receptor antagonist on PGD2
Pharmacol 96(2): 291300, 1989. and allergen-induced bronchoconstriction. J Appl Physiol 66(4): 1685
19. Shiraishi Y, Asano K, Nakajima T, Oguma T, Suzuki Y, Shiomi T 93, 1989.
et al. Prostaglandin D2-induced eosinophilic airway inammation is 36. Walters EH. The inuence of airways smooth muscle tone upon the
mediated by CRTH2 receptor. J Pharmacol Exp Ther 312(3): 95460, response to inhaled PGE2. Agents Actions Suppl 13: 18588, 1983.
2005. 37. Manning PJ, Jones GL, OByrne PM. Tachyphylaxis to inhaled hista-
20. Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori Y mine in asthmatic subjects. J Appl Physiol 63(4): 157277, 1987.
et al. Prostaglandin D2 selectively induces chemotaxis in T helper type 38. OByrne PM, Jones GL. The eect of indomethacin on exercise-
2 cells, eosinophils, and basophils via seven-transmembrane receptor induced bronchoconstriction and refractoriness after exercise. Am Rev
CRTH2. J Exp Med 193(2): 25561, 2001. Respir Dis 134(1): 6972, 1986.
21. Abe H, Takeshita T, Nagata K, Arita T, Endo Y, Fujita T et al. 39. Manning PJ, Lane CG, OByrne PM. The eect of oral prostaglandin
Molecular cloning, chromosome mapping and characterization of the E1 on airway responsiveness in asthmatic subjects. Pulm Pharmacol
mouse CRTH2 gene, a putative member of the leukocyte chemoat- 2(3): 12124, 1989.
tractant receptor family. Gene 227(1): 7177, 1999. 40. Melillo E, Woolley KL, Manning PJ, Watson RM, Obyrne PM. Eect
22. Nagata K, Hirai H, Tanaka K, Ogawa K, Aso T, Sugamura K et al. of inhaled PGE2 on exercise-induced bronchoconstriction in asthmatic
CRTH2, an orphan receptor of T-helper-2-cells, is expressed on subjects. Am J Respir Crit Care Med 149(5): 113841, 1994.
basophils and eosinophils and responds to mast cell-derived factor(s). 41. Anderson WH, Krzanowski JJ, Polson JB, Szentivanyi A. Prostaglandins
FEBS Lett 459(2): 19599, 1999. as mediators of tachyphylaxis to histamine in canine tracheal smooth
23. Xue L, Gyles SL, Wettey FR, Gazi L, Townsend E, Hunter MG et muscle. Adv Prostaglandin Thromboxane Res 7: 9951001, 1980.
al. Prostaglandin D2 causes preferential induction of proinammatory 42. Shore S, Irvin CG, Shenkier T, Martin JG. Mechanisms of histamine-
Th2 cytokine production through an action on chemoattractant recep- induced contraction of canine airway smooth muscle. J Appl Physiol
tor-like molecule expressed on Th2 cells. J Immunol 175(10): 653136, 55(1 Pt 1): 2226, 1983.
2005. 43. Jackson PJ, Manning PJ, OByrne PM. A new role for histamine H2-
24. Hammad H, Kool M, Soullie T, Narumiya S, Trottein F, Hoogsteden receptors in asthmatic airways. Am Rev Respir Dis 138(4): 78488, 1988.
HC et al. Activation of the D prostanoid 1 receptor suppresses asthma 44. Hamielec CM, Manning PJ, OByrne PM. Exercise refractoriness after
by modulation of lung dendritic cell function and induction of regula- histamine inhalation in asthmatic subjects. Am Rev Respir Dis 138(4):
tory T cells. J Exp Med 204(2): 35767, 2007. 79498, 1988.
25. Fuller RW, Dixon CM, Dollery CT, Barnes PJ. Prostaglandin D2 45. Manning PJ, Watson RM, Margolskee DJ, Williams VC, Schwartz JI,
potentiates airway responsiveness to histamine and methacholine. Am OByrne PM. Inhibition of exercise-induced bronchoconstriction by
Rev Respir Dis 133(2): 25254, 1986. MK-571, a potent leukotriene D4-receptor antagonist. N Engl J Med
26. Thomson NC, Roberts R, Bandouvakis J, Newball H, Hargreave FE. 323(25): 173639, 1990.
Comparison of bronchial responses to prostaglandin F2 alpha and 46. Manning PJ, Watson R, OByrne PM. The eects of H2-receptor
methacholine. J Allergy Clin Immunol 68(5): 39298, 1981. antagonists on exercise refractoriness in asthma. J Allergy Clin Immunol
27. OByrne PM, Aizawa H, Bethel RA, Chung KF, Nadel JA, 90(1): 12526, 1992.
Holtzman MJ. Prostaglandin F2 alpha increases responsiveness of pul- 47. Manning PJ, Watson RM, OByrne PM. Exercise-induced refractori-
monary airways in dogs. Prostaglandins 28(4): 53743, 1984. ness in asthmatic subjects involves leukotriene and prostaglandin inter-
28. Walters EH, OByrne PM, Fabbri LM, Graf PD, Holtzman MJ, dependent mechanisms. Am Rev Respir Dis 148(4 Pt 1): 95054, 1993.
Nadel JA. Control of neurotransmission by prostaglandins in canine 48. Cowburn AS, Sladek K, Soja J, Adamek L, Nizankowska E, Szczeklik A
trachealis smooth muscle. J Appl Physiol 57(1): 12934, 1984. et al. Overexpression of leukotriene C4 synthase in bronchial biopsies
29. Aizawa H, Chung KF, Leikauf GD, Ueki I, Bethel RA, OByrne PM from patients with aspirin-intolerant asthma. J Clin Invest 101(4):
et al. Signicance of thromboxane generation in ozone-induced airway 83446, 1998.
hyperresponsiveness in dogs. J Appl Physiol 59(6): 191823, 1985. 49. Sestini P, Armetti L, Gambaro G, Pieroni MG, Reni RM, Sala A
30. Letts GL, McFarlane CS. Thromboxane A2 and airway responsiveness et al. Inhaled PGE2 prevents aspirin-induced bronchoconstriction and
to acetylcholine aerosol in the conscious primate. Prog Clin Biol Res urinary LTE4 excretion in aspirin-sensitive asthma. Am J Respir Crit
263: 9198, 1988. Care Med 153(2): 57275, 1996.
31. Dorsch W, Ring J, Melzer H. A selective inhibitor of thromboxane 50. Pavord ID, Wong CS, Williams J, Tatterseld AE. Eect of inhaled
biosynthesis enhances immediate and inhibits late cutaneous allergic prostaglandin E2 on allergen-induced asthma. Am Rev Respir Dis
reactions in man. J Allergy Clin Immunol 72(2): 16874, 1983. 148(1): 8790, 1993.
32. Shephard EG, Malan L, Macfarlane CM, Mouton W, Joubert JR. Lung 51. Gauvreau GM, Watson RM, OByrne PM. Protective eects of inhaled
function and plasma levels of thromboxane B2, 6-ketoprostaglandin F1 PGE2 on allergen-induced airway responses and airway inammation.
alpha and beta-thromboglobulin in antigen-induced asthma before and Am J Respir Crit Care Med 159(1): 3136, 1999.

282
Leukotrienes and Lipoxins
24CHAPTER

The leukotrienes (LTB4, LTC4, LTD4, and In one major pathway, found in a variety of cells
LTE4) and lipoxins (LXA4 and LXB4) are but most notably neutrophilic polymorphonu-
Bruce D. Levy1 and
molecules derived by lipoxygenation of ara- clear leukocytes (PMN), the addition of water to Jeffrey M. Drazen1,2
chidonic acid. There is evidence that each of form LTB4 is catalyzed by LTA4 hydrolase. [9]. 1
Department of Medicine, Pulmonary
these molecules is potentially important in the In addition to LTB4, the 5-LO product
Division, Brigham and Womens
pathogenesis of asthma and chronic obstruc- (LTA4) can also be enzymatically transformed
tive pulmonary disease (COPD); this chapter to several other bioactive compounds [1]. Each Hospital, Boston, MA, USA
2
reviews the biology of these molecules as related of the other two major lipoxygenases in human Harvard Medical School, Editorial
to these disease entities. tissues, namely 12-LO and 15-LO, can convert Office New England Journal of Medicine,
LTA4 to a distinct class of mediators termed Boston, MA, USA
lipoxins for lipoxygenase interaction products
FORMATION AND METABOLISM OF [10]. Lipoxins carry 3 alcohol groups on arachi-
THE LEUKOTRIENES donic acids 20 carbon backbone with 4 conju-
gated double bonds. The redundant pathways
for lipoxin (LX) biosynthesis indicate the func-
Arachidonic acid, a normal component of many tional importance of these compounds. Because
cell membrane phospholipids, is commonly lipoxygenases are most commonly compartmen-
found esteried to these phospholipids in the talized into distinct cell types, LX biosynthesis
middle or sn2 position. In the presence of an principally occurs during cellcell interactions.
appropriately activated phospholipase A2, ara- For example, at sites of vascular injury or
chidonic acid is cleaved from internal and exter- inammation, PMN 5-LO and platelet 12-LO
nal cell membranes and serves as the primary can interact to generate LXA4 and LXB4 [11].
substrate for subsequent eicosanoid metabolism This transcellular LX generation is also bidi-
(Fig. 24.1) [1]; there is a family of phospholipase rectional as arachidonic acid can originate from
A2 (PLA2) molecules with this catalytic capacity either cell type [12]. Because the airway is lined
[2, 3]. The two major forms of interest in leukot- with epithelial cells that are enriched with 15-
riene and lipoxin biosynthesis are cytosolic PLA2 LO activity, LX generation also occurs when
(cPLA2) and secretory PLA2 (sPLA2) [4, 5]. leukocytes inltrate airways during inamma-
Arachidonic acid, released as a result of tory responses [13]. Of interest, interactions
PLA2 action, enters into a series of reactions between 5-LO and 12-LO generate an equal
at the perinuclear membrane. The rst of these amount of LXA4 and LXB4, whereas 5-LO and
requires a specic 5-lipoxygenase activating 15-LO interactions strongly favor LXA4 pro-
protein (FLAP) [6, 7], which allows arachido- duction [14].
nate to serve as a substrate for the enzyme Aspirin inhibits prostaglandin formation
5-lipoxygenase [8]. 5-Lipoxygenase sequentially and triggers the formation of lipoxin epim-
catalyzes the addition of molecular oxygen to ers [15] as well. Acetylation of cyclooxygen-
arachidonic acid to form 5-hydroperoxy eico- ase-2 at serine 516 blocks formation of PGG2.
satetraenoic acid (5-HPETE) and leukotriene When this occurs, enzymatic action results in
A4 (LTA4), respectively; LTA4 is a major branch arachidonic acids being converted to 15(R)-
point in the formation of the leukotrienes [1]. hydroxyeicosatetraenoic acid (15(R)-HETE).

283
Asthma and COPD: Basic Mechanisms and Clinical Management

Cytosol Extracellular
Cytosolic space
phospholipase A2 Chemotaxis

BLT
receptor
Arachidonic
acid
Leukotriene B4
5-Lipoxygenase Transmembrane
activating transporter
protein Epoxide Leukotriene D4
hydrolase
Nucleus

Zileuton Leukotriene C4 Leukotriene E4


5-Lipoxygenase

Leukotriene C4 Montelukast
Leukotriene A4 Pranlukast
Zafirlukast

CysLT1
receptor

Nuclear Leukotriene C4
membrane synthase Cell Airway
membranes Smooth-muscle
constriction
Eosinophil migration
Edema

FIG. 24.1 Schematic showing the synthesis of leukotrienes from arachidonic acid and the site of action of various drugs in the pathway. Reproduced from
Ref. [75], with permission.

This molecule can be a substrate for conversion of 5-LO to transmembrane transporter [2325], and when it is out-
15-epimer-LXA4 (15-epi-LXA4), a compound also known side the cell, LTC4 serves as a substrate for -glutamyl
as aspirin-triggered LXA4. This seemingly subtle switch in transpeptidase [26, 27] or a specic leukotrienase [28], which
stereochemistry at carbon-15 leads to a twofold increase in cleaves the glutamic acid moiety from its peptide chain to
circulating half-life and potency compared to the 15(S)- form leukotriene D4 (LTD4; LTD4 is further processed by
LXA4 product of 15-LO [16]. Because cytochrome p450 the removal of the glycine moiety from its peptide chain
enzymes can also catalyze 15(R)-HETE formation from to form leukotriene E4, LTE4) [29, 30]. LTC4, LTD4, and
arachidonate, aspirin is not required for 15-epi-LXA4 pro- LTE4 make up the material formerly known as slow-react-
duction [17]. ing substance of anaphylaxis or SRS-A and are collectively
Similar to LTs, LXs are autacoids that are rapidly known as the cysteinyl leukotrienes.
formed and rapidly, enzymatically inactivated [18]. The In the presence of appropriately activated PMNs, the
major route for LX inactivation is dehydrogenation by 15- cysteinyl leukotrienes are degraded to their respective sul-
hydroxy/oxo-eicosanoid oxidoreductase (15-PGDH), lead- foxides and 6-trans diastereoisomers of LTB4 [31]. In the
ing to the conversion of LXA4 to the biologically inert absence of such cells the major degradation and excretion
15-oxo-LXA4. This compound is further converted to 13, products of the cysteinyl leukotrienes are native LTE4,
14-dihydro-15-oxo-LXA4 by LXA4/PGE 13,14-reductase/ N-acetyl LTE4, or the products resulting from -oxidation
LTB4 12-hydroxydehydrogenase (PGR/LTB4DH). This and -elimination of LTE4 [32].
compound is subject to additional reduction of the 15-oxo
group to 13,14-dihydro-LXA4 by 15-PGDH. Moreover,
LXs also undergo -oxidation in vivo [19].
Other cells of myeloid origin including eosinophils,
mast cells, and alveolar macrophages, not only have the
LEUKOTRIENES IN ASTHMA
capacity to form LTA4 from arachidonic acid, but also they
possess a unique and specic glutathionyl-S-transferase, LTC4 Since the structural identication of the leukotrienes, a
synthase, which catalyzes the conjugation of glutathione number of lines of evidence have accrued indicating a causal
to LTA4, at carbon 6, to form leukotriene C4 (LTC4) role for the cysteinyl leukotrienes in aspects of the asthmatic
[2022]. Once formed, LTC4 exits the cell via a specic response. These are reviewed in the following sections.

284
Leukotrienes and Lipoxins 24
Cysteinyl leukotrienes in the pathology of greater amounts in the bronchoalveolar lavage (BAL) uid
chronic mild asthma from subjects with symptomatic asthma than from subjects
with asymptomatic asthma or normal subjects [5760], as
The cysteinyl leukotrienes are synthesized and exported into well as from exhaled breath condensate [6163], suggest-
the microenvironment by constitutive and inltrating cells ing that the leukotrienes are produced locally in the lungs
including mast cells and eosinophils [33, 34]. Cysteinyl leu- of patients with asthma.
kotrienes produced by these cells have numerous actions but The profusion of leukotriene production can be esti-
the predominant ones in asthma are their ligation of leuko- mated from measurements made on urine samples [6469].
triene receptors on airway smooth muscle and the bronchial In normal human subjects, after intravenous administration
vasculature mediating airway obstruction and microvascular of radiolabeled LTC4, 1248% of the counts are recovered
leak [3537]. in the urine with 413% as intact LTE4 [70, 71], and it is
now common to measure leukotriene recovery from the
urine as an index of endogenous production of leukotrienes.
Asano et al. [72] examined urinary LTE4 excretion rates
Biological effects of cysteinyl leukotrienes in eight patients with mild chronic stable asthma (FEV1
relevant to the asthmatic response 70% predicted, inhaled -agonists as the only asthma treat-
ment) followed on a metabolic ward for 4 days. They
The leukotrienes are known to have profound biochemical, demonstrated that, on average, patients with asthma have sig-
immunological, and physiological eects, even in picomolar nicantly higher urinary LTE4 levels than normal subjects.
concentrations, including induction of airway obstruction, However, among the asthmatics studied there were indi-
tissue edema, and expression of bronchial mucus from sub- viduals who were persistent hyperexcretors of urinary LTE4
mucosal glands [3840]; these pathobiological eects make and others with urinary LTE4 levels that were persistently
them part of the panel of mediators which characterize within the normal range. Among the explanations for this
asthmatic responses. nding is the possibility that, among patients with asthma,
Although these mediators have pleotropic biological whose clinical phenotype is similar, there are individuals
actions, the ability of the cysteinyl leukotrienes to mediate whose asthma is associated with leukotriene production
airway narrowing in normal individuals and persons with and others for whom this is not the case. These data clearly
asthma is critical to the biology of asthma. Inhalation of indicate that, during spontaneous, induced, or chronic stable
aerosols of leukotrienes by normal subjects results in airway asthma, at least in some individuals, there is enhanced uri-
obstruction as manifested by a decrease in ow rates during nary LTE4 excretion and by inference an increased cysteinyl
a forced exhalation [4152]. In addition, among normal leukotriene production. Among patients with severe asthma,
subjects there is a relationship between responsiveness to who are receiving treatment with corticosteroids, there are
the leukotrienes and responsiveness to reference agonists substantially increased levels of LTE4 in the urine [73].
such as histamine or methacholine [46, 47]. Subjects that
are more responsive to histamine are those that are more
responsive to the leukotrienes. Leukotriene receptor blockade and
In subjects with asthma, LTC4, LTD4, and LTE4 are
potent bronchoconstrictor agonists when administered by
synthesis inhibition
aerosol, as indicated by induced decrements in the FEV1,
in the V , or in specic conductance [4648, 50, 53, 54]. Two classes of pharmacological agents have been demon-
30P
strated to have a salutary eect on airway obstruction in
When the latter outcome indicator is used, the nebulizer
patients with asthma: leukotriene receptor antagonists and
concentrations required to narrow airways in asthmatics are
leukotriene synthesis inhibitors. As noted above there are at
approximately one-tenth of those required by normal sub-
least two distinct receptors that when ligated transduce the
jects to achieve the same decrement in airow rates. Since
biological activity of the cysteinyl leukotrienes; there are the
normal subjects are approximately 100-fold less sensitive to
cysteinyl leukotriene receptors type 1 and type 2 (CysLT1
histamine or methacholine than asthmatic subjects, whereas
and CysLT2) [37, 74]. Although many chemically distinct
they are only 10-fold less responsive to the cysteinyl leuko-
antagonists at the CysLT1 receptor have been recognized,
trienes, this indicates that the relative degree of hyperre-
as of the present time, only three have been introduced
sponsiveness to the cysteinyl leukotrienes is less than that
into medical use; namely, pranlukast, zarlukast, and
observed when histamine or methacholine is used as the
montelukast [75].
contractile agonist.
With regard to leukotriene synthesis inhibitors,
agents have been developed which are direct inhibitors of
5-LO, such as zileuton [76], as well as agents that inhibit
Leukotriene recovery in asthma the interaction between arachidonic acid and FLAP (MK-
0591 and Bay x1005) [77, 78] (Fig. 24.2).
Cysteinyl leukotrienes have been recovered after experi-
mental challenges, which elicit clinical symptoms similar to
those that occur in spontaneously occurring asthmatic con- Induced asthma
ditions. Leukotrienes have also been recovered in the nasal Leukotriene receptor antagonists and synthesis inhibitors
lavage uid after intranasal challenge with either antigen or have been shown to inhibit various types of laboratory-
cold air [55, 56]. Leukotrienes are recovered in signicantly induced asthma [8084]. Current data indicate that most

285
Asthma and COPD: Basic Mechanisms and Clinical Management

Zileuton, 600 mg
Zileuton, 400 mg
Placebo
20 20

FEV1, % Change

FEV1, % Change
15 * 15
*
10 10

5 5

0 0
0 2 4 6 8 10 12 14 0 2 4 6 8 10 12 14
Treatment week

FIG. 24.2 Mean percentage change in forced expiratory volume rate in the first second (FEV1) (SE) from pre-randomization baseline during 13 weeks of
treatment with 600 mg of zileuton, 400 mg of zileuton, or placebo, each given four times daily. Asterisk indicates p  0.05 versus placebo; and the dagger,
p  0.01 versus placebo. Left panel, average FEV1 24 h after drug ingestion (expected time of peak drug levels). Right panel, FEV, before ingestion of
morning dose of drug (expected time of low drug levels). Reproduced from Ref. [79], with permission.

TABLE 24.1 Early studies of leukotriene modifiers in patients with chronic persistent asthmaa

Zafirlukast Montelukast Pranlukast Zileuton

Number of patientsb 70 408 45 122

Length of study 6 week 12 week 6 week 6 mo

Dose (oral) 20 mg b.i.d. 10 mg q.d. 337.5 mg b.i.d. 600 mg q.i.d.

Baseline FEV1 (% of predicted) 66% 66% 66% 62%

Trough improvement in FEV1 litersc 0.23 lg 0.31 lg 0.34 l

Trough improvement in FEV1 percentc,d 11% (1314%)g 11.5%g,h 15(18)%

Peak improvement in FEV1d,e 13% 20%i (23%)g

-adrenergic agonist use reductiond 31% 27%j NC (30%)

Improvement in AM PEFRd 6% 6.1% 5%g 7.1% (8.5%)

Treatment failure or glucocorticoid rescue therapy 2% versus 10% 8.3% versus 21.5%g
(treatment versus placebo)f

Decrease in symptoms: day/night (%) 28/46 20/NC NC/28 36/33

Reproduced from Ref. [75]. 1999 Massachusetts Medical Society. All rights reserved with permission. NC: no significant change; PEFR: peak expiratory flow rate.
a
Compared with pretreatment values. Only data from double-blind, randomized, and placebo-controlled studies are included. Although additional studies with each drug have
been reported, the study chosen for display is representative. All values were statistically significant from placebo unless otherwise indicated.
b
Number of patients receiving active treatment at the dose indicated. Zafirlukast was administered at twice the currently recommended dose.
c
Trough values: values immediately before next dose or those values not reported as obtained at times of expected peak plasma concentrations or effects.
d
Figures not in parentheses represent means over the study period or end-point analyses. Figures in parentheses represent maximum effect among the study observation
intervals reported.
e
Values recorded at or near time of drugs expected peak plasma concentration or effects.
f
Treatment failure for zafirlukast; glucocorticoid rescue therapy for zileuton.
g
Derived from figures or statistics.
h
225 mg dose.
i
Endpoint value: NS versus placebo at week 26.
j
Percentage difference compared with placebo.

but not all of the physiological eects of aspirin-induced anti-leukotriene treatment can prevent all the clinical
asthma derive from the action of the cysteinyl leukot- manifestations of aspirin ingestion in patients with aspirin-
rienes [82, 85]. It is important to note that, although all sensitive asthma. Indeed, there have been reports of aspirin-
the physiological eects of aspirin-induced asthma derive induced asthma in patients receiving treatment with CysLT1
from the eects of the leukotrienes, this does not mean that receptor antagonists [86, 87]. These ndings are consistent

286
Leukotrienes and Lipoxins 24
with the known competitive nature of CysLT1 antagonists respiratory tissues [13] and block airway inammation
and the massive release of cysteinyl leukotrienes after aspirin and hyperresponsiveness in an experimental model of
ingestion in patients with aspirin-induced aspirin. asthma [102].

Leukotriene inhibition in chronic stable asthma


Multiple double-blind and placebo-controlled and active Biological effects of lipoxins relevant
compartor clinical trials in both adult and pediatric patients to the asthmatic response
with mild-to-moderate chronic stable asthma reported in
the archival literature; these have been the subject of multi- Lipoxin A4 displays cell-type selective actions [10]. On the
ple reviews [8892]. one hand, LXA4 blocks PMN activation and transmigration
The general design of these trials has been similar. across endothelial and epithelial cells [104107]. Distinct
They have recruited and enrolled patients whose asthma was from these stop signals for PMNs, LXA4 stimulates mono-
marginally controlled solely by use of inhaled -agonists, cyte locomotion and macrophage phagocytosis of apoptotic
who had FEV1 values most often between 40% and 80% of PMNs [108, 109]. LXA4 also reduces eosinophil tissue
predicted, who had moderate asthma symptoms as judged accumulation [102], dendritic cell (DC) mobilization [110],
from daily symptom diaries. Each trial incorporated a 13 T-lymphocyte cytokine release [103], and NK cell cytotox-
week run-in period when all patients had their asthma icity [111]. In sum, LXs regulate both innate and acquired
control monitored while on single-blind oral placebo. immune eector cells to reduce leukocyte entry into tissues
During this period baseline lung function, -agonist use, and promote clearance of apoptotic cells, important mecha-
and symptom data were gathered. Patients then entered a nisms in the resolution of acute inammation.
period in which they received randomized treatment with LXs also regulate airway epithelial cells [112]. LXA4
an active agent or placebo. Patients returned on a regular blocks proinammatory cytokine and chemokine release
basis to their clinical centers where their lung function and and gene expression via NFB [113]. Of interest, LX sign-
asthma symptom data were recorded; the results from these aling increases expression of bacterial permeability-inducing
trials are summarized in Table 24.1. protein in epithelial cells to enhance mucosal bacterial kill-
In the course of the performance of these trials, it has ing [114]. Thus, in addition to anti-inammation, LXA4
been noted by many investigators that there is a marked signals carry host protective actions.
variability among individuals in the response to anti- Lipoxins also potently regulate pulmonary responses.
leukotriene treatment. One interpretation of this obser- LXA4 relaxes precontracted pulmonary arteries and bronchi
vation is that there could be genetic dierences among [115]. In addition, a LX stable analog, designed using the
individuals that lead to dierences in the role of leukotrienes 15-epi-LXA4 structure as a template, markedly inhibits (with
in a given individuals asthmatic responses. Polymorphisms, as little as 1 g; 0.05 mg/kg) allergen-driven airway hyper-
both with and without known functional consequences responsiveness and inammation in an experimental model
have been identied in a number of the genes encoding the of asthma [102]. Asthma is frequently worsened by aspira-
proteins that make up the 5-lipoxygenase pathway [9499]. tion of gastric acid, and LX and 15-epi-LX signaling pro-
The gene encoding 5-lipoxygenase has been shown to har- mote restitution of injured airway epithelium and resolution
bor variability that is associated with the asthma treatment of acid-initiated acute airway injury [112, 116]. Together,
response, but the specic functional variants within that the impact of LXs on both cellular eectors and airway
gene have not been identied [97, 100]. responses indicate protective roles for these counter-regula-
tory mediators in airway inammation and hyperresponsive-
ness, important processes linked to asthma pathogenesis.
As a consequence of LXs rapid inactivation, there is
only limited information on their actions in humans in vivo.
LIPOXINS IN ASTHMA In one clinical trial, LXA4 was administered to individuals
with asthma, leading to protection from LTC4-induced bron-
choconstriction [117]. The recent development of new LX
Lipoxins are derived from arachidonic acid, yet structurally stable analogs that are topically and orally active should ena-
and functionally distinct from prostaglandins and leukot- ble further investigation on LX regulation of human illness.
rienes [1]. There is a growing body of evidence linking these
counter-regulatory eicosanoid mediators to homeostatic air-
way responses including control of airway inammation and Lipoxin recovery in asthma
reactivity [101].
Lipoxins have been identied in human subjects with a
wide range of respiratory illnesses involving leukocyte acti-
Lipoxins in the pathology of chronic vation [13]. Both LXs and 15-epi-LXs are generated during
mild asthma respiratory inammation [101, 118]. LXA4 (0.42.8 ng/ml)
has been detected in BAL and pleural uid from individu-
Lipoxins and 15-epi-LXs display several features of media- als with inammatory lung disease [13, 119]. In a murine
tors inhibiting airway inammation [10] including inhibi- experimental model of asthma, LX formation at peak air-
tion in vivo of eosinophil and T-lymphocyte activation and way inammation is similar in magnitude to the amount of
TH2 cytokine release [102, 103]. LXs are generated in LTB4 formed, yet one to two log orders less than cysteinyl

287
Asthma and COPD: Basic Mechanisms and Clinical Management

LTs and PGE2 [102]. Of interest, diminished formation of The leukotrienes have pleomorphic inammatory actions
these counter-regulatory mediators has been identied in that are relevant to the microenvironment of the airway in
severe forms of human respiratory illness, including aspi- COPD. Although a complete evaluation of these actions is
rin-intolerant asthma [120] and severe, steroid-dependent beyond the scope of this chapter, there are excellent review
asthma [121]. Decreased production of these counter-regu- articles that reect this biology [39, 40, 132134]. These
latory substances could predispose the host to more severe, data indicate that if leukotrienes were to become available
inammatory responses in the lung. in the microenvironment of the airway that they have the
potential to contribute to aspects of the bronchotic pheno-
type; at this time, their exact role is not known.
Lipoxin A4 receptors
Lipoxins interact with one or more specic receptors to sig- Recovery of leukotrienes in COPD
nal their counter-regulatory actions. LXA4 can serve as a
high anity ligand for the LXA4 receptor (ALX) [122, 123] Leukotrienes have been recovered in greater quantities from
and a receptor-level antagonist for a subclass of LTD4 recep- the airways of patients with COPD. For example they are
tors (i.e. CysLT1) [124]. ALX is a seven-transmembrane found in the sputum, BAL uid, or exhaled breath conden-
spanning, G-protein coupled protein, binding LXA4 with sate of patients with chronic bronchitis at higher levels than
high anity (KD 1.7 nM) [125]. The human, mouse from similar samples obtained from normal subjects [135
and rat ALX amino acid sequences share 74% or greater 140]. In one study the concentration of LTB4 in sputum was
homology with the highest homology found in their sec- closely correlated with the number of polymorphonuclear
ond intracellular loop (100% identical) and sixth transmem- leukocytes recovered in the sputum [140]. When patients
brane segments (93% identical) [126]. ALX receptors are with COPD were treated with ibuprofen, the amount of
expressed in several mammalian cells and tissues including LTB4 recovered in the exhaled breath condensate increased;
human PMNs, monocytes, epithelial cells, and human and there was no eect of rofecoxib on this measure [141].
murine spleen. Interleukin-13 and interferon- dramatically Sputum obtained from patients in the midst of an infec-
induce ALX expression in vitro in airway epithelial cells tive exacerbation of chronic bronchitis contained high levels
[124]. Cytokine regulation of ALX is also evident in vivo of LTB4, which fell with treatment [142]. In these samples
during disease states, as ALX is upregulated in airway epi- LTB4 constituted about one-third of the total chemotactic
thelial and inammatory cells during allergic airway inam- activity at the time of disease presentation, and the impor-
mation in a murine model of asthma [102]. ALX was the tance of LTB4 as a chemotactic factor fell with antibiotic
initial receptor identied that binds to both lipid and pep- treatment. These data indicate that leukotrienes, both cystei-
tide ligands [123]. Of interest for asthma therapy, corticos- nyl and di-hydroxy moieties, are present and likely active
teroids induce expression of annexin 1 that is enzymatically in the airway microenvironment in COPD. While limited
cleaved to peptides that can also interact with ALX to initi- information is currently available on LXs and COPD, LXA4
ate anti-inammatory circuits [127]. In PMNs, ALX signals and 15-epi-LXA4 are both found in increased amounts in
for LXs anti-inammatory eects, in part, by polyisoprenyl sputum from subjects with COPD [118].
phosphate remodeling [107] and by inhibition of leukocyte- There are few data in the archival literature from stud-
specic protein-1 phosphorylation, a downstream regulator ies in which agents active on various components of the
of the p38-MAPK cascade [128]. Targeted expression of 5-lipoxygenase pathway have been studied on lung function
human LXA4 receptors to murine leukocytes in transgenic in patients with COPD. Small studies, with varying quality
mice also dramatically inhibits allergen sensitization, eosi- of controls, show that treatment of patients with leukotriene
nophil tracking, and airway inammation [102]. receptor antagonists modies a number of outcome indica-
tors in COPD. In most studies the eects are salutary, but
the magnitude of clinical improvement is marginal at best
[93, 143146]. Since these data are not biologically informa-
tive with respect to the role the leukotrienes in COPD, larger
LEUKOTRIENES AND LIPOXINS IN COPD better-controlled studies are needed to resolve this issue.

Leukotrienes and the biology of the


bronchitic airway CONCLUSIONS
There are fewer data concerning the role of leukotrienes in
the pathogenesis of COPD than are available about this The available data support the following conclusions as to
class of mediators in asthma. Introduction of leukotrienes the role of leukotrienes and lipoxins in bronchial asthma
into the microenvironment of the airway is associated with and COPD:
expression of mucus from glands [129, 130]. The receptor
for LTB4 has been detected in greater profusion on alveo- They are produced by constitutive cells (mast cells/
lar macrophages from patients who smoke and have airway macrophages) and inltrating cells (eosinophils,
obstruction than from either normal subjects or patients neutrophils) implicated in asthma and COPD.
who smoke and do not have airway obstruction [131]. They are potent bronchoconstrictor agonists.

288
Leukotrienes and Lipoxins 24
Their administration results in the expression of mucus 15. Claria J, Serhan CN. Aspirin triggers previously undescribed bioactive
from mucous glands. eicosanoids by human endothelial cell-leukocyte interactions. Proc Natl
Acad Sci USA 92: 947579, 1995.
Laboratory-induced asthma and spontaneous asthma 16. Clish CB, OBrien JA, Gronert K, Stahl GL, Petasis NA, Serhan
or COPD is associated with an enhanced recovery of CN. Local and systemic delivery of a stable aspirin-triggered lipoxin
leukotrienes in the urine and exhaled breath condensate prevents neutrophil recruitment in vivo. Proc Natl Acad Sci USA 96(14):
of subjects with asthma and sputum, BAL or exhaled 824752, 1999.
breath condensate leukotrienes in COPD. 17. Claria J, Lee MH, Serhan CN. Aspirin-triggered lipoxins (15-epi-LX)
are generated by the human lung adenocarcinoma cell line (A549)-
Asthma control is enhanced (compared with placebo) by neutrophil interactions and are potent inhibitors of cell proliferation.
agents capable of interfering with leukotriene action or Mol Med 2(5): 58396, 1996.
synthesis. Similar data are not available for COPD. 18. Serhan CN, Fiore S, Brezinski DA, Lynch S. Lipoxin-A(4) metabolism
by dierentiated HL-60 cells and human monocytes conversion to
The data indicate that the leukotrienes play an important novel 15-oxo and dihydro products. Biochem 32: 631319, 1993.
role in the asthmatic response. We are not sure of the 19. Guilford WJ, Parkinson JF. Second-generation beta-oxidation resist-
importance of the pathogenetic role of the leukotrienes ant 3-oxa-lipoxin A4 analogs. Prostaglandins Leukot Essent Fatty Acids
in COPD. 73(34): 24550, 2005.
20. Lam BK, Penrose JF, Freeman GJ, Austen KF. Expression cloning of a
Lipoxins are generated in respiratory tissues in asthma cDNA for human leukotriene C4 synthase, an integral membrane pro-
and COPD and can decrease airway inammation and tein conjugating reduced glutathione to leukotriene A4. Proc Natl Acad
hyperresponsiveness in laboratory-induced asthma. Sci USA 91: 766367, 1994.
21. Welsch DJ, Creely DP, Hauser SD, Mathis KJ, Krivi GG, Isakson PC.
Molecular cloning end expression of human leukotriene-C4 synthase.
Proc Natl Acad Sci USA 91: 974549, 1994.
22. Lam BK. Leukotriene C-4 synthase. Prostagland Leuk Essent Fatty
References Acids 69(23): 11116, 2003.
23. Loe DW, Almquist KC, Deely RG, Coles SPC. Multidrug resistance
1. Samuelsson B, Dahlen SE, Lindgren JA, Rouzer CA, Serhan CN. protein (MRP)-mediated transport of leukotriene C4 and chemothera-
Leukotrienes and lipoxins: Structures, biosynthesis, and biological peutic agents in membrane vesicles. J Biol Chem 271: 967582, 1996.
eects. Science 237: 117176, 1987. 24. Gao M, Yamazaki M, Loe DW et al. Multidrug resistant protein:
2. Dennis EA. The growing phospholipase A2 superfamily of signal Identication of regions required for active transport of leukotriene
transduction enzymes. Trends Biochem Sci 22: 12, 1999. C4. J Biol Chem 273: 1073340, 1998.
3. Murakami M, Kudo I. Phospholipase A(2). J Biochem 131(3): 28592, 25. Qian YM, Qiu W, Gao M, Westlake CJ, Cole SPC, Deeley RG.
2002. Characterization of binding of leukotriene C-4 by human multidrug
4. DeMarino V, Gentile M, Granata F, Marone G, Triggiani M. Secretory resistance protein Evidence of dierential interactions with NH2-
phospholipase A(2): A putative mediator of airway inammation. Int and COOH-proximal halves of the protein. J Biol Chem 276(42):
Arch Allergy Immunol 118(24): 2001, 1999. 3863644, 2001.
5. Leslie CC. Properties and regulation of cytosolic phospholipase A2. 26. Bernstrom K, Orning L, Hammarstrom S. Gamma-glutamyl transpepti-
J Biol Chem 272: 1606972, 1997. dase, a leukotriene metabolizing enzyme. Methods Enzymol 86: 3845,
6. Dixon RA, Diehl RE, Opas E et al. Requirement of a 5-lipoxygenase- 1982.
activating protein for leukotriene synthesis. Nature 343: 28284, 27. Hanigan MH, Frierson HF. Immunohistochemical detection of
1990. gamma-glutamyl transpeptidase in normal human tissue. J Histochem
7. Reid GK, Kargman S, Vickers PJ et al. Correlation between expression Cytochem 44(10): 11018, 1996.
of 5-lipoxygenase-activating protein, 5-lipoxygenase, and cellular leu- 28. Han B, Luo GY, Shi ZZ et al. gamma-Glutamyl leukotrienase, a novel
kotriene synthesis. J Biol Chem 265: 1981823, 1990. endothelial membrane protein, is specically responsible for leukotriene
8. Petersgolden M, Brock TG. 5-lipoxygenase and FLAP. Prostagland D-4 formation in vivo. Amer J Pathol 161(2): 48190, 2002.
Leuk Essent Fatty Acids 69(23): 99109, 2003. 29. Lewis RA, Drazen JM, Austen KF, Clark DA, Corey EJ. Identication
9. Haeggstrom JZ. Leukotriene A(4) hydrolase and the committed step of the C(6)-S-conjugate of leukotriene A with cysteine as a naturally
in leukotriene B-4 biosynthesis. Clin Rev Allergy Immunol 17(12): occurring slow reacting substance of anaphylaxis (SRS-A). Importance
11131, 1999. of the 11-cis-geometry for biological activity. Biochem Biophys Res
10. Serhan CN. Resolution phases of inammation: Novel endogenous Commun 96: 27177, 1980.
anti-inammatory and proresolving lipid mediators and pathways. 30. Parker CW, Koch D, Huber MM, Falkenstein SF. Formation of
Annu Rev Immunol, 2006. the cysteinyl form of slow-reacting substance of analyphylaxis
11. Serhan CN, Sheppard KA. Lipoxin formation during human neutrophil- (leukotriene E) on human plasma. Biochem Biophys Res Commun 97:
platelet interactions. Evidence for the transformation of leukotriene A4 103846, 1980.
by platelet 12- lipoxygenase in vitro. J Clin Invest 85: 77280, 1990. 31. Murphy RC, Wheelan P. Pathways of Leukotriene Metabolism in
12. Fiore S, Serhan CN. Formation of lipoxins and leukotrienes during receptor- Isolated Cell Models and Human Subjects. New York: Macel Dekker,
mediated interactions of human platelets and recombinant human granu- Inc, 1998.
locyte/macrophage colony-stimulating factor-primed neutrophils. J Exp 32. Sala A, Voelkel N, Maclouf J, Murphy RC. Leukotriene E4 elimination
Med 172(5): 145157, 1990. and metabolism in normal human subjects. J Biol Chem 265: 2177178,
13. Lee TH, Crea AE, Gant V et al. Identication of lipoxin A4 and its 1990.
relationship to the suldopeptide leukotrienes C4, D4, and E4 in the 33. BandeiraMelo C, Weller PF. Eosinophils and cysteinyl leukotrienes.
bronchoalveolar lavage uids obtained from patients with selected pul- Prostagland Leuk Essent Fatty Acids 69(23): 13543, 2003.
monary diseases. Am Rev Respir Dis 141: 145358, 1990. 34. Boyce JA. The role of mast cells in asthma. Prostagland Leuk Essent
14. Serhan CN. Lipoxins and novel aspirin-triggered 15-epi-lipoxins Fatty Acids 69(23): 195205, 2003.
(ATL): A jungle of cellcell interactions or a therapeutic opportunity? 35. Lynch KR, ONeill GP, Liu Q et al. Characterization of the human
Prostaglandins 53(2): 10737, 1997. cysteinyl leukotriene CysLT1 receptor. Nature 399: 78993, 1999.

289
Asthma and COPD: Basic Mechanisms and Clinical Management

36. Heise CE, ODowd BF, Figueroa DJ et al. Characterization of the mediators from the lungs of asthmatic and control subjects. Chest 95:
human cysteinyl leukotriene 2 receptor. J Biol Chem 275(39): 3053136, 105963, 1989.
2000. 59. Wenzel SE, Larsen GL, Johnston K, Voelkel NF, Westcott JY. Elevated
37. Evans JF. The cysteinyl leukotriene receptors. Prostagland Leuk Essent levels of leukotriene C4 in bronchoalveolar lavage uid from atopic
Fatty Acids 69(23): 11722, 2003. asthmatics after endobronchial allergen challenge. Am Rev Respir Dis
38. Drazen JM. Leukotrienes as mediators of airway obstruction. Amer 142: 11219, 1990.
J Respir Crit Care Med 158(5): S193S200, 1998. 60. Kane GC, Tollino M, Pollice M et al. Insights into IgE-mediated lung
39. Petersgolden M, Canetti C, Mancuso P, Coey MJ. Leukotrienes: inammation derived from a study employing a 5-lipoxygenase inhibi-
Underappreciated mediators of innate immune responses. J Immunol tor. Prostaglandins 50(1): 118, 1995.
174(2): 58994, 2005. 61. Csoma Z, Kharitonov SA, Balint B, Bush A, Wilson NM, Barnes PJ.
40. Kanaoka Y, Boyce JA. Cysteinyl leukotrienes and their receptors: Increased leukotrienes in exhaled breath condensate in childhood
Cellular distribution and function in immune and inammatory asthma. Amer J Respir Crit Care Med 166(10): 134549, 2002.
responses. J Immunol 173: 150310, 2004. 62. Baraldi E, Carraro S, Alinovi R et al. Cysteinyl leukotrienes and
41. Holroyde MC, Altounyan RE, Cole M, Dixon M, Elliott EV. 8-isoprostane in exhaled breath condensate of children with asthma
Bronchoconstriction produced in man by leukotrienes C and D. Lancet exacerbations. Thorax 58(6): 5059, 2003.
2: 1718, 1981. 63. Carraro S, Corradi M, Zanconato S et al. Exhaled breath condensate
42. Weiss JW, Drazen JM, Coles N et al. Bronchoconstrictor eects of leu- cysteinyl leukotrienes are increased in children with exercise-induced
kotriene C in humans. Science 216: 19698, 1982. bronchoconstriction. J Allergy Clin Immunol 115(4): 76470, 2005.
43. Bisgaard H, Groth S, Dirksen H. Leukotriene D4 induces bronchoc- 64. Tagari P, Ethier D, Carry M et al. Measurement of urinary leukotrienes
onstriction in man. Allergy 38: 44143, 1983. by reversed-phase liquid chromatography and radioimmunoassay. Clin
44. Weiss JW, Drazen JM, McFadden ER Jr. et al. Airway constriction Chem 35: 38891, 1989.
in normal humans produced by inhalation of leukotriene D. 65. Taylor GW, Taylor I, Black P et al. Urinary leukotriene E4 after
Potency, time course, and eect of aspirin therapy. JAMA 249: 281417, antigen challenge and in acute asthma and allergic rhinitis. Lancet 1:
1983. 58488, 1989.
45. Barnes NC, Piper PJ, Costello JF. Comparative eects of inhaled leu- 66. Westcott JY. The measurement of leukotrienes in human uids. Clin
kotriene C4, leukotriene D4, and histamine in normal human subjects. Rev Allergy Immunol 17(12): 15377, 1999.
Thorax 39: 5004, 1984. 67. Wu YH, Li LYT, Henion JD, Krol GJ. Determination of LTE(4) in
46. Smith LJ, Greenberger PA, Patterson R, Krell RD, Bernstein PR. The human urine by liquid chromatography coupled with ionspray tandem
eect of inhaled leukotriene D4 in humans. Am Rev Respir Dis 131: mass spectrometry. J Mass Spectrom 31(9): 98793, 1996.
36872, 1985. 68. Higashi N, Taniguchi M, Mita H, Osame M, Akiyama K. A compara-
47. Adelroth E, Morris MM, Hargreave FE, OByrne PM. Airway tive study of eicosanoid concentrations in sputum and urine in patients
responsiveness to leukotrienes C4 and D4 and to methacholine in with aspirin-intolerant asthma. Clin Exp Allergy 32(10): 148490,
patients with asthma and normal controls. N Engl J Med 315: 48084, 2002.
1986. 69. Rabinovitch N, Zhang L, Gelfand EW. Urine leukotriene E-4 levels
48. Roberts JA, Rodger IW, Thomson NC. Eect of verapamil and sodium are associated with decreased pulmonary function in children with
cromoglycate on leukotriene D4 induced bronchoconstriction in persistent airway obstruction. J Allergy Clin Immunol 118(3): 63540,
patients with asthma. Thorax 41: 75358, 1986. 2006.
49. Bisgaard H, Groth S. Bronchial eects of leukotriene D4 inhalation in 70. Maltby NH, Taylor GW, Ritter JM, Moore K, Fuller RW, Dollery CT.
normal human lung. Clin Sci 72: 58592, 1987. Leukotriene C4 elimination and metabolism in man. J Allergy Clin
50. Davidson AB, Lee TH, Scanlon PD et al. Bronchoconstrictor eects Immunol 85: 39, 1990.
of leukotriene E4 in normal and asthmatic subjects. Am Rev Respir Dis 71. Sladek K, Dworski R, Fitzgerald GA et al. Allergen-stimulated release
135: 33337, 1987. of thromboxane A2 and leukotriene E4 in humans Eect of indometh-
51. Smith LJ, Geller S, Ebright L, Glass M, Thyrum PT. Inhibition of leu- acin. Am Rev Respir Dis 141: 144145, 1990.
kotriene D4-induced bronchoconstriction in normal subjects by the 72. Asano K, Lilly CM, Odonnell WJ et al. Diurnal variation of urinary
oral LTD4 receptor antagonist ICI 204, 219. Am Rev Respir Dis 141: leukotriene E4 and histamine excretion rates in normal subjects and
98892, 1990. patients with mild-to- moderate asthma. J Allergy Clin Immunol 96(5
52. Smith LJ, Kern R, Patterson R, Krell RD, Bernstein PR. Mechanism Part 1): 64351, 1995.
of leukotriene D4-induced bronchoconstriction in normal subject. 73. Vachier I, Kumlin M, Dahlen SE, Bousquet J, Godard P, Chanez P.
J Allergy Clin Immunol 80: 34047, 1987. High levels of urinary leukotriene E-4 excretion in steroid treated
53. Grin M, Weiss JW, Leitch AG et al. Eects of leukotriene D on the patients with severe asthma. Respir Med 97(11): 122529, 2003.
airways in asthma. N Engl J Med 308: 43639, 1983. 74. Capra V. Molecular and functional aspects of human cysteinyl leukot-
54. Pichurko BM, Ingram RHJ, Sperling RI et al. Localization of the site riene receptors. Pharmacol Res 50: 111, 2004.
of the bronchoconstrictor eects of leukotriene C4 compared with that 75. Drazen JM, Israel E, Obyrne PM. Treatment of asthma with drugs
of histamine in asthmatic subjects. Am Rev Respir Dis 140: 33439, modifying the leukotriene pathway. N Engl J Med 340(3): 197206,
1989. 1999.
55. Togias AG, Naclerio RM, Peters SP et al. Local generation of sul- 76. Carter GW, Young PR, Albert DH et al. 5-lipoxygenase inhibitory
dopeptide leukotrienes upon nasal provocation with cold, dry air. Am activity of zileuton. J Pharmacol Exp Ther 256: 92937, 1991.
Rev Respir Dis 133: 113337, 1986. 77. Depre M, Friedman B, Vanhecken A et al. Pharmacokinetics and phar-
56. Silber G, Proud D, Warner J et al. In vivo release of inammatory macodynamics of multiple oral doses of MK-0591, a 5-lipoxygenase-
mediators by hyperosmolar solutions. Am Rev Respir Dis 137: 60612, activating protein inhibitor. Clin Pharmacol Ther 56: 2230, 1994.
1988. 78. Gardiner PJ, Cuthbert NJ, Francis HP et al. Inhibition of antigen-
57. Lam S, Chan H, LeRiche JC, Chan-Yeung M, Salari H. Release of induced contraction of guinea-pig airways by a leukotriene synthesis
leukotrienes in patients with bronchial asthma. J Allergy Clin Immunol inhibitor, BAY x1005. Eur J Pharmacol 258: 95102, 1994.
81: 71117, 1988. 79. Israel E, Cohn J, Dube L, Drazen JM. Eect of treatment with zileu-
58. Zehr BB, Casale TB, Wood D, Floerchinger C, Richerson HB, ton, a 5-lipoxygenase inhibitor, in patients with asthma: A randomized
Hunninghake GW. Use of segmental airway lavage to obtain relevant controlled trial. JAMA 275(12): 93136, 1996.

290
Leukotrienes and Lipoxins 24
80. Fuller RW, Black PN, Dollery CT. Eect of the oral leukotriene D4 102. Levy BD, Desanctis GT, Devchand PR et al. Multi-pronged inhibi-
antagonist LY171883 on inhaled and intradermal challenge with tion of airway hyper-responsiveness and inammation by lipoxin
antigen and leukotriene D4 in atopic subjects. J Allergy Clin Immunol A(4). Nat Med 8(9): 101823, 2002.
83: 93944, 1989. 103. Ariel A, Chiang N, Arita M, Petasis NA, Serhan CN. Aspirin-
81. Israel E, Dermarkarian R, Rosenberg M et al. The eects of a triggered lipoxin A4 and B4 analogs block extracellular signal-regulated
5-lipoxygenase inhibitor on asthma induced by cold, dry air. N Engl J kinase-dependent TNF-alpha secretion from human T cells.
Med 323: 174044, 1990. J Immunol 170(12): 626672, 2003.
82. Israel E, Fischer AR, Rosenberg MA et al. The pivotal role of 5-lipox- 104. Colgan SP, Serhan CN, Parkos CA, Delp-Archer C, Madara JL.
ygenase products in the reaction of aspirin-sensitive asthmatics to Lipoxin A4 modulates transmigration of human neutrophils across
aspirin. Am Rev Respir Dis 148: 144751, 1993. intestinal epithelial monolayers. J Clin Invest 92(1): 7582, 1993.
83. Dahlen B, Kumlin M, Margolskee DJ et al. The leukotriene-receptor 105. Serhan CN, Maddox JF, Petasis NA et al. Design of lipoxin A4 stable
antagonist MK-0679 blocks airway obstruction induced by inhaled analogs that block transmigration and adhesion of human neutrophils.
lysine-aspirin in aspirin-sensitive asthmatics. Eur Respir J 6: 101826, Biochemistry 34(44): 1460915, 1995.
1993. 106. Takano T, Fiore S, Maddox JF, Brady HR, Petasis NA, Serhan CN.
84. Dahlen B, Zetterstrom O, Bjorck T, Dahlen SE. The leukotriene- Aspirin-triggered 15-epi-lipoxin A4 (LXA4) and LXA4 stable ana-
antagonist ICI-204,219 inhibits the early airway reaction to cumula- logues are potent inhibitors of acute inammation: Evidence for anti-
tive bronchial challenge with allergen in atopic asthmatics. Eur Respir inammatory receptors. J Exp Med 185(9): 1693704, 1997.
J 7: 32431, 1994. 107. Levy BD, Fokin VV, Clark JM, Wakelam MJ, Petasis NA, Serhan CN.
85. Dahlen SE, Malmstrom K, Nizankowska E et al. Improvement of Polyisoprenyl phosphate (PIPP) signaling regulates phospholipase D
asprinin-intolerant asthma by montelukast, a leukotriene antagonist activity: A stop signaling switch for aspirin-triggered lipoxin A4.
A randomized, double-blink, placebo-controlled trial. Amer J Respir FASEB J 13(8): 90311, 1999.
Crit Care Med 165(1): 914, 2002. 108. Maddox JF, Serhan CN. Lipoxin A4 and B4 are potent stimuli for
86. Menendez R, Venzor J, Ortiz G. Failure of zarlukast to prevent human monocyte migration and adhesion: Selective inactivation by
ibuprofen-induced anaphylaxis. Ann Allergy Asthma Immunol 80(3): dehydrogenation and reduction. J Exp Med 183(1): 13746, 1996.
22526, 1998. 109. Godson C, Mitchell S, Harvey K, Petasis NA, Hogg N, Brady HR.
87. Enrique E, GarciaOrtega P, Gaig P, SanMiguel MM. Failure of monte- Cutting edge: Lipoxins rapidly stimulate nonphlogistic phagocy-
lukast to prevent anaphylaxis to diclofenac. Allergy 54(5): 52930, 1999. tosis of apoptotic neutrophils by monocyte-derived macrophages.
88. Barnes NC. Eects of antileukotrienes in the treatment of asthma. J Immunol 164(4): 166367, 2000.
Amer J Respir Crit Care Med 161(2): S7376, 2000. 110. Aliberti J, Hieny S, Reis eSousa, Serhan CN, Sher A. Lipoxin-mediated
89. Wenzel SE. The role of leukotrienes in asthma. Prostagland Leuk inhibition of IL-12 production by DCs: A mechanism for regulation
Essent Fatty Acids 69(23): 14555, 2003. of microbial immunity. Nat Immunol 3(1): 7682, 2002.
90. Busse WW, Mcgill KA, Horwitz RJ. Leukotriene pathway inhibi- 111. Ramstedt U, Ng J, Wigzell H, Serhan CN, Samuelsson B. Action
tors in asthma and chronic obstructive pulmonary disease. Clin Exp of novel eicosanoids lipoxin A and B on human natural killer cell
Allergy 29: 11015, 1999. cytotoxicity: Eects on intracellular cAMP and target cell binding.
91. Obyrne PM, Israel E, Drazen JM. Antileukotrienes in the treatment J Immunol 135(5): 343438, 1985.
of asthma. Ann Intern Med 127(6): 47280, 1997. 112. Bonnans C, Fukunaga K, Levy MA, Levy BD. Lipoxin A(4) regulates
92. Bisgaard H. Leukotriene modiers in pediatric asthma management. bronchial epithelial cell responses to acid injury. Am J Pathol 68(4):
Pediatrics 107(2): 38190, 2001. 106472, 2006.
93. Celik P, Sakar A, Havlucu Y, Yuksel H, Turkdogan P, Yorgancioglu A. 113. Gewirtz AT, Collier-Hyams LS, Young AN et al. Lipoxin a4 analogs
Short-term eects of montelukast in stable patients with moderate to attenuate induction of intestinal epithelial proinammatory gene
severe COPD. Respir Med 99(4): 44450, 2005. expression and reduce the severity of dextran sodium sulfate-induced
94. In KH, Asano K, Beier D et al. Naturally occurring mutations in the colitis. J Immunol 168(10): 526067, 2002.
human 5-lipoxygenase gene promoter that modify transcriptio factor 114. Canny G, Levy O, Furuta GT et al. Lipid mediator-induced expres-
binding and reporter gene transcription. J Clin Invest 99: 113037, 1997. sion of bactericidal/permeability-increasing protein (BPI) in human
95. Silverman ES, Du J, Desanctis GT et al. Egr-1 and Sp1 interact func- mucosal epithelia. Proc Natl Acad Sci USA 99(6): 39027, 2002.
tionally with the 5-lipoxygenase promoter and its naturally occurring 115. Dahlen SE, Raud J, Serhan CN, Bjork J, Samuelsson B. Biological
mutants. Amer J Respir Cell Mol Biol 19(2): 31623, 1998. activities of lipoxin A include lung strip contraction and dilation of
96. Sanak M, Simon HU, Szczeklik A. Leukotriene C4 synthase pro- arterioles in vivo. Acta Physiol Scand 130(4): 64347, 1987.
moter polymorphism and risk of aspirin-induced asthma. Lancet 350: 116. Fukunaga K, Kohli P, Bonnans C, Fredenburgh LE, Levy BD.
1599600, 1997. Cyclooxygenase 2 plays a pivotal role in the resolution of acute lung
97. Asano K, Shiomi T, Hasegawa N et al. Leukotriene C4 synthase gene injury. J Immunol 174(8): 503339, 2005.
A(444)C polymorphism and clinical response to a CYS-LT1 antag- 117. Christie PE, Spur BW, Lee TH. The eects of lipoxin A4 on airway
onist, in Japanese patients with moderate asthma. Pharmacogenetics 12: responses in asthmatic subjects. Am Rev Respir Dis 145(6): 128184,
16, 2002. 1992.
98. Sayers I, Barton S, Rorke S et al. Promoter polymorphism in the 118. Vachier I, Bonnans C, Chavis C et al. Severe asthma is associated with
5-lipoxygenase (ALOX5) and 5-lipoxygenase-activating protein a loss of LX4, an endogenous anti- inammatory compound. J Allergy
(ALOX5AP) genes and asthma susceptibility in a Caucasian popula- Clin Immunol 115(1): 5560, 2005.
tion. Clin Exp Allergy 33(8): 110310, 2003. 119. Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid medi-
99. Lima JJ, Zhang S, Grant A et al. Inuence of leukotriene pathway ator class switching during acute inammation: Signals in resolution.
polymorphisms on response to montelukast in asthma. Am J Respir Nat Immunol 2(7): 61219, 2001.
Crit Care Med 173(4): 37985, 2006. 120. Sanak M, Levy BD, Clish CB et al. Aspirin-tolerant asthmatics gen-
100. Drazen JM, Yandava C, Dube L et al. Pharmacogenetic association erate more lipoxins than aspirin-intolerant asthmatics. Eur Respir J
between ALOX5 promoter genotype and the response to anti-asthma 16(1): 4449, 2000.
treatment. Nat Gen 22: 17072, 1999. 121. Levy BD, Bonnans C, Silverman ES, Palmer LJ, Marigowda G, Israel E.
101. Levy BD. Lipoxins and lipoxin analogs in asthma. Prostaglandins Diminished lipoxin biosynthesis in severe asthma. Am J Respir Crit
Leukot Essent Fatty Acids 73(34): 23137, 2005. Care Med 172(7): 82430, 2005.

291
Asthma and COPD: Basic Mechanisms and Clinical Management

122. Fiore S, Romano M, Reardon EM, Serhan CN. Induction of functional 135. Kostikas K, Gaga M, Papatheodorou G, Karamanis T, Orphanidou D,
lipoxin A4 receptors in HL-60 cells. Blood 81(12): 3395403, 1993. Loukides S. Leukotriene B4 in exhaled breath condensate and spu-
123. Chiang N, Fierro IM, Gronert K, Serhan CN. Activation of lipoxin tum supernatant in patients with COPD and asthma. Chest 27(5):
A(4) receptors by aspirin-triggered lipoxins and select peptides evokes 155359, 2005.
ligand-specic responses in inammation. J Exp Med 191(7): 1197 136. Wardlaw AJ, Hay H, Cromwell O, Collins JV, Kay AB.
208, 2000. Leukotrienes, LTC4 and LTB4, in bronchoalveolar lavage in bron-
124. Gronert K, Martinsson-Niskanen T, Ravasi S, Chiang N, Serhan CN. chial asthma and other respiratory diseases. J Allergy Clin Immunol 84:
Selectivity of recombinant human leukotriene D(4), leukotriene B(4), 1926, 1989.
and lipoxin A(4) receptors with aspirin-triggered 15-epi-LXA(4) 137. Emov W, Blazhko VI, Voeikova LS, Karanysheva SA, Bondar TN.
and regulation of vascular and inammatory responses. Am J Pathol The leukotriene B4 content of the bronchoalveolar lavage uid and
158(1): 39, 2001. the function of the prostacyclin-thromboxane system in patients with
125. Fiore S, Serhan CN. Lipoxin A4 receptor activation is distinct from variants of chronic bronchitis. Terapevticheskii Arkhiv 62(4): 9496,
that of the formyl peptide receptor in myeloid cells: Inhibition of 1990.
CD11/18 expression by lipoxin A4-lipoxin A4 receptor interaction. 138. Birring SS, Parker D, Brightling CE, Bradding P, Wardlaw AJ,
Biochemistry 34(51): 1667886, 1995. Pavord ID. Induced sputum inammatory mediator concentrations in
126. Chiang N, Serhan CN, Dahlen SE et al. The lipoxin receptor ALX: chronic cough. Am J Respir Crit Care Med 169(1): 1519, 2004.
Potent ligand-specic and stereoselective actions in vivo. Pharmacol 139. Montuschi P. Exhaled breath condensate analysis in patients with
Rev 58(3): 46387, 2006. COPD. Clin Chim Acta 356(12): 2234, 2005.
127. Perretti M, Chiang N, La M et al. Endogenous lipid- and peptide- 140. Prota M, Giorgi RD, Sala A et al. Muscarinic receptors, leukotriene
derived anti-inammatory pathways generated with glucocorticoid B4 production and neutrophilic inammation in COPD patients.
and aspirin treatment activate the lipoxin A4 receptor. Nat Med 8(11): Allergy 60(11): 136169, 2005.
1296302, 2002. 141. Montuschi P, Macagno F, Parente P et al. Eects of cyclo-oxygenase
128. Ohira T, Bannenberg G, Arita M et al. A stable aspirin-triggered inhibition on exhaled eicosanoids in patients with COPD. Thorax
lipoxin A4 analog blocks phosphorylation of leukocyte-specic pro- 60(10): 82733, 2005.
tein 1 in human neutrophils. J Immunol 173(3): 209198, 2004. 142. Crooks SW, Bayley DL, Hill SL, Stockley RA. Bronchial inamma-
129. Coles SJ, Neill KH, Reid LM et al. Eects of leukotrienes C4 and D4 tion in acute bacterial exacerbations of chronic bronchitis: The role of
on glycoprotein and lysozyme secretion by human bronchial mucosa. leukotriene B-4. Eur Respir J 15(2): 27480, 2000.
Prostaglandins 25: 15570, 1983. 143. Matsuyama W, Mitsuyama H, Watanabe M et al. Eects of omega-3
130. Piacentini GL, Kaliner MA. The potential roles of leukotrienes in polyunsaturated fatty acids on inammatory markers in COPD. Chest
bronchial asthma. Am Rev Respir Dis 143: S96S99, 1991. 128(6): 381727, 2005.
131. Marian E, Baraldo S, Visentin A et al. Up-regulated membrane and 144. Rubinstein I, Kumar B, Schriever C. Long-term montelukast therapy
nuclear leukotriene B4 receptors in COPD. Chest 129(6): 152330, 2006. in moderate to severe COPD a preliminary observation. Respir Med
132. Peters-Golden M, Coey M. Role of leukotrienes in antimicrobial 98(2): 13438, 2004.
host defense of the lung. Clin Rev Allergy Immunol 17(12): 26169, 145. Zuhlke IE, Kanniess F, Richter K et al. Montelukast attenuates the
1999. airway response to hypertonic saline in moderate-to-severe COPD.
133. Hoshino M. Impact of inhaled corticosteroids and leukotriene recep- Eur Respir J 22(6): 92630, 2003.
tor antagonists on airway remodeling. Clin Rev Allergy Immunol 27(1): 146. Nannini LJ, Flores DM. Bronchodilator eect of zarlukast in sub-
5964, 2004. jects with chronic obstructive pulmonary disease. Pulm Pharmacol Ther
134. Busse W, Kraft M. Cysteinyl leukotrienes in allergic inammation 16(5): 30711, 2003.
Strategic target for therapy. Chest 127(4): 131226, 2005.

292
Reactive Oxygen Species and Antioxidant
Therapeutic Approaches
25 CHAPTER

INTRODUCTION sites of inammation are a major cause of the Irfan Rahman


cell and tissue damage associated with many
Department of Environmental Medicine,
chronic inammatory lung diseases, including
asthma and chronic obstructive pulmonary dis- Lung Biology and Disease Program,
The lung is constantly exposed to a high-oxygen
environment and, owing to its large surface area ease (COPD) [59] (Fig. 25.1). University of Rochester Medical Center,
and blood supply, is highly susceptible to injury The composition of inammatory cell Rochester, NY, USA
mediated by a phenomenon known as oxidative types that invade tissues varies widely in asthma
stress. Oxidative stress is described simply as an and COPD; this suggests that there are dier-
increase in the oxidant to antioxidant ratio in ences in the characteristics of the ROS pro-
the lung. Reactive oxygen species (ROS) such as duced in these diseases [1012]. This chapter
superoxide anion (O2) and the hydroxyl radi- reviews the evidence for the role of ROS in the
cal (OH) are unstable molecules with unpaired pathogenesis of asthma and COPD, and dis-
electrons, capable of initiating the oxidation of cusses the molecular mechanisms (cell signaling
various molecules in the airway and airspace and gene expression) and pathophysiological
structures. consequences of increased ROS release in these
Biological systems are continuously conditions. Moreover, it also highlights the anti-
exposed to oxidants that are generated either oxidant mechanisms in place to protect against
endogenously by metabolic reactions (e.g., from the damaging eects of ROS. Finally, it goes on
mitochondrial electron transport during res- to explore possible therapeutic approaches that
piration or during activation of phagocytes) or involve the use of a variety of antioxidants.
exogenously (such as by air pollutants or ciga-
rette smoke). For example, production of ROS
has been directly linked to oxidation of proteins,
DNA, and lipids which may cause direct lung
injury or induce a variety of cellular responses,
CELL-DERIVED ROS
through the generation of secondary metabolic
reactive species. ROS may result in remod- A common feature of all inammatory lung dis-
eling of extracellular matrix, cause apoptosis eases is the development of an inammatory
and mitochondrial respiration, and regulate cell immune response, characterized by activation of
proliferation [1, 2]. Alveolar repair responses epithelial cells, and resident macrophages, and
and immune modulation in the lung may also the recruitment and activation of neutrophils,
be inuenced by ROS [1, 2]. Furthermore, high eosinophils, monocytes, and lymphocytes. The
levels of ROS have been implicated in initiating degree to which this occurs and the cell types
inammatory responses in the lungs through the involved vary widely in asthma and COPD.
activation of transcription factors such as nuclear Inammatory cells once recruited in the air-
factor-B (NF-B) and activator protein-1 space become activated and generate ROS in
(AP-1); their activation leads to signal transduc- response to a sucient stimulus (threshold
tion and gene expression of pro-inammatory concentration). The activation of macrophages,
mediators [3, 4]. It is proposed that ROS pro- neutrophils, and eosinophils generates O2,
duced by phagocytes that have been recruited to which is rapidly converted to H2O2 through

293
Asthma and COPD: Basic Mechanisms and Clinical Management

Environmental the catalytic action of superoxide dismutase (SOD); OH is


Cellular Cigarette smoke, formed nonenzymatically in the presence of Fe2 as a sec-
NO2, SO2, ondary reaction. ROS and reactive nitrogen species (RNS)
MPO EPO particulates can also be generated intracellularly from several sources
PMNs Eos AMs such as mitochondrial respiration, the NADPH oxidase
system, and xanthine/xanthine oxidase (XO) (Fig. 25.2).
Epithelium However, the primary ROS generating system is NADPH
oxidase, a complex enzyme system that is present in phago-
cytes and epithelial cells.
Activation of this enzyme system involves a com-
Mitochondrial plex mechanism with the assembly of various cytosolic
NADPH respiration
oxidase
and membrane-associated subunits, resulting in the one-
electron reduction of oxygen to O2 using NADPH as the
Xanthine electron donor. In addition to NADPH oxidase, phagocytes
oxidase Oxidative
use heme peroxidases such as myeloperoxidase (MPO) or
stress eosinophil peroxidase (EPO) to produce ROS. Activation
of EPO results in the formation of the potent oxidants
hypochlorous acid (HOCl) and hypobromous acid (HOBr)
from H2O2 in the presence of chloride (Cl) and bromide
(Br) ions, respectively. It is believed that the oxidant bur-
den produced by eosinophils is substantial because these
Inflammation cells possess several times greater capacity to generate O2
and H2O2 than do neutrophils, and the content of EPO in
eosinophils is 310 times higher than the amount of MPO
FIG. 25.1 Sources of oxidative stress. Oxidative stress derived from either present in neutrophils [1315] (Fig. 25.3).
environmental or cellular origins results in inflammation. Cellular-derived The physiological consequences of EPO-dependent
oxidative stress can be produced through mitochondrial respiration, the formation of brominating oxidants such as HOBr in vivo
NADPH oxidase system, or the xanthine oxidase system. Inflammation itself are unknown. HOBr reacts rapidly with a variety of nucle-
can have a feed-forward effect, triggering inflammatory cells to produce ophilic targets such as thiols, thiol ethers, amines, unsatu-
yet more oxidative stress, exacerbating and intensifying the inflammatory rated groups, and aromatic compounds [16].
response. PMNs: polymorphonucleocytes; Eos: eosinophils; AMs: alveolar
macrophages.

Protein nitration and


Cellular NO ONOO nitrosylation, lipid peroxidation,
ROS tissue destruction

O2
SOD
Environmental 2H2OO2
ROS Catalase
O2 Haber-
Weiss H2O2
chemistry Glutathione
peroxidase
X Fe2
X GSSG
Fe3
Myeloperoxidase  2H2O
Glutathione Fenton
transferase chemistory

Fe3 Fe2
OH HOCI
GS-X O2

Tissue dysfunction/Destruction
protein, lipids, carbohydrates, DNA

FIG. 25.2 Molecular consequences of oxidative stress. The relatively weak superoxide anion from both cellular and environmental sources can be
transformed into more damaging and potent reactive oxygen and reactive nitrogen species, such as HOCl, the hydroxyl radical and peroxynitrite, through
a series of enzymatic and nonenzynmatic steps. Xenobiotic radicals from the environment can be long lived and undergo redox cycling, such as the
semiquinones, and result in further superoxide anion formation as well as more powerful radical formation in the presence of free metal ions through
HaberWeiss and Fenton chemistry. Endogenous antioxidant defenses glutathione transferase, GPx, SOD, and catalase neutralize and remove these ROS and
RNS. ROS: reactive oxygen species; RNS: reactive nitrogen species; X: xenobiotic radical; O2 : superoxide anion; H2O2: hydrogen peroxide; OH: hydroxide
radical; HOCl: hypochlorous acid; NO: nitric oxide; ONOO: peroxynitrite; GSH: reduced glutathione; GSSG: oxidized glutathione dimers.

294
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
O2

O2 H2O2 NO2 [NOx]

Eosinophil peroxidase

Br H2O2 HOBr


Eosinophil Bromination

Nitration

O2 CO2

NO ONOO
Epithelium

FIG. 25.3 Generation of reactive nitrogen species. Model of potential pathways used by eosinophils and airway epithelial cells for generation of NO-derived
reactive nitrogen and halogen species leading to nitration of proteins.

Several transition metal salts react with H2O2 to form Quinone (Q), hydroquinone (QH2), and semiquinone

OH. Most attention in vivo for the generation of OH has (QH) in the tar phase are present in equilibrium:
focused on the role of iron [17]. Iron is a critical element in
many oxidative reactions. Free iron in the ferrous form cata- Q  QH 2 2H  2Q.
lyzes the Fenton reaction and the superoxide-driven Haber
Weiss reaction, which generate the OH, a ROS which Aqueous extracts of cigarette tar contain the quinone
damages tissues, particularly cell membranes by peroxidation radical (Q), which can reduce oxygen to form O2, which
of their lipid membranes. MPO- and EPO-derived ROS can may dismutate in the presence of H to form H2O2:
also interact with nitrite (NO2) and H2O2 to promote for-
mation of RNS. ROS may also be released by lung epithelial Q  O 2 Q  O 2
cells [18] and stimulate inammatory cells directly, thereby
amplifying lung inammatory and oxidant events. ROS inter- 2O 2  2H O 2  H 2 O 2 .
act with a variety of molecules and donate electrons in biolog-
ical systems. Reactive oxygen and nitrogen species also act on Furthermore, since both cigarette tar and lung ELF
certain amino acids such as methionine, tyrosine, and cysteine contain metal ions, such as iron, Fenton chemistry will
in proteins (e.g. enzymes, kinases), profoundly altering the result in the production of the OH which is a highly reac-
function of these proteins in inammatory lung diseases [19]. tive and potent ROS. Thus, oxidants and electrophilic
compounds may be the initiating or triggering factor in
the underlying inammation seen in the pathogenesis of
COPD as detailed below.
INHALED OXIDANTS AND CIGARETTE SMOKE
Cigarette smoking, or inhalation of airborne pollutants
that may be either oxidant gases [such as ozone, nitrogen
ROS AND MEMBRANE LIPID PEROXIDATION
dioxide (NO2), sulfur dioxide (SO2)] or particulate air pol-
lution, results in direct lung damage as well as the activation Oxygen species such as O2 and OH are highly reactive,
of inammatory responses in the lungs. Cigarette smoke and when generated close to cell membranes they oxidize
is a complex mixture of over 4700 chemical compounds, membrane phospholipids (lipid peroxidation), a proc-
including high concentrations of oxidants (1014 molecules ess which may continue as a chain reaction. Thus, a single
per pu ) [20]. Short-lived oxidants such as O2 and nitric
OH can result in the formation of many molecules of lipid
oxide (NO) are predominantly found in the gas phase. NO hydroperoxides in the cell membrane [19]. The peroxidative
and O2 immediately react to form the highly reactive per- breakdown of polyunsaturated fatty acids impairs mem-
oxynitrite (ONOO) molecule. The radicals in the tar phase brane function, inactivates membrane-bound receptors
of cigarette smoke are organic in nature, such as long-lived and enzymes, and increases tissue permeability. Each of
semiquinone radicals, which can react with O2 to form these processes has been implicated in the pathogenesis of

OH and H2O2 [21]. The aqueous phase of cigarette smoke many forms of lung injury. There is increasing evidence that
condensate may undergo redox recycling for a considerable aldehydes, generated endogenously during the process of
period of time in the epithelial lining uid (ELF) of smok- lipid peroxidation, are involved in many of the pathophysi-
ers [22, 23]. The tar phase is also an eective metal chelator ological eects associated with oxidative stress in cells and
and can bind iron to produce tar-semiquinone  tar-Fe2, tissues [19]. Compared with free radicals, lipid peroxidation
which can generate H2O2 continuously [22, 23]. aldehydes are generally stable, can diuse within, or even

295
Asthma and COPD: Basic Mechanisms and Clinical Management

escape from the cell, and attack targets far from the site of proteins (CREB), CREB-binding protein (CBP), and Elk-
the original free radical event. In addition to their cytotoxic 1 [3740]. This eventually results in chromatin remodeling
properties, lipid peroxides are increasingly recognized as and expression of genes regulating a battery of distinct
being important in signal transduction for a number of pro-inammatory and antioxidant genes involved in sev-
important events in the inammatory response [24]. eral cellular events, including apoptosis, proliferation, trans-
Many of the eects of ROS in airways may be medi- formation, and dierentiation. The intracellular molecular
ated by the secondary release of inammatory lipid media- mechanisms responsible for these actions of ROS have not
tors such as 4-hydroxy-2-nonenal (4-HNE). 4-HNE, a been completely characterized.
highly reactive diusible end-product of lipid peroxidation, Redox-sensitive molecular targets usually contain
is known to induce/regulate various cellular events, such as highly conserved cysteine residues; their oxidation, nitration,
proliferation, apoptosis, and activation of signaling path- and the formation of disulde links are crucial events in
ways [24, 25]. 4-HNE has a high anity toward cysteine, oxidant/redox signaling. It is hypothesized that oxidation of
histidine, and lysine residues. It forms adducts with pro- sulde groups in signaling proteins causes structural modi-
teins, altering their function. Acrolein is another example cations, resulting in the exposure of active sites and con-
of reactive aldehyde. Both acrolein and 4-HNE have a high sequent protein activation. Such molecular targets include
anity toward cysteine, histidine, and lysine residues. These transcription factors (NF-B, AP-1), signaling molecules
reactive aldehydes can form adducts with both intracellu- such as ras/rac or JNK, protein tyrosine phosphatases, and
lar proteins, such as histone deacetylase (HDAC) 2 [26] p21ras. Thiol molecules such as intracellular glutathione
and extracellular proteins, such as collagen and bronec- (GSH) and thioredoxin are of central importance in regu-
tin [27] altering their function which in turn can then lating such redox signaling pathways, by reducing disulde
impact on cell function [27, 28]. In contrast, the isopros- bridges or oxidized cysteine residues [37, 41].
tanes, are ROS-catalyzed oxidative products of arachidonic In response to tumor necrosis factor (TNF-) and
acid metabolism and are stable lipid peroxidation products, lipopolysaccharide (LPS), which are relevant stimuli for the
which circulate in plasma and are excreted in the urine inammatory response in COPD, airway epithelial cells
[29, 30]. It is because of their relative stability in vivo that can concurrently produce increased amounts of intracellular
they have been used as markers of oxidative stress in both ROS and RNS [18]. This intracellular production of oxi-
asthma and COPD [31]. However, one isoprostane mem- dants and the subsequent changes in intracellular redox
ber, 8-isoprostane, has been shown to possess a very potent status are important in the molecular events controling the
biological activity. It has been demonstrated that 8-isopros- expression of genes for inammatory mediators [3]. The
tane is a very potent stimulus for smooth muscle contrac- signaling pathways and activation of transcription factors in
tion through initiation of transduction at the thromboxane response to ROS are the subject of rigorous investigation.
A2 receptor; this could be one of the many causes of small
airway contraction in asthma and COPD [32, 33]. ROS-
induced degradation of arachidonate-based phospholipids
can also produce other bio-active molecules. They are pro-
inammatory aecting both monocytes and neutrophils
causing increased endothelial cell interaction [34] as well as
ROS IN ASTHMA AND COPD
increased cytokine release [35, 36], and it is proposed that
they may play an important role in various chronic inam- In asthma, the airways are the major site of action, char-
matory diseases [34]. acterized by reversible airow obstruction, airway hyper-
responsiveness and hyperreactivity, chronic inammation
characterized by a predominant infusion of lungs with eosi-
nophils, and the inux and activation of inammatory cells
ROLE OF ROS IN SIGNAL TRANSDUCTION such as macrophages, neutrophils, lymphocytes, and mast
cells. Airway smooth muscle contraction, increased airway
reactivity and secretions, increased vascular permeability,
ROS have been implicated in the activation of transcrip- and increased generation of chemoattractants are the major
tion factors such as NF-B and AP-1, and in the signal features of an asthmatic response.
transduction and gene expression involved in cellular pro- In COPD, the major site of attack is the alveo-
inammatory actions [37]. Both environmental and inam- lar spaces. Inammatory cells become compartmental-
matory cell-derived ROS can lead to the activation and ized depending on the cell type. For example, neutrophils
phosphorylation of the mitogen-activated protein kinase are mainly located in the airspaces of the lung, and mac-
(MAPK) family, including extracellular signal regulated rophages accumulate in the tissue matrix in COPD.
kinase (ERK), c-Jun N-terminal kinase ( JNK), p38 kinase, Progression of COPD has been strongly associated with the
and P1-3K, via sensitive cysteine-rich domains; activation accumulation of inammatory mucus exudates in the lumen
of the sphingomyelinaseceramide pathway also occurs of the small airways. In other phenotypes of COPD, oxida-
leading to increased gene transcription [3, 4, 37]. Activation tive stress occurs in the small airways, lung parenchyma, and
of members of the MAPK family leads to the transactiva- alveolar regions. Hence the compartmentalization of dier-
tion of transcription factors such as c-Jun, activating fac- ent inammatory cells in the lungs may help distinguish the
tor-2 (ATF2), cyclic AMP response element binding oxidative stress response in asthma and COPD.

296
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
OXIDATIVE STRESS IN ASTHMA when the epithelium is injured or removed. This observa-
tion might provide a mechanistic link between epithelial
injury arising from a variety of causes and airway hyper-
It has been shown that inammation driven by increased responsiveness [47]. ROS also stimulate histamine release
oxidative stress occurs in the airways of patients with from mast cells and mucus secretion from airway epithelial
asthma [8]. Recent evidence indicates that increased oxi- cells [63].
dative stress occurs in the airways of patients with asthma ROS generation is thought to be a nonspecic proc-
[8]. Inammatory and immune cells in the airways, such as ess initiated by the concurrent action of numerous inam-
macrophages, neutrophils, and eosinophils, release increased matory mediators that have been shown to be present in
amounts of ROS [4245]. ROS can result in lung injury as increased amounts in asthmatics. Several mediators, includ-
a result of direct oxidative damage to epithelial cells and cell ing lipid mediators, chemokines, adhesion molecules, and
shedding [46, 47]. eosinophil granule proteins (EPO), are potential stimuli or
ROS have been shown to be associated with the promoters of ROS production in the airways of asthmatic
pathogenesis of asthma by evoking bronchial hyperreactiv- patients [64, 65].
ity [48, 49]. Viral infections, ozone, and cigarette smoke, Numerous surrogate markers of oxidative stress have
potential triggers for asthma, may serve as sources of ROS been measured in exhaled air or breath condensate. The con-
which enhance inammation and asthmatic symptoms. centration of H2O2 in exhaled air condensate is increased
Animal studies suggest that ROS may contribute to airway in asthmatics [66], and it has been suggested that airway
hyperresponsiveness by increasing vagal tone due to inhi- inammation increases exhaled peroxides. The increased
bition of -adrenergic receptors and by decreasing muco- levels of exhaled H2O2 may be due to decreased dismuta-
ciliary clearance [50, 51]. The actions of ROS can produce tion of O2 since SOD activity is reduced in lung cells of
many of the pathophysiological features of asthma [51], patients with asthma [57]. The signicance of elevated lev-
including changes in the biochemical microenvironment els of various ROS markers to the disease pathogenesis has
with enhanced arachidonic acid release, increased synthesis not been studied.
of chemoattractants, glucocorticoid resistance, and impaired
-adrenergic responsiveness. These and other eects lead to
the physiological changes of airway smooth muscle con- The role of EPO-derived ROS
traction, increased airway reactivity and secretions, and
increased vascular permeability. A specic role for EPO in the generation of oxidants
ROS-mediated injury to the airway epithelium pro- by the phagocytes and in protein oxidation has been
duces hyperresponsiveness of human peripheral airways, described [67]. Eosinophil activation (peroxidase H2O2
suggesting that ROS may play a role in the pathogenesis system  halides) in vivo results in oxidative damage
of asthma [47]. Much of the evidence for this is indirect, to proteins through bromination of tyrosine residues, as
since there are no specic and reliable methods to assess shown by the formation of 3-bromotyrosine in BAL uid
oxidative stress in vivo. Neutrophils isolated from periph- of patients with asthma [16]. The formation of 3-bromoty-
eral blood of asthmatic patients generate greater amounts rosine is a specic response to the release of oxidants from
of O2 and H2O2 than do cells from normal subjects, and eosinophils. Neutrophil- and monocyte-derived MPO,
their ability to produce O2 is related directly to the degree which are increased in smokers and patients with COPD,
of airway hyperresponsiveness to inhaled methacholine [52, produce 3-chlorotyrosine [16]. Thus, distinction between
53]. Inammatory cells obtained from asthmatic patients, these footprints might be useful in assessing the ROS bur-
particularly eosinophils derived from peripheral blood, den in patients with asthma and COPD. A specic marker
produce increased amounts of ROS and RNS such as NO of protein modication by reactive brominating species
spontaneously and after stimulation ex vivo [4246, 54]. 3-bromotyrosine has been shown to be markedly increased
This observation suggests that the inammatory milieu in in BAL proteins obtained from asthmatics [16]. EPO-
asthma contains factors that may prime ROS generation. generated oxidants can interact with RNS present in asthmat-
Other studies have shown that impaired SOD activity is ics and promote protein nitration [15]. Thus, oxidative modi-
associated with airow obstruction, airway hyperresposive- cation of critical biological targets in asthmatic airways may
ness, and remodeling [55, 56]. This inactivation of SOD in contribute to the pathophysiological features of asthma, such
asthmatics, which has been reported in other studies [57, as epithelial cell damage, airway hyperreactivity, bronchocon-
58], was as a result of increased ROS causing tyrosine nitra- striction, -adrenergic receptor dysfunction, mucus hyperse-
tion of SOD. cretion, microvascular leak, and airway edema [68, 69].
Eosinophils are thought to play a critical role in the
inammation of asthma. They are present in large numbers
in bronchoalveolar lavage (BAL) uid and blood, and the Interaction between ROS and RNS
number of cells correlates with bronchial hyperresponsive-
ness [59, 60]. BAL uid eosinophils, alveolar macrophages, The levels of nitric oxide are elevated in the exhaled air of
and neutrophils from asthmatic patients produce more patients with asthma [7073]. Increased levels of exhaled
ROS (O2, H2O2, hypothalites) than do those from nor- NO, together with increased exhaled H2O2 in asthmatic
mal subjects [61, 62]. ROS cause direct contraction of air- patients, are associated with a pro-oxidant activity in air-
way smooth muscle preparations, and this eect is enhanced way walls resulting in lipid peroxidation and nitration of

297
Asthma and COPD: Basic Mechanisms and Clinical Management

proteins [15, 16, 70, 74, 75]. The nding that NO reduces TBARS) are increased in EBC of patients with asthma
the potency of -adrenergic signaling pathways may be an [8689]. The levels of 8-isoprostane are also increased in
important deleterious eect of elevated RNS in asthma BAL uid of patients with asthma [90]. Urinary excretion
[50]. The presence of allergic inammation, involving the of 15-F2t-isoprostane (8-isoprostaglandin2 , family of F2-
recruitment and activation of eosinophils, may be a contrib- isoprostanes) was increased in mild atopic asthmatics fol-
uting factor in these pro-oxidant eects. Eosinophil granule lowing inhaled allergen provocation, whereas no increase
proteins may participate in the formation of nitrating oxi- in the urinary excretion of 15-F2t-isoprostane was observed
dants as well as unique molecules such as brominated prod- after inhalation of methacholine [91]. The presence of vari-
ucts, which have been shown to be elevated in patients with ous markers of oxidative stress provide evidence that oxida-
asthma [15, 16]. However, the signicance of these inam- tive stress is present in the airspaces in asthma, but do not
matory ROS in the etiology of asthma is not established. provide denitive evidence for a role for oxidative stress in
A reaction between NO and O2 results in the forma- the pathogenesis of asthma.
tion of peroxynitrite anions (ONOO), a highly reactive oxi- Measurement of systemic or exhaled isoprostane 8-iso-
dant species. ONOO adds a nitro group to the 3-position PGF2 levels may provide a useful tool in monitoring clinical
adjacent to the hydroxyl group of tyrosine to produce the status in asthma [92]. The level of plasma F2-isoprostanes (an
stable product nitrotyrosine. ONOO induces hyperre- 8-isoPGF2 isomer) is signicantly increased in asthmatics
sponsiveness in airways of guinea pigs, inhibits pulmonary and is related to disease severity [92]. Similarly, arachidonic
surfactant function, induces membrane lipid peroxidation, acid is increased in airway inammatory cells of patients
results in tyrosine/MAP kinase activation, and damages with bronchial asthma [93]. Arachidonic acid may undergo
pulmonary epithelial cells [73, 7578]. The levels of 3- oxidation to produce an end-product of lipid peroxidation
nitrotyrosine are elevated in the exhaled breath of asthmatic such as 4-HNE [24, 25]. The levels of plasma lipid perox-
patients [79]. Furthermore, there is strong immunoreactiv- ides TBARS have been shown to be elevated in asthma, and
ity for nitrotyrosine in the airway epithelium, lung paren- are negatively correlated with the Forced Expiratory Volume
chyma, and inammatory cells in the airways of patients in one sec (FEV1), suggesting a role for increased oxidative
with asthma [79, 80]. stress in the pathogenesis of the airway obstruction of asthma
It has been postulated that increased levels of HOBr [94, 95]. Further studies are needed to dene the source, sig-
production as a result of EPO release from eosinophils nicance, and specicity of these peroxidation products.
result in increased peroxynitrite formation by interaction
of HOBr with NO. NO itself can be stored in cells as s-
nitrosothiols [81], and this may regulate cellular apoptosis
through inactivation of caspases by s-nitrosylation of critical
cysteine residues [82]. Consequently, increased ONOO OXIDATIVE STRESS IN COPD
formation could deplete intracellular stores of NO, lib-
erating active caspases that can then induce epithelial cell More than 90% of patients with COPD are smokers, but
apoptosis. This is supported by the evidence showing that s- not all smokers develop COPD [9698]. However, 1520%
nitrosothiol levels in asthmatics are signicantly depressed of cigarette smokers show a rapid decline in FEV1 over time
[83, 84]. However, there are other data, which show that and develop COPD. An increased oxidant burden in smokers
levels of NO in exhaled breath condensate (EBC) are derives from the fact that cigarette smoke contains an esti-
increased in asthmatics, that appear to conict with this mated 1014 oxidants per pu, and many of these are relatively
supposition [71]. This may simply reect a compartmentali- long-lived such as tar-semiquinone that can generate OH
zation eect in that dierent mechanisms are operating in and H2O2 by the Fenton reaction [2023]. Other factors that
an intracellular versus extracellular environment. The precise may exacerbate COPD, such as air pollutants, infections, and
mechanistic role that SOD inactivation by ONOO plays occupational dusts, also have the potential to produce oxida-
in asthma is as yet unclear. However, it is postulated that tive stress [5, 99]. These include oxidative stress, which has
SOD inactivation by ONOO could increase the over- important consequences on lung physiology and for the
all redox state by allowing higher levels of H2O2 to persist pathogenesis of COPD including increased sequestration
thereby depleting intracellular NO stores through increased of neutrophils in the pulmonary microvasculature, increased
ONOO formation and lowering s-nitrosothiol levels [85]. oxidative inactivation of antiproteases, mucus hypersecretion,
Overall, this clearly shows that oxidant stress occurs in alteration in mitochondrial respiration, alveolar epithelial
asthma which can be reected/detected in the lungs both injury or permeability, breakdown and remodeling of extra-
systemically and locally. cellular matrix, apoptosis of epithelial and endothelial cells,
skeletal muscle dysfunction fatigue and exercise limitation,
and glucocorticoid resistance.
Generation of ROS-mediated lipid
peroxidation products
ROS in the alveolar space
The measurement of aldehydes in exhaled breath has been
proposed as a means to assess lipid peroxidation in vivo. The The oxidant burden in the lungs is enhanced in smokers by
levels of lipid peroxidation products (8-isoprostanes), hydro- the release of ROS from macrophages and neutrophils [5].
carbons (ethane and pentane), and nonspecic products of Oxidants present in cigarette smoke can stimulate alveo-
lipid peroxidation (thiobarbituric acid reactive substances, lar macrophages to produce ROS and to release a host of

298
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
mediators, some of which attract neutrophils and other of brominating oxidant in eosinophil-mediated ROS damage
inammatory cells into the lungs. Both neutrophils and mac- in COPD.
rophages, which are known to migrate in increased numbers Superoxide anion and H2O2 can be generated by the
into the lungs of cigarette smokers compared with nonsmok- xanthine/XO reaction. XO activity has been shown to be
ers [5], can generate ROS via the NADPH oxidase system. increased in cell-free BAL uid and plasma from COPD
Moreover, the lungs of smokers with airway obstruction have patients, compared with normal subjects; and this has been
more neutrophils than smokers without airway obstruction associated with increased O2 and lipid peroxide levels
[100]. Circulating neutrophils from cigarette smokers and [117119].
patients with exacerbations of COPD release more O2
[95]. Cigarette smoking is associated with increased content
of MPO in neutrophils, which correlates with the degree of ROS in blood
pulmonary dysfunction [101, 102]. MPO activity is nega-
tively correlated with FEV1 in patients with COPD, sug- The neutrophil appears to be a critical cell in the pathogen-
gesting that neutrophil MPO-mediated oxidative stress may esis of COPD [120]. Previous epidemiological studies have
play a role in the pathogenesis of the airway obstruction in shown a relationship between circulating neutrophil numbers
COPD [103]. and FEV1 [121, 122]. A relationship has also been shown
Alveolar macrophages obtained by BAL uid from between the changes in peripheral blood neutrophil count
the lungs of smokers release increased amounts of ROS such and in airow limitation over time [122]. Similarly, a corre-
as O2 and H2O2 in response to stimulation [5, 104, 105]. lation between O2 release by peripheral blood neutrophils
Exposure to cigarette smoke in vitro has also been shown to and bronchial hyperreactivity in patients with COPD has
increase the oxidative metabolism of alveolar macrophages been shown, suggesting a role for systemic ROS in the
[106]. Subpopulations of alveolar macrophages with a higher pathogenesis of the airway abnormalities in COPD [123].
granular density appear to be more prevalent in the lungs of Another study has shown a relationship between peripheral
smokers and are responsible for the increased O2 produc- blood neutrophil luminol-enhanced chemiluminescence, as a
tion of smokers macrophages [106, 107]. measure of the release of ROS, and measurements of airow
Hydrogen peroxide, measured in exhaled breath, is limitation in young cigarette smokers [124].
thought to be a direct measurement of oxidant burden in Various studies have demonstrated increased produc-
the airspaces. Smokers and patients with COPD have tion of O2 from peripheral blood neutrophils obtained
higher levels of exhaled H2O2 than nonsmokers [108110], from patients during acute exacerbations of COPD; these
and levels are even higher during exacerbations of COPD levels returned to normal when the patients were clinically
[110]. The source of the increased H2O2 is unknown but stable [95, 125, 126]. Other studies have shown that circu-
may in part derive from increased release of O2 from alve- lating neutrophils from patients with COPD show upregu-
olar macrophages in smokers [110], However, in one study lation of their surface adhesion molecules, which may also
smoking did not appear to inuence the levels of exhaled be an oxidant-mediated eect [95, 127]. Activation may be
H2O2 [108]; the levels of exhaled H2O2 in this study corre- even more pronounced in neutrophils that are sequestered in
lated with the degree of airow obstruction as measured by the pulmonary microcirculation in smokers and in patients
the FEV1. However, the variability of the measurement of with COPD, since neutrophils sequestered in the pulmonary
exhaled H2O2, along with the presence of other confound- microcirculation in animal models of lung inammation
ing factors, such as cigarette smoking and caeine intake, release more ROS than circulating neutrophils [128]. Thus,
has led to concerns over the reproducibility of the use of neutrophils sequestered in the pulmonary microcirculation
exhaled H2O2 as a marker for oxidative stress in smokers may be a source of ROS and may have a role in inducing
and in patients with COPD. The generation of ROS in endothelial adhesion molecule expression in COPD.
ELF may be further enhanced by the presence of increased
amounts of free iron in the airspaces in smokers [111, 112].
This is relevant to COPD since the intracellular iron con- Interaction between ROS and RNS
tent of alveolar macrophages is increased in cigarette smok-
ers and is increased further in those who develop chronic Nitric oxide has been used as a marker of airway inam-
bronchitis, compared with nonsmokers [113]. In addition, mation and indirectly as a measure of oxidative stress. There
macrophages obtained from smokers release more free iron have been reports of increased levels of NO in exhaled
in vitro than those from nonsmokers [114]. breath in patients with COPD, but not so high as the lev-
In some studies, in both stable bronchitis [12] and mild els reported in asthmatics [129131]. One study failed to
exacerbations [10], eosinophils have been shown to be promi- conrm this result [132]. Smoking cessation increases NO
nent in the airways. BAL uid from patients with COPD has levels in exhaled air [133], and the reaction of NO with
also been shown to contain increased eosinophilic cationic O2 limits the usefulness of this marker in COPD, except
protein [102]. Furthermore, peripheral blood eosinophilia is perhaps to dierentiate from patients with COPD from
also considered to be a risk factor for the development of air- those with asthma (Table 25.1).
way obstruction in patients with chronic bronchitis and is an Cigarette smoking increases the formation of RNS
adverse prognostic sign [115, 116]. However, despite the pres- and results in nitration and oxidation of plasma proteins.
ence of an increased number of eosinophils, EPO-mediated The levels of nitrated proteins (brinogen, transferrin,
generation of 3-bromotyrosine has not been detected in plasminogen, and ceruloplasmin) were higher in smokers
COPD patients [16]. This does not provide support for a role than in nonsmokers [142]. Evidence of enhanced NO and

299
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 25.1 ROS markers in asthma and COPD.

References

Biochemical marker Asthma COPD

Elevated hydrogen peroxide level in exhaled breath [66, 75] [108110]

Release of ROS from peripheral blood neutrophils, eosinophils, and macrophages [4246, 54, 61] [95, 121, 123, 126]

Increased release of ROS from alveolar macrophages, eosinophils, and neutrophils [60, 62] [104]

Increased MPO and EPO levels [15] [101103]

Increased BAL fluid xanthine/XO activity [117119]

Elevated plasma and exhaled F2-isoprostane, ethane, and pentane levels [8688, 90, 92] [134136]

Elevated plasma and exhaled lipid peroxide (TBARS) levels [89, 94, 95] [95, 112, 137, 138]

Increased formation of 4-hydroxy-2-nonenal-protein adducts in lungs [139]

Elevated plasma protein carbonyls [140]

Increased exhaled CO [141] [141]

Increased exhaled NO [70, 71] [129132]

Increased 3-nitrotyrosine in plasma, BAL fluid, and exhaled breath [74, 79, 80] [142144]

Increased 3-chlorotyrosine and 3-bromotyrosine [15, 16] [16]

ONOO activity in plasma has been shown in cigarette hydrocarbons, such as ethane and pentane, are increased
smokers [143]. In vitro, exposure to gas-phase cigarette in exhaled air condensate in smokers and in patients with
smoke results in increased lipid peroxidation and protein COPD [134136]. Furthermore, the levels of these lipid
carbonyl formation in plasma [140]. It is likely that alpha, peroxidation products have been correlated with airway
beta-unsaturated aldehydes (acrolein, acetaldehyde, and cro- obstruction [136]. Urinary levels of isoprostane F2-III have
tonaldehyde) that are abundantly present in cigarette smoke been shown to be elevated in patients with COPD com-
may react with protein-SH and -NH2 groups leading to the pared with control subjects and are even more elevated dur-
formation of a protein-bound aldehyde functional group ing exacerbations of COPD [30]. These studies indicate that
and are capable of converting tyrosine to 3-nitrotyrosine there is increased lipid peroxidation in patients with COPD.
and dityrosine [140]. Nitric oxide and ONOO-mediated However, it is not known whether the increased level of lipid
formation of 3-nitrotyrosine in plasma and free catalytic peroxidation products found in these diseases is the result of
iron (Fe2) levels in ELF are elevated in chronic smokers primary lung-associated processes such as alveolar macro-
[144]. Nitration of tyrosine residues or proteins in plasma phage activation, neutrophil activity, or the ongoing lipid per-
leads to the production of 3-nitrotyrosine [145]. The levels oxidative chain reaction in the alveoli, parenchyma, or airways,
of nitrotyrosine and inducible NO synthase (iNOS) were which are induced by inhaled oxidants/cigarette smoke [141].
higher in airway inammatory cells obtained by induced Indirect and nonspecic measurements of lipid per-
sputum from patients with COPD, compared to those with oxidation products, such as TBARS, have also been shown
asthma [144]. The levels of nitrotyrosine were negatively to be elevated in breath condensate and in lungs of patients
correlated with the percentage predicted FEV1 in patients with stable COPD [112, 137, 138]. The levels of plasma
with COPD. These direct and indirect studies indicate that lipid peroxides have been shown to be elevated in COPD
increased RNS- and ROS-mediated protein nitration and and negatively correlated with the FEV1 [94]. Oxidative
lipid peroxidation respectively may play a role in the patho- stress, measured as lipid peroxidation products in plasma,
genesis of COPD. has also been shown to correlate inversely with the percent-
age predicted FEV1 in a population study [146], suggesting
that in patients with COPD lipid peroxidation may play a
Generation of ROS-mediated lipid role in the progression of the disease.
peroxidation products: Carbonyl stress 4-HNE is a highly reactive and specic diusible
end-product of lipid peroxidation. Increased 4-HNE-modi-
Formation of protein carbonyls (aldehyde protein adducts) ed protein levels (protein carbonyls) are present in airway
in response to cigarette smoke-derived lipid peroxides (alde- and alveolar epithelial cells and endothelial cells, and in
hydes) has been implicated in pathogenesis of COPD. neutrophils in smokers with airway obstruction compared to
The levels of lipid peroxides, such as 8-isoprostane, and subjects without airway obstruction [139]. This demonstrates

300
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
not only the presence of 4-HNE but that 4-HNE modi-
es proteins in lung cells to a greater extent in patients Depletion of antioxidant enzymes in
with COPD. The increased level of 4-HNE adducts in asthma and COPD
alveolar epithelium, airway endothelium, and neutrophils
was inversely correlated with FEV1, suggesting a role for 4- An important eect of oxidative stress and inamma-
HNE in the pathogenesis of COPD. The formation of 4- tion is the upregulation of protective antioxidant genes.
HNE may have detrimental eect on a variety of signaling Amongst antioxidants, glutathione (GSH), and its redox
molecules, such as Nrf2, HDAC2, and IB kinase (NF-B enzymes have an important protective role in the airspaces
activation). Hence, the carbonyl stress that predominantly and intracellularly in lung epithelial cells. Indeed, GSH
occurs in response to cigarette smoke may be a dierentiat- levels are increased in the ELF of both asthmatics and
ing factor in distinguishing the oxidative stress response in chronic cigarette smokers [150, 151]. GSH is a tripeptide
the airways of asthma and COPD. (l--glutamyl-l-cysteinyl-glycine) that contains a thiol
group. It functions as an antioxidant by acting as a sacri-
cial target for ROS and other products of lipid peroxida-
tion, such as reactive carbonyls. In so doing, GSH becomes
ALTERATION IN ENDOGENOUS ANTIOXIDANT oxidized to its dimeric form (glutathione disulde, GSSG)
DEFENSES WITHIN THE LUNG or forms adducts with reactive carbonyls and other reac-
tive xenobiotics (GS-X). Furthermore, enzymes such as
GPx and glutathione transferase can facilitate this proc-
To combat and neutralize the deleterious eects of ROS, ess. GSSG can itself be reduced back to GSH by glutath-
various endogenous antioxidant strategies have evolved ione reductase using NADPH generated from the pentose
which employ both enzymatic and nonenzymatic mecha- phosphate pathway. Oxidative stress causes upregulation
nisms. Within the lung lining uid, several nonenzymatic of glutamate cysteine ligase, GCL (formerly known as
antioxidant species exist which include, glutathione, ascorbic -glutamylcysteine synthetase) [3, 152], an important
acid (vitamin C), uric acid, -tocopherol (vitamin E), and enzyme involved in the synthesis of GSH, as an adap-
albumin. The relative abundance of these antioxidants can tive mechanism against subsequent oxidative stress. We
dier from that observed in blood plasma [147]. Nevertheless, have shown the increased expression of GCL mRNA in
ROS-induced lung injury can increase lung epithelial lungs of smokers which is even more pronounced in smok-
permeability [148] allowing leakage of plasma constituents, ers with COPD [153]. This implies that GSH synthesis is
which will also contain antioxidants, into the lung lining uid upregulated in lungs of smokers with and without COPD.
providing additional antioxidant protection. Similarly, enzy- Similarly, bronchial epithelial cells of rats exposed to ciga-
matic antioxidant defenses can also dier in both anatomical rette smoke have shown increased expression of the anti-
and subcellular localization based on expression. This includes oxidant genes such as manganese superoxide dismutase
enzymes such as SOD, catalase, thioredoxin, glutathione (MnSOD), metallothionein, and GPx [154]. This would
peroxidase (GPx), and glutathione-S-transferase. Of these, suggest the importance of an adaptive antioxidant gene
enzymes such as SOD can be found as dierent isoforms response against the injurious eects of cigarette smoke
expressed either intracellularly or extracellularly. Moreover, [154]. However, Harju and colleagues have found that
extracellular-SOD is highly expressed in the lungs mainly GCL immunoreactivity was decreased (possibly lead-
around blood vessels and airways [149]. ing to decreased GSH levels) in the airways of smok-
Table 25.2 highlights the level of dierent nonen- ers compared to nonsmokers, suggesting that cigarette
zymatic antioxidants found in the ELF compared to that smoke predisposes lung cells to ongoing oxidant stress
in plasma. While dierences are clearly evident, both [155]. In addition, Neurohr and colleagues showed that
enzymatic and nonenzymatic antioxidant strategies are decreased GSH levels in BAL uid cells of chronic smok-
employed within the lung. ers were associated with a decreased expression of GCL-
light subunit without a change in GCL-heavy subunit
expression [156].
Important protective antioxidant genes such as
TABLE 25.2 Antioxidant constituents of plasma and lung epithelial MnSOD, GCL, heme oxygenase-1 (HO-1), GPx, thiore-
lining fluid (ELF).
doxin reductase, and metallothionein are similarly induced
by various oxidative stresses including hyperoxia and inam-
Antioxidant Plasma (M) ELF (M)
matory mediators, such as TNF- and LPS in lung cells
Ascorbic acid 40 100 [3740]. Indeed, the transcription factor, Nrf2, is redox sen-
sitive (contains SH groups) and binds to the antioxidant
Glutathione 1.5 100 response element (ARE) within DNA regulating a variety
of antioxidant genes. The importance of Nrf2 can be gauged
Uric acid 300 90
from a report by Rangasamy et al., where they have shown
Albumin-SH 500 70 that disruption of the Nrf2 gene in mice lead to an early and
a more intense emphysema in response to cigarette smoke
-tocopherol 25 2.5 compared to mice with an intact Nrf2 gene [157]. In the
-carotene 0.4
same study they have shown that the expression of nearly
50 antioxidant and cytoprotective genes in the lungs may

301
Asthma and COPD: Basic Mechanisms and Clinical Management

be transcriptionally controlled by Nrf2 and all genes may can then be measured in various body uids as a marker of
work in concert to overcome the eects of cigarette smoke. oxidative stress [164]. The antioxidant properties of albu-
Moreover, studies examining polymorphisms in these pro- min, as well as mucins, come from the presence of exposed
tective antioxidant genes, such as glutathione transferase SH groups. Their protective eects are solely extracellu-
and HO-1, have suggested a link to onset of COPD and lar. However, albumin exists in a high concentration and
emphysema [158, 159]. Interestingly, Rangasamy and col- acts as a sacricial substrate able to react quickly with the
leagues have recently showed that disruption of Nrf2 also very damaging and potent oxidizing ONOO and HOCl
leads to susceptibility to severe allergen-induced asthma in [165].
mice. These ndings indicate that though Nrf2 is important The remaining two protective antioxidant molecules,
for the control of antioxidant genes, it is not specic to one ascorbic acid and -tocopherol, are derived from the diet.
inammatory disease [160]. Recent studies showed deple- Both antioxidants are decreased in chronic cigarette smok-
tion of various Nrf-2 dependent Phase II antioxidant genes ers [5]. Furthermore, the total antioxidant capacity of
in lungs of patients with COPD [161]. The compartmen- plasma is decreased in smokers, and in patients with asthma
talization and localization of various antioxidant enzymes and COPD [95]. It is interesting to note that the decrease
and their functions/alterations in asthma and COPD are in antioxidant capacity in smokers occurs transiently dur-
described in Table 25.3. ing smoking and resolves rapidly after smoking cessation.
Depletion of total antioxidant capacity in smokers is asso-
ciated with decreased levels of major plasma antioxidants
Depletion of antioxidant small (e.g. ascorbic acid, vitamin E, -carotene, and selenium).
molecules in asthma and COPD The depletion of antioxidants may thus be a reection of
ongoing oxidative stress due to underlying inammation
Apart from GSH, several other antioxidants, such as ascor- in these diseases. There is, however, a limitation of oxida-
bic acid (vitamin C), uric acid, -tocopherol (vitamin E), tive stress and antioxidant biomarkers at present due to the
and albumin, are present in the lung. Of these, the major lack of longitudinal studies, correlation with disease sever-
antioxidants in lung lining uid are GSH, ascorbic acid, ity or outcome, and the variations observed in the control
and uric acid [147]. Like GSH, uric acid can also be found subjects.
intracellularly, although at lower concentrations than that Ascorbic acid, otherwise known as vitamin C, is water
seen in plasma. It is a powerful scavenger of both ROS and soluble. It functions as an antioxidant by accepting free
RNS and can protect proteins against nitration [162] par- radical electrons, thus forming the ascorbyl radical which
ticularly from the nitrogen dioxide radical, nitrous oxide, is relatively unreactive. However, the ascorbyl radical will
found in cigarette smoke [163]. Oxidation of uric acid by undergo a disproportionation reaction to regenerate ascor-
ROS or RNS results in the formation of allantoin that bate and dehydroascorbate, the latter rapidly breaking down

TABLE 25.3 Antioxidant enzymes of the lungs, their localization, and functions in asthma and COPD.

Enzyme Lung localization Function Expression/Activity

Cu, ZnSOD Bronchial, alveolar epithelium, Scavenges O2 Decreased in COPD


macrophages, fibroblasts, pneumocytes

EC-SOD Bronchial epithelium, macrophages, Scavenges O2 Decreased in COPD


neutrophils, vascular walls, pneumocytes

MnSOD Bronchial epithelium, macrophages, Scavenges O2 Decreased in Asthma


neutrophils, vascular walls, pneumocytes Decreased in Smokers

Catalase macrophages, fibroblasts, pneumocytes Hydrogen peroxide to water Decreased in Asthma

Glutathione ELF cells, epithelium, macrophages, Organic hydroperoxides to organic Decreased in COPD
peroxidase other lung cells hydroxides Decreased in Asthma

Heme Alveolar, bronchial epithelium, Heme to CO and biliverdin Decreased in Emphysema


oxygenase-1 macrophages, inflammatory cells of Decreased in COPD
lungs

Thioredoxin bronchial epithelium, macrophages Transcriptional modulation, thiol Decreased in COPD


dithiol exchange, Prot-S-S-Prot to
Prot-SH

GCL (catalytic Alveolar, bronchial epithelium, Synthesis of glutathione (first rate Decreased in COPD
subunit) macrophages limiting step) Decreased in Smokers

302
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
to form oxalic acid and l-threonine [166]. Ascorbic acid has
been shown to possess a whole array of antioxidant proper-
THERAPEUTIC INTERVENTION WITH
ties in vitro, from scavenging ROS and RNS, preventing ANTIOXIDANTS IN ASTHMA AND COPD
lipid peroxidation to regenerating other antioxidants, such
as uric acid and -tocopherol. Like uric acid, ascorbic acid In view of the evidence implicating oxidative stress in the
is also a powerful scavenger of nitrous oxide which is a pathogenesis of chronic airway diseases, one rational approach
potent promoter of both protein nitration and lipid peroxi- would be to consider antioxidant intervention to neutralize the
dation [167, 168]. Indeed, increases in F2-isoprostane lipid increased oxidative stress, and the subsequent inammatory
peroxidation products are decreased in smokers who con- response. Several small molecular weight compounds that tar-
sume more ascorbate [169]. Further evidence of asorbates get oxidant signaling, or quench oxidants/aldehydes/carbonyls
antioxidant properties in vivo is provided by the evidence derived from cigarette smoke or boost antioxidant potential in
highlighting its depletion in clinical conditions associated the lung are currently being tested clinically [178]. Antioxidant
with a high oxidative stress burden [170]. Intriguingly, there agents such as thiol molecules (GSH/cysteine and mucolytic
is also a dark side to ascorbic acid in that in vitro it can drugs, such as N-acetyl-l-cysteine (NAC) and nacystelyn
reduce Fe3 to Fe2 thereby facilitating OH radical forma- (NAL)), dietary polyphenols (curcumin, resveratrol, green tea,
tion through Fenton chemistry. However, the relevance of and quercetin), erdosteine, fudosteine, and carbocysteine lysine
this in vivo is still a subject of investigation. salt, all have been reported to increase intracellular thiol anti-
oxidant levels and induction of GSH biosynthesis genes. The
various antioxidant strategies used have been divided into six
categories; dietary, thiols, spin traps, enzyme mimetics, and
IS THERE A CO-RELATIONSHIP BETWEEN polyphenols as shown in Table 25.4.
OXIDATIVE STRESS, ANTIOXIDANT DEPLETION
AND DECLINE IN LUNG FUNCTION IN ASTHMA Dietary
AND COPD? Reports of clinical benet in asthma and COPD for
increased vitamin C, E, and other dietary antioxidants have
been varied. Nevertheless, epidemiological studies have
Emerging evidence suggests that oxidative stress is directly shown that there are decreased vitamin C, E, -carotene, and
correlated with the decline in lung function in asthma and selenium levels in cigarette smokers [179181]. Similarly,
COPD. Epidemiological studies have shown a relation- other studies have reported a link with decreased lung func-
ship between circulating increase in neutrophil numbers tion [182] and the presence and severity of asthma to dietary
and decline in FEV1. Oxidative stress, measured as lipid antioxidant intake, such as vitamins C and E, -carotene
peroxidation products in plasma, has also been shown to and selenium [183, 184]. One small trial has shown that
correlate inversely with the FEV1 percentage in a population there was a clinical improvement in asthmatic symptoms
study [171]. after being given selenium supplementation compared to
There is an association between dietary intake of anti- placebo [185]. In addition, one very early study incorporat-
oxidant vitamins and polyphenols and lung function in the ing selenium supplementation in smokers demonstrated
general population as well as in obstructive airway disease, that this resulted in reduced superoxide release from leu-
supporting the hypothesis that oxidative stress may have kocytes [186]. Moreover, two studies by the same group
a role in decline in lung function, and chronic cough and have shown no clinical benet of supplemental intake of
breathlessness (but not chronic phlegm) in COPD [146, vitamin C or E when compared to current standard thera-
172174]. This is substantiated by a study highlighting the pies for mild-to-moderate asthma [187, 188]. This is in
benecial protective eect of fruit containing polyphenols contrast with earlier studies, showing decreased lipid per-
and vitamin E intake against COPD symptoms in 20-year oxidation and a corresponding improvement in lung func-
COPD mortality from three European countries consist- tion with increased vitamin C intake in smokers [177, 189].
ing of Finnish, Italian, and Dutch cohorts [175]. It has Supplementation with either vitamin E or -carotene alone
been shown that there is a correlation between increased resulted in no clinical benet in COPD [190]. Moreover,
dietary antioxidant intake and improved lung function -carotene supplementation may have a detrimental side
[146]. Moreover, increased dietary antioxidant status, par-
ticularly vitamin E (-tocopherol), correlated with lower
levels of lipid peroxidation [176]. -Tocopherol is a lipid TABLE 25.4 Antioxidant therapeutic interventions in asthma and COPD.
soluble antioxidant and as such is probably one of the most
important scavengers and inhibitors of lipid peroxidation. Antioxidant compounds
It largely achieves this by reacting faster with the lipid per- Thiol compounds-N-acetyl-L-cysteine (NAC), nacystelyn (NAL),
oxyl radicals than these radicals can react with other lipid glutathione esters, thioredoxin, Procysteine, erdosteine, fudosteine,
molecules. Similarly, another study reported that increased carbocysteine, N-isobutyrylcysteine, ebselen
dietary intake of ascorbic acid led to an improvement in Antioxidant vitamins (vitamin A, E, C), -carotene, CoQ10
lung function in both smokers and asthmatics [177]. These Polyphenols (curcumin, resveratrol, quercetin, and green tea-catechins)
studies support the concept that oxidative stress plays an Nitrone spin traps
important role in decline in lung function in smokers, and Enzyme mimetics: SOD and GPx mimetics, porphyrins
possibly in asthma patients with COPD.

303
Asthma and COPD: Basic Mechanisms and Clinical Management

eect in that it may accelerate the onset of lung cancer in cig- more promising than NAC in reducing the oxidant burden
arette smokers [189]. Hence, dietary antioxidants intake/sup- in the lung in chronic airways disease.
plementation may not be so eective as therapeutic option to The potential of other thiols as antioxidants are also
control the declining lung function in airway diseases. being explored. These include erdosteine, fudosteine, and
carbocysteine, which have mucoactive properties as well as
reduce bacterial adhesiveness. In the Equalife randomized
Thiols placebo controlled trial, erdosteine was dosed orally 300 mg
In the lung, thiols in the form of GSH constitute one of the b.i.d. for a period of 8 months [217]. Patients receiving
main antioxidant defenses in both intracellular and extra- erdosteine had signicantly fewer exacerbations and spent
cellular compartments. Attempts to raise GSH levels in few days in hospital than the placebo group. Moreover,
the lung through administration of GSH itself have been patients receiving erdosteine showed no reduction in lung
tried [191]. Unfortunately, aerosolization of GSH into the function over this period and showed a signicant improve-
lung was characterized by a poor half life [192] and led to ment in health-related quality of life. Similarly, fudosteine
induction of bronchial hyperreactivity [193]. Intracellular has shown to possess promising potential in treatment of
antioxidant protection after GSH oral administration was patients with COPD. A clinical trial on the combination of
also limited, due to its inecient cellular uptake [194]. steroids and erdosteine/fudosteine/carbosyteine in patients
Alternative methods of raising GSH levels have involved with COPD is needed. A recent study has shown that
supplementing the GSH precursor cysteine, through the long-term use of carbocysteine reduced the rate of exacer-
use of NAC. This approach has met with varying success bations in patients with COPD, but there was no dier-
in the past [195]. A Cochrane systematic review and other ence in exacerbation rate between the carbocysteine group
systematic reviews demonstrated that NAC treatment was and placebo group at an early treatment (3 months) [218].
associated with a signicant reduction of 0.79 exacerbations/ However, 1-year treatment of carbocysteine (1,500 mg/day)
patient/year compared with placebo, and a 29% decrease in was eective in COPD patients in terms of reductions in
COPD patients [196200]. Similarly, randomized, double- numbers of exacerbations and improvements in quality of
blind, placebo-controlled Phase II trials of a 6-12 month life. Hence, it was suggested that longer use of carbocisteine
oral dose (of 600 mg, twice a day) showed reduction in vari- is more eective for preventing exacerbations of COPD.
ous plasma and BAL uid oxidative biomarkers in smok-
ers [201, 202]. Similarly, two further studies showed a clear
benet of NAC in reducing oxidant burden [203, 204]. Spin traps
While there is a body of evidence that the administration of Spin traps are based around a nitrone or nitroxide contain-
NAC provides a potential benet for COPD patients, this ing molecule such as isoindole-based nitrones [219] and
evidence is not clinically directive. For example, in a recently azulenyl-based nitrones [220]. One such azulenyl nitrone,
conducted Phase III multicenter Bronchitis Randomized STANZ, may prove promising for in vivo use as it exhib-
on NAC Cost-Utility Study (BRONCUS) [205], NAC its very potent antioxidant activity. Surprisingly, no studies
(600 mg oral, daily) was ineective in halting the decline have been performed in asthma and COPD, looking at the
in lung function, but did lead to a reduction in hyperina- impact of spin traps on clinical endpoints, such as FEV1. A
tion in patients with severe COPD and decreased the exac- phenyl-based nitrone spin trap developed by AstraZeneca,
erbation rate in patients who were not treated with inhaled NXY-059, is due to enter Phase III clinical trials for use
glucocorticoids [205] (Table 25.5). The variability in all the in acute ischemic stroke. The utility of this compound in
current studies using NAC at 600 mg, oral, daily may simply SAINTII trial was unsuccessul though the drugh was safe.
reect the fact that the dose was not high enough. Indeed Nevertheless, it remains to be seen whether such com-
the pharmacokinetics support this explanation. Given orally, pounds could be developed for more long-term use in
600 mg of NAC is rapidly absorbed by the gut but has a asthma and COPD.
bioavailability of only 10% with a plasma half life of 6.3 h;
this led to a transient increase in lung GSH levels after daily
dosing for 2 weeks [206, 207]. Though oral intake of NAC Enzyme mimetics
(600 mg daily) led to decreased markers of oxidative stress in Enzyme mimetics are generally small compounds that possess
smokers and patients with COPD, perhaps higher doses of catalytic activity that mimics the activity of larger enzyme-
NAC (12001800 mg/day) are needed to observe any clini- based molecules. In the case of antioxidants, this includes the
cal benet on lung function. SOD, catalase, and GPx-like activities. A number of SOD
An alternative to NAC is the lysine salt of NAC, mimetics based around organomanganese complexes have
nacystelyn (NAL). It has been used as a mucolytic for cystic been developed which retain their antioxidant properties
brosis. However, it also possesses antioxidant properties in vivo. These include a series of manganese-based mac-
and can reduce both ROS levels and ROS-mediated inam- rocyclic ligands, such as M40401, M40403, and M40419
matory events in vitro [213] and in vivo [214]. NAL has [221223]. The second class of SOD mimetics are the
several advantages over NAC, rstly it can enhance GSH manganesemetalloporphyrin based compounds as exem-
levels twice as eectively as NAC and secondly it forms a plied by AEOL-10113 and AEOL-10150 [224, 225]. The
neutral pH when in solution, unlike NAC which is acidic third class of manganese-based SOD mimetic are the Salens.
[215]. This has meant that when delivered directly into the These are generally aromatic substituted ethylenediamine
lung by aerosol in healthy volunteers, it did not cause any metal complexes [226], such as EUK134 [227, 228]. The
irritation or other side eects [216]. Therefore NAL may be Salens also possess some catalytic activity and can therefore

304
TABLE 25.5 Clinical trials conducted for the efficacy of antioxidants in asthma and COPD. 25
Disease/
Trial name Antioxidant used Aim of study Condition Outcome References

BRONCUS N-acetyl-L-cysteine Effect of NAC COPD 30% reduction in COPD [205]


on FEV1 hospitalization obtained without
change on decline in FEV1.

Systematic Cochrane N-acetyl-L-cysteine Effect of NAC and COPD Significant reduction in days [197, 198]
review of 23 (2 months of oral NAC antibiotics on number of disability (0.65 day/patient/
randomized controlled therapy) of days of disability month) and 29% reduction in
trails exacerbations.
No difference in lung function

A Cochrane systematic N-acetyl-L-cysteine Used a validated score COPD 9 trials showed prevention of [199]
review of randomized, to evaluate the quality exacerbation and 5 of which
controlled trials of 11 of of each study addressed improvement of
39 retrieved trials symptoms compared with 34.6%
of patients receiving placebo

A meta-analysis of N-acetyl-L-cysteine To assess the possible COPD 23% decrease in number of acute [200]
published trials prophylactic benefit of exacerbations
prolonged treatment

N-acetyl-L-cysteine Effect of NAC on H2O2 COPD No change in TBARS reduce H2O2 [202204]
(600 mg once a day for and TBARS in EBC levels
12 months)
-carotene (20 mg/day) Effect on the symptoms COPD No benefit on symptoms [208]
and -tocopherol (chronic cough,
(50 mg/day) phlegm, or dyspnea)
-carotene (20 mg daily Effect on lipid peroxides Smokers Reduce lipid peroxidation (pentane [190, 209,
for 4 weeks) Levels in exhaled breath levels) in exhaled breath 210]

The MORGEN Diet rich in Effect on FEV1, chronic COPD Positively associated with decline [174]
study polyphenols/ cough, breathlessness, in FEV1 and inversely associated
bioflavonoids (catechin, and chronic phlegm with chronic cough and
flavonol, and flavone) breathlessness, but not chronic
(58 mg/day) phlegm

European countries Diet rich in fruits, Effect on 20-year COPD COPD A 24% lower COPD mortality risk [175]
(Finnish, Italian, and vegetables and fish mortality
Dutch cohorts) intake

Vitamin C (500 mg daily Effect on lipid peroxides Smokers No change in lung function [209]
for 4 weeks) Levels in breath and No change in lipid peroxidation
plasma (breath pentane and plasma MDA
levels)
Vitamin C (600 mg) Effect on lipid Smokers Reduced lipid peroxidation [211, 212]
Vitamin E (400 IU) peroxidation
-carotene (30 mg)
Vitamin E (400 IU b.i.d. Effect on breath ethane Smokers No effect on breath ethane levels [208]
For 3 weeks) levels
Vitamin C and E, Effect on lung function Asthmatics 50% reduction in asthma [185188]
-carotene, selenium and oxidative stress prevalence and clinical
biomarkers improvement whereas other
studies showed no clinical
improvement

Peace study Carbocysteine Effect on rate of COPD Long-term (one year) use of [218]
(Carbocisteine) exacerbations in COPD carbocysteine (1,500 mg/day)
produced reduction in numbers
of exacerbations in patients with
COPD

BONCUS: Bronchitis randomized on NAC cost utility study.

305
Asthma and COPD: Basic Mechanisms and Clinical Management

also scavenge hydrogen peroxide, a product of SOD activity. patients [237]. Moreover, Birrell and colleagues have showed
Moreover, they are also able to decompose peroxynitrite [229]. that in vivo, resveratrol can inhibit inammatory cytokine
Within the various classes of SOD mimetic, only expression in response to LPS challenge in rat lungs [238].
the metalloporphyrin-based compounds AEOL-10150 Furthermore, in both monocytic U937 and alveolar epithe-
and AEOL-10113 (iron-containing porphyrin complexes lial A549 cells, resveratrol inhibits NF-B and AP-1 activa-
that decomposes ONOO into innocuous nitrate) have tion [239, 240]. However, the clinical utility of resveratrol in
been studied in models of airway inammation [230]. In patients with asthma and COPD is not known.
one study, AEOL-10113 was shown to inhibit both airway Another well-studied polyphenol is curcumin. It is
inammation and bronchial hyperreactivity in an ovalbu- the active constituent of Curcuma longa, commonly known
min challenge model of airway inammation [224]. This as turmeric. Like resveratrol, it has also been reported
highlighted an important therapeutic benet in asthma. In to inhibit NF-B activation, along with IL-8 release,
another study, AEOL-10150 was demonstrated to inhibit Cyclooxygenase-2 expression, and neutrophil recruitment in
cigarette smoke-induced lung inammation [225] suggest- the lungs [241]. Interestingly, one study postulates that cur-
ing a potential therapeutic benet in COPD. cumin prevents cigarette smoke-induced NF-B activation
Another type of catalytic antioxidant is the GPx through the inhibition of IB kinase in human lung epi-
mimetic ebselen. This is a selenium-based organic com- thelial cells [242]. Recently, we have observed that curcumin
plex and has been shown to be a very powerful antioxidant can inhibit inammation and restore glucocorticoid ecacy
against the highly reactive and destructive ONOO [231]. in response to oxidative stress, through the upregulation/
It is able to prevent both NF-B/AP-1 activation and pro- restoration of HDAC2 activity in macrophages (U937 and
inammatory gene expression in human leukocytes exposed MonoMac6 cells) [243]. This would facilitate steroid-medi-
to ONOO. Other studies have shown that ebselen is also ated HDAC2 recruitment in attenuating NF-B-mediated
active in vivo in preventing LPS-induced airway inamma- chromatin acetylation and subsequent pro-inammatory
tion [23, 232]. However, no studies have been reported so gene expression. As glucocorticoids are the main thrust of
far on the protective eects of ebselen in cigarette smoke- anti-inammatory treatment, any therapeutics that can
induced lung inammation. In contrast, the GPx mimetic be used as an add-on to improve steroid responsiveness in
BXT-51072 (Oxis, USA) and the lipid peroxidation inhibi- COPD and severe asthma would be of signicant clini-
tor BO-653 (Chugai Pharma, Japan) are either currently in cal benet. Clearly clinical trials by using a combination
Phase I or pre-clinical trials in patients with COPD. A more approach of a steroid with polyphenols are warranted.
detailed review of the development and properties of these
catalytic antioxidants is described elsewhere [233].

Polyphenols SUMMARY
Several epidemiological studies have been undertaken, which
have established a benecial link between polyphenol intake There is now increasing evidence that ROS generation plays
and lower disease risk with many of the clinical benets a major pathophysiological role in asthma and COPD, and
being attributed to both the antioxidant and anti-inamma- it is important for the severity of these conditions. ROS
tory properties of polyphenols [234]. In one Finnish study generation through endogenous mechanisms or exogenous
with over 10,000 participants, a signicant inverse correla- cigarette smoke/environmental oxidants is critical to the
tion was observed between polyphenol intake and the inci- inammatory response through activation of redox-sensitive
dence of asthma [235]. Similar benecial associations were transcription factors and pro-inammatory signaling path-
also observed for COPD in a study encompassing over ways. At the same time, endogenous antioxidant mechanisms
13,000 adults. This study reported that increased polyphenol are present to attenuate this redox-mediated inammatory
intake correlated with improved symptoms, as assessed by response. It is when these two opposing mechanisms are
cough, phlegm production, and breathlessness and improved out of balance that a chronic and more severe inamma-
lung function as measured by FEV1 [174]. Two further tory state becomes apparent. The use of antioxidants or other
studies appeared to corroborate these ndings. The rst pharmacological agents to boost the endogenous antioxidant
study showed a benecial protective eect against COPD system could be used to redress this imbalance. In so doing,
symptoms for increased fruit intake, high in polyphenol and this would provide therapeutic benet in damping down and
vitamin E content [175]. In the second more recent study, curtailing the severity and chronicity of the inammatory
a standardized polyphenol extract administered orally was response in asthma and COPD patients.
shown to be eective in reducing oxidant stress and increas- A variety of oxidants, free radicals, and aldehydes are
ing PaO2, as well as improvements in FEV1 between the implicated in the pathogenesis of COPD, therefore it is pos-
enrollment and the end of the study [236]. sible that the therapeutic administration of multiple antioxi-
While the above studies would appear to demonstrate dants will be eective in the treatment of COPD. An eective
an epidemiological link between polyphenol intake and clin- wide spectrum antioxidant therapy that has good bioavailabil-
ical benet in asthma and COPD, other studies have tried to ity and potency is needed to control the localized oxidative
show a direct impact of specic polyphenolic compounds on and inammatory processes that occur in the pathogenesis
inammation in vitro and in vivo. For example, the avonoid of asthma and COPD. Furthermore, human clinical trials of
resveratrol, a constituent of red wine, inhibits inammatory newly developed small molecular antioxidants (alone or in
cytokine release from macrophages isolated from COPD combination with anti-inammatory agents), or higher doses

306
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
of NAC, are urgently needed to validate these compounds as 20. Church T, Pryor WA. Free radical chemistry of cigarette smoke and its
clinical therapies and to provide the proof of concept that oxi- toxicological implications. Environ Health Perspect 64: 11126, 1985.
dative stress is involved in the pathogenesis of COPD. 21. Pryor WA, Stone K. Oxidants in cigarette smoke: Radicals, hydro-
gen peroxides, peroxynitrate, and peroxynitrite. Ann NY Acad Sci 686:
Clearly, further studies are required to understand the
1228, 1993.
eect of ROS on basic cellular functions and the dierential 22. Nakayama T, Church DF, Pryor WA. Quantitative analysis of the
responses seen in dierent cell types and how this in turn hydrogen peroxide formed in aqueous cigarette tar extracts. Free Radic
impacts on the pathology of dierent inammatory disease Biol Med 7: 915, 1989.
states. At the same time, endeavors into identifying new and 23. Zang LY, Stone K, Pryor WA. Detection of free radicals in aqueous
more ecacious antioxidants, radical quenchers, inhibitors extracts of cigarette tar by electron spin resonance. Free Radic Biol Med
of lipid peroxidation, and agents that can reverse protein 19: 16167, 1995.
carbonyl protein damage as a therapeutic strategy should 24. Uchida K, Shiraishi M, Naito Y et al. Activation of stress signaling
continue. Indeed, elucidating the mechanism of action for pathways by the end product of lipid peroxidation. J Biol Chem 274:
some of the naturally occurring antioxidants, such as the 223442, 1999.
25. Parola M, Bellomo G, Robino G, Barrera G, Dianzani MU. 4-hydrox-
potent enzyme mimetics and polyphenols, may lead to
ynonenal as a biological signal: Molecular basis and pathophysiological
new therapeutic targets that can be antagonized/agonized implications. Antioxid Redox Signal 1: 25584, 1999.
through more conventional pharmacological approaches. 26. Marwick JA, Kirkham PA, Stevenson CS, Danahay H, Giddings J,
Butler K, Donaldson K, MacNee W, Rahman I. Cigarette smoke alters
chromatin remodeling and induces proinammatory genes in rat lungs.
Am J Respir Cell Mol Biol 31: 63342, 2004.
References 27. Kirkham PA, Spooner G, Rahman I, Rossi AG. Macrophage phago-
cytosis of apoptotic neutrophils is compromised by matrix proteins
1. Gutteridge JM, Halliwell B. Free radicals and antioxidants in the modied by cigarette smoke and lipid peroxidation products. Biochem
year 2000: A historic look to the future. Ann NY Acad Sci 899: Biophys Res Commun 318: 3237, 2004.
13647, 2000. 28. Kirkham PA, Spooner G, Ffoulkes-Jones C, Calvez R. Cigarette
2. Richer C, Cogvadze V, La ranchi R et al. Oxidants in mitochondria: smoke triggers macrophage adhesion and activation: Role of lipid
From physiology to diseases. Biochim Biophys Acta 127: 6774, 1995. peroxidation products and scavenger receptor. Free Radic Biol Med 35:
3. Rahman I, MacNee W. Role of transcription factors in inammatory 697710, 2003.
lung diseases. Thorax 53: 60112, 1998. 29. Reilly M, Delanty N, Lawson JA, FitzGerald GA. Modulation of
4. Guyton KZ, Liu Y, Gorospe M et al. Activation of mitogenactivated oxidant stress in vivo in chronic cigarette smokers. Circulation 94:
protein kinase by H2O2. J Biol Chem 271: 413842, 1996. 1925, 1996.
5. Rahman I, MacNee W. Role of oxidants/antioxidants in smoking- 30. Pratico D, Basili S, Vieri M et al. Chronic obstructive pulmonary dis-
induced airway diseases. Free Radic Biol Med 21: 66981, 1996. ease is associated with an increase in urinary levels of isoprostane
6. Rahman I, MacNee W. Lung glutathione and oxidative stress: F2-III, an index of oxidant stress. Am J Respir Crit Care Med 158:
Implications in cigarette smoke-induced airways disease. Am J Physiol 170914, 1998.
277: L1067L1088, 1999. 31. Morrow JD, Roberts LJ. The isoprostanes: Unique bioactive products
7. Rahman I, MacNee W. Oxidative stress and regulation of glutathione of lipid peroxidation. Prog Lipid Res 36: 121, 1997.
synthesis in lung inammation. Eur Respir J 16: 53454, 2000. 32. Kinsella BT, OMahony DJ, Fitzgerald GA. The human thromboxane
8. Dworski R. Oxidant stress in asthma. Thorax 55: S5153, 2000. A2 receptor alpha isoform (TP alpha) functionally couples to the G
9. Hatch GE. Asthma: Inhaled oxidants and dietary antioxidants. J Clin proteins Gq and G11 in vivo and is activated by the isoprostane 8-epi
Nutr 61: 625S630S, 1995. prostaglandin F2 alpha. J Pharmacol Exp Ther 281: 95764, 1997.
10. Saetta M. Airway inammation in chronic obstructive pulmonary dis- 33. Okazawa A, Kawikova I, Cui ZH, Skoogh BE, Lotvall J.
ease. Am J Respir Crit Care Med 160: S1720, 1999. 8-Epi-PGF2alpha induces airow obstruction and airway plasma exu-
11. Barnes PJ. Mechanisms in COPD: Dierences from asthma. Chest dation in vivo. Am J Respir Crit Care Med 155: 43641, 1997.
117: 10S14S, 2000. 34. Leitinger N, Tyner TR, Oslund L, Rizza C, Subbanagounder G, Lee H,
12. Jeery PK. Structural and inammatory changes in COPD: A com- Shih PT, Mackman N, Tigyi G, Territo MC, Berliner JA, Vora DK.
parison with asthma. Thorax 53: 12936, 1998. Structurally similar oxidized phospholipids dierentially regulate
13. Walsh GM. Advances in the immunobiology of eosinophils and their endothelial binding of monocytes and neutrophils. Proc Natl Acad Sci
role in disease. Crit Rev Clin Lab Sci 36: 45396, 1999. USA 96: 1201015, 1999.
14. Eiserich JP, Hristova M, Cross CE et al. Formation of nitric oxide 35. Lee H, Shi W, Tontonoz P, Wang S, Subbanagounder G, Hedrick CC,
derived inammatory oxidants by myeloperoxidase in neutrophils. Hama S, Borromeo C, Evans RM, Berliner JA, Nagy L. Role for
Nature 391: 39397, 1998. peroxisome proliferator-activated receptor alpha in oxidized phos-
15. MacPherson JC, Comhair SAAA, Eruzurum SC et al. Eosinophils pholipid-induced synthesis of monocyte chemotactic protein-1 and
are a major source of nitric oxide-derived oxidants in severe asthma: interleukin-8 by endothelial cells. Circ Res 87: 51621, 2000.
Characterization of pathways available to eosinophils for generating 36. Yeh M, Leitinger N, de MR, Onai N, Matsushima K, Vora DK,
reactive nitrogen species. J Immunol 166: 576372, 2001. Berliner JA, Reddy ST. Increased transcription of IL-8 in endothelial
16. Wu W, Samoszuk MK, Comhair SAAA et al. Eosinophils generate cells is dierentially regulated by TNF-alpha and oxidized phospholip-
brominating oxidants in allergen-induced asthma. J Clin Invest 105: ids. Arterioscler Thromb Vasc Biol 21: 158591, 2001.
145563, 2000. 37. Sen CK. Redox signaling and the emerging therapeutic potential of
17. Halliwell B, Gutteridge JMC. Role of free radicals and catalytic metal thiol antioxidants. Biochem Pharmacol 55: 174758, 1998.
ions in human disease: An overow. Methods Enzymol 186: 185, 1990. 38. Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling.
18. Rochelle LG, Fischer BM, Adler KB. Concurrent production of reac- Am J Physiol Lung Cell Mol Physiol 279: L100528, 2000.
tive oxygen and nitrogen species by airway epithelial cells in vitro. Free 39. Adler V, Yin Z, Tew KD, Ronai Z. Role of redox potential and reactive
Radic Biol Med 24: 86368, 1998. oxygen species in stress signaling. Oncogene 18: 610411, 1999.
19. Gutteridge JMC. Lipid peroxidation and antioxidants as biomarkers of 40. Rahman I, Adcock M. Oxidative stress and redox signaling of lung
tissue damage. Clin Chem 41: 181928, 1995. inammation in COPD. Eur Respir J 28: 21942, 2006.

307
Asthma and COPD: Basic Mechanisms and Clinical Management

41. Rahman I, MacNee W. Regulation of redox glutathione levels and gene 61. Teramoto S, Shu CY, Ouchi Y, Fukuchi Y. Increased spontaneous pro-
transcription in lung inammation: Therapeutic approaches. Free Radic duction and generation of superoxide anion by blood neutrophils in
Biol Med 28: 140520, 2000. patients with asthma. J Asthma 33: 14955, 1996.
42. Sedgewick JB, Geiger KM, Busse WW. Superoxide generation by 62. Shauer U, Leinhaas C, Jager R, Rieger CH. Enhanced superoxide gen-
hypodense eosinophils from patients with asthma. Am Rev Respir Dis eration by eosinophils from asthmatic children. Int Arch Allergy Appl
80: 195201, 1990. Immunol 96: 31721, 1991.
43. Kanazawa H, Kurihara N, Hirata K, Takeda T. The role of free 63. Krishna MT, Madden J, Teran LM et al. Eects of 0.2 ppm ozone on
radicals in airway obstruction in asthmatic patients. Chest 100: 1319 biomarkers of inammation in bronchoalveolar lavage uid and bron-
22, 1991. chial mucosa of healthy subjects. Eur Respir J 11: 1294300, 1998.
44. Calhoun WJ, Reed HE, Moest DR, Stevens CA. Enhanced superoxide 64. Agosti JM, Altman LC, Avars GH et al. The injurious eect of eosi-
production by alveolar macrophages and air-space cells, airway inam- nophil peroxidase, hydrogen peroxide, and halides on pneumocytes
mation, and alveolar macrophage density changes after segmental anti- in vitro. J Allergy Clin Immunol 79: 496504, 1987.
gen bronchoprovocation in allergic subjects. Am Rev Respir Dis 145: 65. McBride DE, Koenig JQ, Luchtel DL, Williams PV, Henderson WR.
31725, 1992. Inammatory eects of ozone in the upper airways of subjects with
45. Vachier I, Damon M, Le Doucen C et al. Increased oxygen species asthma. Am J Respir Crit Care Med 149: 119297, 1994.
generation in blood monocytes of asthmatic patients. Am Rev Respir 66. Emelyanov A, Fedoseev G, Abulimity A et al. Elevated concentra-
Dis 146(1): 16166, 1992. tions of exhaled hydrogen peroxide in asthmatic patients. Chest 120:
46. Cluzel M, Damon M, Chanez P et al. Enhanced alveolar cell luminol- 113639, 2001.
dependent chemiluminescence in asthma. J Allergy Clin Immunol 80: 67. Mitra SN, Slungaard A, Hazen SL. Role of eosinophil peroxidase in
195201, 1987. the origins of protein oxidation in asthma. Redox Rep 5: 21524, 2000.
47. Hulsmann AR, Raatgeep HR, den Hollander JC et al. Oxidative epi- 68. Motojima S, Fukuda T, Makino S. Eect of eosinophil peroxidase on
thelial damage produces hyperresponsiveness of human peripheral air- beta-adrenergic receptor density on guinea pig lung. Biochem Biophys
ways. Am J Respir Crit Care Med 149: 51925, 1994. Res Commun 189: 161319, 1992.
48. Sadeghi-Hashjin G, Folkerts G, Henricks PA et al. Peroxynitrite 69. Yoshikawa S, Kayes SG, Parker JC. Eosinophils increase lung micro-
induces airway hyperresponsiveness in guinea pigs in vitro and in vivo. vascular permeability via the peroxidase-hydrogen peroxide-halide
Am J Respir Crit Care Med 153: 1697701, 1996. system: Bronchoconstriction and vasoconstriction unaected by eosi-
49. Cortijo J, Marti-Cabrera M, de la Asuncion JG et al. Contraction of nophil peroxidase inhibition. Am Rev Respir Dis 147: 91420, 1993.
human airways by oxidative stress protection by N-acetylcysteine. Free 70. Alving K, Weitzberg E, Lundberg JM. Increased amount of nitric
Radic Biol Med 27: 392400, 1999. oxide in exhaled air of asthmatics. Eur Respir J 6: 136870, 1993.
50. Adam L, Bouvier M, Jones TLZ. Nitric oxide modulates 71. Silko PE, Sylvester JT, Zamel N, Permutt S. Airway nitric oxide dif-
2-adrenergic receptor palmitoylation and signaling. J Biol Chem 274: fusion in asthma: Role in pulmonary function and bronchial respon-
2633743, 1999. siveness. Am J Respir Crit Care Med 161: 121828, 2000.
51. Owen S, Pearson D, ODriscoll R. Evidence of free-radical activity in 72. Barnes PJ. NO or no NO in asthma? Thorax 51: 21820, 1996.
asthma. N Engl J Med 325: 58687, 1991. 73. Beckman DL, Mehta P, Hanks V, Rowan WH, Liu L. Eects of per-
52. Seltzer J, Bigby BG, Stulbarg M et al. O3-induced change in bronchial oxynitrite on pulmonary edema and the oxidative state. Exp Lung Res
reactivity to methacholine and airway inammation in humans. J Appl 26: 34959, 2000.
Physiol 60: 132126, 1986. 74. Saleh D, Ernst P, Lim S, Barnes PJ, Giaid A. Increased formation of
53. Hiltermann TJ, Peters EA, Alberts B et al. Ozone-induced airway the potent oxidant peroxynitrite in the airways of asthmatic patients
hyperresponsiveness in patients with asthma: Role of neutrophil- is associated with induction of nitric oxide synthase: Eect of inhaled
derived serine proteinases. Free Radic Biol Med 24: 95258, 1998. glucocorticoid. FASEB J 12: 92937, 1998.
54. Sanders SP, Zweier JL, Harrison SL et al. Spontaneous oxygen radical 75. Dohlman AW, Black HR, Royall JA. Expired breath hydrogen perox-
product at sites of antigen challenge in allergic subjects. Am J Respir ide is a marker of acute airway inammation in paediatric patients with
Crit Care Med 151: 172533, 1995. asthma. Am Rev Respir Dis 148: 95560, 1993.
55. Comhair SA, Ricci KS, Arroliga M, Lara AR, Dweik RA, Song W, 76. Zhang P, Wang YZ, Kagan E, Bonner JC. Peroxynitrite targets the
Hazen SL, Bleecker ER, Busse WW, Chung KF, Gaston B, Hastie A, epidermal growth factor receptor, Raf-1, and MEK independently to
Hew M, Jarjour N, Moore W, Peters S, Teague WG, Wenzel SE, activate MAPK. J Biol Chem 275: 2247986, 2000.
Erzurum SC. Correlation of systemic superoxide dismutase deciency 77. Groves JT. Peroxynitrite: Reactive, invasive and enigmatic. Curr Opin
to airow obstruction in asthma. Am J Respir Crit Care Med 172: Chem Biol 3: 22635, 1999.
30613, 2005a. 78. Hogg N, Kalyanaraman B. Nitric oxide and lipid peroxidation. Biochim
56. Comhair SA, Xu W, Ghosh S, Thunnissen FB, Almasan A, Calhoun WJ, Biophys Acta 1411: 37884, 1999.
Janocha AJ, Zheng L, Hazen SL, Erzurum SC. Superoxide dismutase 79. Hanazawa T, Kharitonov SA, Barnes PJ. Increased nitrotyrosine in
inactivation in pathophysiology of asthmatic airway remodeling and exhaled breath condensate of patients with asthma. Am J Respir Crit
reactivity. Am J Pathol 166: 66374, 2005b. Care Med 162(4 Pt.1): 127376, 2000.
57. Smith LJ, Shamsuddin M, Sporn PH, Denenberg M, Anderson J. 80. Kaminsky DA, Mitchell J, Carroll N et al. Nitrotyrosine formation in
Reduced superoxide dismutase in lung cells of patients with asthma. the airways and lung parenchyma of patients with asthma. J Allergy
Free Radic Biol Med 22: 13017, 1997. Clin Immunol 104: 74754, 1999.
58. Comhair SA, Bhathena PR, Dweik RA, Kavuru M, Erzurum SC. 81. Foster MW, McMahon TJ, Stamler JS. S-Nitrosylation in health and
Rapid loss of superoxide dismutase activity during antigen-induced disease. Trends Mol Med 9: 16068, 2003.
asthmatic response. Lancet 355: 624, 2000. 82. Liu L, Stamler JS. NO: An inhibitor of cell death. Cell Death Dier
59. Foreman RC, Mercer PF, Kroegel C, Warner JA. Role of the eosinophil 6: 93742, 1999.
in protein oxidation in asthma: Possible eects on proteinase/antiopro- 83. Gaston B, Sears S, Woods J, Hunt J, Ponaman M, McMahon T,
teinase balance. Int Arch Allergy Immunol 118: 18386, 1999. Stamler JS. Bronchodilator S-nitrosothiol deciency in asthmatic res-
60. Wardlaw AJ, Dunnette S, Gleich GJ, Collins JV, Kay AB. Eosinophils piratory failure. Lancet 351: 131719, 1998.
and mast cells in bronchoalveolar lavage in subjects with mild asthma: 84. Dweik RA, Comhair SA, Gaston B, Thunnissen FB, Farver C,
Relationship to bronchial hyperreactivity. Am Rev Respir Dis 137: Thomassen MJ, Kavuru M, Hammel J, Abu-Soud HM, Erzurum SC.
6269, 1988. NO chemical events in the human airway during the immediate and

308
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
late antigen-induced asthmatic response. Proc Natl Acad Sci USA 98: 107. Schaberg T, Klein U, Rau M, Eller J, Lode H. Subpopulation of alve-
262227, 2001. olar macrophages in smoker and nonsmokers: Relation to the expres-
85. Janssen-Heininger Y, Ckless K, Reynaert N, van der Vliet A. SOD sion of CD 11/CD 18 molecules and superoxide anion production.
inactivation in asthma: Bad news or no news? Am J Pathol 166: 649 Am J Respir Crit Care Med 151: 155158, 1995.
52, 2005. 108. Nowak D, Kasielski M, Pietras T, Bialasiewicz P, Antczak A. Cigarette
86. Montuschi P, Corradi M, Ciabattoni G et al. Increased 8-isoprostane, smoking does not increase hydrogen peroxide levels in expired breath
a marker of oxidative stress, in exhaled condensate of asthma patients. condensate of patients with stable COPD. Monaldi Arch Chest Dis 53:
Am J Respir Crit Care Med 160: 21620, 1999. 26873, 1998.
87. Paredi P, Kharitonov SA, Barnes PJ. Elevation of exhaled ethane 109. Nowak D, Antczak A, Krol M et al. Increased content of hydrogen
concentration in asthma. Am J Respir Crit Care Med 162: 145054, 2000. peroxide in expired breath of cigarette smokers. Eur Respir J 9: 652
88. Olepade CO, Zakkar M, Swedler WI. Exhaled pentane levels in acute 57, 1996.
asthma. Chest 111: 86265, 1997. 110. Dekhuijzen PNR, Aben KKH, Dekke I et al. Increased exhalation
89. Antczak A, Nowak D, Shariati B et al. Increased hydrogen peroxide of hydrogen peroxide in patients with stable and unstable chronic
and thiobarbituric acid reactive products in expired breath condensate obstructive pulmonary disease. Am J Respir Crit Care Med 154:
of asthma patients. Eur Respir J 10: 123541, 1997. 81316, 1996.
90. Dworski R, Murry JJ, Roberts LJ et al. Allergen-induced synthesis 111. Mateos F, Brock JF, Perez-Arellano JL. Iron metabolism in the lower
of F2-isoprostanes in atopic asthmatics: Evidence for oxidant stress. respiratory tract. Thorax 53: 594600, 1998.
Am J Respir Crit Care Med 160: 194751, 1999. 112. Lapenna D, Gioia SD, Mezzetti A et al. Cigarette smoke, ferritin, and
91. Dworski R, Roberts LJ, Murry JJ et al. Assessment of oxidant stress lipid peroxidation. Am J Respir Crit Care Med 151: 43135, 1995.
in allergic asthma by measurement of the major urinary metabolite of 113. Thompson AB, Bohling T, Heires A, Linder J, Rennard SI. Lower
F2-isoprostane, 15-F2t-IsoP (8-iso-PGF2alpha). Clin Exp Allergy 31: respiratory tract iron burden is increased in association with cigarette
38790, 2001. smoking. J Lab Clin Med 117: 49499, 1991.
92. Wood LG, Fitzgerald DA, Gibson PG, Cooper DM, Garg ML. 114. Wesselius LJ, Nelson ME, Skikne BS. Increased release of ferritin and
Lipid peroxidation as determined by plasma isoprostanes is related to iron by iron loaded alveolar macrophages in cigarette smokers. Am J
disease severity in mild asthma. Lipids 35: 96774, 2000. Respir Crit Care Med 150: 69095, 1994.
93. Calabrese C, Triggiani M, Marone G, Mazzarella G. Arachidonic acid 115. Lacoste JY, Bousquet J, Chanez P et al. Eosinophilic and neutrophilic
metabolism in inammatory cells of patients with bronchial asthma. inammation in asthma, chronic bronchitis, and chronic obstructive
Allergy 55: 2730, 2000. pulmonary disease. J Allergy Clin Immunol 149: 80310, 1993.
94. Tsukagoshi H, Shimizu Y, Iwamae S et al. Evidence of oxidative stress 116. Lebowitz MD, Postma DS. Adverse eects of eosinophilic and smok-
in asthma and COPD: Potential inhibitory eect of theophylline. ing on the natural history of newly diagnosed chronic bronchitis.
Respir Med 94: 58488, 2000. Chest 108: 5561, 1995.
95. Rahman I, Morrison D, Donaldson K, MacNee W. Systemic oxida- 117. Pinamonti S, Muzzuli M, Chicca C et al. Xanthine oxidase activity
tive stress in asthma, COPD, and smokers. Am J Respir Crit Care Med in bronchoalveolar lavage uid from patients with chronic obstructive
154: 105560, 1996. lung disease. Free Radic Biol Med 21: 14755, 1996.
96. British Thoracic Society guidelines for the management of chronic 118. Heunks LM, Vina J, van Herwaarden CL et al. Xanthine oxidase is
obstructive pulmonary disease. Thorax 52:S128, 1997. involved in exercise-induced oxidative stress in chronic obstructive
97. American Thoracic Society Standards for the diagnosis and care of pulmonary disease. Am J Physiol 277: R1697704, 1999.
patients with chronic obstructive pulmonary disease. Am J Respir Crit 119. Pinamonti S, Leis M, Barbieri A et al. Detection of xanthine oxidase
Care Med 152:S77120, 1995. activity products by EPR and HPLC in bronchoalveolar lavage uid
98. Snider G. Chronic obstructive pulmonary disease: Risk factors, patho- from patients with chronic obstructive pulmonary disease. Free Radic
physiology and pathogenesis. Ann Rev Med 40: 41129, 1989. Biol Med 25: 77179, 1998.
99. Repine JE, Bast A, Lankhorst Ithe Oxidative Stress Study Group. 120. Chan-Yeung M, Dybuncio A. Leucocyte count, smoking and lung
Oxidative stress in chronic obstructive pulmonary disease. Am J Respir function. Am J Med 76: 3137, 1984.
Crit Care Med 156: 34157, 1997. 121. Van Antwerpen VL, Theron AJ, Richards GA et al. Vitamin E, pul-
100. Bosken CH, Hards J, Gatter K, Hogg JC. Characterization of the monary functions, and phagocyte-mediated oxidative stress in smok-
inammatory reaction in the peripheral airways of cigarette smokers ers and nonsmokers. Free Radic Biol Med 18: 93543, 1995.
using immunocytochemistry. Am Rev Respir Dis 145: 91117, 1992. 122. Chan-Yeung M, Abboud R, Dybuncio A, Vedal S. Peripheral leu-
101. Aaron SD, Angel JB, Lunau M et al. Granulocyte inammatory cocyte count and longitudinal decline in lung function. Thorax 43:
markers and airway infection during acute exacerbation of chronic 42668, 1988.
obstructive pulmonary disease. Am J Respir Crit Care Med 163: 123. Postma DS, Renkema TEJ, Noordhoek JA et al. Association between
34955, 2001. nonspecic bronchial hyperreactivity and superoxide anion produc-
102. Fiorini G, Crespi S, Rinaldi M et al. Serum ECP and MPO are tion by polymorphonuclear leukocytes in chronic airow obstruction.
increased during exacerbations of chronic bronchitis with airway Am Rev Respir Dis 137: 5761, 1988.
obstruction. Biomed Pharmacother 54: 27478, 2000. 124. Richards GA, Theron AJ, van der Merwe CA, Anderson R.
103. Gompertz S, Bayley DL, Hill SL, Stockley RA. Relationship between Spirometric abnormalities in young smokers correlate with increased
airway inammation and the frequency of exacerbations in patients chemiluminescence responses of activated blood phagocytes. Am Rev
with smoking related COPD. Thorax 56: 3641, 2001. Respir Dis 139: 18187, 1989.
104. Morrison D, Rahman I, Lannan S, MacNee W. Epithelial permeabil- 125. Rahman I, Skwarska E, MacNee W. Attenuation of oxidant/antioxidant
ity, inammation and oxidant stress in the airspaces of smokers. Am J imbalance during treatment of exacerbations of chronic obstructive
Respir Crit Care Med 159: 47379, 1999. pulmonary disease. Thorax 52: 56568, 1997.
105. Nakashima H, Ando M, Sugimoto M et al. Receptor-mediated O2 126. Muns G, Rubinstein I, Bergmann KC. Phagocytosis and oxidative
release by alveolar macrophages and peripheral blood monocytes from bursts of blood phagocytes in chronic obstructive airway disease.
smokers and nonsmokers. Am Rev Respir Dis 136: 31015, 1987. Scand J Infect Dis 27: 36973, 1995.
106. Drath DB, Larnovsky ML, Huber GL. The eects of experimental 127. Noguera A, Busquets X, Sauleda J et al. Expression of adhesion mole-
exposure to tobacco smoke on the oxidative metabolism of alveolar cules and G-proteins in circulating neutrophils in COPD. Am J Respir
macrophages. J Reticul Soc 25: 597604, 1970. Crit Care Med 158: 166468, 1998.

309
Asthma and COPD: Basic Mechanisms and Clinical Management

128. Brown DM, Drost E, Donaldson K, MacNee W. Deformability and cigarette smoke in vivo and in vitro. Am J Respir Crit Care Med 149:
CD 11/CD 18 expression of sequestered neutrophils in normal and 151825, 1994.
inamed lungs. Am J Respir Cell Mol Biol 13: 53139, 1995. 149. Su WY, Folz R, Chen JS, Crapo JD, Chang LY. Extracellular super-
129. Maziak W, Loukides S, Culpitt S et al. Exhaled nitric oxide in chronic oxide dismutase mRNA expressions in the human lung by in situ
obstructive pulmonary disease. Am J Respir Crit Care Med 147: hybridization. Am J Respir Cell Mol Biol 16: 16270, 1997.
9981002, 1998. 150. Cantin AM, North SL, Hubbard RC, Crystal RG. Normal alveo-
130. Corradi M, Majori M, Cacciani GC et al. Increased exhaled nitric lar epithelial lining uid contains high levels of glutathione. J Appl
oxide in patients with stable chronic obstructive pulmonary disease. Physiol 63: 15257, 1987.
Thorax 54: 57275, 1999. 151. Smith LJ, Houston M, Anderson J. Increased levels of glutathione
131. Delen FM, Sippel JM, Osborne ML et al. Increased exhaled nitric in bronchoalveolar lavage uid from patients with asthma. Am Rev
oxide in chronic bronchitis: Comparison with asthma and COPD. Respir Dis 147: 146164, 1993.
Chest 117: 695701, 2000. 152. Rahman I, Smith CA, Lawson MF, Harrison DJ, MacNee W.
132. Rutgers SR, van der Mark TW, Coers W et al. Markers of nitric oxide Induction of gamma-glutamylcysteine synthetase by cigarette smoke
metabolism in sputum and exhaled air are not increased in chronic is associated with AP-1 in human alveolar epithelial cells. FEBS Lett
obstructive pulmonary disease. Thorax 54: 576680, 1999. 396: 2125, 1996.
133. Robbins RA, Millatmal T, Lassi K, Rennard S, Daughton D. Smoking 153. Rahman I, van Schadewijk AM, Hiemstra PS et al. Localisation of
cessation is associated with an increase in exhaled nitric oxide. Chest -glutamylcysteine synthetase messenger RNA expression in lungs
112: 31318, 1997. of smokers and patients with chronic obstructive pulmonary disease.
134. Paredi P, Kharitonov SA, Leak D, Ward S, Cramer D, Barnes PJ. Free Radic Biol Med 28: 92025, 2000.
Exhaled ethane, a marker of lipid peroxidation, is elevated in chronic 154. Gilks CB, Price K, Wright JL, Churg A. Antioxidant gene expres-
obstructive pulmonary disease. Am J Respir Crit Care Med 162: sion in rat lung after exposure to cigarette smoke. Am J Pathol 152:
36973, 2000. 26978, 1998.
135. Euler DE, Dave SJ, Guo H. Eect of cigarette smoking on pentane 155. Harju T, Kaarteenaho-Wiik R, Soini Y, Sormunen R, Kinnula VL.
excretion in alveolar breath. Clin Chem 42: 3038, 1996. Diminished immunoreactivity of gamma-glutamylcysteine synthetase
136. Montuschi P, Collins JV, Ciabattoni G et al. Exhaled 8-isoprostane in the airways of smokers lung. Am J Respir Crit Care Med 166:
as an in-vivo biomarker of lung oxidative stress in patients with 75459, 2002.
COPD and healthy smokers. Am J Respir Crit Care Med 162: 156. Neurohr C, Lenz AG, Ding I, Leuchte H, Kolbe T, Behr J.
117577, 2000. Glutamate-cysteine ligase modulatory subunit in BAL alveolar mac-
137. Nowak D, Kasielski M, Antczak A, Pietras T, Bialasiewicz P. rophages of healthy smokers. Eur Respir J 22: 8287, 2003.
Increased content of thiobarbiturate reactive acid substances in hydro- 157. Rangasamy T, Cho CY, Thimmulappa RK, Zhen L, Srisuma SS,
gen peroxide in the expired breath condensate of patients with stable Kensler TW, Yamamoto M, Petrache I, Tuder RM, Biswal S. Genetic
chronic obstructive pulmonary disease: No signicant eect of ciga- ablation of Nrf2 enhances susceptibility to cigarette smoke-induced
rette smoking. Respir Med 93: 38996, 1999. emphysema in mice. J Clin Invest 114: 124859, 2004.
138. Fahn H, Wang L, Kao S et al. Smoking-associated mitochondrial 158. Harrison DJ, Cantlay AM, Rae F, Lamb D, Smith CA. Frequency of
DNA mutation and lipid peroxidation in human lung tissue. Am J glutathione s-transferase M1 deletion in smokers with emphysema
Respir Cell Mol Biol 19: 9019, 1998. and lung cancer. Hum Exp Toxicol 16: 35660, 1997.
139. Rahman I, Crowther A, de Boer WI et al. 4-hydroxy-2-nonenal, a 159. Exner M, Minar E, Wagner O, Schillinger M. The role of heme oxy-
specic lipid peroxidation product is elevated in lungs of patients with genase-1promoter polymorphisms in human disease. Free Radic Biol
chronic obstructive pulmonary disease. Am J Respir Crit Care Med Med 37: 1097104, 2004.
163: A31, 2001. 160. Rangasamy T, Guo J, Mitzner WA, Roman J, Singh A, Fryer AD,
140. Eiserich JP, van der Vliet A, Handelman GJ, Halliwell B, Cross CE. Yamamoto M, Kensler TW, Tuder RM, Georas SN, Biswal S.
Dietary antioxidants and cigarette smoke-induced biomolecular dam- Disruption of Nrf2 enhances susceptibility to severe airway inam-
age: A complex interaction. Am J Clin Nutr 62: 1490S500S, 1995. mation and asthma in mice. J Exp Med 202: 4759, 2005.
141. Rahman I, Kelly F. Biomarkers in breath condensate: A promising 161. Tomaki M, Sugiura H, Koarai A, Komaki Y, Akita T, Matsumoto T,
non-invasive technique in free radical research. Free Radic Res 37: Nakanishi A, Ogawa H, Hattori T, Ichinose M. Decreased expression
125366, 2003. of antioxidant enzymes and increased expression of chemokines in
142. Pignatelli B, Li CG, Boetta P et al. Nitrated and oxidized plasma COPD lung. Pulm Pharmacol Ther, 20: 596-605, 2007.
proteins in smokers and lung cancer patients. Cancer Res 61: 162. Simic, Michael G, Jovanovic , Slobodan V. Antioxidation mechanisms
77884, 2001. of uric acid. J Am Chem Soc 111: 577882, 1989.
143. Petruzzelli S, Puntoni R, Mimotti P et al. Plasma 3-nitrotyrosine in 163. Jano A, Pryor WA, Bengali ZH. NHLBI Workshop Summary.
cigarette smokers. Am J Respir Crit Care Med 156: 19027, 1997. Eects of tobacco smoke components on cellular and biochemical
144. Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato K. Increase in processes in the lung. Am Rev Respir Dis 136: 105864, 1987.
reactive nitrogen species production in chronic obstructive pulmonary 164. Grootveld M, Halliwell B. Measurement of allantoin and uric acid
disease airways. Am J Respir Crit Care Med 162: 7016, 2000. in human body uids. A potential index of free-radical reactions
145. Van der Vliet A, Smith D, ONeill CA et al. Interactions of peroxyni- in vivo? Biochem J 243: 8038, 1987.
trite and human plasma and its constituents: Oxidative damage and 165. Halliwell B, Gutteridge JM. The antioxidants of human extracellular
antioxidant depletion. Biochem J 303: 295301, 1994. uids. Arch Biochem Biophys 280: 18, 1990b.
146. Britton JR, Pavord ID, Richards KA, Knox AJ, Wisniewski AF, 166. Halliwell B, Gutteridge JM. Free Radicals in Biology and Medicine. ,
Lewis SA, Tatterseld AE, Weiss ST. Dietary antioxidant vitamin 3rd edn. New York: Oxford University Press Inc, 1999.
intake and lung function in the general population. Am J Respir Crit 167. Spencer JP, Jenner A, Chimel K, Aruoma OI, Cross CE, Wu R,
Care Med 151: 138387, 1995. , 174. Halliwell B. DNA damage in human respiratory tract epithelial cells:
147. Cross CE, van dV, ONeill CA, Louie S, Halliwell B. Oxidants, anti- Damage by gas phase cigarette smoke apparently involves attack by
oxidants, and respiratory tract lining uids. Environ Health Perspect reactive nitrogen species in addition to oxygen radicals. FEBS Lett
102(Suppl 10): 18591, 1994. 375: 17982, 1995.
148. Li XY, Donaldson K, Rahman I, MacNee W. An investigation of the 168. Halliwell B. Vitamin C: Antioxidant or pro-oxidant in vivo?.
role of glutathione in increased epithelial permeability induced by Free Radic Res 25: 43954, 1996.

310
Reactive Oxygen Species and Antioxidant Therapeutic Approaches 25
169. Morrow JD, Frei B, Longmire AW, Gaziano JM, Lynch SM, Shyr Y, carotene supplementation on COPD symptoms. Am J Respir Crit
Strauss WE, Oates JA, Roberts LJ. Increase in circulating products of Care Med 156: 144752, 1997.
lipid peroxidation (F2-isoprostanes) in smokers, smoking as a cause of 191. MacNee W, Rahman I. Oxidants and antioxidants as therapeutic tar-
oxidative damage. N Engl J Med 332: 1198203, 1995. gets in chronic obstructive pulmonary disease. Am J Respir Crit Care
170. Schorah CJ, Downing C, Piripitsi A, Gallivan L, Al-Hazaa AH, Med 160: S58S65, 1999.
Sanderson MJ, Bodenham A. Total vitamin C, ascorbic acid, and 192. Borok Z, Buhl R, Grimes GJ, Bokser AD, Hubbard RC, Holroyd KJ,
dehydroascorbic acid concentrations in plasma of critically ill patients. Roum JH, Czerski DB, Cantin AM, Crystal RG. Eect of glutath-
Am J Clin Nutr 6: 76065, 1996. ione aerosol on oxidant-antioxidant imbalance in idiopathic pulmo-
171. Schunemann HJ, Muti P, Freudenheim JL et al. Oxidative stress and nary brosis. Lancet 338: 21516, 1991.
lung function. Am J Epidemiol 146: 93948, 1997. 193. Gillissen A, Roum JH, Hoyt RF, Crystal RG. Aerosolization of
172. Grievink L, Smit HA, Ocke MC et al. Dietary intake of antioxidant superoxide dismutase. Augmentation of respiratory epithelial lin-
(pro)-vitamins, respiratory symptoms and pulmonary function: The ing uid antioxidant screen by aerosolization of recombinant human
MORGEN Study. Thorax 53: 16671, 1998. Cu/Zn superoxide dismutase. Chest 104: 81115, 1993.
173. Sargeant LA, Jaeckel A, Wareham NJ. Interaction of vitamin C with 194. MacNee W, Bridgeman MM, Marsden M, Drost E, Lannan S, Selby C,
the relation between smoking and obstructive airways disease in Donaldson K. The eects of N-acetylcysteine and glutathione on
EPIC Norfolk. European Prospective Investigation into Cancer and smoke-induced changes in lung phagocytes and epithelial cells. Am J
Nutrition. Eur Respir J 16: 397403, 2000. Med 1(91): 60S66S, 1991.
174. Tabak C, Ilja C, Arts CW et al. Chronic obstructive pulmonary dis- 195. Dueholm M, Nielson C, Thorshauge H et al. N-acetylcysteine by
ease and intake of catechins, avonols, and avones: The MORGEN metered dose inhaler in the treatment of chronic bronchitis: A multi-
Study. Am J Respir Crit Care Med 164: 6164, 2001. centre study. Respir Med 86: 8992, 1992.
175. Walda IC, Tabak C, Smit HA. Diet and 20-year chronic obstruc- 196. Dekhuijzen PN. Antioxidant properties of N-acetylcysteine: Their
tive pulmonary disease mortality in middle-aged men from three relevance in relation to chronic obstructive pulmonary disease. Eur
European countries. Eur J Clin Nutr 56: 63843, 2002. Respir J 23: 62936, 2004.
176. Mezzetti A, Lapenna D, Pierdomenico SD, Calaore AM, Costantini 197. Poole PJ, Black PN. Oral mucolytic drugs for exacerbations of
F, Riario-Sforza G, Imbastaro T, Neri M, Cuccurullo F. Vitamins E, C chronic obstructive pulmonary disease: Systematic review. BMJ 322:
and lipid peroxidation in plasma and arterial tissue of smokers and 127174, 2001.
non-smokers. Atherosclerosis 112: 9199, 1995. 198. Poole PJ, Black PN. Preventing exacerbations of chronic bronchitis
177. Schwartz J, Weiss ST. Relationship between dietary vitamin C intake and COPD: Therapeutic potential of mucolytic agents. Am J Respir
and pulmonary function in the First National Health and Nutrition Med 2: 36770, 2003.
Examination Survey (NHANES I). Am J Clin Nutr 59: 11014, 1994. 199. Stey C, Steurer J, Bachmann S, Medici TC, Tramer MR. The eect of
178. Rahman I. Antioxidant therapies in COPD. Int J COPD 1: oral N-acetylcysteine in chronic bronchitis: A quantitative systematic
1530, 2006. review. Eur Respir J 16: 25362, 2006.
179. Anderson R. Assessment of the roles of vitamin C, vitamin E, and 200. Grandjean EM, Berthet P, Rumann R, Leuenberger P. Ecacy of
beta-carotene in the modulation of oxidant stress mediated by cigarette oral long-term N-acetylcysteine in chronic bronchopulmonary dis-
smoke-activated phagocytes. Am J Clin Nutr 53: 358S61S, 1991. ease: A meta-analysis of published double-blind, placebo-controlled
180. Romieu I, Trenga C. Diet and obstructive lung diseases. Epidemiol clinical trials. Clin Ther 22: 20921, 2000.
Rev 2: 26887, 2001. 201. Van Schooten FJ, Nia AB, De Flora S et al. Eects of oral adminis-
181. Santos MC, Oliveira AL, Viegas-Crespo AM, Vicente L, Barreiros A, tration of N-acetyl-l-cysteine: A multi-biomarker study in smokers.
Monteiro P, Pinheiro T, Bugalho De AA. Systemic markers of the Cancer Epidemiol Biomarkers Prev 11: 16775, 2002.
redox balance in chronic obstructive pulmonary disease. Biomarkers 202. Kasielski M, Nowak D. Long-term administration of N-acetylcysteine
9: 46169, 2004. decreases hydrogen peroxide exhalation in subjects with chronic
182. Gilliland FD, Berhane KT, Li YF, Gauderman WJ, McConnell R, obstructive pulmonary disease. Respir Med 95: 44856, 2001, 200.
Peters J. Childrens lung function and antioxidant vitamin, fruit, juice, 203. De Benedetto F, Aceto A, Dragani B, Spacone A, Formisano S, Pela
and vegetable intake. Am J Epidemiol 158: 57684, 2003. R, Donner CF, Sanguinetti CM. Long-term oral N-acetylcysteine
183. Harik-Khan RI, Muller DC, Wise RA. Serum vitamin levels and the reduces exhaled hydrogen peroxide in stable COPD. Pulm Pharmacol
risk of asthma in children. Am J Epidemiol 159: 35157, 2004. Ther 18: 4147, 2005.
184. Rubin RN, Navon L, Cassano PA. Relationship of serum antioxi- 204. Sadowska AM, van Overveld FJ, Gorecka D, Zdral A, Filewska
dants to asthma prevalence in youth. Am J Respir Crit Care Med 169: M, Demkow UA, Luyten C, Saenen E, Zielinski J, De Backer WA.
39398, 2004. The interrelationship between markers of inammation and oxida-
185. Allam MF, Lucane RA. Selenium supplementation for asthma. tive stress in chronic obstructive pulmonary disease: Modulation by
Cochrane Database Syst Rev, 2004, 2: CD003538, 2004 inhaled steroids and antioxidant. Respir Med 99: 24149, 2005.
186. Clausen J. The inuence of selenium and vitamin E on the enhanced 205. Decramer M, Rutten-van Molken M, Dekhuijzen PN, Troosters T,
respiratory burst reaction in smokers. Biol Trace Elem Res 31: van Herwaarden C, Pellegrino R, van Schayck CP, Olivieri D,
28191, 1991. Del Donno M, De Backer W, Lankhorst I, Ardia A. Eects of
187. Fogarty A, Lewis SA, Scrivener SL, Antoniak M, Pacey S, Pringle M, N-acetylcysteine on outcomes in chronic obstructive pulmonary disease
Britton J. Oral magnesium and vitamin C supplements in asthma: A (Bronchitis Randomized on NAC Cost-Utility Study, BRONCUS): A
parallel group randomized placebo-controlled trial. Clin Exp Allergy randomised placebo-controlled trial. Lancet 365: 155260, 2005.
33: 135559, 2003. 206. Bridgeman MM, Marsden M, MacNee W, Flenley DC, Ryle AP.
188. Pearson PJ, Lewis SA, Britton J, Fogarty A. Vitamin E supplements Fluid after treatment with N-acetylcysteine. Thorax 46: 3942, 1991.
in asthma: A parallel group randomised placebo controlled trial. 207. Bridgeman MM, Marsden M, Selby C, Morrison D, MacNee W.
Thorax 59: 65256, 2004. Eect of N-acetyl cysteine on the concentrations of thiols in plasma,
189. Bucca C, Rolla G, Arossa W, Caria E, Elia C, Nebiolo F, Baldi S. bronchoalveolar lavage uid, and lung tissue. Thorax 49: 67075, 1994.
Eect of ascorbic acid on increased bronchial responsiveness during 208. Habib MP, Tank LJ, Lane LC, Garewal HS. Eect of vitamin E on
upper airway infection. Respiration 55: 21419, 1989. exhaled ethane in cigarette smokers. Chest 115: 68490, 1999.
190. Rautalahti M, Virtamo J, Haukka J, Heinonen OP, Sundvall J, 209. Allard JP, Royall D, Kurian R, Muggli R, Jeejeebhoy KN. Critical
Albanes D, Huttunen JK. The eect of alpha-tocopherol and beta- assessment of body-composition measurements in malnourished

311
Asthma and COPD: Basic Mechanisms and Clinical Management

subjects with Crohns disease: The role of bioelectric impedance analy- 227. Izumi M, McDonald MC, Sharpe MA, Chatterjee PK,Thiemermann C.
sis. Am J Clin Nutr 59: 88490, 1994. Superoxide dismutase mimetics with catalase activity reduce the organ
210. Steinberg FM, Chait A. Antioxidant vitamin supplementation and injury in hemorrhagic shock. Shock 18: 23035, 2002.
lipid peroxidation in smokers. Am J Clin Nutr 68: 31927, 1998. 228. Chatterjee PK, Patel NS, Kvale EO, Brown PA, Stewart KN, Mota-
211. Aghdassi E, Royall D, Allard JP. Oxidative stress in smokers supple- Filipe H, Sharpe MA, Di Paola R, Cuzzocrea S, Thiemermann C.
mented with vitamin C. Int J Vitam Nutr Res 6(9): 4551, 1999. EUK-134 reduces renal dysfunction and injury caused by oxidative
212. Lykkesfeldt J, Christen S, Wallock LM, Chang HH, Jacob RA, and nitrosative stress of the kidney. Am J Nephrol 24: 16577, 2004.
Ames BN. Ascorbate is depleted by smoking and repleted by mod- 229. Sharpe MA, Ollosson R, Stewart VC, Clark JB. Oxidation of nitric
erate supplementation: A study in male smokers and nonsmok- oxide by oxomanganese-salen complexes: A new mechanism for cel-
ers with matched dietary antioxidant intakes. Am J Clin Nutr 71: lular protection by superoxide dismutase/catalase mimetics. Biochem J
53036, 2000. 366: 97107, 2002.
213. Antonicelli F, Parmentier M, Drost EM, Hirani N, Rahman I, 230. Cuzzocrea S, Thiemermann C, Salvemini D. Potential therapeutic
Donaldson K, MacNee W. Nacystelyn inhibits oxidant-mediated eect of antioxidant therapy in shock and inammation. Curr Med
interleukin-8 expression and NF-kappab nuclear binding in alveolar Chem 11: 114762, 2004.
epithelial cells. Free Radic Biol Med 32: 492502, 2002. 231. Jozsef L, Filep JG. Selenium-containing compounds attenuate perox-
214. Antonicelli F, Brown D, Parmentier M, Drost EM, Hirani N, ynitrite-mediated NF-KappaB and AP-1 activation and interleukin-8
Rahman I, Donaldson K, MacNee W. Regulation of LPS-mediated gene and protein expression in human leukocytes. Free Radic Biol Med
inammation in vivo and in vitro by the thiol antioxidant nacystelyn. 35: 101827, 2003.
Am J Physiol Lung Cell Mol Physiol 286: L1319L1327, 2004. 232. Haddad E, McCluskie K, Birrell MA, Dabrowski D, Pecoraro M,
215. Gillissen A, Jaworska M, Orth M, Coner M, Maes P, App EM, Underwood S, Chen B, De Sanctis GT, Webber SE, Foster ML,
Cantin AM, Schultze-Werninghaus G. Nacystelyn, a novel lysine salt Belvisi MG. Dierential eects of ebselen on neutrophil recruitment,
of N-acetylcysteine, to augment cellular antioxidant defence in vitro. chemokine, and inammatory mediator expression in a rat model of
Respir Med 91: 15968, 1997. lipopolysaccharide-induced pulmonary inammation. J Immunol 169:
216. App EM, Baran D, Dab I, Malfroot A, Coner M, Vanderbist F, 97482, 2002.
King M. Dose-nding and 24-h monitoring for ecacy and 233. Day BJ. Catalytic antioxidants: A radical approach to new therapeu-
safety of aerosolized nacystelyn in cystic brosis. Eur Respir J 19: tics. Drug Discov Today 9: 55766, 2004.
294302, 2002. 234. Arts IC, Hollman PC. Polyphenols and disease risk in epidemiologic
217. Moretti M, Bottrighi P, Dallari R, Da PR, Dolcetti A, Grandi P, studies. Am J Clin Nutr 81: 317S325S, 2005.
Garuti G, Guanti E, Roversi P, De GM, Potena A. The eect of 235. Knekt P, Kumpulainen J, Jarvinen R, Rissanen H, Heliovaara M,
long-term treatment with erdosteine on chronic obstructive pul- Reunanen A, Hakulinen T, Aromaa A. Flavonoid intake and risk of
monary disease: The EQUALIFE Study. Drugs Exp Clin Res 30: chronic diseases. Am J Clin Nutr 76: 56068, 2002.
14352, 2004. 236. Santus P, Sola A, Carlucci P, Fumagalli F, Di GA, Mondoni M,
218. Zheng JP, Kang J, Huang SG, Chen P, Yao WZ, Yang L, Bai, CX, Carnini C, Centanni S, Sala A. Lipid peroxidation and 5-lipoxygen-
Wang CZ, Wang C, Chen BY, Shi Y, Liu CT, Chen P, Li Q, Wang ase activity in chronic obstructive pulmonary disease. Am J Respir Crit
ZS, Huang YJ, Luo ZY, Chen FP, Yuan JZ, Yuan BT, Qian HP, Zhi Care Med 171: 83843, 2005.
RC, Zhong NS. Lancet. 371: 2013-8, 2008. 237. Culpitt SV, Rogers DF, Fenwick PS, Shah P, De Matos C, Russell RE,
219. Bottle SE, Micallef AS. Synthesis and EPR spin trapping properties Barnes PJ, Donnelly LE. Inhibition by red wine extract, resveratrol,
of a new isoindole-based nitrone: 1,1,3-Trimethylisoindole N-oxide of cytokine release by alveolar macrophages in COPD. Thorax 58:
(TMINO). Org Biomol Chem 1: 258184, 2003. 94246, 2003.
220. Becker DA, Ley JJ, Echegoyen L, Alvarado R. Stilbazulenyl nitrone 238. Birrell MA, McCluskie K, Wong S, Donnelly LE, Barnes PJ,
(STAZN): A nitronyl-substituted hydrocarbon with the potency of Belvisi MG. Resveratrol, an extract of red wine, inhibits lipopoly-
classical phenolic chain-breaking antioxidants. J Am Chem Soc 124: saccharide induced airway neutrophilia and inammatory media-
467884, 2002. tors through an NF-KappaB-independent mechanism. FASEB J 19:
221. Salvemini D, Wang ZQ, Zweier JL, Samouilov A, Macarthur H, 84041, 2005.
Misko TP, Currie MG, Cuzzocrea S, Sikorski JA, Riley DP. A non- 239. Manna SK, Mukhopadhyay A, Aggarwal BB. Resveratrol suppresses
peptidyl mimic of superoxide dismutase with therapeutic activity in TNF-induced activation of nuclear transcription factors NF-kappa B,
rats. Science 286: 3046, 1999. activator protein-1, and apoptosis: Potential role of reactive oxygen
222. Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y, intermediates and lipid peroxidation. J Immunol 164: 650919, 2000.
Salvemini D, Voelkel NF, Flores SC. Oxidative stress and apoptosis 240. Donnelly LE, Newton R, Kennedy GE, Fenwick PS, Leung RH, Ito K,
interact and cause emphysema due to vascular endothelial growth fac- Russell RE, Barnes PJ. Anti-inammatory eects of resveratrol in
tor receptor blockade. Am J Respir Cell Mol Biol 29: 8897, 2003. lung epithelial cells: Molecular mechanisms. Am J Physiol Lung Cell
223. Muscoli C, Sacco I, Alecce W, Palma E, Nistico R, Costa N, Clementi F, Mol Physiol 287: L77483, 2004.
Rotiroti D, Romeo F, Salvemini D, Mehta JL, Mollace V. The pro- 241. Biswas SK, McClure D, Jimenez LA, Megson IL, Rahman I.
tective eect of superoxide dismutase mimetic M40401 on bal- Curcumin induces glutathione biosynthesis and inhibits NF-kappaB
loon injury-related neointima formation: Role of the lectin-like activation and interleukin-8 release in alveolar epithelial cells:
oxidized low-density lipoprotein receptor-1. J Pharmacol Exp Ther 311: Mechanism of free radical scavenging activity. Antioxid Redox Signal
4450, 2004. 7: 3241, 2005.
224. Chang LY, Crapo JD. Inhibition of airway inammation and hyperreac- 242. Shishodia S, Potdar P, Gairola CG, Aggarwal BB. Curcumin (diferu-
tivity by an antioxidant mimetic. Free Radic Biol Med 33: 37986, 2002. loylmethane) Down-regulates cigarette smoke-induced NF-kappaB
225. Smith KR, Uyeminami DL, Kodavanti UP, Crapo JD, Chang LY, activation through inhibition of IkappaBalpha kinase in human lung
Pinkerton KE. Inhibition of tobacco smoke-induced lung inamma- epithelial cells: Correlation with suppression of COX-2, MMP-9 and
tion by a catalytic antioxidant. Free Radic Biol Med 33: 110614, 2002. cyclin D1. Carcinogenesis 24: 126979, 2003.
226. Doctrow SR, Human K, Marcus CB, Musleh W, Bruce A, Baudry M, 243. Meja KK, Rajendrasozhan S, Adenuga D, Biswas SK, Sundar IK,
Malfroy B. Salen-manganese complexes: Combined superoxide dis- Spooner G, Marwick JA, Chakravarty P, Fletcher D, Whittaker P,
mutase/catalase mimics with broad pharmacological ecacy. Adv Megson IL, Kirkham PA, Rahman I. Am J Respir Cell Mol Biol. 2008
Pharmacol 38: 24769, 1997. (In press).

312
Chemokines
26 CHAPTER

Tissue infection or injury induces a local inam- recruitment of these cells to sites of injury or James E. Pease and
matory response in which the accumulation of infection. Timothy J. Williams
white blood cells from the blood is an essen- The chemokine gradient, which the cells
tial feature. The type of infection or injury follow, is produced constitutively in specialized Leukocyte Biology Section, National
determines the types of leukocyte which are locations, or upregulated at sites of inamma- Heart and Lung Institute, Imperial
recruited as appropriate for the necessary local tion, such as in asthma or chronic obstructive College London, London, UK
host defense and repair response. Inammatory pulmonary disease (COPD). The chemokines
diseases can result from an inappropriate or produced locally stimulate integrins on the leu-
overzealous inammatory response. Such kocyte to attach to complementary receptors on
inammation, when it occurs in the lung, can the microvascular endothelium and cytoskeletal
seriously compromise the normal function of the changes necessary for attachment and migration
organ and can be life-threatening. Thus, there is through the barrier. Similar processes mediate
considerable interest in the mechanisms under- the movement of the cell in the tissue toward
lying leukocyte recruitment and the endog- the inammatory focus and elsewhere if appro-
enous molecules regulating the process, with an priate (e.g. to lymph nodes).
ultimate aim of designing targeted therapy to Until recently, little was known about
attenuate the tracking of specic cell types. how the immune system organizes its geogra-
Leukocyte recruitment represents a rene- phy under basal conditions or how specic cell
ment of a fundamental property of a motile cell types can be recruited to sites of inammation.
to detect a chemical gradient, orient itself, and Chemoattractants, such as complement-derived
move along the gradient. C5a, leukotriene B4, and platelet activity factor,
This process, known as chemotaxis, has had been discovered to be chemotactic in vitro
been elegantly described and analyzed in primi- in Boyden chamber chemotaxis assays, but none
tive single-celled organisms and is an essential had the specicity necessary for many of the
property of cells at dierent stages of their life migratory responses seen in the body. This situ-
history in multicellular organisms. In higher ation changed dramatically with the discovery
animals, this property has been adapted to trap of the rst chemokines in the late 1980s and
specic leukocyte types on the inner surface the growing number of family members that
of small blood vessels and induce migration have been discovered over the last decade.
through the vessel wall into the tissues. Pivotal
to the process of chemoattraction of leukocytes
are the chemokines, a large family of chemo-
tactic proteins, typically of 10 kDa; see Rot and
Von Andrian [1] for a review. These proteins
CHEMOKINES
act upon specic G-protein-coupled receptors
(GPCRs) found on the surface of cells and have Chemokines have multiple roles in the organi-
a prominent role in the immune system, where zation of the immune system under basal condi-
they are essential for both the localization of tions and during infection, and are also involved
dierent types of leukocytes in tissue compart- in angiogenesis. A crucial role of chemokines is
ments under basal conditions and the rapid the recruitment of dierent types of leukocytes

313
Asthma and COPD: Basic Mechanisms and Clinical Management

from the blood to the sites of inammation. Selective cell has only one pair of cysteines and a single disulde bond [3]
recruitment is achieved by means of locally produced chem- and fractalkine has a CXXXC motif at the N-terminus [4].
okines, the particular chemokine secreted depends on the Fractalkine is also unusual in that it is produced attached
type of stimulus and the tissue involved. Specicity is pro- to a cell-bound mucin stalk, a property shared by the
vided by the expression of dierent types of receptor on dif- CXC chemokine CXCL16 [5]. This allows them to also
ferent leukocyte types. function as adhesion molecules, tethering cells expressing
Most of the known chemokines (Table 26.1) belong the cognate receptor.
to two major families dened by the position of four con- Chemokines are thought to bind to presenting mol-
served cysteine residues (Fig. 26.1). The largest family is the ecules on the luminal surface of the venular endothelium,
CC chemokine family (28 in number) which possess two such as glycosaminoglycans, where the chemokines engage
adjacent cysteines in the vicinity of the N-terminus of the their receptors on the leukocyte surface. Leukocytes typi-
mature peptide. The CXC family (16 members) have two cally roll on the endothelium through low anity intermo-
cysteine residues in this same region, but with an inter- lecular interactions mediated by selectins. Stimulation of
posed amino acid. In both families, the rst cysteine forms chemokine receptors induces the upregulation of adhesion
a disulde bond with the third, and the second with the molecules on the leukocyte surface and cytoskeletal changes
fourth cysteine. CXC chemokines are characteristically necessary for rm attachment, followed by emigration.
chemotactic for neutrophils and lymphocytes, whereas CC Chemokines bind to and transduce their actions via
chemokines act upon the majority of leukocytes, but gen- specic GPCRs [7, 8]. To date, 6 CXC and 10 CC chem-
erally have little activity on neutrophils. CXC chemokines okine receptors have been characterized, with single recep-
can be further subdivided into those possessing a gluta- tors identied for fractalkine and for lymphotactin. The
mateleucinearginine (ELR) motif immediately before the majority of receptors bind and signal in response to several
rst cysteine, and those lacking the ELR motif. Members chemokine ligands, but such promiscuity is generally class-
of the former, such as CXCL1, CXCL8, and CXCL7, are restricted, with CC chemokines binding only to CC chem-
associated with angiogenic properties, while those lacking okine receptors. One exception to this is the Duy antigen
the ELR motif are typically inducible by IFN- and are receptor for chemokines (DARC) which can bind chemok-
angiostatic, such as CXCL9, CXCL10, and CXCL11 [2]. ines of both classes and has a role in clearance of chemok-
There are some nonconforming chemokines: lymphotactin ines. This receptor is present on erythrocytes and endothelial

TABLE 26.1 CXC Chemokines, and their receptors.

Chemokine Original name Chemokine receptor(s) Chemokine Original name Chemokine receptor(s)

CXCL1 GRO-/MGSA- CXCR2  CXCR1 CCL8 MCP-2 CCR3

CXCL2 GRO-/MGSA- CXCR2 (CCL9/10) Unknown Unknown

CXCL3 GRO-/MGSA- CXCR2 CCL11 Eotaxin CCR3

CXCL4 PF4 Unknown (CCL12) Unknown Unknown

CXCL5 ENA-78 CXCR2 CCL13 MCP-4 CCR2, CCR3

CXCL6 GCP-2 CXCR1, CXCR2 CCL14 HCC-1 CCR1

CXCL7 NAP-2 CXCR2 CCL15 HCC-2/Lkn-1/MIP-1 CCR1, CCR3

CXCL8 IL-8 CXCR1, CXCR2 CCL16 HCC-4/LEC CCR1

CXCL9 Mig CXCR3 CCL17 TARC CCR4

CXCL10 IP-10 CXCR3 CCL18 DC-CK1/PARC AMAC-1 Unknown

CXCL11 I-TAC CXCR3 CCL19 MIP-3/ELC/exodus-3 CCL7

CXCL12 SDF-1/ CXCR4 CCL20 MIP-3/LARC/exodus-1 CCR6

CXCL13 BLC/BCA-1 CXCR5 CCL21 6Ckine/SLC/exodus-2 CCR7

CXCL14 BRAK/bolekine Unknown CCL22 MDC/STCP-1 CCR4

(CXCL15) Unknown Unknown CCL23 MPIF-1 CCR1

CXCL16 SexCkine CXCR6 CCL24 MPIF-2/eotaxin-2 CCR3

The systematic names, together with the colloquial names and agoinst activity of known receptors ae shown. Some chemokines appear to be missing from the list, e.g. CCL6. In
these cases please note that whilst a chemokinase of that name has been identified in the mouse, no human orthologue has been documented.

314
Chemokines 26
cells, whereas a second receptor, D6 is located primarily on associated with multiple colloquial names for the same
the lymphatic endothelium and is involved in the selective ligands. Chemokine receptors are designated as CXCRn,
uptake of inammatory chemokines [9]. CCRn, CX3CR1, and XCR1 with the chemokine ligands
The chemokines and their receptors have now been that stimulate these receptors now designated CXCLn,
given a nomenclature to alleviate some of the problems CCLn, CX3CLn, and XCLn, respectively (Table 26.1).

(A)
CC C C
NH2 CC or Class COOH
CHEMOKINES IN COPD
CC C C
NH2 CXC or Class COOH
C C Neutrophils have been strongly implicated in the patho-
NH2 C or Class COOH genesis of COPD and are found in high numbers in lung
C C C C tissue and sputum [10]. It is believed that activation of
NH2 CX3C or Class COOH these cells to release activated oxygen species and proteases,
such as elastase, is important in the specic pattern of lung
(B) damage and chronic dysfunction that characterize COPD.
Consequently, there is considerable interest in the mecha-
nisms involved in the recruitment of these cells to the lung
with the aim of identifying opportunities for potential ther-
apeutic intervention (Fig. 26.2).
CXCL8/IL-8 is a CXC chemokine that is a potent
chemoattractant for neutrophils and was amongst the
rst of the chemokines to be discovered and characterized
[1113]. Early studies showed that this chemokine was
(C) present in the inammatory exudate induced in a model of
NH2
microbial infection in vivo [14, 15] and its production was
often preceded by a phase of appearance of the complement
fragment, C5a, which is also a potent neutrophil attract-
ant but is generated in tissue uid rather than as a secretory
product of a cell [16]. Neutrophils themselves have been
shown to produce CXCL8 during phagocytosis [17, 18],
these eects can be blocked by antibodies to integrins and
COOH
platelet-activating factor antagonists [19]. A similar pattern
of chemoattractant production, C5a followed by CXCL8,
FIG. 26.1 (A) Linear representations of the major chemokine classes. has also been observed in response to myocardial ischemia,
(B) Primary structure of the human homolog of CCL11/Eotaxin showing and interestingly the CXCL8 production is dependent on
disulfide bridges. (C) The secondary structure of human CCL11 is the presence of neutrophils in the heart implying that
represented as a ribbon model, modeled from the coordinates given in neutrophils are the source of the chemokine [20]. In a lung
Crump et al. using the Pymol software [6]. model of ischemia, neutralization of CXCL8 by an antibody

Cigarette smoke /irritants

Epithelium

M
CXCL1 CXCL9 CCL2
CXCL6 CXCL10 CXCL1
CXCL8 CXCL11

Endothelium R2 CX
CXCR2 CXCR3
CC CR
2
Venule
FIG. 26.2 Mechanisms underlying leukocyte recruitment
in COPD. The roles of CXC chemokines in recruiting
Neutrophil CD8 T-cell Monocyte
neutrophils, monocytes and T-cells to the COPD lung are
illustrated.

315
Asthma and COPD: Basic Mechanisms and Clinical Management

suppresses tissue damage, thus providing a link between of CXCR3 by peripheral blood CD8 T-cells has been
neutrophils and injury to lung tissues [21]. reported to be increased in smokers with COPD compared
CXCL8 was rst discovered as a secretory product of to controls [46]. Similarly, increased numbers of CXCR3
stimulated macrophages and other cells [11]. It stimulates CD8 T-cells have been reported within the epithelium
neutrophils via two dierent receptors namely CXCR1 and and submucosa of lung biopsies from smokers with COPD,
CXCR2 [22, 23]; another related chemokine, CXCL6/ compared with nonsmoking control subjects [47]. This sug-
GCP-2 can stimulate both receptors [24, 25]. There are gests that this receptor and its ligands may play a role in
several other chemokines that can stimulate CXCR2 only their recruitment to the lung. Since the CXCR3 ligands
(e.g. CXCL1/GRO-, CXCL2/GRO-, CXCL3/GRO-, CXCL9, CXCL10, and CXCL11 are all induced by IFN-
and CXCL7/NAP-2). CXCL8 appears to be an important and CD8 T-cells are themselves a source of IFN-, there
mediator of neutrophil accumulation in COPD. Elevated is a great potential for positive feedback. A more recent
levels of CXCL8 have been detected in the bronchoalveo- study examining 11 dierent chemokine receptors, found
lar lavage (BAL) uid, induced sputum, and in the circu- that in addition to CXCR3, CCR5, and CXCR6 expression
lation of patients with the disease compared with smoking by lung CD8 T-cells was also increased in COPD patients
and nonsmoking controls [2629] with the levels of and the levels of expression correlated with COPD severity
CXCL8 correlating with neutrophil accumulation [3032]. [48], the cells displaying a Tc1, CD45RA eector memory
Increases in CXCL8 levels in the sputum from COPD phenotype. Since the LTB4 receptor BLT1 has also been
patients have also been documented during disease exacer- shown to be expressed by this subset [49], an additional role
bation [33, 34] although there appears to be some debate for leukotrienes in cell recruitment may be likely.
as to whether there is correlation with acute inammation
and viral/bacterial airway infections. Likewise, elevated lev-
els of CXCL1 [35] and CXCL5 [36] have been detected
in the sputum and BAL uid of COPD patients com-
pared with smoking and nonsmoking controls, although
CHEMOKINES IN ASTHMA
levels of the latter chemokine were not signicantly dif-
ferent in normal smokers versus patients with emphysema Considerable evidence underpins the link between the inl-
[36]. The source of CXCL1 in COPD patients appears to tration of inammatory cells in the lung and the progressive
be predominantly bronchial epithelial cells under the inu- changes in lung pathophysiology that characterize asthma.
ence of TNF- [37]. The chemotactic activity of CXCL1 For example, the late asthmatic response to allergen expo-
for monocytes in addition to neutrophils, may explain in sure is associated with activation of Th2-lymphocytes that are
part the marked increase in alveolar macrophage numbers believed to regulate the massive eosinophil inux that occurs.
observed in COPD patients compared with normal con- Eosinophils are thought to make an important contribution to
trols [38]. The upregulation of MCP-1/CCL2 in the spu- lung damage and dysfunction by releasing their toxic granular
tum and BAL of patients with COPD [35, 39] may also contents which directly induce tissue damage and also by the
play a role in this process as it too is a potent recruiter of release of cytokines, such as TGF-, which plays an impor-
monocytes [40]. Since the engagement of chemokines with tant role in tissue remodeling [50]. The recent generation of
their cognate receptor usually leads to receptor endocytosis, mice decient in eosinophils has allowed the role of the cell
this is the likely explanation for the nding that, compared to be probed. Data from allergen-challenge models with these
with control subjects, the cell surface expression of CXCR2 mice implicate the eosinophil in both airways remodeling and
on peripheral blood neutrophils is lower in patients with airway hyperresposiveness [5153], although this is the sub-
COPD than healthy controls [41]. Enhanced chemotaxis ject of controversy [54]. For this reason, there is considerable
of monocytes from COPD patients in response to the interest in the endogenous chemoattractants involved in the
CXC chemokines CXCL1/GRO- and NAP-2/CXCL7, recruitment of eosinophils to the lung. Since eosinophils are
compared with monocytes from smoking and nonsmoking believed to have evolved in host defense to parasitic worms,
control subjects has also been reported, although this was these cells are an attractive therapeutic target in the Western
apparently not due to dierences in expression of either world. In contrast, neutrophils present diculties as a target,
CXCR1 or CXCR2 on the cell surface [42]. as they are essential for survival against microbial infection.
In addition to the neutrophil inux observed in the
lungs of COPD patients, increased numbers of CD8
T-cells have also been reported [43]. These cells are a major
source of IFN- which itself, when expressed as a lung- EOSINOPHIL CHEMOATTRACTION
targeted transgene in mice, can induce emphysema with
classical symptoms of alveolar enlargement, enhanced lung
volumes, enhanced pulmonary compliance, and an inam- The selective recruitment of eosinophils during the late
matory inltrate rich in both macrophages and neutrophils response to allergen in sensitized individuals implies the
[44]. This suggests a primary role for CD8 T-cells in the existence of endogenous selective eosinophil chemoattract-
orchestration of leukocyte recruitment and subsequent ants (Fig. 26.3). A protein was puried from BAL uid of
pathology. Supportive of this, a recent study showed that allergen-challenged, sensitized guinea pigs that had this prop-
CD8 T-cell-decient mice had a blunted inammatory erty [55]. This protein was sequenced and named Eotaxin
response and did not develop emphysema when exposed as it was found to exhibit high specically in recruiting eosi-
to long-term cigarette smoke [45]. Increased expression nophils when intradermally injected into nave recipients [55].

316
Chemokines 26
Allergen

Epithelium

APC

IL-4
IL-13
Smooth muscle IL-5 Eosinophil survival,
priming
CCL11 Th-2 cell
CCL24
Rolling CCL26
Adhesion Emigration

Selectins 41 VCAM
CCR3 Venule
FIG. 26.3 Mechanisms underlying eosinophil
Eosinophil recruitment in asthma. An example of the links
between the cytokine, chemokine and cellular
adhesion systems mediating the recruitment
Endothelium of eosinophils to sites of allergic inflammation .

Eotaxin/CCL11 is expressed by many cells [5666] and that can be recruited to a site of allergic inammation [87].
its generation within the lung appears to be dependent upon In addition to the eotaxins, an increasing number of CC
the presence of T-cells [67]. CCL11 orthologs have been chemokines, have been found to activate CCR3 with vary-
discovered in several species, including human, mouse, rat, ing potency, including CCL5/RANTES and members of the
and macaque [6872]. CCL11 is a highly potent chemoat- monocyte chemotactic proteins such as CCL2, CCL7, and
tractant for eosinophils acting via a single chemokine recep- CCL13, but as these chemokines also activate other chemok-
tor, CCR3, that is highly expressed on eosinophils [72], but ine receptors, they are relatively nonselective (Table 26.2).
also on Th2 cells [73], basophils [74], and mast cells [75, 76], Dierent approaches have been made to determine
supportive of a critical role for this molecule in the process of the role of particular chemokines in eosinophil recruitment
allergic inammation. Two additional functional homologs, in vivo, including the use of neutralizing antibodies against
with low sequence similarity but closely related biological both CCR3 and CCL11 which inhibited the migration of
properties, have been described in the human and are named eosinophils in vivo [90, 91]. Studies with mice harboring a
CCL24/Eotaxin-2 and CCL26/Eotaxin-3 [7781]. Despite disrupted gene for CCL11 have shown conicting results;
sharing the same colloquial name, both CCL24 and CCL26 BALB/c mice decient in CCL11 showed a reduction in
are only distantly related to CCL11, being encoded in dier- ovalbumin-induced lung eosinophilia [92] whilst, in contrast,
ent chromosomal locations and sharing only low identity at CCL11-decient mice of the outbred ICR strain exhibited
the amino acid level. In terms of their ability to recruit eosi- no dierence in allergen-induced eosinophil recruitment.
nophils in assays of chemotaxis in vitro, CCL11 and CCL24 Use of CCR3-decient mice has also been informa-
exhibit similar potencies, whilst CCL26 has been reported to tive. Reduced eosinophil numbers in the airways of
be an order of magnitude less potent [77]. All three signal intraperitoneal-sensitized, aerosol-challenged CCR3-
specically via CCR3. Animal studies have highlighted the decient mice have been reported by Humbles et al., with
production of both CCL11 mRNA and CCL11 protein in the eosinophils trapped within the vasculature, unable
the lung during the early response to allergen exposure [55, to migrate into the lung parenchyma [93]. Intriguingly,
8284]. In humans, increased expression of CCL11 and CCR3-decient mice also exhibited elevated airway hyper-
CCL24 at both the mRNA and the protein level has been responsiveness (AHR) to methacholine upon ovalbu-
observed in the allergic lung of both atopic and nonatopic min challenge which was attributed to increased numbers
asthmatics [56, 59, 60, 85] and the proteins are also found in of intraepithelial mast cells within the trachea. The routes
induced sputum [60, 86]. In both animal and human studies of immunization used for allergen sensitization appear
of allergic airways disease, airway epithelium, microvascular to make an important contribution, as intradermal sen-
endothelium, tissue macrophages, and inltrating inamma- sitization of mice with ovalbumin led to reduced AHR
tory cells have been shown to express eotaxins [56, 57, 59, responses in CCR3-decient mice following methacholine
60]. CCL11 has also been shown to act in concert with the and ovalbumin challenge [94]. More recently, a study using
important eosinophil survival factor IL-5, promoting eosi- Aspergillus as the challenge, rather than ovalbumin, found
nophil recruitment from the microcirculation [87] and eosi- reduced numbers of perivascular and peribronchial eosi-
nophil mobilization from the bone marrow [84, 88, 89]. Thus, nophils in both CCR3-decient mice and double knockout
CCL11 along with other cytokines may further contribute CCL11/CCL24-decient mice [52]. Deletion of the genes
to eosinophilia. Indeed, the number of eosinophils in the for CCL11 and the Th2 cytokine IL-5 in double knock-
blood appears to be an important determinant of the number out mice appears to be particularly eective in suppressing

317
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 26.2 Cellular specificity of chemokines.

Systematic Receptor Principle cells Systematic Receptor Principle cells


name Colloquial name usage recruited name Colloquial name usage recruited

CCL1 I-309 CCR8 Mo, T, NK CXCL1 GRO-/MGSA- CXCR2 No, Mo


CCL2 MCP-1/MCAF CCR2 Mo, DC, T, Bs CXCL2 GRO-/MGSA- CXCR2 No, Mo
CCL3 MIP-1/LD78 CCR1, 5 Mo, DC, Eo, Bs, T, No, NK CXCL3 GRO-/MGSA- CXCR2 No, Mo
CCL3L1 LD78 CCR1, 5 Mo, No, DC, Eo, Bs, T, NK CXCL4 PF-4 Unknown T, No, Mo
CCL4 MIP-1 CCR5 Mo, DC, T CXCL5 ENA-78 CXCR2 No, Mo
CCL4L1 LAG-1 CCR5 Mo, DC, T CXCL6 GCP-2 CXCR1, 2 No, Mo
CCL5 RANTES CCR1, 3, 5 Mo, DC, Eo, Bs, T, No, NK CXCL7 NAP-2 CXCR2 No, Mo
CCL7 MCP-3 CCR1, 2, 3 Mo, DC, Eo, Bs, T, No, NK CXCL8 IL-8 CXCR1, 2 No, Mo
CCL8 MCP-2 CCR3 Eo, T, Bs, Mc CXCL9 Mig CXCR3 T, B
CCL11 Eotaxin CCR3 Eo, Bs, T CXCL10 IP-10 CXCR3 T, B
CCL13 MCP-4 CCR2, 3 Eo, T, Bs, Mc Mo, DC, CXCL11 I-TAC CXCR3 T, B
CCL14 HCC-1 CCR1 Mo, DC, Eo, Bs, T, No, NK CXCL12 SDF-1/ CXCR4 T, B, DC, Mo
CCL15 HCC-2/Lkn- CCR1, 3 Eo, T, Bs, Mc CXCL13 BLC/BCA-1 CXCR5 T, B
1/MIP-1/MIP-5
CCL16 HCC-4/LEC CCR1 Mo, DC, Eo, Bs, T, No, NK CXCL14 BRAK/bolekine unknown
CCL17 TARC CCR4 DC, T, Bs, NK CXCL16 SR-PSOX CXCR6 T
CCL18 DC-CK 1/AMAC- unknown T
1/MIP-5/PARC
CCL19 MIP-3/ELC/ CCR7 DC, T, B, NK XCL1 Lymphotactin/ SCM-1/ATAC XCR1 T, NK
CCL20 MIP-3/LARC CCR6 DC, T XCL2 SCM-1 XCR1 T, NK
CCL21 SLC/6Ckine CCR7 DC, T, B, NK
CCL22 MDC/STCP-1 CCR4 DC, T, Bs, NK CX3CL1 Fractalkine, neurotactin CX3CR1 T, NK, DC, Mo
CCL23 MPIF-1 CCR1 Mo, DC, Eo, Bs, T, No, NK
CCL24 MPIF-2/eotaxin-2 CCR3 Eo, Bs, T
CCL25 TECK CCR9 T
CCL26 Eotaxin-3 CCR3 Eo, Bs, T
CCL27 CTACK/ALP/ILC/ CCR10 T
ESkine

CCL28 MEC CCR10, 3 Eo, T, Bs, Mc

B: B-lymphocyte; Bs: basophil; DC: dendritic cell: Eo, eosinophil; Mc: mast cell; Mo: monocyte; NK: natural killer cell; No: neutrophil; T: T-lymphocyte.

airway eosinophilia and hyperresponsiveness [95]; interest- [96] allowing them to be recruited by the chemokine CCL20,
ingly, IL-13 levels were markedly reduced in these animals. produced by lymphocytes and monocytes and markedly
upregulated by tumor necrosis factor or lipopolysaccharide
[97, 98]. This receptor is downregulated during maturation
CHEMOATTRACTION OF DENDRITIC CELLS and is replaced by CCR7, allowing the cells to respond to
the chemokines CCL19 [99, 100] and CCL21 produced in
the lymphatic system [101]. This maturation is accompanied
Dendritic cells (DCs) are important sentinels of the immune by the production of chemokines, such as CCL22, which
system, and their migration to lymph nodes following acti- can recruit activated T-cells expressing CCR4 [102]. The
vation and encounter with antigens is crucial to the adaptive importance of CCR7 in animal models of allergy has been
immune response. In the context of allergic lung inammation, demonstrated by the nding that mAb-mediated neutrali-
CCR6 has been shown to be expressed by lung-derived DCs zation of CCR7 in a Severe Combined Immunodeciency

318
Chemokines 26
mouse reconstituted with Peripheral blood mononu- toward Th2 cells via a contact-dependent mechanism [127].
clear cells from allergic humans, impaired DC homing to Similarly in the human, CD4CD25 regulatory T-cells
lymph nodes following antigen challenge, accompanied by a puried from peripheral blood have been reported to express
decrease in both Th2 cytokine production and T-cell recruit- CCR4 and CCR8 which is likely to facilitate their migration
ment [103]. Likewise, mice decient in the transcription toward antigen-presenting cells (APCs) and activated T-cells,
factor Runx3, which have enhanced expression of CCR7 on enabling them to inhibit APC function or suppress respond-
alveolar DCs, have an accumulation of activated DCs within ing T-cells respectively [128]. T-cell polarization may also be
the lymph nodes which is associated with features character- amplied by chemokines acting as antagonists for example,
istic of allergic asthma, such as increased serum IgE levels and CXCL9, CXCL10, and CXCL11, ligands for the Th1 asso-
AHR to methacholine [104]. ciated receptor CXCR3, can also act as antagonists of CCR3
both in vitro [129] and in vivo [130].

CHEMOATTRACTION OF T-LYMPHOCYTES
CHEMOATTRACTION OF BASOPHILS
T-cell depletion has been shown to prevent allergen-
induced CCL11 generation and eosinophil accumulation in Basophils are the least common granulocyte in peripheral
the lungs of sensitized mice, emphasizing the pivotal role blood, but they accumulate in signicant numbers in certain
for T-cells in allergic disease [67]. A link between Th2 cells types of inammatory responses [131]. Basophils contribute
and eosinophil recruitment is well established, with the Th2 to the cellular inltrate in asthmatic lung and are promi-
cytokines, IL-4, and IL-13 stimulating CCL11 and CCL26 nent in late-phase cutaneous allergic reactions. Although,
production by other cells in vitro [58, 63, 105107]. In relative to eosinophils and neutrophils, basophils contain
addition, expression of CXCR4 [108] by nave T-cells can small amounts of cytotoxic proteins and proteases, they are
be upregulated by IL-4 [109]. In murine models of allergic able to release mediators, such as histamine, in response to
airway disease, neutralizing mAbs to CXCR4 and its with cross-linking of their Fc R1 receptors by allergens. Further,
ligand CXCL12 were reported to reduce lung eosinophilia basophils are thought to contribute large amounts of the
and airway hyperresponsiveness [110]. Likewise, treatment Th2 cytokines, IL-4 and IL-13 [132].
of allergic mice with the CXCR4 antagonist AMD3100 A major observation was that basophils express the
resulted in signicant reduction in airway hyperresponsive- CCR3 receptor and CCL11 is highly chemotactic for these
ness, eosinophilia, and the production of Th2 associated cells [74, 133]. However, the preferential recruitment of
cytokines such as IL-4, IL-5, CCL17, and CCL22 [111] basophils in certain types of inammatory response (e.g. the
supportive of a role for the CXCR4:CXCL12 axis in allergic cutaneous response to ectoparasitic ticks) suggests that more
airways disease. Th2 cells polarized in vitro express the selective basophil chemoattractant molecules exist. Despite
chemokine receptors CCR3, CCR4, and CCR8 [73, 112, this, no such molecule has been identied. One possibility
113]. Such polarization has also been observed in vivo, is that two or more chemokines acting in combination con-
with T-cells recovered from the allergic airways of humans trol basophil recruitment in vivo. Using a system for meas-
shown to preferentially express CCR3, CCR4, and CCR8 uring shape change in basophils with ow cytometry, some
[114116]. Consistent with this, the CCR4 ligands CCL22 evidence has been provided for this [134]. Combinations of
and CCL17 are upregulated in the human lung following ligands acting via CCR2 and CCR3 were the most eective
allergen challenge [117, 118]. in this system. Alternatively, nonchemokine molecules may
In mice, mAb-mediated neutralization of CCL11 and have potent stimulatory eects on basophils. In this context,
CCL22 has been shown to impede the early recruitment of an extract of nasal polyps was found to have potent stim-
Th2 cells to the allergic lung with CCL22 responsible for ulatory eects on basophils. This activity was found to be
Th2 cell recruitment following repeated antigen stimulation due to insulin-like growth factor-1 and -2 (IGF-1 and -2)
[119]. Studies using mice decient in CCR4 and CCR8 [135]. The IGFs induce increase random migration
have failed to conclusively prove a requirement for either (i.e. chemokinesis). This action, not observed with eosi-
receptor in allergic airways disease [120122] and likewise, nophils, had a marked potentiating eect on chemotaxis
mAb-mediated neutralization of CCR4 in guinea pigs or along a gradient induced by CCL11.
CCL1 in mice had little eect upon T helper cell recruit-
ment following allergen challenge [123, 124].

Regulatory T-cells CHEMOATTRACTION OF MAST CELLS


An inbalance or defect in the activity of regulatory Mast cells pose a particular challenge in the identication
CD4CD25 T-cells may result in allergic disease [125, of chemoattractant mechanisms as recruitment is from a
126]. CCR4 and CCR8 are also reported to be co-expressed small circulating pool of progenitors in the blood. Recently,
by CD4CD25 regulatory T-cells, in both murine and it has been found that mast cell progenitors express the
human systems. In the former, these cells have been reported high anity BLT1 receptor [136]. As the ligand for this
to suppress the dierentiation of murine CD4 T-cells receptor, LTB4, is secreted by activated mature mast cells,

319
Asthma and COPD: Basic Mechanisms and Clinical Management

this may explain part of the mechanism underlying local A selection of prototypic chemokine receptor antagonists of
mast cell hyperplasia associated with allergic reactions. The relevance to this chapter are shown in Fig. 26.4.
cells mature and proliferate in the tissue under the inuence The rst low-molecular-weight compound to be
of local factors, such as stem cell factor. During maturation, developed against a chemokine receptor was a CXCR2
the BTL1 receptor is downregulated [136], and it is likely antagonist [142]. This compound, SB225002, is a potent
that other chemoattractants and their receptors are involved antagonist that blocks CXCL8 binding and inhibits neu-
in moving the cells along local tissue gradients to mediate trophil chemotaxis in vitro and neutrophil accumulation
the localization of the long-lived tissue-resident mature in vivo . It is therefore plausible that CXCR2 antagonists
cells. Several chemokine receptors have been identied on could be developed to prevent neutrophil accumulation as
mast cells [76, 137, 138], and their ligands may be impor- observed in the lung of COPD patients. However, such
tant in this localization in the tissues. Of particular inter- eects would have to be monitored carefully because neu-
est is the nding that mast cells move into airway smooth trophils are essential for host defense against microbial
muscle layers in asthmatic patients [139]. These mast cells pathogens, as previously discussed. The SB225002 com-
have been found to express CXCR3 and one of its ligands, pound has shown ecacy in several animal models of dis-
CCL10, is produced by asthmatic smooth muscle cells ease including bronchopulmonary dysplasia [143] and
[140]. These observations may indicate an important mech- arthritis [144] although some of this ecacy may be due to
anism underlying lung dysfunction in asthmatic patients. o-target eects as recent proling of the compound sug-
gests it has potency at several additional targets, including
histamine and prostanoid receptors [145].
Because of its prominence on cells associated with
Th2 cell-driven allergic reactions, CCR3 has become a
CHEMOKINES AND THEIR RECEPTORS AS prime target for the development of antagonists. The rst
THERAPEUTIC TARGETS publication of such an antagonist described the eects of
a low-molecular-weight compound (UCB 35625, a struc-
ture based on a compound produced by Banyu, Japan) that
The observations outlined above provide the basis for the antagonizes CCR3 [146]. This compound also antagonizes
development of low-molecular-weight compounds aimed at CCR1, a receptor for CCL3 and CCL5. Such a bispecic
selectively preventing the recruitment of a particular leuko- compound may be advantageous as around 1520% of indi-
cyte type to a site of inammation and thus suppressing the viduals also express CCR1 at high levels on their eosinophils
pathogenesis associated with that cell type. High through- [147, 148]. UCB 35625 is unusual amongst chemokine
put screening of vast libraries comprising several millions receptor antagonists as, although it blocks the activation
of small molecules have identied many molecules that can of chemokine receptors at nanomolar concentrations, it
modify the interaction between GPCRs and their ligands; does not readily displace chemokine from the receptor. It
such compounds, either agonists or antagonists, provide appears that the antagonist binding site is located within
a large proportion of the drugs currently prescribed [141]. the transmembrane helices of the receptor [149], whilst

(A) SB 225002 (B) UCB 35625

Br OH
NH NH
N I-
O
NO2

HN O
Cl Cl
(C) A-122058

(D) GW-766994

O
N Cl FIG. 26.4 Chemical structures of the
O N N N CXCR2 antagonist (A) the bispecific CCR1
H H
O NH2 and CCR3 antagonist UCB 35625 (B), and
Cl the CCR3 antagonists A-122058 (C) and
N O GW-701897B (D).

320
Chemokines 26
the ligand binding sites are located extracellularly [150]. 4. Bazan JF, Bacon KB, Hardiman G et al. A new class of membrane-
Despite several publications describing the in vitro ecacy bound chemokine with a CX3C motif. Nature 385: 64044, 1997.
of CCR3 specic antagonists [151154], descriptions of 5. Matloubian M, David A, Engel S et al. A transmembrane CXC chem-
their ecacy in vivo lag well behind, as the molecules gen- okine is a ligand for HIV-coreceptor Bonzo. Nat Immunol 1: 298304,
erally have poor cross-reactivity with the rodent ortholog 2000.
receptors. The small molecule CCR3 antagonist A-122058 6. DeLano WL. The PyMOL Molecular Graphics System on World
(Abbott Laboratories) has been reported to be ecacious in Wide Web, 2002; Available from: http://www.pymol.org.
7. Murphy PM, Baggiolini M, Charo IF et al. International union
reducing the number of eosinophils in a mouse peritoneal
of pharmacology. XXII. Nomenclature for chemokine receptors.
model of eosinophil recruitment following injection with Pharmacol Rev 52: 14576, 2000.
CCL11 [155]. Likewise, the Yamanouchi Pharmaceutical 8. Murphy PM. International Union of Pharmacology. XXX. Update on
Company have recently described compounds with ecacy chemokine receptor nomenclature. Pharmacol Rev 54: 22729, 2002.
in a murine model of cutaneous inammation [156] and in 9. Mantovani A, Bonecchi R, Locati M. Tuning inammation and immu-
a macaque model of eosinophil recruitment to the lung fol- nity by chemokine sequestration: Decoys and more. Nat Rev Immunol
lowing bronchoprovocation with CCL11 [151]. 6: 90718, 2006.
To date, the only small molecule antagonist reported 10. Barnes PJ, Shapiro SD, Pauwels RA. Chronic obstructive pulmonary
to have entered Phase II trails is the compound GW- disease: Molecular and cellular mechanisms. Eur Respir J 22: 67288,
766994 from GlaxoSmithKline, with little apparent ecacy 2003.
11. Yoshimura T, Matsushima K, Tanaka S et al. Purication of a human
in an allergic rhinitis study [157]. This follows on from the
monocyte-derived neutrophil chemotactic factor that has peptide
recent reports of the ecacy of the GlaxoSmithKline com- sequence similarity to other host defense cytokines. Proc Natl Acad Sci
pound GW-701897B in reducing vagally mediated bron- USA 84: 923337, 1987.
choconstriction in antigen-challenged guinea pigs [158]. In 12. Schrder J-M, Mrowietz U, Christophers E. Purication and partial
an alternative approach, neutralization of CCL11 has been biological characterization of a human lymphocyte-derived peptide
achieved by the generation of a human single-chain frag- with potent neutrophil-stimulating activity. J Immunol 140: 353440,
ment variable antibody named CAT-213, isolated by phage 1988.
display methodologies [159]. CAT-213 is selective for 13. Walz A, Peveri P, Aschauer H, Baggiolini M. Purication and amino
CCL11 over closely related chemokines, such as CCL2, with acid sequencing of NAF, a novel neutrophil-activating factor produced
sub-nanomolar IC50 values. In a Phase I single-dose clini- by monocytes. Biochem Biophys Res Commun 149: 75561, 1987.
14. Beaubien BC, Collins PD, Jose PJ et al. A novel neutrophil chem-
cal study, CAT-213 was reported to be well tolerated, and
oattractant generated during an inammatory reaction in the rabbit
long-lived, remaining in an active form in serum for over 2 peritoneal cavity in vivo: Purication, partial amino acid sequence
months, following intravenous administration [160]. In sub- and structural relationship to interleukin 8. Biochem J 271: 797801,
sequent Phase II studies, administration of CAT-213 prior 1990.
to the allergen challenge of rhinitic subjects was observed to 15. Jose PJ, Collins PD, Perkins JA et al. Identication of a second neu-
reduce the mucosal eosinophil inltration [161, 162]. trophil chemoattractant cytokine generated during an inammatory
reaction in the rabbit peritoneal cavity in vivo: Purication, partial
amino acid sequence and structural relationship to melanoma growth
stimulatory activity. Biochem J 278: 49397, 1991.
CONCLUSION 16. Collins PD, Jose PJ, Williams TJ. The sequential generation of
neutrophil chemoattractant proteins in acute inammation in the rab-
bit in vivo. Relationship between C5a and proteins with the character-
A combination of clinical observations, in vitro cell biol- istics of IL-8/neutrophil-activating protein 1. J Immunol 146: 67784,
ogy and in vivo animal modeling has delineated potentially 1991.
important mechanisms underlying lung inammation in 17. Bazzoni F, Cassatella MA, Rossi F et al. Phagocytosing neutrophils
COPD and asthma. These extensive studies support the produce and release high amounts of the neutrophil-activating peptide
hypothesis that chemokines have a fundamental role in reg- 1/interleukin 8. J Exp Med 173: 77174, 1991.
ulating leukocyte tracking in inammatory disease. The 18. Au B-T, Williams TJ, Collins PD. Zymosan-induced interleukin-8
release from human neutrophils involves activation via the CD11b/
chemokine receptors represent novel targets for treatment
CD18 receptor and endogenous platelet activating factor as an auto-
and therapeutic compounds have been described with e- crine modulator. J Immunol 152: 541119, 1994.
cacy in vitro and in vivo. Clinical trials will ultimately pro- 19. Au B-T, Teixeira MM, Collins PD, Williams TJ. Eect of PDE4
vide the crucial information that will dene the chemokine inhibitors on zymosan-induced IL-8 release from human neutrophils:
receptors that can be blocked to provide future therapy in Synergism with prostanoids and salbutamol. Br J Pharmacol 123:
these diseases. 126066, 1998.
20. Ivey CL, Williams FM, Collins PD et al. Neutrophil chemoattractants
generated in two phases during reperfusion of ischemic myocardium in
References the rabbit: Evidence for a role for C5a and interleukin-8. J Clin Invest
95: 272028, 1995.
1. Rot A, Von Andrian UH. Chemokines in innate and adaptive host 21. Sekido N, Mukaida N, Harada A et al. Prevention of lung reperfusion
defense: Basic chemokinese grammar for immune cells. Annu Rev injury in rabbits by a monoclonal antibody against interleukin-8.
Immunol 22: 891928, 2004. Nature 365: 65457, 1993.
2 Strieter RM, Belperio JA, Keane MP. CXC chemokines in angiogenesis 22. Holmes WLJ, Kuang WJ, Rice G, Wood W. Structure and functional
related to pulmonary brosis. Chest 122: 298S301S, 2002. expression of a human interleukin08 receptor. Science 253: 1278, 1991.
3 Kelner GS, Kennedy J, Bacon KB et al. Lymphotactin: A cytokine that 23. Murphy P, Tiany H. Cloning of complimentary DNA encoding a
represents a new class of chemokine. Science 266: 139599, 1994. functional human IL-8 receptor. Science 253: 1280, 1991.

321
Asthma and COPD: Basic Mechanisms and Clinical Management

24. Proost P, De Wolf-Peeters C, Conings R et al. Identication of a novel 45. Maeno T, Houghton AM, Quintero PA et al. CD8 T Cells are
granulocyte chemotactic protein (GCP-2) from human tumor cells. required for inammation and destruction in cigarette smoke-induced
J Immunol 150: 100010, 1993. emphysema in mice. J Immunol 178: 809096, 2007.
25 Ahuja SK, Lee JC, Murphy PM. CXC chemokines bind to unique sets 46. Koch A, Gaczkowski M, Sturton G et al. Modication of surface anti-
of selectivity determinants that can function independently and are gens in blood CD8 T-lymphocytes in COPD: Eects of smoking.
broadly distributed on multiple domains of human interleukin-8 recep- Eur Respir J 29: 4250, 2007.
tor B: Determinants of high anity binding and receptor activation are 47. Saetta M, Mariani M, Panina-Bordignon P et al. Increased expres-
distinct. J Biol Chem 271: 22532, 1996. sion of the chemokine receptor CXCR3 and its ligand CXCL10 in
26. Soler N, Ewig S, Torres A et al. Airway inammation and bronchial peripheral airways of smokers with chronic obstructive pulmonary dis-
microbial patterns in patients with stable chronic obstructive pulmo- ease. Am J Respir Crit Care Med 165: 14049, 2002.
nary disease. Eur Respir J 14: 101522, 1999. 48. Freeman CM, Curtis JL, Chensue SW. CC chemokine receptor 5 and
27. Pesci A, Balbi B, Majori M et al. Inammatory cells and mediators CXC chemokine receptor 6 expression by lung CD8 cells correlates
in bronchial lavage of patients with chronic obstructive pulmonary with chronic obstructive pulmonary disease severity. Am J Pathol 171:
disease. Eur Respir J 12: 38086, 1998. 76776, 2007.
28. Nocker RE, Schoonbrood DF, van de Graaf EA et al. Interleukin-8 in 49. Goodarzi K, Goodarzi M, Tager AM et al. Leukotriene B4 and BLT1
airway inammation in patients with asthma and chronic obstructive control cytotoxic eector T cell recruitment to inamed tissues.
pulmonary disease. Int Arch Allergy Immunol 109: 18391, 1996. Nat Immunol 4: 96573, 2003.
29. Keatings VM, Collins PD, Scott DM, Barnes PJ. Dierences in 50. Kay AB, Phipps S, Robinson DS. A role for eosinophils in airway
interleukin-8 and tumor necrosis factor-alpha in induced sputum from remodelling in asthma. Trends Immunol 25: 47782, 2004.
patients with chronic obstructive pulmonary disease or asthma. Am J 51. Humbles AA, Lloyd CM, McMillan SJ et al. A critical role for eosi-
Respir Crit Care Med 153: 53034, 1996. nophils in allergic airways remodeling. Science 305: 177679, 2004.
30. Keman S, Willemse B, Tollerud DJ et al. Blood interleukin-8 produc- 52. Fulkerson PC, Fischetti CA, McBride ML et al. A central regulatory
tion is increased in chemical workers with bronchitic symptoms. Am J role for eosinophils and the eotaxin/CCR3 axis in chronic experi-
Ind Med 32: 67073, 1997. mental allergic airway inammation. Proc Natl Acad Sci USA 103:
31. Yamamoto C, Yoneda T, Yoshikawa M et al. Airway inammation in 1641823, 2006.
COPD assessed by sputum levels of interleukin-8. Chest 112: 50510, 53. Lee JJ, Dimina D, Macias MP et al. Dening a link with asthma in
1997. mice congenitally decient in eosinophils. Science 305: 177376,
32. Woolhouse IS, Bayley DL, Stockley RA. Sputum chemotactic activ- 2004.
ity in chronic obstructive pulmonary disease: Eect of alpha(1)- 54. Wills-Karp M, Karp CL. Biomedicine. Eosinophils in asthma:
antitrypsin deciency and the role of leukotriene B(4) and interleukin Remodeling a tangled tale. Science 305: 172629, 2004.
8. Thorax 57: 70914, 2002. 55. Jose PJ, Griths-Johnson DA, Collins PD et al. Eotaxin: A potent
33. Aaron SD, Angel JB, Lunau M et al. Granulocyte inammatory mark- eosinophil chemoattractant cytokine detected in a guinea-pig model of
ers and airway infection during acute exacerbation of chronic obstruc- allergic airways inammation. J Exp Med 179: 88187, 1994.
tive pulmonary disease. Am J Respir Crit Care Med 163: 34955, 2001. 56. Ying S, Robinson DS, Meng Q et al. Enhanced expression of eotaxin
34. Sethi S, Muscarella K, Evans N et al. Airway inammation and etiol- and CCR3 mRNA and protein in atopic asthma. Association with air-
ogy of acute exacerbations of chronic bronchitis. Chest 118: 155765, way hyperresponsiveness and predominant co-localization of eotaxin
2000. mRNA to bronchial epithelial and endothelial cells. Eur J Immunol 27:
35. Traves SL, Culpitt SV, Russell RE et al. Increased levels of the chem- 350716, 1997.
okines GROalpha and MCP-1 in sputum samples from patients with 57. Komiya A, Nagase H, Yamada H et al. Concerted expression of
COPD. Thorax 57: 59095, 2002. eotaxin-1, eotaxin-2, and eotaxin-3 in human bronchial epithelial cells.
36. Tanino M, Betsuyaku T, Takeyabu K et al. Increased levels of inter- Cell Immunol 225: 91100, 2003.
leukin-8 in BAL uid from smokers susceptible to pulmonary emphy- 58. Kobayashi I, Yamamoto S, Nishi N et al. Regulatory mechanisms of
sema. Thorax 57: 40511, 2002. Th2 cytokine-induced eotaxin-3 production in bronchial epithelial
37. Schulz C, Kratzel K, Wolf K et al. Activation of bronchial epithelial cells: Possible role of interleukin 4 receptor and nuclear factor-kappaB.
cells in smokers without airway obstruction and patients with COPD. Ann Allergy Asthma Immunol 93: 39097, 2004.
Chest 125: 170613, 2004. 59. Ying S, Meng Q , Zeibecoglou K et al. Eosinophil chemotactic chem-
38. Turato G, Zuin R, Miniati M et al. Airway inammation in severe okines (eotaxin, eotaxin-2, RANTES, monocyte chemoattractant
chronic obstructive pulmonary disease: Relationship with lung function protein-3 (MCP-3), and MCP-4), and C-C chemokine receptor 3
and radiologic emphysema. Am J Respir Crit Care Med 166: 10510, 2002. expression in bronchial biopsies from atopic and nonatopic (Intrinsic)
39. Capelli A, Di Stefano A, Gnemmi I et al. Increased MCP-1 and asthmatics. J Immunol 163: 632129, 1999.
MIP-1beta in bronchoalveolar lavage uid of chronic bronchitics. 60. Zeibecoglou K, Macfarlane AJ, Ying S et al. Increases in eotaxin-
Eur Respir J 14: 16065, 1999. positive cells in induced sputum from atopic asthmatic subjects after
40. Rollins BJ, Walz A, Baggiolini M. Recombinant human MCP-1/JE inhalational allergen challenge. Allergy 54: 73035, 1999.
induces chemotaxis, calcium ux, and the respiratory burst in human 61. Abonyo BO, Alexander MS, Heiman AS. Autoregulation of CCL26
monocytes. Blood 78: 111216, 1991. synthesis and secretion in A549 cells: A possible mechanism by which
41. Pignatti P, Moscato G, Casarini S et al. Downmodulation of CXCL8/ alveolar epithelial cells modulate airway inammation. Am J Physiol
IL-8 receptors on neutrophils after recruitment in the airways. J Allergy Lung Cell Mol Physiol 289: L478L488, 2005.
Clin Immunol 115: 8894, 2005. 62. Miyamasu M, Yamaguchi M, Nakajima T et al. Th1-derived cytokine
42. Traves SL, Smith SJ, Barnes PJ, Donnelly LE. Specic CXC but not IFN-gamma is a potent inhibitor of eotaxin synthesis in vitro.
CC chemokines cause elevated monocyte migration in COPD: A role Int Immunol 11: 10014, 1999.
for CXCR2. J Leukoc Biol 76: 44150, 2004. 63. Teran LM, Mochizuki M, Bartels J et al. Th1- and Th2-type
43. Saetta M, Di Stefano A, Turato G et al. CD8 T-lymphocytes in cytokines regulate the expression and production of eotaxin and
peripheral airways of smokers with chronic obstructive pulmonary dis- RANTES by human lung broblasts. Am J Respir Cell Mol Biol 20:
ease. Am J Respir Crit Care Med 157: 82226, 1998. 77786, 1999.
44. Wang Z, Zheng T, Zhu Z et al. Interferon gamma induction of pulmo- 64. Stellato C, Matsukura S, Fal A et al. Dierential regulation of epithelial-
nary emphysema in the adult murine lung. J Exp Med 192: 1587600, derived C-C chemokine expression by IL-4 and the glucocorticoid
2000. budesonide. J Immunol 163: 562432, 1999.

322
Chemokines 26
65. Ghaar O, Hamid Q, Renzi PM et al. Constitutive and cytokine- airways disease: Analysis in a guinea pig model in vivo. J Exp Med
stimulated expression of eotaxin by human airway smooth muscle cells. 186: 60112, 1997.
Am J Respir Crit Care Med 159: 193342, 1999. 85. Yamada H, Yamaguchi M, Yamamoto K et al. Eotaxin in induced
66. Fukagawa K, Nakajima T, Saito H et al. IL-4 induces eotaxin produc- sputum of asthmatics: Relationship with eosinophils and eosinophil
tion in corneal keratocytes but not in epithelial cells. Int Arch Allergy cationic protein in sputum. Allergy 55: 39297, 2000.
Immunol 121: 14450, 2000. 86. Yamada H, Yamaguchi M, Yamamoto K et al. Eotaxin in induced
67. MacLean JA, Ownbey R, Luster AD. T cell-dependent regulation of sputum of asthmatics: Relationship with eosinophils and eosinophil
eotaxin in antigen-induced pulmonary eosinophilia. J Exp Med 184: cationic proteins in sputum. Allergy 55: 39297, 2000.
146169, 1996. 87. Collins PD, Marleau S, Griths-Johnson DA et al. Co-operation
68. Ponath PD, Qin S, Ringler DJ et al. Cloning of the human eosinophil between interleukin-5 and the chemokine eotaxin to induce eosi-
chemoattractant, eotaxin. Expression, receptor binding and functional nophil accumulation in vivo. J Exp Med 182: 116974, 1995.
properties suggest a mechanism for the selective recruitment of eosi- 88. Palframan RT, Collins PD, Severs NJ et al. Mechanisms of acute eosi-
nophils. J Clin Invest 97: 60412, 1996. nophil mobilization from the bone marrow stimulated by interleukin
69. Rothenberg ME, Luster AD, Leder P. Murine eotaxin: An eosinophil 5: The role of specic adhesion molecules and phosphatidylinositol
chemoattractant inducible in endothelial cells and in interleukin 3-kinase. J Exp Med 188: 162132, 1998.
4-induced tumor suppression. Proc Natl Acad Sci USA 92: 896064, 89. Palframan RT, Collins PD, Williams TJ, Rankin SM. Eotaxin
1995. induces a rapid release of eosinophils and their progenitors from the
70. Williams CMM, Newton DJ, Wilson SA et al. Conserved bone marrow. Blood 91: 224048, 1998.
structure and tissue expression of rat eotaxin. Immunogenetics 47: 17880, 90. Sabroe I, Conroy DM, Gerard NP et al. Cloning and characterisation
1998. of the guinea pig eosinophil eotaxin receptor, CCR3: Blockade using
71. Zhang L, Soares MP, Guan Y et al. Functional expression and char- a monoclonal antibody in vivo. J Immunol 161: 613947, 1998.
acterization of macaque C-C chemokine receptor 3 (CCR3) and 91. Campbell EM, Kunkel SL, Strieter RM, Lukacs NW. Temporal role
generation of potent antagonistic anti-macaque CCR3 monoclonal of chemokines in a murine model of cockroach allergen-induced
antibodies. J Biol Chem 277: 33799810, 2002. airway hyperreactivity and eosinophilia. J Immunol 161: 704753,
72. Ponath PD, Qin S, Post TW et al. Molecular cloning and characteriza- 1998.
tion of a human eotaxin receptor expressed selectively on eosinophils. J 92. Rothenberg ME, MacLean JA, Pearlman E et al. Targeted disruption
Exp Med 183: 24372448, 1996. of the chemokine eotaxin partially reduces antigen-induced tissue
73. Sallusto F, Mackay CR, Lanzavecchia A. Selective expression of the eosinophilia. J Exp Med 185: 78590, 1997.
eotaxin receptor CCR3 by human T helper 2 cells. Science 277: 20057, 93. Humbles AA, Lu B, Friend DS et al. The murine CCR3 receptor reg-
1997. ulates both the role of eosinophils and mast cells in allergen-induced
74. Uguccioni M, Mackay CR, Ochensberger B et al. High expression airway inammation and hyperresponsiveness. Proc Natl Acad Sci USA
of the chemokine receptor CCR3 in human blood basophils. Role in 99: 147984, 2002.
activation by eotaxin, MCP-4, and other chemokines. J Clin Invest 94. Ma W, Bryce PJ, Humbles AA et al. CCR3 is essential for skin eosi-
100: 113743, 1997. nophilia and airway hyperresponsiveness in a murine model of allergic
75. Romagnani P, De Paulis A, Beltrame C et al. Tryptasechymase skin inammation. J Clin Invest 109: 62128, 2002.
double-positive human mast cells express the eotaxin receptor CCR3 95. Mattes J, Yang M, Mahalingam S et al. Intrinsic defect in T cell
and are attracted by CCR3-binding chemokines. Am J Pathol 155: production of interleukin (IL)-13 in the absence of both IL-5 and
1195204, 1999. eotaxin precludes the development of eosinophilia and airways hyper-
76. Ochi H, Hirani WM, Yuan Q et al. T helper cell type 2 cytokine- reactivity in experimental asthma. J Exp Med 195: 143344, 2002.
mediated comitogenic responses and CCR3 expression during dier- 96. Power CA, Church DJ, Meyer A et al. Cloning and characterization
entiation of human mast cells in vitro. J Exp Med 190: 26780, 1999. of a specic receptor for the novel CC chemokine MIP-3alpha from
77. Kitaura M, Suzuki N, Imai T et al. Molecular cloning of a novel human lung dendritic cells. J Exp Med 186: 82535, 1997.
CC chemokine (Eotaxin-3) that is a functional ligand of CC chemok- 97. Greaves DR, Wang W, Dairaghi DJ et al. CCR6, a CC chemokine
ine receptor 3. J Biol Chem 274: 2797580, 1999. receptor that interacts with macrophage inammatory protein 3alpha
78. Patel VP, Kreider BL, Li Y et al. Molecular and functional characteri- and is highly expressed in human dendritic cells. J Exp Med 186:
zation of two novel human C-C chemokines as inhibitors of two dis- 83744, 1997.
tinct classes of myeloid progenitors. J Exp Med 185: 116372, 1997. 98. Hromas R, Gray PW, Chantry D et al. Cloning and characterization
79. White JR, Imburgia C, Dul E et al. Cloning and functional charac- of exodus, a novel beta-chemokine. Blood 89: 331522, 1997.
terization of a novel human CC chemokine that binds to the CCR3 99. Dieu MC, Vanbervliet B, Vicari A et al. Selective recruitment of
receptor and activates human eosinophils. J Leukoc Biol 62: 66775, immature and mature dendritic cells by distinct chemokines expressed
1997. in dierent anatomic sites. J Exp Med 188: 37386, 1998.
80. Forssmann U, Uguccioni M, Loetscher P et al. Eotaxin-2, a novel CC 100. Sozzani S, Allavena P, DAmico G et al. Dierential regulation of
chemokine that is selective for the chemokine receptor CCR3, and acts chemokine receptors during dendritic cell maturation: A model for
like eotaxin on human eosinophil and basophil leukocytes. J Exp Med their tracking properties. J Immunol 161: 108386, 1998.
185: 217176, 1997. 101. Yoshida R, Imai T, Hieshima K et al. Molecular cloning of a novel
81. Shinkai A, Yoshisue H, Koike M et al. A novel human CC chemokine, human CC chemokine EBI1-ligand chemokine that is a specic
eotaxin-3, which is expressed in IL-4- stimulated vascular endothe- functional ligand for EBI1, CCR7. J Biol Chem: 138039, 1997.
lial cells, exhibits potent activity toward eosinophils. J Immunol 163: 102. Tang HL, Cyster JG. Chemokine up-regulation and activated T cell
160210, 1999. attraction by maturing dendritic cells. Science 284: 81922, 1999.
82. Rothenberg ME, Luster AD, Lilly CM et al. Constitutive and 103. Hammad H, Lambrecht BN, Pochard P et al. Monocyte-derived
allergen-induced expression of eotaxin mRNA in the guinea pig lung. dendritic cells induce a house dust mite-specic Th2 allergic inam-
J Exp Med 181: 121116, 1995. mation in the lung of humanized SCID mice: Involvement of CCR7.
83. Gonzalo J-A, Jia G-Q, Aguirre V et al. Mouse eotaxin expression J Immunol 169: 152434, 2002.
parallels eosinophil accumulation during lung allergic inammation 104. Fainaru O, Shseyov D, Hantisteanu S, Groner Y. Accelerated chem-
but it is not restricted to a Th2-type response. Immunity 4: 114, 1996. okine receptor 7-mediated dendritic cell migration in Runx3 knock-
84. Humbles AA, Conroy DM, Marleau S et al. Kinetics of eotaxin out mice and the spontaneous development of asthma-like disease.
generation and its relationship to eosinophil accumulation in allergic Proc Natl Acad Sci USA 102: 10598603, 2005.

323
Asthma and COPD: Basic Mechanisms and Clinical Management

105. Li L, Xia Y, Nguyen A et al. Eects of Th2 cytokines on chemokine 126. Ling EM, Smith T, Nguyen XD et al. Relation of CD4  CD25
expression in the lung: IL-13 potently induces eotaxin expression by regulatory T-cell suppression of allergen-driven T-cell activation to
airway epithelial cells. J Immunol 162: 247787, 1999. atopic status and expression of allergic disease. Lancet 363: 60815,
106. Wenzel SE, Trudeau JB, Barnes S et al. TGF-beta and IL-13 syner- 2004.
gistically increase eotaxin-1 production in human airway broblasts. 127. Stassen M, Jonuleit H, Muller C et al. Dierential regulatory capacity
J Immunol JID 2985117R 169: 461319, 2002. of CD25 T regulatory cells and preactivated CD25 T regulatory
107. Zuyderduyn S, Hiemstra PS, Rabe KF. TGF-beta dierentially regu- cells on development, functional activation, and proliferation of Th2
lates TH2 cytokine-induced eotaxin and eotaxin-3 release by human cells. J Immunol 173: 26774, 2004.
airway smooth muscle cells. J Allergy Clin Immunol 114: 79198, 2004. 128. Iellem A, Mariani M, Lang R et al. Unique chemotactic response pro-
108. Bleul CC, Wu L, Hoxie JA et al. The HIV coreceptors CXCR4 and le and specic expression of chemokine receptors CCR4 and CCR8
CCR5 are dierentially expressed and regulated on human T lym- by CD4()CD25() regulatory T cells. J Exp Med 194: 84753,
phocytes. Proc Natl Acad Sci USA 94: 192530, 1997. 2001.
109. Jourdan P, Abbal C, Noraz N et al. IL-4 induces functional cell-surface 129. Xanthou G, Duchesnes CE, Williams TJ, Pease JE. CCR3 functional
expression of CXCR4 on human T cells. J Immunol 160: 415357, responses are regulated by both CXCR3 and its ligands CXCL9,
1998. CXCL10 and CXCL11. Eur J Immunol 33: 224150, 2003.
110. Gonzalo JA, Lloyd CM, Peled A et al. Critical involvement of the 130. Fulkerson PC, Zimmermann N, Brandt EB et al. Negative regulation
chemotactic axis CXCR4/stromal cell-derived factor-1 alpha in the of eosinophil recruitment to the lung by the chemokine monokine
inammatory component of allergic airway disease. J Immunol 165: induced by IFN-gamma (Mig, CXCL9). Proc Natl Acad Sci USA 101:
499508, 2000. 198792, 2004.
111. Lukacs NW, Berlin A, Schols D et al. AMD3100, a CxCR4 antago- 131. Falcone FH, Haas H, Gibbs BF. The human basophil: A new appre-
nist, attenuates allergic lung inammation and airway hyperreactivity. ciation of its role in immune responses. Blood 96: 402838, 2000.
Am J Pathol 160: 135360, 2002. 132. Gibbs BF, Haas H, Falcone FH et al. Puried human peripheral blood
112. Bonecchi R, Bianchi G, Bordignon PP et al. Dierential expression basophils release interleukin-13 and preformed interleukin-4 following
of chemokine receptors and chemotactic responsiveness of type 1 T immunological activation. Eur J Immunol 26: 249398, 1996.
helper cells (Th1s) and Th2s. J Exp Med 187: 12934, 1998. 133. Yamada H, Hirai K, Miyamasu M et al. Eotaxin is a potent chemo-
113. Sallusto F, Lenig D, Mackay CR, Lanzavecchia A. Flexible programs taxin for human basophils. Biochem Biophys Res Commun 231: 36568,
of chemokine receptor expression on human polarised T helper 1 and 1997.
2 lymphocytes. J Exp Med 187: 87583, 1998. 134. Heinemann A, Hartnell A, Stubbs VE et al. Basophil responses to
114. Morgan AJ, Symon FA, Berry MA et al. IL-4-expressing bronchoal- chemokines are regulated by both sequential and cooperative receptor
veolar T cells from asthmatic and healthy subjects preferentially express signaling. J Immunol 165: 722433, 2000.
CCR 3 and CCR 4. J Allergy Clin Immunol 116: 594600, 2005. 135. Hartnell A, Heinemann A, Conroy DM et al. Identication of selec-
115. Panina-Bordignon P, Papi A, Mariani M et al. The C-C chemok- tive basophil chemoattractants in human nasal polyps as insulin-like
ine receptors CCR4 and CCR8 identify airway T cells of allergen- growth factor-1 and insulin-like growth factor-2. J Immunol 173:
challenged atopic asthmatics. J Clin Invest 107: 135764, 2001. 644857, 2004.
116. Banwell ME, Robinson DS, Lloyd CM. Adenoid-derived TH2 cells 136. Weller CL, Collington SJ, Brown JK et al. Leukotriene B4, an activa-
reactive to allergen and recall antigen express CC chemokine tion product of mast cells, is a chemoattractant for their progenitors.
receptor 4. J Allergy Clin Immunol 112: 115561, 2003. J Exp Med 201: 196171, 2005.
117. Pilette C, Francis JN, Till SJ, Durham SR. CCR4 ligands are up- 137. Oliveira SH, Lukacs NW. Stem cell factor and IgE-stimulated
regulated in the airways of atopic asthmatics after segmental allergen murine mast cells produce chemokines (CCL2, CCL17, CCL22) and
challenge. Eur Respir J 23: 87684, 2004. express chemokine receptors. Inamm Res 50: 16874, 2001.
118. Bochner BS, Hudson SA, Xiao HQ, Liu MC. Release of both 138. Juremalm M, Hjertson M, Olsson N et al. The chemokine receptor
CCR4-active and CXCR3-active chemokines during human allergic CXCR4 is expressed within the mast cell lineage and its ligand stromal
pulmonary late-phase reactions. J Allergy Clin Immunol 112: 93034, cell-derived factor-1alpha acts as a mast cell chemotaxin. Eur J
2003. Immunol 30: 361422, 2000.
119. Lloyd CM, Delaney T, Nguyen T et al. CC Chemokine recep- 139. Brightling CE, Bradding P, Symon FA et al. Mast-cell inltration
tor (CCR)3/eotaxin is followed by CCR4/monocyte-derived of airway smooth muscle in asthma. N Engl J Med 346: 1699705,
chemokine in mediating pulmonary T helper lymphocyte type 2 2002.
recruitment after serial antigen challenge in vivo. J Exp Med 191: 140. Brightling CE, Ammit AJ, Kaur D et al. The CXCL10/CXCR3 axis
26573, 2000. mediates human lung mast cell migration to asthmatic airway smooth
120. Chvatchko Y, Hoogewerf AJ, Meyer A et al. A key role for CC chem- muscle. Am J Respir Crit Care Med 171: 11038, 2005.
okine receptor 4 in lipopolysaccharide-induced endotoxic shock. J 141. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB. The
Exp Med 191: 175564, 2000. G-protein-coupled receptors in the human genome form ve main
121. Chung CD, Kuo F, Kumer J et al. CCR8 is not essential for the devel- families. Phylogenetic analysis, paralogon groups, and ngerprints.
opment of inammation in a mouse model of allergic airway disease. Mol Pharmacol 63: 125672, 2003.
J Immunol 170: 58187, 2003. 142. White JR, Lee JM, Young PR et al. Identication of a potent, selective
122. Goya I, Villares R, Zaballos A et al. Absence of CCR8 does not non-peptide CXCR2 antagonist that inhibits interleukin-8-induced
impair the response to ovalbumin-induced allergic airway disease. neutrophil migration. J Biol Chem 273: 1009598, 1998.
J Immunol 170: 213846, 2003. 143. Auten RL, Richardson RM, White JR et al. Nonpeptide CXCR2
123. Conroy DM, Jopling LA, Lloyd CM et al. CCR4 blockade does not antagonist prevents neutrophil accumulation in hyperoxia-exposed
inhibit allergic airways inammation. J Leukoc Biol 74: 55863, 2003. newborn rats. J Pharmacol Exp Ther 299: 9095, 2001.
124. Bishop B, Lloyd CM. CC chemokine ligand 1 promotes recruitment 144. Podolin PL, Bolognese BJ, Foley JJ et al. A potent and selective non-
of eosinophils but not Th2 cells during the development of allergic peptide antagonist of CXCR2 inhibits acute and chronic models of
airways disease. J Immunol 170: 481017, 2003. arthritis in the rabbit. J Immunol 169: 643544, 2002.
125. Bellinghausen I, Klostermann B, Knop J, Saloga J. Human 145. Validation of dierent FLIPR calcium no wash dyes for performance
CD4 CD25 T cells derived from the majority of atopic donors through Millipores GPCRProlerTM service. Oral presentation
are able to suppress TH1 and TH2 cytokine production. J Allergy Clin SBS 13th Annual Conference & Exhibition, Montreal, Canada April
Immunol 111: 86268, 2003. 15th19th, 2007.

324
Chemokines 26
146. Sabroe I, Peck MJ, Jan Van Keulen B et al. A small molecule antago- 155. Warrior U, McKeegan EM, Rottinghaus SM et al. Identication and
nist of the chemokine receptors CCR1 and CCR3 : Potent inhibi- characterization of novel antagonists of the CCR3 receptor. J Biomol
tion of Eosinophil function and CCR3-mediated HIV-1 entry. J Biol Screen 8: 32431, 2003.
Chem 275: 2598592, 2000. 156. Suzuki K, Morokata T, Morihira K et al. In vitro and in vivo charac-
147. Sabroe I, Hartnell A, Jopling LA et al. Dierential regulation of eosi- terization of a novel CCR3 antagonist, YM-344031. Biochem Biophys
nophil chemokine signaling via CCR3 and non-CCR3 pathways. Res Commun 339: 121723, 2006.
J Immunol 162: 294655, 1999. 157. Murdoch RD. The Challenges of Drug Discovery & Development.
148. Phillips R, Stubbs VELS, Henson MR, Williams TJ, Pease JE, Sabroe Oral presentation, 4th James Black Conference, University of
I. Variations in eosinophil chemokine responses: An investigation of Hertfordshire, September 13, 2006.
CCR1 and CCR3 function, expression in atopy, and identication of 158. Fryer AD, Stein LH, Nie Z et al. Neuronal eotaxin and the eects
a functional CCR1 promoter. J Immunol 170: 6190201, 2003. of CCR3 antagonist on airway hyperreactivity and M2 receptor dys-
149. de Mendonca FL, da Fonseca PC, Phillips RM et al. Site-directed function. J Clin Invest 116: 22836, 2006.
mutagenesis of CC chemokine receptor 1 reveals the mechanism of 159. Main S, Handy R, Wilton J et al. A potent human anti-eotaxin1 anti-
action of UCB 35625, a small molecule chemokine receptor antago- body, CAT-213: Isolation by phage display and in vitro and in vivo
nist. J Biol Chem 280: 480816, 2005. ecacy. J Pharmacol Exp Ther 319: 1395404, 2006.
150. Pease JE, Wang J, Ponath PD, Murphy PM. The N-terminal extra- 160. Brennan N, Case N, Meyers T, Amakye D, Doughty J, Forward JA,
cellular segments of the chemokine receptors CCR1 and CCR3 are Varley P, Powell J, Glover DR. Pharmacokinetics of CAT-123, a
determinants for MIP-1 and eotaxin binding, respectively, but a human anti-eotaxin-1 monoclonal antibody, following single intra-
second domain is essential for receptor activation. J Biol Chem 273: venous administration to healthy volunteers. Br J Clin Pharmacol 53:
1997276, 1998. 441P, 2002.
151. Morokata T, Suzuki K, Masunaga Y et al. A novel, selective, and orally 161. Pereira S, Clark T, Darby Y, Salib R, Salagean M, Hewitt L, Powell J,
available antagonist for CC chemokine receptor 3. J Pharmacol Exp Howarth P, Scadding G. Eects of anti-eotaxin monoclonal antibody
Ther 317: 24450, 2006. CAT-213 on allergen-induced rhinitis. J Allergy Clin Immunol 111:
152. Anderskewitz R, Bauer R, Bodenbach G et al. Pyrrolidinohydroquin S268, 2003.
azolines a novel class of CCR3 modulators. Bioorg Med Chem Lett 162. Salib R, Salagean M, Lau L, Di Giovanna I, Brennan N, Scadding G,
15: 66973, 2005. Howarth P. The anti-inammatory response of anti-eotaxin mono-
153. De Lucca GV, Kim UT, Vargo BJ et al. Discovery of CC chemokine clonal antibody CAT-213 on nasal allergen-induced cell inltration
receptor-3 (CCR3) antagonists with picomolar potency. J Med Chem and activation. J Allergy Clin Immunol 111: S347, 2003.
48: 2194211, 2005.
154. Naya A, Kobayashi K, Ishikawa M et al. Structureactivity relation-
ships of 2-(benzothiazolylthio)acetamide class of CCR3 selective
antagonist. Chem Pharm Bull (Tokyo) 51: 697701, 2003.

325
Cytokines
27 CHAPTER

INTRODUCTION context of airways disease, nevertheless, a broad


functional classication remains helpful (Table
Kian Fan Chung
27.1). The major classes of cytokines include: pro- Department of Thoracic Medicine,
Cytokines are signaling proteins, usually less and anti-inammatory cytokines, cytokines of National Heart and Lung Institute,
than 80 kDa in size, which regulate a wide range neutrophil and eosinophil recruitment and acti- Imperial College, London, UK
of biological functions including innate and vation, cytokines derived from T-helper (Th) and
acquired immunity, hematopoiesis, inamma- T-regulatory (Tregs) cells, and cytokines of T-cell
tion and repair, and proliferation through mostly recruitment and growth factors.
extracellular signaling. They are secreted by many The receptors for many cytokines have been
cell types at local high concentrations and are grouped into superfamilies based on the pres-
involved in cell-to-cell interactions, have an eect ence of common homology regions (Table 27.2).
on closely adjacent cells, and therefore func- Cytokine receptors are linked to multiple signal-
tion in a predominantly paracrine fashion. They ing pathways in the cytoplasm and nucleus, lead-
may also act at a distance by secretion of soluble ing to transcriptional and post-transcriptional
products into the circulation (endocrine or sys- activation of many factors including cytokines.
temic eect) and may have eects on the cell of Some cytokines may stimulate their own produc-
origin itself (autocrine eect). The rst group of tion in an autocrine manner while others stimu-
cytokines described was immune-response factors, late the synthesis of dierent cytokines that have
labeled as lymphokines that were soluble products a stimulatory feedback eect on the rst cytokine.
released from activated lymphocytes in response Cytokines may also induce the expression of
to specic or polyclonal antigen. Other groups of receptors which may change the responsiveness
cytokines were named according to their actions of both source and target cells. This complexity of
such as the colony stimulating factors (e.g., granu- cytokine interactions is referred to as the cytokine
locyte colony stimulating factor, G-CSF or gran- network, the overall eect of which will depend
ulocyte-macrophage colony stimulating factor, on the type of cytokines and cytokine receptors
GM-CSF), the chemoattractants now grouped involved and the general milieu and context of the
under chemokines with a special nomenclature response.
(e.g., CCL11/eotaxin or CXCL1/MIP-1), or The potential contribution of cytokines
the growth factors that mediate proliferation, dif- to disease has been explored in studies using
ferentiation, and survival of cells (e.g. transform- cytokines as agonists (mainly in cell-based stud-
ing growth factor-, TGF-). However, the eect ies), by blocking the eects of specic cytokines
of a particular cytokine is not restricted to only mainly in animal models, by conditional overex-
one set of biological functions but often has more pression and deletion of cytokines in transgenic
than one function extending beyond the function mice, and by genetic studies. In humans, impli-
that may be implied in its name. The wide pleiot- cation for particular cytokines in disease has
ropy and element of redundancy in the cytokine rested on the ability to detect their expression
family, with each cytokine having many overlap- in the tissue of interest, and rarely on the eect
ping functions, and with each function potentially of selective inhibition of specic cytokines. The
mediated by more than one cytokine make the sources and eects of cytokines are summarized
classication of cytokines a dicult task. In the in Table 27.2. In this chapter, the cytokines will

327
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 27.1 Classification of cytokines and cytokine receptors.

Cytokines

Proinflammatory cytokines IL-1/, TNF-/, IL-6, IL-11, IL-18, IFN-

Anti-inflammatory cytokines IL-10, TGF, IL-1ra

Cytokines of neutrophil recruitment and activation CXCL8/IL-8, IL-1/, TNF-/, G-CSF, IL-17A, IL-17F

Cytokines of eosinophil recruitment and activation IL-2, IL-3, IL-4, IL-5, GM-CSF, CCL5/RANTES, CCL11/eotaxin, CCL7/MCP-3,
CCL13/MCP-4

Cytokines from T-helper (Th) cells Th1: IFN-, IL-2, IL-12, GM-CSF, TNF-, TNF-, IL-18

Th2: IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, IL-25 (IL-17E)

Cytokines from T-regulatory cells (Tregs) IL-10, TGF

Cytokines of T-cell recruitment IL-16, CCL5/RANTES, CCL3/MIP-1, CCL4/MIP-1, TSLP, CCL17/TARC, CCL22/MDC

Growth factors PDGF, VEGF, TGF-, FGF, EGF, SCF

Cytokine receptor superfamilies

Cytokine receptor superfamily IL-2R and chains, IL-4R, IL-3R and chains, IL-5 and chains, IL-6R, gp130,
IL-12R, GM-CSFR Soluble forms by alternative splicing (e.g. IL-4R)

Immunoglobulin superfamily IL-1R, IL-6R, PDGFR, M-CSFR

Protein kinase receptor superfamily PDGFR, EGFR, FGFR

Interferon receptor superfamily IFN-/ receptor, IFN- receptor, IL-10 receptor

Nerve growth factor superfamily NGFR, TNFR-I (p55), TNFR-II (p75)

Seven-transmembrane G-protein-coupled receptor superfamily Chemokine receptors: CXCR1 to CXCR7, CCR1 to CCR10, CX3CR1

EGF epidermal growth factor; FGF fibroblast growth factor; GM-CSF granulocytemacrophage colony stimulating factor; IFN interferon; IL interleukin; MCP
monocyte chemotactic protein; MDC macrophage-derived chemokine; MIP macrophage inflammatory protein; NGFR nerve growth factor receptor; PDGF platelet-
derived growth factor; R receptor; RANTES regulated on activation, normal T-cell expressed, and secreted; SCF stem cell factor; TARC thymus and activation regulated
chemokine; TGF transforming growth factor; TNF tumor necrosis factor; TSLP thymic stromal lymphopoietin; VEGF vascular endothelial growth factor.

TABLE 27.2 Sources and effects of cytokines.

Cytokine Sources Important cellular and mediator effects

Th2 cytokines

IL-3 Th2 cell Eosinophilia in vivo


Mast cells Pluripotential hematopoietic factor
Eosinophils

IL-4 Th2 cell Eosinophil growth


Mast cells Th2; Th1
Basophils IgE
Eosinophils Mucin expression and goblet cells

IL-5 Th2 cell Eosinophil maturation


Mast cells Apoptosis
Eosinophils Th2 cells
BHR

IL-9 Th2 cells Activated T-cells and IgE from B-cells


Eosinophils Mast cell growth and differentiation
Mucin expression and goblet cells
Causes eosinophilic inflammation and BHR

(Continued)

328
Cytokines 27
TABLE 27.2 (Continued)

Cytokine Sources Important cellular and mediator effects

IL-13 Th2 cells Activates eosinophils


Basophils Apoptosis
Eosinophils IgE
Natural killer cells Mucin expression and goblet cells

IL-15 T-cells As for IL-2


Lung fibroblasts Growth and differentiation of T-cells
Monocytes

IL-23 Antigen presenting cells Induces IL-17A from activated memory CD4 or CD8 T-cells
Induces secretion of IL-17F, IL-6, TNF in Th17 cells

IL-25 (IL-17E) Th2 cells Limits Th1-induced inflammation


Mast cells Causes eosinophilic inflammation and bbronhicl hyperresponsiveness
Increases IL-5, IL-13, CXCL5/ENA-78, CCL11/eotaxin expression
Increases mucus secretion

Th1 cytokines

IFN- Th1 cells Eosinophil influx after allergen


Natural killer cells Th2 cells
Activates endothelial cells, epithelial cells, alveolar macrophages/monocytes
IgE
BHR

IL-2 Th0 & Th1 cells Eosinophilia in vivo


Eosinophils Growth and differentiation of T-cells
Airway epithelial cells

IL-12 B-cells Regulates Th1 cell differentiation


Monocytes/macrophages Expansion of Th2 cells
Dendritic cells IL-4-induced IgE synthesis
Eosinophils Stimulates NK cells and T-cells to produce IFN-

IL-18 Th1 cells Induces IFN- release from mitogen-stimulated blood mononuclear cells
Airway epithelium Induces Th1 cell development together with IL-12
Causes release of CXCL8/IL-8, CCL1/MIP1 and CCL2/MCP-1 from mononuclear cells

Proinflammatory

IL-1 Monocytes/macrophages Adhesion to vascular endothelium; eosinophil accumulation in vivo


Fibroblasts Growth factor for Th2 cells
B-cells B-cell growth factor; neutrophil chemoattractant; T-cell and epithelial activation
Th1 and Th2 cells BHR
Neutrophils
Endothelial cells
Airway epithelial cells
Airway smooth muscle cells

TNF- Macrophages Activates epithelium, endothelium, antigen-presenting cells, monocytes/macrophages


T-cells BHR
Mast cells CXCL8/IL-8 from epithelial cells
Airway epithelial cells Matrix metalloproteinases from macrophages

IL-6 Monocytes/macrophages T-cell growth factor


T-cells B-cell growth factor
B-cells IgE
Fibroblasts
Airway epithelial cells

(Continued)

329
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 27.2 (Continued)

Cytokine Sources Important cellular and mediator effects

CXCL-8/IL-8 Monocytes/macrophages Neutrophil chemoattractant and activator


Airway epithelial cells Chemotactic for CD8 T-cells
Airway smooth muscle cells Activates 5-lipoxygenase in neutrophils
Eosinophils Induces release of histamine and cys-leukotrienes from basophils

IL-11 Airway epithelial cells B-cell growth factor


Airway smooth muscle cells Activates fibroblast
Fibroblasts BHR
Eosinophils

IL-16 CD8 T-cells Eosinophil migration


Epithelial cells Growth factor and chemotaxis of CD4 T-cells
Eosinophils
Mast cells

IL-17A Activated CD4 and CD8 memory Activates epithelial and endothelial cells, and fibroblasts
T-cells Induces release of IL-6, CXCL5 (ENA-78), CXCL8 (IL-8), CCL1 (GRO) and GM-CSF
Neutrophils Neutrophil chemoattractant and activator
Monocytes Induces mucins, Muc5Ac and 5B from epithelial cells

GM-CSF T-cells Eosinophil apoptosis and activation; induces release of leukotrienes


Macrophages Proliferation and maturation of hematopoietic cells
Eosinophils Endothelial cell migration
Fibroblasts BHR
Endothelial cells
Airway smooth muscle cells
Airway epithelial cells

SCF Bone marrow stromal cells VCAM-1 on eosinophils


Fibroblasts Growth factor for mast cells
Epithelial cells

Chemokines

CXCL1/GRO Macrophages/monocytes Neutrophil chemoattractant through CXCR2 receptor


Neutrophils Angiogenesis
Airway smooth muscle cells

CXCL10/IP10 Epithelial cells Chemoattractant for CXCR3-bearing cells including Th1 cells
Monocytes Chemoattractant for mast cells and monocytes
Endothelial cells Promotes T-cell adhesion to endothelial cells
Fibroblasts Inhibits angiogenesis

CCL2/MCP-1 Monocytes/macrophages Activates CCR2 and CCR5 receptors


Vascular and airway smooth muscle Monocyte chemoattractant
Induces MMP seceretion from macrophages
Recruits NK T-cells

CCL5/ Epithelial cells Activates CCR3 receptors, and also CCR1, CCR4 and CCR5
RANTES Fibroblasts Chemoattractant for eosinophil, basophils, memory T-cells, NK T-cells and monocytes
CD8 T-cells
Airway smooth muscle cells

CCL11/ Epithelial cells Activates CCR3 and CCR5 receptors


eotaxin Fibroblasts Eosinophils and monocytes
Vascular endothelium Mobilises eosinophils and its precursors from bone marrow
Eosinophil chemoattractant
Adhesion of eosinophils to endothelium

(Continued)

330
Cytokines 27
TABLE 27.2 (Continued)

Cytokine Sources Important cellular and mediator effects

Inhibitory cytokines

IL-10 Th2 cells Eosinophil survival


Tregs (CD4) Th1 and Th2
CD8 T-cells Monocyte/macrophage activation; B-cell; Mast cell growth and release of
Monocytes/macrophages pro-inflammatory cytokines
Suppresses allergen-specific IgE; induces allergen-specific IgG4
BHR

IL-1ra Monocytes/macrophages Th2 proliferation


BHR

IL-18 Dendritic cells Enhances Th1 cells via IFN- release


Monocytes Releases IFN- from Th1 cells
NK cells Activates NK cells, monocytes
IgE

Growth factors

PDGF Macrophages Fibroblast and airway smooth muscle proliferation


Airway epithelium Release of collagen
Fibroblasts

TGF- Macrophages allergen-specific IgE, and Th1 and Th2 cells


Eosinophils Blocks IL-2 effects
Airway epithelium Down-regulates FceRI expression and co-stimulatory ligand expression
Tregs Fibroblast proliferation
Chemoattractant for monocytes, fibroblasts, mast cells
ASM proliferation

VEGF Macrophages Angiogenic factor


Airway epithelium Increases vascular permeability
Airway and vascular smooth muscle Eosinophil chemotaxis
Mesenchymal cells Apoptosis of lung parenchymal cells
Neutrophils
Eosinophils

ASM airway smooth muscle; BHR bronchial hyperresponsiveness; GM-CSF granulocytemacrophage colony stimulating factor; IgE immunoglobulin E;
IFN interferon; IL interleukin; IL-1ra interleukin-1 receptor antagonist; MCP monocyte chemoattractant protein; MIP macrophage inflammatory factor; NK natural
killer; TNF tumor necrosis factor; Treg CD4CD25 regulatory T-cells; VCAM vascular adhesion molecule.

be considered separately in terms of asthma and chronic the disease, with an increase in eosinophils and neutrophils
obstructive pulmonary disease (COPD), although there is in the airway submucosa and release of mediators such as
often a clear overlap in the role of many cytokines in the histamine and cysteinyl leukotrienes from eosinophils and
context of these two airway diseases. mast cells to induce bronchoconstriction, airway edema, and
mucus secretion. Airway wall remodeling changes include
increase in the thickness of the airway smooth muscle
(ASM) layer result from both hypertrophy and hyperplasia
INFLAMMATION AND CYTOKINES IN ASTHMA and thickening of the lamina reticularis with increased col-
lagen and tenascin deposition, in blood vessels and goblet
cell numbers in the airway epithelium [1].
T-cell-derived cytokine expression Cytokines play an integral role in the coordination and
persistence of the chronic allergic inammatory process in
The chronic airway inammation of asthma is character- asthma (Fig. 27.1). Particular subsets of CD4 T-cells may
ized by an inltration of T-cells, eosinophils, macrophages/ be induced as a result of microenvironmental events resulting
monocytes, and mast cells, and sometimes neutrophils; the the preferential secretion of dened patterns of cytokines,
latter are a possible marker of severity. Acute-on-chronic leading to initiation and propagation of distinct immune
inammation may be observed with acute exacerbations of eector mechanisms. Studies in mouse CD4 T-cell clones,

331
Asthma and COPD: Basic Mechanisms and Clinical Management

Allergen
GM-CSF, CCL11/Eotaxin
CCL5/RANTES

Epithelial cells

F
EG GF
IL-1 Dendritic cells PD F-
TNF- TG
MHC II B7.2
Peptide CD28
TCR
Th1
IL-12 IFN-
IL-12
Macrophage IL-23
Thp
Fibroblast
Th17
IL-4 ASM
CCL2/MCP-1 IL-17 CXCL8/IL-8
IL

IL-13
-1
0

Monocyte
GM-C
SF Neutrophil GM-CSF
Th2 CCL5/RANTES
TGF-
CCL11/Eotaxin TGF-
- IL-10
IL- Treg
5
IL-4
IgE
3 IL-13
TNF- IL-
IL-4
IL-5
Mast cell Eosinophil
IL5 GM-CSF

B-cell
IgE

FIG. 27.1 Interactions of cells and cytokines in airway inflammation of asthma. Antigen presentation by dendritic cells to T-cells with the subsequent
polarization to Th2 cells appears to be important initial process. The roles of other cells are also crucial, such as airway epithelium, eosinophils, neutrophils,
macrophages, and ASM cells. Arrows shown in red with dashed lines indicate potential interactions of allergens through high-affinity IgE receptors with
macrophages and mast cells. GM-CSF: granulocyte-macrophage colony stimulating factors; IFN interferon; IgE immunoglobulin E; IL interleukin;
MCP-1 monocyte chemoattractant protein-1; PDGF platelet-derived growth factor; RANTES regulated on activation, normal T-cell expressed, and
secreted; TCR T-cell receptor; Th2 T-cell helper type 2; Th17 T-cell helper producing IL-17; Treg CD4 regulatory T-cell; TGF transforming
growth factor; TNF tumor necrosis factor.

and later in human CD4 T-cells, indicate the presence of asthmatics [5]. Thus, lung T-cells are activated and express
two basic functional polarized subsets, termed T-helpers and release high levels of Th2-type cytokines.
(Th1 and Th2). The Th2-associated cytokines are one of the A subset of CD4 T-cells expressing CD25, termed
more important classes of cytokines identied in the airway T-regulatory cells (Tregs), are capable of suppressing both
mucosa of patients with asthma. By using in situ hybridiza- Th1 and Th2-mediated adaptive immune responses, and
tion techniques, increased proportions of cells in bronchial these cells may mediate their suppressive actions through
biopsies and in bronchoalveolar lavage (BAL) uid from release of TGF- or IL-10 [6]. An imbalance between aller-
patients with atopic asthma express mRNA for IL-3, IL-4, gen-specic Tregs and eector Th2 cells has been shown in
IL-5, IL-13, and granulocyte-macrophage colony-stimulating allergic diseases [7].
factor (GM-CSF) that is the Th2 gene cluster, when com- Recruitment of Th2 cells to the airways may occur
pared with those of nonatopic control subjects [24]. In through certain chemokines such as CCL17/TARC and
a model of inhalational allergen challenge, an increase in CCL22/MDC which are ligands for CCR4 expressed on Th2
mRNA for these Th2 cytokines, but not for IFN-, has been T-cells. Increased expression of these chemokines in the bron-
demonstrated in bronchial biopsies and BAL cells of atopic chial mucosa of asthma patients has been reported, together

332
Cytokines 27
with the expression of thymic stromal lymphopoietin that can The immunoglobulin-E response and
increase the expression of these Th2 chemoattractants [8]. mast cells
Although CD4 T-cells appear to be the main cell
expressing these Th2 cytokine transcripts, mast cells and eosi- IL-4 is the most important cytokine mediating IgE synthesis
nophils also express IL-4 and IL-5 mRNA [911]. Mast cells in through isotype switching by B-cells, but IL-13 is also capable
mucosal biopsies from atopic asthmatics were positive for IL-3, of a similar action on B-cells [19]. IL-4 also activates B-cells
IL-4, IL-5, IL-6, and tumor necrosis factor- (TNF-) by through increasing the expression of class II MHC molecules,
immunohistochemistry, while immunoreactive IL-5 and GM- as well as enhancing the expression of CD23, the low-anity
CSF in eosinophils have been detected after endobronchial IgE (Fc RII) receptor. CD40 antigen is expressed on B-cells
allergen challenge in asthmatics [10]. Alveolar macrophages after antigen recognition. IL-4 together with the engage-
from patients with asthma release more proinammatory ment of CD40 antigen with its ligand, CD40-L on activated
cytokines (i.e. IL-1, TNF-, GM-CSF, and MIP-1) than T-cells, promotes IgE class switching and B-cell growth.
do macrophages from normal subjects [12, 13]. Because IL-4 can also be produced by basophils, mast cells, and
eosinophils and also have surface CD40-L, these cells may con-
Antigen presentation and tribute to the amplication of IgE responses. IL-4 and IL-13
share the -chain of the IL-4 receptor (IL-4R). On engage-
release of cytokines ment of the ligand with IL-4R, signal transducer and activa-
tor of transcription 6 (STAT 6) translocates to the nucleus, and
The primary signals that activate Th2 cells are related to the
germline mRNA transcription is initiated together with -
presentation of a restricted panel of antigens in the presence
class switching of immunoglobulin genes. Individuals with gain
of appropriate cytokines. Allergens are taken up and proc-
of function mutations in both the extracytoplasmic and intra-
essed by specialized cells within the mucosa, such as dendritic
cellular domains of IL-4R show an enhanced IgE response
cells (antigen presenting cells, APC), followed by presentation
and predisposition to atopic disease [20].
of peptide fragments to naive T-cells. The activation of naive
IgE produced in asthmatic airways binds to Fc RI
T-cells occurs rstly via the T-cell receptor on CD4 cells
receptors (high anity IgE receptors) on mast cells and
through the APC-bound antigen to major histocompatibility
basophils, priming them for activation by antigen. Cross-
complex (MHC)-II complex and, secondly, via the costimula-
linking of Fc RI receptors upregulates its own expression and
tory pathway linked by the B7 family and T-cell-bound CD28
leads to mast cell degranulation, with the release of mediators
[14]. CD28 itself has two major ligands: B7.1 that inhibits Th2
such as histamine and cysteinyl leukotrienes. The importance
cell activation and development, and B7.2 which induces T-cell
of IgE in asthmatic inammation has been emphasized by
activation and Th2 cell proliferation. Under the inuence of
the demonstration that a humanized monoclonal antibody to
IL-12 and IFN-, Th cells are polarised toward Th1 which pre-
IgE inhibits eosinophilic inammation and reduced CD4
dominantly secrete interleukin (IL-2), interferon (IFN-), and
T-cells, IL-4 staining cells and B-cells in the airway mucosa
tumor necrosis factor (TNF), triggering both cell-mediated
of asthmatic patients [21].
immunity and production of opsonizing antibodies. Th2 cells
The maturation and expansion of mast cells from bone
secrete IL-4, IL-5, IL-10, and IL-13, responsible for IgE and
marrow cells involves growth factors and cytokines such as
IgG4 antibody production, and activation of mast cells and
stem cell factor (SCF) and IL-3 derived from structural cells.
eosinophils [15]. A third subset of T-helper cells, Th0, shows
Bronchoalveolar mast cells from asthmatics show enhanced
a composite prole producing both Th1- and Th2-associated
release of mediators such as histamine. Mast cells also elabo-
cytokines. These soluble factors can also be expressed by mac-
rate IL-4 and IL-5 [9]. IL-4 also increases the expression
rophages, epithelial cells, and mast cells and may take part in
of an inducible form of the low-anity receptor for IgE
more general inammatory processes.
(Fc RII or CD23) on B lymphocytes and macrophages [22].
Cytokines may play an important role in antigen presen-
IL-4 drives the dierentiation of CD4 T-helper precursors
tation. Airway macrophages are usually poor at antigen presen-
into Th2-like cells.
tation and suppress T-cell proliferative responses (possibly via
release of cytokines such as IL-1 receptor antagonist), but in
asthma there is reduced suppression after exposure to allergen
[16]. Both GM-CSF and IFN- increase the ability of mac- Eosinophil-associated cytokines
rophages to present allergen and express HLA-DR [17]. IL-1
is important in activating T lymphocytes and is an important The dierentiation, migration, and pathobiological eects
costimulator of the expansion of Th2 cells after antigen pres- of eosinophils may occur through the actions of GM-CSF,
entation [18]. IL-3, IL-5, and certain chemokines such as CCL11/eotaxin
Airway macrophages may also be an important source [2325]. IL-5 inuences the production, maturation, and
of rst wave cytokines, such as IL-1, TNF-, and IL-6, activation of eosinophils, acting predominantly at the later
which may be released on exposure to inhaled allergens via stages of eosinophil maturation and activation, and can also
transduction mediated by Fc RI receptors. These cytokines prolong the survival of eosinophils. IL-5 causes eosinophils
may then act on epithelial cells to cause release of a second to be released from the bone marrow, while the local release
wave of cytokines, including GM-CSF, CCL11/eotaxin, of an eosinophil chemoattractant such as eotaxin may be
and CCL5/RANTES, which then leads to inux of second- necessary for the tissue localization of eosinophils [26].
ary cells, such as eosinophils, which themselves may release Mature eosinophils may show increased survival in bron-
multiple cytokines. chial tissue, secondary to the eects of GM-CSF, IL-3, and

333
Asthma and COPD: Basic Mechanisms and Clinical Management

IL-5 [27]. Eosinophils themselves may also generate other Overexpression of these cytokines in the airway epithelium
cytokines such as IL-3, IL-5, and GM-CSF [11]. causes various features of airway wall remodeling. A CC10-
Cytokines such as IL-4 may also exert an important driven overexpression of IL-13 in the lungs caused eosinophilic
regulatory eect on the expression of adhesion molecules and mononuclear inammation with goblet cell hyperplasia,
such as VCAM-1, both on endothelial cells of bronchial subepithelial brosis, airway obstruction, and airway hyper-
blood vessels and on airway epithelial cells. IL-1 and TNF- responsiveness [34]. IL-4, IL-5, and IL-9 overexpression lead
increase the expression of intercellular adhesion molecule 1 to substantial mucus metaplasia, while IL-9 and IL-5 overex-
(ICAM-1) in both vascular endothelium and airway epi- pression also caused subepithelial brosis and airways hyper-
thelium [28]. An anti-IL5 monoclonal antibody adminis- responsiveness [3537]. ASM hyperplasia occurs following
tered to mild allergic asthmatics has been shown to suppress IL-11 overexpression [38]. IL-5 and IL-13 have been associ-
allergen-induced blood and sputum eosinophilia, without ated with the production of TGF- in asthma [39, 40].
aecting the degree of the late-phase response [29], indi- Proliferation of myobroblasts and the hyperplasia of
cating that IL-5 may not be the only cytokine involved in ASM may also occur through the action of several growth
allergen-induced late-phase responses. Eotaxin is a chem- factors, such as platelet-derived growth factor (PDGF) and
oattractant cytokine (chemokine) selective for eosinophils TGF-. They may be released from inammatory cells in
and acts through the chemokine receptor CCR3 present on the airways, such as macrophages and eosinophils, but also
eosinophils, basophils, and T-cells. Cooperation between by structural cells such as airway epithelium, endothelial
IL-5 and eotaxin appears necessary for the mobilization of cells, and broblasts. These growth factors may stimulate
eosinophils from the bone marrow during allergic reactions brogenesis by recruiting and activating broblasts or trans-
and for the local release of chemokines to induce homing forming myobroblasts [41]. Epithelial cells may release
and migration into tissues. growth factors, since collagen deposition occurs underneath
the basement membrane of the airway epithelium [42].
Growth factors may also stimulate the proliferation
Th17 cytokines: IL-17 family and growth of ASM cells. PDGF, TGF-, and epidermal
growth factor (EGF) are potent stimulants of human ASM
A separate lineage of eector T-helper cells termed Th-17 proliferation [43, 44]. Pro-angiogenic factors such as ang-
cells produced by the action of IL-15 or IL-23 on CD4 or iogenin, vascular endothelial growth factor (VEGF), and
CD8 memory T-cells has been described. Several members CCL2/MCP-1 may be involved in angiogenesis of chronic
of the IL-17 family have been described, with the founder asthma [45]. There is greater expression of airway mucosal
member, IL-17 or IL-17A, originally reported from CD4 eosinophils expressing TGF- mRNA and protein, corre-
memory T-cells. Together with IL-17F, these two members lating with the severity of asthma and the degree of sub-
are the most studied and have overlapping functions par- epithelial brosis [46]. Increased expression of TGF- is
ticularly mediating neutrophil mobilization through activa- also observed in epithelial cells and airway smooth cells of
tion of T-cells (Th17). IL-17A activates neutrophils to cause patients with asthma [47]. EGF expression is increased in
release of neutrophil elastase and myeloperoxidase activity and bronchial epithelium, smooth muscle, and the bronchial
regulate the recruitment of neutrophils in response to aller- submucosa of asthmatic patients [48, 49].
gen, while attenuating allergen-induced increase in IL-5 and
eosinophils [30]. IL-17A may be necessary for the prolifera-
tion of CD4 T-cells for the production of IL4 and IL5 and Airway smooth muscle-associated cytokines
for bronchial hyperresponsiveness [31]. IL-17A induces the
release of CXCL1/GRO, CXCL8/IL-8 and G-CSF, and Airway smooth muscle (ASM) cells have the capacity to
GM-CSF from the airway epithelium, which may be a mech- elaborate a range of cytokines, including IL-4, IL-5, GM-
anism for neutrophilic chemotaxis and activation, as is found CSF, CXCL8/IL-8, CCL5/RANTES, CCL11/eotaxin,
in patients with severe asthma. IL-17A induces the expres- CCL2/MCP-1, and TGF- and therefore may play a role
sion of the mucins, MUC-5AC and MUC-5B, in bronchial in the induction of local inammatory and remodeling
epithelial cells. An increase in the expression of IL-17A in responses [50]. In addition, these cytokines may provide
eosinophils has been shown in patients with asthma [32]. the mechanisms for interacting with cells within the ASM
Unlike IL-17A and IL-17F, IL-17E, also known as bundle of asthmatic patients, such as mast cells [51]. Thus,
IL-25, is a Th2-like cytokine produced by Th2 cells and increased production of CXCL10/IP-10 from ASM cells
mast cells. IL-17E induces overproduction of Th2 cytokines, of asthmatics could be the cause for enhanced chemotactic
IL-4 and IL-13, production of IgA and IgE, mucus, epithe- activity of lung mast cells toward ASM cells in asthma [52].
lial cell hyperplasia, and eosinophilia. IL-17E cells, prob-
ably eosinophils, were detected in the bronchial submucosa
of patients with asthma [33].
INFLAMMATION AND CYTOKINES IN COPD
Airway wall remodeling cytokines
Cytokine profile in COPD
Experimental approaches made possible by genetically
modied mice have highlighted the potential role of various The characteristic pathology of stable COPD includes chronic
T-cell-derived cytokines in aspects of airway wall remodeling. inammation of the small bronchi and bronchioles less than

334
Cytokines 27
2 mm diameter, together with structural changes to all the air- (MMP) proteases. In COPD, as contrasted to healthy smok-
ways and lungs with alveolar wall destruction or emphysema. ers also exposed to cigarette smoke, the release of cytokines
The inammation is characterized by an accumulation of may be amplied by the eect of oxidative stress, perhaps
neutrophils, macrophages, B-cells, lymphoid aggregates, and viruses and by unknown genetic factors (Fig. 27.2).
CD8 T-cells particularly in the small airways and is pro- Cytokines in COPD may be classied as (i) proin-
portionate to the severity of disease [53]. Chronic bronchitis ammatory cytokines including chemokines involved in
characterized by glandular hypertrophy and hyperplasia with the recruitment of neutrophils, macrophages, T-cells, and
dilated ducts of 24 mm internal diameter may be present B-cells (ii) cytokines of airway wall remodeling, including
with mucus plugging, goblet cell hyperplasia, increased goblet cell, ASM hyperplasia (iii) cyokines of emphysema
ASM mass, and distortion due to brosis of bronchioles. (iv) systemic cytokines, that is extrapulmonary source of
Neutrophils are localized particularly to the bronchial epithe- cytokines involved in systemic inammatory responses.
lium, the bronchial glands [54], and also in close apposition to
ASM bundles [55]. Some patients with COPD have a pre-
ponderance of eosinophils in sputum, which has been associ- Proinflammatory cytokines and
ated with improvement of FEV1 with corticosteroid therapy chemokines
[56]. CD8 T-cells expressing IFN- and CXCR3, prob-
ably of the type 1 cytotoxic (Tc1) T-cells [57], are increased Increased levels of IL-6, IL-1, TNF-, CXCL8/IL-8, and
throughout the airways and in lung parenchyma [58]. CCL2/MCP-1 have been observed in induced sputum of
In COPD the basic abnormality is thought to be the patients with stable COPD [59, 60]. Increased release of
response of airway cells, particularly epithelial cells and mac- proinammatory cytokines CXCL8/IL-8, CCL1/GRO,
rophages to toxic gases and particulates in cigarette smoke, CXCL5/ENA-78, IL-1, TNF- and of the anti-inam-
which generate an inammatory and immune response with matory cytokine, IL-10 from alveolar macrophages of cig-
the release of proinammatory cytokines, chemokines, growth arette smokers and of COPD patients have been reported
factors, cathepsins, and serine and matrix metalloproteinase [61, 62]. Levels of TNF-, IL-1, IL-6, CXCL8/IL-8, and

Cigarette smoke, Macrophage


particles, gases Neutrophil elastase
Neutrophil

Bacteria
Amplification
Viruses IL-1, TNF-
Oxidative stress TNF-
Genetic factors Macrophage

Epithelium

CXCL10/IP10, RANTES Fibrogenic cytokines


eg. TGF-, EGF, VEGF
Neutrophil IL-13
CD4/CD8 IL-6, Fibroblast
T-cell CXCL8/IL-8, proliferation,
CCL2/MCP-1, Goblet cell Oxidants
LTB4 Proteases hyperplasis, MMP
Th1 Oxidants mucin genes TIMP
(IFN-) Defensins
CXCL8/IL-8,
is
motax LTB4
Eosinophil ration/che
(Acute) oph il emig Tissue damage (emphysema)
Neutr
Inflammation & Remodeling
Mucus hypersecretion
Airflow obstruction
Airway smooth muscle

COPD

FIG. 27.2 Interaction of cells and cytokines in airway inflammation of COPD. The initiating factors include cigarette smoke with macrophages and airway
epithelium, inducing the release of chemotactic factors for neutrophils, which in turn are important effector cells for inflammation, tissue damage, and repair.
Potential amplification factors include viruses, oxidative stress, and genetic factors. EGF epidermal growth factor; IL interleukin; IP-10 IFN--induced
protein 10; LTB4 leukotriene B4; MCP monocyte chemotactic protein; MMP matrix metalloproteinase; TGF transforming growth factor; TIMP
tissue inhibitor of matrix metalloproteinase; TNF tumor necrosis factor; VEGF vascular endothelial growth factor. Black arrows indicate interactions
with inflammatory cells leading to release of cytokines and mediators; red arrows indicate interactions of bacteria with macrophages and epithelial cells that
could amplify the inflammatory process.

335
Asthma and COPD: Basic Mechanisms and Clinical Management

CCL2/MCP-1 were increased in BAL uid of chronic emphysema in these mice was related to the release of metallo-
smokers compared to non-smokers [63], and of CXCL8/ and cysteine-proteases [78]. Interestingly, the proinam-
IL-8 and GM-CSF levels in COPD patients [64, 65]. In matory cytokine, IL-18, can induce lung inammation and
general, the expression of cytokines from COPD patients is emphysema through the production of IL-13 but not through
higher than of asymptomatic smokers. Sputum neutrophil IFN- [79], despite IFN-s capability of inducing emphy-
counts and level of CXCL8/IL-8 and circulating levels of sema [80]. Although IL-18 expression appears to be increased
TNF- and C-reactive protein (CRP) have been reported in pulmonary macrophages of COPD patients, IL-13 expres-
to be the best markers for distinguishing the various levels sion is decreased in emphysema lung [81]. IL-9-mRNA is
of severity of COPD [66]. overexpressed in CD3 T-cells in patients with COPD [82].
Cigarette smoke increases CXCL8/IL-8 gene expres-
sion and release by bronchial epithelial cells, and TNF-
and IL-6 by alveolar macrophages [67, 68], through oxidant Systemic cytokines
mechanisms that include activation of the transcription fac-
tor, nuclear factor-B (NF-B) [69]. Exposure of lung epi- Evidence of systemic inammatory processes in COPD can
thelial cells to smoke extract causes the release of neutrophil be judged from increased circulating levels of cytokines such as
and monocytic chemotactic activities with CXCL8/IL-8 IL-6, CXCL8/IL-8, TNF-, and TNF receptors 55 and 75,
and G-CSF accounting for the neutrophilic activity and together with CRP levels [83]. The source of these circulating
CCL-2/MCP-1 for the monocytic activity [70]. cytokines may represent a spill-over from pulmonary sources
A higher expression of CCL2/MCP-1, TGF-1 but may also originate from other sources such as circulating
and CXCL8/IL-8 mRNA and protein has been observed blood monocytes [84], striated muscle or from concomitant
in bronchiolar epithelium in macrophages of smokers with atherosclerotic lesions. In stable COPD patients with respira-
COPD compared with those smokers without COPD [71]; tory muscle impairment, increased TNF- and IL-6 gene and
a similar dierence has been found for CCR2. Since CCL2/ protein expression in muscle has been reported [85].
MCP-1 binds to CCR2 and can induce T-cell and mono-
cytic migration, CCL2 [72] may contribute to the recruit-
ment of these cells in COPD. Exacerbations of COPD
Dierent chemokines in induced sputum from COPD
patients have been found to be involved in the chemotaxis of Increases in plasma levels of IL-6, CXCL8/IL-8, and LTB4
monocytes. Thus, there is increased chemotaxis of monocytes but not of TNF- have been reported during COPD exacer-
in COPD for CCL1/GRO or CXCL7/NAP2 but not for bations severe enough to require the patient to be hospitalized
CXCL8/IL-8 or CXCL5/ENA-78, possibly due to dier- [86]. During exacerbations of COPD associated with neu-
ential regulation of CXCR2 receptors in COPD [73]. CD8 trophilia requiring mechanical ventilatory support, gene expres-
T-cells expressing IFN- inltrate the peripheral airways of sion for CXCL5/ENA78, CXCL8/IL-8, and the receptor
smokers with COPD and have an increased expression of the CXCR2 is increased in tissue, particularly in the bronchiolar
chemokine receptor, CXCR3, which is paralleled by a strong epithelium [87]. Both CXCL5/ENA-78 and CXCL8/IL-8
epithelial expression of its ligand, CXCL10/IP-10 [57], a che- bind to CXCR2 and are neutrophil chemoattractants.
moattractant for Th1 cells. The chemokine ligand 20, CCL20, In acute exacerbations of COPD, eosinophils may be
is the most potent known chemoattractant for dendritic cells prominent among the cells recovered in sputum or in bron-
and is upregulated in lung and in induced sputum of patients chial biopsies, but there is no increased expression of IL-5 pro-
with COPD [74]. This could account for increased number tein in tissues [88]. Higher levels of IL-6 and CXCL8/IL-8
of dendritic cells observed in the small airways of COPD as in induced sputum of patients with recurrent exacerbations of
these cells express CCR6 which is the receptor for CCL20. COPD (3 per year) have been reported [89]. In patients with
Microarray analysis of lung tissues from COPD patients stable COPD, lymphomononuclear cells expressing eotaxin
revealed the expression of CX3CL1/fractalkine [75]. mRNA was increased compared to healthy non-smokers. In
patients with an exacerbation of chronic bronchitis, RANTES
mRNA expression was upregulated and strongly expressed on
Th2 cytokines the surface epithelium and subepithelial lympho-mononuclear
cells, together with increased number of eosinophils [76].
Expression of Th2 cytokines is less prominent in COPD as CCL11/eotaxin and its receptor, CCR3, are upregulated dur-
compared to asthma. IL-5 expression is absent in COPD ing exacerbations of chronic bronchitis, with CCR3 colocal-
airways associated with eosinophilia, but other eosinophil ized mainly to eosinophils [90]. The increased expression of
chemoattractants such as eotaxin or RANTES may be CCL11/eotaxin and CCL5/RANTES may underlie the eosi-
implicated, particularly during exacerbations [76]. Increased nophilia sometimes observed during exacerbations of COPD.
IL-4 cells in mucus secreting cells and associated plasma
cells of the airway mucosa of chronic bronchitis patients Properties of some specific
have been reported [77].
Overexpression of the Th2 cytokine, IL-13, in lungs of cytokines in COPD
adult mice induces emphysema, mucus goblet cell hyperplasia,
and airway inammation with macrophages, lymphocytes, TNF-
and eosinophils, and increased MMPs, which are many of TNF- is produced by many cells including macrophages,
the features associated with COPD [78]. The induction of T-cells, mast cells, and epithelial cells, but the principal

336
Cytokines 27
source is the macrophage. The secretion of TNF- by from intracellular storage organelles, and respiratory burst.
monocytes/macrophages is greatly enhanced by other It also upregulates the expression of two integrins (CD11b/
cytokines such as IL-1, GM-CSF, and IFN-. TNF- CD18 and CD11c/CD18) during exocytosis of specic
activates NFB that switches on the transcription of the granules. CXCL8/IL-8 activates neutrophil 5-lipoxygenase
CXCL8/IL-8 gene and increases CXCL8/IL-8 release from with the formation of LTB4 and 5-hydroxy-eicosanotetrae-
the airway epithelium and neutrophils. Bacteria and bacterial noic acid (5-HETE) and together with LTB4 contributes to
products such as lipopolysaccharide can induce CXCL8/IL-8 the neutrophil chemotactic activity of sputum from COPD
expression, probably as a result of initial TNF- production patients [104, 105]. CXCL8/IL-8 also has chemoattractant
[91, 92]. TNF- may be important in inducing the expres- properties for T-cells. Bacteria for example due to a soluble
sion of CXCL2/MIP-2 and CCL2/MCP-1, and in connec- cytoplasmic factor of nontypeable Haemophilus Inuenzae
tive tissue breakdown following cigarette smoke exposure can induce CXCL8/IL-8 expression in epithelial cells [106].
[93]. TNF- through the activation of EGF receptor leads to The increased levels of CXCL8/IL-8 found in sputum sam-
the expression of mucin genes, in particular MUC5AC [94]. ples of COPD patients correlate with airway bacterial load
TNF- increases the expression of ICAM-1, which and with myeloperoxidase released from activated neu-
is increased in serum of COPD patients [95]. TNF- may trophils. Elastase released from neutrophils may also stimu-
activate macrophages to produce MMPs. This eect is inhib- late epithelial cells to produce more CXCL8/IL-8 [107]. A
ited by the anti-inammatory cytokine, IL-10, which also study of a blocking anti-IL-8 antibody in moderate COPD
enhances the release of tissue inhibitor of metalloproteinases patients showed a small eect in improving dyspnea [108].
(TIMP) in macrophages from normal volunteers. In smok-
ers IL-10 increases TIMP-1 release, without modifying TGF-
MMP-9 release, from alveolar macrophages [61]. TNF- TGF- is a multifunctional growth factor with biological
also stimulates bronchial epithelial cells to produce tenascin, eects on broblast proliferation, deposition of extracellular
an extracellular matrix glycoprotein that may contribute to matrix, epithelial repair, chemotactic activities, and also as
airway remodeling. In animal models, MMP-12 induces the an immune suppressor. TGF- is secreted as latent TGF-
release of TNF- from lung macrophages exposed to ciga- that is sequestered by many binding proteins including
rette smoke, causing endothelial activation and neutrophil decorin, biglycan, bronectin, and elastin. TGF- can be
inux [96]. activated in vivo by mechanisms including thrombospondin,
Increased serum concentrations of TNF- have been acid pH, reactive oxygen species, and nitric oxide nitrosyla-
measured in patients with COPD with weight loss [97, tion. TGF- regulates epithelial repair by inhibiting pro-
98], and increased TNF- production has been noted by liferation but increasing epithelial cell migration [109].
peripheral blood monocytes from COPD patients during TGF-1 is involved in the transformation of broblasts to
the catabolic phase of their illness leading to weight loss myobroblasts, and enhancing myobroblast survival. In
[99]. TNF- has direct eects on skeletal muscle cells and addition, TGF-1 and TGF-2 can convert myobroblasts
reduces adult myosin heavy chain content and induces apop- into smooth muscle cells [110]. TGF- induces hypertro-
tosis of skeletal muscle via several signaling pathways [100, phy and hyperplasia of ASM cells [47, 111]. TGF- induces
101]. Given the potential role for TNF- in the inamma- collagen production in human lung broblasts [112] and
tion of COPD, the eect of TNF- blockers in this disease ASM cells [113]. Therefore, TGF- is involved in remod-
is of therapeutic interest. However, a multicenter randomized eling of the airway wall in both asthma and COPD.
trial of an anti-TNF- antibody, iniximab, found no treat- Increased expression of a number of related growth
ment benet in terms of quality of life, lung function, and factors, CTGF, TGF-1, and PDGFRA in the lungs using
the 6-min walking test [102]. gene microarray analysis of lungs from smoking COPD
patients compared to healthy smokers has been reported
IL-1 [75]. Expression of TGF- in the bronchiolar and alveolar
IL-1 induces leukocytosis by the release of neutrophils epithelium and submucosal cells of patients with COPD has
from the bone marrow and induces the release of many been reported [114116]. In addition, increased release of
other cytokines such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, TGF- and of bronectin from alveolar macrophages from
CXCL8/IL-8, CCL5/RANTES, GM-CSF, IFN-, TNF, patients with chronic bronchitis has been observed [117].
and PDGF from a variety of cells. It induces broblasts to
proliferate, and increases prostaglandin secretion, and the VEGF
synthesis of bronectin and collagen. Together with TNF-,
IL-1 induces ICAM-1 expression on endothelial cells. IL-1 Increased levels of VEGF have been observed in sputum of
can induce attenuation of -adrenergic receptor-induced patients with COPD [118], and its augmented expression
airway relaxation through reduction of -adrenoreceptors, appears to be localized to pulmonary arteries [119]. Inhibition
an increase in the G-protein, Gi subunit, and a defect in of VEGF receptor activation in mice led to the induction of
adenylate cyclase activity [103]. enlargement of air spaces, and induced alveolar septal cell
apoptosis, indicating that VEGF receptor signaling is required
for maintenance of alveolar structures [120]. Oxidative stress
CXCL8/IL-8 and apoptosis may be the down-stream eects of VEGF
CXCL8/IL-8 is a neutrophil chemoattractant and activa- receptor activation [121]. The nding that VEGF levels are
tor that induces a transient shape change, rise in intracellu- increased in patients with COPD reinforces a potential non-
lar calcium, exocytosis with release of enzymes and proteins proteolytic mechanism for emphysema through VEGF.

337
Asthma and COPD: Basic Mechanisms and Clinical Management

CONCLUSION that may reect similarities in established disease but not


necessarily during the initial phase of these diseases. For
example, severe asthma may resemble more closely COPD
Because there has been little direct comparison of tissues with a neutrophilic inammatory response, and remodeling
between asthma and COPD patients, it is not possible to aspects of the airways may be similar despite being dier-
state categorically the degree of overlap of cytokine proles ently localized in the airways in terms of the cytokines and
or networks that exist between the two diseases. However, growth factors involved. Further analysis of cytokine expres-
analysis of cytokine expression in asthma and COPD from sion in asthma and COPD may help determine whether
the published literature reveals many similarities in terms these conditions have common mechanisms or similar pre-
of increased expression of many cytokines (Table 27.3) disposing factors. Analysis of cytokine expression may help
determine which ones may be useful to target for therapeu-
tic eects, although the limited experience so far has been
disappointing.
TABLE 27.3 Expression of cytokines in asthma and COPDa.

Asthma COPD
References
IL-3  ?
1. Bousquet J, Jeery PK, Busse WW, Johnson M, Vignola AM. Asthma.
IL-4   From bronchoconstriction to airways inammation and remodeling.
Am J Respir Crit Care Med 161(5): 172045, 2000.
IL-5  2. Hamid Q, Azzawi M, Ying S, Moqbel R, Wardlaw AJ, Corrigan CJ
et al. Expression of mRNA for interleukins in mucosal bronchial biop-
IL-9  
sies from asthma. J Clin Invest 87: 154146, 1991.
IL-10  or  3. Broide DH, Lotz M, Cuomo AJ, Coburn DA, Federman EC,
Wasserman SI. Cytokines in symptomatic asthmatic airways. J Allergy
IL-13   Clin Immunol 89: 95867, 1992.
4. Humbert M, Durham SR, Kimmitt P, Powell N, Assou B, Pster R
IL-17A  ? et al. Elevated expression of messenger ribonucleic acid encoding IL-13
in the bronchial mucosa of atopic and nonatopic subjects with asthma.
IL-25 (IL-17E)  ? J Allergy Clin Immunol 99(5): 65765, 1997.
5. Robinson DS, Hamid Q, Ying S, Tsigopoulos A, Barkans T, Bentley
IFN 
AM et al. Predominant Th2-type bronchoalveolar lavage T-lymphocyte
CCL2/MCP-1   population in atopic asthma. New Engl J Med 326: 298304, 1992.
6. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms
CCL5/RANTES  * of immune suppression by interleukin-10 and transforming growth
factor-beta: The role of T regulatory cells. Immunology 117(4): 43342,
CCL11/Eotaxin  * 2006.
7. Ling EM, Smith T, Nguyen XD, Pridgeon C, Dallman M, Arbery J
CCL17/TARC  ? et al. Relation of CD4CD25 regulatory T-cell suppression of
allergen-driven T-cell activation to atopic status and expression of
CCL20 ? 
allergic disease. Lancet 363(9409): 60815, 2004.
CCL22/MDC  ? 8. Ying S, OConnor B, Rato J, Meng Q, Mallett K, Cousins D et al.
Thymic stromal lymphopoietin expression is increased in asthmatic air-
CXCL1/GRO ?  ways and correlates with expression of Th2-attracting chemokines and
disease severity. J Immunol 174(12): 818390, 2005.
CXCL8/IL-8   9. Bradding P, Roberts JA, Britten KM, Montefort S, Djukanovic R,
Mueller R et al. Interleukin-4, -5 and -6 and tumor necrosts factor-
CXCL10/IP10   in normal and asthmatic airways: Evidence for the human mast cell as
a source of these cytokines. Am J Respir Cell Mol Biol 10: 47180, 1994.
IL-1  
10. Broide D, Paine MM, Firestein GS. Eosinophils express interleukin S
TNF-   and granulocyte-macrophage colony-stimulating factor mRNA at sites
of allergic inammation in asthmatics. J Clin Invest 90: 141424, 1992.
TGF-1   11. Moqbel R, Hamid Q, Ying S, Barkans J, Hartnell A, Tsicopoulos A
et al. Expression of mRNA and immunoreactivity for the granulocyte/
CTGF ?  macrophage colony-stimulating factor in activated human eosinophils.
J Exp Med 174: 74952, 1991.
GM-CSF   12. John M, Lim S, Seybold J, Jose P, Robichaud A, OConnor B et al. Inhaled
corticosteroids increase interleukin-10 but reduce macrophage inamma-
EGF  
tory protein-1alpha, granulocyte-macrophage colony-stimulating factor,
VEGF   and interferon-gamma release from alveolar macrophages in asthma. Am J
Respir Crit Care Med 157(1): 25662, 1998.
a
From review of literature: expression is reported as gene or protein in airway cells or 13. Hallsworth MP, Soh CPC, Lane SJ, Arm JP, Lee TH. Selective
tissues, in addition to measurement of protein release in airway fluids. The degree of enhancement of GM-CSF, TNF-, IL-1 and IL-8 production by
expression has been arbitrarily assessed as: reduced;  or   increased; ? monocytes and macrophages of asthmatic subjects. Eur Respir J 7: 1096
not known or uncertain; * associated with exacerbations. 102, 1994.

338
Cytokines 27
14. Green JM. The B7/CD28/CTLA4 T-cell activation pathway. 33. Letuve S, Lajoie-Kadoch S, Audusseau S, Rothenberg ME, Fiset PO,
Implications for inammatory lung disease. Am J Respir Cell Mol Biol Ludwig MS et al. IL-17E upregulates the expression of proinamma-
22(3): 26164, 2000. tory cytokines in lung broblasts. J Allergy Clin Immunol 117(3): 590
15. Mossman TR, Coman RL. TH1 and TH2 cells : Dierent patterns 96, 2006.
of lymphokine secretion lead to dierent functional properties. Ann 34. Zhu Z, Homer RJ, Wang Z, Chen Q, Geba GP, Wang J et al.
Rev Immunol 7: 14573, 1989. Pulmonary expression of interleukin-13 causes inammation, mucus
16. Spiteri M, Knight RA, Jeremy JY, Barnes PJ, Chung KF. Alveolar hypersecretion, subepithelial brosis, physiologic abnormalities, and
macrophage-induced suppression of T-cell hyperresponsiveness in eotaxin production. J Clin Invest 103(6): 77988, 1999.
bronchial asthma is reversed by allergen exposure. Eur Respir J 7: 1431 35. Temann UA, Geba GP, Rankin JA, Flavell RA. Expression of interleukin
38, 1994. 9 in the lungs of transgenic mice causes airway inammation, mast cell
17. Fischer HG, Frosch S, Reske K, Reske-Kunz AB. Granulocyte- hyperplasia, and bronchial hyperresponsiveness. J Exp Med 188: 130720,
macrophage colony-stimulating factor activates macrophages derived 1998.
from bone marrow cultures to synthesis of MHC class II molecules 36. Rankin JA, Picarella DE, Geba GP, Temann UA, Prasad B, DiCosmo B
and to augmented antigen presentation function. J Immunol 141: 3882 et al. Phenotypic and physiologic characterization of transgenic mice
88, 1988. expressing interleukin 4 in the lung: Lymphocytic and eosinophilic
18. Chang TL, Shea CH, Urioste S, Thompson RC, Boom WH, Abbas AK. inammation without airway hyperreactivity. Proc Natl Acad Sci USA
Heterogeneity of helper/inducer T lymphocytes: Lymphokine produc- 93(15): 782125, 1996.
tion and lymphokine responsiveness. J Immunol 145: 28038, 1990. 37. Lee JJ, McGarry MP, Farmer SC, Denzler KL, Larson KA, Carrigan PE
19. Minty A, Chalon P, Derocq J-M, Dumont X, Guillemot J-C, Kaghad M et al. Interleukin-5 expression in the lung epithelium of transgenic
et al. Interleukin-13 is a new human lymphokine regulating inamma- mice leads to pulmonary changes pathognomonic of asthma. J Exp
tory and immune responses. Nature 362: 24850, 1993. Med 185(12): 214356, 1997.
20. Khuruna Hershey GK, Friedrich MF, Esswein LA, Thomas ML, 38. Einarsson O, Geba GP, Zhu Z, Landry M, Elias JA. Interleukin-11:
Chatila TA. The association of atopy with a gain-of-function mutation in Stimulation in vivo and in vitro by respiratory viruses and induction of
the -subunit of the IL-4 receptor. New Eng J Med 337: 172025, 1997. airways hyperresponsiveness. J Clin Invest 97(4): 91524, 1996.
21. Djukanovic R, Wilson SJ, Kraft M, Jarjour NN, Steel M, Chung KF 39. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A,
et al. Eects of treatment with anti-immunoglobulin E antibody oma- Ludwig MS et al. Anti-IL-5 treatment reduces deposition of ECM
lizumab on airway inammation in allergic asthma. Am J Respir Crit proteins in the bronchial subepithelial basement membrane of mild
Care Med 170(6): 58393, 2004. atopic asthmatics. J Clin Invest 112(7): 102936, 2003.
22. Vercelli D, Jabara HH, Lee BW, Woodland N, Geha RS, Leung DY. 40. Lee CG, Homer RJ, Zhu Z, Lanone S, Wang X, Koteliansky V et al.
Human recombinant interleukin-4 induces FC4RII/CD23 on normal Interleukin-13 induces tissue brosis by selectively stimulating and
human monocytes. J Exp Med 167: 140616, 1988. activating transforming growth factor beta(1). J Exp Med 194(6): 809
23. Sanderson CJ, Warren DJ, Strath M. Identication of a lumphokine 21, 2001.
that stimulates eosinophil dierentiation in vitro. Its relationship to 41. Boxall C, Holgate ST, Davies DE. The contribution of transforming
interleukin 3, and functional properties of eosinophils produced in growth factor-beta and epidermal growth factor signalling to airway
cultures. J Exp Med 162: 6074, 1985. remodelling in chronic asthma. Eur Respir J 27(1): 20829, 2006.
24. Jose PJ, Griths-Johnson DA, Collins PD, Walsh DT, Moqbel R, 42. Brewster CEP, Howarth PH, Djukanovic R, Wilson J, Holgate ST,
Totty NF et al. Eotaxin: A potent eosinophil chemoattractant cytokine Roche WR. Myobroblasts and subepithelial brosis in bronchial
detected in a guinea pig model of allergic airways inammation. J Exp asthma. Am J Resp Cell Mol Biol 3: 50711, 1990.
Med 179: 88187, 1994. 43. Hirst SJ, Martin JG, Bonacci JV, Chan V, Fixman ED, Hamid QA et al.
25. Ponath PD, Qin S, Post TW, Wang J, Wu L, Gerard N et al. Molecular Proliferative aspects of airway smooth muscle. J Allergy Clin Immunol
cloning and characterization of a human eotaxin receptor expressed 114(2 Suppl): S217, 2004.
selectively on eosinophils. J Exp Med 183: 243748, 1996. 44. Leung SY, Niimi A, Noble A, Oates T, Williams AS, Medicherla S
26. Collins PD, Griths-Johnson DA, Jose PJ, Williams TJ, Marleau S. et al. Eect of transforming growth factor-beta receptor I kinase inhib-
Co-operation between interleukin-5 and the chemokine, eotaxin, to itor 2,4-disubstituted pteridine (SD-208) in chronic allergic airway
induce eosinophil accumulation in vivo. J Exp Med 182: 116974, 1995. inammation and remodeling. J Pharmacol Exp Ther 319(2): 58694,
27. Rothenberg ME, Owen WFJ, Siberstein DS. Human eosinophils have 2006.
prolonged survival, enhanced functional properties and become hypodense 45. Simcock DE, Kanabar V, Clarke GW, OConnor BJ, Lee TH, Hirst SJ.
when exposed to human interleukin. J Clin Invest 81: 198692, 1988. Proangiogenic activity in bronchoalveolar lavage uid from patients with
28. Tosi MF, Stark JM, Smith WC, Hamedani A, Gruenert DC, asthma. Am J Respir Crit Care Med 176(2): 14653, 2007.
Infeld MD. Induction of ICAM-1 expression on human airway epi- 46. Minshall EM, Leung DYM, Martin RJ, Song YL, Cameron L, Ernst P
thelial cells by inammatory cytokines: Eects on neutrophil-epithelial et al. Eosinophil-associated TGF1 mRNA expression and airways bro-
cell adhesion. Am J Respir Cell Mol Biol 7: 21421, 1992. sis in asthma. Am J Respir Cell Mol Biol 17: 32633, 1997.
29. Leckie MJ, ten BA, Khan J, Diamant Z, OConnor BJ, Walls CM et al. 47. Xie S, Sukkar MB, Issa R, Khorasani NM, Chung KF. Mechanisms
Eects of an interleukin-5 blocking monoclonal antibody on eosi- of induction of airway smooth muscle hyperplasia by transform-
nophils, airway hyper-responsiveness, and the late asthmatic response. ing growth factor-beta. Am J Physiol Lung Cell Mol Physiol 293(1):
Lancet 356(9248): 214448, 2000. L24553, 2007.
30. Hellings PW, Kasran A, Liu Z, Vandekerckhove P, Wuyts A, 48. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo A
Overbergh L et al. Interleukin-17 orchestrates the granulocyte inux et al. Transforming growth factor-beta expression in mucosal biopsies in
into airways after allergen inhalation in a mouse model of allergic asthma and chronic bronchitis. Am J Respir Crit Care Med 156(2 Pt 1):
asthma. Am J Respir Cell Mol Biol 28(1): 4250, 2003. 59199, 1997.
31. Nakae S, Komiyama Y, Nambu A, Sudo K, Iwase M, Homma I et al. 49. Amishima M, Munakata M, Nasuhara Y, Sato A, Takahashi T,
Antigen-specic T cell sensitization is impaired in IL-17-decient mice, Homma Y et al. Expression of epidermal growth factor and epidermal
causing suppression of allergic cellular and humoral responses. Immunity growth factor receptor immunoreactivity in the asthmatic human air-
17(3): 37587, 2002. way. Am J Respir Crit Care Med 157(6 Pt 1): 190712, 1998.
32. Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Page N et al. IL-17 is 50. Howarth PH, Knox AJ, Amrani Y, Tliba O, Panettieri RA Jr., Johnson M.
increased in asthmatic airways and induces human bronchial broblasts Synthetic responses in airway smooth muscle. J Allergy Clin Immunol
to produce cytokines. J Allergy Clin Immunol 108(3): 43038, 2001. 114(2 Suppl): S3250, 2004.

339
Asthma and COPD: Basic Mechanisms and Clinical Management

51. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, into the airways through nuclear factor-kappaB activation and IL-8
Pavord ID. Mast-cell inltration of airway smooth muscle in asthma. mRNA expression in guinea pigs in vivo. Am J Respir Cell Mol Biol 20(2):
New Engl J Med 346(22): 1699705, 2002. 18998, 1999.
52. Brightling CE, Ammit AJ, Kaur D, Black JL, Wardlaw AJ, Hughes JM 70. Masubuchi T, Koyama S, Sato E, Takamizawa A, Kubo K, Sekiguchi M
et al. The CXCL10/CXCR3 axis mediates human lung mast cell et al. Smoke extract stimulates lung epithelial cells to release neutrophil
migration to asthmatic airway smooth muscle. Am J Respir Crit Care and monocyte chemotactic activity. Am J Pathol 153(6): 190312, 1998.
Med 171(10): 11038, 2005. 71. de Boer WI, Sont JK, van Schadewijk A, Stolk J, van Krieken JH,
53. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L et al. Hiemstra PS. Monocyte chemoattractant protein 1, interleukin 8,
The nature of small-airway obstruction in chronic obstructive pulmo- and chronic airways inammation in COPD. J Pathol 190(5): 61926,
nary disease. New Engl J Med 350(26): 264553, 2004. 2000.
54. Saetta M, Turato G, Facchini FM, Corbino L, Lucchini RE, Casoni G 72. Premack BA, Schall TJ. Chemokine receptors: Gateways to inamma-
et al. Inammatory cells in the bronchial glands of smokers with tion and infection. Nat Med 2(11): 117478, 1996.
chronic bronchitis. Am J Respir Crit Care Med 156(5): 163339, 1997. 73. Traves SL, Smith SJ, Barnes PJ, Donnelly LE. Specic CXC but not
55. Baraldo S, Turato G, Badin C, Bazzan E, Beghe B, Zuin R et al. CC chemokines cause elevated monocyte migration in COPD: A role
Neutrophilic inltration within the airway smooth muscle in patients for CXCR2. J Leukoc Biol 76(2): 44150, 2004.
with COPD. Thorax 59(4): 30812, 2004. 74. Demedts IK, Bracke KR, Van Pottelberge G, Testelmans D, Verleden GM,
56. Chanez P, Vignola AM, OShaugnessy T, Enander I, Li D, Jeery PK Vermassen FE et al. Accumulation of dendritic cells and increased CCL20
et al. Corticosteroid reversibility in COPD is related to features of levels in the airways of patients with chronic obstructive pulmonary dis-
asthma. Am J Respir Crit Care Med 155(5): 152934, 1997. ease. Am J Respir Crit Care Med 175(10): 9981005, 2007.
57. Saetta M, Mariani M, Panina-Bordignon P, Turato G, Buonsanti C, 75. Ning W, Li CJ, Kaminski N, Feghali-Bostwick CA, Alber SM, Di YP
Baraldo S et al. Increased Expression of the Chemokine Receptor et al. Comprehensive gene expression proles reveal pathways related
CXCR3 and Its Ligand CXCL10 in Peripheral Airways of Smokers to the pathogenesis of chronic obstructive pulmonary disease. Proc Natl
with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Acad Sci USA 101(41): 14895900, 2004.
Med 165(10): 14049, 2002. 76. Zhu J, Qiu YS, Majumdar S, Gamble E, Matin D, Turato G et al.
58. OShaughnessy TC, Ansari TW, Barnes NC, Jeery PK. Inammation Exacerbations of Bronchitis: Bronchial eosinophilia and gene expres-
in bronchial biopsies of subjects with chronic bronchitis: Inverse rela- sion for interleukin-4, interleukin-5, and eosinophil chemoattractants.
tionship of CD8 T lymphocytes with FEV1. Am J Respir Crit Care Am J Respir Crit Care Med 164(1): 10916, 2001.
Med 155: 85257, 1997. 77. Zhu J, Qiu Y, Valobra M, Qiu S, Majumdar S, Matin D et al. Plasma
59. Keatings VM, Collins PD, Scott DM, Barnes PJ. Dierences in cells and IL-4 in chronic bronchitis and chronic obstructive pulmonary
interleukin-8 and tumor necrosis factor-alpha in induced sputum from disease. Am J Respir Crit Care Med 175(11): 112533, 2007.
patients with chronic obstructive pulmonary disease or asthma. Am J 78. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, Riese RJ Jr. et al. Inducible
Respir Crit Care Med 153(2): 53034, 1996. targeting of IL-13 to the adult lung causes matrix metalloproteinase- and
60. Traves SL, Culpitt SV, Russell RE, Barnes PJ, Donnelly LE. Increased cathepsin-dependent emphysema. J Clin Invest 106(9): 108193, 2000.
levels of the chemokines GROalpha and MCP-1 in sputum samples 79. Hoshino T, Kato S, Oka N, Imaoka H, Kinoshita T, Takei S et al.
from patients with COPD. Thorax 57(7): 59095, 2002. Pulmonary Inammation and Emphysema: Role of the Cytokines
61. Lim S, Roche N, Oliver BG, Mattos W, Barnes PJ, Chung KF. Balance IL-18 and IL-13. Am J Respir Crit Care Med 176(1): 4962, 2007.
of matrix metalloprotease-9 and tissue inhibitor of metalloprotease-1 from 80. Wang Z, Zheng T, Zhu Z, Homer RJ, Riese RJ, Chapman HA Jr.
alveolar macrophages in cigarette smokers. Regulation by interleukin-10. et al. Interferon gamma induction of pulmonary emphysema in the
Am J Respir Crit Care Med 162(4 Pt 1): 135560, 2000. adult murine lung. J Exp Med 192(11): 1587600, 2000.
62. Morrison D, Strieter RM, Donnelly SC, Burdick MD, Kunkel SL, 81. Boutten A, Bonay M, Laribe S, Leseche G, Castier Y, Lecon-Malas V
MacNee W. Neutrophil chemokines in bronchoalveolar lavage uid et al. Decreased expression of interleukin 13 in human lung emphysema.
and leukocyte-conditioned medium from nonsmokers and smokers. Thorax 59(10): 85054, 2004.
Eur Respir J 12(5): 106772, 1998. 82. Panzner P, Latte JJ, Tsicopoulos A, Hamid Q, Tulic MK. Marked up-
63. Kuschner WG, DAlessandro A, Wong H, Blanc PD. Dose-dependent regulation of T lymphocytes and expression of interleukin-9 in bron-
cigarette smoking-related inammatory responses in healthy adults. chial biopsies from patients with chronic bronchitis with obstruction.
Eur Respir J 9(10): 198994, 1996. Chest 124(5): 190915, 2003.
64. Pesci A, Balbi B, Majori M, Cacciani G, Bertacco S, Alciato P et al. 83. Gan WQ, Man SF, Senthilselvan A, Sin DD. Association between
Inammatory cells and mediators in bronchial lavage of patients with chronic obstructive pulmonary disease and systemic inammation: A
chronic obstructive pulmonary disease. Eur Respir J 12(2): 38086, systematic review and a meta-analysis. Thorax 59(7): 57480, 2004.
1998. 84. de GI, Donahoe M, Calhoun WJ, Mancino J, Rogers RM. Elevated
65. Balbi B, Bason C, Balleari E, Fiasella F, Pesci A, Ghio R et al. Increased TNF-alpha production by peripheral blood monocytes of weight-losing
bronchoalveolar granulocytes and granulocyte/macrophage colony- COPD patients. Am J Respir Crit Care Med 153(2): 63337, 1996.
stimulating factor during exacerbations of chronic bronchitis. Eur Respir 85. Casadevall C, Coronell C, Ramirez-Sarmiento AL, Martinez-Llorens J,
J 10(4): 84650, 1997. Barreiro E, Orozco-Levi M et al. Upregulation of proinammatory
66. Franciosi LG, Page CP, Celli BR, Cazzola M, Walker MJ, Danhof M cytokines in the intercostal muscles of COPD patients. Eur Respir J,
et al. Markers of disease severity in chronic obstructive pulmonary dis- 2007.
ease. Pulm Pharmacol Ther 19(3): 18999, 2006. 86. Pinto-Plata VM, Livnat G, Girish M, Cabral H, Masdin P, Linacre P
67. Mio T, Romberger DJ, Thompson AB, Robbins RA, Heires A, et al. Systemic cytokines, clinical and physiological changes in patients
Rennard SI. Cigarette smoke induces interleukin-8 release from human hospitalized for exacerbation of COPD. Chest 131(1): 3743, 2007.
bronchial epithelial cells. Am J Respir Crit Care Med 155(5): 177076, 87. Qiu Y, Zhu J, Bandi V, Atmar RL, Hattotuwa K, Guntupalli KK et al.
1997. Biopsy neutrophilia, neutrophil chemokine and receptor gene expres-
68. Dubar V, Gosset P, Aerts C, Voisin C, Wallaert B, Tonnel AB. In vitro sion in severe exacerbations of chronic obstructive pulmonary disease.
acute eects of tobacco smoke on tumor necrosis factor alpha and Am J Respir Crit Care Med 168(8): 96875, 2003.
interleukin-6 production by alveolar macrophages. Exp Lung Res 19(3): 88. Saetta M, Di SA, Maestrelli P, Turato G, Mapp CE, Pieno M et al.
34559, 1993. Airway eosinophilia and expression of interleukin-5 protein in asthma
69. Nishikawa M, Kakemizu N, Ito T, Kudo M, Kaneko T, Suzuki M et al. and in exacerbations of chronic bronchitis [see comments]. Clin Exp
Superoxide mediates cigarette smoke-induced inltration of neutrophils Allergy 26(7): 76674, 1996.

340
Cytokines 27
89. Wedzicha JA, Seemungal TA, MacCallum PK, Paul EA, 106. Wang B, Cleary PP, Xu H, Li JD. Up-regulation of interleukin-8 by
Donaldson GC, Bhowmik A et al. Acute exacerbations of chronic novel small cytoplasmic molecules of nontypeable Haemophilus inu-
obstructive pulmonary disease are accompanied by elevations of plasma enzae via p38 and extracellular signal-regulated kinase pathways. Infect
brinogen and serum IL-6 levels. Thromb Haemost 84(2): 21015, Immun 71(10): 552330, 2003.
2000. 107. Nakamura H, Yoshimura K, McElvaney NG, Crystal RG. Neutrophil
90. Bocchino V, Bertorelli G, Bertrand CP, Ponath PD, Newman W, elastase in respiratory epithelial lining uid of individuals with cystic
Franco C et al. Eotaxin and CCR3 are up-regulated in exacerbations brosis induces interleukin-8 gene expression in a human bronchial
of chronic bronchitis. Allergy 57(1): 1722, 2002. epithelial cell line. J Clin Invest 89: 147884, 1992.
91. Khair OA, Devalia JL, Abdelaziz MM, Sapsford RJ, Tarraf H, 108. Mahler DA, Huang S, Tabrizi M, Bell GM. Ecacy and safety of a
Davies RJ. Eect of Haemophilus inuenzae endotoxin on the synthe- monoclonal antibody recognizing interleukin-8 in COPD: A pilot
sis of IL-6, IL-8, TNF-alpha and expression of ICAM-1 in cultured study. Chest 126(3): 92634, 2004.
human bronchial epithelial cells. Eur Respir J 7(12): 210916, 1994. 109. Howat WJ, Holgate ST, Lackie PM. TGF-beta isoform release and
92. Inoue H, Hara M, Massion PP, Grattan KM, Lausier JA, Chan B et al. activation during in vitro bronchial epithelial wound repair. Am J
Role of recruited neutrophils in interleukin-8 production in dog tra- Physiol Lung Cell Mol Physiol 282(1): L11523, 2002.
chea after stimulation with Pseudomonas in vivo. Am J Respir Cell Mol 110. Chambers RC, Leoni P, Kaminski N, Laurent GJ, Heller RA. Global
Biol 13(5): 57077, 1995. expression proling of broblast responses to transforming growth
93. Churg A, Dai J, Tai H, Xie C, Wright JL. Tumor necrosis factor-alpha factor-beta1 reveals the induction of inhibitor of dierentiation-1 and
is central to acute cigarette smoke-induced inammation and connec- provides evidence of smooth muscle cell phenotypic switching. Am J
tive tissue breakdown. Am J Respir Crit Care Med 166(6): 84954, 2002. Pathol 162(2): 53346, 2003.
94. Takeyama K, Dabbagh K, Lee HM, Agusti C, Lausier JA, Ueki IF 111. Goldsmith AM, Bentley JK, Zhou L, Jia Y, Bitar KN, Fingar DC et al.
et al. Epidermal growth factor system regulates mucin production in Transforming growth factor-beta induces airway smooth muscle hyper-
airways. Proc Natl Acad Sci USA 96(6): 308186, 1999. trophy. Am J Respir Cell Mol Biol 34(2): 24754, 2006.
95. Riise GC, Larsson S, Lofdahl CG, Andersson BA. Circulating cell 112. Eickelberg O, Kohler E, Reichenberger F, Bertschin S, Woodtli T,
adhesion molecules in bronchial lavage and serum in COPD patients Erne P et al. Extracellular matrix deposition by primary human lung
with chronic bronchitis. Eur Respir J 7(9): 167377, 1994. broblasts in response to TGF-beta1 and TGF-beta3. Am J Physiol
96. Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J et al. Macrophage 276(5 Pt 1): L81424, 1999.
metalloelastase mediates acute cigarette smoke-induced inammation 113. Coutts A, Chen G, Stephens N, Hirst S, Douglas D, Eichholtz T et al.
via tumor necrosis factor-alpha release. Am J Respir Crit Care Med Release of biologically active TGF-beta from airway smooth muscle
167(8): 108389, 2003. cells induces autocrine synthesis of collagen. Am J Physiol Lung Cell Mol
97. Di FM, Barbier D, Mege JL, Orehek J. Tumor necrosis factor-alpha Physiol 280(5): L9991008, 2001.
levels and weight loss in chronic obstructive pulmonary disease. Am J 114. Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M et al.
Respir Crit Care Med 150(5 Pt 1): 145355, 1994. Increased expression of transforming growth factor-beta1 in small
98. Schols AM, Buurman WA, Staal van den Brekel AJ, Dentener MA, airway epithelium from tobacco smokers and patients with chronic
Wouters EF. Evidence for a relation between metabolic derangements obstructive pulmonary disease (COPD). Am J Respir Crit Care Med
and increased levels of inammatory mediators in a subgroup of patients 163(6): 147683, 2001.
with chronic obstructive pulmonary disease. Thorax 51(8): 81924, 1996. 115. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo A
99. de GI, Donahoe M, Calhoun WJ, Mancino J, Rogers RM. Elevated et al. Transforming growth factor-beta expression in mucosal biopsies in
TNF-alpha production by peripheral blood monocytes of weight-losing asthma and chronic bronchitis. Am J Respir Crit Care Med 156(2 Pt 1):
COPD patients. Am J Respir Crit Care Med 153(2): 63337, 1996. 59199, 1997.
100. Reid MB, Lannergren J, Westerblad H. Respiratory and limb muscle 116. de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J,
weakness induced by tumor necrosis factor-alpha: Involvement of mus- Hiemstra PS et al. Transforming growth factor beta1 and recruitment
cle myolaments. Am J Respir Crit Care Med 166(4): 47984, 2002. of macrophages and mast cells in airways in chronic obstructive pul-
101. Li YP, Lecker SH, Chen Y, Waddell ID, Goldberg AL, Reid MB. monary disease. Am J Respir Crit Care Med 158(6): 195157, 1998.
TNF-alpha increases ubiquitin-conjugating activity in skeletal muscle 117. Vignola AM, Chanez P, Chiappara G, Merendino A, Zinnanti E,
by up-regulating UbcH2/E220k. FASEB J 17(9): 104857, 2003. Bousquet J et al. Release of transforming growth factor-beta (TGF-
102. Rennard SI, Fogarty C, Kelsen S, Long W, Ramsdell J, Allison J beta) and bronectin by alveolar macrophages in airway diseases. Clin
et al. The safety and ecacy of iniximab in moderate to severe chronic Exp Immunol 106(1): 11419, 1996.
obstructive pulmonary disease. Am J Respir Crit Care Med 175(9): 926 118. Kanazawa H, Asai K, Hirata K, Yoshikawa J. Possible eects of vascu-
34, 2007. lar endothelial growth factor in the pathogenesis of chronic obstruc-
103. Koto H, Mak JC, Haddad EB, Xu WB, Salmon M, Barnes PJ et al. tive pulmonary disease. Am J Med 114(5): 35458, 2003.
Mechanisms of impaired beta-adrenoceptor-induced airway relaxa- 119. Santos S, Peinado VI, Ramirez J, Morales-Blanhir J, Bastos R, Roca J
tion by interleukin-1beta in vivo in the rat. J Clin Invest 98(8): 1780 et al. Enhanced expression of vascular endothelial growth factor in pulmo-
87, 1996. nary arteries of smokers and patients with moderate chronic obstructive
104. Beeh KM, Kornmann O, Buhl R, Culpitt SV, Giembycz MA, pulmonary disease. Am J Respir Crit Care Med 167(9): 125056, 2003.
Barnes PJ. Neutrophil chemotactic activity of sputum from patients 120. Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD,
with COPD: Role of interleukin 8 and leukotriene B4. Chest 123(4): Abman S, Hirth PK et al. Inhibition of VEGF receptors causes lung
124047, 2003. cell apoptosis and emphysema. J Clin Invest 106(11): 131119, 2000.
105. Woolhouse IS, Bayley DL, Stockley RA. Sputum chemotactic activity 121. Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y,
in chronic obstructive pulmonary disease: Eect of alpha(1)-antitrypsin Salvemini D et al. Oxidative stress and apoptosis interact and cause
deciency and the role of leukotriene B(4) and interleukin 8. Thorax emphysema due to vascular endothelial growth factor receptor block-
57(8): 70914, 2002. ade. Am J Respir Cell Mol Biol 29(1): 8897, 2003.

341
Matrix Degrading Proteinases
in COPD and Asthma
28 CHAPTER

INTRODUCTION been derived (Table 28.1). Host serine protein- Steven D. Shapiro
ases associated with COPD and asthma largely
belong to the SA clan, S1(trypsin/chymot- Department of Medicine, University of
In chronic obstructive pulmonary disease rypsin) family. S1 serine proteinases are charac- Pittsburgh, Pittsburgh, PA, USA
(COPD), we now appreciate that cigarette terized by conserved His, Asp, and Ser residues
smoke sets into motion a complex inamma- that form a charge-relay system that functions
tory network leading to inammation, destruc- by transfer of electrons from the carboxyl group
tion, and when coupled with abnormal repair of Asp to the oxygen of Ser which then becomes
results in COPD. Elastolytic proteinases are a powerful nucleophile able to attack the car-
well established in the destructive phase. Recent bonyl carbon atom of the peptide bond of the
studies have focused on the roles of proteinases substrate. These enzymes are synthesized as pre-
in control of inammation and repair. With proenzymes in the endoplasmic reticulum and
respect to asthma, proteinases are prominently processed by cleavage of the signal peptide (pre-
expressed on many airway cells and also appear ) and removal of a dipeptide (pro-) by cathep-
be involved in controlling inammation as well sin C, and stored in granules as active packaged
as participating in other aspects of remodeling proteins. Distinct subsets of serine proteinases
such as subepithelial brosis, mucus metaplasia, are expressed in a lineage-restricted manner in
and smooth muscle hypertophy. This discussion immune and inammatory cells. Serine protein-
will be limited to host proteinases with poten- ases are also expressed in a developmentally spe-
tial extracellular matrix degrading capacity, cic manner. For example, neutrophil elastase
excluding intracellular cysteine proteinases, such (NE), proteinase 3, and cathepsin G are major
as caspases, that regulate cell death, and non- components of primary or azurophil granules
host serine proteinases of dust mites and other that are formed during a very specic stage dur-
allergens that may also play a role in asthma. ing the development of myeloid cells.
Neutrophil elastase (NE), cathepsin G, and
proteinase 3 are 30 kDa glycoprotein contain-
ing 20% neutral sugars. NE is a more potent
PROTEINASES elastase than the other serine proteinases and
has received the most attention in COPD.
NEs tertiary structure is similar to other
Serine proteinases chymotrypsin-like serine proteinases with two
interacting antiparallel -barrel cylindrical
Serine proteinases have diverged evolution- domains that form a crevice encompassing the
arily from a single gene product; as a result of catalytic triad [1]. NE prefers substrates with
duplications and mutations, enzymes with Val  Ala  Ser, Cys at the P1 position. NE
diverse biological functions including digestive has activity against a broad range of extracellu-
enzymes of exocrine glands, clotting factors, lar matrix proteins including the highly resistant
and leukocyte granule associated proteinases elastin. NE expression is localized to primary or
some of which degrade extracellular matrix pro- azurophil granules of neutrophils and a subset
teins and are of relevance to emphysema have of proinammatory monocytes (Fig. 28.1) [2].

343
Asthma and COPD: Basic Mechanisms and Clinical Management

Neutrophil 1-AT TIMP Macrophage Serine proteinase inhibitors


Serine proteinase inhibitors are abundant in the plasma.
Alpha-2 macroglobulin, a large protein usually restricted to
NE MMP-12
the bloodstream because of its mass, 725,000 kDa, inhibits
proteinases of several classes by entrapping proteinases
following cleavage of susceptible regions of the molecule.
At a concentration of 150350 mg/dl, 1-AT has the high-
chemotaxis chemotaxis
est concentration of the plasma inhibitors. 1-AT belongs
ECM to a family of serine proteinase inhibitors called the serpins.
ECM Serpins have considerable sequence homology, particularly
fragments around their reactive sites. They are important for homeos-
tasis, since they exert some control over such major proteo-
FIG. 28.1 Interactions between neutrophil and macrophage proteinases lytic cascades as the complement system and coagulation.
in emphysema. Neutrophil elastase (NE) originating from azurophil 1-AT is a 52 kDa glycoprotein synthesized primarily by
granules directly degrades elastin and indirectly augments elastolysis via the liver, consisting of a single polypeptide chain of 394
degradation of TIMP(s). Macrophage MMP-12 also directly degrades elastin amino acids. Proteolytic inhibition by 1-AT involves cleav-
and also indirectly augments NE activity by degrading its inhibitor 1-AT.
age of the strained reactive open center of 1-AT between
Extracellular matrix (ECM) fragments released during proteolysis play a role
in perpetuating inflammatory cell recruitment.
Met358 and Ser359, resulting in an altered, relaxed 1-AT
conformation in complex with the proteinase. Formation
of the complex renders the proteinase inactive and, because
the complex is quite stable, inactivation is essentially per-
manent. The association and inhibition of NE by 1-AT is
much faster than with other serine proteinases including
Studies from several laboratories have shown that NE trypsin, yet the name 1-antitrypsin is retained for histori-
is responsible for most of the degradative activity of neu- cal respect. 1-AT is the major inhibitor of serine protei-
trophils toward extracellular matrix structures. However, nases in the lower airspace. As discussed below, inherited
when optimally primed and stimulated by biologically rel- deciency of 1-AT represents the only genetic abnormal-
evant agonists, neutrophils release less than 2% of their ity to date associated with COPD.
content of NE freely into the extracellular space, and that Additional low molecular weight serine proteinase
they are able to translocate as much as 12% of their total inhibitors are abundant in airway uid and hence thought to
NE to the cell membrane, where it is catalytically active represent the primary defense against proteinase-mediated
and resistant to inhibitors [3]. The high concentration of airway damage. Secretory leukoprotease inhibitor (SLPI) is
NE (5 mM) within azurophil granules transiently over- a12 kDa protein produced by mucus-secreting and epithe-
whelm local NE inhibitors, resulting in a quantum burst lial cells in the airway as well as type 2 pneumocytes [10].
of obligate catalytic activity near the cell surface when each SLPI inhibits NE and cathepsin G and many other serine
granule is released [4]. These mechanisms allow NE to pro- proteinases, but not proteinase 3. Elan, also produced by
teolyze extracellular matrix (ECM) in a focused and pro- airway secretory and epithelial cells is released as a 12 kDa
tected manner while limiting widespread tissue destruction. precursor which is processed to a 6 kDa form that speci-
NE might also be freely released during frustrated phago- cally inhibits NE and proteinase 3 [11]. These inhibitors
cytosis, or by necrotic or apoptotic neutrophils if the lat- are able to inhibit NE bound to substrate giving them an
ter are not cleared eciently by macrophages. Regardless added dimension that 1-AT lacks. An additional serpin,
of the mechanisms by which active NE reaches ECM, it serpin B1 or monocyte/neutrophil elastase inhibitor, also
is relatively protected from inhibitors following binding has considerable activity against NE [12]. Airway mucus
to insoluble macromolecules, and NE is intimately associ- contains several other substances that partially inhibit NE
ated with elastic bers in emphysematous tissue [5]. During including polyanionic molecules such as mucins, other
bacterial exacerbations of COPD, overwhelming (micromo- glycosaminoglycans, and fatty acids. DNA, released from
lar) concentrations of active NE can be found as a result of inammatory leukocytes, binds to SLPI greatly enhancing
brisk neutrophil inux, and active NE can often be found its rate of association with NE. The relative contribution of
within the airways of patients with alpha-1-antitrypsin each of these molecules to proteinase inhibition is unknown.
(1-AT) deciency. NE is also a potent secretagogue and
induces mucin gene expression [6], which might aggravate
airow obstruction in COPD. Neutrophil inux and NE is Matrix metalloproteinases
also believed to be important in severe, steroid-resistant
asthma. Matrix metalloproteinases (MMPs) [13] comprise a fam-
NE decient mice have demonstrated a role for NE ily of 23 human matrix degrading enzymes believed to be
in killing gram-negative bacteria [7]. NE-mediated bac- essential for development and physiologic tissue remodeling
terial killing is related to proteolytic degradation of Omp and repair (Table 28.1). Abnormal expression of MMPs has
proteins on the outer wall of gram-negative bacteria [8]. been implicated in many destructive processes, including
Application of NE/ mice to cigarette smoke also demon- tumor cell invasion and angiogenesis, arthritis, atheroscle-
strates a role for NE in the development of emphysema as rosis, arterial aneurysms, and pulmonary emphysema [14].
discussed below [9]. MMPs are secreted as inactive proenzymes that are activated

344
Matrix Degrading Proteinases in COPD and Asthma 28
at the cell membrane surface or within the extracellular space many interesting vascular phenotypes, such as a requirement
by proteolytic cleavage of the N-terminal domain. Catalytic for MMP-9, for abdominal aortic aneurysms [28].
activity is dependent upon binding of a zinc ion at the active Macrophage elastase (MMP-12) is characterized by
site and is specically inhibited by members of another gene macrophage-specic expression and broad potent matrix
family, called tissue inhibitor of metalloproteinases (TIMPs) degrading capacity for an MMP [29]. MMP-12, like many
for tissue inhibitors of MMPs. Currently, four TIMPs have MMPs also has important non-matrix substrates. MMP-
been described. Optimal activity of MMPs is around pH 7.4. 12 is required for TNF- shedding following smoke expo-
MMP family members share 4050% identity at the amino sure [30], and MMP-12 to a greater degree than other
acid level, and they possess common structural domains. MMPs degrades and inactivates 1-AT [29], thus indirectly
Domains include a proenzyme domain that maintains enhancing the activity of NE.
the enzyme in its latent form, an active domain that coor- MMP-12 is predominantly a macrophage product
dinates binding of the catalytic zinc molecule, and (except and it has been found in macrophages in association with
for MMP-7) a C-terminal domain involved in substrate, cigarette smoke exposure and COPD. In fact, it was the
cell and TIMP binding. The gelatinases A and B (MMP-2 most highly upregulated gene by expression proling in
and MMP-9, respectively) have an additional bronectin- macrophages obtained from both murine and human smok-
like domain which mediates their high binding anity to ers [31]. MMP-12 has also been found in asthmatic lungs,
gelatins and elastin. MMP-9 has one more domain with and has even been detected in airway epithelial cells [32]
homology to type V collagen. Membrane-type MMPs and smooth muscle cells as well [33].
(MT1-6-MMP or MMP-14 to -19) have an additional C- Macrophages of MMP-12/ mice have a markedly
terminal membrane-spanning domain. diminished capacity to degrade extracellular matrix compo-
Individual members of the MMP family can be nents and are essentially unable to penetrate reconstituted
loosely divided into groups based on their matrix degrading basement membranes both in vitro and in vivo [34]. As
capacity. As a whole, they are able to cleave all extracellu- discussed below unlike wild-type (WT) mice, MMP-12-
lar matrix components. Those MMPs implicated in COPD decient mice were protected from macrophage accumu-
and asthma include the collagenases (MMPs -1, -8, -13) lation and the development of emphysema despite heavy
that have the unique capacity to cleave native triple heli- long-term smoke exposure [35]. MMP-12 appears to be
cal interstitial collagens but not elastin. MMP-1 is promi- important in inammatory cell recruitment in several murine
nently expressed in epithelial cells in patients with asthma models of asthma described below as well (Fig. 28.2).
and COPD [15]. However, mice do not have MMP-1 Membrane-type metalloproteinases (MT-MMPs) rep-
but only MMP-8 and -13. Gene targeting of MMP-8 has resent six MMPs that are localized at the cell surface and
shown that this neutrophil-derived proteinase is involved in at least one MT-MMP, MT1-MMP, activates MMP-2.
turning o neutrophil recruitment, likely via degradation of MT-MMPs also appear to directly degrade ECM proteins,
neutrophil chemokines [16]. MMP-13/ mice have devel- but their catalytic capacities is not well dened at present.
opmental bone defects [17]. MT1-MMP is among the MMPs associated with COPD
Gelatinases of 72 kDa (gelatinase A, MMP-2) and [36].
92 kDa (gelatinase B, MMP-9) dier in their cellular origin ADAMs (a disintegrin and metalloprotease domain) rep-
and regulation, but share the capacity to degrade gelatins resents a family of related metalloproteases that are bound
(denatured collagens), type IV collagen, elastin, and other to the cell surface. ADAM-33 was described as an asthma
matrix proteins. The gelatinases are by far the most thor- gene in humans following an extensive genetic linkage anal-
oughly studied members of the MMP family and MMP- ysis [37]. Subsequently, ADAM-33 has been replicated in
9 clearly the most thoroughly investigated in COPD and some but not all asthma populations, as well as in COPD.
asthma. Research focus to gelatinases is in part due to their The role of ADAM-33 is not clear, in fact it does not
importance in biology, but also reects the ease of detect- appear to be catalytically active due to splicing. Expressed
ing them by the exquisitely sensitive technique of gelatin by broblasts and smooth muscle cells, it might be involved
zymography in which the active moiety is detected by virtue in airway remodeling and hyperresponsiveness [38].
of its cleavage of an articial substrate.
In both COPD [18] and asthma [19, 20] many stud-
ies have found increased expression of MMP-9, often in Tissue inhibitors of metalloproteinases
association with decrease in its inhibitor TIMP-1, in spu- Tissue inhibitors of metalloproteinases (TIMPS) comprise a
tum, bronchoalveolar lavage (BAL), lung tissue, and cells family of four with molecular masses ranging between 21
obtained from asthmatic patients. For example, airway epi- (TIMP-2, nonglycosylated) and 27.5 (TIMP-1, glyco-
thelial cells produce MMP-9 upon injury and proportional sylated). Each TIMP inhibits MMPs via tight, non-covalent
to the severity of asthma [21]. Inammatory and immune binding with 1:1 stoichiometry. TIMP-1 binds to the C-
cells are a rich source of MMP-9 particularly eosinophils, terminal domain of MMPs, but how this leads to inhibi-
neutrophils, macrophages [22,23], and DCs [24,25]. tion of catalysis is unknown. Those MMPs that lack the
MMP-9/ mice are protected from airway subepithe- C-terminal domain, including MMP-7 and fully processed
lial brosis in models of asthma [26]. Yet, they have no role form of MMP-12 are still susceptible to TIMP inhibition
in cigarette smoke-induced airspace enlargement (Shapiro although with a lower Ki. TIMP-2 is secreted complexed to
unpublished). Other phenotypes of MMP-9/ mice include MMP-2 in broblasts. TIMP-2, not only inhibits MMP-2,
abnormal long bone development (through failed activa- but also is involved in docking pro-MMP-2 to the cell sur-
tion of vascular endothelial growth factor, VEGF) [27], and face where the enzyme is activated by MT1-MMP.

345
Asthma and COPD: Basic Mechanisms and Clinical Management

Smooth muscle
hypertrophy

- ADAM-33?

Asthma FIG. 28.2 Potential functions of proteinases in asthma.


Airway Subepithelial fibrosis Tryptase mediates bronchoconstriction and fibroblast
MMP-9 - MMP-9 activates proliferation, most likely via proteolytic activation of
TGF - protease activated receptor-2. MMPs have been linked to
both fibroblast proliferation (MMP-2) and collagenolysis
MMP-12 (MMP-1). MMP-9 might promote eosinophil transvascular
Inflammation migration. MMPs and related ADAMs (a disintegrin and
- MMp-12 Products metalloproteinase domain) might regulate cell survival
Eosinophil Macrophage
Eosinophil & Macrophag via shedding of surface molecules such as Fas, TNF/TNF
Migration receptor.

TIMPs are secreted from many cell types and are enzymes clearly have the capacity to cause lung destruction
abundant in tissues. For example, alveolar macrophages if targeted to the cell surface or extracellular space particu-
secrete both a variety of metalloproteinases and TIMP-1 larly in acidic microenvironments.
and TIMP-2. TIMP-3 is expressed predominantly by epi- The role of cysteine proteinases in COPD and asthma
thelial cells and binds to extracellular matrix and thus may remains speculative. However, cathepsin S was shown to
be important in preventing emphysema and in allowing be involved in the emphysematous process associated with
excess matrix accumulation in asthmatic airways. Expression IFN- transgenic mice [42].
of MMPs and TIMPs may depend on the inammatory
stimulus, MMPs, and TIMPs may be coordinately regu- Cysteine proteinase inhibitors
lated, perhaps to limit tissue injury during normal remod- Cysteine proteinases are inhibited by cystatins. Some cystatins
eling associated with inammation, or regulation may be are strictly intracellular, while others, such as cystatin C, pos-
discoordinate, potentially leading to tissue injury. sess a signal peptide and are secreted by a variety of cells into
the extracellular uid. Cystatin C, comprised of a single non-
glycosylated 120 amino acid peptide chain (13 kDa) forms
Cysteine proteinases reversible 1:1 complexes with enzymes in competition with
substrates. Cystatin C, the most ubiquitous cystatin, is found
Cysteine proteinases utilize the sulfhydryl group of Cys as in all human tissues and body uids tested, providing general
a nucleophile. Similar to serine proteinases a proton donor protection against tissue destruction by intracellular cathepsin
from His form a catalytic dyad (and in some cases triads) enzymes leaking from dying cells. Lack of cystatin C has been
required for endopeptidase activity (Table 28.1). Cysteine associated with destructive lesions in the vasculature [43].
proteinase represent a large, diverse group of plant and
animal enzymes with amino acid homology at the active
site only [39]. Human alveolar macrophages produce the
lysosomal thiol proteinases, cathepsins B, H, L, and S that
have been implicated in COPD [40]. CD4 cells express COPD
cathepsin S which functions in antigen processing [41] and
thus might prove to play a role in asthma . These enzymes COPD is currently dened by the Global Initiative for
share similar sizes of 2432 kDa and high mannose side Chronic Obstructive Lung Disease (GOLD) as a dis-
chains (typical of proteins targeted for lysosomal accumu- ease state characterized by exposure to a noxious agent
lation). Cathepsins B and H have little endopeptidase activ- resulting in airow limitation that is not fully reversible
ity and may function to activate other proteins similar to a (http://www.goldcopd.com/) [44]. Airow limitation may be
distant relative; interleukin converting enzyme. Cathepsin secondary to airway obstruction or alveolar destruction with
C or dipeptidyl peptidase I has limited extracellular matrix loss of elastic recoil. In fact, most patients have an admixture
degrading activity but is required for activation of nearly of large airway changes (accounting for symptoms of chronic
all matrix degrading serine proteinase proenzymes to their bronchitis), small airway changes, and parenchymal lung
active form. Cathepsins L and S have large active pock- involvement.
ets with relatively indiscriminate substrate specicities
that include elastin and other matrix components. These
enzymes have an acidic pH optima but cathepsin S retains Small airway disease
25% of its elastolytic capacity at neutral pH (making it
approximately equal to NE). Cathepsin K is a potent elastase The small airways (diameter 2 mm) are the major sites
predominantly expressed in osteoclasts but also by macro- of airway obstruction in COPD. Characteristic cellular
phages in the vasculature and perhaps other tissues. These changes leading to luminal narrowing include goblet cell

346
Matrix Degrading Proteinases in COPD and Asthma 28
TABLE 28.1 Main inflammatory cell matrix degrading proteinases in the lung.

Molecular Other cells expressing


Cell (major source) Proteinase Class mass *(kDa) Matrix substrates proteinase

Parenchyma
Neutrophil Neutrophil elastase Serine 2731 Elastin, bm components Proinflammatory
monocyte
Proteinase 3 Serine 2834 Elastin, bm components Monocyte
Mast cell
Cathepsin G Serine 2732 (elastin) bm components Monocyte
Mast cell
MMP-8 MMP #Pro-55 Interstitial collagens

MMP-9 MMP Pro-9295 Denatured collagens, types Macrophage


IV, V, and VII collagen Endothelial cell

Macrophage MMP-12 MMP Pro-54 Elastin, bm components


MMP-1 MMP Pro-55 Interstitial collagens Fibroblasts
Epithelial cells
Cathepsin L Cysteine 29 Elastin (at acidic pH)
Cathepsin S Cysteine 28 Elastin CD4 T-cell

*denotes (pre)proenzyme forms. basement membrane (bm) components include fibronectin, laminin, entactin, vitronectin, and type IV collagen (non-helical domains). #Pro,
molecular mass of proenzyme form.
Note: parentheses denote minor cellular sources.

metaplasia with excess mucus production, smooth muscle foundation of which was built upon two seminal observa-
hypertrophy, inltrating mononuclear inammatory cells, tions made over 35 years, one experimental and the other
and submucosal brosis. Surrounding lung parenchymal clinical. First, in 1963 Gross instilled papain into the lungs
elastin provides radial traction on bronchioles at points of rodents which resulted in emphysema [45]. Subsequently,
where alveolar septa attach. Hence, elastolysis may lead to investigators have instilled a variety of proteinases into ani-
loss of alveolar attachments with airway distortion and nar- mal lungs, yet only elastases result in emphysema. Second,
rowing in COPD. Destruction of small airways may directly in 1964 Laurel and Eriksson described ve patients with
contribute to an increase in ow resistance through destruc- deciency of 1-AT, the physiologic inhibitor of NE. Three
tion of parallel conducting pathways. of these initial subjects had emphysema [46].
The role of proteinases in small airways disease in Over time, there have been revisions to the elastase:
COPD is less well appreciated than in the lung paren- antielastase hypothesis, but the basic concept has stood the
chyma. Proteinase expression has been well studied in test of time remarkably well [47]. A current rendition of the
asthma (see below) where there may be some common pathogenesis of emphysema includes the following steps.
mechanisms of airway remodeling. One distinction is that First, cigarette smoke causes inammatory cell recruitment.
neutrophil inux is a hallmark of COPD, while prominent Next, these inammatory cells release matrix degrading
only in severe asthma. Neutrophil inux in COPD is likely proteinases that locally overwhelm or evade inhibitors, caus-
related directly to both cigarette smoke exposure and colo- ing destruction of lung elastin and other extracellular matrix
nization by pathogens later in the disease process. In addi- (ECM) proteins. In addition to loss of ECM, alveolar cells
tion to its elastolytic activity, NE is a potent secretagogue. die by apoptosis. Whether this is a primary or secondary
As in asthma, MMP-9 is expressed by several cell types in event remains a point of controversy. Finally, lung destruc-
the airway might paradoxically lead to increased, rather than tion coupled with failure to repair abnormal structures
decreased, collagen accumulation in the airway. Similarly, leads to airspace enlargement characteristic of emphysema.
despite collagenase-1 (MMP-1) expression by airway epi-
thelial cells [15], collagen accumulates in the airways, high-
lighting the complexity of matrix turnover. Inflammation and proteinases in
human COPD
Emphysema
A major achievement in our understanding of emphysema has
Emphysema is characterized by abnormal, permanent been our appreciation of the complex inammatory milieu
enlargement of airspaces distal to the terminal bronchiole, and the interrelationships between inammatory/immune
accompanied by destruction of their walls with coalescence cells leading to lung destruction [48]. Macrophages patrol the
into larger airspaces. The explanation for this pathol- lower airspace under normal conditions. Acutely following
ogy stems from the elastase:antielastase hypothesis, the cigarette smoke exposure, macrophages may become activated

347
Asthma and COPD: Basic Mechanisms and Clinical Management

and neutrophils quickly arrive. Subacutely, macrophages accu- lung destruction. These concepts were conrmed in a mouse
mulate in respiratory bronchioles. Chronically, macrophages, model where cigarette exposure to mice decient in CD8
neutrophils, and CD8  CD4 T-cells as well as B-cells T-cells fails to produce IP-10, activate/recruit macrophages,
accumulate in the lung. Moreover, loss of cilia predisposes to produce MMP-12, or develop airspace enlargement [61].
airway infection with a prominent neutrophilic response.
While cigarette smoke initiates the inammation and
destruction in emphysema, in severe disease inammation Emphysema associated with 1-AT
is sustained by factors other than cigarette smoke. Indeed deficiency
intense inammation with many cell types was observed in
lung tissue taken from patients with end-stage lung COPD The clearest example of the association of proteinase
undergoing volume reduction surgery, who had discontin- antiproteinase imbalance and emphysema occurs with
ued smoking on average 9 years earlier [49]. inherited deciency of 1-AT. Several abnormal 1-AT
A more careful examination of inammatory cell alleles are associated with very low serum concentrations
interrelationships demonstrates that proteinases are not of 1-AT and enhanced risk for emphysema. Of these, the
only involved in lung destruction, but also in regula- Pi Z variant is by far the most common, and greater than
tion of inammation. Upon initial exposure to cigarette 95% of 1-AT decient individuals have only the Z vari-
smoke, constitutive macrophages secrete MMP-12 which ant detectable. A variety of rare deciency variants, includ-
is responsible for shedding of active TNF- from the ing non-expressing alleles that do not result in detectable
cell surface and subsequent recruitment of neutrophils 1-AT in plasma, comprise the remaining individuals. Pi Z
in response to cigarette smoke in mice [50]. In addition, individuals have about 15% of the normal serum concentra-
cigarette smoke related oxidant activity, inactivates mac- tion of 1-AT. The abnormality leading to the Pi Z variant
rophage histone deacetylase-2 promoting transcription of is a point mutation involving a single nucleotide at codon
MMPs and neutrophil chemokines including IL-8 [51]. 342 that results in coding for Lys instead of Glx. This amino
Both MMP-12 and neutrophil NE contribute to lung acid substitution changes the charge attraction between the
destruction in murine models of cigarette smoke-induced amino acids at positions 342 and 290 present in the nor-
emphysema with MMP-12/ mice being fully protected mal form of 1-AT and prevents the formation of a fold
and NE/ mice being two-third protected from airspace in the molecule. With this change in tertiary structure, the
enlargement [9]. These two proteolytic systems interact to molecule is susceptible to loop sheet polymerization of
achieve maximal lung damage. NE degrades TIMP-1 aug- 1-AT in the endoplasmic reticulum that impedes secretion
menting MMP activity [52] and MMPs degrade 1-AT, of the protein from the hepatocyte [62]. In addition, its rate
indirectly augmenting NE activity [53]. of association with NE is slightly but signicantly slower
Elastin degradation products generated by these than the association rate of normal 1-AT with NE. The
enzymes themselves appear to be a critical chemokine in prevalence of the Pi Z phenotype in the United States is
further macrophage accumulation in COPD [54]. Hence, about one in 2800 people [63]. The Z allele is not found in
a positive feedback loop is established between elastoly- Asians and African populations at measurable frequencies.
sis, macrophage accumulation, and neutrophil recruitment. Most Pi Z individuals eventually become sympto-
Most recently, elastin fragments have also been implicated matic with COPD, but there is considerable variation and
an auto-immune target in COPD [55]. Interest in B-cells some individuals reach advanced age with minimal symp-
in COPD has grown with the appreciation that the amount toms. Smoking has a marked eect on the age at which
of BALT (bronchial associated lymphoid tissue) correlates shortness of breath appears. On the average Pi Z smokers
with the severity of COPD [56]. BALT could simply be have symptoms by age 40, about 15 years earlier than Pi Z
associated with airway remodeling and bacterial coloniza- nonsmokers [63]. Additional genetic modiers, such as pol-
tion. However, patients with COPD have been show to ymorphisms in IL-10 [64], also help explain dierences in
develop antibodies to elastin and this might be involved the clinical course of patients with 1-AT deciency.
in disease progression [55]. Hence, these new wrinkles
continue to strengthen the elastase:antielastase hypoth-
esis demonstrating that not only elastin is critical to the
structure and function of the lung, but by-products of its
dissolution are both proinammatory and may induce auto-
ASTHMA
immune disease.
Currently, focus has shifted to understanding the role Human asthma is characterized by persistent airway hyper-
of adaptive immunity in COPD, particularly the CD8 responsiveness (AHR) and episodic airow obstruction.
T-cell. CD8 T-cells, are known to be increased in lungs Asthma is also associated with an immune/inammatory
of patients with COPD [57]. Transgenic mice overexpress- response characterized by activated T-cells with Th2 cytokines
ing IFN- develop inammation, apoptosis, and consequent (IL-4, -5, -9, -13), eosinophils, and mast cells that inltrate
emphysema [58]. Humans with COPD have increased the mucosa and submucosa. The AHR may be a distinct
amounts of IP-10 and its receptor CXCR3 [59]. Induction trait, separate and preceding the inammation. While several
of IP-10 in CD8 T-cells and bronchiolar epithelium signals proteinases, particularly MMPs have been associated with
through CXCR3 expressed on alveolar macrophages to pro- asthma, their role in disease pathogenesis is still emerging.
duce MMP-12 [60]. As discussed above, MMP-12 produc- Proteinases might be particularly important in controlling
tion leads to elastolysis and sets in motion a cycle of further inammatory cell migration and airway remodeling.

348
Matrix Degrading Proteinases in COPD and Asthma 28
Initiation of asthma OVA 10 days in the context of adenovirus-mediated
GMCSF [71], resulted in a Th2 cytokine prole, OVA-
The initiation of asthma is driven, in large part, by stimu- specic IgE, and airway eosinophilia. In WT mice, MMP-
lation of allergen-specic CD4 T helper cells by profes- 12 mRNA and protein were upregulated 70-fold in OVA
sional antigen presenting cells (APCs), most notably lung treated versus saline controls. MMP-12/ mice had an 80%
dendritic cells (DCs). DCs play a prominent role in the reduction in eosinophil accumulation in BAL compared to
pathogenesis of asthma because they are able to eciently WT mice. It is noteworthy that, MMP-12 production was
present major histocompatibility complex (MHC) class IL-13 dependent, since it was abrogated in similar experi-
II-restricted antigenic determinants to nave T-cells to ments using IL-13/ mice.
direct CD4 T-cell polarization to promote a Th2-type
immune response within the lung.
MMP-9 is expressed in DCs and has been shown Airway remodeling
to be promote DC tracking [24] lending support for its
potential importance in the initiation of asthma. Cathepsin Inammation is generally thought to drive other aspects of
S, a potent matrix degrading cysteine proteinase in its own airway remodeling. For example, eosinophils were shown
right, has been shown to be required for normal MHC class to be required for brosis and smooth muscle hypertrophy,
II tracking and function within DCs [65]. Pharmacological but not AHR or mucus production [72]. In turn, smooth
inhibition of cathepsin S attenuated lung eosinophilia and muscle changes might also regulate inammation [73].
IgE generation in a mouse model of asthma [41]. Alternatively, many changes related to airway remodeling
have been replicated by pathological stretch applied purely
to airway epithelial cells (i.e. in the absence of inamma-
Asthma progression tion) [74]. Expression proling, followed by conrmative
studies demonstrated that mechanical deformation of air-
Chronic asthma is characterized by airway remodeling, way epithelial cells induces production of MMP-9 as well
a term that incorporates chronic airway inammation, as its activator plasmin(ogen) [75].
marked smooth muscle hyperplasia (with mast cell inltra- MMP-9 has been implicated in subepithelial air-
tion), mucus gland and goblet cell hyperplasia, myobroblast way brosis. The IL-13 transgenic mice described above
accumulation with subepithelial brosis, and angiogenesis. also have features of airway remodeling including mucus
cell hyperplasia and subepithelial brosis [76]. The airway
changes were attenuated upon back-crossing to MMP-
Inflammation 9/ mice [26]. This appears to work through the capacity
of MMP-9 to activate TGF- promoting collagen produc-
Although MMP-9 is expressed in a variety of inammatory tion. NE also has been shown to activate latent TGF- in a
cells including eosinophils, macrophages, and neutrophils, model of bleomycin-induced lung brosis. It is noteworthy
the role of MMP-9 deciency in asthma pathogenesis has that, NE also induces epithelial cell production of MUC5
been confusing to date. Application of MMP-9/ mice [77] and mucus cell metaplasia in mouse lungs [78]. Hence,
to acute allergic/inammatory models have reported both in addition to the local concentration of proteinases and
proinammatory [66] and anti-inammatory roles [67,68]. their inhibitors, additional non-matrix substrates will inu-
It is likely that depending upon the protocol, amount of ence the eect of a proteinase is turnover.
lipopolysaccharide (LPS) and other conditions, one might
suppress or exacerbate inammation in asthma. Future stud-
ies will require control for these factors. Moreover, the role
of MMPs in newer models of chronic asthma with repeti-
References
tive antigen stimulation in susceptible strains will allow us
1. Bode W, Meyer EJ, Powers JC. Human leukocyte elastase: X-ray crys-
to establish roles for MMPs in models of chronic asthma,
tal structures, mechanism, substrate specicity, and mechanism-based
with potential human applicability. inhibitors. Biochemistry 28: 195163, 1989.
Application of MMP-12/ mice to several models 2. Owen CA, Campbell MA, Boukedes SS, Stockley RA, Campbell EJ.
of asthma has established the importance of the enzyme A discrete subpopulation of human monocytes expresses a neutrophil-
in inammatory cell migration in asthma. For example, like proinammatory (P) phenotype. Am J Physiol 267: 77585, 1994.
IL-13 inducible transgenic mice, described above, develop 3. Owen CA, Campbell MA, Sannes PL, Boukedes SS, Campbell EJ. Cell
both inammation and emphysema. Inammation and surface-bound elastase and cathepsin G on human neutrophils: A novel,
emphysema were dependent upon MMP-12 [69]. In addi- non-oxidative mechanism by which neutrophils focus and preserve cata-
tion, in mice sensitized and challenged with cockroach lytic activity of serine proteinases. J Cell Biol 131(3): 77589, 1995.
antigen (CRA) [70], MMP-12 deciency resulted in fewer 4. Campbell EJ, Campbell MA, Boukedes SS, Owen CA. Quantum pro-
teolysis by neutrophils: Implications for pulmonary emphysema in alpha
peribronchial leukocytes and correspondingly less protein
1-antitrypsin deciency. J Clin Invest 104(3): 33744, 1999.
and fewer cells in BAL compared to WT mice. Eosinophils 5. Damiano VV, Tsang A, Kucich U, Abrams WR, Rosenbloom J, Kimbel P,
were reduced to the greatest degree (80% reduction). In Fallahnejed M, Weinbaum G. Immunolocalization of elastase in human
addition, there were marked decreases in IL-5, MIP-1, emphysematous lungs. J Clin Invest 78: 48293, 1986.
MCP-1, and TNF- in MMP-12/ BAL. AHR did not 6. Fahy JV, Schuster A, Ueki I, Boushey HA, Nadel JA. Mucus hypersecre-
dier between groups. Another model of acute allergic air- tion in bronchiectasis. The role of neutrophil proteases. Am Rev Respir
way inammation that involved sensitization by aerosolized Dis 146(6): 143033, 1992.

349
Asthma and COPD: Basic Mechanisms and Clinical Management

7. Belaaouaj A, McCarthy R, Baumann M, Gao Z, Ley TJ, Abraham SN, Interleukin-13 induces tissue brosis by selectively stimulating and
Shapiro SD. Impaired host defense against bacterial infection in mice activating transforming growth factor beta(1). J Exp Med 194(6):
lacking neutrophil elastase. Nat Med, 1998. 80921, 2001.
8. Belaaouaj A, Kim KS, Shapiro SD. Degradation of outer membrane 27. Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA, Hanahan D,
protein A in Escherichia coli killing by neutrophil elastase. Science Shapiro SD, Senior RM, Werb Z. MMP-9/gelatinase B is a key
289(5482): 118588, 2000. regulator of growth plate angiogenesis and apoptosis of hypertrophic
9. Shapiro SD, Goldstein NM, Houghton AM, Kobayashi DK, Kelley D, chondrocytes. Cell 93(3): 41122, 1998.
Belaaouaj A. Neutrophil elastase contributes to cigarette smoke- 28. Pyo R, Lee JK, Shipley JM, Curci JA, Mao D, Ziporin SJ, Ennis TL,
induced emphysema in mice. Am J Pathol 163(6): 232935, 2003. Shapiro SD, Senior RM, Thompson RW. Targeted gene disruption
10. Sallenave JM. The role of secretory leukocyte proteinase inhibitor and of matrix metalloproteinase-9 (gelatinase B) suppresses development
elan (elastase-specic inhibitor/skin-derived antileukoprotease) as of experimental abdominal aortic aneurysms. J Clin Invest 105(11):
alarm antiproteinases in inammatory lung disease. Respir Res 1(2): 164149, 2000.
8792, 2000. 29. Gronski TJ, Martin R, Kobayashi DK, Walsh BC, Holman MC, Van
11. Sallenave JM, Silva A. Characterization and gene sequence of the pre- Wart E, Shapiro SD. Hydrolysis of a broad spectrum of extracellu-
cursor of elan, an elastase-specic inhibitor in bronchial secretions. lar matrix proteins by human macrophage elastase. J Biol Chem 272:
Am J Respir Cell Mol Biol 8(4): 43945, 1993. 1218994, 1997.
12. Cooley J, Takayama TK, Shapiro SD, Schechter NM, Remold- 30. Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, Shapiro SD,
ODonnell E. The serpin MNEI inhibits elastase-like and chymotrypsin- Wright JL. Macrophage metalloelastase mediates acute cigarette
like serine proteases through ecient reactions at two active sites. smoke-induced inammation via tumor necrosis factor-alpha release.
Biochemistry 40(51): 1576270, 2001. Am J Respir Crit Care Med 167(8): 108389, 2003.
13. Parks WC, Shapiro SD. Matrix metalloproteinases in lung biology. 31. Woodru PG, Koth LL, Yang YH, Rodriguez MW, Favoreto S,
Respir Res 2(1): 1019, 2001. Dolganov GM, Paquet AC, Erle DJ. A distinctive alveolar macrophage
14. Shapiro SD. Matrix metalloproteinase degradation of extracellular activation state induced by cigarette smoking. Am J Respir Crit Care
matrix: Biological consequences. Curr Opin Cell Biol 10(5): 6028, Med 172(11): 138392, 2005.
1998. 32. Lavigne MC, Thakker P, Gunn J, Wong A, Miyashiro JS,
15. Imai K, Dalal SS, Chen ES, Downey R, Schulman LL, Ginsburg M, Wasserman AM, Wei SQ, Pelker JW, Kobayashi M, Eppihimer MJ.
DArmiento J. Human collagenase (matrix metalloproteinase-1) Human bronchial epithelial cells express and secrete MMP-12. Biochem
expression in the lungs of patients with emphysema. Am J Respir Crit Biophys Res Commun 324(2): 53446, 2004.
Care Med 163(3 Pt 1): 78691, 2001. 33. Xie S, Issa R, Sukkar MB, Oltmanns U, Bhavsar PK, Papi A,
16. Balbin M, Fueyo A, Tester AM, Pendas AM, Pitiot AS, Astudillo A, Caramori G, Adcock I, Chung KF. Induction and regulation of matrix
Overall CM, Shapiro SD, Lopez-Otin C. Loss of collagenase-2 con- metalloproteinase-12 in human airway smooth muscle cells. Respir Res
fers increased skin tumor susceptibility to male mice. Nat Genet 35(3): 6: 148, 2005.
25257, 2003. 34. Shipley JM, Wesselschmidt RL, Kobayashi DK, Ley TJ, Shapiro SD.
17. Deguchi JO, Aikawa E, Libby P, Vachon JR, Inada M, Krane SM, Metalloelastase is required for macrophage-mediated proteolysis and
Whittaker P, Aikawa M. Matrix metalloproteinase-13/collagenase-3 matrix invasion in mice. Proc Natl Acad Sci 93: 394246, 1996.
deletion promotes collagen accumulation and organization in mouse 35. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD. Macrophage
atherosclerotic plaques. Circulation 112(17): 270815, 2005. elastase is required for cigarette smoke-induced emphysema in mice.
18. Atkinson JJ, Senior RM. Matrix metalloproteinase-9 in lung remod- Science 277: 20024, 1997.
eling. Am J Respir Cell Mol Biol 28(1): 1224, 2003. 36. Ohnishi K, Takagi M, Kurokawa Y, Satomi S, Konttinen YT. Matrix
19. Cundall M, Sun Y, Miranda C, Trudeau JB, Barnes S, Wenzel SE. metalloproteinase-mediated extracellular matrix protein degradation in
Neutrophil-derived matrix metalloproteinase-9 is increased in severe human pulmonary emphysema. Lab Invest 78(9): 107787, 1998.
asthma and poorly inhibited by glucocorticoids. J Allergy Clin Immunol 37. Van Eerdewegh P, Little RD, Dupuis J, Del Mastro RG, Falls K,
112(6): 106471, 2003. Simon J, Torrey D, Pandit S, McKenny J, Braunschweiger K, Walsh A,
20. Wenzel SE, Balzar S, Cundall M, Chu HW. Subepithelial base- Liu Z, Hayward B, Folz C, Manning SP, Bawa A, Saracino L,
ment membrane immunoreactivity for matrix metalloproteinase 9: Thackston M, Benchekroun Y, Capparell N, Wang M, Adair R,
Association with asthma severity, neutrophilic inammation, and Feng Y, Dubois J, FitzGerald MG, Huang H, Gibson R, Allen KM,
wound repair. J Allergy Clin Immunol 111(6): 134552, 2003. Pedan A, Danzig MR, Umland SP, Egan RW, Cuss FM, Rorke S,
21. Mattos W, Lim S, Russell R, Jatakanon A, Chung KF, Barnes PJ. Clough JB, Holloway JW, Holgate ST, Keith TP. Association of the
Matrix metalloproteinase-9 expression in asthma: Eect of asthma ADAM33 gene with asthma and bronchial hyperresponsiveness.
severity, allergen challenge, and inhaled corticosteroids. Chest 122(5): Nature 418(6896): 42630, 2002.
154352, 2002. 38. Holgate ST, Yang Y, Haitchi HM, Powell RM, Holloway JW,
22. Shapiro SD. Biological consequences of extracellular matrix cleavage Yoshisue H, Pang YY, Cakebread J, Davies DE. The genetics of asthma:
by matrix metalloproteinases. In Extracellular matrix and cell-to-cell ADAM33 as an example of a susceptibility gene. Proc Am Thorac Soc
contact. Curr Opin Cell Biol 10: 6028, 1998. 3(5): 44043, 2006.
23. Parks W. Matrix metalloproteinases. San Diego, CA: Academic Press, 39. Kung TT, Jones H, Adams GK, Umland SP, Kreutner W, Egan RW,
1998. Chapman RW, Watnick AS. Characterization of a murine model of aller-
24. Ichiyasu H, McCormack JM, McCarthy KM, Dombkowski D, gic pulmonary inammation. Int Arch Allergy Immunol 105: 8390, 1994.
Preer FI, Schneeberger EE. Matrix metalloproteinase-9-decient 40. Chapman HA, Riese RJ, Shi GP. Emerging roles for cysteine proteases
dendritic cells have impaired migration through tracheal epithelial in human biology. Annu Rev Physiol 59: 6388, 1997.
tight junctions. Am J Respir Cell Mol Biol 30(6): 76170, 2004. 41. Riese RJ, Mitchell RN, Villadangos JA, Shi GP, Palmer JT, Karp ER,
25. Vermaelen KY, Cataldo D, Tournoy K, Maes T, Dhulst A, Louis R, De Sanctis GT, Ploegh HL, Chapman HA. Cathepsin S activity regu-
Foidart JM, Noel A, Pauwels R. Matrix metalloproteinase-9-mediated lates antigen presentation and immunity. J Clin Invest 101(11): 235163,
dendritic cell recruitment into the airways is a critical step in a mouse 1998.
model of asthma. J Immunol 171(2): 101622, 2003. 42. Zheng T, Kang MJ, Crothers K, Zhu Z, Liu W, Lee CG, Rabach LA,
26. Lee CG, Homer RJ, Zhu Z, Lanone S, Wang X, Koteliansky V, Chapman HA, Homer RJ, Aldous D, De Sanctis GT, Underwood S,
Shipley JM, Gotwals P, Noble P, Chen Q , Senior RM, Elias JA. Graupe M, Flavell RA, Schmidt JA, Elias JA. Role of cathepsin

350
Matrix Degrading Proteinases in COPD and Asthma 28
S-dependent epithelial cell apoptosis in IFN-gamma-induced alveolar obstructive pulmonary disease. Am J Respir Crit Care Med 165(10):
remodeling and pulmonary emphysema. J Immunol 174(12): 810615, 14049, 2002.
2005. 60. Grumelli S, Corry DB, Song LZ, Song L, Green L, Huh J, Hacken J,
43. Shi GP, Sukhova GK, Grubb A, Ducharme A, Rhode LH, Lee RT, Espada R, Bag R, Lewis DE, Kheradmand F. An immune basis for
Ridker PM, Libby P, Chapman HA. Cystatin C deciency in human lung parenchymal destruction in chronic obstructive pulmonary disease
atherosclerosis and aortic aneurysms. J Clin Invest 104(9): 119197, and emphysema. PLoS Med 1(1): e8, 2004.
1999. 61. Maeno T, Houghton AM, Quintero PA, Grumelli S, Owen CA,
44. Pauwels R, Buist A, Calverley P, Jenkins C, Hurd S. Global strategy Shapiro SD. CD8 T Cells are required for inammation and
for the diagnosis, management, and prevention of chronic obstruc- destruction in cigarette smoke-induced emphysema in mice. J Immunol
tive pulmonary disease. NHLBI/WHO Global Initiative for Chronic 178(12): 809096, 2007.
Obstructive Lung Disease (GOLD) Workshop summary. Am J Respir 62. Lomas DA, Evans DL, Stone SR, Chang W-SW, Carre RW. Eect of
Crit Care Med 163: 125676, 2001. the Z mutation on the physical and inhibitory properties of alpha-1-
45. Gross P, Ptzer E, Tolker E, Babyak M, Kaschak M. Experimental antitrypsin. Biochemistry 32: 5008, 1993.
emphysema: its production with papain in normal and silicotic rats. 63. Silverman EK, Pierce JA, Province MA, Rao DC, Campbell EJ.
Arch Environ Health 11: 5058, 1965. Variability of pulmonary function in alpha-1-antitrypsin deciency:
46. Laurell. C. B. a. S. E. The electrophoretic alpha-globulin pattern of Clinical correlates. Ann Intern Med 111(12): 98291, 1989.
serum in alpha-antitrypsin deciency. Scand J Clin Invest 15: 13240, 64. Demeo DL, Campbell EJ, Barker AF, Brantly ML, Eden E,
1963. McElvaney NG, Rennard SI, Sandhaus RA, Stocks JM, Stoller JK,
47. Shapiro SD, Ingenito EP. The pathogenesis of chronic obstructive pul- Strange C, Turino G, Silverman EK. IL10 polymorphisms are associ-
monary disease: Advances in the past 100 years. Am J Respir Cell Mol ated with airow obstruction in severe alpha 1-antitrypsin deciency.
Biol 32(5): 36772, 2005. Am J Respir Cell Mol Biol 38(1): 11420, 2008.
48. Shapiro SD. COPD unwound. N Engl J Med 352(19): 201619, 65. Riese RJ, Wolf PR, Bromme D, Natkin LR, Villadangos JA, Ploegh HL,
2005. Chapman HA. Essential role for cathepsin S in MHC class II-associated
49. Retamales I, Elliott WM, Meshi B, Coxson HO, Pare PD, Sciurba FC, invariant chain processing and peptide loading. Immunity 4(4): 35766,
Rogers RM, Hayashi S, Hogg JC. Amplication of inammation in 1996.
emphysema and its association with latent adenoviral infection. Am J 66. Cataldo DD, Tournoy KG, Vermaelen K, Munaut C, Foidart JM,
Respir Crit Care Med 164(3): 46973, 2001. Louis R, Noel A, Pauwels RA. Matrix metalloproteinase-9 deciency
50. Churg A, Dai J, Zay K, Karsan A, Hendricks R, Yee C, Martin R, impairs cellular inltration and bronchial hyperresponsiveness during
MacKenzie R, Xie C, Zhang L, Shapiro S, Wright JL. Alpha-1- allergen-induced airway inammation. Am J Pathol 161(2): 49198, 2002.
antitrypsin and a broad spectrum metalloprotease inhibitor, RS113456, 67. McMillan SJ, Kearley J, Campbell JD, Zhu XW, Larbi KY, Shipley JM,
have similar acute anti-inammatory eects. Lab Invest 81(8): 111931, Senior RM, Nourshargh S, Lloyd CM. Matrix metalloproteinase-9
2001. deciency results in enhanced allergen-induced airway inammation.
51. Tomita K, Barnes PJ, Adcock IM. The eect of oxidative stress on his- J Immunol 172(4): 258694, 2004.
tone acetylation and IL-8 release. Biochem Biophys Res Commun 301(2): 68. Greenlee KJ, Corry DB, Engler DA, Matsunami RK, Tessier P,
57277, 2003. Cook RG, Werb Z, Kheradmand F. Proteomic identication of in vivo
52. Itoh Y, Nagase H. Preferential inactivation of tissue inhibitor of metal- substrates for matrix metalloproteinases 2 and 9 reveals a mechanism
loproteinases-1 that is bound to the precursor of matrix metallopro- for resolution of inammation. J Immunol 177(10): 731221, 2006.
teinase 9 (progelatinase B) by human neutrophil elastase. J Biol Chem 69. Zheng T, Zhu Z, Wang Z, Homer RJ, Ma B, Riese RJ Jr., Chapman HA
270(28): 1651821, 1995. Jr., Shapiro SD, Elias JA. Inducible targeting of IL-13 to the adult lung
53. Desrochers PE, Je rey JJ, Weiss SJ. Interstitial collagenase (matrix causes matrix metalloproteinase- and cathepsin-dependent emphy-
metalloproteinase-1) expresses serpinase activity. J Clin Invest 87(6): sema. J Clin Invest 106(9): 108193, 2000.
225865, 1991. 70. Warner RL, Lukacs NW, Shapiro SD, Bhagarvathula N, Nerusu KC,
54. Houghton AM, Quintero PA, Perkins DL, Kobayashi DK, Kelley DG, Varani J, Johnson KJ. Role of metalloelastase in a model of allergic lung
Marconcini LA, Mecham RP, Senior RM, Shapiro SD. Elastin frag- responses induced by cockroach allergen. Am J Pathol 165(6): 192130,
ments drive disease progression in a murine model of emphysema. 2004.
J Clin Invest 116(3): 75359, 2006. 71. Pouladi MA, Robbins CS, Swirski FK, Cundall M, McKenzie AN,
55. Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S, Jordana M, Shapiro SD, Stampi MR. Interleukin-13-dependent
Green L, Hacken-Bitar J, Huh J, Bakaeen F, Coxson HO, Cogswell S, expression of matrix metalloproteinase-12 is required for the develop-
Storness-Bliss C, Corry DB, Kheradmand F. Antielastin autoimmu- ment of airway eosinophilia in mice. Am J Respir Cell Mol Biol 30(1):
nity in tobacco smoking-induced emphysema. Nat Med 13(5): 56769, 8490, 2004.
2007. 72. Humble A, Lloyd C, McMillan S, Friend D, Zanthou G, McKenna E,
56. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Ghiran S, Gerard N, Yu C, Orkin S, Gerard C. A critical role for eosi-
Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The nophils in allergic airways remodeling. Science 305: 177679, 2004.
nature of small-airway obstruction in chronic obstructive pulmonary 73. Lazaar A, Panettieri RJ. Airway smooth muscle: A modulator of airway
disease. N Engl J Med 350(26): 264553, 2004. remodeling in asthma. J Allergy Clin Immunol 116: 48895, 2005.
57. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L, Mapp CE, 74. Tschumperlin DJ, Shively JD, Kikuchi T, Drazen JM. Mechanical
Maestrelli P, Ciaccia A, Fabbri LM. CD8 T-lymphocytes in periph- stress triggers selective release of brotic mediators from bronchial epi-
eral airways of smokers with chronic obstructive pulmonary disease. thelium. Am J Respir Cell Mol Biol 28(2): 14249, 2003.
Am J Respir Crit Care Med 157(3 Pt 1): 82226, 1998. 75. Chu EK, Cheng J, Foley JS, Mecham BH, Owen CA, Haley KJ,
58. Wang Z, Zheng T, Zhu Z, Homer RJ, Riese RJ, Chapman HA Jr., Mariani TJ, Kohane IS, Tschumperlin DJ, Drazen JM. Induction of
Shapiro SD, Elias JA. Interferon gamma induction of pulmonary emphy- the plasminogen activator system by mechanical stimulation of human
sema in the adult murine lung. J Exp Med 192(11): 1587600, 2000. bronchial epithelial cells. Am J Respir Cell Mol Biol 35(6): 62838,
59. Saetta M, Mariani M, Panina-Bordignon P, Turato G, Buonsanti C, 2006.
Baraldo S, Bellettato CM, Papi A, Corbetta L, Zuin R, Sinigaglia F, 76. Cho JY, Miller M, McElwain K, McElwain S, Shim JY, Raz E,
Fabbri LM. Increased expression of the chemokine receptor CXCR3 Broide DH. Remodeling associated expression of matrix metallo-
and its ligand CXCL10 in peripheral airways of smokers with chronic proteinase 9 but not tissue inhibitor of metalloproteinase 1 in airway

351
Asthma and COPD: Basic Mechanisms and Clinical Management

epithelium: Modulation by immunostimulatory DNA. J Allergy Clin 78. Voynow JA, Fischer BM, Malarkey DE, Burch LH, Wong T,
Immunol 117(3): 61825, 2006. Longphre M, Ho SB, Foster WM. Neutrophil elastase induces mucus
77. Voynow JA, Young LR, Wang Y, Horger T, Rose MC, Fischer BM. cell metaplasia in mouse lung. Am J Physiol Lung Cell Mol Physiol
Neutrophil elastase increases MUC5AC mRNA and protein 287(6): L1293302, 2004.
expression in respiratory epithelial cells. Am J Physiol 276(5 Pt 1):
L83543, 1999.

352
Growth Factors
29CHAPTER

INTRODUCTION acute inammation, such as in ARDS, or more Martin Kolb, Zhou Xing,
chronic inammation states such as COPD and
usual interstitial pneumonia (UIP) are usually
Kjetil Ask and Jack
It has long been recognized that after acute injury associated with the sequelae of tissue remodeling. Gauldie
to the lung, the tissue repair process is engaged to However, there may be situations involving struc- Centre for Gene Therapeutics,
return the organ to normal function. In chronic tural cell phenotype or extra cellular matrix alter-
Department of Pathology and Molecular
tissue injury, it appears the process of repair loses ations that can propagate the remodeling process
Medicine, McMaster University
many of the control mechanisms, and continued through autocrine and paracrine pathways inde-
pendent of the state of inammation that may Hamilton, ON, Canada
repair results in remodeling of the tissue with
alteration of normal structure and compromise have preceded the alteration. Thus, identica-
of normal lung function. The remodeling process tion of growth factors known to modulate the
involves excess matrix synthesis along with dis- synthesis and deposition of matrix or inuence
torted deposition of that matrix; in addition there the make-up of the cell phenotype in the tissue
is the appearance of altered tissue cell phenotypes, may be crucial in dening the therapy that halts
most notably that of the myobroblast. These cells remodeling and progression and/or induces the
are derived from existing broblasts under the tissue to return to normal structure and function.
inuence of cytokines and growth factors, such as
transforming growth factor- (TGF-), or possi-
bly from circulating (stem cell like) precursors that
have migrated into the tissue injury site. Given
the morphology seen in lung disorders, asthma is GROWTH FACTORS IN HUMAN LUNG
mostly associated with minor, primarily peribron-
chial, matrix deposition, while chronic obstructive
AND AIRWAY DISEASE
pulmonary disease (COPD) has wide evidence
of remodeling, primarily at the alveolar level and A large number of studies have investigated the
brosis of the small airways [1]. role of growth factors in human lung disease in
Many cytokines and growth factors can which tissue remodeling is a prominent feature.
be found at the site of tissue remodeling using Most of these studies are based on immuno-
immunohistochemistry, in situ hybridization, and histochemical analysis of bronchial and pulmo-
gene expression assessments, so much so, that it nary tissue, obtained by biopsy, and on analysis
is dicult to determine causative versus second- of bronchoalveolar lavage (BAL) uid and cells.
ary presence. However, some, in comparison to Unfortunately there is minimal information avail-
others, have recently been shown to play a pri- able about growth factors in airway disease, but the
mary role in induction, and thus represent more presence of brotic responses and mesenchymal
likely targets for developing therapeutic inter- cell proliferation would suggest factors that are
ventions. Transgenic and gene knockout mod- prevalent in brogenesis. Three dierent factors
els in mice point to possible primary targets in are considered to play a prominent role: epider-
growth factors such as TGF-, IL-13, or hepato- mal growth factor (EGF), GM-CSF, and TGF-
cyte growth factor (HGF), while others implicate [2, 3]. Increased EGF and EGF receptor are
factors such as IL-1 or TNF-. The presence of present in the submucosa of asthmatic airways

353
Asthma and COPD: Basic Mechanisms and Clinical Management

[4, 5] and it is suggested that the epithelium does not respond present in the lung but is secreted rapidly upon a variety of
adequately to EGF after damage in asthma [3]. GM-CSF stimuli, mainly by alveolar macrophages and type II epithelial
expression is increased in asthmatic epithelium and lym- cells [18]. TNF- can bind to two dierent types of recep-
phocytes after allergen challenge [4] and GM-CSF can medi- tors, which are expressed on most cells and signals through
ate airway remodeling through its survival eect on eosinophils various intracellular pathways [27]. It is a potent proinam-
[2] and release of probrotic cytokines [6]. TGF- has received matory cytokine and exerts a variety of eects, which may
particular interest. In one study TGF- expression in airways is contribute to the process of remodeling and brosis: TNF-
shown to correlate with the degree of subepithelial brosis in induces inammatory cell migration and adhesion, initiates
asthma, although the data are controversial [4, 7]. More tell- a cytokine cascade, and regulates apoptosis [27]. Further,
ing is the presence of TGF- in BAL uid after segmental it is mitogenic for mesenchymal cells and inuences colla-
allergen challenge [5]. Moreover, neutralization of TGF- in gen metabolism, being either pro- or antibrotic [18, 28].
an animal model of repeat allergen exposure showed decreased Numerous studies have demonstrated that TNF- is
bronchial brotic sequelae [8] and TNF- and IL-1 are found involved in acute and chronic tissue changes seen after
at elevated levels in acutely inamed asthmatic airways [4]. bleomycin, asbestos, silica, and irradiation damage. Animal
Since both acute inammatory cytokines are able to trigger strains that do not develop brosis following exposure to
probrotic tissue reactions [911], this strongly suggests a role these agents show less TNF- upregulation. Transient over-
in airway remodeling. Other growth factors detected, such as expression of TNF- in the lung induces a limited brosis,
platelet-derived growth factor (PDGF) [2], insulin growth fac- likely by upregulation of TGF- [29]. Others have shown
tor-I (IGF-I), and basic broblast growth factors (bFGF) [3], that TNF- can act also through PDGF pathways [18].
could also participate in the repair process in airways. TNF- was detected in BAL uid and biopsies of patients
Most knowledge about growth factors in lung tis- with IPF, BOOP, and asbestosis, and likely plays a role in
sue remodeling has been accumulated through studies of many pulmonary diseases [18, 30].
parenchymal lung disease, such as asbestosis, sarcoidosis, or In asthma, TNF- can amplify the inammatory
idiopathic pulmonary brosis (IPF). We include these dis- process and have indirect inuence on airway remodeling
orders in our discussion to provide the reader perspective through induction of other growth factors [4]. It has been
on the potential biological actions of these factors and thus detected in BAL uid of asthmatics, in alveolar macro-
to provide a better perspective on asthma and COPD. The phages after allergen challenge and in bronchial mucosa of
presence of two key probrotic growth factors, TGF- and asthma patients [4] and can be rapidly released from mast
PDGF, has been demonstrated in various brosing disorders. cells on degranulation [31].
In IPF, TGF- was shown to be elevated in BAL uid [12,
13] and can be expressed by bronchial and alveolar epithe-
lial cells, alveolar macrophages and broblast foci [1417]. Interleukin-1
PDGF-mRNA was shown to be upregulated in BAL cells
retrieved from patients with IPF and in brotic areas of the Two forms of IL-1 ( and ) are known with almost iden-
lung, while lungs aected by asbestosis and silicosis express tical biological properties. In the lung, many parenchymal
increased TGF- mRNA [18]. In human brotic disorders and epithelial cell types are able to produce IL-1; how-
TNF- and IL-1 are present in BAL uid or BAL cell ever, the major sources are activated macrophages [4]. IL-1
supernatants [19]. In lungs of patients with progressive pul- is a pleiotropic proinammatory cytokine, often acting
monary brosis, TNF- and G-CSF are expressed at higher synergistically with TNF- [32]. In remodeling and bro-
than normal levels [19, 20], while similar tissue expresses sis, several actions of IL-1 are important. IL-1 stimulates
lower than normal levels of HGF [21]. An important con- broblasts to secrete other cytokines including IL-1, IL-8,
sideration is the relationship between growth factors and MCP-1, PDGF, and TGF- [33, 34], while the direct eect
extracellular matrix (ECM). Many growth factors can induce on broblast proliferation and ECM synthesis is controver-
ECM synthesis, but many are also associated with the matrix sial [28, 32, 35, 36]. In vivo, IL-1 is elevated in BAL uid
through adherence via RGD or related sequences to prote- and alveolar macrophages of patients with ARDS, but not
oglycans, collagens, and bronectins [22] and require acti- in IPF [19, 33], while in animal studies IL-1 is involved
vation (release) through interaction with cell integrins and in the early stage of pulmonary bleomycin injury [33], and
ECM components [2325] or via proteolysis, as seen with IL-1 receptor antagonist ameliorates the brotic response
plasminogen-mediated activation and release of HGF from following silica and bleomycin administration [37]. Recent
ECM [26]. (Table 29.1 and Fig. 29.1) studies show that transient transgene expression of IL-1
in the lung causes marked alveolar damage, induction of
TGF- and broblast foci with progressive brosis [38],
and similar ndings are seen in mice with inducible IL-1
SPECIFIC GROWTH FACTORS transgene [11]. Moreover, IL-1 is a potent stimulator of
osteopontin expression in lung broblasts, one of the most
highly expressed genes found in IPF tissues [39, 40] and a
Tumor necrosis factor- cytokine that recently has been shown to play a critical role
in allergic airway disease [41].
Tumor necrosis factor- (TNF-) is a peptide secreted as In asthma, IL-1 was found in BAL uid and mac-
membrane bound form and released after cleavage by TNF- rophages of asthma patients [4]. Its role in airway disease
-converting enzyme [27]. The cytokine is not constitutively is probably similar to TNF-, perpetuating primarily acute

354
Growth Factors 29
TABLE 29.1 Growth factors in pulmonary tissue remodeling.

Induce fibroblast accumulation Induce myofibroblasts Induce matrix expression

In vitro In vivo In vitro In vivo In vitro In vivo

TNF /   / 

IL-1 /   / 

GM-CSF   

PDGF  /   /

IGF / ? 

FGFs  ? ? ?  ?

KGF  ? ? ? ? ?

HGF      

TGF- /  ?   

TGF-      

CTGF  ? ? ?  ?

TNF: tumor necrosis factor; IL: interleukin; GM-CSF: granulocyte/macrophage colony stimulating factor; PDGF: platelet-derived growth factor; IGF: insulin-like growth factor;
FGF: fibroblast growth factor; TGF: transforming growth factor; CTGF: connective tissue growth factor; KGF: keratinocyte growth factor (FGF7); HGF: hepatocyte growth factor;
 not detectable; / detectable;  low;  moderate;  medium;  strong; ? unknown.

Interplay between growth factors and pulmonary parenchymal cells.


Injury depend on dose and time of expression and on concomitant
TNF IL-I epithelial cell damage. In the model of transient transgene
Remodeling
overexpression, low levels of GM-CSF induce little peri-
bronchial inammation and facilitate allergic reactions to
exogenous allergen [42]. At higher levels, GM-CSF in the
airways results in sustained eosinophilia and macrophage
accumulation with moderate development of tissue brosis
Myofibroblast mediated by induction of TGF-[43] and may represent a
target for manipulation in diseases such as COPD [44]. In
GM-CSF TGF- the bleomycin model, early upregulation of GM-CSF in
FGF PDGF Matrix
pulmonary cells is present and followed by enhanced TGF-
TGF- CTGF expression [45], while other studies imply a protective eect
of GM-CSF on the development of brosis in rats [46] and
FIG. 29.1 Growth factors released from injured epithelial and mice [47]. The probrotic response may be due to stimula-
parenchymal cells modify the behaviour and phenotype of mesenchymal tion and enhanced survival of macrophages and eosinophils,
cells in the parenchyma to induce myofibroblast differentiation and matrix both cells major sources for TGF-[4, 43, 45]. In the set-
deposition.
ting of damaged bronchial and alveolar epithelium, the pro-
liferative response of epithelium to GM-CSF stimulation
inammatory processes and aecting remodeling directly, might result in a more protective eect [46].
through modication of the matrix microenvironment, or GM-CSF is present in asthmatic airways of humans;
indirectly via induction of growth factors, such as PDGF there are increased levels in BAL after allergen challenge;
and TGF-. signicant levels of GM-CSF are found in plasma of
patients with severe asthma [4]. These observations indicate
that GM-CSF likely contributes to airway remodeling and
Granulocyte macrophage-colony plays a major role in the pathogenesis of asthma [2].
stimulating factor
GM-CSF is a colony-stimulating factor that regulates Platelet-derived growth factor
growth and dierentiation of hematopoietic cells. The lung
is a major source of GM-CSF and most pulmonary cells are PDGF is a glycoprotein homo- and heterodimer composed of
able to synthesize this cytokine in response to various stim- A and B chains, and is secreted by epithelial and endothelial
uli [4]. In the context of tissue remodeling and brosis, the cells, macrophages, and broblasts. PDGF-A chains can bind
eects of GM-CSF in the local environment of the lung to both PDGF-receptors and , whereas PDGF-B bind only

355
Asthma and COPD: Basic Mechanisms and Clinical Management

PDGF-receptor [48]. Signal transduction proceeds through broblasts [58]. Major sources for FGF-2 in the lung are
tyrosine kinases, a major pharmacologic target [49, 50]. PDGF mast cells [59]. In humans, FGF-2 was found in BAL and
isoforms are chemoattractive not only for broblasts, but also for serum of patients with IPF and scleroderma [60] and in
neutrophils and macrophages and they upregulate bronec- human airway smooth muscle cells; FGF-2 increases the
tin and procollagen gene expression and synthesis. PDGF can expression of PDGF-receptor and therefore indirectly
induce TGF- expression, suggesting that some of the long- stimulates proliferation [4].
term eects are partly mediated through this cytokine. However, KGF (FGF-7) is primarily produced by broblasts
PDGF and PDGF-receptor expression is stimulated by TGF-, and is mitogenic mainly for alveolar type II epithelial cells
IL-1, TNF-, and bFGF, indicating that parts of the pro- through the FGFR2-IIIb receptor, essentially expressed only
brotic activities of these molecules are due to PDGF-depend- by epithelial cells [57, 61]. IL-1 appears to be the most
ent pathways [18]. In animals that are exposed to asbestos, both potent inducer of KGF from broblasts [35] and in the
PDGF and its receptors are upregulated in bronchial bifurcations context of epithelial-mesenchymal cell interaction, IL-1
with developing broproliferative lesions [51]. PDGF-A knock- from epithelial cells stimulates the release of KGF from
out mice die from pulmonary emphysema apparently because of broblasts and results in the stimulation of the epithelium
impaired alveolar development and lack of myobroblasts [52]. in a positive paracrine system [62]. In the bleomycin model
Interference with PDGF activity, either through transgene over- of brosis instillation of KGF prevented the progression to
expression of a truncated receptor or through inhibition of tyro- brogenesis [63] and KGF was able to limit tissue damage
sine kinase results in amelioration of the brotic response in the and enhance epithelial repair in a syngeneic tracheal trans-
bleomycin model and in the bronchiolitis obliterans model [18]. plant model in rats [64]. In humans with ARDS, KGF in
In human brotic disorders of the lung, such as IPF, scleroderma BAL uid was taken as a marker of the severity of tissue
and bronchiolitis obliterans, PDGF genes were shown to be injury and correlated with poor prognosis [57], while KGF
upregulated in BAL cells or in aected tissues [18]. administration limited the decreased lung permeability and
In asthma and airway remodeling, a contribution of inammation seen on allergen challenge in an OVA-sensitized
PDGF to the pathogenesis is likely, but available data are rat model [65].
controversial. Eosinophils in biopsies of asthmatic airways
have been shown to produce PDGF-B chain [4]. Bronchial
broblasts from asthma patients show enhanced responsive- Epidermal growth factor
ness to the mitogenic eects of PDGF, but increased levels
of PDGF in BAL are either not present or do not correlate The EGF family is an enlarging group of related proteins and
with airway brosis [2]. includes EGF and transforming growth factor- (TGF-).
Both factors have potential roles in wound healing and
remodeling. They share 42% homology and signal through
Insulin-like growth factor the EGF receptor that activates tyrosine kinase [49, 66].
EGFs are important in the repair of epithelial injury.
IGF-I and II are single chain peptides with structural It has been shown in damaged bronchial epithelial cells that
homology to pro-insulin [53]. In the lung, the major sources EGF receptors become phosphorylated and consequently
of IGFs are macrophages, but mesenchymal and bronchial the defect is repaired and supplementation with exogenous
epithelial cells are also able to produce IGF [4, 54]. IGFs can EGF further enhances this process [3]. In asthma, EGF
bind to two receptors, type I and II, and biological activity receptor is highly expressed in airway epithelium, but epi-
is mainly determined by IGF-binding protein that releases thelial cell proliferation is still impaired [3], and impaired
IGF [55]. IGFs regulate proliferation and dierentiation of re-epithelialization induces the formation of granulation
a variety of cells [53], being strong mitogens for broblasts tissue [67]. This suggests that an abnormal response of the
and causing enhanced collagen synthesis [54]. IGFs are also epithelium to growth factors could be a central factor in the
found in tissue and BAL uids of various broproliferative pathogenesis of asthma and airway remodeling. In humans,
diseases [12, 56]. However, little information is available EGF and EGF receptor are present in the submucosa of
about the role of IGFs in asthma or COPD, but IGFs can asthmatic airways [3, 4].
stimulate the proliferation of airway smooth muscle cells and Similar to EGF, TGF- is able to promote wound
may be involved in airway brosis [4]. epithelialization [53], while tissue specic overexpression of
TGF- in the lung of mice leads to alveolar enlargement
and interstitial and pleural brosis. Conversely, elimination
Fibroblast growth factors of the TGF- gene signicantly decreases the brogenic
tissue response to bleomycin [66]. In humans, TGF- pro-
FGFs are a group of nine heparin-binding peptides, tein is elevated in BAL uid of patients with ARDS and
amongst them acidic and basic FGF (now FGF-1 and 2) IPF [68], and TGF- and EGF receptor expression is seen
and keratinocyte growth factor (KGF or FGF-7) [53, 57]. in biopsy material in IPF [69].
FGF-1 and 2 have been shown to accelerate granulation
tissue formation, broblast proliferation and collagen syn-
thesis [53]. They are strong mitogens for angiogenesis and Hepatocyte growth factor
endothelial cell migration [4]. FGF-1 and 2 are not present
at signicant levels in normal lungs but are produced after HGF was originally identied as a potent stimulator of
tissue injury by alveolar macrophages, epithelial cells, and mature hepatocytes, but more recently is recognized as

356
Growth Factors 29
a participant in many tissue responses to injury and is a Extra cellular matrix
potentially important stimulator of alveolar epithelial cells deposition
and vascular endothelial cells [70, 71]. In lung development,
HGF is shown to be required for alveologenesis in the
rodent [72], and recent studies indicate this growth factor
may play a role in airway sensitization through modula- Ap
te TGF- op
tion of dendritic cell activation [73]. Importantly, there are mula tos
defects in HGF production from broblasts in tissue sam- Sti is

ples from both emphysema and IPF patients [21, 74]. Most
impressive is the experimental attenuation of both elastase-
induced emphysema and bleomycin-induced brosis by
Ap
administration of either recombinant HGF or by gene op late
tos mu
transfer to the lung [71, 75, 76]. These data suggest that is
HGF Sti
HGF has activities that could balance the probrotic activi-
ties of factors such as TGF- and establishes an interplay
between these powerful factors that could distinguish cor-
rect tissue repair from disordered pathology (Fig. 29.2).

Fibroblast Epithelium
Transforming growth factor- Myofibroblast
Inflammation
The TGF- family includes 5 isoforms, of which mamma- &
lian cells express three. The isoforms TGF- 1 to 3 reveal Tissue damage
considerable sequence homology, and their biological prop-
FIG. 29.2 Two main growth factors balance cell survival and tissue
erties in wound healing and tissue remodeling are similar.
remodeling. TGF-, released from the matrix in the parenchyma during
Most studies have been performed with TGF-1, which tissue injury, is a major stimulator of myofibroblast and fibroblast survival
is the most abundant and best-characterized isoform [77]. and ECM production. TGF- also induces apoptosis in pulmonary epithelial
TGF- is probrotic by various eects on ECM turnover cells. HGF, released from fibroblasts in the parenchyma by molecules
and stromal cell biology. The immunomodulatory activities such as IL-1, balance the pro-fibrotic effects of TGF- by stimulating the
of TGF- are crucial and aim mainly at the limitation of survival of epithelial cells and inducing the apoptosis of myofibroblasts, as
inammatory processes. Many dierent cells secrete TGF-, well as dampening the inflammatory response that accompanies tissue
in the lung mainly macrophages, epithelial cells, and brob- injury. Both are autocrine and paracrine in function, and have an apparent
lasts. The secreted molecule is inactive and has to be acti- inhibition effect on release of the counterpart.
vated by cleavage of the latency associated peptide (LAP)
through oxygen radicals or proteases (e.g. thrombospondin The role of TGF- in pulmonary remodeling and
or plasmin) [78] or through interaction with cell integrins, brosis has been investigated mainly in interstitial lung
most notably V6 and V8 on epithelial cells or brob- disease, where the key role of this growth factor is widely
lasts [24, 79]. TGF- binds to two receptors, type I and II, accepted, with evidence from a large number of animal
and signals are transduced by serine/threonine kinases to models and human studies [18, 53, 83, 85]. In models using
dierent intracellular pathways [80, 81]. In the context of chemical agents such as bleomycin, asbestos or silica, or
tissue remodeling, the Smad-signaling pathway is the most irradiation to induce progressive tissue damage, TGF- was
prominent and a potential target for pharmacologic inter- shown to be upregulated at the sites of developing bro-
vention in brotic disorders [82, 83]. TGF- stimulates sis. In our own studies, in which transient overexpression
ECM production, predominantly collagen and bronectin, of active TGF-1 is achieved by adenoviral gene transfer,
and reduces matrix degradation by changing the balance severe and diuse pulmonary brosis developed in rats and
of collagenases and collagenase inhibitors (TIMP-1) [78]. mice [78], mediated through the Smad3-signaling pathway
It is anti-proliferative for epithelial cells, inducing apopto- [83]. The importance of TGF- in tissue remodeling after
sis and subsequent brotic responses in the lung [84, 85]. pulmonary injury is highlighted by several experimental set-
The mitogenic eect on broblasts is not uniform, but it is tings that demonstrate the benecial eect of anti-TGF-
apparent that TGF- induces the transformation of brob- agents on the course of brosis. Neutralizing antibodies,
lasts into myobroblasts [86]. Myobroblasts are contractile dominant negative TGF--receptors or natural antago-
cells and synthesize most matrix proteins, both important nists such as the proteoglycan decorin have all been able to
factors in the nal step of wound healing, but potentially reduce brotic reactions in the lung [87, 88] and TGF-
a pathogenic process in brosis. In the remodeling proc- was shown to be present in a variety of human lung dis-
ess, TGF- acts in concert with many other cytokines and eases, including asthma and IPF. Also, as COPD could be
growth factors. On the aerent, it can be upregulated by considered a disorder with inadequate tissue repair [89], the
TNF-, IL-1, GM-CSF, PDGF, and TGF- itself. On the genetic association of the TGF gene with COPD disease is
eector, TGF- not only acts through direct interference a positive indication of involvement [90].
with collagen metabolism but may induce the expression The importance of TGF- in pulmonary remodeling
and secretion of FGF, PDGF, and connective tissue growth is not only restricted to the interstitium, but is also present
factor (CTGF), as well as its own upregulation [9, 18, 86]. in airways. TGF- genes are upregulated in bronchial walls

357
Asthma and COPD: Basic Mechanisms and Clinical Management

in a model of chronic airway disease [91], and anti-TGF- by repeat allergen exposure with Aspergillus, ovalbumin or
is shown to modulate the subepithelial brotic response house dust mite antigens. These models produce in mice
seen in a chronic allergen challenge model in the mouse changes similar to human asthmatic airway remodeling, and
[8]. Interestingly, TGF-1 and TGF-3 may have opposite airway brosis, accompanied by upregulation of TGF- and
eects on connective tissue synthesis in cultured airway GM-CSF [8, 17, 91, 99, 100].
smooth muscle cells, suggesting a protective role for TGF-3
in airway remodeling [92]. In asthma patients, TGF-
was found in airway walls and correlated with the degree Transgene animal models
of subepithelial brosis [7], but others have failed to nd a
correlation to disease severity [93], while TGF- is released In transgenic animal models certain genes are deleted, over-
into BAL uid after endobronchial allergen challenge [3]. expressed, or mutated. Transgenic animals, either developed
There are ndings that link the pathogenesis of brogen- by gene insertion (or deletion) in embryonic stem cells or
esis to that of emphysema; interference with either the acti- developed by transient transgene expression in adult ani-
vation of TGF- (V6 null mouse) or signaling through mals by gene transfer (e.g. adenoviral vectors) have proven
the receptor (Smad3 null mouse) leads to spontaneous useful to answer questions about the role of specic growth
airspace enlargement and aspects of emphysema [89, 94]. factors in the pathogenesis of pulmonary disease. Animals
These changes are accompanied by enhanced expression of transiently overexpressing IL-1 or TGF- develop pro-
MMP12 by macrophages, normally controlled by exposure gressive brotic lesions in the lung, IL-1, likely through
of these cells to TGF- [89], implying tissue repair abnor- induction of TGF- [10, 29, 43, 78]. Conversely, TNF-
malities lie at the center of both disorders. -receptor and TGF- knockout mice are resistant to
bleomycin-induced pulmonary brosis [18, 66]. Also impli-
cated by transgenic studies are IL-11 and IL-13; these mol-
Connective tissue growth factor ecules were not previously known to be factors involved
in tissue remodeling and repair, but showing evidence of
Connective tissue growth factor is a growth factor with brotic changes, emphysematous lesions, and deranged
similar activities on collagen metabolism as TGF-. It is a mucus production [101, 102].
cysteine-rich peptide produced by broblasts and endothelial
cells, but not by leukocytes or epithelial cells [18, 95]. CTGF
is constitutively expressed in human lung broblasts and
is stimulated by TGF-, but not TNF- or IL-1 [96] In
mouse lungs, TGF- and CTGF are upregulated following SUMMARY
bleomycin injury. It has been suggested that TGF- exerts
its probrotic eects in part through PDGF pathways and Of the many cytokines and growth factors that are found
through a PDGF-independent pathway using CTGF [18]. within the tissue or surrounding uids in COPD and
Less information is known about CTGF in airway asthma, only a few can be shown to have direct impact on
remodeling. However, the association with TGF- makes it the process of tissue remodeling. In vitro and in vivo studies
likely that CTGF might have similar eects in the airway outlined above indicate that factors such as TGF-, which
as TGF-. Indeed, it has been recently shown that TGF- induces chronic repair without accompanying tissue injury,
stimulates expression of CTGF in cultured airway smooth and IL-1, which induces tissue injury and chronic repair,
muscle cells [97]. likely through induction of TGF-, may be considered the
most critical targets for intervention. The fact that these
growth factors act mainly at a local site in association with
matrix helps explain the progressive and tissue-restricted
ANIMAL MODELS nature of remodeling. Development of potent inhibitors of
these growth factors or of genes activated downstream of
them could prove benecial in modifying the altered tis-
Chronic airway challenge sue in asthma and COPD allowing conjoint therapy with
anti-inammatory drugs, preferably delivered to the local
Airway remodeling in human chronic airway disease is a remodeled site, to halt the destructive process and return
concept not yet clearly dened [2]. The principal morpho- the lung to normal function.
logic features are subepithelial brosis, myobroblast hyper-
plasia, airway smooth muscle hypertrophy, mucus gland and
goblet cell hyperplasia, and epithelial disruption [98]. The References
mechanisms leading to persistent airway changes are poorly
characterized because of a lack of simple and suitable ani-
1. Hogg JC, Chu F, Utokaparch S et al. The nature of small-airway obstruc-
mal models. An impaired response of asthmatic bronchi to tion in chronic obstructive pulmonary disease. N Engl J Med 350(26):
EGF after epithelial injury seems to be important to initiate 264553, 2004.
the process, while other growth factors, such as TGF- 2. Fish JE, Peters SP. Airway remodeling and persistent airway obstruction
isoforms, PDGF, and bFGF might be responsible for the in asthma. J Allergy Clin Immunol 104(3 Pt 1): 50916, 1999.
proliferative response [3]. Recently described animal mod- 3. Holgate ST. Epithelial damage and response. Clin Exp Allergy 30
els resemble certain aspects of human asthma induced (Suppl 1): 3741, 2000.

358
Growth Factors 29
4. Chung KF, Barnes PJ. Cytokines in asthma. Thorax 54(9): 82557, 24. Araya J, Cambier S, Morris A, Finkbeiner W, Nishimura SL. Integrin-
1999. mediated transforming growth factor-beta activation regulates home-
5. Redington AE, Madden J, Frew AJ et al. Transforming growth factor- ostasis of the pulmonary epithelial-mesenchymal trophic unit. Am J
beta 1 in asthma. Measurement in bronchoalveolar lavage uid. Am J Pathol 169(2): 40515, 2006.
Respir Crit Care Med 156(2 Pt 1): 64247, 1997. 25. Parameswaran K, Willems-Widyastuti A, Alagappan VK, Radford K,
6. Xing Z, Ohkawara Y, Jordana M, Graham FL, Gauldie J. Transfer of Kranenburg AR, Sharma HS. Role of extracellular matrix and its regu-
granulocyte-macrophage colony-stimulating factor gene to rat lung lators in human airway smooth muscle biology. Cell Biochem Biophys
induces eosinophilia, monocytosis, and brotic reactions. J Clin Invest 44(1): 13946, 2006.
97(4): 110210, 1996. 26. Matsuoka H, Sisson TH, Nishiuma T, Simon RH. Plasminogen-mediated
7. Minshall EM, Leung DY, Martin RJ et al. Eosinophil-associated TGF- activation and release of hepatocyte growth factor from extracellular
beta1 mRNA expression and airways brosis in bronchial asthma. Am J matrix. Am J Respir Cell Mol Biol 35(6): 70513, 2006.
Respir Cell Mol Biol 17(3): 32633, 1997. 27. Sporn MB, Roberts AB (eds). Peptide Growth Factors and Their
8. McMillan SJ, Xanthou G, Lloyd CM. Manipulation of allergen- Receptors. New York: Springer-Verlag, 1991.
induced airway remodeling by treatment with anti-TGF-{beta} 28. Tufvesson E, Westergren-Thorsson G. Alteration of proteoglycan
antibody: Eect on the Smad signaling pathway. J Immunol 174(9): synthesis in human lung broblasts induced by interleukin-1beta and
577480, 2005. tumor necrosis factor-alpha. J Cell Biochem 77(2): 298309, 2000.
9. Xing Z, Jordana M, Gauldie J, Wang J. Cytokines and pulmonary 29. Sime PJ, Marr RA, Gauldie D et al. Transfer of tumor necrosis factor-
inammatory and immune diseases. Histol Histopathol 14(1): 185201, alpha to rat lung induces severe pulmonary inammation and patchy
1999. interstitial brogenesis with induction of transforming growth factor-
10. Kolb M, Margetts PJ, Anthony DC, Pitossi F, Gauldie J. Transient beta 1 and myobroblasts. Am J Pathol 153(3): 82532, 1998.
expression of IL-1 beta induces acute lung injury and chronic repair 30. Mukhopadhyay S, Hoidal J, Mukherjee T. Role of TNFalpha in pul-
leading to pulmonary brosis. J Clin Invest 107(12): 152936, 2001. monary pathophysiology. Respir Res 7(1): 125, 2006.
11. Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K. 31. Kendall JC, Li XH, Galli SJ, Gordon JR. Promotion of mouse brob-
Interleukin-1beta causes pulmonary inammation, emphysema, and last proliferation by IgE-dependent activation of mouse mast cells:
airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol Role for mast cell tumor necrosis factor-alpha and transforming
32(4): 31118, 2005. growth factor-beta 1. J Allergy Clin Immunol 99(1 Pt 1): 11323, 1997.
12. Vanhee D, Gosset P, Wallaert B, Voisin C, Tonnel AB. Mechanisms of 32. Rochester CL et al. Cytokines and cytokine networking in the patho-
brosis in coal workerspneumoconiosis. Increased production of platelet- genesis in interstitial and brotic lung disorders. Semin Respir Crit Care
derived growth factor, insulin-like growth factor type I, and transform- Med(14): 389416, 1994.
ing growth factor beta and relationship to disease severity. Am J Respir 33. Sime PJ, Gauldie J. Mechanisms of scarring. In: Tw E, Haslett C
Crit Care Med 150(4): 104955, 1994. (eds). ARDS: Acute Respiratory Distress in Adults, pp. 21531. London:
13. Hiwatari N, Shimura S, Yamauchi K, Nara M, Hida W, Shirato K. Chapman and Hall Medical, 1996.
Signicance of elevated procollagen-III-peptide and transforming 34. Boyle JE, Lindroos PM, Rice AB, Zhang L, Zeldin DC, Bonner JC.
growth factor-beta levels of bronchoalveolar lavage uids from idi- Prostaglandin-E2 counteracts interleukin-1beta-stimulated upregula-
opathic pulmonary brosis patients. Tohoku J Exp Med 181(2): 28595, tion of platelet-derived growth factor alpha-receptor on rat pulmonary
1997. myobroblasts. Am J Respir Cell Mol Biol 20(3): 43340, 1999.
14. Broekelmann TJ, Limper AH, Colby TV, McDonald JA. Transforming 35. Chedid M, Rubin JS, Csaky KG, Aaronson SA. Regulation of kerati-
growth factor beta 1 is present at sites of extracellular matrix gene nocyte growth factor gene expression by interleukin 1. J Biol Chem
expression in human pulmonary brosis. Proc Natl Acad Sci USA 269(14): 1075357, 1994.
88(15): 664246, 1991. 36. Postlethwaite AE, Raghow R, Stricklin GP, Poppleton H, Seyer
15. Khalil N, Greenberg AH. The role of TGF-beta in pulmonary brosis. JM, Kang AH. Modulation of broblast functions by interleukin 1:
Ciba Found Symp 157: 194211, 1991. Increased steady-state accumulation of type I procollagen messen-
16. Corrin B, Butcher D, McAnulty BJ et al. Immunohistochemical locali- ger RNAs and stimulation of other functions but not chemotaxis by
zation of transforming growth factor-beta 1 in the lungs of patients human recombinant interleukin 1 alpha and beta. J Cell Biol 106(2):
with systemic sclerosis, cryptogenic brosing alveolitis and other lung 31118, 1988.
disorders. Histopathology 24(2): 14550, 1994. 37. Piguet PF, Vesin C, Grau GE, Thompson RC. Interleukin 1 receptor
17. Kelly MM, Leigh R, Bonniaud P et al. Epithelial expression of pro- antagonist (IL-1ra) prevents or cures pulmonary brosis elicited in
brotic mediators in a model of allergen-induced airway remodeling. mice by bleomycin or silica. Cytokine 5(1): 5761, 1993.
Am J Respir Cell Mol Biol 32(2): 99107, 2005. 38. Kolb M, Bonniaud P, Galt T et al. Dierences in the brogenic
18. Lasky JA, Brody AR. Interstitial brosis and growth factors. Environ response after transfer of active transforming growth factor-beta1 gene
Health Perspect 108(Suppl 4): 75162, 2000. to lungs of brosis-prone and brosis-resistant mouse strains. Am J
19. Ziegenhagen MW, Schrum S, Zissel G, Zipfel PF, Schlaak M, Muller- Respir Cell Mol Biol 27(2): 14150, 2002.
Quernheim J. Increased expression of proinammatory chemokines 39. Pardo A, Gibson K, Cisneros J et al. Up-regulation and probrotic role
in bronchoalveolar lavage cells of patients with progressing idiopathic of osteopontin in human idiopathic pulmonary brosis. PLoS Med 2(9),
pulmonary brosis and sarcoidosis. J Investig Med 46(5): 22331, 2005.
1998. 40. Serlin DM, Kuang PP, Subramanian M et al. Interleukin-1beta induces
20. Ashitani J, Mukae H, Taniguchi H et al. Granulocyte-colony stimulat- osteopontin expression in pulmonary broblasts. J Cell Biochem(97):
ing factor levels in bronchoalveolar lavage uid from patients with idi- 51929, 2006.
opathic pulmonary brosis. Thorax 54(11): 101520, 1999. 41. Xanthou G, Alissa T, Semitekolou M et al. Osteopontin has a crucial
21. Marchand-Adam S, Marchal J, Cohen M et al. Defect of hepatocyte role in allergic airway disease through regulation of dendritic cell sub-
growth factor secretion by broblasts in idiopathic pulmonary brosis. sets. Nat Med 13(5): 57078, 2007.
Am J Respir Crit Care Med 168(10): 115661, 2003. 42. Stampi MR, Wiley RE, Neigh GS et al. GM-CSF transgene expres-
22. Postma DS, Timens W. Remodeling in asthma and chronic obstructive sion in the airway allows aerosolized ovalbumin to induce allergic sen-
pulmonary disease. Proc Am Thorac Soc 3(5): 43439, 2006. sitization in mice. J Clin Invest 102(9): 170414, 1998.
23. Munger JS, Huang X, Kawakatsu H et al. The integrin alpha v beta 43. Xing Z, Tremblay GM, Sime PJ, Gauldie J. Overexpression of
6 binds and activates latent TGF beta 1: A mechanism for regulating granulocyte-macrophage colony-stimulating factor induces pulmonary
pulmonary inammation and brosis. Cell 96(3): 31928, 1999. granulation tissue formation and brosis by induction of transforming

359
Asthma and COPD: Basic Mechanisms and Clinical Management

growth factor-beta 1 and myobroblast accumulation. Am J Pathol 64. Gomperts BN, Belperio JA, Fishbein MC, Keane MP, Burdick MD,
150(1): 5966, 1997. Strieter RM. Keratinocyte growth factor improves repair in the injured
44. Vlahos R, Bozinovski S, Hamilton JA, Anderson GP. Therapeutic tracheal epithelium. Am J Respir Cell Mol Biol 37(1): 4856, 2007.
potential of treating chronic obstructive pulmonary disease (COPD) 65. Tillie-Leblond I, Gosset P, Le Berre R et al. Keratinocyte growth fac-
by neutralising granulocyte macrophage-colony stimulating factor tor improves alterations of lung permeability and bronchial epithelium
(GM-CSF). Pharmacol Ther 112(1): 10615, 2006. in allergic rats. Eur Respir J 30(1): 131725, 2007.
45. Andreutti D, Gabbiani G, Neuville P. Early granulocyte-macrophage 66. Madtes DK, Elston AL, Hackman RC, Dunn AR, Clark JG.
colony-stimulating factor expression by alveolar inammatory cells Transforming growth factor-alpha deciency reduces pulmonary brosis
during bleomycin-induced rat lung brosis. Lab Invest 78(12): 1493 in transgenic mice. Am J Respir Cell Mol Biol 20(5): 92434, 1999.
502, 1998. 67. Khalil N, OConnor RN, Flanders KC, Shing W, Whitman CI.
46. Christensen PJ, Bailie MB, Goodman RE, OBrien AD, Toews GB, Regulation of type II alveolar epithelial cell proliferation by TGF-beta
Paine R III.. Role of diminished epithelial GM-CSF in the pathogenesis during bleomycin-induced lung injury in rats. Am J Physiol 267(5 Pt 1):
of bleomycin-induced pulmonary brosis. Am J Physiol Lung Cell Mol L498507, 1994.
Physiol 279(3): L487lL495, 2000. 68. Madtes DK, Rubenfeld G, Klima LD et al. Elevated transforming
47. Piguet PF, Grau GE, de Kossodo S. Role of granulocyte-macrophage growth factor-alpha levels in bronchoalveolar lavage uid of patients
colony-stimulating factor in pulmonary brosis induced in mice by with acute respiratory distress syndrome. Am J Respir Crit Care Med
bleomycin. Exp Lung Res 19(5): 57987, 1993. 158(2): 42430, 1998.
48. Fabisiak JK, Kelley JP. Platelet derived growth factor. In: Kelley J (ed.), 69. Baughman RP, Lower EE, Miller MA, Bejarano PA, Heelnger SC.
Cytokines of the Lung, pp. 339. New York: Marcel Dekker, 1992. Overexpression of transforming growth factor-alpha and epidermal
49. Rice AB, Moomaw CR, Morgan DL, Bonner JC. Specic inhibitors growth factor-receptor in idiopathic pulmonary brosis. Sarcoidosis Vasc
of platelet-derived growth factor or epidermal growth factor receptor Diuse Lung Dis 16(1): 5761, 1999.
tyrosine kinase reduce pulmonary brosis in rats. Am J Pathol 155(1): 70. Ware LB, Matthay MA. Keratinocyte and hepatocyte growth factors
21321, 1999. in the lung: Roles in lung development, inammation, and repair. Am J
50. Ingram JL, Bonner JC. EGF and PDGF receptor tyrosine kinases as Physiol Lung Cell Mol Physiol 282(5): L924L940, 2002.
therapeutic targets for chronic lung diseases. Curr Mol Med 6(4): 409 71. Shigemura N, Sawa Y, Mizuno S et al. Amelioration of pulmonary
21, 2006. emphysema by in vivo gene transfection with hepatocyte growth factor
51. Lasky JA, Tonthat B, Liu JY, Friedman M, Brody AR. Upregulation in rats. Circulation 111(11): 140714, 2005.
of the PDGF-alpha receptor precedes asbestos-induced lung brosis in 72. Padela S, Cabacungan J, Shek S et al. Hepatocyte growth factor is
rats. Am J Respir Crit Care Med 157(5 Pt 1): 165257, 1998. required for alveologenesis in the neonatal rat. Am J Respir Crit Care
52. Bostrom H, Willetts K, Pekny M et al. PDGF-A signaling is a critical Med 172(7): 90714, 2005.
event in lung alveolar myobroblast development and alveogenesis. Cell 73. Okunishi K, Dohi M, Nakagome K et al. A novel role of hepatocyte
85(6): 86373, 1996. growth factor as an immune regulator through suppressing dendritic
53. Mutsaers SE, Bishop JE, McGrouther G, Laurent GJ. Mechanisms of cell function. J Immunol 175(7): 474553, 2005.
tissue repair: From wound healing to brosis. Int J Biochem Cell Biol 74. Plantier L, Marchand-Adam S, Marchal-Somme J et al. Defect of
29(1): 517, 1997. hepatocyte growth factor production by broblasts in human pulmo-
54. Uh ST, Inoue Y, King TE Jr., Chan ED, Newman LS, Riches DW. nary emphysema. Am J Physiol Lung Cell Mol Physiol 288(4): L641
Morphometric analysis of insulin-like growth factor-I localization in L647, 2005.
lung tissues of patients with idiopathic pulmonary brosis. Am J Respir 75. Mizuno S, Matsumoto K, Li MY, Nakamura T. HGF reduces advanc-
Crit Care Med 158(5 Pt 1): 162635, 1998. ing lung brosis in mice: A potential role for MMP-dependent myo-
55. Bayes-Genis A, Conover CA, Schwartz RS. The insulin-like growth broblast apoptosis. FASEB J 19(6): 58082, 2005.
factor axis: A review of atherosclerosis and restenosis. Circ Res 86(2): 76. Gazdhar A, Fachinger P, van Leer C et al. Gene transfer of hepato-
12530, 2000. cyte growth factor by electroporation reduces bleomycin-induced lung
56. Krein PM, Sabatini PJ, Tinmouth W, Green FH, Winston BW. brosis. Am J Physiol Lung Cell Mol Physiol 292(2): L529L536, 2007.
Localization of insulin-like growth factor-I in lung tissues of patients 77. OKane S, Ferguson MW. Transforming growth factor beta s and
with broproliferative acute respiratory distress syndrome. Am J Respir wound healing. Int J Biochem Cell Biol 29(1): 6378, 1997.
Crit Care Med 167(1): 8390, 2003. 78. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector-
57. Stern JB, Fierobe L, Paugam C et al. Keratinocyte growth factor and mediated gene transfer of active transforming growth factor-beta1
hepatocyte growth factor in bronchoalveolar lavage uid in acute respi- induces prolonged severe brosis in rat lung. J Clin Invest 100(4): 768
ratory distress syndrome patients. Crit Care Med 28(7): 232633, 2000. 76, 1997.
58. Barrios R, Pardo A, Ramos C, Montano M, Ramirez R, Selman M. 79. Munger JS, Huang X, Kawakatsu H et al. A mechanism for regulat-
Upregulation of acidic broblast growth factor during development of ing pulmonary inammation and brosis: The integrin [alpha]v[beta]6
experimental lung brosis. Am J Physiol 273(2 Pt 1): L451L458, 1997. binds and activates latent TGF [beta]1. Cell 96(3): 319, 1999.
59. Liebler JM, Picou MA, Qu Z, Powers MR, Rosenbaum JT. Altered 80. Derynck R, Zhang YE. Smad-dependent and Smad-independent
immunohistochemical localization of basic broblast growth factor after pathways in TGF-beta family signalling. Nature 425(6958): 57784,
bleomycin-induced lung injury. Growth Factors 14(1): 2538, 1997. 2003.
60. Kadono T, Kikuchi K, Kubo M, Fujimoto M, Tamaki K. Serum con- 81. Feng XH, Derynck R. Specicity and versatility in tgf-beta signaling
centrations of basic broblast growth factor in collagen diseases. J Am through Smads. Annu Rev Cell Dev Biol 21: 65993, 2005.
Acad Dermatol 35(3 Pt 1): 39297, 1996. 82. Bonniaud P, Margetts PJ, Kolb M et al. Progressive transforming
61. Finch PW, Rubin JS. Keratinocyte growth factor/broblast growth fac- growth factor {beta}1-induced lung brosis is blocked by an orally
tor 7, a homeostatic factor with therapeutic potential for epithelial pro- active ALK5 kinase inhibitor. Am J Respir Crit Care Med 171(8): 889
tection and repair. Adv Cancer Res 91: 69136, 2004. 98, 2005.
62. Marchand-Adam S, Plantier L, Bernuau D et al. Keratinocyte growth 83. Gauldie J, Kolb M, Ask K, Martin G, Bonniaud P, Warburton D.
factor expression by broblasts in pulmonary brosis: Poor response to Smad3 signaling involved in pulmonary brosis and emphysema. Proc
interleukin-1 {beta}. Am J Respir Cell Mol Biol 32(5): 47077, 2005. Am Thorac Soc 3(8): 696702, 2006.
63. Sugahara K, Iyama K, Kuroda MJ, Sano K. Double intratracheal instil- 84. Lee CG, Cho SJ, Kang MJ et al. Early growth response gene 1-medi-
lation of keratinocyte growth factor prevents bleomycin-induced lung ated apoptosis is essential for transforming growth factor beta1-
brosis in rats. J Pathol 186(1): 9098, 1998. induced pulmonary brosis. J Exp Med 200(3): 37789, 2004.

360
Growth Factors 29
85. Lee CG, Kang HR, Homer RJ, Chupp G, Elias JA. Transgenic mode- 94. Bonniaud P, Kolb M, Galt T et al. Smad3 null mice develop airspace
ling of transforming growth factor-beta(1): Role of apoptosis in brosis enlargement and are resistant to TGF-beta-mediated pulmonary
and alveolar remodeling. Proc Am Thorac Soc 3(5): 41823, 2006. brosis. J Immunol 173(3): 2099108, 2004.
86. Gauldie J et al. TGFb gene transfer to the lung induces myobro-blast 95. Grotendorst GR. Connective tissue growth factor: A mediator of
presence and pulmonary brosis. In: Desmoliere A, Tuchweber B (eds). TGF-beta action on broblasts. Cytokine Growth Factor Rev 8(3):
Tissue Repair and Fibrosis: Current Topics in Pathology. Berlin: Springer- 17179, 1997.
Verlag, 1999. 96. Lasky JA, Ortiz LA, Tonthat B et al. Connective tissue growth factor
87. Giri SN, Hyde DM, Braun RK, Gaarde W, Harper JR, Pierschbacher mRNA expression is upregulated in bleomycin-induced lung brosis.
MD. Antibrotic eect of decorin in a bleomycin hamster model of Am J Physiol 275(2 Pt 1): L365L371, 1998.
lung brosis. Biochem Pharmacol 54(11): 120516, 1997. 97. Douglas DA. TGFb stimulates the expression of CTGF by human bron-
88. Kolb M, Margetts PJ, Galt T et al. Transient transgene expression of chial smooth muscle cells. Am J Respir Crit Care Med 161: A699, 2000.
decorin in the lung reduces the brotic response to bleomycin. Am J 98. Redington AE. Fibrosis and airway remodelling. Clin Exp Allergy
Respir Crit Care Med 163(3): 77077, 2001. 30(Suppl 1): 4245, 2000.
89. Morris DG, Sheppard D. Pulmonary emphysema: When more is less. 99. Cates EC, Fattouh R, Wattie J et al. Intranasal exposure of mice to
Physiology (Bethesda) 21: 396403, 2006. house dust mite elicits allergic airway inammation via a GM-CSF-
90. Celedon JC, Lange C, Raby BA et al. The transforming growth factor- mediated mechanism. J Immunol 173(10): 638492, 2004.
beta1 (TGFB1) gene is associated with chronic obstructive pulmonary 100. Johnson JR, Wiley RE, Fattouh R et al. Continuous exposure to
disease (COPD). Hum Mol Genet 13(15): 164956, 2004. house dust mite elicits chronic airway inammation and structural
91. Hogaboam CM, Blease K, Mehrad B et al. Chronic airway hyper- remodeling. Am J Respir Crit Care Med 169(3): 37885, 2004.
reactivity, goblet cell hyperplasia, and peribronchial brosis during 101. Zhu Z, Homer RJ, Wang Z et al. Pulmonary expression of inter-
allergic airway disease induced by Aspergillus fumigatus. Am J Pathol leukin-13 causes inammation, mucus hypersecretion, subepithelial
156(2): 72332, 2000. brosis, physiologic abnormalities, and eotaxin production. J Clin
92. Coutts A et al. TGF-beta3 inhibits connective tissue synthesis by air- Invest 103(6): 77988, 1999.
way smooth muscle cells. Am J Respir Crit Care Med 161: A699, 2000. 102. Lee PJ, Zhang X, Shan P et al. ERK1/2 mitogen-activated protein
93. Chu HW, Halliday JL, Martin RJ, Leung DY, Szeer SJ, Wenzel kinase selectively mediates IL-13-induced lung inammation and
SE. Collagen deposition in large airways may not dierentiate severe remodeling in vivo. J Clin Invest 116(1): 16373, 2006.
asthma from milder forms of the disease. Am J Respir Crit Care Med
158(6): 193644, 1998.

361
Nitric Oxide
30CHAPTER

three isoforms: (1) constitutive neuronal NOS


INTRODUCTION (NOS I or nNOS); (2) inducible NOS (NOS Sergei A. Kharitonov
II or iNOS); and (3) constitutive endothelial and Kazuhiro Ito
Nitric oxide (NO) in human biology was NOS (NOS III or eNOS). The existence of a
mostly considered to be a by-product of the genetic association between a polymorphism Section of Airway Disease, National
combustion of fossil fuels and therefore as an in the nNOS gene and the diagnosis of asthma Heart and Lung Institute, Imperial
air pollutant, until the mid- to late 1980s when has led to the speculation that nNOS is a can- College, London, UK
it was discovered that this free radical was the didate gene for asthma [4].
previously uncharacterized endothelial-derived Functionally, constitutive (or c) NOSs
relaxing factor [1]. (nNOS and eNOS) are Ca2- and calmodulin-
NO is now known to play a central role dependent enzymes and rapidly release low con-
in the physiology and the pathophysiology of centrations of NO upon receptor stimulation by
many human organ systems. Within the res- selective agonists. The activity of iNOS is largely
piratory system, NO promotes vascular and regulated at a pretranslational level and can be
bronchial dilation, is a key mediator of the induced by proinammatory cytokines, such as
coordinated beating of ciliated epithelial cells, tumor necrosis factor- (TNF-), interferon-
promotes mucus secretion, and is an important (IFN-), and interleukin (IL)-1 [5]. Several
neurotransmitter for nonadrenergic, nonchol- hours after exposure to these inammatory
inergic neurons in the bronchial wall [2, 3]. stimuli, iNOS action releases large quantities of
Additionally, NO is a mediator of inammatory NO, which may continue in a sustained manner
phenomena within the lung by virtue of its abil- (hours or days).
ity to inuence the phenotype of inammatory All the three NOS isoforms are found in
cells and its contribution to the formation of the respiratory system and may co-operatively
reactive nitrogen products. Given its wide dis- regulate various functions in the respiratory
tribution within the lung and airway, it is not tract from bronchomotor tone to immunologic/
surprising that NO can be detected in exhaled inammatory responses:
gas in levels that we now know vary in health a. eNOS is expressed in human bronchial
and disease. epithelium [6]and in type II human alveolar
This chapter reviews the formation of epithelial cells [7, 8].
NO, novel approaches of its measurements, and
its role in the pathobiology and the manage- b. nNOS is localized in the airway nerves and
ment of asthma and chronic obstructive pulmo- in the airway smooth muscle where NO is
nary disease (COPD). the major mediator for the neuronal smooth
muscle relaxation [9], mediated by the
inhibitory nonadrenergic, noncholinergic
(iNANC) system [10]. In the human lung
SOURCE OF NO IN ASTHMA AND COPD nNOS is expressed in capillary endothelial
cells of alveolar septa and plays a role as an
endothelium-derived regulator of capillary
The enzyme system responsible for producing permeability, a modulator of cholinergic
NO is NO synthase (NOS), consists of at least neuronal transmission, or an inhibitor of

363
Asthma and COPD: Basic Mechanisms and Clinical Management

platelet aggregation [11]. These low NO concentrations modied by glutathione S-transferase or glutathione per-
produced by nNOS are detectable in exhaled air [12], oxidase via either of the following:
and contributes to bronchial responsiveness [13]. We
a. converting NO2 to NH2 in tyrosine residues
recently found that nNOS is upregulated with increased
disease severity and dominant isoform as a source of NO b. denitrating NO2 directly/indirectly in tyrosine residues
in COPD (Ito et al., in preparation). c. changing SNO to SH in cysteine residues, or
c. iNOS is expressed in alveolar type II endothelial cells, denitrosation.
lung broblasts, airway and vascular smooth muscle
Nitration of tyrosine residues by lipopolysaccha-
cells, airway epithelial cells, mast cells, endothelial cells,
ride (LPS) [30], for example, induces the formation of
neutrophils, and chondrocytes.
hydrophilic negatively charged nitrotyrosine residue which
The increased levels of exhaled NO in asthma, for alters the function [31], catalytic activity, structure, sus-
example, have a predominant lower airway origin and ceptibility to proteolytic degradation, and reduces the
are most likely due to activation of NOS2 in airway kinase substrate eciency of proteins [3234]. Nitration of
epithelial and inammatory cells [1418], with a small mitochondrial proteins may change the activity of several
contribution from NOS [14]. enzymes involved in energy production (glutamate dehy-
Exhaled NO levels in stable COPD [19, 20] are lower drogenase), or in the electron transport chain (cytochrome
than in either smoking or nonsmoking asthmatics oxidase and ATPase), or in energy production (creatine
[21] and are not dierent from normal subjects. kinase) [30] (Fig. 30.1).
This reduction in exhaled NO is due to the eect of Interestingly, both energy production and apoptosis
tobacco smoking, which downregulates eNOS [22] are aected by NO and related oxides [35] and are dierent
and reduces exhaled NO [23], suggesting that this in asthma and COPD.
may contribute to the high risk of pulmonary and Nonasthmatic lungs showed little or no nitrotyrosine
cardiovascular disease in cigarette smokers. In addition staining, whereas lungs of patients even with mild asthma
to the eects of cigarette smoking, a relatively low [15] and especially who died of status asthmaticus [36]
value of exhaled NO in COPD may reect more have high presence of nitrotyrosine in both the airways and
peripheral inammation than in asthma, low NOS2 the lung parenchyma. Nevertheless, nitrotyrosine formation
expression [24], and increased oxidative stress that may in airway epithelial and inammatory cells is signicantly
consume NO in the formation of peroxynitrite [25]. higher in COPD than in asthma [37] and is related to
COPD severity [38]. This exaggerated level of nitration may
explain low protective properties of antioxidant enzymes
[30] in the central bronchial epithelium in COPD [39].
On the other hand, protein nitration may be also
POTENTIAL MECHANISMS OF NO-RELATED benecial:
DYSFUNCTION IN ASTHMA AND COPD a. Surfactant protein A (SP-A), a product of tyrosine
nitration of human pulmonary surfactant, downregulates
T-cell-dependent alveolar inammation and protect
Nitration of proteins against idiopathic pneumonia injury [40].
NO production and release are directly linked to the forma- b. Tyrosine nitration in proteins is also sucient to induce
tion of reactive nitrogen species (RNS) that modify proteins an accelerated degradation of the modied proteins by
in asthma and COPD. The magnitude of this modica- the proteasome, which may be critical for the removal of
tion correlates with the degree of oxidative and nitrosative nitrated proteins in vivo [41].
stresses. c. Nitration of tyrosine residues in tyrosine kinase
Protein nitration is unique among posttranslational substrates may prevent phosphorylation and, therefore,
modications in its dependency on reactivity of tyrosine inhibit tyrosine kinase function in cellular signaling [27];
residues in the protein target that may be achieved: this mechanism remains highly speculative.
d. Despite the discovery of enzymatic nitrotyrosine
a. by peroxynitrite (formed from the reaction between NO dinitrase activity in lung homogenates [42], suggesting
and O2), the potential role of nitration (and dinitration) as a
b. through the reaction of NO with protein tyrosyl radicals, signaling mechanism, it remains to be established
c. by the reaction of nitrite with peroxidases [26, 27] whether nitrotyrosine is merely a biomarker of increased
including myeloperoxidase and eosinophil peroxidase [28], nitrosative stress or whether it actively contributes to
cellular dysfunction and development of the airway
d. by combination of all the above pathways. inammatory processes in asthma or COPD.
Nitration of proteins by peroxynitrite is a concentra-
tion-dependent process related to formation of two dis- Tyrosine hydroxylase
tinctive forms of nitrated proteins: stable 3-nitrotyrosine
(nitration) and labile S-nitrosocysteine (S-nitrosation) [29]. Tyrosine hydroxylase (TH), the enzyme which catalyses the
Both of these nitrated proteins can be further enzymatically production of L-dihydroxyphenylalanine (L-DOPA) is also

364
Nitric Oxide 30
MnSOD Oxidative stress

COX2 PGs

Prostacyclin PGI2
synthase
O2  NO Peroxynitrite
Tyrosine NE2
hydroxylate

Glutamine
Glutamatine
syntase

IL-8 chemotactic activity

Myelo- Tyrosine Glucocorticoid Steroid sensitive


peroxidase nitration receptor or or resistance

HDAC2 Steroid insensitive, inflammation


FIG. 30.1 Effect of
peroxynitrite or impact of
NF-B or Inflammation or anti-inflammation
tyrosine nitration on various
Activate procollagenase molecules. PG: prostaglandin,
MMP3
Degradation of geratine, collagen, etc. NE: norepinephrine.

reported to be inactivated following tyrosine nitration [43]. maximum at about 6 days [46]. This suggests that presence
The loss in TH activity and expression is thought to con- of nitrated histones in tissues may reect the long-term
tribute to the L-DOPA deciency observed in certain neu- exposure to RNS [47].
rological disorders such as Parkinsons disease [43, 44].
Repeated exposures to cigarette smoke cause a variety
of molecular neuroadaptations in the cAMP signaling path- Histone deacetylase
way in postmortem tissue from the brains of human smok-
ers and former smokers [45]. Activity levels of two major Inducible NOS and other NF-B-dependent genes
components of cAMP signaling, cAMP-dependent protein involved in inammation may be regulated by the specic
kinase A (PKA), and adenylate cyclase, have been found recruitment of histone deacetylases (HDAC) [48], that may
abnormally elevated in nucleus accumbens of smokers and further enhance cytokine induction of both the iNOS and
in ventral midbrain dopaminergic region of both smokers the NF-B. HDAC activity/expression is reduced in air-
and former smokers. Although protein levels of other can- ways of mild asthmatics [49] compared with normal sub-
didate neuroadaptations, including glutamate receptor sub- jects, but may be restored by steroids that activate histone
units, TH, and other protein kinases, were within normal acetyltransferases [49]. It has been shown that in subjects
range, protein levels of the catalytic subunit of PKA were with severe asthma and COPD with reduced HDAC activ-
correspondingly higher in the ventral midbrain dopaminer- ity, the ability of inhaled steroids to control inammation
gic region of both smokers and former smokers [45]. These may be lost [49].
ndings suggest that smoking-induced brain neuroadapta- Molecular mechanisms underlying this reduction in
tions can persist for signicant periods in former smokers. HDAC2 activity is currently believed to be linked to nitra-
tive stress. Peroxynitrite has been reported to alter HDAC2
function and stability via nitration of tyrosine residues on
Histones proteins in vitro and in vivo [50, 51].

Histones are appreciably more stable than most cellular pro-


teins and their slower turnover may permit them to accu- Fate of nitrated protein
mulate 3-nitrotyrosine more than high turnover proteins
[46]. Since nitrotyrosine can readily be detected immuno- Protein nitration is reported to be more sensitive to
histochemically, nitrated histones may prove to be useful as trypsinization, and TH, once nitrated by peroxynitrite, is
a marker of extended exposure of cells or tissues to reactive eliminated by proteasomal degradation [41, 52]. An increase
nitrogen oxygen species. in free nitrotyrosine is also an evidence of nitrated protein
The fact that only a limited number of the tyrosine degradation. Oxidative/nitrative stress may delay the deg-
residues were nitrated in histones [46] allows us to consider radation of nitrated proteins due to the inactivation of the
nitrotyrosine as one of the posttranslational modications of proteasome [53].
histones that can occur in vivo. Nitration of histones using Interestingly, protein tyrosine nitration is a reversible
Mutatect cells (contain a relatively low level of nitrated pro- process catalysed by an unknown enzyme [54] that removes
teins) cultured under standard conditions was not apparent the nitro (NO2) group on nitrated proteins and has been
until 3 days of sodium nitroprusside exposure, reaching a referred to as a denitrase [42, 55].

365
Asthma and COPD: Basic Mechanisms and Clinical Management

We have found that denitrase activity in human (Fig. 30.2) that allows FENO measurements with sucient
cells and lung tissue is reduced in COPD (Ito and Osoata, accuracy and reproducibility in both children and adults
unpublished), suggesting that reduced denitration activity [68, 69], may considerably change current management
could be involved in accumulation of n-tyrosine in the lung of asthma, as FENO can be measured on a daily basis by
as well as elevated NO production. patients at home and frequently during their regular visits
to their general practitioner [62].

Asthma
MEASUREMENT OF NO IN EXHALED AIR
Increased levels of exhaled NO of a predominant lower air-
way origin [70], have been widely documented in patients
Several techniques are currently used to measure the frac- with asthma [17, 71], and are likely related to activation of
tion of NO in the exhaled air (Fig. 30.2); these are pub- NOS2 in airway epithelial and inammatory cells [1418],
lished in internationally recognized guidelines [56, 57]. The with a small contribution from NOS1 [4].
major issues are to control the expiratory ow rate [58] and
to use an orapharnygeal pressure that prevents contamina-
tion of the expirate with high NO gas derived from the Diagnosis
nasopharynx [59].
Measurement of exhaled NO from a single expiration The diagnostic value of exhaled NO measurements (with
at a xed ow are simple, highly reproducible [60], have 90% specicity and 95% positive predictive value) to dif-
been used to monitor FENO in asthma research [61] and are ferentiate between healthy subjects with or without respi-
now moving into clinical practice [62]. ratory symptoms and patients with conrmed asthma has
Methods for measuring exhaled NO at multiple been reported by several groups [72, 73]. This suggests that
expiratory ows (ME FENO) [63, 64] have been used to the simple measurement of exhaled NO can be used as an
detect elevated levels of alveolar NO in asthma [65, 66] additional diagnostic tool for the screening of patients with
leading to the renement of analytical methods to poten- a suspected diagnosis of asthma. Importantly, normal values
tially discriminate exhaled NO sources in the lung [67] of FENO for adults have been established and can be pre-
(Fig. 30.2). dicted on the basis of age and height [74].
Until recently, no day-to-day and home FENO moni- It has been conrmed that the diagnostic utility of
toring was possible, as portable and simple NO analysers exhaled NO and induced sputum are superior to the con-
were not available. The arrival of the rst hand-held port- ventional tests (peak ow measurements, spirometry, and
able NO analyser (NIOX MINO, Aerocrine, Sweden) changes in these parameters after a trial of steroid) [75],

Primary ciliary dyskinesia, CF, allergic rhinitis


Diagnosis (PCD), CF (?), management (?)
Upper airways

Nasal
technique
Stationary NO analyser
Nasal NO

Small airways Larger airways

MEF SEF
technique technique

Small airways Larger airways


COPD, severe asthma Asthma diagnosis, monitoring
Disease severity (?), management (?) Asthma management (hospital, GP, home)

Alveolar NO Bronchial NO

Stationary NO analyser Stationary Portable


NO analyser

FIG. 30.2 Exhaled NO: A marker of airway (SEF technique) and lung inflammation (MEF technique). MEF: multiple exhalation flow; SEF: single
exhalation flow.

366
Nitric Oxide 30
with exhaled NO being most advantageous because the test nebulized budesonide [81], or within 23 days [16, 82]
is quick and easy to perform. after treatment with inhaled corticosteroids (ICS) from a
metered-dose or dry-powder inhaler.
We observed that the onset of action of inhaled
Asthma control budesonide on exhaled NO was dose dependent, both
within the initial phase (rst 35 days of treatment) and
Monitoring of asthma may be much more conclusive [76] during treatment weeks 1, 2, and 3 [82]. Cessation of the
when repetitive FENO measurements, especially made at actions of ICS is a rapid process too and can also be moni-
home by a portable NO analyzer, instead of single assess- tored by repetitive NO during the rst 35 days in patients
ment were used, changes in FENO correlates signicantly not who stopped inhaling budesonide [82].
only with changes in sputum eosinophils and hyperrespon-
siveness, but also with lung function and asthma symptoms.
Jones et al. [77] have demonstrated that repetitive Dose dependency
exhaled NO measurements have a positive predictive value We have shown that the acute reduction in exhaled NO
between 80% and 90% for predicting and diagnosing loss (within the rst 35 days of treatment) and the chronic
of control in asthma and are as useful as induced sputum reduction (days 721) are dose dependent in patients with
eosinophils and airway hyperresponsiveness to hypertonic mild asthma who are treated with low doses of budesonide
saline, but with the enormous advantage that they are easy [82]. Serial exhaled NO measurements, as we recently sug-
to perform. gested [61], may therefore be useful in studying the onset
An advantage of FENO as a loss-of-control-marker and duration of action of ICS, as well as in monitoring
[78] is that increase in FENO and asthma symptoms may be patient compliance.
seen before any signicant deterioration in airway hyper- It is unclear, however, whether exhaled NO can guide
responsiveness, sputum eosinophils, or lung function during an asthma treatment strategy. Thus, recent study based on
asthma exacerbation induced by steroid reduction [18, 79]. the measurement of exhaled NO did not result in a large
Exhaled NO levels were (median, interquartile range) 11 ppb reduction in asthma exacerbations, but resulted in more
(921) in children who had good asthma control, 15 ppb patients on lower doses of ICS over 12 months, when com-
(1126) acceptable asthma control, and 28 ppb (1933) with pared with management by current asthma guidelines and
insuciently controlled asthma [80], suggesting NO measure- no measure of FENO [83].
ments may be useful for monitoring pediatric asthma in clinic.

iNOS inhibitors
Inhaled corticosteroids
The ux of NO through the airways can be decreased by a
Onset and cessation of action single dose of inhaled NOS inhibitors in both asthma and
Exhaled NO behaves as a rapid response marker that is healthy controls subjects with a greater reduction in patients
extremely sensitive to steroid treatment, because it may with asthma (80 versus 61%) [84] (Fig. 30.3). This nd-
be signicantly reduced even 6 h after a single dose of ing supports the hypothesis that iNOS induction may be

Alveolar NO Bronchial NO

5 5 2500 2500

4 4 2000 2000
JNO (pL /s)

JNO (pL /s)


Calv (ppb)

Calv (ppb)

3 3 1500 1500
NS
2 2 1000 1000
84%
1 1 NS 500 500

p  0.0001 p  0.0001
0 0 0 0

0 60 0 60 0 60 0 60
Time (min) Time (min) Time (min) Time (min)
Asthma Healthy Asthma Healthy

Aminoguanidine Placebo

FIG. 30.3 Fast and differential effect of inhaled aminoguanidine versus placebo on bronchial NO and alveolar NO in asthma and healthy.
From Ref. [84].

367
Asthma and COPD: Basic Mechanisms and Clinical Management

p  0.05 p  0.05

4 3000
Alveolar NO Bronchial NO
3
2000

JNO (pL/s)
Calv (ppb)

1000
1

0 0
2

ild

re

ild

re
lth

lth
1

at

at
ve

ve
M

M
er

er
ea

ea
LD

LD

Se

LD

LD

Se
od

od
H

H
O

O
M

M
G

G
COPD Asthma COPD Asthma

FIG. 30.4 Exhaled NO measured with the MEF technique: Alveolar versus bronchial inflammation is asthma and COPD of different stage and severity.
From Ref. [84, 90].

responsible for the raised levels of NO in asthma, whereas [91] (Fig. 30.4) suggesting similar mechanisms may be
the levels seen in normal subjects seem due to cNOS and responsible.
a low level of iNOS activity.

MONITORING OF SMALL AIRWAY


COPD INFLAMMATION IN BOTH COPD AND
SEVERE ASTHMA
Exhaled NO levels in stable COPD [19, 20] are lower than
in either smoking or nonsmoking asthmatics [21] and are not
dierent from normal subjects. Patients with unstable COPD, MEFT FENO measurements are highly reproducible, free
however, have high NO levels compared with stable smokers of diurnal variation, unaected by smoking, bronchodilator,
or ex-smokers with COPD [19], which are still lower than or ICS [90] and therefore could be useful for COPD moni-
expired NO in nonsteroid-treated asthmatic subjects [85]. toring (Fig. 30.5).
Data from experiments using multiple expiratory Interestingly, the elevated levels of alveolar NO are sim-
ows have demonstrated that the peripheral airways/alve- ilar in COPD [90] and severe asthma [91], supporting our
olar region is the predominant source of elevated exhaled previous nding of elevated exhaled FENO levels in patients
NO in COPD. In contrast, increased exhaled NO levels in with severe [92] and dicult steroid-resistant asthma [93].
asthma (Fig. 30.4) are mainly of larger airways/bronchial Therefore, dierential ow analysis of exhaled NO
origin [86]. Prevalence of alveolar-derived NO in COPD is provides additional information about the sites of inam-
possibly related to the iNOS in macrophages, alveolar walls, mation in asthma and COPD which are clearly dierent
and bronchial epithelium of COPD patients [87]. (Figs. 30.3, 30.4, 30.6), may be useful in assessing the
response of peripheral inammation to therapy, including
its combination with iNOS inhibitors [94].
Disease severity and progression
Severity-related increase of the inferred concentration of iNOS inhibitors
NO in the alveolar zone, Calv, may be due to higher pres-
ence of iNOS-positive cells in alveolar walls in more severe Under conditions of inammation and oxidative stress,
COPD patients [88], but not with severe emphysema superoxide anion will preferentially react with available NO
that shows a lower percentage of iNOS-positive alveo- rather than its endogenous neutralizer superoxide dismutase,
lar macrophages than patients with milder disease [89]. thus increasing the formation of peroxynitrite in tissues [95].
Patient with COPD rated as stage Global Initiative for Diusion of peroxynitrite through biomembranes can
Chronic Obstructive Lung Disease II (GOLD II) have Calv cause oxidative damage at one to two cell diameters from its
levels [90] similar to those of more severe asthma patients site of formation [96] that may be of importance in COPD

368
Nitric Oxide 30
COPD severity
p  0.0001
5

4 GOLD 34
GOLD 12
GOLD 0

Calv (ppb)
3
GOLD 4 GOLD 3 GOLD 2 GOLD 0 1, Smokers

6 2

5 Smokers 1

GOLD 0
4 0
Calv (ppb)

Non-
GOLD 1 smokers
3 GOLD 2
GOLD 3 Smoking ICS
2 5
GOLD 4
1 r 0.6; p  0.0001 4 Smokers Steroids

Calv (ppb)
3
0 20 40 60 80 100 120
FEV1 (% pred) 2

0
Ex Current No Yes

FIG. 30.5 Alveolar NO in COPD: relationship to the disease stage and severity, smoking and treatment with inhaled corticosteroids. ICS: inhaled
corticosteroids. From Ref. [90].

Alveolar NO Bronchial NO with its high local NO production [90]. Therefore, the use
of iNOS inhibitors by reducing available NO may restore
5 2500
the preferred pathway of superoxide radicals detoxication,
via superoxide dismutase.
4 2000
We have reported this selective inhibition by amino-
guanidine versus placebo of bronchial NO ux ( JNO) in
JNO (pL /s)
Calv (ppb)

3 1500
COPD patients but not in smokers or normal volunteers [97]
2 1000
(Fig. 30.6). Aminoguanidine also caused a decrease in alveolar
p  0.01 (COPD) NO (Calv) in smokers and in COPD but not in healthy con-
p  0.001

1 32% 500
trols. These ndings give potential new insights into the dif-
ferences between asthma (Fig. 30.3) and COPD (Fig. 30.6).
0 0

0 60 0 60 SUMMARY
Time (min) Time (min)
COPD and smokers COPD and smokers
The precise inammatory pathways and cells, NOS enzyme
isoforms, and anatomical compartments responsible for
Aminoguanidine (COPD) production of the NO captured in the expirate are areas of
Aminoguanidine (smokers) active investigation (Fig. 30.7).
Placebo There are two interlinked and equally important
future areas of NO research: exploring further potential and
FIG. 30.6 Fast and differential effect of inhaled aminoguanidine versus prevalent sources of NO and its role in dierent diseases
placebo on bronchial and alveolar NO in COPD and healthy smokers. From and the use of NO in clinical medicine, including home
Ref. [97]. measurements and routine asthma monitoring.

369
Asthma and COPD: Basic Mechanisms and Clinical Management

Normal Asthma COPD

Exhaled NO Alveolar Bronchial Bronchial (JNO) Alveolar (Calv)

4 3000 3000 4

3 3
2000 2000

JNO (pL/s)
Calv (ppb)

JNO (pL/s)

Calv (ppb)
2 2

1000 1000
1 1

0 0 0 0
y

ild

re

ild

re
lth

lth

at

at
ve

ve
M

M
er

er
ea

ea

LD

LD

Se

LD

LD

Se
Sources od

od
H

O
M

M
G

G
eNOS
nNOS
iNOS or

FIG. 30.7 Prevalent anatomical and enzymatic sources of NO in exhaled air of normal subjects and patients with asthma and COPD.

References 8. Pechkovsky DV, Zissel G, Goldmann T, Einhaus M, Taube C,


Magnussen H, Schlaak M, Muller-Quernheim J. Pattern of NOS2 and
1. Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for NOS3 mRNA expression in human A549 cells and primary cultured
the biological activity of endothelium-derived relaxing factor. Nature AEC II. Am J Physiol Lung Cell Mol Physiol 282(4): L68492, 2002.
327(6122): 52426, 1987. 9. Fischer A, Homann B. Nitric oxide synthase in neurons and nerve
2. Ialenti A, Ianaro A, Moncada S, Di Rosa M. Modulation of acute bers of lower airways and in vagal sensory ganglia of man. Correlation
inammation by endogenous nitric oxide. Eur J Pharmacol 211(2): 177 with neuropeptides. Am J Respir Crit Care Med 154(1): 20916, 1996.
82, 1992. 10. Widdicombe JG. Autonomic regulation. i-NANC/e-NANC. Am J
3. Guo FH, De Raeve HR, Rice TW, Stuehr DJ, Thunnissen FB, Erzurum Respir Crit Care Med 158(5 Pt 3): S17175, 1998.
SC. Continuous nitric oxide synthesis by inducible nitric oxide synthase 11. Luhrs H, Papadopoulos T, Schmidt HH, Menzel T. Type I nitric oxide
in normal human airway epithelium in vivo. Proc Natl Acad Sci USA synthase in the human lung is predominantly expressed in capillary
92(17): 780913, 1995. endothelial cells. Respir Physiol 129(3): 36774, 2002.
4. Wechsler ME, Grasemann H, Deykin A, Silverman EK, Yandava CN, 12. De Sanctis GT, Mehta S, Kobzik L, Yandava C, Jiao A, Huang PL,
Israel E, Wand M, Drazen JM. Exhaled nitric oxide in patients with Drazen JM. Contribution of type I NOS to expired gas NO and
asthma. Association with NOS1 genotype. Am J Respir Crit Care Med bronchial responsiveness in mice. Am. J Physiol 273(4 Pt 1): L88388,
162: 204347, 2000. 1997.
5. Morris SM Jr, Billiar TR. New insights into the regulation of 13. Jang AS, Choi IS, Lee JU, Park SW, Lee JH, Park CS. Changes in the
inducible nitric oxide synthesis. Am. J Physiol 266(6 Pt 1): E82939, expression of NO synthase isoforms after ozone: The eects of allergen
1994. exposure. Respir Res 5: 5, 2004.
6. Shaul PW, North AJ, Wu LC, Wells LB, Brannon TS, Lau KS, Michel 14. Hamid Q, Springall DR, Riveros-Moreno V, Chanez P, Howarth PH,
T, Margraf LR, Star RA. Endothelial nitric oxide synthase is expressed Redington A, Bousquet J, Godard P, Holgate S, Polak JM. Induction
in cultured human bronchiolar epithelium. J Clin Invest 94(6): 223136, of nitric oxide synthase in asthma. Lancet 342(8886-8887): 151013,
1994. 1993.
7. Pechkovsky DV, Zissel G, Stamme C, Goldmann T, Ari JH, Einhaus 15. Saleh D, Ernst P, Lim S, Barnes PJ, Giaid A. Increased formation of
M, Taube C, Magnussen H, Schlaak M, Muller-Quernheim J. Human the potent oxidant peroxynitrite in the airways of asthmatic patients
alveolar epithelial cells induce nitric oxide synthase-2 expression in alve- is associated with induction of nitric oxide synthase: Eect of inhaled
olar macrophages. Eur Respir J 19(4): 67283, 2002. glucocorticoid. FASEB J 12(11): 92937, 1998.

370
Nitric Oxide 30
16. Kharitonov SA, Yates DH, Barnes PJ. Inhaled glucocorticoids decrease 38. Ricciardolo FL, Caramori G, Ito K, Capelli A, Brun P, Abatangelo G,
nitric oxide in exhaled air of asthmatic patients. Am J Respir Crit Care Papi A, Chung KF, Adcock I, Barnes PJ, Donner CF, Rossi A, Di
Med 153(1): 45457, 1996. Stefano A. Nitrosative stress in the bronchial mucosa of severe chronic
17. Kharitonov SA, Yates DH, Robbins RA, Logan-Sinclair R, obstructive pulmonary disease. J Allergy Clin Immunol 116(5): 102835,
Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of 2005.
asthmatic patients. Lancet 343(8890): 13335, 1994. 39. Harju T, Kaarteenaho-Wiik R, Soini Y, Sormunen R, Kinnula VL.
18. Kharitonov SA, Yates DH, Chung KF, Barnes PJ. Changes in the Diminished immunoreactivity of gamma-glutamylcysteine synthetase
dose of inhaled steroid aect exhaled nitric oxide levels in asthmatic in the airways of smokers lung. Am J Respir Crit Care Med 166(5):
patients. Eur Respir J 9: 196201, 1996. 75459, 2002.
19. Maziak W, Loukides S, Culpitt SV, Sullivan P, Kharitonov SA, Barnes 40. Yang S, Milla C, Panoskaltsis-Mortari A, Ingbar DH, Blazar BR,
PJ. Exhaled nitric oxide in chronic obstructive pulmonary disease. Am J Haddad IY. Human surfactant protein a suppresses T cell-dependent
Respir Crit Care Med 157(3 Pt 1): 9981002, 1998. inammation and attenuates the manifestations of idiopathic pneumo-
20. Robbins RA, Floreani AA, Von Essen SG, Sisson JH, Hill GE, nia syndrome in mice. Am J Respir Cell Mol Biol 24(5): 52736, 2001.
Rubinstein I, Townley R. Measurement of exhaled nitric oxide by three 41. Souza JM, Choi I, Chen Q, Weisse M, Daikhin E, Yudko M, Obin M,
dierent techniques. Am J Respir Crit Care Med 153: 163135, 1996. Ara J, Horwitz J, Ischiropoulos H. Proteolytic degradation of tyrosine
21. Verleden GM, Dupont LJ, Verpeut AC, Demedts MG. The eect of nitrated proteins. Arch Biochem Biophys 380(2): 36066, 2000.
cigarette smoking on exhaled nitric oxide in mild steroid- naive asth- 42. Kamisaki Y, Wada K, Bian K, Balabanli B, Davis K, Martin E, Behbod F,
matics. Chest 116(1): 5964, 1999. Lee YC, Murad F. An activity in rat tissues that modies nitrotyrosine-
22. Su Y, Han W, Giraldo C, De Li Y, Block ER. Eect of cigarette smoke containing proteins. Proc Natl Acad Sci USA 95(20): 1158489, 1998.
extract on nitric oxide synthase in pulmonary artery endothelial cells. 43. Blanchard-Fillion B, Souza JM, Friel T, Jiang GC, Vrana K, Sharov V,
Am J Respir Crit Care Med 19: 81925, 1998. Barron L, Schoneich C, Quijano C, Alvarez B, Radi R, Przedborski S,
23. Kharitonov SA, Robbins RA, Yates D, Keatings V, Barnes PJ. Acute Fernando GS, Horwitz J, Ischiropoulos H. Nitration and inactivation
and chronic eects of cigarette smoking on exhaled nitric oxide. Am J of tyrosine hydroxylase by peroxynitrite. J Biol Chem 276(49): 46017
Respir Crit Care Med 152(2): 60912, 1995. 23, 2001.
24. Rutgers SR, van der Mark TW, Coers W, Moshage H, Timens W, 44. Souza JM, Chen Q, Blanchard-Fillion B, Lorch SA, Hertkorn C,
Kauman HF, Ko Postma,DS. Markers of nitric oxide metabolism in Lightfoot R, Weisse M, Friel T, Paxinou E, Themistocleous M, Chov S,
sputum and exhaled air are not increased in chronic obstructive pulmo- Ischiropoulos H. Reactive nitrogen species and proteins: Biological sig-
nary disease. Thorax 54(7): 57680, 1999. nicance and clinical relevance. Adv Exp Med Biol 500: 16974, 2001.
25. Eiserich JP, Hristova M, Cross CE, Jones AD, Freeman BA, Halliwell B, 45. Hope BT, Nagarkar D, Leonard S, Wise RA. Long-term upregulation
van-der VA. Formation of nitric oxide-derived inammatory oxidants by of protein kinase A and adenylate cyclase levels in human smokers.
myeloperoxidase in neutrophils. Nature 391(6665): 39397, 1998. J Neurosci 27(8): 196472, 2007.
26. Gunther MR, Sturgeon BE, Mason RP. Nitric oxide trapping of the 46. Haqqani AS, Kelly JF, Birnboim HC. Selective nitration of histone
tyrosyl radical-chemistry and biochemistry. Toxicology 177(1): 19, tyrosine residues in vivo in mutatect tumors. J Biol Chem 277(5): 3614
2002. 21, 2002.
27. Davis KL, Martin E, Turko IV, Murad F. Novel eects of nitric oxide. 47. Hanazawa T, Kharitonov SA, Barnes PJ. Increased nitrotyrosine in
Ann Rev Pharmacol Toxicol 41: 20336, 2001. exhaled breath condensate of patients with asthma. Am J Respir Crit
28. Eiserich JP, Baldus S, Brennan ML, Ma W, Zhang C, Tousson A, Care Med 162(4 Pt 1): 127376, 2000.
Castro L, Lusis AJ, Nauseef WM, White CR, Freeman BA. 48. Ito K, Adcock IM. Histone acetylation and histone deacetylation.
Myeloperoxidase, a leukocyte-derived vascular NO oxidase. Science Mol Biotechnol 20(1): 99106, 2002.
296(5577): 239194, 2002. 49. Ito K, Caramori G, Lim S, Oates T, Chung KF, Barnes PJ, Adcock IM.
29. Kuo WN, Kocis JM. Nitration/S-nitrosation of proteins by Expression and activity of histone deacetylases in human asthmatic
peroxynitrite-treatment and subsequent modication by glutathione airways. Am J Respir Crit Care Med 166(3): 39296, 2002.
S-transferase and glutathione peroxidase. Mol. Cell Biochem. 233(1-2): 50. Ito K, Hanazawa T, Tomita K, Barnes PJ, Adcock IM. Oxidative stress
5763, 2002. reduces histone deacetylase 2 activity and enhances IL-8 gene expression:
30. Aulak KS, Miyagi M, Yan L, West KA, Massillon D, Crabb JW, Role of tyrosine nitration. Biochem Biophys Res Commun 315(1): 24045,
Stuehr DJ. Proteomic method identies proteins nitrated in vivo during 2004.
inammatory challenge. Proc Natl Acad Sci USA 98(21): 1205661, 2001. 51. Marwick JA, Kirkham PA, Stevenson CS, Danahay H, Giddings J,
31. Turko IV, Murad F. Protein nitration in cardiovascular diseases. Butler K, Donaldson K, MacNee W, Rahman I. Cigarette smoke alters
Pharmacol Rev 54(4): 61934, 2002. chromatin remodeling and induces proinammatory genes in rat lungs.
32. Berlett BS, Friguet B, Yim MB, Chock PB, Stadtman ER. Am J Respir Cell Mol Biol 31(6): 63342, 2004.
Peroxynitrite-mediated nitration of tyrosine residues in Escherichia coli 52. Gole MD, Souza JM, Choi I, Hertkorn C, Malcolm S, Foust RF III,
glutamine synthetase mimics adenylylation: Relevance to signal trans- Finkel B, Lanken PN, Ischiropoulos H. Plasma proteins modied by
duction. Proc Natl Acad Sci USA 93(5): 177680, 1996. tyrosine nitration in acute respiratory distress syndrome. Am J Physiol
33. Lennon CW, Cox HD, Hennelly SP, Chelmo SJ, McGuirl MA. Lung Cell Mol. Physiol 278(5): L96167, 2000.
Probing structural dierences in prion protein isoforms by tyrosine 53. Reinheckel T, Sitte N, Ullrich O, Kuckelkorn U, Davies KJ, Grune T.
nitration. Biochemistry 46(16): 485060, 2007. Comparative resistance of the 20S and 26S proteasome to oxidative
34. Salvemini D, Doyle TM, Cuzzocrea S. Superoxide, peroxynitrite and stress. Biochem J 335(Pt 3): 63742, 1998.
oxidative/nitrative stress in inammation. Biochem Soc Trans 34(Pt 5): 54. Gow AJ, Duran D, Malcolm S, Ischiropoulos H. Eects of peroxynitrite-
96570, 2006. induced protein modications on tyrosine phosphorylation and degrada-
35. Brown GC, Borutaite V. Nitric oxide, mitochondria, and cell death. tion. FEBS Lett 385(1-2): 6366, 1996.
IUBMB Life 52(3-5): 18995, 2001. 55. Irie Y, Saeki M, Kamisaki Y, Martin E, Murad F. Histone H1.2 is a
36. Kaminsky DA, Mitchell J, Carroll N, James A, Soultanakis R, Janssen Y. substrate for denitrase, an activity that reduces nitrotyrosine immuno-
Nitrotyrosine formation in the airways and lung parenchyma of patients reactivity in proteins. Proc Natl Acad Sci USA 100(10): 563439, 2003.
with asthma. J Allergy Clin Immunol 104(4 Pt 1): 74754, 1999. 56. Kharitonov SA, Alving K, Barnes PJ. Exhaled and nasal nitric oxide
37. Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato K. Increase in measurements: Recommendations. Eur Respir J 10: 168393, 1997.
reactive nitrogen species production in chronic obstructive pulmonary 57. ATS/ERS. Recommendations for Standardized Procedures for the
disease airways. Am J Respir Crit Care Med 162(2): 7016, 2000. Online and Oine Measurement of Exhaled Lower Respiratory

371
Asthma and COPD: Basic Mechanisms and Clinical Management

Nitric Oxide and Nasal Nitric Oxide, 2005. Am J Respir Crit Care Med 79. Jatakanon A, Lim S, Barnes PJ. Changes in sputum eosinophils predict
171(8): 91230, 2005. loss of asthma control. Am J Respir Crit Care Med 161(1): 6472, 2000.
58. Silko PE, McClean PA, Slutsky AS, Furlott HG, Hostein E, 80. Meyts I, Proesmans M, De Boeck K. Exhaled nitric oxide corresponds
Wakita S, Chapman KR, Szalai JP, Zamel N. Marked ow-dependence with oce evaluation of asthma control. Pediatr Pulmonol 36(4):
of exhaled nitric oxide using a new technique to exclude nasal nitric 28389, 2003.
oxide. Am J Respir Crit Care Med 155(1): 26067, 1997. 81. Kharitonov SA, Barnes PJ, OConnor BJ. Reduction in exhaled nitric
59. Kharitonov SA, Barnes PJ. Nasal contribution to exhaled nitric oxide oxide after a single dose of nebulised budesonide in patients with
during exhalation against resistance or during breath holding. Thorax asthma. Am J Respir Crit Care Med 153: A799, 1996.
52(6): 54044, 1997. 82. Kharitonov SA, Donnelly LE, Montuschi P, Corradi M, Collins JV,
60. Kharitonov SA, Gonio F, Kelly C, Meah S, Barnes PJ. Reproducibility Barnes PJ. Dose-dependent onset and cessation of action of inhaled
of exhaled nitric oxide measurements in healthy and asthmatic adults budesonide on exhaled nitric oxide and symptoms in mild asthma.
and children. Eur Respir J 21(3): 43338, 2003. Thorax 57(10): 88996, 2002.
61. Kharitonov SA, Barnes PJ. Exhaled markers of pulmonary disease. 83. Shaw DE, Berry MA, Thomas M, Green RH, Brightling CE,
Am J Respir Crit Care Med 163(7): 1693722, 2001. Wardlaw AJ, Pavord ID. The use of exhaled nitric oxide to guide
62. Kharitonov SA. Exhaled markers of inammatory lung diseases: Ready asthma management: A randomized controlled trial. Am J Respir Crit
for routine monitoring?. Swiss Med Wkly 134(13-14): 17592, 2004. Care Med 176(3): 23137, 2007.
63. Lehtimaki L, Turjanmaa V, Kankaanranta H, Saarelainen S, Hahtola P, 84. Brindicci C, Ito K, Barnes PJ, Kharitonov SA. Eect of an inducible
Moilanen E. Increased bronchial nitric oxide production in patients nitric oxide synthase inhibitor on dierential ow exhaled nitric oxide
with asthma measured with a novel method of dierent exhalation in asthmatic patients and healthy volunteers. Chest, 2007.
ow rates. Ann Med 32(6): 41723, 2000. 85. Massaro AF, Mehta S, Lilly CM, Kobzik L, Reilly JJ, Drazen JM.
64. Lehtimaki L, Kankaanranta H, Saarelainen S, Hahtola P, Jarvenpaa R, Elevated nitric oxide concentrations in isolated lower airway gas of
Koivula T, Turjanmaa V, Moilanen E. Extended exhaled NO measure- asthmatic subjects. Am J Respir Crit Care Med 153: 151014, 1996.
ment dierentiates between alveolar and bronchial inammation. Am J 86. Brindicci C, Cosio B, Gajdocsi R, Collins JV, Bush A, Abdallah S,
Respir Crit Care Med 163(7): 155761, 2001. Barnes PJ, Kharitonov SA. Extended exhaled NO measurements at
65. Silko PE, Sylvester JT, Zamel N, Permutt S. Airway Nitric Oxide dierent exhalation ows may dierentiate between bronchial and
Diusion in Asthma. Role in pulmonary function and bronchial respon- alveolar inammation in patients with asthma and COPD. Eur Respir
siveness. Am J Respir Crit Care Med 161(4 Part 1): 121828, 2000. J 20: 174s, 2002.
66. Lehtimaki L, Kankaanranta H, Saarelainen S, Turjanmaa V, Moilanen 87. Paska C, Maestrelli P, Formichi B, Monti S, Baldi S, Miniati M, Saetta
E. Increased alveolar nitric oxide concentration in asthmatic patients M, Fabbri LM. Increased expression of inducible NOS in peripheral
with nocturnal symptoms. Eur Respir J 20(4): 84145, 2002. lung of severe COPD patients. Eur Respir J 20: 95s, 2002.
67. George SC, Hogman M, Permutt S, Silko PE. Modeling pulmonary 88. Maestrelli P, Paska C, Saetta M, Turato G, Nowicki Y, Monti S,
nitric oxide exchange. J Appl Physiol 96(3): 83139, 2004. Formichi B, Miniati M, Fabbri LM. Decreased haem oxygenase-1 and
68. Vahlkvist S, Sinding M, Skamstrup K, Bisgaard H. Daily home meas- increased inducible nitric oxide synthase in the lung of severe COPD
urements of exhaled nitric oxide in asthmatic children during natural patients. Eur Respir J 21(6): 97176, 2003.
birch pollen exposure. J Allergy Clin Immunol 117(6): 127276, 2006. 89. van Straaten JF, Postma DS, Coers W, Noordhoek JA, Kauman HF,
69. Gill M, Gra GR, Adler AJ, Dweik RA. Validation study of fractional Timens W. Macrophages in lung tissue from patients with pulmonary
exhaled nitric oxide measurements using a handheld monitoring device. emphysema express both inducible and endothelial nitric oxide syn-
J Asthma 43(10): 73134, 2006. thase. Mod Pathol 11(7): 64855, 1998.
70. Kharitonov SA, Chung FK, Evans DJ, OConnor BJ, Barnes PJ. The 90. Brindicci C, Ito K, Resta O, Pride NB, Barnes PJ, Kharitonov SA.
elevated level of exhaled nitric oxide in asthmatic patients is mainly Exhaled nitric oxide from lung periphery is increased in COPD. Eur
derived from the lower respiratory tract. Am J Respir Crit Care Med Respir J 26(1): 5259, 2005.
153: 177380, 1996. 91. Brindicci C, Ito K, Barnes PJ, Kharitonov SA. Dierential ow analysis
71. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JM. of exhaled nitric oxide in patients with asthma of diering severity.
Expired nitric oxide levels during treatment of acute asthma. Am J Chest 131(5): 135362, 2007.
Respir Crit Care Med 152(2): 8003, 1995. 92. Jatakanon A, Lim S, Kharitonov SA, Chung KF, Barnes PJ. Correlation
72. Dupont LJ, Demedts MG, Verleden GM. Prospective evaluation of between exhaled nitric oxide, sputum eosinophils, and methacholine
the accuracy of exhaled nitric oxide for the diagnosis of asthma. Am J responsiveness in patients with mild asthma. Thorax 53(2): 9195,
Respir Crit Care Med 159: A861, 1999. 1998.
73. Dupont LJ, Demedts MG, Verleden GM. Prospective evaluation of 93. Stirling RG, Kharitonov SA, Campbell D, Robinson D, Durham SR,
the validity of exhaled nitric oxide for the diagnosis of asthma. Chest Chung KF, Barnes PJ. Exhaled NO is elevated in dicult asthma and
123(3): 75156, 2003. correlates with symptoms and disease severity despite treatment with
74. Olin AC, Bake B, Toren K. Fraction of exhaled nitric oxide at 50 mL/s: oral and inhaled corticosteroids. Thorax 53: 103034, 1998.
Reference values for adult lifelong never-smokers. Chest 131(6): 94. Kharitonov SA, Barnes PJ. Nitric oxide, nitrotyrosine, and nitric oxide
185256, 2007. modulators in asthma and chronic obstructive pulmonary disease. Curr
75. Smith AD, Cowan JO, Filsell S, McLachlan C, Monti-Sheehan G, Allergy Asthma Rep 3(2): 12129, 2003.
Jackson P, Taylor DR. Diagnosing Asthma: Comparisons between 95. Cooke CL, Davidge ST. Peroxynitrite increases iNOS through
Exhaled Nitric Oxide Measurements and Conventional Tests. Am J NF-kappaB and decreases prostacyclin synthase in endothelial cells.
Respir Crit Care Med 169(4): 47378, 2004. Am J Physiol Cell Physiol 282(2): C395402, 2002.
76. Kharitonov SA, Barnes PJ. Does Exhaled Nitric Oxide Reect Asthma 96. Szabo C, Ischiropoulos H, Radi R. Peroxynitrite: Biochemistry, patho-
Control?. Yes, it does!. Am J Respir Crit Care Med 164(5): 72728, physiology and development of therapeutics. Nat Rev Drug Discov
2001. 6: 66280, 2007.
77. Jones SL, Kittelson J, Cowan JO, Flannery EM, Hancox RJ, 97. Brindicci C, Torre O, Barnes PJ, Ito K, Kharitonov SA. Eects of
McLachlan CR, Taylor DR. The predictive value of exhaled nitric oxide aminoguanidine, an inhibitor of inducible nitric oxide synthase, on
measurements in assessing changes in asthma control. Am J Respir Crit nitric oxide production and its metabolites in healthy controls, healthy
Care Med 164(5): 73843, 2001. smokers and COPD patients. Chest, 2008.
78. Kharitonov SA. Exhaled nitric oxide and carbon monoxide in asthma.
Eur Respir J 9(68): 21218, 1999.

372
Transcription Factors
31 CHAPTER

INTRODUCTION Several new compounds based on inter- Ian M. Adcock and


acting with specic transcription factors or their Gaetano Caramori
activation pathways are now in development for
Inammation is a central feature of asthma and the treatment of asthma and COPD, and some National Heart and Lung Institute,
chronic obstructive pulmonary disease (COPD). drugs already in clinical use (such as glucocor- Imperial College School of Medicine,
The specic characteristics of the inammatory ticoids) are thought to work via transcription London, UK
response in each disease and the site of inam- factors. Glucocorticoids are eective therapy in
mation dier, but both involve the recruitment the long-term control of asthma. One of their
and activation of inammatory cells and changes mechanisms of action in asthmatic airways is
in the structural cells of the lung [1]. Asthma by inhibiting the action of transcription fac-
and COPD are characterized by an increased tors that regulate inammatory gene expression
expression of components of the inammatory [5]. Glucocorticoids may also have a benecial
cascade. These inammatory proteins include eect in COPD during exacerbations, although
cytokines, chemokines, growth factors, enzymes, this is less marked than in asthma, indicating
receptors, and adhesion molecules [2]. The that dierent genes and transcription factors
increased expression of these proteins seen in are involved and emphasizing the importance
asthma and COPD is the result of enhanced of cell-specic transcription factors.
gene transcription since many of the genes are One concern about this approach is the
not expressed in normal cells but are induced in specicity of such drugs, but it is clear that
a cell-specic manner during the inammatory transcription factors have selective eects on
process [2]. the expression of certain genes and this may
Changes in gene transcription are regu- make it possible to be more selective. In addi-
lated by transcription factors, which are pro- tion, there are cell-specic transcription factors
teins that bind to DNA and modulate the that may be targeted for inhibition, which could
transcriptional apparatus. Transcription factors provide selectivity of drug action. One such
regulate the expression of many genes, includ- example is GATA3, which has been reported
ing inammatory genes, and may play a key role to have a restricted cellular distribution [6]. In
in the pathogenesis of asthma and COPD since asthma and COPD it may be possible to tar-
they regulate the increased gene expression that get drugs to the airways by inhalation, as is cur-
may underlie the acute and chronic inamma- rently performed for inhaled glucocorticoids, to
tory mechanisms that characterize these dis- systemic eects.
eases [1, 3]. Transcription factors may amplify Despite the fact that many transcription
and perpetuate the inammatory process, so it factors have now been discovered there is still
is possible that abnormal functioning of tran- a paucity of data concerning the regulation of
scription factors may determine disease severity transcription factors in the human lungs. This
and response to treatment. The increased under- chapter briey reviews the physiological func-
standing of the role of transcription factors in tion of the transcription factors in the normal
the pathogenesis of asthma and COPD has also cells and the role of some transcription factors
opened an opportunity for the development of that may be relevant in the pathogenesis of
new potential anti-inammatory drugs [4]. asthma and COPD.

373
Asthma and COPD: Basic Mechanisms and Clinical Management

TRANSCRIPTION FACTORS nucleus [5]. It is becoming clear that post-translational


modications of transcription factors by phosphoryla-
tion, acetylation, and nitration can have profound eects
Transcription factors are proteins that bind to DNA-regu- on either the DNA-binding capacity or the transcriptional
latory sequences (enhancers and silencers), usually local- activity of a transcription factor [10]. This is exemplied
ized in the 5
-upstream region of target genes, to modulate by changes in p65 NF-B phosphorylation and acetyla-
the rate of gene transcription. This may result in increased tion which alter its association with transcriptional coacti-
or decreased gene transcription, protein synthesis, and sub- vator and repressor complexes resulting in changes in gene
sequent altered cellular function. Many transcription fac- expression prole [1113].
tors have now been identied and a large proportion of the
human genome appears to code for these proteins.
Several families of transcription factors exist and Cross-talk
members of each family may share structural characteristics.
These families include One of the most important concepts to have emerged is
helix-turn-helix (e.g. Oct-1), the demonstration that transcription factors may physically
interact with each other to form homodimers or heterodim-
helix-loop-helix (e.g. E2A), ers, resulting in inhibition or enhancement of transcrip-
zinc nger (e.g. glucocorticoid receptors, GATA tional activity at a site distinct from the consensus target for
proteins), a particular transcription factor (Fig. 31.1). This then allows
basic protein-leucine zipper [cyclic AMP response cross-talk between dierent signal transduction pathways
element-binding factor (CREB), activator protein-1 at the level of gene expression. Generally it is necessary to
(AP-1)], have coincident activation of several transcription factors
in order to have maximal gene expression. This may explain
-sheet motifs [e.g. nuclear factor-B (NF-B)] how transcription factors that are ubiquitous may regulate
[4, 7] (Table 31.1). particular genes in certain types of cells [4, 10].
Many transcription factors are common to several The complexity of the activation pathways and their
cell types (ubiquitous), such as AP-1 and NF-B, and ability to engage in cross-talk enables cells to overcome
may play a general role in the regulation of inammatory inhibition of one pathway and retain a capacity to activate
genes, whereas others are cell-specic and may determine specic transcription factors. This cross-talk and redundancy
the phenotypic characteristics of a cell. Transcription fac- may also hinder the search for novel anti-inammatory
tor activation is complex and may involve multiple intrac- agents targeted toward transcription factor activation.
ellular signal transduction pathways, including the kinases Binding of transcription factors to their specic bind-
PKA, MAPKs, JAKs, and PKCs, stimulated by cell-surface ing motifs in the promoter region may alter transcription
receptors [8, 9]. Transcription factors may also be directly by interacting directly with components of the basal tran-
activated by ligands such as glucocorticoids and vitamins scription apparatus or via cofactors that link the transcrip-
A and D [5]. Transcription factors may therefore convert tion factor to the basal transcription apparatus [14]. Large
transient environmental signals at the cell surface into long- proteins that bind to the basal transcription apparatus bind
term changes in gene transcription, thus acting as nuclear many transcription factors and thus act as integrators of
messengers. Transcription factors may be activated within gene transcription. These coactivator molecules include
the nucleus, often with the transcription factor already CREB-binding protein (CBP), and the related p300, thus
bound to DNA, or within the cytoplasm, resulting in expo- allowing complex interactions between dierent signaling
sure of nuclear localization signals and targeting to the pathways [14].

TABLE 31.1 Transcription factor families involved in the pathogenesis of


Histone acetylation
asthma and COPD.
DNA is wound around histone proteins to form nucleo-
GR NFATs somes and the chromatin ber in chromosomes [15].
It has long been recognized at a microscopic level that
NF-B GATA chromatin may become dense or opaque owing to the
AP-1 HIF-1
winding or unwinding of DNA around the histone core
[16]. CBP, p300, and other coactivators have histone acety-
CREB C/EBP lase activity (HAT) which is activated by the binding of
transcription factors, such as AP-1, NF-B, and STATs
STATs SP1 (Fig. 31.2) [14, 15]. Acetylation of histone residues results
GR: glucocorticoids receptor; NF-B: nuclear factor-kappa B; AP-1: activator protein 1;
in unwinding of DNA coiled around the histone core, thus
CREB: cyclic AMP response element binding protein; STATs: signal transducers and opening up the chromatin structure, allowing increased
activators of transcription; NFATs: nuclear factors of activated T cells; GATA: GATA transcription. Histone deacetylation by specic histone
binding proteins; HIF-1: hypoxia-inducible factor-1; C/EBP: CAAT/enhancer-binding deacetylases (HDACs) reverses this process, leading to gene
protein; SP1: specificity protein 1. repression [17].

374
Transcription Factors 31
Stimuli TNF- Cytokines Cytokines TNF- Steroid
IL-1

IB Kinase Ras
JAK
P
P CaN
MEKK-1
IB STAT
NF-ATc
P JNKK Steroid
P
NF-B receptor
Ub- JNK
Cytoplasm NF-ATc
proteasome

JUN P

AP-1 FOS

Nucleus
P

NF-B STAT NF-AT AP-1 GR


(A) (B) (C) (D) (E)

FIG. 31.1 Multiple pathways mediating transcription factor modulation of inflammatory genes, (A) Inflammatory mediator signal transduction activation.
The binding of cytokines, growth factors, or chemokines to their respective receptors sets in train the activation of a number of signal transduction
pathways, including the receptor tyrosine kinases, mitogen-activated protein kinases (MAPKs including MEKK1 and JNK), Janus kinases (JAKs), and other
kinase pathways involved in NF-B activation. Activation of nuclear factor-B involves phosphorylation of the inhibitory protein IB by specific kinase(s),
with subsequent ubiquitination and proteolytic degradation by the proteasome. The free NF-B then translocates to the nucleus, where it binds to B
sites in the promoter regions of inflammatory genes. Activation of the IB gene results in increased synthesis of IB to terminate the activation NF-B. (B)
JAK-STAT pathways. Cytokine binding to its receptor results in activation of JAK which phosphorylate intracellular domains of the receptor, resulting in
phosphorylation of signal transduction-activated transcription factors (STATs). Activated STATs dimerize and translocate to the nucleus where they bind
to recognition elements on certain genes. (C and D) Nuclear factor of activated T-cells (NF-AT) is activated via dephosphorylation by calcineurin (CaN)
and translocates to the nucleus where it interacts with AP-1 to induce gene transcription. (E) Classical mechanism of steroid action. Glucocorticoids are
lipophilic molecules which diffuse readily through cell membranes to interact with cytoplasmic receptors. Upon ligand binding receptors are activated and
translocate into the nucleus where they bind to specific DNA elements. The foregoing pathways can interact so that the final signal may be amplified or
altered depending upon the exact combination of stimuli. The final response to each stimulus or combination of stimuli by a particular cell depends upon
the receptors present in a particular cell along with the exact intracellular transduction pathway activated.

Cytokine stimulation
TRANSCRIPTION FACTORS IN ASTHMA
PCAF

Asthma is a complex chronic inammatory disease of the


m T
x

TF airways that involves the activation of many inamma-


ple
co HA

CBP SWI/SNF tory and structural cells, all of which release inamma-
RNA tory mediators that result in the typical clinicopathological
TAFs

polymerase II changes of asthma. The chronic airway inammation of


Nucleosomes asthma is unique in that the airway wall is inltrated by
TF TBP T-lymphocytes of the T-helper (Th) type 2 phenotype,
eosinophils, macrophages/monocytes, and mast cells. In
addition, an acute-on-chronic inammation may be
observed during exacerbations, with an increase in eosi-
nophils and sometimes also neutrophils [5].
FIG. 31.2 Histone acetylation by proinflammatory transcription factors.
In response to stress and other stimuli, such as cytokines, various second
messenger systems are upregulated, leading to activation of signal-
dependent transcription factors such as CREB, NF-B, AP-1, and STAT The role of transcription factors in
proteins. Binding of these factors leads to recruitment of CBP and/or other differentiation of Th1/Th2 cells
coactivators to signal-dependent promoters and acetylation of histones by
an intrinsic acetylase activity (HAT). Induction of histone acetylation allows
the formation of a more loosely packed nucleosome structure which enables
CD4 T-helper (Th) cells can be divided into three major
access to TATA box-binding protein (TBP) and associated factors (TAFs) and subsets, termed Th1, Th2, and Th0 based on the pattern of
the recruitment of further remodeling factors including switch/sucrose cytokines they produce [18]. Th1 cells produce predominantly
nonfermentable (SWI/SNF). Remodeling thereby allows RNA polymerase II IL-2 and interferon gamma (IFN-) and predominantly
recruitment and the activation of inflammatory gene transcription. promote cell-mediated immune responses. Th2 cells, which

375
Asthma and COPD: Basic Mechanisms and Clinical Management

IFN- Determination of asthma severity

STAT4 Activation of NF-B leads to the coordinated induction


JNK2 of multiple genes that are expressed in inammatory and
IL-12R P38 immune responses. Many of these genes are induced in
inammatory and structural cells and play an important
TCR NFATc2 role in the inammatory process.
Th0 NFATc3 Th1 cell
cell JNK1 NF-B
While NF-B is not the only transcription factor involved
c-Maf in regulation of the expression of these genes, it often
GATA3 appears to have a decisive regulatory role. NF-B often
IL-4R STAT6 functions in cooperation with other transcription factors,
NFATc1 Th2 cell such as AP-1 and C/EBP, which are also involved in regu-
lation of inammatory and immune genes [10, 26]. Genes
induced by NF-B include those for the proinammatory
IL-4, 5, 6, 9, 10, 13
cytokines IL-1, TNF-, and GM-CSF and the chemok-
FIG. 31.3 Differentiation of T-cell subtypes depends upon the action ines IL-8, macrophage inammatory protein-1 (MIP-1),
of a number of transcription factors. Th0 cells can differentiate into Th1 macrophage chemotactic protein-1 (MCP-1), RANTES,
and Th2 cells following IL-12 or IL-4 stimulation. Stimulation through and eotaxins, that are largely responsible for attracting
the IL-12 receptor (IL-12) activates the transcription factor STAT4 and the inammatory cells into sites of inammation [4, 7]. NF-
MAPK pathways JNK2 and p38 and drives Th1 cell differentiation, leading B also regulates the expression of inammatory enzymes,
to cells capable of releasing IFN-. In contrast, activation of the IL-4R in including the inducible form of nitric oxide synthase
concert with T-cell receptor (TCR) stimulation leads to activation of the (NOS2) that produces large amounts of nitric oxide (NO).
transcription factors STAT6, GATA3, c-Maf, and NFATc1, and drives Th2 NF-B also plays an important role in regulating expres-
cell differentiation. Th2 cells can release a number of cytokines including
sion of adhesion molecules, such as vascular cell adhesion
interleukins 4, 5, 6, 9, 10, and 13. In addition, activation of JNK1, NFATc2,
and NFATc3 can prevent Th2 cell differentiation.
molecule-1 (VCAM-1) and intercellular adhesion molecule-1
(ICAM-1), that are expressed on endothelial and epithelial
cells at inammatory sites and play a key role in the initial
recruitment of inammatory cells [4, 7].
Data also suggest that NF-B may be activated by
produce mainly interleukins (IL-4, IL-5, and IL-13), aug- many of the stimuli that exacerbate asthmatic inammation
ment certain B-cell responses. IL-4 in particular is the major (e.g. rhinovirus infection, allergen exposure, proinamma-
inducer of B-cell switching to immunoglobulin E (IgE) pro- tory cytokines, and oxidants) [2729]. There is also evidence
duction, and therefore plays a crucial role in allergic reactions for activation of NF-B in bronchial epithelial and sputum
involving IgE and mast cells. Th cells producing cytokines cells of patients with asthma [30]. NF-B is an ampli-
typical of both Th1 and Th2 clones have also been described fying and perpetuating mechanism that exaggerates the
and they have been named Th0 [1]. disease-specic inammatory process through the coordi-
Evidence indicates that Th1 and Th2 cells dierenti- nated activation of multiple inammatory genes. The degree
ate from a common precursor, naive T-cells. This appears to of NF-B phosphorylation and acetylation in asthma is
be a multistep process, in which naive T-cells pass through unknown as is the specic transcriptome associated with
an intermediate stage (Th0) at which both Th1 and Th2 each modied NF-B [10]. Inhibitors of the control-
cytokines are produced (Fig. 31.3). Transcription factors, ling kinase (Inhibitor of B kinase 2, IKK2) are in clinical
including GATA3, T-bet, c-maf, and STAT6, are involved development for asthma and other inammatory diseases
in the molecular mechanisms by which Th1 and Th2 with the hope of achieving as good an anti-inammatory
cells dierentially express the Th1 and Th2 cytokines eect as glucocorticoids without the side eects [31].
[19, 20] and that this may have diering consequences on
subepithelial brosis, airway smooth muscle hyperplasia AP-1
and mucus hyperplasia as a consequence of eects on AP-1 is a collection of related transcription factors belong-
TGF- and CCL11 expression [21]. However, most of ing to the Fos (c-Fos, FosB) and Jun (c-Jun, JunB, JunD)
these studies were conducted on murine T-cells, and the sit- families which dimerize in various combinations through
uation in human T-cells, in physiological and pathological their leucine zipper region. Fos/Jun form heterodimers
conditions (e.g. bronchial asthma) remain largely unknown. with high anity and are the predominant form of AP-1
A key role for regulatory T-cells (e.g. Treg and Th-17) has in most cells, whereas Jun/Jun homodimers bind with low
also been proposed in asthma, particularly in those patients anity and are less abundant [32]. AP-1 may be activated
with severe disease [22, 23]. Production of the Treg phe- by various cytokines, including TNF- and IL-1, via sev-
notype is associated with the transcription factor Foxp3 eral types of protein tyrosine kinase (PTK) and mitogen-
whose DNA binding to gene promoters has recently been activated protein (MAP) kinases in particular JUN-N-
mapped [24] and whose activity is also dependent upon terminal kinase ( JNK), which themselves activate a cascade
NF-AT [25]. of intracellular kinases [9]. AP-1, like NF-B, regulates

376
Transcription Factors 31
many of the inammatory and immune genes that are over-
expressed in asthma. Indeed many of these genes require the
simultaneous activation of both transcription factors that CBP ex
work together cooperatively. There is evidence for increased pl
 co
m
expression of c-Fos in bronchial epithelial cells in asthmatic AF
PC AC
airways [33] and an even greater expression in patients with HAT HD
severe, treatment-insensitive asthma [34, 35], and many of
the stimuli relevant to asthma that activate NF-B will also GR

activate AP-1. Inhibitors of JNK are being developed [32] p50 p65
and these show good eectiveness against bronchial hyper-
responsiveness, bronchoalveolar lavage (BAL) inammatory
cells, and airway remodeling in animal models of asthma
[36]. Interestingly, JNKs are also involved in T-cell class Nucleosomes
switching and their inhibitors may also have a profound
immunomodulatory role [32]. FIG. 31.4 The actions of the dexamethasone/GR complex on inhibition
of IL-1-stimulated histone acetylation. DNA-bound p65 induces histone
acetylation via activation of CBP and a CBP-associated HAT complex
STATs and other transcription factors to occur. This causes local unwinding of DNA and increased gene
Signal transduction-activated transcription factor-6 (STAT6) transcription. GR, acting as a monomer, interacts with CBP causing an
also provides a target as a potential treatment for allergic inhibition of CBP-mediated HAT activity. In addition, GR also recruits HDAC2
asthma. STAT6 knockout mice have no response to IL- to the p65/CBP complex, further reducing local HAT activity, leading to
4, do not develop Th2 cells in response to IL-4, and fail to enhanced nucleosome compaction and repression of transcription.
produce IgE, bronchial hyperresponsiveness, or BAL eosi-
nophilia after allergen sensitization indicating the critical act as important regulators of the inammatory reaction,
role of STAT6 in allergic responses [37]. STAT6 has been responding to various inammatory mediators, such as
reported to be overexpressed in bronchial biopsies from cytokines, by the production of a wide range of cytokines,
asthmatic patients [38]. In addition, evidence exists for chemokines, and other inammatory mediators [5].
enhanced expression of STAT1 in the airways of asthmatic, Glucocorticoids bind to and activate a cytosolic recep-
but not of COPD subjects [39]. Indeed, decoy oligonucle- tor (GR) which then translocates to the nucleus. Within the
otides against STAT1 are in early clinical development for nucleus two GR subunits form a dimer and bind to specic
asthma [40]. DNA elements (glucocorticoid response elements or GREs)
Cyclosporin has been used as an immunomodulator in the promoter regions of glucocorticoid-responsive genes,
for many years although its side eect prole limits its util- resulting in modulation of transcription [5]. Several genes
ity in asthma [1]. Its target is cyclophilin which controls the are upregulated by glucocorticoids, including the 2-receptor,
dephosphorylation of NF-AT (nuclear factor of activated MAPK phosphatase (MKP1), and serum leukoprotease
T-cells) and thereby the expression of key Th2 cytokines inhibitor (SLPI). However, in inammation the major role
including IL-13 [4, 7]. of glucocorticoids appears to be gene repression. Evidence
Sp1 is a ubiquitous transcription factor that binds accumulated over the past few years has suggested that a
to GC-boxes and related motifs, which are frequently major mechanism of glucocorticoids is repression of the
occurring DNA elements present in many promoters and actions of AP-1 and NF-B [5].
enhancers. Modication of these sites within the IL-10 and The major eect of glucocorticoids is on gene tran-
IL-4 and the 5-lipoxygenase promoters has been associated scription although actions on mRNA stability do occur with
with altered expression of these genes in distinct patient specic genes under the control of MKP-1 [43]. Several
groups [41, 42]. molecular mechanisms for the suppressive actions of the
activated GR on NF-B-mediated gene transcription have
been proposed, but it is likely that a combination of inhibi-
Transcription factors and glucocorticoid tion of histone H4 acetylation on lysines 8 and 12 induced
action by inammatory mediators, such as IL-1 and TNF-, lead
to changes in the ability of RNA polymerase II to tran-
Glucocorticoids belong to the family of nuclear steroid hor- scribe mRNA [5, 4446]. This eect on histone acetylation
mone receptors and are important anti-inammatory agents is achieved by a direct inhibition of CBP-associated histone
used in the treatment of chronic inammatory diseases such acetylation and by recruitment of HDAC2 to the activated
as asthma [5]. Functionally they act by suppressing airway DNA-bound NF-B complex [44] (Fig. 31.4). Of inter-
hyperresponsiveness, reducing airway edema and the inl- est is that oxidative stress that modulates HDAC activity is
tration of inammatory cells from the blood to the airway able to markedly alter the expression of some inammatory
and thereby reducing the airway inammatory response [5]. mediators and reduce glucocorticoid responsiveness [47].
Glucocorticoid receptors (GR) are predominantly local- We have recently reported that the expression of HDAC2
ized to the airway epithelium, alveolar macrophages and is reduced in the lung of patients with COPD [48] and that
endothelium [5], which are, therefore, probably important overexpression of HDAC2 restores glucocorticoid sensitivity
sites for the anti-inammatory action of steroids, especially in BAL macrophages from these patients [49]. The failure of
those delivered by the inhaled route. Airway epithelial cells glucocorticoids to regulate proopiomelanocortin (POMC)

377
Asthma and COPD: Basic Mechanisms and Clinical Management

expression in Cushings disease has also been related to a that multiple genes are operating and that the inuence of
lack of HDAC2 [46] and targeting of these enzymes may each gene in isolation may be relatively weak. The suscepti-
provide another novel target for future drug discovery. bility to develop COPD with smoking is likely to depend
Glucocorticoids are able to activate the expression of on the coincidence of several gene polymorphisms that act
a number of anti-inammatory genes due to GR binding to together [55].
specic GREs in the promoter regions of responsive genes The only clearly established, but rare, genetic risk fac-
[5]. This is often in conjunction with other transcription tor for COPD is 1-antitrypsin deciency (1-AT) [56].
factors and activation of CAAT/enhancer-binding protein Approximately 95% of cases of clinical 1-AT deciency
(C/EBP) by long-acting 2-agonists in airway smooth are caused by a single amino acid substitution at position
muscle cells has been proposed to account, at least in part, 342 (lysine for glutamic acid) in the coding region of the
for the enhanced eect of combination treatment in asth- 1-AT gene (Z allele). This genotype is uncommon, but a
matic patients [50]. Loss of C/EBP expression in some Taq1 polymorphism in the 3-anking region of the 1-
asthmatic patients may account for steroid eectiveness in AT gene has been reported to be present in 18% of COPD
these subjects [50]. patients but in only 5% of the general population in the
A very small number of asthmatic patients are United Kingdom [56]. Although this mutation is not asso-
steroid-resistant and fail to respond to even high doses of ciated with an abnormal 1-AT protein expression, this
oral glucocorticoids (so called steroid-resistant asthma). genetic variant is within an enhancer sequence (which con-
This defect is also present in their peripheral blood mono- tains C/EBP-binding sites) and may impair the acute-phase
nuclear cells (PBMCs) and T-lymphocytes [35]. Although increase in 1-AT gene expression in response to IL-6 [57].
ligand binding data show no change in GR number in these
patients, immunocytochemistry indicates a reduced GR
nuclear translocation which may account for the reduced Transcription factors and clinical
binding to DNA seen in these patients [51]. Altered manifestations of COPD
nuclear translocation is aected by GR phosphorylation
status [52], which may also be a target for long-acting The chronic airow obstruction in tobacco smoking-related
-agonists [5]. In the same patients there is a reduced COPD results from a combination of airway disease, which
inhibitory eect of glucocorticoids on AP-1 activation, particularly aects small airways, and loss of lung elasticity
but not on NF-B. Furthermore, there is an increase in the because of destruction of the lung parenchyma. The modu-
baseline activity of AP-1 which appears to be due to exces- lation of the inammatory response in COPD by genetic
sive activation of JNK [53, 54]. This resistance will be seen factors associated with transcription factors may, in part,
at the site of inammation where AP-1 is activated but not cause of the dierent clinical phenotypes of patients with
at uninamed sites. This may explain why patients with COPD [4]; however, it is clear that COPD patients have
steroid-resistant asthma are not resistant to the endocrine marked inammation, which increases with disease stage
and metabolic eects of glucocorticoids, and thus develop [58]. Patients with COPD are reported to have increased
the drugs systemic side eects. activation of NF-B in sputum and lung tissue macro-
phages and bronchial epithelial cells [5961]. The activation
status of NF-B and its repression by glucocorticoids may
also be aected by altered expression of HDAC2 [48, 49]
TRANSCRIPTION FACTORS IN COPD and of the protein deacetylase sirtuin (SIRT1) [62].
Hogg and colleagues [58] were able to show that the
mRNA expression of the transcription factors early growth
In contrast to the enormous increase in our understand- response (Egr)-1 and c-Fos were increased in bronchial
ing of the pathogenesis of asthma, less is known about the biopsies and that exposure of broblasts to cigarette smoke
molecular pathogenesis of COPD [55]. enhanced Egr-1 expression [63]. This results in subsequent
induction of MMP2 expression in vitro and in vivo [64].
In contrast to the tissue destruction seen in the alveoli in
Transcription factors and disease COPD patients, the small airways are characterized by
susceptibility brosis and increased smooth muscle. Activation of Smad
factors by growth factors, such as TGF, may be important
The etiology of COPD appears to be due to interactions in this process and Smad3 knockout mice develop emphy-
between environmental factors (particularly cigarette smok- sema [65]. In addition, expression of the inhibitory Smad7
ing) and genetic factors. Chronic heavy cigarette smoking is reduced in epithelial cells of COPD (Global Initiative
is currently the cause of more than 90% of cases of COPD for Chronic Obstructive Lung Disease, GOLD stage II)
in Westernized countries, so environmental factors are patients [66].
clearly very important. However, it is important to iden- In contrast to asthma, where C/EBP is abnormally
tify the factors that determine why only 1520% of chronic expressed, Borger and colleagues have reported a reduc-
heavy cigarette smokers develop symptomatic COPD [55]. tion of C/EBP in patients with COPD [67]. Activation of
So far, this is little understood, although it is likely that STAT4 is critical for the dierentiation of Th1/Tc1 cells T-
genetic factors are important. There is convincing evidence cells and the production of IFN- [68]. We have reported
that several genes inuence the development of COPD. increased expression of activated phospho-STAT4 in bron-
In a complex polygene disease such as COPD, it is likely chial biopsies and BAL lymphocytes of COPD patients and

378
Transcription Factors 31
that this correlated with the degree of airow obstruction 15. Kouzarides T. Chromatin modications and their function. Cell 128:
and the number of IFN- cells. In contrast, T-bet expres- 693705, 2007.
sion was not altered [68]. 16. Allfrey VG, Faulkner R, Mirsky AE. Acetylation and methylation of
histones and their possible role in the regulation of RNA synthesis.
Recently it has been suggested that in patients with
Proc Natl Acad Sci USA 51: 78694, 1964.
COPD and symptoms of chronic bronchitis there is an 17. de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB.
increased prevalence of a common polymorphism in the Histone deacetylases (HDACs): Characterization of the classical
5-anking region of the TNF- gene, which was previ- HDAC family. Biochem J 370: 73749, 2003.
ously found to be associated with high baseline and induced 18. Neurath MF, Finotto S, Glimcher LH. The role of Th1/Th2 polariza-
TNF- expression [69]. Unfortunately, recent results indi- tion in mucosal immunity. Nat Med 8: 56773, 2002.
cate that anti-TNF therapy is ineective in COPD [70]. 19. Zheng W, Flavell RA. The transcription factor GATA-3 is necessary
and sucient for Th2 cytokine gene expression in CD4 T cells. Cell
89: 58796, 1997.
20. Zhang S, Lukacs NW, Lawless VA, Kunkel SL, Kaplan MH. Cutting
edge: Dierential expression of chemokines in Th1 and Th2 cells is
CONCLUSION dependent on STAT6 but not STAT4. J Immunol 165: 1014, 2000.
21. Kiwamoto T, Ishii Y, Morishima Y et al. Transcription factors T-bet
and GATA-3 regulate development of airway remodeling. Am J Respir
In the future the role of transcription factors and the genetic Crit Care Med 174: 14251, 2006.
regulation of their expression in asthma and COPD may be 22. Xystrakis E, Kusumakar S, Boswell S et al. Reversing the defective
an increasingly important aspect of research, as this may be induction of IL-10-secreting regulatory T cells in glucocorticoid-
one of the critical mechanisms regulating the expression resistant asthma patients. J Clin Invest 116: 14655, 2006.
of clinical phenotypes and their responsiveness to therapy. 23. Bullens DM, Truyen E, Coteur L et al. IL-17 mRNA in sputum of
Despite recent advances in the knowledge of the patho- asthmatic patients: Linking T cell driven inammation and granulo-
cytic inux? Respir Res 7: 135, 2006.
genesis of asthma and COPD, much more research on the
24. Marson A, Kretschmer K, Frampton GM et al. Foxp3 occupancy and
molecular mechanisms of asthma and COPD are needed to regulation of key target genes during T-cell stimulation. Nature 445:
aid the logical development of new therapies for these com- 93135, 2007.
mon and important diseases, particularly in COPD where 25. Wu Y, Borde M, Heissmeyer V et al. FOXP3 controls regulatory T cell
no eective treatments currently exist. function through cooperation with NFAT. Cell 126: 37587, 2006.
26. Baldwin AS Jr. Series introduction: The transcription factor
NF-kappaB and human disease. J Clin Invest 107: 36, 2001.
References 27. Papi A, Johnston SL. Rhinovirus infection induces expression of its
own receptor intercellular adhesion molecule 1 (ICAM-1) via increased
1. Caramori G, Adcock I. Pharmacology of airway inammation in NF-kappaB-mediated transcription. J Biol Chem 274: 970720, 1999.
asthma and COPD. Pulm Pharmacol Ther 16: 24777, 2003. 28. Poynter ME, Irvin CG, Janssen-Heininger YM. Rapid activation of
2. Barnes PJ. Mechanisms in COPD: Dierences from asthma. Chest nuclear factor-kappaB in airway epithelium in a murine model of aller-
117: 10S14S, 2000. gic airway inammation. Am J Pathol 160: 132534, 2002.
3. Barnes PJ, Chung KF, Page CP. Inammatory mediators of asthma: An 29. Rahman I. Redox signaling in the lungs. Antioxid Redox Signal 7: 15,
update. Pharmacol Rev 50: 51596, 1998. 2005.
4. Barnes PJ. Transcription factors in airway diseases. Lab Invest 86: 867 30. Hart LA, Krishnan VL, Adcock IM, Barnes PJ, Chung KF. Activation
72, 2006. and localization of transcription factor, nuclear factor-kappaB, in
5. Ito K, Chung KF, Adcock IM. Update on glucocorticoid action and asthma. Am J Respir Crit Care Med 158: 158592, 1998.
resistance. J Allergy Clin Immunol 117: 52243, 2006. 31. Adcock IM, Chung KF, Caramori G, Ito K. Kinase inhibitors and air-
6. Caramori G, Lim S, Ito K et al. Expression of GATA family of tran- way inammation. Eur J Pharmacol 533: 11832, 2006.
scription factors in T-cells, monocytes and bronchial biopsies. Eur 32. Manning AM, Davis RJ. Targeting JNK for therapeutic benet: From
Respir J 18: 46673, 2001. junk to gold? Nat Rev Drug Discov 2: 55465, 2003.
7. Caramori G, Ito K, Adcock IM. Transcription factors in asthma and 33. Demoly P, Basset-Seguin N, Chanez P et al. c-fos proto-oncogene
COPD. IDrugs 7: 76470, 2004. expression in bronchial biopsies of asthmatics. Am J Respir Cell Mol
8. Gilmore TD. Introduction to NF-kappaB: Players, pathways, perspec- Biol 7: 12833, 1992.
tives. Oncogene 25: 668084, 2006. 34. Loke TK, Mallett KH, Rato J et al. Systemic glucocorticoid reduces
9. Saklatvala J. The p38 MAP kinase pathway as a therapeutic target in bronchial mucosal activation of activator protein 1 components in
inammatory disease. Curr Opin Pharmacol 4: 37277, 2004. glucocorticoid-sensitive but not glucocorticoid-resistant asthmatic
10. Perkins ND. Post-translational modications regulating the activity patients. J Allergy Clin Immunol 118: 36875, 2006.
and function of the nuclear factor kappaB pathway. Oncogene 25: 6717 35. Adcock IM, Lane SJ. Corticosteroid-insensitive asthma: Molecular
30, 2006. mechanisms. J Endocrinol 178: 34755, 2003.
11. Zhong H, May MJ, Jimi E, Ghosh S. The phosphorylation status of 36. Eynott PR, Nath P, Leung SY et al. Allergen-induced inammation
nuclear NF-kappaB determines its association with CBP/p300 or and airway epithelial and smooth muscle cell proliferation: Role of
HDAC-1. Mol Cell 9: 62536, 2002. JUN N-terminal kinase. Br J Pharmacol 140: 137380, 2003.
12. Chen L, Fischle W, Verdin E, Greene WC. Duration of nuclear NF- 37. Tomkinson A, Kanehiro A, Rabinovitch N et al. The failure of STAT6-
kappaB action regulated by reversible acetylation. Science 293: 165357, decient mice to develop airway eosinophilia and airway hyperrespon-
2001. siveness is overcome by interleukin-5. Am J Respir Crit Care Med 160:
13. Ogawa S, Lozach J, Benner C et al. Molecular determinants of cross- 128391, 1999.
talk between nuclear receptors and toll-like receptors. Cell 122: 70721, 38. Mullings RE, Wilson SJ, Puddicombe SM et al. Signal transducer
2005. and activator of transcription 6 (STAT-6) expression and function
14. Li B, Carey M, Workman JL. The Role of Chromatin during in asthmatic bronchial epithelium. J Allergy Clin Immunol 108:
Transcription. Cell 128: 70719, 2007. 83238, 2001.

379
Asthma and COPD: Basic Mechanisms and Clinical Management

39. Sampath D, Castro M, Look DC, Holtzman MJ. Constitutive acti- 55. Barnes PJ, Kleinert S. COPD a neglected disease. Lancet 364: 564
vation of an epithelial signal transducer and activator of transcription 65, 2004.
(STAT) pathway in asthma. J Clin Invest 103: 135361, 1999. 56. Kalsheker NA, Morgan K. Regulation of the alpha 1-antitrypsin gene
40. Quarcoo D, Weixler S, Groneberg D et al. Inhibition of signal trans- and a disease-associated mutation in a related enhancer sequence. Am J
ducer and activator of transcription 1 attenuates allergen-induced Respir Crit Care Med 150: S183S189, 1994.
airway inammation and hyperreactivity. J Allergy Clin Immunol 114: 57. Morgan K, Scobie G, Marsters P, Kalsheker NA. Mutation in an
28895, 2004. alpha1-antitrypsin enhancer results in an interleukin-6 decient acute-
41. Hobbs K, Negri J, Klinnert M, Rosenwasser LJ, Borish L. Interleukin- phase response due to loss of cooperativity between transcription fac-
10 and transforming growth factor-beta promoter polymorphisms in tors. Biochim Biophys Acta 1362: 6776, 1997.
allergies and asthma. Am J Respir Crit Care Med 158: 195862, 1958. 58. Hogg JC, Chu F, Utokaparch S et al. The nature of small-airway
42. Lim S, Crawley E, Woo P, Barnes PJ. Haplotype associated with low obstruction in chronic obstructive pulmonary disease. N Engl J Med
interleukin-10 production in patients with severe asthma. Lancet 352: 350: 264553, 2004.
113, 1998. 59. Caramori G, Romagnoli M, Casolari P et al. Nuclear localisation of
43. Abraham SM, Lawrence T, Kleiman A et al. Antiinammatory eects p65 in sputum macrophages but not in sputum neutrophils during
of dexamethasone are partly dependent on induction of dual specicity COPD exacerbations. Thorax 58: 34851, 2003.
phosphatase 1. J Exp Med 203: 188389, 2006. 60. Di Stefano A, Caramori G, Oates T et al. Increased expression of
44. Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of nuclear factor-kappaB in bronchial biopsies from smokers and patients
histone deacetylase 2 inhibits interleukin-1beta-induced histone H4 with COPD. Eur Respir J 20: 55663, 2002.
acetylation on lysines 8 and 12. Mol Cell Biol 20: 6891903, 2000. 61. Szulakowski P, Crowther AJ, Jimenez LA et al. The eect of smoking
45. Luecke HF, Yamamoto KR. The glucocorticoid receptor blocks P- on the transcriptional regulation of lung inammation in patients with
TEFb recruitment by NFkappaB to eect promoter-specic transcrip- chronic obstructive pulmonary disease. Am J Respir Crit Care Med 174:
tional repression. Genes Dev 19: 111627, 2005. 4150, 2006.
46. Bilodeau S, Vallette-Kasic S, Gauthier Y et al. Role of Brg1 and 62. Rajendrasozhan S, Yang SR, Kinnula VL, Rahman I. SIRT1, an anti-
HDAC2 in GR trans-repression of the pituitary POMC gene and inammatory and antiaging protein, is decreased in lungs of patients
misexpression in Cushing disease. Genes Dev 20: 287186, 2006. with chronic obstructive pulmonary disease. Am J Respir Crit Care Med
47. Ito K, Lim S, Caramori G et al. Cigarette smoking reduces histone 177: 861870, 2008.
deacetylase 2 expression, enhances cytokine expression, and inhibits 63. Ning W, Li CJ, Kaminski N et al. Comprehensive gene expression pro-
glucocorticoid actions in alveolar macrophages. FASEB J 15: 111012, les reveal pathways related to the pathogenesis of chronic obstructive
2001. pulmonary disease. Proc Natl Acad Sci USA 101: 14895900, 2004.
48. Ito K, Ito M, Elliott WM et al. Decreased histone deacetylase activity 64. Ning W, Dong Y, Sun J et al. Cigarette Smoke Stimulates Matrix
in chronic obstructive pulmonary disease. N Engl J Med 352: 196776, Metalloproteinase-2 Activity via EGR-1 in Human Lung Fibroblasts.
2005. Am J Respir Cell Mol Biol, 2006.
49. Ito K, Yamamura S, Essile-Quaye S et al. Histone deacetylase 2-medi- 65. Chen H, Sun J, Buckley S et al. Abnormal mouse lung alveolarization
ated deacetylation of the glucocorticoid receptor enables NF-kappaB caused by Smad3 deciency is a developmental antecedent of centri-
suppression. J Exp Med 203: 713, 2006. lobular emphysema. Am J Physiol Lung Cell Mol Physiol 288: L68391,
50. Roth M, Johnson PR, Borger P et al. Dysfunctional interaction of 2005.
C/EBPalpha and the glucocorticoid receptor in asthmatic bronchial 66. Zandvoort A, Postma DS, Jonker MR et al. Altered expression of the
smooth-muscle cells. N Engl J Med 351: 56074, 2004. Smad signalling pathway: Implications for COPD pathogenesis. Eur
51. Matthews JG, Ito K, Barnes PJ, Adcock IM. Defective glucocorticoid Respir J 28: 53341, 2006.
receptor nuclear translocation and altered histone acetylation patterns 67. Borger P, Matsumoto H, Boustany S et al. Disease-specic expression
in glucocorticoid-resistant patients. J Allergy Clin Immunol 113: 1100 and regulation of CCAAT/enhancer-binding proteins in asthma and
8, 2004. chronic obstructive pulmonary disease. J Allergy Clin Immunol 119:
52. Irusen E, Matthews JG, Takahashi A et al. p38 Mitogen-activated pro- 98105, 2007.
tein kinase-induced glucocorticoid receptor phosphorylation reduces 68. Di Stefano A, Caramori G, Capelli A et al. STAT4 activation in smok-
its activity: Role in steroid-insensitive asthma. J Allergy Clin Immunol ers and patients with chronic obstructive pulmonary disease. Eur Respir
109: 64957, 2002. J 24: 7885, 2004.
53. Lane SJ, Adcock IM, Richards D et al. Corticosteroid-resistant bron- 69. Huang SL, Su CH, Chang SC. Tumor necrosis factor-alpha gene
chial asthma is associated with increased c-fos expression in monocytes polymorphism in chronic bronchitis. Am J Respir Crit Care Med 156:
and T lymphocytes. J Clin Invest 102: 215664, 1998. 143639, 1997.
54. Sousa AR, Lane SJ, Soh C, Lee TH. In vivo resistance to corticosteroids 70. Rennard SI, Fogarty C, Kelsen S et al. The Safety and Ecacy of
in bronchial asthma is associated with enhanced phosyphorylation of JUN Iniximab in Moderate-To-Severe Chronic Obstructive Pulmonary
N-terminal kinase and failure of prednisolone to inhibit JUN N-terminal Disease. Am J Respir Crit Care Med 156: 92634, 2007.
kinase phosphorylation. J Allergy Clin Immunol 104: 56574, 1999.

380
Neural and Humoral Control of the Airways
32CHAPTER

receptors are involved. Three types of airway


INTRODUCTION nerve are recognized: Peter J. Barnes1 and
Neil C. Thomson2
1. parasympathetic nerves which primarily
Airway nerves and circulating hormones regu- release acetylcholine (ACh) 1
National Heart and Lung Institute
late the caliber of the airways and have an inu-
2. sympathetic nerves which primarily release (NHLI), Clinical Studies Unit,
ence on airway smooth muscle tone, airway
norepinephrine (noradrenaline) Imperial College, London, UK
blood ow, and mucus secretion. They may also 2
inuence the inammatory process and play an Department of Respiratory Medicine,
3. aerent (sensory nerves) whose primary
integral role in host defense. There is increasing Division of Immunology, Infection and
transmitter is glutamate.
evidence that neural and humoral mechanisms Inflammation, University of Glasgow,
may play a role in the pathophysiology of asthma In addition to these classical transmitters, Glasgow, UK
and COPD, and several of the treatments used multiple neuropeptides have now been localized
interact with neural or humoral control. to airway nerves and may have potent eects on
airway function [1]. All of these neurotransmit-
ters act on autonomic receptors that are expressed
on the surface of target cells in the airway. It is
increasingly recognized that a single transmitter
NEURAL CONTROL may act on several subtypes of receptor, which
may lead to dierent cellular eects mediated via
dierent second messenger systems.
Overview of airway innervation Several neural mechanisms are involved
in the regulation of airway caliber and abnor-
Neural control of airway function is complex malities in neural control may contribute to
and many neurotransmitters are autonomic airway narrowing in disease (Fig. 32.1). Neural

Cholinergic ACh Muscarinic B/C


receptors

NE -receptors B/C? FIG. 32.1 Autonomic control of airway


Adrenergic smooth muscle tone. Neural mechanisms
E -receptors B/D resulting in bronchoconstriction (B/C) and
bronchodilatation (B/D). Ach: acetylcholine;
NE: norepinephrine; E: epinephrine; VIP:
VIP VIP-receptor vasoactive intestinal peptide; NO: nitric
B/D
NO Guanylyl cyclase oxide; i-NANC: inhibitory non-adrenergic
NANC non-cholinergic nerves; e-NANC: excitatory
SP/NKA NK-receptor B/C non-adrenergic non-cholinergic nerves; NK:
neurokinin.

381
Asthma and COPD: Basic Mechanisms and Clinical Management

mechanisms are involved in the pathophysiology of asthma either facilitatory eects or antagonistic eects on target
and COPD, contributing to the symptoms, and possibly to cells, or may inuence the release of the primary transmitter
the inammatory response [2]. There is a close interrela- via pre-junctional receptors. Thus VIP modulates the release
tionship between inammation and neural responses in the of ACh from airway cholinergic nerves. Sympathetic nerves,
airways, since inammatory mediators may inuence the which release noradrenaline, may also release neuropep-
release of neurotransmitters via activation of sensory nerves tide Y (NPY) and enkephalins, whereas aerent nerves
leading to reex eects and via stimulation of pre-junctional may contain a variety of peptides including substance P
receptors that inuence the release of neurotransmitters [3]. (SP), neurokinin A (NKA), calcitonin gene-related peptide
In turn, neural mechanisms may inuence the nature of the (CGRP), galanin, VIP, and cholecystokinin.
inammatory response, either reducing inammation or The physiological role of neurotransmission may be
exaggerating the inammatory response. Neuroimmmune in ne tuning of neural control. Neuropeptides may
interactions in asthma are increasingly recognized, particu- be preferentially released by high frequency ring of nerves,
larly the role of neurotrophins released from inammatory and their eects may therefore only become manifest under
and immune cells in the airways [4]. condition of excessive nerve stimulation. Neuropeptide
neurotransmitters may also act on target cells dierent from
the primary transmitter, resulting in dierent physiological
Neural interactions eects. Thus in airways ACh causes bronchoconstriction,
but VIP which is co-released may have its major eect on
Complex interactions between various components of the bronchial vessels, thus increasing blood ow to the airways.
autonomic nervous system are now recognized. Adrenergic In chronic inammation the role of co-transmitters may be
nerves may modulate cholinergic neurotransmission in the increased by alterations in the expression of their receptors
airways and sensory nerves may inuence neurotransmission or by increased synthesis of transmitters via increased gene
in parasympathetic ganglia and at post-ganglionic nerves. transcription.
This means that changes in the function of one neural path-
way may have eects on other pathways.

SENSORY NERVES
Co-transmission
The sensory innervation of the respiratory tract is mainly car-
Almost every nerve contains multiple transmitters. Thus
ried in the vagus nerve. The neuronal cell bodies are localized
airway parasympathetic nerves, in which the primary trans-
to the nodose and jugular ganglia and input to the solitary
mitter is ACh, also contain the neuropeptides vasoactive
tract nucleus in the brain stem. A few sensory bers sup-
intestinal polypeptide (VIP), peptide histidine isoleucine/
plying the lower airways enter the spinal cord in the upper
methionine (PHI/M), pituitary adenylyl cyclase activating
thoracic sympathetic trunks, but their contribution to respi-
peptide (PACAP), helodermin, galanin, and neuronal nitric
ratory reexes is minor and it is uncertain whether they are
oxide synthase (Fig. 32.2). These co-transmitters may have
represented in humans. There is a tonic discharge of sensory
nerves that has a regulatory eect on respiratory function
and also triggers powerful protective reex mechanisms in
response to inhaled noxious agents, physical stimuli, or cer-
VIP, PHM, PHV tain inammatory mediators.
PACAP-27 At least three types of aerent ber have been identi-
Parasympathetic Helospectin I, II
Acetylcholine Helodermin ed in the lower airways [5] (Fig. 32.3). Most of the infor-
Galanin mation on their function has been obtained from studies in
(SP, CGRP) anesthetized animals, so it is dicult to know how much
of the information obtained in anesthetized animals can be
Sympathetic NPY
Noradrenaline (Enkephalin) extrapolated to human airways.

SP, NKA, NPK


CGRP Slowly adapting receptors
GRP
Afferent Secretoneurin
Galanin? Myelinated bers associated with smooth muscle of proxi-
Glutamate
Somatostatin? mal airways are probably slowly adapting (pulmonary
CCK-8? stretch) receptors (SARs) that are involved in reex con-
Nociceptin, trol of breathing. Activation of SARs reduces eerent vagal
Endomorphins?
discharge and mediates bronchodilatation. During tracheal
FIG. 32.2 Neurotransmitters and co-transmitters in airway nerves. SP:
constriction the activity of SARs may serve to limit the
substance P, NKA: neurokinin A, CGRP: calcitonin gene-related peptide, VIP: bronchoconstrictor response. SARs may play a role in
vasoactive intestinal peptide, PHI/PHM/PHV: peptide histidine isoleucine/ the cough reex since when these receptors are destroyed by
methionine/valine, PACAP: pituitary adenylate cyclase activating peptide, high concentration of SO2 the cough response to mechani-
NPY: neuropeptide Y. cal stimulation is lost.

382
Neural and Humoral Control of the Airways 32
Mechanical stimuli Capsaicin, bradykinin, H
(bronchocontriction) cigarette smoke, SO2 water,
water, low Cl hyperosmolar solutions

Airway epithelium

C-fiber
RAR NP release
A-fiber SP, NKA, Cough
CGRP Bronchial vessel cholinergic
reflex
Airway smooth muscle FIG. 32.3 Afferent nerves in airways. Slowly
adapting receptors (SARs) are found in airway
smooth muscle, whereas rapidly adapting
myelinated (RAR) and unmyelinated C-fibers
SAR are present in the airway mucosa.

Rapidly adapting receptors serotonin, and prostaglandins. They are selectively stimulated
by capsaicin given either by intravenously or by inhalation and
Myelinated bers in the epithelium, particularly at the branch- are also stimulated by SO2 and cigarette smoke. Since these
ing points of proximal airways, show rapid adaptation. Rapidly bers are relatively unaected by lung mechanics, it is likely
adapting receptors (RARs) account for 1030% of the myeli- that these agents act directly on the unmyelinated endings in
nated nerve endings in the airways. These endings are sensitive the airway epithelium. In the in vitro guinea pig trachea prep-
to mechanical stimulation and to mediators such as histamine. aration C-bers are stimulated by capsaicin and by brady-
The response of RAR to histamine is partly due to mechani- kinin, but not by histamine, serotonin, or prostaglandins (with
cal distortion consequent on bronchoconstriction, although if the possible exception of prostacyclin) [6].
this is prevented by pre-treatment with isoprenaline the RAR RARs and C-bers are sensitive to water and hyper-
response is not abolished, indicating a direct stimulatory eect osmotic solutions, with RARs showing a greater sensitiv-
of histamine. It is likely that mechanical distortion of the air- ity to hypotonic and C-bers to hypertonic saline. In vitro
way may amplify irritant receptor discharge. guinea pig trachea preparation A bers and C-bers are
RAR with widespread arborizations are very numer- stimulated by water and by hyperosmolar solutions; a small
ous in the area of the carina, where they have been termed proportion of A bers are also stimulated by low-chloride
cough receptors as cough can be evoked by even the slight- solutions, whereas the majority of C-bers are [8]. C-bers
est touch in this region. RAR respond to inhaled cigarette are activated by capsaicin, which acts through vanilloid or
smoke, ozone, serotonin, and prostaglandin F2, although TRPV1 receptors, for which small molecule inhibitors are
it is possible that these responses are secondary to the now in development [9].
mechanical distortion produced by the bronchoconstrictor
response to these irritants. Neurophysiological studies using
an in vitro preparation in guinea pig trachea and bronchi Cough
show that a majority of aerent bers are myelinated and
belong to the A-ber group. Although these bers are acti- Cough is an important defense reex, which may be trig-
vated by mechanical stimulation and low pH, they are not gered from either laryngeal or lower airway aerents and is
sensitive to capsaicin, histamine, or bradykinin [6, 7]. an important symptom of asthma and COPD [10]. There is
debate about which are the most important aerents for ini-
tiation of cough and this may be dependent on the stimulus.
C-Fibers Thus RARs are activated by mechanical stimuli (e.g. particu-
late matter), bronchoconstrictors and hypotonic saline and
There is a high density of unmyelinated (C-bers) in the water, whereas C-bers are more sensitive to hypertonic solu-
airways and they greatly outnumber myelinated bers. In tions, bradykinin, and capsaicin. In normal humans inhaled
the bronchi C-bers account for 8090% of all aerent b- capsaicin is a potent tussive stimulus and this is associated
ers in cats. C-bers play an important role in the defense of with a transient bronchoconstrictor reex that is abolished
the lower respiratory tract. C-bers contain neuropeptides, by an anticholinergic drug. It is not certain whether this is
including SP, NKA, and CGRP, that confers a motor func- due to stimulation of C-bers in the larynx, but as these
tion on these nerves. Bronchial C-bers are insensitive to lung are very sparse it is likely that bronchial C-bers are also
ination and deation but typically respond to chemical stim- involved. Citric acid is commonly used to stimulate cough-
ulation. In vivo studies suggest that bronchial C-bers in dogs ing in experimental challenges in human subjects; it is likely
respond to the inammatory mediators histamine, bradykinin, that it produces cough by a combination of low pH (which

383
Asthma and COPD: Basic Mechanisms and Clinical Management

stimulates C-bers) and low chloride (which may stimulate and COPD. PGE2 is a potent sensitizer of airway sensory
laryngeal and lower airway aerents). Inhaled bradykinin nerves and is increased in asthma and COPD. Bradykinin is
causes coughing and a raw sensation retrosternally which also a potent aerent nerve sensitizer [7]. Chronic inam-
may be due to stimulation of C-bers in the lower airways. mation may lead to neural hyperesthesia through mecha-
Bradykinin appears to be a relatively pure stimulant of nisms that may involve cytokines and neurotrophins [5].
C-bers [6]. Prostaglandins E2 and F2 are potent tussive Neurotrophins, such as nerve growth factor (NGF), may
agents in humans and also sensitize the cough reex [11, 12]. result in proliferation of airway sensory nerves and a change
in the nerve phenotype, with a reduced threshold of activa-
tion and increased expression of neuropeptides [14].
Afferent nerves in airway disease
Airway aerent nerves may become sensitized in inam- Neurotrophins
matory airway diseases, resulting in increased symptoms,
such as cough and chest tightness (Fig. 32.4). Cough is a The role of neurotrophins in airway diseases is increasingly
prominent symptom of asthma and COPD, and there is recognized (Fig. 32.5) [15]. NGF, brain-derived neuro-
evidence that cough sensitivity is increased [13]. This may trophic factor, and neurotrophins 3 and 4/5 are nerve-related
be due to senitization of aerent nerves in the airways as a cytokines that play an important role in the function, prolif-
result of inammatory mediators produced during asthma eration, and survival of autonomic nerves. In sensory nerves

Inflammation

Prostaglandins Cytokines Neurotrophins Bradykinin


PGE2, PGI2 IL-1, TNF- NGF, BDNF B1, B2 receptors H

Activation
Sensitization

Symptoms
C-fiber
Cough
A-fiber Neurogenic
Chest tightness
inflammation
AHR

SP, CGRP

FIG. 32.4 Airway hyperaesthesia. Increased sensitivity of airway nerves induced by inflammation as a result of sensitization and activation of airway
sensory nerves, resulting in increased symptoms and possibly neurogenic inflammation through the release of neuropeptides, such as substance P (SP) and
calcitonin gene-related peptide (CGRP). AHR: airway hyperresponsiveness.

Mast cell
Allergen trkA
Growth
Epithelial
cells

NGF

Cough
Eosinophil
survival trkA receptors

FIG. 32.5 Neurotrophin effects in airways. NGF is


Proliferation released from mast cell and epithelial cells and has
Sensitization Airway effects of airway afferent nerves, which may lead to
Tachykinins hyperresponsiveness cough and airway hyperresponsiveness. NGF also
promotes the growth of mast cells through its receptor
Sensory nerves TrkA and is chemotactic for eosinophils.

384
Neural and Humoral Control of the Airways 32
neurotrophins increase responsiveness and may also lead to Cholinergic control of airways
expression of tachykinins. Neurotrophins may be produced
by inammatory cells, such as mast cells, lymphocytes, mac- Cholinergic nerve bers arise in the nucleus ambiguous in
rophages, and eosinophils, as well as structural cells, such as the brain stem and travel down the vagus nerve and synapse
epithelial cells, broblasts, and airway smooth muscle cells in parasympathetic ganglia which are located within the
[16]. Although neurotrophins have predominant eects on airway wall. From these ganglia short post-ganglionic b-
neuronal cells they may also act as growth factors for inam- ers travel to airway smooth muscle and submucosal glands
matory cells such as mast cells, as well as increasing chemo- (Fig. 32.6). In animals, electrical stimulation of the vagus
taxis and survival of eosinophils [4]. NGF induces airway nerve causes release of ACh from cholinergic nerve termi-
hyperresponsiveness in various animal models of asthma, nals, with activation of muscarinic cholinergic receptors on
including guinea pig, mice, and rats and this is blocked by smooth muscle and gland cells, which results in bronchoc-
antibodies to NGF or its receptor TrkA [15]. In guinea pig onstriction and mucus secretion. Prior administration of a
NGF enhances airway hyperresponsiveness by increasing the muscarinic receptor antagonist, such as atropine, prevents
release of SP from sensory nerves [17] and increases sensory vagally induced bronchoconstriction.
nerve activation [4]. The role of neurotrophins in human
asthma is uncertain. However, NGF levels are increased in
bronchoalveolar lavage uid of asthmatic patients and there
is a further increase after allergen challenge [18]. Blocking Non-neuronal cholinergic system
NGF or its receptors may therefore have some therapeutic
potential. A decoy TrkA receptor which blocks NGF in vivo There is increasing evidence that ACh is also synthesized
reduces airway hyperresponsiveness in animal models [19]. and released from several non-neuronal cells, including air-
way epithelial cells and broblasts [20, 21]. Inammatory
signals may increase the synthesis of ACh and thus enhance
cholinergic eects. The contribution of non-neuronal
CHOLINERGIC NERVES ACh to cholinergic responses in the airways is currently
unknown but may play an important role in regulating the
tone of small airways where cholinergic innervation is very
Cholinergic nerves are the major neural bronchoconstrictor sparse. Non-neuronal ACh may have eects on inamma-
mechanism in human airway and are the major determinant tory cells through nicotinic and muscarinic receptors that
of airway caliber in humans. are expressed on their surface [22].

CNS

Nodose
ganglion

Vagus nerve
Laryngeal
Oesophageal
afferents Parasympathetic nerve

ACh
C-fiber
A-fiber Parasympathetic ganglion
Inflammatory
cell
C-fiber
ACh Submucosal
receptors
ACh gland

Irritant
receptors Mediators

Airway
epithelium

Irritants (e.g. cigarette smoke)

FIG. 32.6 Cholinergic control of airway smooth muscle. Pre-ganglionic and post-ganglionic parasympathetic nerves release acetylcholine (ACh) and can be
activated by airway and extra-pulmonary afferent nerves.

385
Asthma and COPD: Basic Mechanisms and Clinical Management

Muscarinic receptors also explain the sometimes catastrophic bronchoconstric-


tion which occurs with -blockers in asthma which, at least
Of the ve known subtypes of muscarinic receptor, four in mild asthmatics, appears to be mediated by cholinergic
have been identied by binding studies and pharmacologi- pathways [27]. Antagonism of inhibitory -receptors on
cally in lung [23]. The muscarinic receptors that mediate cholinergic nerves would result in increased release of ACh
bronchoconstriction in human and animal airways belong to which could not be switched o in the asthmatic patient
the M3-receptor subtype, whereas mucus secretion appears (Fig. 32.8). This explains why anticholinergics prevent -
to be mediated by M1- and M3-receptors. M1-receptors are blocker-induced asthma. The mechanisms which lead to
also localized to parasympathetic ganglia, where they facili- dysfunction of pre-junctional M2-receptors in asthmatic air-
tate the neurotransmission mediated via nicotinic receptors ways are not certain, but it is possible that M2-receptors may
(Fig. 32.7). be more susceptible to damage by oxidants or other products
Inhibitory muscarinic receptors (autoreceptors) have of the inammatory response in the airways. Experimental
been demonstrated on cholinergic nerves of airways in studies have demonstrated that inuenza virus infection and
animals in vivo, and in human bronchi in vitro [24]. These eosinophils in guinea pigs may result in a selective loss of
pre-junctional receptors inhibit ACh release and may serve M2-receptors compared with M3-receptors, resulting in
to limit vagal bronchoconstriction. Autoreceptors in human a loss of autoreceptor function and enhanced cholinergic
airways belong to the M2-receptor subtype, whereas those bronchoconstriction.
on airway smooth muscle and glands belong to the M3- Cholinergic innervation is greatest in large airways
receptor subtype [25]. Drugs such as atropine and ipra- and diminishes peripherally, although in humans muscarinic
tropium bromide, which block both pre-junctional receptors are localized to airway smooth muscle in all air-
M2-receptors and post-junctional M3-receptors on smooth ways [28]. In humans, studies which have tried to distinguish
muscle with equal ecacy, therefore increase in ACh large and small airway eects have shown that cholinergic
release may then overcome the post-junctional blockade. bronchoconstriction predominantly involves larger airways,
This means that such drugs will not be as eective against whereas -agonists are equally eective in large and small air-
vagal bronchoconstriction as against cholinergic agonists, ways. This relative diminution of cholinergic control in small
and it may be necessary to re-evaluate the contribution of airways may have important clinical implications, since anti-
cholinergic nerves when drugs which are selective for the cholinergic drugs are likely to be less useful than -agonists
M3-receptors are developed for clinical use. The presence of when bronchoconstriction involves small airways. Normal
muscarinic autoreceptors has been demonstrated in human human subjects also have resting bronchomotor tone, since
subjects in vivo [26]. A cholinergic agonist, pilocarpine, atropine causes bronchodilatation.
which selectively activates M2-receptors, inhibits cholin-
ergic reex bronchoconstriction induced by sulfur dioxide
in normal subjects, but such an inhibitory mechanism does Cholinergic reflexes
not appear to operate in asthmatic subjects, suggesting that
there may be dysfunction of these autoreceptors. Such a A wide variety of stimuli are able to elicit reex cholinergic
defect in muscarinic autoreceptors may then result in exag- bronchoconstriction through activation of sensory recep-
gerated cholinergic reexes in asthma, since the normal tors in the larynx or lower airways. Activation of cholinergic
feedback inhibition of ACh release may be lost. This might reexes may result in bronchoconstriction and an increase in

Pre-ganglionic nerve

Normal COPD

Vagus nerve
Parasympathetic ACh N (+)
M1 (+) ACh Vagal tone ACh
ganglion
Narrowed airway

M2 ()
Post-ganglionic nerve
ACh
ACh ACh
Anticholinergic Resistance 1/r 4
Airway smooth M3 () 2-Agonist
muscle

FIG. 32.7 Muscarinic receptor subtypes in the airways. Acetylcholine


(ACh) from pre-ganglionic vagal nerves activates nicotinic receptors (N) FIG. 32.8 Cholinergic control of airways in COPD. The normal vagal
and may be facilitated by M1-receptors. ACh release from pre-ganglionic cholinergic tone has a greater effect in COPD airways because of the
nerves activate M3-receptors on airway smooth muscle and feeds back to geometric relationship between airway diameter and resistance, so that
activate pre-junctional M2 receptors (autoreceptors) which inhibit further anticholinergics are effective bronchodilators in COPD. 2-Agonists are also
ACh release. effective and act as functional antagonists of cholinergic tone.

386
Neural and Humoral Control of the Airways 32
airway mucus secretion through the activation of muscarinic narrowed airways. This may explain why anticholinergics are
receptors on airway smooth muscle cells and submucosal often of greater use in chronic asthmatics with a major ele-
glands. Cholinergic reexes may also be activated from ment of xed airway obstruction.
extrapulmonary aerents and these reexes may also con-
tribute to airway defenses. Esophageal reux may be associ- Role in COPD
ated with bronchoconstriction in asthmatic patients. In some The structural narrowing of the airways in COPD means
patients this may be due to aspiration of acid into the air- that even normal vagal tone will exert a greater eect on
ways, in other cases acid reux into the esophagus activates a airway caliber than in normal airways (Fig. 32.8). This may
reex cholinergic bronchoconstriction (the reux reex). account for the ecacy of anticholinergics as bronchodila-
tors in COPD, as cholinergic tone is the only reversible
Modulation of cholinergic neurotransmission element and have similar or even greater eects than 2-
Many agonists may modulate cholinergic neurotransmission agonists. In addition, cholinergic mechanisms may account
via pre-junctional receptors on post-ganglionic nerves [29]. for the mucus hypersecretion of chronic bronchitis.
Some receptors increase (facilitate), whereas others inhibit
the release of ACh. Inammatory mediators may inuence
cholinergic neurotransmission via pre-junctional receptors.
For example, thromboxane and prostaglandin (PG)D2 facili- ADRENERGIC CONTROL
tate ACh release from post-ganglionic nerves in the airways.
Facilitation may also occur at parasympathetic ganglia in the
airways; these structures are surrounded by inammatory The airways are also under adrenergic control, which includes
cells and have an aerent neural input. Electrophysiological sympathetic nerves (which release noradrenaline), circulating
recordings show a prolonged potentiation of neurotransmis- catecholamines (predominantly adrenaline) and - and -
sion in ganglia after allergen exposure in sensitized guinea adrenoceptors (Fig. 32.9). The fact that -adrenergic antago-
pigs [30]. nists cause bronchoconstriction in asthmatic patients, but
not in normal individuals, suggests that adrenergic control of
Role in asthma airway smooth muscle may be abnormal in asthma.
Many of the stimuli which produce bronchospasm in asthma
activate sensory nerves and reex cholinergic bronchocon- Sympathetic innervation
striction in animals and there is some evidence that cholin-
ergic tone is increased in asthmatic airways. There are several Although sympathetic bronchodilator nerves have been dem-
mechanisms by which cholinergic tone might be increased onstrated in several species, including cats, dogs, and guinea
in asthma: pigs, most evidence suggests that adrenergic nerves do not con-
trol human airway smooth muscle directly. However, sympa-
Increased aerent stimulation by inammatory thetic nerves may inuence cholinergic tone of airway smooth
mediators, such as histamine or prostaglandins. muscle via adrenoceptors localized to parasympathetic ganglia
Increased release of ACh from cholinergic nerve and pre-junctionally on post-ganglionic nerves [29], and sym-
terminals by an action on cholinergic nerve endings pathetic nerves may play an important role in the regulation of
themselves, or by an increase in nerve trac through airway blood ow and in mucus secretion.
cholinergic ganglia (local airway reex).
Abnormal muscarinic receptor expression, either via -Adrenoceptors
an increase in M3-receptors or via reduction in M2-
receptors. There is no evidence for increased M1- or M3- -Adrenoceptors regulate many aspects of airway function,
receptor expression in asthmatic lungs [31], but there is including airway smooth muscle tone, mast cell mediator
functional evidence for a defect in M2-receptor function release, and plasma exudation. The possibility that -receptors
that may be secondary to the inammatory process. are abnormal in asthma has been extensively investigated. The
Decrease in the neuromodulators (VIP, NO) that have a suggestion that there is a primary defect in -receptor func-
braking eect on neurotransmission. tion in asthma has not been substantiated and any defect in
-receptors is likely to be secondary to the disease, perhaps
The eect of ACh on asthmatic airways is exagger- as a result of inammation or as a consequence of adrenergic
ated, as a manifestation of non-specic hyperresponsiveness therapy. Some studies have demonstrated that airways from
of the airways. Anticholinergic agents will only counteract asthmatic patients fail to relax normally to isoproterenol,
the cholinergic reex component of bronchoconstriction, suggesting a possible defect in -receptor function in airway
which are less prominent in human airways than animal smooth muscle [32]. Whether this is due to a reduction in -
studies had indicated. By contrast 2-agonists reverse bron- receptors, a defect in receptor coupling, or some abnormality
choconstriction irrespective of the mechanism, since they in the biochemical pathways leading to relaxation, is not yet
act as functional antagonists and always have a much greater known, although the density of -receptors in airway smooth
bronchodilator eect than anticholinergic drugs. Vagal tone muscle appears to be normal [33] and there is no reduction
increases the airway narrowing further, and for geomet- in the density of 1- or 2-receptors in asthmatic lung, either
ric reasons will have a greater eect on airway resistance in at the receptor or at the mRNA level [31].

387
Asthma and COPD: Basic Mechanisms and Clinical Management

CNS M3-receptors
2-receptors
-receptors

Sympathetic Vagus nerve


ganglion

Sympathetic Parasympathetic nerve


nerve

Adrenal NE NE
medulla E
Parasympathetic ganglion

NE ACh
Bronchial
vessel
Airway smooth muscle

FIG. 32.9 Adrenergic control of airway smooth muscle. Sympathetic nerves release norepinephrine (NE), which may modulate cholinergic nerves at the
level of the parasympathetic ganglion or post-ganglionic nerves, rather than directly at smooth muscle in human airways. Circulating epinephrine (E) is
more likely to be important in adrenergic control of airway smooth muscle.

There is some evidence that proinammatory cytokines neuropeptides, nitric oxide (NO), and adenosine triphos-
may aect 2-receptor function. IL-1 reduces the bron- phate (ATP). In the airways both inhibitory NANC (bron-
chodilator eect of isoprenaline in vitro and in vivo and chodilator) and excitatory NANC (bronchoconstrictor)
this appears to be due to uncoupling of 2-receptors due nerves have been described.
to increased expression of the inhibitory G protein, Gi
[34, 35]. However, studies of 2-receptor expression in asth-
matic airways obtained by biopsy have demonstrated only i-NANC nerves
small defects in coupling after local allergen challenge [36].
i-NANC nerves which mediate bronchodilatation have
been described in may species, including humans, in whom
-Adrenoceptors they are of particular importance in the absence of any
direct sympathetic innervation of airway smooth muscle
-Receptors which mediate bronchoconstriction have been [38]. The neurotransmitter for these nerves in some species,
demonstrated in airways of several species, and may only be including guinea pigs and cats, is VIP and related peptides.
demonstrated under certain experimental conditions. There The i-NANC bronchodilator response is blocked by -
is now considerable doubt about the role of -receptors in chymotrypsin, an enzyme, which very eciently degrades
the regulation of tone in human airways, however, since it VIP and by antibodies to VIP. However, although VIP is
has proved dicult to demonstrate their presence function- present in human airways and VIP is a potent bronchodi-
ally or by autoradiography [37]. -Adrenoceptor antago- lator of human airways in vitro, there is no evidence that
nists are not eective as bronchodilators, but it is possible VIP is involved in neurotransmission of i-NANC responses
that -receptors may play an important role in regulating in human airways, and -chymotrypsin that completely
airway blood ow, which may indirectly inuence airway blocks the response to exogenous VIP has no eect on neu-
responsiveness. ral bronchodilator responses [39]. It is likely that VIP and
related peptides may be more important in neural vasodila-
tation responses and may result on increased blood ow to
bronchoconstricted airways.
The predominant neurotransmitter of human air-
NANC NERVES AND NEUROPEPTIDES ways is NO. NO synthase inhibitors, such as NG-l-arginine
methyl ester, virtually abolish the i-NANC response [40].
This eect is more marked in proximal airways, consistent
Neural responses that are not blocked by a combination of with the demonstration that nitrergic innervation is great-
adrenergic and cholinergic antagonists are known as non- est in proximal airways. NO appears to be a co-transmitter
adrenergic non-cholinergic (NANC) nerves. These NANC with ACh, and NO acts as a braking mechanism for the
responses appear to be due to the release of neurotrans- cholinergic system by acting as a functional antagonist to
mitters from classical autonomic nerves, which include ACh at airway smooth muscle [41] (Fig. 32.10).

388
Neural and Humoral Control of the Airways 32
Normal TABLE 32.1 Neuropeptides in the respiratory tract.

Airway smooth Cholinergic Neuropeptide Localization


muscle cell
Vasoactive intestinal peptide Parasympathetic
ACh
NO  Peptide histidine isoleucine/methionine (Afferent)
EFS iNANC Peptide histidine valine-42

Helodermin
Parasympathetic
nerve Helospectins I and II

Inflammation PACAP-27
Cholinergic Galanin

Substance P Afferent
ACh
NO Neurokinin A (C-fibers)

Neuropeptide K
EFS
ONOO O2 Calcitonin gene-related peptide
Bronchoconstriction
Gastrin-releasing peptide

Secretoneurin

Nociceptin
Inflammatory cells
Neuropeptide Y Sympathetic
FIG. 32.10 Nitric oxide (NO) and vasoactive intestinal peptide (VIP)
may modulate cholinergic neural effects mediated via acetylcholine Opioids?
(ACh). In inflammation NO may be removed by superoxide anions (O2)
generated form inflammatory cells and VIP by mast cell tryptase, therefore Somatostatin Afferent/uncertain
diminishing their braking effects, resulting in exaggerated cholinergic
Enkephalin
bronchoconstriction.
Endomorphins Cholecystokinin octapeptide

Airway neuropeptides
Many neuropeptides are localized to sensory, parasympa-
thetic, and sympathetic neurones in the human respiratory Neuropeptides and airway inflammation
tract (Table 32.1). These peptides have potent eects on
bronchomotor tone, airway secretions, bronchial circulation, Neuropeptides have multiple inammatory and immune
and on inammatory and immune cells [1]. Although the eects on the airways, thereby intensifying the ongoing inam-
precise physiological roles of each peptide are not yet fully mation [44]. In turn, inammatory mediators may amplify or
understood, clues are provided by their localization and sometimes dampen neuropeptide eects. Inammatory medi-
functional eects. Recently the development of specic neu- ators may increase the release of neuropeptides from sensory
ropeptide receptor antagonists has provided important new and other nerves, may increase the expression of neuropeptide
insights into the roles of these neurotransmitters. Many of genes in neural and inammatory cells, may increase the
the inammatory and functional eects of neuropeptides expression of neuropeptide receptors and may decrease
are relevant to asthma and there is compelling evidence for the degradation of neuropeptides.
the involvement of neuropeptides in the pathophysiology
and symptomatology of asthma and COPD.
Although classically neuropeptides are released from Vasoactive intestinal peptide and
autonomic nerves, there is increasing evidence that these related peptides
peptides may be synthesized and released from inamma-
tory and non-neural structural cells, particularly in disease. VIP-immunoreactive nerves are widely distributed through-
Inammatory cytokines may increase the expression of neu- out the respiratory tract in humans and there is also evidence
ropeptide genes in inammatory cells, so that inammatory for the presence of several closely related peptides (peptide
become a major source of the neuropeptide at the inam- histidine methionine, peptide histidine valine, helodermin,
matory site. For example, both VIP and SP have been local- helospectins I and II, and pituitary adenylate cyclase acti-
ized to human eosinophils and SP to macrophages [42, 43]. vating peptide (PACAP-38) and PACAP-27 which have

389
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 32.2 Effects of tachykinins on airways. plasma exudation and the vasodilator response to tachyki-
nins. Tachykinins may activate alveolar macrophages and
Effect Neurokinin receptor monocytes to release inammatory cytokines, such as IL-6.
Tachykinins also enhance cholinergic neurotransmission by
Bronchoconstriction (small  large airways) NK2, NK1 facilitating acetylcholine release at cholinergic nerve termi-
nals and by enhancing ganglionic transmission.
Plasma exudation NK1
Tachykinins are subject to degradation by at least
Bronchial vasodilatation NK1 two enzymes, angiotensin-converting enzyme (ACE), and
NEP. ACE is predominantly localized to vascular endothe-
Neurotransmission (ganglia, cholinergic NK1, NK2, NK3 lial cells and therefore degrades intravascular peptides,
nerves) whereas NEP is important for degrading tachykinins in
Cough (peripheral and central mechanisms) NK1, NK2, NK3
the airways. The activity of NEP may therefore determine
tachykinin responsiveness in the airways. Inhibition of
Mucus secretion (submucosal glands and NK1 NEP by phosphoramidon or thiorphan potentiates bron-
goblet cells) choconstriction in vitro in animal and human airways and
after inhalation in vivo. The activity of NEP is reduced by
Adhesion molecules (ICAM-1, E-selectin) NK1 mechanical removal of the epithelium, virus infections, cig-
Activation of inflammatory cells NK1, NK2 arette smoke, and hypertonic saline. Several of the stimuli
(macrophages, T-lymphocytes, eosinophils) known to induce bronchoconstrictor responses in asthmatic
patients have been found to reduce the activity of airway
Angiogenesis NK1 NEP [49].
Fibroblast activation NK1, NK2

Calcitonin gene-related peptide


CGRP-immunoreactive nerves are abundant in the respira-
tory tract of several species and is co-stored and co-localized
similar functional eects. VIP may be localized to para- with SP in aerent nerves. CGRP is a potent vasodilator,
sympathetic and sensory nerves. It is a potent vasodilator, a which has long-lasting eects and potently dilates bron-
bronchodilator, increases mucus secretion and may have anti- chial vessels in vitro and in vivo. It is possible that CGRP
inammatory eects. In some species it is a mediator of neu- may be the predominant mediator of arterial vasodilata-
rogenic bronchodilatation, but this is not the case in human tion and increased blood ow in response to sensory nerve
airways. A defect in VIP has been proposed in asthma, but stimulation in the bronchi. CGRP may be an important
there is little evidence for this [45]. mediator of airway hyperemia in asthma. CGRP has vari-
able eects on airway smooth muscle tone and appears
to act indirectly through the relapse of other constrictors, such
Tachykinins as endothelin. Like tachykinins, CGRP is chemotactic for
eosinophils.
SP and NKA, but not NKB, are localized to C-bers are
abundant in rodent airways, but are sparse in human airways
[46]. Tachykinins are also be expressed human macrophages,
which also express tachykinin receptors. Tachykinins have Neurogenic inflammation in
many dierent eects on the airways which may be relevant airway disease
to asthma and these eects are mediated via NK1-receptors
(preferentially activated by SP) and NK2-receptors (activated Sensory nerves may be involved in inammatory responses
by NKA) (Table 32.2). Tachykinins constrict smooth mus- through the antidromic release of neuropeptides from noci-
cle of human airways in vitro via NK2-receptors. Tachykinin ceptive nerves or C-bers via a local (axon) reex [50] (Fig.
receptors are widely distributed in human airways, with 32.11). The phenomenon is well documented in several
localization predominantly to airway smooth muscle, mucus- organs, including skin, eye, gastrointestinal tract, and blad-
secreting cells and bronchial cells [47]. NKA causes bronchoc- der. Neurogenic inammation occurs in the respiratory tract
onstriction after both intravenous and inhaled administration and may contribute to the inammatory response in asthma.
in asthmatic subjects. Mechanical removal of airway epithe- Neurogenic inammation is well documented in the air-
lium potentiates the bronchoconstrictor response to tachyki- ways of rodents, and there is good evidence that tachykinins
nins, largely because the neutral endopeptidase (NEP), which contribute to the airway hyperresponsiveness in several ani-
is a key enzyme in the degradation of tachykinins in airways mal models of asthma, using capsaicin depletion or specic
is strongly expressed on epithelial cells [48]. tachykinin antagonists. However, although it was proposed
SP stimulates mucus secretion from submucosal several years ago that neurogenic inammation and pep-
glands in human airways in vitro and is a potent stimulant tides released from sensory nerves might be important as an
to goblet cell secretion in guinea pig airways via activation amplifying mechanism in asthmatic inammation, there is
of NK1-receptors. NK1-receptors also mediate the increased little evidence to date to support this idea.

390
Neural and Humoral Control of the Airways 32
Epithelial Eosinophils
shedding

Sensory nerve
activation

Plasma
exudation
Vasodilatation

Mucus
secretion
Neuropeptide release
SP, NKA, CGRP, ....

Bronchoconstriction

Cholinergic
activation
Cholinergic facilitation

FIG. 32.11 Axon reflex in asthma. Possible neurogenic inflammation in asthmatic airways via retrograde release of peptides from sensory nerves via an
axon reflex. Substance P (SP) causes vasodiatation, plasma exudation and mucus secretion, whereas neurokinin A (NKA) causes bronchoconstriction and
enhanced cholinergic reflexes and calcitonin gene-related peptide (CGRP) vasodilatation.

There is some evidence in support of a role for tachy- Other neuropeptides


kinins in asthma:
An increase in SP-immunoreactive nerves has been Several other neuropeptides have been identied in human
described in patients with severe asthma [51]. airways (Table 32.1) [58], but their function is even less well
dened that the neuropeptides discussed. Secretoneurin is a
SP and NKA levels are increased in bronchoalveolar neuropeptide released by sensory nerves in the nose and is
lavage uid and induced sputum of asthmatic patients chemotactic for inammatory cells such as eosinophils [59],
[46]. but its role in asthma is uncertain.
There is increased expression of NK1- and NK2-receptors
in asthmatic lungs and airways [52, 53]. However this is
more evidence against a role for tachykinins:
SP-immunoreactive nerves are sparse in human airways CIRCULATING VASOACTIVE PEPTIDES
and are not increased in lungs and biopsies from AND HORMONES
asthmatic patients [45].
Capsaicin has no eect on human airways in vitro, Airway function can be altered by vasoactive peptides and hor-
whereas it potently constructs guinea pig airways. mones that reach the lungs from the bloodstream, as well as
Similarly, inhaled capsicum causes cough and by neurotransmitters released from nerve endings and by mol-
transient bronchoconstriction, but not prolonged ecules released locally from other cells within the airway [60].
bronchoconstriction as in rodents.
NEP inhibitors have no dierent eects in patients with
asthma than normal subjects [54]. Circulating catecholamines
Tachykinin antagonists are so far ineective in asthma.
Circulating epinephrine (adrenaline) is released from the
Neurogenic inammation plays a role in COPD. SP adrenal medulla into the circulation. It may reduce bronchial
levels are elevated in induced sputum of patients with COPD smooth muscle tone directly by stimulating 2-adrenergic
[55]. Cigarette smoke activates C-bers in airways and may receptors on airway smooth muscle, or indirectly by reducing
result in mucus hypersecretion and goblet cell discharge [56] acetylcholine release from cholinergic nerves. The lack of a
and tachykinins are potent stimuli of mucus secretion in bronchoconstrictor eect of -antagonists in normal subjects
human airways [57]. suggests that in this group, basal concentrations of circulating

391
Asthma and COPD: Basic Mechanisms and Clinical Management

adrenaline are probably not important in the regulation of 3.5


resting bronchomotor tone. In contrast, 2-antagonists cause
bronchoconstriction in some asthmatics; in the absence of 3.0

Plasma adrenaline (nmol/l)


an important sympathetic nerve supply to airway smooth
2.5
muscle, this suggests that basal concentrations of circulating
epinephrine are important in the maintenance of airway tone. 2.0
The controlling inuence of circulating epinephrine on airway
tone might operate particularly in those patients with asthma 1.5
in whom resting airway caliber is already reduced. In patients
with non-asthmatic COPD, however, 2-antagonists do not 1.0
normally cause bronchoconstriction. This nding would sug- 0.5
gest that basal concentrations of circulating adrenaline are not
important in the maintenance of airway tone in COPD. 0.0
Basal epinephrine concentrations and the circadian Base 1 2 3 4 5 2 5 10 20
variation in epinephrine concentrations in asthmatic patients
appear to be similar to those found in normal subjects [61 Stage Bruce Time post-exercise
protocol (min)
63]. Although Bates et al. reported that plasma epinephrine
levels at 10 pm were lower in patients with nocturnal asthma
FIG. 32.12 Plasma epinephrine concentrations during the course of and
than in a non-nocturnal asthma group [64]; correction for 20 min after maximal treadmill exercise in normal subjects (broken line)
of the nocturnal fall in plasma epinephrine does not alter and asthmatic subjects (solid) (no significant difference). Reproduced from
the peak expiratory ow values of patients with nocturnal Ref. [76] with permission.
asthma [65]. These ndings, taken together with the report
of nocturnal asthma occurring in a patient after adrenalec-
tomy [66], suggest that a fall in plasma epinephrine at night
is not a dominant factor in nocturnal asthma. The human natriuretic peptides include atrial natriuretic
Allergen- or pharmacological-induced bronchocon- peptide (ANP), brain natriuretic peptide (BNP), C-type
striction do not cause the release of epinephrine, which sug- natriuretic peptide (CNP), and urodilatin [82]. Most natri-
gests that epinephrine does not appear to have an important uretic peptides are produced primarily in the heart but are
homeostatic role in the regulation of airway caliber during released also in other tissues, including the kidneys, lungs,
bronchoconstriction to these stimuli [67, 68]. Even during and central nervous system. Specic ANP receptors have
acute exacerbations of asthma there may be no elevation been localized to lung (including airway smooth muscle), of
in plasma epinephrine levels [69, 70], although very high- which some may be the ANP-c or clearance receptor sub-
epinephrine concentrations have been found in some patients type [83], although the receptor subtype(s) in human airway
with acute severe asthma. The elevated epinephrine concen- smooth muscle is unknown. In isolated human airway tis-
trations achieved after strenuous exercise [71] cause bron- sue, ANP has a direct relaxant eect and confers protection
chodilation in both normal and asthmatic subjects [62, 63, against agonist-induced contraction [84, 85]. Two princi-
72] and may counteract bronchospasm induced by exercise pal mechanisms have been proposed for the inactivation of
in asthma [73]. Although a blunted catecholamine response ANP: degradation by NEP and binding to a non-guanylyl
to exercise in asthmatic patients has been reported by some cyclase clearance receptor (ANP-c receptor). NEP is widely
investigators, [74] other studies have found no signicant distributed within the airways and plays a role in modulat-
dierence in either the peak plasma catecholamine level ing the eect of ANP on airway smooth muscle [84, 85].
between normal and asthmatic subjects or in the response to An intravenous infusion of exogenous ANP causes
increasing levels of exercise (Fig. 32.12) [71, 75, 76]. bronchodilation and attenuates bronchial reactivity to
Norepinephrine, which has 1- and weak 2- inhaled histamine and to fog challenge [8690]. The rise
adrenergic activity in addition to -adrenergic eects, acts in plasma ANP levels during exercise is similar to these
as a neurotransmitter in the sympathetic nervous system but obtained during the lowest rates of ANP infusion;
overspills into the circulation. The infusion of noradrenaline, these results suggest that the rise in circulating ANP lev-
producing circulating concentrations within the physiologi- els during exercise may attenuation exercise-induced bron-
cal and pathophysiological range, has no eect on airway chospasm [76] (Fig. 32.13). Plasma ANP levels are elevated
caliber in either normal subjects or asthmatic subjects [62, in patients with cardiac failure [91] and pulmonary hyper-
63]. The third catecholamine present in the blood, dopamine, tension secondary to COPD [92]; under these circum-
has no inuence on resting bronchomotor tone in normal stances ANP may also play a protective role on the airways.
or stable asthmatic subjects [77] although it may attenuate Circulating ANP at physiological concentrations, however,
induced bronchoconstriction [78] and can decrease the rate appears unlikely to have any inuence on bronchomotor
of ventilation in animals and man [79]. tone in healthy subjects [88].

Natriuretic peptides Angiotensin II


Natriuretic peptides are a family of hormones that have The renin-angiotensin system plays an important role in
an important role in salt and water homeostasis [80, 81]. uid and electrolyte homeostasis through the actions of the

392
Neural and Humoral Control of the Airways 32
30 140

Plasma All (pg/ml)


AII Placebo
120 *
25 100
Plasma ANP (pmol/l)

80
20 60
*
15
40 *
20
0
10
35

systolic BP (mmHg)
Absolute change in
5 * *
25
0
Base 1 2 3 4 5 2 5 10 20 15 *
Stage Bruce Time post-exercise
protocol (min) 5

FIG. 32.13 Plasma ANP concentrations during exercise after maximal 5


treadmill exercise in normal subjects (broken line) and asthmatic subjects
5
(solid line) (no significant difference). Reproduced from Ref. [76] with

Change in FEV1 (%)


permission.
0

5
*
octapeptide angiotensin II. Angiotensin II is formed from 10
*
angiotensinogen by the action of renin and then ACE, 60
15
80% occurring within the pulmonary vascular endothelium.
An alternative ACE-independent pathway, possibly medi- 20
ated by several inammatory proteases [93], may also cause 0 20 40 60 80 100
the formation of angiotensin II. Time (min)
The eect of physiological concentrations of angio-
tensin II on basal bronchial tone of normal individuals is FIG. 32.14 Effect of infused angiotensin II (AII) on plasma levels of AII,
changes in systolic blood pressure (BP) and change in FEV1 from baseline
not know, whereas infusion of angiotensin II in mild asth-
values in eight asthmatic patients; *p  0.05 versus placebo. Reproduced
matic patients to plasma levels found in acute asthma causes from Ref. [94] with permission.
bronchoconstriction [94] (Fig. 32.14). Angiotensin II,
although causing only weak contraction of isolated human
and bovine bronchial rings, potentates the eects of metha- Adrenomedullin
choline and endothelin-1 in vitro [95, 96]. In patients with
mild asthma, angiotensin II at subthreshold concentrations Adrenomedullin is a 52-amino acid peptide that possesses
potentates methacholine-induced bronchoconstriction [96] vasodilator and natriuretic properties [107]. The chemical
but has no eect on endothelin- or histamine-evoked bron- structure of adrenomedullin is similar to CGRP. Immuno-
choconstriction in vivo [97, 98]. Angiotensin II receptor reactivity to adrenomedullin is found in tissues throughout
antagonists reduce allergen-induced airway hyperrespon- the body including the lung [108]. Adrenomedullin has a
siveness and eosinophil accumulation in guinea pigs [99] long-lasting bronchodilator action in guinea pig trachea [109,
and methacholine-induced bronchoconstriction in asthma 110], inhibits IL-8 secretion from alveolar macrophages
[100]. These results suggest a role for angiotensin II as a [111] and attenuates antigen-induced microvascular leakage
putative mediator in asthma. and bronchoconstriction in guinea pigs [112]. Plasma lev-
The renin-angiotensin system is activated in acute els of adrenomedullin are elevated in hypoxic patients with
severe asthma, but not in stable chronic asthma [70, 94]. The COPD [113] and in severe chronic asthma [114]. Its role
mechanism of activation is unclear, but nebulized 2-agonists in the control of airway smooth tone and inammation in
cause elevation of renin and angiotensin II in normal and asthma or COPD, however, remains uncertain.
mild asthmatic subjects through an ACE-dependent path-
way [101, 102]. This may occur via stimulation of -adreno-
ceptors on juxtaglomerular cells, but the levels of angiotensin Urotensin II
II seen in acute severe asthma are higher, suggesting the
existence of an alternative pathway of angiotensin II forma- Urotensin II is an 11-amino acid cyclic peptide, which acti-
tion. Exercise activates the renin-angiotensin system [103, vates specic receptors that are widely distributed in smooth
104], raising the possibility that elevated angiotensin II levels muscle [115]. It is a potent vasoconstrictor and a more
during exercise could contribute to exercise-induced bron- potent constrictor of airway smooth muscle than endothe-
chospasm. The renin-angiotensin system is also activated in lins [116]. Its role in regulating airway smooth muscle and
patients with COPD and edema [105, 106]. vascular tone in asthma and COPD are currently unknown.

393
Asthma and COPD: Basic Mechanisms and Clinical Management

Cortisol on the airways [126, 127]. However, one report found that
female athletes develop more severe exercise-induced asthma
Pharmacological doses of intravenous cortisol have no during the mid-luteal phase of the menstrual cycle when sali-
short-term eect on airway caliber in normal subjects [117]. vary progesterone levels were at there highest, suggesting that
Although glucocorticoids can potentate the response to cat- the menstrual cycle phase is an important determinant of the
echolamines in isolated bronchial tissue, the eect occurs severity of exercise-induced asthma in female athletes with
only at supraphysiological concentrations [118]. These results mild atopic asthma [128]. It is of interest that intramuscu-
suggest that endogenous cortisol is unlikely to have an lar progesterone has a benecial eect in some women with
important direct modulating eect on airway tone in nor- severe menstrual asthma [129].
mal individuals. Estrogen possesses both immunostimulatory and
In asthma the role of physiological concentrations of immunosuppressive properties and causes increased acetylcho-
circulating cortisol in airway function is uncertain. In noc- line activity in the lungs of animals [130], which could result
turnal asthma, the nadir in the circadian variation in plasma in an increase or decrease in airway tone. Estrogen receptor-
cortisol occurs 4 h before maximal bronchoconstriction decient mice exhibit increased airway responsiveness, pos-
[119], although the delayed action of cortisol means that it sibly due to reduced M2 muscarinic receptor function and
could still have an inuence on airway caliber. A reduced resultant increased acetylcholine release, suggesting a possible
nadir of plasma cortisol is found in patients with nocturnal role for estrogen in regulating airway function [131]. A pre-
asthma compared to a group without nocturnal asthma, but liminary report suggested that estrogen treatment may have
this nding may have been inuenced by previous cortico- steroid-sparing eects in women with severe asthma [132].
steroid therapy [120]. Other studies have found no direct
association between plasma cortisol concentrations and
nocturnal asthma [63]. Furthermore, the infusion of physi- Glucagon
ological concentrations of hydrocortisone, eliminating the
fall in plasma cortisol at night, does not prevent the noctur- Glucagon has bronchodilator actions, but whether it has a role
nal fall in peak ow rate in most asthmatic patients [119], in the control of airway smooth tone in humans is not estab-
suggesting that the circulating cortisol level is not the only lished [133]. Certainly intravenous glucagon has no signicant
factor in determining nocturnal asthma. Endogenous glu- bronchodilation action in patients with acute asthma [134].
cocorticoids rise following allergen challenge in allergic
asthmatic subjects and may play a role in the modulation of
allergen-induced bronchoconstriction [121].
ROLE OF HUMORAL MECHANISMS IN
Thyroid hormones ASTHMA AND COPD
The relationship between asthma and thyroid disease pro- Circulating hormones and vasoactive peptides appear to
vides indirect evidence of a role for thyroid hormones in play a minor role in the physiological regulation of airway
maintaining airway function. The development of hyperthy- tone in normal individuals. Epinephrine is the only hor-
roidism can be associated with deterioration in asthma con- mone known to inuence bronchomotor tone, and it is only
trol, with subsequent improvement in symptoms following during strenuous exercise that concentrations are elevated
appropriate treatment [122, 123]. Conversely the occurrence suciently to cause bronchodilation.
of hypothyroidism has been reported to be associated with Humoral mechanisms play a more important role in
improvement in asthma control, which relapses following the regulation of airway tone in diseased states of the air-
subsequent thyroxine replacement [124]. The possible mech- ways such as asthma and possibly in other disorders such as
anisms by which thyroid hormones could inuence airway COPD, cor pulmonale, congestive cardiac failure, respira-
smooth muscle tone and responsiveness are unclear [60]. tory failure, and thyroid disease. Circulating epinephrine has
a role in the maintenance of resting airway tone in asthma,
perhaps particularly in those patients in whom resting air-
Sex hormones way caliber is already reduced. The elevated epinephrine and
ANP concentrations achieved after vigorous exercise may
Progesterone has an important role in reducing the contrac- act to counteract exercise-induced asthma. It has not been
tility of uterine smooth muscle during pregnancy, and this established in asthma whether elevated circulating angio-
eect may be due to its inuence on gap junction formation tensin II achieved during exercise, or more particularly in
between smooth muscle cells. It has been suggested that pro- acute severe asthma, contributes to bronchospasm.
gesterone might cause similar eects on bronchial smooth
muscle. Progesterone could inuence airway smooth muscle
tone by other mechanisms indirectly either by potentiating References
the eect of catecholamines [118] or by potentiating airway
inammation [125]. Progesterone levels and airway respon- 1. Barnes PJ, Baraniuk J, Belvisi MG. Neuropeptides in the respiratory
siveness do not show a clear relationship during either the tract. Am Rev Respir Dis 144(11871198): 139199, 1991.
pregnancy or the menstrual cycle, although changes in the lev- 2. Barnes PJ. Autonomic Control of the Respiratory System. London:
els of other hormones may obscure an eect of progesterone Harwood, 1997.

394
Neural and Humoral Control of the Airways 32
3. Barnes PJ. Neurogenic inammation in the airways. Respir Physiol 125: 26. Minette PAH, Lammers J, Dixon CMS, McCusker MT, Barnes PJ.
14554, 2001. A muscarinic agonist inhibits reex bronchoconstriction in normal but
4. Nassenstein C, Kutschker J, Tumes D, Braun A. Neuro-immune inter- not in asthmatic subjects. J Appl Physiol 67: 246165, 1989.
action in allergic asthma: Role of neurotrophins. Biochem Soc Trans 34: 27. Ind PW, Dixon CMS, Fuller RW, Barnes PJ. Anticholinergic blockade
59193, 2006. of beta-blocker induced bronchoconstriction. Am Rev Respir Dis 139:
5. Taylor-Clark T, Undem BJ. Transduction mechanisms in airway 139094, 1989.
sensory nerves. J Appl Physiol 101: 95059, 2006. 28. Mak JCW, Baraniuk JN, Barnes PJ. Localization of muscarinic recep-
6. Fox AJ, Barnes PJ, Urban L, Dray A. An in vitro study of the proper- tor subtype mRNAs in human lung. Am J Respir Cell Mol Biol 7:
ties of single vagal aerents innervating guinea-pig airways. J Physiol 34448, 1992.
469: 2135, 1993. 29. Barnes PJ. Modulation of neurotransmission in airways. Physiol Rev 72:
7. Fox AJ, Lalloo UG, Belvisi MG, Bernareggi M, Chung KF, Barnes PJ. 699729, 1992.
Bradykinin-evoked sensitization of airway sensory nerves: A mecha- 30. Undem BJ, Riccio MM, Weinreich D, Ellis JL, Myers AC.
nism for ACE-inhibitor cough. Nature Med 2: 81417, 1996. Neurophysiology of mast cellnerve interactions in the airways. Int
8. Fox AJ, Barnes PJ, Dray A. Stimulation of aerent bres in the guinea Arch Allergy Immunol 107: 199201, 1995.
pig trachea by non-isomotic and low chloride solutions and its modu- 31. Haddad E-B, Mak JCW, Barnes PJ. Expression of -adrenergic and
lation by frusemide. J Physiol 482: 17987, 1995. muscarinic receptors in human lung. Am J Physiol 270: L94753, 1996.
9. Jia Y, Lee LY. Role of TRPV receptors in respiratory diseases. Biochim 32. Bai TR. Abnormalities in airway smooth muscle in fatal asthma: A
Biophys Acta 1772: 91527, 2007. comparison between trachea and bronchus. Am Rev Respir Dis 143:
10. McGarvey LP, Morice AH. Clinical cough and its mechanisms. Respir 44143, 1991.
Physiol Neurobiol 152: 36371, 2006. 33. Spina D, Rigby PJ, Paterson JW, Goldie RG. Autoradiographic locali-
11. Choudry NB, Fuller RW, Pride NB. Sensitivity of the human cough zation of beta-adrenoceptors in asthmatic human lung. Am Rev Respir
reex: Eect of inammatory mediators prostaglandin E2, bradykinin Dis 140: 141015, 1989.
and histamine. Am Rev Respir Dis 140: 13741, 1989. 34. Hakonarson H, Herrick DJ, Serrano PG, Grunstein MM. Mechanism
12. Nicholson CD, Challiss RAJ, Shahid M. Dierential modulation of of cytokine-induced modulation of -adrenoceptor responsiveness in
tissue function and therapeutic potential of selective inhibitors of cyclic airway smooth muscle. J Clin Invest 97: 2593600, 1996.
nucleotide phosphodiesterase isoenzymes. Trends Pharmacol Sci 12: 35. Koto H, Mak JCW, Haddad EB, Xu WB, Salmon M, Barnes PJ et al.
1927, 1991. Mechanisms of impaired -adrenergic receptor relaxation by inter-
13. Doherty MJ, Mister R, Pearson MG, Calverley PM. Capsaicin respon- leukin-1 in vivo in rat. J Clin Invest 98: 178087, 1996.
siveness and cough in asthma and chronic obstructive pulmonary 36. Penn RB, Shaver JR, Zangrilli JG, Pollice M, Fish JE, Peters SP et al.
disease. Thorax 55: 64349, 2000. Eects of inammation and acute -agonist inhalation on 2-AR sign-
14. Carr MJ, Hunter DD, Undem BJ. Neurotrophins and asthma. Curr aling in human airways. Am J Physiol 271: L60108, 1996.
Opin Pulm Med 7: 17, 2001. 37. Spina D, Rigby PJ, Paterson JW, Goldie RG. -Adrenoceptor func-
15. Freund-Michel V, Frossard N. The nerve growth factor and its recep- tion and autoradiographic distribution in human asthmatic lung. Br J
tors in airway inammatory diseases. Pharmacol Ther 117: 5276, 2008. Pharmacol 97: 7018, 1989.
16. Fox AJ, Patel HJ, Barnes PJ, Belvisi MG. Release of nerve growth fac- 38. Lammers JWJ, Barnes PJ, Chung KF. Non-adrenergic, non-cholinergic
tor by human pulmonary epithelial cells: Role in airway inammatory airway inhibitory nerves. Eur Respir J 5: 23946, 1992.
diseases. Eur J Pharmacol 424: 15962, 2001. 39. Belvisi MG, Stretton CD, Miura M, Verleden GM, Tadjarimi S,
17. de Vries A, Engels F, Henricks PA, Leusink-Muis T, McGregor GP, Yacoub MH et al. Inhibitory NANC nerves in human tracheal smooth
Braun A et al. Airway hyper-responsiveness in allergic asthma in guinea- muscle: A quest for the neurotransmitter. J Appl Physiol 73: 250510,
pigs is mediated by nerve growth factor via the induction of substance P: 1992.
A potential role for trkA. Clin Exp Allergy 36: 1192200, 2006. 40. Belvisi MG, Ward JR, Mitchell JA, Barnes PJ. Nitric oxide as a neuro-
18. Kassel O, de Blay F, Duvernelle C, Olgart C, Israel-Biet D, Krieger P transmitter in human airways. Arch Int Pharmacodyn Ther 329: 11120,
et al. Local increase in the number of mast cells and expression of nerve 1995.
growth factor in the bronchus of asthmatic patients after repeated 41. Ward JK, Belvisi MG, Fox AJ, Miura M, Tadjkarimi S, Yacoub MH
inhalation of allergen at low-dose. Clin Exp Allergy 31: 143240, 2001. et al. Modulation of cholinergic neural bronchoconstriction by endog-
19. Watson JJ, Fahey MS, van den WE, Engels F, Nijkamp FP, Stroemer P enous nitric oxide and vasoactive intestinal peptide in human airways
et al. TrkAd5: A novel therapeutic agent for treatment of inammatory in vitro. J Clin Invest 92: 73643, 1993.
pain and asthma. J Pharmacol Exp Ther 316: 112229, 2006. 42. Ho WZ, Lai JP, Zhu XH, Uvaydova M, Douglas SD. Human mono-
20. Grando SA, Kawashima K, Kirkpatrick CJ, Wessler I. Recent progress cytes and macrophages express substance P and neurokinin-1 receptor.
in understanding the non-neuronal cholinergic system in humans. Life J Immunol 159: 565460, 1997.
Sci 80: 218185, 2007. 43. Germonpre PR, Bullock GR, Lambrecht BN, Van DVV, Luyten WH,
21. Gwilt CR, Donnelly LE, Rogers DF. The non-neuronal cholinergic Joos GF et al. Presence of substance P and neurokinin 1 receptors in
system in the airways: An unappreciated regulatory role in pulmonary human sputum macrophages and U-937 cells. Eur Respir J 14: 77682,
inammation? Pharmacol Ther 115: 20822, 2007. 1999.
22. Neumann S, Razen M, Habermehl P, Meyer CU, Zepp F, 44. Barnes PJ. Cytokine modulators as novel therapies for airway disease.
Kirkpatrick CJ et al. The non-neuronal cholinergic system in peripheral Eur Respir J Suppl 34: 67s77s, 2001.
blood cells: Eects of nicotinic and muscarinic receptor antagonists on 45. Lilly CM, Bai TR, Shore SA, Hall AE, Drazen JM. Neuropeptide
phagocytosis, respiratory burst and migration. Life Sci 80: 236164, 2007. content of lungs from asthmatic and nonasthmatic patients. Am J
23. Barnes PJ. Muscarinic receptor subtypes in airways. Life Sci 52: Respir Crit Care Med 151: 54853, 1995.
52128, 1993. 46. De Swert KO, Joos GF. Extending the understanding of sensory neu-
24. Fryer AD, Adamko DJ, Yost BL, Jacoby DB. Eects of inammatory ropeptides. Eur J Pharmacol 533: 17181, 2006.
cells on neuronal M2 muscarinic receptor function in the lung. Life Sci 47. Mapp CE, Miotto D, Braccioni F, Saetta M, Turato G, Maestrelli P
64: 44955, 1999. et al. The distribution of neurokinin-1 and neurokinin-2 receptors in
25. Patel HJ, Barnes PJ, Takahashi T, Tadjkarimi S, Yacoub MH, human central airways. Am J Respir Crit Care Med 161: 20715, 2000.
Belvisi MG. Characterization of prejunctional muscarinic autorecep- 48. Frossard N, Rhoden KJ, Barnes PJ. Inuence of epithelium on guinea
tors in human and guinea-pig trachea in vitro. Am J Respir Crit Care pig airway responses to tachykinins: Role of endopeptidase and
Med 152: 87278, 1995. cyclooxygenase. J Pharmacol Exp Ther 248: 29298, 1989.

395
Asthma and COPD: Basic Mechanisms and Clinical Management

49. Di Maria GU, Belloore S, Geppetti P. Regulation of airway neuro- 73. Knox A, Campos-Gongora H, Wisniewski A, MacDonald I,
genic inammation by neutral endopeptidase [In Process Citation]. Tatterseld A. Modication of bronchial reactivity by physiological
Eur Respir J 12: 145462, 1998. concentrations of plasma epinephrine. J Appl Physiol 73: 10047, 1992.
50. Barnes PJ. Asthma as an axon reex. Lancet i: 24245, 1986. 74. Barnes P, Brown M, Silverman M, Dollery C. Circulating catecho-
51. Ollerenshaw SL, Jarvis D, Sullivan CE, Woolcock AJ. Substance P lamines in exercise and hyperventilation induced asthma. Thorax 36:
immunoreactive nerves in airways from asthmatics and non-asthmatics. 43540, 1981.
Eur Respir J 4: 67382, 1991. 75. Gilbert I, Lenner K, McFadden EJ. Sympathoadrenal response to
52. Adcock IM, Peters M, Gelder C, Shirasaki H, Brown CR, Barnes PJ. repetitive exercise in normal and asthmatic subjects. J Appl Physiol 64:
Increased tachykinin receptor gene expression in asthmatic lung and its 266774, 1988.
modulation by steroids. J Mol Endocrinol 11: 17, 1993. 76. Hulks G, Mohammed A, Jardine A, Connell J, Thomson NC.
53. Bai TR, Zhou D, Weir T, Walker B, Hegele R, Hayashi S et al. Circulating plasma concentrations of atrial natriuretic peptide and cat-
Substance P (NK1)- and neurokinin A (NK2)-receptor gene expression echolamines in response to maximal exercise in normal and asthmatic
in inammatory airway diseases. Am J Physiol 269: L30917, 1995. subjects. Thorax 46: 82428, 1991.
54. Cheung D, Timmers MC, Zwinderman AH, den Hartigh J, 77. Thomson N, Patel K. Eect of dopamine on airways conductance in
Dijkman JH, Sterk PJ. Neonatal endopeptidase activity and airway normals and extrinsic asthmatics. Br J Clin Pharmacol 5: 42124, 1978.
hyperesponsiveness to neurokin A in asthmatic subjects in vivo. Am 78. Michoud M, Amyot R, Jeanneret-Grosjean A. Dopamine eect on
Rev Respir Dis 148: 146773, 1993. bronchomotor tone in vivo. Am Rev Respir Dis 130: 75558, 1984.
55. Tomaki M, Ichinose M, Miura M, Hirayama Y, Yamauchi H, Nakajima N 79. Saaresranta T, Polo O. Hormones and breathing. Chest 122: 216582,
et al. Elevated substance P content in induced sputum from patients 2002.
with asthma and patients with chronic bronchitis. Am J Respir Crit 80. Ruskoaho H. Atrial natriuretic peptide: Synthesis, release, and metabo-
Care Med 151: 61317, 1995. lism. Pharmacol Rev 44: 479602, 1992.
56. Kuo H-P, Rohde JAL, Barnes PJ, Rogers DF. Cigarette smoke induced 81. Levin ER, Gardner DG, Samson WK. Natriuretic peptides. N Engl J
goblet cell secretion: Neural involvement in guinea pig trachea. Eur Med 339: 32128, 1998.
Respir J 3: 1895, 1990. 82. Hamad AM, Clayton A, Islam B, Knox AJ. Guanylyl cyclases, nitric
57. Rogers DF, Aursudkij B, Barnes PJ. Eects of tachykinins on mucus oxide, natriuretic peptides, and airway smooth muscle function. Am J
secretion on human bronchi in vitro. Eur J Pharmacol 174: 28386, Physiol Lung Cell Mol Physiol 285: L97383, 2003.
1989. 83. James S, Burnstock G. Atrial and brain natriuretic peptides share bind-
58. Uddman R, Hakanson R, Luts A, Sundler F. Distribution of neuropep- ing sites on cultured cells from the rat trachea. Cell Tissue Res 265:
tides in airways. In: Barnes PJ (ed.), Autonomic Control of the Respiratory 55565, 1991.
System, pp. 2137. London: Harwood Academic, 1997. 84. Angus R, Nally J, McCall R, Young L, McGrath J, Thomson NC.
59. Korsgren M, Fischer-Colbrie R, Andersson M, Coman WB, Mackay- Modulation of the eect of atrial natriuretic peptide in human and
Sim A, Persson CG et al. Secretoneurin is released into human airways bovine bronchi by phosphoramidon. Clin Sci 86: 29195, 1994.
by topical histamine but not capsaicin. Allergy 60: 45963, 2005. 85. Nally J, Clayton R, Thomson N, McGrath J. The interaction of alpha-
60. Thomson N, Dagg K, Ramsay S. Humoral control of airway tone. Thorax human atrial natriuretic peptide (ANP) with salbutamol, sodium nitro-
51: 46164, 1996. prusside and isosorbide dinitrate in human bronchial smooth muscle.
61. Berkin K, Inglis G, Ball S, Thomson N. Eect of low dose adrenaline Br J Pharmacol 113: 132832, 1994.
and noradrenaline infusions on airway calibre in asthmatic patients. 86. Hulks G, Jardine A, Connell J, Thomson N. Bronchodilator eect of
Clin Sci 70: 34752, 1986. atrial natriuretic peptide in asthma. BMJ 299: 108182, 1989.
62. Berkin K, Inglis G, Ball S, Thomson N. Airway responses to low con- 87. Chanez P, Mann CJB, Chabrier P, Godard P, Braquet P et al. Atrial
centrations of adrenaline and noradrenaline in normal subjects. Q J Exp natriuretic factor (ANF) is a potent bronchodilator in asthma. J Allergy
Physiol 70: 2039, 1985. Clin Immunol 86: 32124, 1990.
63. Barnes P, FitzGerald G, Brown M, Dollery C. Nocturnal asthma and 88. Hulks G, Jardine A, Connell J, Thomson NC. Eect of atrial natriu-
changes in circulating epinephrine, histamine, and cortisol. N Engl J retic factor on bronchomotor tone in the normal human airway. Clin
Med 303: 26367, 1980. Sci 79: 5155, 1990.
64. Bates M, Clayton M, Calhoun W, Jarjour N, Schrader L, Geiger K et al. 89. Mcalpine L, Hulks G, Thomson N. Eect of atrial natriuretic pep-
Relationship of plasma epinephrine and circulating eosinophils to noc- tide given by intravenous infusion on bronchoconstriction induced by
turnal asthma. Am J Respir Crit Care Med 149: 66772, 1994. ultrasonically nebulized distilled water (fog). Am Rev Respir Dis 146:
65. Morrison J, Teale C, Pearson S, Marshall P, Dwyer N, Jones S et al. 91215, 1992.
Adrenaline and nocturnal asthma. BMJ 301: 47376, 1990. 90. Angus R, McCallum M, Hulks G, Thomson N. Bronchodilator, car-
66. Morice A, Sever P, Ind P. Adrenaline, bronchoconstriction, and asthma. diovascular, and cyclic guanylyl monophosphate response to high-dose
BMJ 293: 53940, 1986. infused atrial natriuretic peptide in asthma. Am Rev Respir Dis 147:
67. Arvidsson P, Larsson S, Lfdahl C, Melander B, Whlander L, 112225, 1993.
Svedmyr N. Formoterol, a new long-acting bronchodilator for inhala- 91. Raine A, Erne P, Brgisser E, Mller F, Bolli P, Burkart F et al. Atrial
tion. Eur Respir J 2: 32530, 1989. natriuretic peptide and atrial pressure in patients with congestive heart
68. Larsson K, Gronneberg R, Hjemdahl P. Bronchodilatation and inhi- failure. N Engl J Med 315: 53337, 1986.
bition of allergen-induced bronchoconstriction by circulating epine- 92. Burghuber O, Hartter E, Punzengruber C, Weissel M, Woloszczuk W.
phrine in asthmatic subjects. J Allergy Clin Immunol 75: 58693, 1985. Human atrial natriuretic peptide secretion in precapillary pulmonary
69. Ind P, Causon R, Brown M, Barnes P. Circulating catecholamines in hypertension. Clinical study in patients with COPD and interstitial
acute asthma. BMJ 290: 26769, 1985. brosis. Chest 93: 3137, 1988.
70. Ramsay SG, Dagg KD, McKay IC, Lipworth BJ, McSharry C, 93. Husain A. The chymaseangiotensin system in humans. J Hypertens
Thomson NC. Investigations on the renin-angiotensin system in acute 11: 115559, 1993.
severe asthma. Eur Respir J 10: 276671, 1997. 94. Millar E, Angus R, Hulks G, Morton J, Connell J, Thomson N.
71. Berkin K, Walker G, Inglis G, Ball S, Thomson N. Circulating adrena- Activity of the reninangiotensin system in acute severe asthma and
line and noradrenaline concentrations during exercise in patients with the eect of angiotensin II on lung function. Thorax 49: 49295, 1994.
exercise induced asthma and normal subjects. Thorax 43: 29599, 1988. 95. Nally J, Clayton R, Wakelam M, Thomson N, McGrath J. Angiotensin
72. Warren J, Dalton N. A comparison of the bronchodilator and vasopressor II enhances responses to endothelin-1 in bovine bronchial smooth
eects of exercise levels of adrenaline in man. Clin Sci 64: 47579, 1983. muscle. Pulm Pharmacol 7: 40913, 1994.

396
Neural and Humoral Control of the Airways 32
96. Millar EA, Nally JE, Thomson NC. Angiotensin II potentiates meth- 113. Cheung B, Leung R. Elevated plasma levels of human adrenomedul-
acholine-induced bronchoconstriction in human airway both in vitro lin in cardiovascular, respiratory, hepatic and renal disorders. Clin Sci
and in vivo. Eur Respir J 8: 183841, 1995. 92: 5962, 1997.
97. Chalmers GW, Millar EA, Little SA, Shepherd MC, Thomson NC. 114. Ceyhan B, Karakurt S, Hekim N. Plasma adrenomedullin levels in
Eect of infused angiotensin II on the bronchoconstrictor activity of asthmatic patients. J Asthma 38: 22127, 2001.
inhaled endothelin-1 in asthma. Chest 115: 35256, 1999. 115. Ames RS, Sarau HM, Chambers JK, Willette RN, Aiyar NV,
98. Ramsay S, Clayton R, Dagg K, Thomson L, Nally J, Thomson N. Romanic AM et al. Human urotensin-II is a potent vasoconstrictor
Eect of angiotensin II on histamine-induced bronchoconstriction in and agonist for the orphan receptor GPR14. Nature 401: 28286,
the human airway both in vitro and in vivo. Respir Med 91: 60915, 1999.
1997. 116. Hay DWP, Luttmann MA, Douglas SA. Human urotensin-II is a
99. Myou S, Fujimura M, Kurashima K, Tachibana H, Watanabe K, potent spasmogen of primate airway smooth muscle. Br J Pharmacol
Hirose T. Type 1 angiotensin II receptor antagonism reduces antigen- 131: 1012, 2000.
induced airway reactions. Am J Respir Crit Care Med 162: 4549, 117. Ramsdell JW, Berry CC, Clausen JL. The immediate eects of cor-
2000. tisol on pulmonary function in normals and asthmatics. J Allergy Clin
100. Myou S, Fujimura M, Kamio Y, Ishiura Y, Kurashima K, Tachibana H Immunol 72: 6974, 1983.
et al. Eect of Losartan, a type 1 angiotensin II receptor antagonist, 118. Foster P, Goldie R, Paterson J. Eect of steroids on beta-adrenoceptor-
on bronchial hyperresponsiveness to methacholine in patients with mediated relaxation of pig bronchus. Br J Pharmacol 78: 44145, 1983.
bronchial asthma. Am J Respir Crit Care Med 162: 4044, 2000. 119. Soutar C, Costello J, Ijaduola O, Turner-Warwick M. Nocturnal and
101. Millar E, McInnes G, Thomson N. Investigation of the mechanism morning asthma, relationship to plasma corticosteroids and response
of beta 2-agonist-induced activation of the renin-angiotensin system. to cortisol infusion. Thorax 30: 43640, 1975.
Clin Sci 88: 43337, 1995. 120. Kallenbach JM, Panz VR, Joe BI, Jankelow D, Anderson R, Haitas B
102. Millar EA, Connell JM, Thomson NC. The eect of nebulized et al. Nocturnal events related to morning dipping in bronchial asthma.
albuterol on the activity of the reninangiotensin system in asthma. Chest 93: 75157, 1988.
Chest 111: 7174, 1997. 121. Stokes P, Togias, Bickel, Diemer, Hubbard, Schleimer. Endogenous
103. Kosunen KJ, Pakarinen AJ, Kuoppasalmi K, Adlercreutz H. Plasma glucocorticoids and antigen-induced acute and late phase pulmonary
renin activity, angiotensin II, and aldosterone during intense heat responses. Clin Exp Allergy 30: 125765, 2000.
stress. J Appl Physiol 41: 32327, 1976. 122. Ayres J, Clark T. Asthma and the thyroid. Lancet 2: 111011, 1981.
104. Milledge JS, Catley DM. Renin, aldosterone, and converting enzyme 123. Lipworth B, Dhillon D, Clark R, Newton R. Problems with asthma
during exercise and acute hypoxia in humans. J Appl Physiol 52: following treatment of thyrotoxicosis. Br J Dis Chest 82: 31014, 1988.
32023, 1982. 124. Bush RK, Ehrlich EN, Reed CE. Thyroid disease and asthma.
105. Reihman DH, Farber MO, Weinberger MH, Henry DP, Fineberg NS, J Allergy Clin Immunol 59: 398401, 1977.
Dowdeswell IRG et al. Eect of hypoxemia on sodium and water excre- 125. Hellings PW, Vandekerckhove P, Claeys R, Billen J, Kasran A,
tion in chronic obstructive lung disease. Am J Med 78: 8794, 1985. Ceuppens JL. Progesterone increases airway eosinophilia and hyper-
106. Anand I, Chandrashekhar Y, Ferrari R, Sarma R, Guleria R, Jindal S responsiveness in a murine model of allergic asthma. Clin Exp Allergy
et al. Pathogenesis of congestive state in chronic obstructive pulmo- 33: 145763, 2003.
nary disease. Studies of body water and sodium, renal function, hemo- 126. Juniper E, Daniel E, Roberts R, Kline P, Hargreave F, Newhouse M.
dynamics, and plasma hormones during edema and after recovery. Improvement in airway responsiveness and asthma severity during
Circulation 86: 1221, 1992. pregnancy. A prospective study. Am Rev Respir Dis 140: 92431, 1989.
107. Jougasaki M, Burnett JC. Adrenomedullin: Potential in physiology 127. Tan KS, Thomson NC. Asthma in pregnancy. Am J Med 109: 72733,
and pathophysiology. Life Sci 66: 85572, 2000. 2000.
108. Ichiki Y, Kitamura K, Kangawa K, Kawamoto M, Matsuo H, Eto T. 128. Stanford K, Mickleborough T, Ray S, Lindley M, Koceja D, Stager J.
Distribution and characterization of immunoreactive adrenomedullin Inuence of menstrual cycle phase on pulmonary function in asthmatic
in human tissue and plasma. FEBS Lett 338: 610, 1994. athletes. Eur J Appl Physiol 96: 70310, 2006.
109. Kanazawa H, Kurihara N, Hirata K, Kudoh S, Kawaguchi T, 129. Beynon H, Garbett N, Barnes P. Severe premenstrual exacerbations of
Takeda T. Adrenomedullin, a newly discovered hypotensive peptide, asthma: Eect of intramuscular progesterone. Lancet 2: 37072, 1988.
is a potent bronchodilator. Biochem Biophys Res Commun 205: 25154, 130. Abdul-Karim R, Marshall L, Nesbitt RJ. Inuence of estradiol-17
1994. beta on the acetylcholine content of the lung in the rabbit neonate.
110. Nishimura J, Seguchi H, Sakihara C, Kureishi Y, Yoshimura H, Am J Obstet Gynecol 107: 64144, 1970.
Kobayashi S et al. The relaxant eect of adrenomedullin on particular 131. Carey MA, Card JW, Bradbury JA, Moorman MP, Haykal-Coates N,
smooth muscles despite a general expression of its mRNA in smooth Gavett SH et al. Spontaneous airway hyperresponsiveness in estro-
muscle, endothelial and epithelial cells. Br J Pharmacol 120: 193200, gen receptor-{alpha}-decient mice. Am J Respir Crit Care Med 175:
1997. 12635, 2007.
111. Kamoi H, Kanazawa H, Hirata K, Kurihara N, Yano Y, Otani S. 132. Myers JR, Sherman CB. Should supplemental estrogens be used as
Adrenomedullin inhibits the secretion of cytokine-induced neutrophil steroid-sparing agents in asthmatic women? Chest 106: 31819, 1994.
chemoattractant, a member of the interleukin-8 family, from rat alve- 133. Sherman M, Lazar E, Eichacker P. A bronchodilator action of gluca-
olar macrophages. Biochem Biophys Res Commun 211: 103135, 1995. gon. J Allergy Clin Immunol 81: 90811, 1988.
112. Ohbayashi H, Suito H, Yoshida N, Ilto Y, Kume H, Yamaki K. 134. Wilber ST, Wilson JE, Blanda M, Gerson LW, Meerbaum SO,
Adrenomedullin inhibits ovalbumin-induced bronchoconstriction and Janas G. The bronchodilator eect of intravenous glucagon in asthma
airway microvascular leakage in guinea-pigs. Eur Respir J 14: 107681, exacerbation: A randomized, controlled trial. Ann Emerg Med 36:
1999. 42731, 2000.

397
Pathogenic 5
PART

Mechanisms in
Asthma and COPD
Pathophysiology of Asthma
33CHAPTER

INTRODUCTION ASTHMA AS AN INFLAMMATORY Peter J. Barnes1 and


DISEASE Jeffrey M. Drazen2
This chapter aims to provide a brief overview 1
National Heart and Lung Institute
of disease mechanisms to integrate some of the It had been recognized for many years that (NHLI), Clinical Studies Unit,
detailed information provided in earlier chap- patients who die of asthma attacks have grossly Imperial College, London, UK
ters into a framework with clinical relevance. inamed airways. The airway lumen is occluded 2
Department of Medicine, Pulmonary
Asthma and chronic obstructive pulmonary dis- by tenacious mucus plugs composed of plasma Division, Brigham and Womens
ease (COPD) are both highly complex condi- proteins exuded from airway vessels and mucus
Hospital, Boston, MA, USA
tions whose pathobiology remains inadequately glycoproteins secreted from surface epithelial
dened. In both diseases there are many dier- cells. The airway wall is edematous and inltrated
ent and distinct inammatory cells and multiple with inammatory cells, which are predominantly
mediators with complex acute and chronic eects eosinophils and T-lymphocytes [2]. The airway
on the airways and airspaces. We now appreci- epithelium is invariably shed in a patchy man-
ate that these changes may vary among patients ner and clumps of epithelial cells are found in
because of genetic variance in susceptibility [1]. the airway lumen. Occasionally there have been
Although our understanding of asthma patho- opportunities to examine the airways of asth-
genesis has been enhanced by the application of matic patients who die in accidents thought to be
new molecular, cell biological, and genetic tech- unrelated to their asthma. In this setting inam-
niques to the condition, and the key test of the matory changes have been observed but they are
acquired knowledge is the success of new speci- less marked than those observed in patients with
cally targeted therapies. In this area the successes active asthma [3]. These studies also reveal that
have been few in asthma and are virtually absent the inammation in asthmatic airways involves
in COPD. Thus, even though we have made not only the trachea and bronchi, but also extends
considerable advances in understanding asthma, to the terminal bronchioles [4]; some investiga-
there remain many fundamental questions that tors using transbronchial biopsies have shown
need to be answered. inammatory cells in the parenchyma [5].
Our views on asthma and COPD have It has also been possible to examine the
continued to evolve. Although it is recognized airways of asthmatic patients by beroptic and
that chronic inammation underlies the clinical rigid bronchoscopy, bronchial biopsy, and bron-
syndromes, it has been hard to dene the precise choalveolar lavage (BAL). Direct bronchoscopic
nature of this inammation, much less its pri- examination reveals that the airways of asthmatic
mary etiology. Nevertheless it is appreciated that patients are often erythematous and swollen,
the nal consequence of this chronic inamma- indicating acute inammation. Lavage of the air-
tory response is an abnormal control of airway ways has revealed an increase in the numbers of
caliber in vivo in asthma. In contrast, the evolu- lymphocytes, mast cells, and eosinophils and evi-
tion of understanding in COPD has been much dence for activation of macrophages in compari-
slower; key insights are sought which will open up son with nonasthmatic controls. Biopsies have
the understanding of this complex disorder (see provided evidence for increased numbers and
Chapter 34). activation of mast cells, macrophages, eosinophils,

401
Asthma and COPD: Basic Mechanisms and Clinical Management

T-lymphocytes including regulatory T-cells, and mucin airway cells or that modify the mechanical characteristics
secreting cells [69]. These changes are found even in patients of the airway wall. AHR may be due to increased release of
with mild asthma who have few symptoms [10], suggesting mediators (such as histamine and leukotrienes from mast
that inammation may be found in all asthmatic patients who cells), abnormal behavior of airway smooth muscle, thicken-
are symptomatic. Similar inammatory changes have been ing of the airway wall by reversible (edema), and irreversible
found in bronchial biopsies of children with asthma, even at (airway smooth muscle thickening, brosis) elements. Airway
the onset of symptoms [11, 12]. sensory nerves may also contribute importantly to symptoms,
such as cough and chest tightness, as the nerves become sen-
sitized by the chronic inammation in the airways.
Although most attention has been focused on the acute
AIRWAY HYPERRESPONSIVENESS inammatory changes seen in asthmatic airways (bronchoc-
onstriction, plasma exudation, mucus secretion), asthma is a
chronic disease, persisting over many years in most patients.
The relationship between inammation and clinical symp- Superimposed on this chronic inammatory state are acute
toms of asthma is not clear. There is evidence that the degree inammatory episodes, which correspond to exacerbations
of inammation is loosely related to airway hyperrespon- of asthma. It is clearly important to understand the mecha-
siveness (AHR), as measured by histamine or methacholine nisms of acute and chronic inammation in asthmatic air-
challenge. However, the degree of inammation, as measured ways and to investigate the long-term consequences of this
by the number of various types of inammatory cells in the chronic inammation on airway function.
lesion, is not closely linked to the clinical severity of asthma or
AHR [13]. This suggests that other factors, such as structural
changes in the airway wall, are important in mediating the
clinical symptoms of asthma. The increased airway respon-
siveness in asthma is a striking physiological abnormality that
INFLAMMATORY CELLS
is present even when airway function is otherwise normal. It is
likely that there are several factors that underlie this increased Many dierent inammatory cells are involved in asthma,
responsiveness to constrictor agents, particularly those that although the precise role of each cell type is not yet certain
act indirectly by releasing bronchoconstrictor mediators from (Fig. 33.1). It is evident that no single inammatory cell is

Inhaled allergens

Epithelial cells
CCL11
Mast cell SCF

Histamine TSLP
Cys-LTs
PGD2 Dendritic cell
IL-9
CCL17
CCL22
Bronchoconstriction T-reg
CCR4
IgE
Th2 cell

IL-4, IL-13 IL-5


CCR3

B-Iymphocyte
Eosinophils

FIG. 33.1 A potential schema of Inflammation in asthma. Inhaled allergens activate sensitized mast cells by crosslinking surface-bound IgE molecules to
release several bronchoconstrictor mediators, including cysteinyl-leukotrienes (cys-LT) and prostaglandin D2 (PGD2). Epithelial cells release stem-cell factor
(SCF), which is important for maintaining mucosal mast cells at the airway surface. Allergens are processed by myeloid dendritic cells, which are conditioned
by thymic stromal lymphopoietin (TSLP) secreted by epithelial cells and mast cells to release the chemokines CC-chemokine ligand 17 (CCL17) and CCL22,
which act on CC-chemokine receptor 4 (CCR4) to attract T helper 2 (Th2) cells. Th2 cells have a central role in orchestrating the inflammatory response in
allergy through the release of interleukin-4 (IL-4) and IL-13 (which stimulate B-cells to synthesize IgE), IL-5 (which is necessary for eosinophilic inflammation)
and IL-9 (which stimulates mast-cell proliferation). Epithelial cells release CCL11, which recruits eosinophils via CCR3. Patients with asthma may have a defect
in regulatory T-cells (T-reg), which may favor further Th2-cell proliferation.

402
Pathophysiology of Asthma 33
able to account for the complex pathophysiology of asthma Macrophages
but some cells predominate in asthmatic inammation. The
inammation in asthmatic airways diers strikingly from that Macrophages, which are derived from blood monocytes,
observed in COPD, where there is a predominance of mac- may trac into the airways in asthma and may be activated
rophages, cytotoxic (CD8) T-lymphocytes, and neutrophils, by allergen via low-anity IgE receptors (Fc RII) [23]. The
although both of these common diseases may coexist in some enormous immunological repertoire of macrophages allows
patients. However, although the dierences in inammation these cells to produce many dierent products, including a
between mild asthma and COPD are well described, it is not large variety of cytokines that may orchestrate the inam-
recognized that patients with severe asthma have inamma- matory response. Macrophages have the capacity to initiate
tory changes that are similar to those of COPD, with increased a particular type of inammatory response via the release of
numbers of neutrophils, CD4 Th1 and CD8 cells [14]. a certain pattern of cytokines. Macrophages may increase
or decrease inammation depending on the stimulus. For
example, alveolar macrophages from normal and patients
Mast cells with mild asthma phagocytose apoptic cells resulting from
lipopolysaccharide (LPS) exposure in vitro and produce
Mast cells are clearly important in initiating the acute bron- the anti-inammatory mediator PGE2. In contrast alveolar
choconstrictor responses to allergen and probably to other macrophages from patients with severe asthma are impaired
indirect stimuli, such as exercise and hyperventilation (via with respect to apoptotic cell uptake and production of
osmolality or thermal changes) and fog. Mast cell activation PGE2 [24]. These observations indicate that the severity
is likely to play a key role in the symptoms of asthma and of asthma and perhaps its chronicity can modulate the role
during acute exacerbations. There is inltration of airway of macrophages in asthma. It is also possible that the oppo-
smooth muscle by mast cells in patients with asthma that site is true, that is, patients with severe asthma are the indi-
is associated with the characteristic disordered airway func- viduals with defective macrophage function. It is established
tion of this condition [15]. Treatment of asthmatic patients that repeated short term exposure to ozone recruits alveolar
with prednisone results in a decrease in the number of tryp- macrophages to the airways of patients with mild asthma
tase-only positive mast cells [16]. Furthermore, the number [25] and that there is an association between the amount of
of mast cells in induced sputum in patients with seasonal carbon in alveolar macrophages, which had to be activated
allergic rhinitis is related to the degree of AHR [17]. to ingest this material, and lung function [26]. Thus the
There appears to be an inltration of mast cells into airway role of alveolar macrophages appears to be to modulate the
smooth muscle of asthmatic patients and this is associated expression and severity of asthma.
with AHR [15]. The key role of mast cells in asthma has
been the subject of comprehensive reviews [7, 18]. Mast
cells are maintained at the mucosal surface by the secre- Dendritic cells
tion of stem cell factor, which acts on c-kit receptors, by
epithelial cells [19]. However, it is now clear that mast cells By contrast, dendritic cells (which are specialized macro-
alone cannot account for all the pathobiological changes phage-like cells in the airway epithelium) are very eec-
in asthma and that other cells, such as macrophages, eosi- tive antigen-presenting cells and may therefore play a very
nophils, and T-lymphocytes, play key roles in the chronic important role in the initiation of allergen-induced responses
inammatory process including induction and maintenance in asthma [27, 28]. Dendritic cells take up allergens though
of AHR. their long nger-like projections, which extend into the air-
Classically mast cells are activated by allergens way lumen, process them to peptides, and migrate to local
through an IgE-dependent mechanism. The importance of lymph nodes where they present the allergenic peptides to
IgE in the pathophysiology of asthma has been highlighted uncommitted T-lymphocytes, to program the production of
by clinical studies with humanized anti-IgE antibodies, allergen-specic T-cells. Although there is no established
which inhibit IgE-mediated eects. Although anti-IgE ways to manipulate dendritic cells, it has been shown that
antibody results in a reduction in circulating IgE to unde- during experimental allergen challenge in humans that a sig-
tectable levels, this treatment results in minimal clinical nicantly greater proportion of CD33 cells expressed the
improvement in patients with severe steroid-dependent cys-LT1-receptor compared with CD123 cells. Pranlukast,
asthma [20]. Examination of the patients receiving anti-IgE cys-LT1-receptor antagonist, decreased the allergen-induced
therapy with omalizumab reveals downregulated expres- decrease in CD33 dendritic cells at 3 h compared with
sion of IgE receptors (Fc RI) on mast cells and basophils placebo treatment. Based on this nding it is reasonable to
[21]. Biopsy studies show a profound reduction in tissue speculate that anti-leukotriene treatment could modify the
eosinophilia, together with reductions in T-cell and B-cell initial responses to allergen exposure [29].
numbers in the airway submucosa [22]. These observations
suggest that treatment with anti-IgE may be more eec-
tive in early stage rather than late stage asthma; however, Eosinophils
no clinically directive trials with omalizumab have been
conducted in this population. Furthermore, unless the Eosinophil inltration is a characteristic feature of asthmatic
cost of such therapy is dramatically reduced it is unlikely airways and dierentiates asthma from other non-infectious
that such treatment would be used in patients with mild inammatory conditions of the airway. Indeed, asthma was
disease. described as chronic eosinophilic bronchitis as early as 1916.

403
Asthma and COPD: Basic Mechanisms and Clinical Management

Allergen inhalation results in a marked increase in eosi- require the presence of various growth factors, of which
nophils in BAL uid at the time of the late reaction, and GM-CSF and IL-5 appear to be the most important [47].
there is a correlation between eosinophil counts in periph- In the absence of these growth factors eosinophils undergo
eral blood or bronchial lavage and AHR. Eosinophils are programmed cell death (apoptosis) [48].
linked to the development of AHR through the release of A humanized monoclonal antibody to IL-5 has been
basic proteins and oxygen-derived free radicals [30, 31]. administered to asthmatic patients [49]; and, as in animal
An important area of research is now concerned with studies, there is a profound and prolonged reduction in cir-
the mechanisms involved in recruitment of eosinophils into culating eosinophils. Although the inltration of eosinophils
asthmatic airways. Eosinophils are derived from bone mar- into the airway after inhaled allergen challenge is completely
row precursors; the Th2 cytokine IL-5 is a unique mediator blocked, there is no eect on the response to inhaled aller-
of eosinophil dierentiation and survival in response to aller- gen and no reduction in AHR. Anti-CCR3, anti-integrin,
gen provocation. After allergen challenge eosinophils appear and antiselectin treatments are currently being consid-
in BAL uid during the late response, and this appearance ered as anti-eosinophil strategies for treatment of asthma;
is associated with a decrease in peripheral eosinophil counts the results of these studies should better dene the role of
and with the appearance of eosinophil progenitors in the eosinophils as mediators of asthmatic events. Eosinophils
circulation. The signal for increased eosinophil production may play a role in airway remodeling rather than AHR in
is likely IL-5 derived from the inamed airway. Eosinophil asthma as anti-IL-5 treatment is able to reduce extracellular
recruitment initially involves adhesion of eosinophils to vas- matrix deposition in asthma [50].
cular endothelial cells in the airway circulation, their migra-
tion into the submucosa, and their subsequent activation. The
role of individual adhesion molecules, cytokines, and media- Neutrophils
tors in orchestrating these responses has been extensively
investigated but since there are no pathways that are unique The eosinophil has been the recipient of current attention as
to eosinophils, this aspect of eosinophil biology has not been an eector cell in asthma, but attention has been returning
extensively probed. Adhesion of eosinophils involves the to the role of neutrophils [51, 52]. Although neutrophils are
expression of specic glycoprotein molecules on the surface not a predominant cell type observed in the airways of most
of eosinophils (integrins) and their expression of such mol- patients with mild-to-moderate chronic asthma, they appear
ecules as intercellular adhesion molecule-1 (ICAM-1) on to be a more prominent cell type in airways and induced spu-
vascular endothelial cells. An antibody directed at ICAM-1 tum of patients with more severe asthma [5357]. The mecha-
markedly inhibits eosinophil accumulation in the airways nism of neutrophilic inammation are not yet well understood,
after allergen exposure and also blocks the accompanying but several mediators, including CXCL8 (IL-8) are likely to
hyperresponsiveness [32]. However, ICAM-1 is not selective be involved (Fig. 33.2). Sputum analysis shows that 20%
for eosinophils and cannot account for the selective recruit- of asthmatics, even with mild disease, have a predominantly
ment of eosinophils in allergic inammation. Eosinophil neutrophilic pattern of inammation [58]. Also in patients
migration may be due to the eects of lipid mediators, such who die suddenly of asthma, large numbers of neutrophils are
as leukotrienes [33, 34] and possibly platelet-activating factor found in their airways [59] although this may reect the rapid
(PAF), to the eects of cytokines, such as GM-CSF and IL- kinetics of neutrophil recruitment compared to eosinophil
5 both of which are important for the survival of eosinophils inammation. Rapid withdrawal of corticosteroids results in
in the airways and prime eosinophils to exhibit enhanced the appearance of neutrophils in the airways of patients with
responsiveness. Eosinophils from asthmatic patients show asthma [60]. There are data to suggest that monomeric IgE
exaggerated responses to PAF and phorbol esters, compared may mediate the appearance of neutrophils in the airways
with eosinophils from atopic nonasthmatic individuals [35]. of asthmatics and may represent the anti-apoptic activity of
This is further increased by allergen challenge [36, 37] sug- myeloid cell leukemia-1 protein, decreased caspase-3 activ-
gesting that they may have been primed by exposure to ity, diminished availability of Smac from mitochondria, and
cytokines in the circulation. sequestration of the pro-apoptic protein Bax in the cytoplasm
There are several mediators involved in the migration of [61]. Thus the presence of neutrophils in the airway may reect
eosinophils from the circulation to the surface of the airway. the inammatory microenvironment rather than indicate a
There are data showing that cysteinyl leukotrienes, through causal role for this cell. Further research into the role of neu-
stimulation at the cys-LT1-receptor, are in part responsi- trophils especially in severe asthma is ongoing [62].
ble for eosinophilopoiesis in human airways [38], but other
mediators such as the eotaxins, a family of chemoattractant
cytokines that promote eosinophil recruitment to tissues in
response to allergic provocation via CCR3 receptors also play T-lymphocytes
a role [3942]. Other potent and selective eosinophil chem-
oattractants include chemokines, such as CCL5 (RANTES) Th2 cells
and CCL13 (MCP-4), that are expressed in epithelial cells T-lymphocytes play a very important role in co-ordinating
[40, 43, 44] but data for their involvement in intact humans the inammatory response in asthma through the release of
are minimal. There appears to be a co-operative interaction specic patterns of cytokines, resulting in the recruitment and
between IL-5, IL-13, and chemokines, so that more than one survival of eosinophils and in the maintenance of mast cells
cytokine may be necessary for the eosinophilic response in in the airways. T-lymphocytes are coded to express a distinc-
airways [45, 46]. Once recruited to the airways, eosinophils tive pattern of cytokines, which are similar to that described

404
Pathophysiology of Asthma 33
Viruses Allergens Allergen

Antigen presenting cell


Dendritic cell Dendritic cell, macrophage
TCR
Epithelial cells MHC1
Allergenl B7-2
peptide
TCR CD28
IL-17 IL-23 Mast cell
Th0
TNF- CXCL1, CXCL8 Th17
IL-12 IL-4
IL-18
Neutrophils Th1
IFN- Th2
Neutrophil IL-4
IgE
elastase CXCL8 T-bet IL-13
MMP-9 TGF-
IL-10
TGF- IL-5
Th17 IL-2
Goblet cells IFN-
Fibroblast
Treg
Mucus hypersecretion Fibrosis
Eosinophil
FIG. 33.2 Neutrophilic inflammation in asthma. Viruses, such as rhinovirus,
stimulate the release of CXCL8 (IL-8) and CXCL1 (GRO-) from airway FIG. 33.3 T-lymphocytes in asthma. Asthmatic inflammation is
epithelial cells. Allergens activate dendritic cells to release IL-23, which characterized by a preponderance of T helper 2 (Th2) lymphocytes over
recruits helper T-cells that secrete IL-17 (Th17 cells) to release tumor T helper 1 (Th1) cells. Regulatory T-cells (Treg) have an inhibitory effect,
necrosis factor- (TNF-), which amplifies inflammation and CXCL1 and whereas T helper 17 (Th17) cells have a proinflammatory effects. MHC1:
CXCL8, which recruit neutrophils into the airways. Neutrophils release Class 1 major histocompatibility complex; IL: interleukin; IFN-: interferon
more CXCL8 and also transforming growth factor- (TGF-), which gamma; TGF-: transforming growth factor beta; IgE: immunoglobulin E,
activates fibroblasts to cause fibrosis, and neutrophil elastase and matrix Th0: uncommitted T-cell.
metalloproteinase-9 (MMP-9) which stimulate mucus hypersecretion from
goblet cells.

Regulatory T-cells
A subset of CD4 T-cells expressing CD25, termed
in the murine Th2 type of T-lymphocytes, which characteris- T-regulatory cells (Tregs), are capable of suppressing both
tically express IL-4, IL-5, and IL-13 [6]. This programming Th1 and Th2-mediated adaptive immune responses, and
of T-lymphocytes is presumably due to antigen-presenting these cells may mediate their suppressive actions through
cells such as dendritic cells, which may migrate from the epi- release of TGF- or IL-10 [72]. An imbalance between
thelium to regional lymph nodes or which interact with lym- allergen-specic Tregs and eector Th2 cells has been shown
phocytes resident in the airway mucosa. The naive immune in allergic diseases [73]. There is some evidence that early
system is skewed to express the Th2 phenotype; data now infections or exposure to endotoxins might promote Th1-
indicate that children with atopy are more likely to retain mediated responses to predominate and that a lack of infec-
this skewed phenotype than are normal children [63]. There tion or a clean environment in childhood may favor Th2-cell
is also evidence that chitinase, a hydrolase that cleaves envi- expression and thus atopic diseases [74, 75]. Indeed, the bal-
ronmental chitin, an insect protein, plays a critical role in ance between Th1 cells and Th2 cells is thought to be deter-
murine Th2 dierentiation and has been identied in human mined by locally released cytokines, such as IL-12, which tip
tissues from patients with asthma [64, 65]. The recruitment the balance in favor of Th1 cells, or IL-4 or IL-13, which
of Th2 cells in the airway is dependent on several chemotac- favor the emergence of Th2 cells (Fig. 33.3).
tic mediators, including the chemokines CCL17 (TARC)
and CCL22 (MDC) which act on CCR4 that are selectively
expressed on Th2 cells. These CCR4 ligands show increased Th17 cells
expression in airway epithelial cells, macrophages, and mast Another subset of CD4 T-cells, known as Th17 cells, has
cells of asthmatic patients [66]. PGD2, mainly derived from recently been described and shown to have an important
mast cells, is also an important chemoattractant of Th2 cells role in inammatory and autoimmune diseases [76]. Little is
through the activation of DP2-receptors (or CRTH2) [67]. known about the role of Th17 cells in asthma, but increased
There is some evidence that steroid treatment may dieren- concentrations of IL-17A (the predominant product of Th17
tially aect the balance between IL-12 expression and IL-13 cells) have been reported in the sputum of asthma patients
expression [68]. Data from murine models of asthma [6971] [77]. IL-17A and the closely related cytokine IL-17F have
have strongly suggested that IL-13 is both necessary and suf- been linked to neutrophilic inammation by inducing the
cient for induction of the asthmatic phenotype. One of the release of CXCL1 and CXCL8 from airway epithelial
most important areas of asthma research in the next few years cells [78]. As well as IL-17, Th17 cells also produce IL-21,
will be to establish the importance of IL-13 in the induction which is important for the dierentiation of these cells and
of the Th2 phenotype and asthma in humans. thus acts as a positive autoregulatory mechanism, but it also

405
Asthma and COPD: Basic Mechanisms and Clinical Management

inhibits FOXP3 expression and regulatory T-cell devel- Basophils


opment [79, 80]. Another cytokine IL-22 is also released
by these cells and stimulates the production of IL-10 and The role of basophils in asthma is uncertain, as these cells have
acute-phase proteins [81]. However, more work is needed to previously been dicult to detect by immunocytochemistry
understand the role and regulation of Th17 cells in asthma, [89]; indeed there may be multiple phenotypes of basophils
as they may represent important new targets for future with some able to release histamine on IgE challenge and
therapies. others not able to do so [90]. Using a basophil-specic marker,
a small increase in basophils has been documented in the air-
Natural killer T-cells ways of asthmatic patients, with an increased number after
allergen challenge. However, these cells are far outnumbered
A subset of CD4 T-cells termed invariant natural killer
by eosinophils [91].
T (iNKT) cells, which secrete IL-4 and IL-13, has been
shown to account for 60% of all CD4 T-cells in bronchial
biopsies from asthmatic patients [82], but this has been dis-
puted in another study, which failed to show any increase
in iNKT-cell numbers in bronchial biopsies, BAL or spu- STRUCTURAL CELLS AND AIRWAY
tum of either asthma patients or COPD patients [83]. The REMODELING
role of iNKT cells in asthma is currently uncertain as there
appears to be a discrepancy between the data from murine
models of asthma and humans with the disease [84]. Structural cells of the airways, including epithelial cells, brob-
lasts, and even airway smooth muscle cells are the site of
inuence of many of the cytokines, growth factors, and inam-
CD8 cells matory mediators that characterize the asthmatic diathesis.
CD8 (cytotoxic) T-cells are present in patients with more Interestingly they may also be an important source of these
severe disease and irreversible airow obstruction [85] and same molecular entities [92, 93]. Although the concept is
these cells may be of either the TC1 or the TC2 type, releas- poorly dened, the idea that there are long-lasting changes in
ing the same patterns of cytokines as CD4 cells [86]. the cells that make up the airway wall and that these cells nar-
row the lumen and contribute to chronic airow limitation has
been termed airway remodeling. Given the absence of a clean
B-lymphocytes case denition, most investigators agree that certain changes,
such as thickening of the collagen layer deposited below the
B-cells have an important role in asthma through the release true basement membrane, is a critical component of this aspect
of allergen-specic IgE which binds to Fc RI on mast cells of asthma [9498]. Indeed, because structural cells far outnum-
and basophils and to low-anity Fc RII on other inam- ber inammatory cells they may become the major source of
matory cells, including B-cells, macrophages, and possibly mediators driving chronic inammation in asthmatic airways.
eosinophils [87]. The Th2-type cytokines IL-4 and IL-13 For example, epithelial cells may have a key role in translat-
induce B-cells to undergo class switching to produce IgE. ing inhaled environmental signals into an airway inammatory
In both atopic asthma and non-atopic asthma, IgE may be response and are probably a target cell for inhaled glucocorti-
produced locally by B-cells in the airways [88]. coids and anti-leukotrienes (Fig. 33.4) [99].

Mechanical stress Allergens Viruses


Viruses
O2, NO2
Y

Macrophage
FcRII

TNF-, IL-1, IL-6

Airway
epithelial cells

GM-CSF PDGF
CCL11 CCL5 FGF
PDGF
CCL5 IL-16 IGF-1
EGF FIG. 33.4 Airway epithelial cells in asthma. Airway
CCL13 CCL17, CCL22 IL-11
TGF epithelial cells may play an active role in asthmatic
ET inflammation through the release of many inflammatory
mediators, cytokines, chemokines and growth factors.
TNF-: tumor necrosis factor alpha; IL: interleukin,
Eosinophil Lymphocyte Smooth muscle Fibroblast CCL: C-C chemokine, PDGF: platelet-derived growth factor;
survival activation hyperplasia activation EGF: epidermal growth factor; FGF: fibroblast growth factor;
chemotaxis IGF: insulin-like growth factor.

406
Pathophysiology of Asthma 33
Structural changes in the airways may account for other inammatory cells. Because each mediator has many
the accelerated decline in airway function seen in asthmatic eects in the role of individual mediators in the pathophysi-
patients over several years [100, 101]. Structural changes in ology of asthma is not yet clear. Although the multiplicity of
asthmatic airways include mediators makes it unlikely that preventing the synthesis or
action of a single mediator will have a major impact in clinical
increased thickness of airway smooth muscle; asthma, recent clinical studies with anti-leukotrienes suggest
inltration of the airway smooth muscle with mast cells that cysteinyl leukotrienes have a clinically important eect.
brosis (which is predominantly subepithelial);
increased mucus-secreting cells; Leukotrienes
increased numbers of blood vessels (angiogenesis).
The cysteinyl leukotrienes LTC4, LTD4, and LTE4 are potent
It is not known if these changes are reversible with constrictors of human airways and have been reported to
therapy. These changes may occur in some patients to a greater increase AHR and may play an important role in asthma
extent than others and may be increased by other factors such [107110] (see Chapter 24). The recent development of
as concomitant cigarette smoking. It is likely that genetic fac- potent specic leukotriene antagonists has made it possible to
tors will inuence the extent of remodeling that occurs in indi- evaluate the role of these mediators in asthma. Potent LTD4
vidual patients. antagonists protect (by about 50%) against exercise- and
There are good data to support the idea that mechani- allergen-induced bronchoconstriction [111114], suggesting
cal deformation of the airway epithelium as a result of airway that leukotrienes contribute to bronchoconstrictor responses.
smooth muscle constriction results in the creation of a micro- Combined treatment with an anti-histamine and an anti-
environment that promotes airway remodeling. Deformed air- leukotriene is particularly eective [115, 116] Chronic treat-
way epithelial cells activate signaling via the epidermal growth ment with anti-leukotrienes improves lung function and
factor receptor (EGFR) and the urokinase plasminogen acti- symptoms in asthmatic patients, although the degree of lung
vator system. Experimental data show that soluble signals function improvement is not as great as that seen with an
generated by airway epithelial cells can inuence the pheno- inhaled corticosteroid. It is only through the use of specic
type of broblasts to produce collagen similar to that found in antagonists that the role of individual mediators of asthma
remodeled airways [102104]. These data could help explain may be dened. In the future, pharmaceuticals with specic
why combination treatments with inhaled steroids and long- targets of action will be of special value in providing patho-
acting -agonists are eective asthma therapies. biological insights into the basic mechanisms of asthma;
their potential role in the treatment of asthma remains to be
determined.

INFLAMMATORY MEDIATORS Platelet-activating factor


Many dierent mediators have been implicated in asthma. Platelet-activating factor (PAF) and PAF-acetylhydrolase,
They may have a variety of eects on the airways, which could the enzyme that degrades PAF, constitute a potent inam-
account for the pathological features of asthma (Fig. 33.5) matory mediator system that mimics many of the features
[105, 106]. Mediators such as histamine, prostaglandins, and of asthma, including eosinophil recruitment and activation
leukotrienes contract airway smooth muscle, increase micro- and induction of AHR; yet even potent PAF antagonists,
vascular leakage, increase airway mucus secretion, and attract such as modipafant, do not control asthma symptoms, at
least in chronic asthma [117120]. However, genetic stud-
ies in Japan, where there is a high frequency of a genetic
mutation which disables the PAF metabolizing enzyme,
Inflammatory cells
Mediators
PAF-acetylhydrolase, have shown that there is an association
Mast cells between the presence of the mutant form of the enzyme and
Eosinophils Histamine
Leukotrienes severe asthma [121, 122]. In addition, there are data showing
Th2 cells
Basophils Prostanoids Effects that patients with a deciency of the enzyme that degrades
Neutrophils PAF Bronchospasm PAF is associated with enhanced asthma severity [123].
Platelets Kinins Plasma exudation These data suggest that there may be certain conditions
Adenosine Mucus secretion
Structural cells associated with a signicant role for PAF in asthma.
Endothelins AHR
Epithelial cells Nitric oxide Structural changes
Sm muscle cells Cytokines
Endothelial cells
Fibroblasts
Chemokines Cytokines
Growth factors
Nerves
Cytokines are increasingly recognized to be important in
FIG. 33.5 Multiple cells, mediators and effects. Many cells and mediators chronic inammation and play a critical role in orchestrating
are involved in asthma and lead to several effects on the airways. Th2: the type of inammatory response (see Chapter 27) (Fig. 33.6)
T helper 2 cells; Sm: smooth; PAF: platelet-activating factor; AHR: airway [124]. Many inammatory cells (macrophages, mast cells,
hyperresponsiveness. eosinophils, and lymphocytes) are capable of synthesizing

407
Asthma and COPD: Basic Mechanisms and Clinical Management

Allergen

Epithelial cells GM-CSF, IL-6, IL-11, IL-16


CCL11, CCL5, CXCL8, CCL17
IL-1
TNF-
Dendritic cell
Smooth muscle
Macrophage IL-12, IL-18
Th0 cell

IL-10
CCL1 IL-4
IL-13

GM-CSF GM-CSF
Monocyte Th2 cell CCL5, CCL11
IL-3
IL-9 IL-4 IL-5
FIG. 33.6 The cytokine network in
IL-13
asthma. Many inflammatory cytokines
TNF- are released from inflammatory and
IL-4 IL5, GM-CSI structural cells in the airway and
Y

IL-5 IgE orchestrate and perpetuate the


Mast cell B-Iymphocyte Eosinophil inflammatory response.

and releasing these proteins, and structural cells such as epi- patients was associated with a decrease in eosinophil counts
thelial cells and endothelial cells may also release a variety of in the blood and BAL uid [49]. Interestingly in this study
cytokines and may therefore participate in the chronic inam- there was no eect on the physiology of the allergen-
matory response [125, 126]. There are number of major classes induced asthmatic response; although this is not the last
of cytokines. One class includes T-cell derived cytokines, word on eosinophils in asthma, it provides evidence that the
examples of which include interferon (IFN) and TNF eosinophil, as recruited by IL-5, is not the major pathoge-
from Th1 cells and IL-4 and IL-5 from Th2 cells. Another netic cell in asthma. This has been conrmed by a study of
class includes pro- and anti-inammatory cytokines with IL- the same antibody in symptomatic asthmatic patients, who
1 and IL-10 being respective examples. Chemoattractant showed no improvement in asthma control despite a pro-
cytokines, such as CCL11 and CXCL8 (IL-8), and growth found reduction in circulating eosinophils [137]. Another
factors cytokines, including PDGF, TGF-, and VEGF are Th2 cytokine produced by peripheral blood cells of patients
other major classes. It is now clear that cytokines play a domi- with asthma, IL-9, may play a critical role in airway remod-
nant role in chronic inammation of asthma. eling, eosinophilopoiesis, and may interact with the IL-4
T-cell-derived cytokines play a key role in creating the receptor in mediating some of the pathobiological features
microenvironment that lends itself to initiating and perpet- of asthma [138141].
uating the chronic inammatory response of asthma [127]. Other cytokines, such as IL-1, IL-6, TNF-, TGF-
The key cytokines here are Th2 cytokines, IL-3, IL-4, and , and GM-CSF, are released from a variety of cells, includ-
IL-5. IL-3 induces eosinophilia in vivo and maintains mast ing macrophages and epithelial cells and may be important
cell viability. IL-4 induces dierentiation of Th2 cells and in amplifying the inammatory response. TNF- may be an
creates the microenvironment in which IgE antibody is syn- amplifying mediator in asthma and is produced in increased
thesized [128] and enhances mucin expression in goblet cells amounts in asthmatic airways. Inhalation of TNF- increased
[129]. IL-5 enhances eosinophil maturation and prevents airway responsiveness in normal individuals [142]. TNF-
apoptosis of inammatory cells [130]. The recently described and IL-1 both activate the proinammatory transcription
IL-17 family of cytokines, produced by cells as a result of the factors, nuclear factor-B (NF-B) and activator protein-1
action of IL-15 or IL-23 on CD4 or CD8 memory cells may (AP-1) which then switch on many inammatory genes in
be important regulators of the asthmatic response [131133]. the asthmatic airway [143]. TGF-, through ligation of two
A lack of specic small molecule antagonists has made receptors, type I and II, has been linked to tissue remodeling,
cytokine related research in humans dicult. Although and potentially to subepithelial brosis through the Smad-
important observations have been made using specic neu- signaling pathway which could be a site for therapeutic inter-
tralizing antibodies [134], interventions involving immune vention [144146].
function can be dicult. Thymic stromal lymphopoeitin (TSLP) is a cytokine
There is increased gene expression of IL-5 in lym- that shows a marked increase in expression in airway epi-
phocytes in bronchial biopsies of patients with sympto- thelium and mast cells of asthmatic patients [66]. TSLP
matic asthma [135, 136]. The role of IL-5 in eosinophil appears to play a key role in programming airway dendritic
recruitment in asthma has been conrmed in a study in cells to release CCL17 and CCL22 to attract Th2 cells (Fig.
which administration of an anti-IL-5 antibody to asthmatic 33.7) [147]. It therefore acts as an upstream cytokine which

408
Pathophysiology of Asthma 33
peroxidation of their lipid membranes. Phagocytes use heme
peroxidases (myeloperoxidase, MPO) or eosinophil peroxi-
Epithelial cells Mast cell dase (EPO) to produce ROS; these moieties can also inter-

Y
act with nitrite (NO2) and hydrogen peroxide (H2O2)
TSLP to promote formation of RNS. Eosinophils, a key cell in
asthma, have several times greater capacity to generate O2
Myeloid dendritic cell
and H2O2 than do neutrophils, and the content of EPO in
eosinophils is 310 times higher than the amount of MPO
present in neutrophils [153155].
CCL17 Evidence for increased oxidative stress in asthma is
provided by the increased concentrations of 8-isoprostane
CCR4 (a product of oxidized arachidonic acid) in exhaled breath
Th2 cell
condensates [156], and increased ethane (a product of oxi-
dative lipoid peroxidation) in exhaled breath of asthmatic
patients [156158]. Airow obstruction, airway hyperre-
IL-5 IL-4, IL-13 sponsiveness and airway remodeling have been associated
with impaired SOD activity [159, 160]. Oxidative stress is
IgE
increased particularly in patients with severe asthma (perhaps
as a result of the neutrophilic inltration in these patients)
Eosinophil B-lymphocyte
and this may reduce the activity of histone deacetylase
FIG. 33.7 Thymic stromal lymphopoetin in asthma. TSLP is an upstream (HDAC), which may account for the reduced response to
cytokine produced by airway epithelial cells and mast cells in asthma that corticosteroids in these patients [161, 162].
acts on immature dendritic cells to mature and release CCL17 (TARC),
which attracts Th2 cells via CCR4. Th2: T helper 2; CCL: chemokine; CCR:
chemokine receptor. Endothelins
Endothelins are potent peptide mediators that are vasocon-
may play a key role in orchestrating an allergic pattern of strictors, bronchoconstrictors and promote brosis [163,
inammation in asthmatic airways. 164]. Endothelin-1 levels are increased in the sputum, serum,
and BAL uid of patients with asthma; these levels are mod-
ulated by allergen exposure and steroid treatment [165167].
Complement Endothelins also induce airway smooth muscle cell prolifera-
tion and promote a probrotic phenotype and may therefore
A decade ago it was postulated that complement had little play a role in the chronic inammation of asthma [164, 168].
role in the biology of asthma. However, studies in mice har-
boring a targeted deletion of the C5a receptor showed dimin-
ished bronchial hyperresponsiveness induced after allergen Nitric oxide
challenge [148]. In addition, genetic linkage studies in mice
have linked regions of the mouse genome containing the Nitric oxide (NO) is produced by several cells in the airway
gene for C5a to the phenotype of AHR [149]. Furthermore, by NO synthases [169, 170] (see Chapter 30). Although the
the complement peptide C5a has been recovered from BAL cellular source of NO within the lung is not known, infer-
uid of patients after allergen challenge [148]. Current think- ences based on mathematical models suggest that it is the
ing is that complement contributes to the airway inam- large airways which are the source of NO [171]. However, in
mation, AHR and mucus production observed in human patients with severe asthma NO is also derived form periph-
asthma and may also prevent the formation of a Th2-directed eral airways [172]. Current data indicate that the level of
immune response to allergens [150152]. NO in the exhaled air of patients with asthma is higher than
the level of NO in the exhaled air of normal subjects (see
Chapter 30) [173177]. The elevated levels of NO in asthma
Oxidative stress are closely linked to airway pH [178180] and thus are more
likely reective of inammatory mechanisms than of a direct
In asthma, activation of epithelial cells, and resident macro- pathogenetic role of this gas in asthma [181, 182]. The com-
phages, and the recruitment and activation of neutrophils, bination of increased oxidative stress and NO may lead to
eosinophils, monocytes, and lymphocytes leads to the gen- the formation of the potent radical peroxynitrite that may
eration of reactive oxygen species (ROS) and reactive nitro- result in nitrosylation of proteins in the airways [183, 184].
gen species (RNS). Action of many systems but primarily Asthmatics managed by measurements of exhaled nitric
NADPH oxidase results in the microenvironmental availabil- oxide achieved equivalent asthma control with less inhaled
ity of superoxide anions (O2), which are rapidly converted corticosteroids than those managed using conventional algo-
to H2O2 through the catalytic action of superoxide dismutase rithms [185]; the availability of portable devices to measure
(SOD). Hydroxyl ion (OH) is then formed nonenzymati- exhaled nitric oxide may allow management of asthma in
cally; this damages tissues, particularly cell membranes by manner similar to the self management of diabetics [186].

409
Asthma and COPD: Basic Mechanisms and Clinical Management

inammatory cells, and activated complement. Epithelial


EFFECTS OF INFLAMMATION ON cells shed from the airway wall may be found in clumps in
THE AIRWAYS the BAL uid or sputum (Creola bodies) of asthmatics, sug-
gesting that there has been a loss of attachment to the basal
The idea that the chronic inammatory response results in layer or basement membrane. The damaged epithelium may
the characteristic pathophysiological changes associated contribute to AHR in a number of ways, including
with asthma has been widely promulgated. However, biop- loss of its barrier function to allow penetration of
sies from children with wheezing show many of the charac- allergens and irritants;
teristic features of asthma [12]. Thus it is probable that some
loss of enzymes (such as neutral endopeptidase) which
of the dierences between the condition of the airways in
normally degrade inammatory mediators such as kinins;
normal and asthmatic subjects reects the genetic makeup
of the patient inuenced by environmental factors such as loss of a relaxant factor (so-called epithelial-derived
airway infections and allergen exposures. The descriptions relaxant factor);
below present the observations without adequate evidence edema and swelling that promotes obstruction by
to support whether changes are markers for or the result of a lumenal encroachment;
chronic inammatory response. The inammatory response
has many eects on structural cells of the airways result- loss of surfactant function leading to increased increased
ing an amplication and perpetuation of the inammatory recoil of the airway;
response (Fig. 33.8). exposure of sensory nerves which may lead to reex
neural eects on the airway.

Airway epithelium
Fibrosis
Airway epithelial cells play a critical role in asthma and
link environmental inuences to inammatory responses in The basement membrane in asthma appears on light micros-
the airway (Fig. 33.4). Airway epithelial shedding may be copy to be thickened; on closer inspection by electron micro-
important in contributing to AHR and may explain how scopy it has been demonstrated that this apparent thickening
several dierent mechanisms, such as ozone exposure, certain is due to subepithelial brosis with deposition of types III
virus infections, chemical sensitizers and allergen exposure and V collagen below the true basement membrane; however,
can lead to its development, since all these stimuli may lead the ratio of interstitial collagen brils to matrix is not dif-
to epithelial disruption. Epithelium may be shed because of ferent from that found in normal subjects [187189]. Data
an inammatory milieu made up of eosinophil basic pro- from a number of investigative groups show that the thick-
teins, oxygen-derived free radicals, proteases released from ness of the deposited collagen is related to airway obstruction

Allergen

Macrophage/
dendritic cell Mast cell

Th2 cell
Neutrophil

Eosinophil
Mucus plug
Epithelial compression and loss
Nerve activation

Subepithelial
fibrosis
Myofibroblast
Plasma leak Sensory nerve
Edema activation
Mucus
Cholinergic
hypersecretion Vasodilatation
reflex
hyperplasia New vessels
Bronchoconstriction
Hypertrophy/hyperplasia

FIG. 33.8 The complexity of asthma. The pathophysiology of asthma is complex, with participation of several interacting inflammatory cells which result in
acute and chronic inflammatory effects on the airway.

410
Pathophysiology of Asthma 33
and airway responsiveness [190193]. The mechanism of smooth muscle modulates the remodeling process by secret-
the collagen deposition is not known. However, it is known ing multiple cytokines, growth factors, or matrix proteins
that several probrotic cytokines, including TGF- and and by expressing cell adhesion molecules and other poten-
PDGF, and mediators such as endothelin-1, can be pro- tial costimulatory molecules [196]. For example, increased
duced by epithelial cells or macrophages in the inamed numbers of mast cells have been identied in airway smooth
airway [126]. Even simple mechanical manipulations can muscle of patients with asthma [15, 198].
alter the phenotype of airway epithelial cells in a probrotic The importance of airway smooth muscle in chronic
fashion [102104, 194]. The role of airway brosis in asthma asthma was highlighted by a recent trial in which airway
is unclear, as subepithelial brosis has been observed even smooth muscle was rendered dysfunctional by bronchial
in mild asthmatics at the onset of disease; it is not certain thermoplasty. Although this intervention was limited to rel-
whether the collagen deposition has any functional conse- atively large airways there was a measurable, although not
quences. Subepitheial brosis is also seen in patients with overwhelming, clinical benet [203]. The idea of manipu-
chronic eosinophilic bronchitis, who usually present with lating the mass or contractile capacity of airway smooth
cough and have an inltration of eosinophils in the airways muscle in asthma remains an important and unexplored
but no AHR or airway obstruction [195]. This suggests that avenue of asthma therapeutics [199, 204].
the subepithelial brosis seen in asthmatics is likely to be
secondary to eosinophilic inammation in the airways and
may not have important functional consequences. Vascular responses
It has been known for decades that the subepithelial con-
Airway smooth muscle nective tissue of the asthmatic airway has many more blood
vessels than are found in similar locations in normal subjects
When the concept that asthma was primarily an inamma- [205]. It is now recognized that bronchial vessels play a key
tory disease was developed in the late 1980s to early 1990s, role in the pathophysiology of asthma (Fig. 33.10). Despite
airway smooth muscle was thought to be predominantly a this anatomic knowledge, little is known about the role of
bystander tissue. That is the inammatory process resulted the bronchial circulation in asthma. This is partly because
in the availability of mediators that led to smooth muscle of the diculties involved in measuring airway blood ow.
constriction and expression of an asthma phenotype. There Recent studies using an inhaled absorbable gas have dem-
are now substantial data to support the concept that airway onstrated increased airway mucosal blood ow in asthma
smooth muscle in asthma is phenotypically dierent from [206208].
normal airway smooth muscle and that these changes reect The bronchial circulation may also play an important
the impact of the inammatory asthmatic airway microenvi- role in regulating airway caliber, since an increase in the vas-
ronment on smooth muscle [15, 196201]. Although some cular volume may contribute to airway narrowing. Vascular
of the abnormalities identied to date in asthma impact on blanching of the skin is used to measure the potency of
the contractility of individual units of airway smooth muscle, topical steroids and a similar mechanism may be involved
the more recently identied ones reect dierences in the with the eects of inhaled steroids in asthma [207, 209].
ability of this tissue to proliferate or to function as an endo- Increased airway blood ow may be important in removing
crine tissue (Fig. 33.9). For example, airway smooth muscle inammatory mediators from the airway, and may play a
may increase in mass because of an increased tendency to role in the development of exercise-induced asthma [210].
proliferate due to the absence of an inhibitory transcription Increased shear stress due to high expiratory pressures may
factor C/EBP- [197]. lead to gene transduction and enhanced production of nitric
Airway smooth muscle is not only inuenced by oxide by type III (endothelial) NO synthase [211, 212];
the inammatory mediators and cytokines in which it is there are in vivo experimental data in sheep that support the
immersed [202], but also recent studies suggest that airway importance of this mechanism [213]. There may also be an

Inflammatory cells

Mediators

Airway smooth muscle

FIG. 33.9 Airway smooth muscle in asthma.


Inflammation has several effects on airway smooth
muscle cells, resulting in contraction, proliferation and
Contraction Proliferation Secretion secretion of inflammatory mediators. Cys: Cysteinyl;
Histamine, cys -leukotrienes PDGF, EGF, cytokines, chemokines, PDGF: platelet-derived growth factor; EGF: epidermal
kinins, prostanoids, endothelin endothelin-1 prostanoids growth factor.

411
Asthma and COPD: Basic Mechanisms and Clinical Management

increase in the number of blood vessels in asthmatic airways Mucus hypersecretion


as a result of angiogenesis in response to VEGF [214216].
Microvascular leakage is an essential component of Mucus hypersecretion is a common inammatory response
the inammatory response and many of the inammatory in secretory tissues and is an important component of the
mediators implicated in asthma produce this leakage [217, inammatory response in asthma (Fig. 33.11). Increased
218]. There is a good evidence for microvascular leakage mucus secretion contributes to the viscid mucus plugs
in asthma, and it may have several consequences on airway which occlude asthmatic airways, particularly in fatal
function, including increased airway secretions, impaired asthma. There is evidence for hyperplasia of submucosal
mucociliary clearance, formation of new mediators from glands which are conned to large airways, and of increased
plasma precursors (such as kinins), and mucosal edema numbers of epithelial goblet cells in patients with asthma,
which may contribute to airway narrowing and increased especially those with severe asthma [8]. One of the major
AHR [219221]. mucin genes, MUC5AC is induced by complement factor
C3a [222], NO [223], TNF- [224], EGF family ligands
[225], or neutrophil elastase [226]. Since both neutrophils
and enhanced expression of mucin genes have been found
Inflammatory in great profusion in the airways of patients with severe
mediators asthma [52], it is tempting to speculate that the neutrophils
may have led to the enhanced mucin gene expression. Thus
enhanced mucin expression in epithelial cells can be viewed
as an indicator of the inammatory state of the airways.
These ndings are not limited to cell culture, since in intact
humans with asthma, exercise-induced bronchospasm is
associated with enhanced expression of MUC5AC [227].
It is highly likely that methods to manipulate mucin gene
expression in response to inammatory stimuli will be iden-
tied and could be of value in the treatment of asthma as
Vasodilatation Plasma exudation Angiogenesis well as COPD.
NO, CGRP histamine, cys-leukotrienes VEGF, TNF-
histamine, PGE2 kinins, PAF

FIG. 33.10 Vascular abnormalities in asthma. Allergic inflammation has


Neural effects
several vascular effects, including vasodilatation, plasma exudation from
post-capillary venules and new vessel formation (angiogenesis). NO: Nitric Neural mechanisms play an important role in the patho-
oxide; CGRP: calcitonin gene-related peptide; PGE2: prostaglandin E2; PAF: genesis in asthma as discussed in Chapter 32 [228].
platelet-activating factor; VEGF: vascular-endothelial growth factor; TNF-: Autonomic nervous control of the airways is complex; in
tumor necrosis factor-alpha. addition to classical cholinergic and adrenergic mechanisms,

Neutrophil Th2 cell


Oxidative stress
IL-4, IL-13, IL-9

Neutrophil
elastase

EGFR

MUC5AC

Goblet cells

Goblet cells Mucus hyperplasia

Mucus hypersecretion
Submucosal gland

FIG. 33.11 Mucus hypersecretion in asthma. Increased mucus secretion in asthma may be stimulated by neutrophils through the release of neutrophil
elastase, T helper 2 (Th2) cytokines and oxidative stress. This may be mediated via epidermal growth factor receptors (EGFR) which may result in mucus
hyperplasia and increased expression of the mucin gene MUC5AC. Mucus hypersecretion is also enhanced by neural mechanisms through the release of
acetylcholine (ACh) and substance P (SP).

412
Pathophysiology of Asthma 33
nonadrenergic noncholinergic (NANC) nerves and several related peptide may be released from sensitized inamma-
neuropeptides have been identied in the respiratory tract tory nerves in the airways which increase and extend the
[229231]. Many studies have investigated the possibility ongoing inammatory response [237]. There is evidence
that defects in autonomic control may contribute to AHR for an increase in SP-immunoreactive nerves in airways
in asthma, and abnormalities of autonomic function, such as of patients with severe asthma [238], which may be due
enhanced cholinergic and -adrenergic responses or reduced to proliferation of sensory nerves and increased synthesis
-adrenergic responses, have been proposed. Current think- of sensory neuropeptides as a result of nerve growth fac-
ing suggests that these abnormalities are likely to be second- tors released during chronic inammation although this
ary to the disease, rather than primary defects [228]. It is has not been conrmed in milder asthmatic patients [239].
possible that airway inammation may interact with auto- There may also be a reduction in the activity of enzymes,
nomic control by several mechanisms. such as neutral endopeptidase, which degrade neuropeptides
There is a close interaction between airway inamma- such as SP [240]. Thus chronic asthma may be associated
tion and airway nerves (Fig. 33.12). Inammatory media- with increased neurogenic inammation, which may provide
tors may act on various prejunctional receptors on a mechanism for perpetuating the inammatory response
airway nerves to modulate the release of neurotransmitters even in the absence of initiating inammatory stimuli.
[232, 233]. Thus thromboxane and PGD2 facilitate the
release of acetylcholine from cholinergic nerves in canine
airways, whereas histamine inhibits cholinergic neurotrans-
mission at both parasympathetic ganglia and postganglionic TRANSCRIPTION FACTORS
nerves via histamine H3-receptors. Inammatory media-
tors may also activate sensory nerves, resulting in reex
cholinergic bronchoconstriction or release of inammatory The chronic inammation of asthma is due to increased
neuropeptides. Inammatory products may also sensitize expression of multiple inammatory proteins (cytokines,
sensory nerve endings in the airway epithelium, so that the enzymes, receptors, adhesion molecules). In many cases
nerves become hyperalgesic. Hyperalgesia and pain (dolor) these inammatory proteins are induced by transcription
are cardinal signs of inammation, and in the asthmatic air- factors, DNA-binding factors that increase the transcrip-
way may mediate cough and chest tightness, which are such tion of selected target genes (Fig. 33.13) [143].
characteristic symptoms of asthma. The precise mechanisms A proinammatory transcription factor that may play
of hyperalgesia are not yet certain, but mediators such as a critical role in asthma is nuclear factor-kappa B (NF-B)
prostaglandins, certain cytokines, and neurotrophins may which can be activated by multiple stimuli, including protein
be important. Neurotrophins, which may be released from kinase C activators, oxidants, and proinammatory cytokines
various cell types in peripheral tissues, may cause prolifera- (such as IL-1 and TNF-) [241243]. There is evidence
tion and sensitization of airway sensory nerves [234]. for increased activation of NF-B in asthmatic airways, par-
Bronchodilator nerves which are nonadrenergic are ticularly in epithelial cells and macrophages [244]. Although
prominent in human airways and may be dysfunctional in many aspects of the asthmatic airway are inuenced by the
asthma [235]. In human airways, the bronchodilator neuro- expression of transcription factors, there are data linking
transmitter appears to be NO [236]. the mechanical deformation of the airway to expression of
Airway nerves may also release neurotransmitters proinammatory transcription factors; these ndings have
which have proinammatory eects. Thus neuropeptides direct implications for the mechanism of exercise-induced
such as substance P (SP), neurokinin A, and calcitonin-gene asthma and may explain how this physical stimulus could

Inflammatory mediators
Neutrotrophins

Airway nerves
Inflammatory
cells

Neurotransmitters
Neuropeptides

Neurogenic inflammation

FIG. 33.12 Neural mechanism in asthma. There is a close interaction between asthmatic inflammation and neural mechanisms.

413
Asthma and COPD: Basic Mechanisms and Clinical Management

Inflammatory Inflammatory
stimuli proteins
allergens, viruses, cytokines, enzymes, receptors,
cytokines adhesion molecules

Receptor

Kinases Inactive
transcription
factor
mRNA

Activated
transcription factor
P (NF-B, AP-1, STATs)
Nucleus

Inflammatory Promoter Coding region


gene

FIG. 33.13 Proinflammatory transcription factors in asthma. Transcription factors play a key role in amplifying and perpetuating the inflammatory response
in asthma. Transcription factors, including nuclear factor kappa-B (NF-B), activator protein-1 (AP-1), and signal transduction-activated transcription factors
(STATs), are activated by inflammatory stimuli and increase the expression of multiple inflammatory genes.

IL-27 IL-12 IL-4 IL-33 naive T-cells into Th2 cells and it regulates the secretion
of TH2-type cytokines [248]. There is an increase in the
number of GATA-3 T-cells in the airways of asthmatic
subjects compared with normal subjects [249]. Following
STAT1 STAT4 STAT6
simultaneous ligation of the T-cell receptor (TCR) and
co-receptor CD28 by antigen-presenting cells, GATA-3
is phosphorylated and activated by p38 mitogen-activated
Th1 cells T-bet GATA-3 Th2 cells
protein kinase. Activated GATA-3 then translocates from
the cytoplasm to the nucleus, where it activates gene tran-
scription [250]. GATA-3 expression in T-cells is regulated
by the transcription factor STAT-6 (signal transducer and
Th1 cytokines Th2 cytokines
activator of transcription 6) via IL-4 receptor activation.
(IL-2, IFN-) (IL-4, IL-5, IL-9, IL-13) Another transcription factor T-bet has the opposite
eect and promotes Th1 cells and suppresses Th2 cells.
Allergic inflammation T-bet expression is reduced in T-cells from the airways of
asthmatic patients compared with nonasthmatic patients
FIG. 33.14 GATA-3 in asthma. The transcription factor GATA-3 (GATA- [251]. When phosphorylated, T-bet can associate with
binding protein-3) is regulated by interleukin-4 (IL-4) via STAT-6 (signal and inhibit the function of GATA-3, by preventing it
transducer and activator of transcription 6) and regulates the expression from binding to its DNA target sequences [252]. GATA-
of IL-4, IL-5, IL-9, and IL-13 from T helper 2 (Th2) cells and inhibits the 3 inhibits the production of Th1-type cytokines by inhibit-
expression of T-bet via inhibition of STAT4. IL-33 enhances the actions of ing STAT-4, the major transcription factor activated by the
GATA-3. T-bet regulates TH1-cell secretion of IL-2 and interferon- (IFN-) T-bet-inducing cytokine IL-12 [253] (Fig. 33.14). Nuclear
and has an inhibitory action on GATA-3. T-bet is regulated by IL-12 via
factor of activated T-cells (NFAT) is a T-cell-specic tran-
STAT4 and by IL-27 via STAT1. This demonstrates the complex interplay of
cytokines and transcription factors in asthma.
scription factor and appears to enhance the transcriptional
activation of GATA-3 at the IL-4 promoter [254]. FOXP3
is a transcription factor that is expressed in Tregs, whereas
modify the inammatory microenvironment of the airway the equivalent transcription factor in Th17 cells is RORt.
[104, 245247]. There appears to be a negative interaction between these
The transcription factor GATA-3 (GATA-binding transcription factors so that a reduction of Tregs in asthma
protein 3) is crucial for the dierentiation of uncommitted may be a factor increasing Th17 cells [255].

414
Pathophysiology of Asthma 33
exhaled breath condensates from healthy subjects but levels
Anti- were signicantly lower in condensates from patients with
inflammatory asthma exacerbations [266]. PD1 when administered before
mediators inhaled allergen challenge, in mice led to decreased airway
IL-10, IL-1ra eosinophil and T-lymphocyte recruitment, decreased airway
IFN-, IL-12, IL-18
Lipoxins, resolvins mucus, and less of an increment in AHR than without the
Inflammatory Protectins PD1. These data suggest that endogenous PD1 is a novel
mediators PGE2 and important counterregulatory signal in allergic airway
Lipid mediators inammation.
Cytokines
Peptides
Oxidants
IL-10
Airway and alveolar macrophages have a predominantly
suppressive eect in asthma and inhibit T-cell proliferation.
The mechanism of macrophage-induced immunosuppression
is not yet certain, but PGE2 and IL-10 secretion may con-
tribute [267]. IL-10 inhibits many of the inammatory
mechanisms in asthma and its secretion is defective from
macrophages of asthmatic patients [268]. IL-10 is secreted
by a subset of Tregs and this is enhanced by specic allergen
FIG. 33.15 Anti-inflammatory mediators in asthma. There may be an
imbalance between increased proinflammatory mediators and a deficiency
immunotherapy [267]. Interestingly treatment of asthmatic
in anti-inflammatory mediators. IL: interleukin; IL-1ra: interleukin-1 receptor children with an inhaled corticosteroid or with montelukast
antagonist; IFN-: interferon gamma; PGE2: prostaglandin E2. increases serum levels of IL-10 [269]; these ndings sug-
gest that increasing IL-10 may be an important therapeutic
approach in asthma.

ANTI-INFLAMMATORY MECHANISMS IN
ASTHMA GENETIC INFLUENCES
Although most emphasis has been placed on inammatory Over the past 15 years there have been multiple studies on
mechanisms, there may be important anti-inammatory the genetics of asthma (see Chapter 4). A number of poten-
mechanisms that may be defective in asthma, resulting in tial asthma genes have been identied [270], but very few
increased inammatory responses in the airways (Fig. 33.15). have been replicated in multiple populations [271]. However
all the data suggest that there is not a single major asthma
susceptibility gene, but rather there are multiple genes each
Lipid anti-inflammatory mediators explaining only a small proportion of the total variance in
the asthma phenotype. The keys to nd the most important
Several lipid mediators appear to have anti-inammatory asthma genes will be to use comprehensive methods and to
eects, including PGE2, lipoxins, resolvins, and protectins replicate the ndings in an individual population in multiple
[256, 257]. There is a strong evidence supporting the role of distinct populations.
signaling through the lipoxin A receptor as being decient ADAM33 is an asthma gene, discovered by positional
in severe asthma in general and aspirin-sensitive asthma in cloning, that has been replicated in many but not all popu-
particular [258263]. In addition it has now been estab- lations tested [272]. It was initially identied in Caucasian
lished that presqualene diphosphate (PSDP) is an endog- families from the United States and United Kingdom and
enous regulator of phosphatidylinositol 3-kinase (PI3K). was associated with asthma characterized by bronchial hyper-
PSDP, but not presqualene monophosphate (PSMP), responsiveness. There were many variants within ADAM33
directly inhibits recombinant human PI3K- activity and that were signicantly associated with these phenotypes.
a new PSDP structural mimetic blocks human neutrophil Subsequently, there have been a large number of case-control
activation and murine lung PI3K activity and inamma- and family-based association studies focused on ADAM33,
tion [264]. Resolvin E1 (RvE1: 5S,12R,18R-trihydroxyei with the majority but not all, conrming the original nding
cosapentaenoic acid) is an anti-inammatory lipid media- [273]. Current data suggest that ADAM33 may be involved
tor derived from the -3 fatty acid eicosapentaenoic acid. in lung development and remodeling.
RvE1 is involved in the resolution of inammation and In genome wide-association studies microarrays capa-
reduces allergic inammation in mice exposed to inhaled ble of typing 500,000 to 1 million single nucleotide poly-
allergen [265]. morphisms are used to identify associations between a given
Another important anti-inammatory mediator is phenotype and genotype. This technology has been applied
protectin D1, a natural lipid based chemical mediator that to childhood asthma [274]. These investigators identi-
counters leukocyte activation. PD1 has been recovered from ed a strong statistical association between the diagnosis of

415
Asthma and COPD: Basic Mechanisms and Clinical Management

asthma in childhood in families from the United Kingdom central heating providing more favorable environment), cig-
and markers located on chromosome 17q21. The results were arette smoking (pregnancy and early childhood exposure),
replicated in two European populations. They then used the and possibly air pollution due to road trac. The role of
genetic data to examine transcript levels in immortalized lym- exposures to pets in early childhood remains unresolved and
phocytes from children harboring the implicated variants and because of confounding will be dicult to ascertain using an
showed a strong association between variants in ORMDL3 epidemiological approach.
and asthma. ORMDL3 is a member of a family of genes that
encode endoplasmic reticulum proteins of unknown function. Why does asthma once established become chronic in some indi-
If these studies are replicated, then the community of asthma viduals but remain intermittent and episodic in others?
researchers will need to determine how variants in the func- Is there a genetic dierence among people or an environ-
tion of this gene could lead to childhood asthma. mental dierence among stimuli that results in full resolu-
An area of importance is the relationship between tion of airway obstruction in some patients and the failure
clinical responses to anti-asthma treatments and genotype for this to occur in others? For example, occupational asthma
at various drug targets [275]. For example, there are known due to chemical sensitizers, such as toluene diisocyanate,
to be a number of SNPs in the 2-adrenoceptor gene that may remit if the patient is removed from exposure to the
result in structural changes in the 2-receptor structure asso- sensitizer within 6 months of development of asthma symp-
ciated with functional changes in isolated cell systems [276]. toms, whereas longer exposure is often associated with per-
These are related to the acute response to bronchodilator sistent asthma even when avoidance of exposure is complete
aerosols acting via this receptor [277, 278] and with del- [289, 290]. This suggests that once inammation is estab-
eterious eects of chronic use of albuterol [279, 280]. In a lished it may continue independently of a causal mechanism,
double blind randomized controlled trial, the rst in a non- whereas in others this self perpetuation does not occur.
malignant condition where genotype at a specic locus was
an entry criterion, those patients harboring the alleles encod- Are there dierent types of asthma, each characterized by a com-
ing for homozygous expression of arginine rather than gly- mon genetic background with its attendant immunological and
cine at the 16th amino acid in the 2-adrenergic receptor inammatory response?
had diminished morning and evening peak ow and lower Asthma remains a syndromic diagnosis. How can we get
FEV1 after the regularly scheduled use of albuterol than beyond a physiological and descriptive denition to one in
when albuterol was used on an as needed only basis [281]. which the primary disease pathobiology, for each of the many
In a retrospective analysis of two other randomized clinical asthma syndromes, can be developed?
trials similar changes have been observed by some groups
[282] but not by others [283]. However, other retrospective
analyses of long-acting 2-agonists have not demonstrated How does inammation of the airways translate into clinical
symptoms of asthma?
a functional eect of these SNPs [284]. Similarly, there are
Airway thickening, as a consequence of the inammatory
genetically based functional variants in the regulation of the
response, may contribute to increased responsiveness to spas-
ALOX-5 gene [285]. Although these variants were originally
mogens [193, 291]. However, there is no obvious relationship
associated with the response to anti-leukotriene treatment
between the inammatory response in airways and asthma
[286], current studies suggest that it is not these variants per
severity; patients with mild asthma may have a similar eosi-
se, but variants in the ALOX-5 gene in general that may
nophil response to patients with severe asthma, suggesting
confer dierences in treatment response [287].
that there are other factors that determine clinical severity.
The nature of these factors remains elusive, but we must
identify them if we are to make true scientic progress.
UNANSWERED QUESTIONS How important are genetic factors (genetic polymorphisms) in
determining the phenotype of asthma?
As a syndrome much about asthma remains an enigma. Asthma is a complex genetic disease but likely one in which
Although we have reasonably eective treatments and a there is both dierences among individuals in the genes that
general idea of it pathophysiology, there are still many unan- pre-dispose to asthma and a requirement for more than one
swered questions about its pathobiology. genetic variant in a given person for asthma to be manifest.
Since each asthma gene may account for such a small pro-
Why is the prevalence of asthma increasing throughout the portion of the total genetic variance in asthma, how can we
world as a consequence of Westernization? use genetics to develop better asthma diagnostics? How can
It is clear that allergic diseases appear as societies become we use genetics to help stratify patients to achieve uniform
more Westernized, but what environmental factors are cohorts for clinical trials?
responsible is not known. It is likely that several factors are
operating together [288]. Among the factors identied to Are asthma and atopy causally related or just closely linked?
date are diet (reduced intake of antioxidants, reduced unsatu- Much of our understanding of asthma derives from study of
rated fats), lack of early childhood infections (with conse- models of allergy. Although these models are of great value
quent tendency to develop Th2-driven responses), greater in dening the pathobiology of asthma, how closely will they
exposure to allergens in the home (tight housing, mattresses, relate to the human disease?

416
Pathophysiology of Asthma 33
References 25. Arjomandi M, Witten A, Abbritti E et al. Repeated exposure to ozone
increases alveolar macrophage recruitment into asthmatic airways.
Am J Respir Crit Care Med 172(4): 42732, 2005.
1. Bosse Y, Hudson TJ. Toward a comprehensive set of asthma suscepti- 26. Kulkarni N, Pierse N, Rushton L, Grigg J. Carbon in airway macro-
bility genes. Annu Rev Med 58: 17184, 2007. phages and lung function in children. N Engl J Med 355(1): 2130, 2006.
2. Hogg JC. The pathology of asthma. APMIS 105(10): 73545, 1997. 27. Hammad H, Lambrecht BN. Recent progress in the biology of airway
3. Dunnill MS. The pathology of asthma, with special reference to the dendritic cells and implications for understanding the regulation of asth-
changes in the bronchial vasculature. J Clin Pathol 13: 2733, 1960. matic inammation. J Allergy Clin Immunol 118(2): 33136, 2006.
4. Hamid Q, Song YL, Kotsimbos TC et al. Inammation of small air- 28. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells:
ways in asthma. J Allergy Clin Immunol 100(1): 4451, 1997. Linking innate and adaptive immunity in asthma. Nat Rev Immunol
5. Martin RJ. Small airway and alveolar tissue changes in nocturnal 8(3): 193204, 2008.
asthma. Am J Respir Crit Care Med 157(5 Suppl.): S18890, 1998. 29. Parameswaran K, Liang H, Fanat A, Watson R, Snider DP, Obyrne PM.
6. Larche M, Robinson DS, Kay AB. The role of T lymphocytes in the Role for cysteinyl leukotrienes in allergen-induced change in circulating
pathogenesis of asthma. J Allergy Clin Immunol 111(3): 45063, 2003. dendritic cell number in asthma. J Allergy Clin Immunol 114: 7379, 2004.
7. Bradding P, Walls AF, Holgate ST. The role of the mast cell in the patho- 30. Gleich GJ. Mechanisms of eosinophil associated inammation.
physiology of asthma. J Allergy Clin Immunol 117(6): 127784, 2006. J Allergy Clin Immunol 195: 65163, 2000.
8. Ordonez CL, Khashayar R, Wong HH et al. Mild and moderate asthma 31. Rosenberg HF, Phipps S, Foster PS. Eosinophil tracking in allergy and
is associated with airway goblet cell hyperplasia and abnormalities in asthma. J Allergy Clin Immunol 119(6): 130310, 2007.
mucin gene expression. Am J Respir Crit Care Med 163(2): 51723, 2001. 32. Wegner CD, Gundel RH, Reilly P, Haynes N, Letts LG, Rothlein R.
9. Seroogy CM, Gern JE. The role of T regulatory cells in asthma. Intercellular adhesion molecule-1 (ICAM-1) in the pathogenesis of
J Allergy Clin Immunol 116(5): 99699, 2005. asthma. Science 247(4941): 45659, 1990.
10. Laitinen LA, Laitinen A, Altraja A et al. Bronchial biopsy ndings in 33. Laitinen LA, Laitinen A, Haahtela T, Vilkka V, Spur BW, Lee TH.
intermittent or early asthma. J Allergy Clin Immunol, 1996. Leukotriene-E(4) and granulocytic inltration into asthmatic airways.
11. Payne DN, Rogers AV, Adelroth E et al. Early thickening of the reticular Lancet 341: 98990, 1993.
basement membrane in children with dicult asthma. Am J Respir Crit 34. Laitinen A, Lindqvist A, Halme M, Altraja A, Laitinen LA. Leukotriene
Care Med 167(1): 7882, 2003. E-4-induced persistent eosinophilia and airway obstruction are reversed
12. Saglani S, Payne DN, Zhu J et al. Early detection of airway wall by zarlukast in patients with asthma. J Allergy Clin Immunol 115(2):
remodelling and eosinophilic inammation in preschool wheezers. 25965, 2005.
Am J Respir Crit Care Med, 2007. 35. Chanez P, Dent G, Yukawa T, Barnes PJ, Chung KF. Generation
13. Brusasco V, Pellegrino R. Complexity of factors modulating airway of oxygen free radicals from blood eosinophils from asthma patients
narrowing in vivo: Relevance to assessment of airway hyperresponsive- after stimulation with PAF or phorbol ester. Eur Respir J 3(9): 10027,
ness. J Appl Physiol 95(3): 130513, 2003. 1990.
14. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary 36. Evans DJ, Barnes PJ, Spaethe SM, Vanalstyne EL, Mitchell MI,
disease. Nat Rev Immunol 8(3): 18392, 2008. Oconnor BJ. Eect of a leukotriene B4 receptor antagonist, LY293111,
15. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, on allergen induced responses in asthma. Thorax 51(12): 117884, 1996.
Pavord ID. Mast-cell inltration of airway smooth muscle in asthma. 37. Evans DJ, Lindsay MA, OConnor BJ, Barnes PJ. Priming of circu-
N Engl J Med 346(22): 1699705, 2002. lating human eosinophils following late response to allergen challenge.
16. Bentley AM, Hamid Q, Robinson DS et al. Prednisolone treatment in Eur Respir J 9(4): 7038, 1996.
asthma. Reduction in the numbers of eosinophils, T cells, tryptase-only 38. Parameswaran K, Watson R, Gauvreau GM, Sehmi R, OByrne PM.
positive mast cells, and modulation of IL-4, IL-5, and interferon-gamma The eect of pranlukast on allergen-induced bone marrow eosinophi-
cytokine gene expression within the bronchial mucosa. Am J Respir Crit lopoiesis in subjects with asthma. Am J Respir Crit Care Med 169(8):
Care Med 153(2): 55156, 1996. 91520, 2004.
17. Foresi A, Leone C, Pelucchi A. Eosinophils, mast cells, and basophils in 39. Grithsjohnson DA, Collins PD, Rossi AG, Jose PJ, Williams TJ.
induced sputum from patients with seasonal allergic rhinitis and peren- The chemokine, eotaxin, activates guinea-pig eosinophils in vitro and
nial asthma: Relationship to methacholine responsiveness. J Allergy Clin causes their accumulation into the lung in vivo. Biochem Biophys Res
Immunol 100(5): 5864, 1997. Commun 197: 116772, 1993.
18. Boyce JA. The role of mast cells in asthma. Prostaglandins Leukot Essent 40. Ponath PD, Qin S, Ringler DJ et al. Cloning of the human eosinophil
Fatty Acids 69(23): 195205, 2003. chemoattractant, eotaxin. Expression, receptor binding, and functional
19. Reber L, Da Silva CA, Frossard N. Stem cell factor and its receptor properties suggest a mechanism for the selective recruitment of eosi-
c-Kit as targets for inammatory diseases. Eur J Pharmacol 533(13): nophils. J Clin Invest 97(3): 60412, 1996.
32740, 2006. 41. Luster AD, Rothenberg ME. Role of the monocyte chemoattractant
20. Milgrom H, Fick RB, Su JQ et al. Treatment of allergic asthma with protein and eotaxin subfamily of chemokines in allergic inammation.
monoclonal anti-IgE antibody. N Engl J Med 341(26): 196673, 1999. J Leukocyte Biol 62(5): 62033, 1997.
21. Avila PC. Does anti-IgE therapy help in asthma? Ecacy and contro- 42. Ravensberg AJ, Ricciardolo FL, van Schadewijk A et al. Eotaxin-2 and
versies. Annu Rev Med 58: 185203, 2007. eotaxin-3 expression is associated with persistent eosinophilic bron-
22. Holgate S, Casale T, Wenzel S, Bousquet J, Deniz Y, Reisner C. The chial inammation in patients with asthma after allergen challenge.
anti-inammatory eects of omalizumab conrm the central role of IgE J Allergy Clin Immunol 115(4): 77985, 2005.
in allergic inammation. J Allergy Clin Immunol 115(3): 45965, 2005. 43. Berkman N, Krishnan VL, Gilbey T et al. Expression of RANTES
23. Sabroe I, Parker LC, Dockrell DH, Davies DE, Dower SK, Whyte mRNA and protein in airways of patients with mild asthma. Am J
MK. Targeting the networks that underpin contiguous immunity in Respir Crit Care Med 154(6): 180411, 1996.
asthma and chronic obstructive pulmonary disease. Am J Respir Crit 44. Ponath PD, Qin SX, Post TW et al. Molecular cloning and characteri-
Care Med 175(4): 30611, 2007. zation of a human eotaxin receptor expressed selectively on eosinophils.
24. Huynh ML, Malcolm KC, Kotaru C et al. Defective apoptotic cell J Exp Med 183(6): 243748, 1996.
phagocytosis attenuates prostaglandin E2 and 15-hydroxyeicosatetrae- 45. Collins PD, Marleau S, Grithsjohnson DA, Jose PJ, Williams TJ.
noic acid in severe asthma alveolar macrophages. Am J Respir Crit Care Cooperation between interleukin-5 and the chemokine eotaxin to
Med 172(8): 97279, 2005. induce eosinophil accumulation in vivo. J Exp Med 182: 116974, 1995.

417
Asthma and COPD: Basic Mechanisms and Clinical Management

46. Zimmermann N, King NE, Laporte J et al. Dissection of experimen- 68. Naseer T, Minshall EM, Leung DY et al. Expression of IL-12 and
tal asthma with DNA microarray analysis identies arginase in asthma IL-13 mRNA in asthma and their modulation in response to steroid
pathogenesis. J Clin Invest 111(12): 186374, 2003. therapy. Am J Respir Crit Care Med 155(3): 84551, 1997.
47. Park CS, Choi YS, Ki SY et al. Granulocyte macrophage colony- 69. Grunig G, Warnock M, Wakil AE et al. Requirement for IL-13 inde-
stimulating factor is the main cytokine enhancing survival of eosi- pendently of IL-4 in experimental asthma. Science 282(5397): 226163,
nophils in asthmatic airways. Eur Respir J 12(4): 87278, 1998. 1998.
48. Walsh GM. Mechanisms of human eosinophil survival and apoptosis. 70. Wills-Karp M, Luyimbazi J, Xu X et al. Interleukin-13: Central medi-
Clin Exp Allergy 27(5): 48287, 1997. ator of allergic asthma. Science 282(5397): 225861, 1998.
49. Leckie MJ, ten Brincke A, khan j et al. Eects of an interleukin-5 71. Zhou Z, Homer RJ, Wang ZD et al. Pulmonary expression of interleukin-
blocking monoclonal antibody on eosinophils, airway hyperresponsive- 13 causes inammation, mucus hypersecretion, subepithelial brosis, phys-
ness and the late asthmatic response. Lancet 356: 214448, 2000. iologic abnormalities, and eotaxin production. J Clin Invest 103(6): 77988,
50. FloodPage P, MenziesGow A, Phipps S et al. Anti-IL-5 treatment 1999.
reduces deposition of ECM proteins in the bronchial subepithelial 72. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms
basement membrane of mild atopic asthmatics. J Clin Invest 112(7): of immune suppression by interleukin-10 and transforming growth
102936, 2003. factor-beta: The role of T regulatory cells. Immunology 117(4): 43342,
51. Empey DW, Laitinen LA, Jacobs L, Gold WM, Nadel JA. Mechanisms 2006.
of bronchial hyperreactivity in normal subjects after upper respiratory 73. Ling EM, Smith T, Nguyen XD et al. Relation of CD4CD25 regula-
tract infection. Am Rev Respir Dis 113: 13139, 1976. tory T-cell suppression of allergen-driven T-cell activation to atopic sta-
52. Wenzel SE. Asthma: Dening of the persistent adult phenotypes. tus and expression of allergic disease. Lancet 363(9409): 60815, 2004.
Lancet 368: 80413, 2007. 74. Ball TM, Castro-Rodriguez JA, Grith KA, Holberg CJ,
53. Wenzel S. Severe asthma in adults. Am J Respir Crit Care Med 172(2): Martinez FD, Wright AL. Siblings, day-care attendance, and the risk
14960, 2005. of asthma and wheezing during childhood. N Engl J Med 343(8):
54. Comhair SA, Ricci KS, Arroliga M et al. Correlation of systemic 53843, 2000.
superoxide dismutase deciency to airow obstruction in asthma. Am J 75. Braun-Fahrlander C, Riedler J, Herz U et al. Environmental exposure
Respir Crit Care Med 172(3): 30613, 2005. to endotoxin and its relation to asthma in school-age children. N Engl
55. Chu HW, Balzar S, Seedorf GJ et al. Transforming growth factor-beta J Med 347(12): 869, 2002.
2 induces bronchial epithelial mucin expression in asthma. Am J Pathol 76. Stockinger B, Veldhoen M. Dierentiation and function of Th17 T
165(4): 1097106, 2004. cells. Curr Opin Immunol 19(3): 28186, 2007.
56. Wenzel SE. The role of leukotrienes in asthma. Prostaglandins Leukot 77. Bullens DM, Truyen E, Coteur L et al. IL-17 mRNA in sputum of
Essent Fatty Acids 69(23): 14555, 2003. asthmatic patients: Linking T cell driven inammation and granulo-
57. Wenzel SE, Szeer SJ, Leung DY, Sloan SI, Rex MD, Martin RJ. cytic inux?. Respir Res 7: 135, 2006.
Bronchoscopic evaluation of severe asthma. Persistent inammation 78. Laan M, Lotvall J, Chung KF, Linden A. IL-17-induced cytokine
associated with high dose glucocorticoids. Am J Respir Crit Care Med release in human bronchial epithelial cells in vitro: Role of mitogen-
156(3 Pt 1): 73743, 1997. activated protein (MAP) kinases. Br J Pharmacol 133(1): 2006, 2001.
58. Simpson JL, Scott R, Boyle MJ, Gibson PG. Inammatory subtypes in 79. Nurieva R, Yang XO, Martinez G et al. Essential autocrine regulation
asthma: Assessment and identication using induced sputum. Respirology by IL-21 in the generation of inammatory T cells. Nature 448(7152):
11(1): 5461, 2006. 48083, 2007.
59. Sur S, Crotty TB, Kephart GM et al. Sudden-onset fatal asthma: 80. Spolski R, Leonard WJ. Interleukin-21: Basic biology and implications
A distinct entity with few eosinophils and relatively more neutrophils for cancer and autoimmunity. Annu Rev Immunol 26: 5779, 2008.
in the airway submucosa. Am Rev Respir Dis 148: 71319, 1993. 81. Wolk K, Sabat R. Interleukin-22: A novel T- and NK-cell derived
60. Maneechotesuwan K, Essile-Quaye S, Kharitonov SA, Adcock IM, cytokine that regulates the biology of tissue cells. Cytokine Growth
Barnes PJ. Loss of control of asthma following inhaled corticosteroid Factor Rev 17(5): 36780, 2006.
withdrawal is associated with increased sputum interleukin-8 and neu- 82. Akbari O, Faul JL, Hoyte EG et al. CD4 invariant T-cell-receptor
trophils. Chest 132(1): 98105, 2007. natural killer T cells in bronchial asthma. N Engl J Med 354(11):
61. Saar AS, Alphonse MP, Shan L, Hayglass KT, Simons FE, Gounni 111729, 2006.
AS. IgE modulates neutrophil survival in asthma: Role of mitochon- 83. Vijayanand P, Seumois G, Pickard C et al. Invariant natural killer
drial pathway. J Immunol 178(4): 253541, 2007. T cells in asthma and chronic obstructive pulmonary disease. N Engl J
62. Holgate ST, Holloway J, Wilson S et al. Understanding the patho- Med 356(14): 141022, 2007.
physiology of severe asthma to generate new therapeutic opportunities. 84. Meyer EH, DeKruy RH, Umetsu DT. T cells and NKT cells in the
J Allergy Clin Immunol 117(3): 496506, 2006. pathogenesis of asthma. Annu Rev Med 59: 28192, 2008.
63. Holt PG, Upham JW, Sly PD. Contemporaneous maturation of immu- 85. van Rensen EL, Sont JK, Evertse CE et al. Bronchial CD8 cell inl-
nologic and respiratory functions during early childhood: Implications trate and lung function decline in asthma. Am J Respir Crit Care Med
for development of asthma prevention strategies. J Allergy Clin Immunol 172(7): 83741, 2005.
116: 1624, 2005. 86. Cho SH, Stanciu LA, Holgate ST, Johnston SL. Increased interleukin-
64. Zhu Z, Zheng T, Homer RJ et al. Acidic mammalian chitinase in asth- 4, interleukin-5, and interferon-gamma in airway CD4 and CD8 T
matic Th2 inammation and IL-13 pathway activation. Science 304(5677): cells in atopic asthma. Am J Respir Crit Care Med 171(3): 22430, 2005.
167882, 2004. 87. Gould HJ, Beavil RL, Vercelli D. IgE isotype determination: Epsilon-
65. Elias JA, Homer RJ, Hamid Q, Lee CG. Chitinases and chitinase-like germline gene transcription, DNA recombination and B-cell dieren-
proteins in T(H)2 inammation and asthma. J Allergy Clin Immunol tiation. Br Med Bull 56(4): 90824, 2000.
116(3): 497500, 2005. 88. Takhar P, Corrigan CJ, Smurthwaite L et al. Class switch recombina-
66. Ying S, OConnor B, Rato J et al. Thymic stromal lymphopoietin expres- tion to IgE in the bronchial mucosa of atopic and nonatopic patients
sion is increased in asthmatic airways and correlates with expression of with asthma. J Allergy Clin Immunol 119(1): 21318, 2007.
Th2-attracting chemokines and disease severity. J Immunol 174(12): 89. Costa JJ, Weller PF, Galli SJ. The cells of the allergic response: Mast
818390, 2005. cells, basophils, and eosinophils. JAMA 278(22): 181522, 1997.
67. Kostenis E, Ulven T. Emerging roles of DP and CRTH2 in allergic 90. Youssef LA, Schuyler M, Gilmartin L et al. Histamine release from the
inammation. Trends Mol Med 12(4): 14858, 2006. basophils of control and asthmatic subjects and a comparison of gene

418
Pathophysiology of Asthma 33
expression between releaser and nonreleaser basophils. J Immunol 115. Dahlen B, Roquet A, Inman MD et al. Inuence of zarlukast and
178(7): 458494, 2007. loratadine on exercise-induced bronchoconstriction. J Allergy Clin
91. Macfarlane AJ, Kon OM, Smith SJ et al. Basophils, eosinophils, and mast Immunol 109(5): 78993, 2002.
cells in atopic and nonatopic asthma and in late-phase allergic reactions 116. Roquet A, Dahlen B, Kumlin M et al. Combined antagonism of leu-
in the lung and skin. J Allergy Clin Immunol 105(1 Pt 1): 99107, 2000. kotrienes and histamine produces predominant inhibition of allergen-
92. Levine SJ. Bronchial epithelial cell-cytokine interactions in airway induced early and late phase airway obstruction in asthmatics. Am J
inammation. J Invest Med 43(3): 24149, 1995. Respir Crit Care Med 155(6): 185663, 1997.
93. Boxall C, Holgate ST, Davies DE. The contribution of transforming 117. Freitag A, Watson RM, Matsos G, Eastwood C, Obyrne PM. Eect of
growth factor-beta and epidermal growth factor signalling to airway a platelet activating factor antagonist, WEB-2086, on allergen induced
remodelling in chronic asthma. Eur Respir J 27(1): 20829, 2006. asthmatic responses. Thorax 48: 59498, 1993.
94. Bai TR, Roberts CR, Pare PD. Airway remodelling. Asthma: Basic 118. Karasawa K. Clinical aspects of plasma platelet-activating factor-
Mechanisms and Clinical Management, 3rd edn, pp. 475486, London; acetylhydrolase. Biochim Biophys Acta 176: 135972, 2006.
San Diego: Academic press, 1998. 119. Kuitert LM, Angus RM, Barnes NC et al. Eect of a novel potent
95. Pascual RM, Peters SP. Airway remodeling contributes to the pro- platelet-activating factor antagonist, modipafant, in clinical asthma.
gressive loss of lung function in asthma: An overview. J Allergy Clin Am J Respir Crit Care Med 151(5): 133135, 1995.
Immunol 116(3): 47786, 2005. 120. Spence DP, Johnston SL, Calverley PM et al. The eect of the orally
96. Homer RJ, Elias JA. Airway remodeling in asthma: Therapeutic active platelet-activating factor antagonist WEB 2086 in the treat-
implications of mechanisms. Physiology 20(1): 2835, 2005. ment of asthma. Am J Respir Crit Care Med 149(5): 114248, 1994.
97. Bergeron C, Page N, Barbeau B, Chakir J. Interleukin-4 promotes 121. Staorini DM, Satoh K, Atkinson DL et al. Platelet-activating factor
airway remodeling in asthma-regulation of procollagen I (Alpha(1)) acetylhydrolase deciency: A missense mutation near the active site
gene by interleukin-4. Chest 123(3): 424S, 2003. of an anti-inammatory phospholipase. J Clin Invest 97(12): 278491,
98. James AL, Wenzel S. Clinical relevance of airway remodelling in air- 1996.
way diseases. Eur Respir J 30(1): 13455, 2007. 122. Staorini DM, Numao T, Tsodikov A et al. Deciency of platelet-
99. Hoshino M. Impact of inhaled corticosteroids and leukotriene recep- activating factor acetylhydrolase is a severity factor for asthma. J Clin
tor antagonists on airway remodeling. Clin Rev Allergy Immunol 27(1): Invest 103(7): 98997, 1999.
5964, 2004. 123. Ito S, Noguchi E, Shibasaki M, YamakawaKobayashi K, Watanabe H,
100. Lange P, Parner J, Vestbo J, Schnohr P, Jensen G. A 15-year follow- Arinami T. Evidence for an association between plasma platelet-
up study of ventilatory function in adults with asthma. N Engl J Med activating factor acetylhydrolase deciency and increased risk of
339(17): 1194200, 1998. childhood atopic asthma. J Hum Genet 47(2): 99101, 2002.
101. Mcfadden ER. Natural history of chronic asthma and its long-term 124. OByrne PM. Cytokines or their antagonists for the treatment of
eects on pulmonary function. J Allergy Clin Immunol 105(2): S535 asthma. Chest 130(1): 24450, 2006.
39, 2000. 125. Barnes PJ, Lim S. Inhibitory cytokines in asthma. Mol Med Today
102. Tschumperlin DJ, Dai G, Maly IV et al. Mechanotransduction 4(10): 45258, 1998.
through growth-factor shedding into the extracellular space. Nature 126. Elias JA. Cytokines in inammation. Chest, 2002.
429(6987): 8386, 2004. 127. Meiler F, Zimmermann M, Blaser K, Akdis CA, Akdis M. T-cell
103. Chu EK, Foley JS, Cheng J, Patel AS, Drazen JM, Tschumperlin DJ. subsets in the pathogenesis of human asthma. Curr Allergy Asthma
Bronchial epithelial compression regulates epidermal growth factor Rep 6(2): 9196, 2006.
receptor family ligand expression in an autocrine manner. Am J Respir 128. Oettgen HC, Geha RS. IgE regulation and roles in asthma pathogen-
Cell Mol Biol 32(5): 37380, 2005. esis. J Allergy Clin Immunol 107(3): 42940, 2001.
104. Tschumperlin DJ, Drazen JM. Chronic eects of mechanical force on 129. Karras JG, Crosby JRGM, Tung D et al. Anti-inammatory activity
airways. Annu Rev Physiol 68: 56383, 2006. of inhaled IL-4 receptor-alpha antisense oligonucleotide in mice. Am
105. Barnes PJ, Chung KF, Page CP. Inammatory mediators of asthma: J Respir Cell Mol Biol 36: 27685, 2007.
An update. Pharmacol Rev 50(4): 51596, 1998. 130. Chung KF, van Rensen E, Stirling R, Barnes PJ. IL-5 in asthma.
106. Spina D. Asthma mediators: Current views. J Pharm Pharmacol 52(2): Thorax 57(8): 751, 2002.
12545, 2000. 131. Chakir J, shannon J, Molet S et al. Airway remodeling-associated media-
107. Samuelsson B, Dahlen SE, Lindgren JA, Rouzer CA, Serhan CN. tors in moderate to severe asthma: eect of steroids on TGF-beta, IL-11,
Leukotrienes and lipoxins: Structures, biosynthesis, and biological IL-17, and type I and type III collagen expression. J Allergy Clin
eects. Science 237: 117176, 1987. Immunol 111: 129398, 2003.
108. Drazen JM, Israel E, Obyrne PM. Treatment of asthma with drugs mod- 132. Molet S, Hamid Q, davoine F et al. IL-17 is increased in asthmatic
ifying the leukotriene pathway. N Engl J Med 340(3): 197206, 1999. airways and induces human bronchial broblasts to produce cytokines.
109. Ogawa Y, Calhoun WJ. The role of leukotrienes in airway inamma- J Allergy Clin Immunol 108: 43038, 2001.
tion. J Allergy Clin Immunol 118(4): 78998, 2006. 133. Letuve S, Lajoie-Kadoch S, Audusseau S et al. IL-17E upregulates
110. Obyrne PM. Leukotriene bronchoconstriction induced by allergen the expression of proinammatory cytokines in lung broblasts.
and exercise. Am J Respir Crit Care Med 161(2): S6872, 2000. J Allergy Clin Immunol 117(3): 59096, 2006.
111. Le JA, Busse WW, Pearlman D et al. Montelukast, a leukotriene- 134. Berry MA, Hargadon B, Shelley M et al. Evidence of a role of tumor
receptor antagonist, for the treatment of mild asthma and exercise- necrosis factor alpha in refractory asthma. N Engl J Med 354(7): 697
induced bronchoconstriction. N Engl J Med 339(3): 14752, 1998. 708, 2006.
112. Adelroth E, Inman MD, Summers E, Pace D, Modi M, Obyrne PM. 135. Hamid Q, Azzawi M, Ying S et al. Expression of mRNA for inter-
Prolonged protection against exercise-induced bronchoconstriction leukin-5 in mucosal bronchial biopsies from asthma. J Clin Invest 87:
by the leukotriene D4-receptor antagonist cinalukast. J Allergy Clin 154146, 1991.
Immunol 99(2): 21015, 1997. 136. Teran LM. Chemokines and IL-5: Major players of eosinophil
113. Taylor IK, OShaughnessy KM, Fuller RW, Dollery CT. Eect of recruitment in asthma. Clin Exp Allergy 29(3): 28790, 1999.
cysteinyl-leukotriene receptor antagonist ICI 204.219 on allergen- 137. Flood-Page P, Swenson C, Faiferman I et al. A study to evaluate
induced bronchoconstriction and airway hyperreactivity in atopic safety and ecacy of mepolizumab in patients with moderate persist-
subjects. Lancet 337: 69094, 1991. ent asthma. Am J Respir Crit Care Med 176(11): 106271, 2007.
114. Wenzel SE. The role of leukotrienes in asthma. Prostaglandins Leukot 138. Umezu-Goto M, Kajiyama Y, Kobayashi N, Kaminuma O, Suko M,
Essent Fatty Acids 69(23): 14555, 2003. Mori A. IL-9 production by peripheral blood mononuclear cells of

419
Asthma and COPD: Basic Mechanisms and Clinical Management

atopic asthmatics. Int Arch Allergy Immunol 143(Suppl 1): 7679, 2007, 162. Hew M, Bhavsar P, Torrego A et al. Relative corticosteroid insensi-
Epub@2007 May 1 tivity of peripheral blood mononuclear cells in severe asthma. Am J
139. van den BS, Heymans J, Havaux X et al. Probrotic eect of IL-9 Respir Crit Care Med 174(2): 13441, 2006.
overexpression in a model of airway remodeling. Am J Respir Cell Mol 163. Barnes PJ. Endothelins and pulmonary diseases. J Appl Physiol 77:
Biol 37(2): 2029, 2007. 105159, 1994.
140. Melen E, Umerkaje S, Nyberg F et al. Interaction between variants 164. Dosanjh A, Zuraw B. Endothelin-1 (ET-1) decreases human bron-
in the interleukin-4 receptor alpha and interleukin-9 receptor genes chial epithelial cell migration and proliferation: Implications for air-
in childhood wheezing: Evidence from a birth cohort study. Clin Exp way remodeling in asthma. J Asthma 40(8): 88386, 2003.
Allergy 36(11): 139198, 2006. 165. Redington AE, Springall DR, Meng QH et al. Immunoreactive endothe-
141. Sitkauskiene B, Radinger M, Bossios A, Johansson AK, Sakalauskas R, lin in bronchial biopsy specimens: Increased expression in asthma and
Lotvall J. Airway allergen exposure stimulates bone marrow eosi- modulation by corticosteroid therapy. J Allergy Clin Immunol 100(4):
nophilia partly via IL-9. Respir Res 6: 33, 2005. 54452, 1997.
142. Thomas PS, Yates DH, Barnes PJ. Tumor necrosis factor-alpha 166. Chalmers GW, Thomson L, Macleod KJ et al. Endothelin-1 levels in
increases airway responsiveness and sputum neutrophilia in normal induced sputum samples from asthmatic and normal subjects. Thorax
human subjects. Am J Respir Crit Care Med 152: 7680, 1995. 52(7): 62527, 1997.
143. Barnes PJ. Transcription factors in airway diseases. Lab Invest 86(9): 167. Gawlik R, Jastrzebski D, Ziora D, Jarzab J. Concentration of
86772, 2006. endothelin in plasma and BAL uid from asthmatic patients. J Physiol
144. Gauldie J, Kolb M, Ask K, Martin G, Bonniaud P, Warburton D. Pharmacol 57(Suppl 4): 10310, 2006.
Smad3 signaling involved in pulmonary brosis and emphysema. 168. Shi-Wen X, Chen Y, Denton CP et al. Endothelin-1 promotes myo-
Proc Am Thorac Soc 3(8): 696702, 2006. broblast induction through the ETA receptor via a rac/phosphoinositide
145. Kelly MM, Leigh R, Bonniaud P et al. Epithelial expression of pro- 3-Kinase/Akt-dependent pathway and is essential for the enhanced
brotic mediators in a model of allergen-induced airway remodeling. contractile phenotype of brotic broblasts. Mol Biol Cell 15(6): 2707
Am J Respir Cell Mol Biol 32(2): 99107, 2005. 19, 2004.
146. Bonniaud P, Margetts PJ, Ask K, Flanders K, Gauldie J, Kolb M. 169. Barnes PJ. Nitric oxide and airway disease. Ann Med 27(3): 38993,
TGF-beta and Smad3 signaling link inammation to chronic bro- 1995.
genesis. J Immunol 175(8): 539095, 2005. 170. De Sanctis G, Maclean JA, Hamada K et al. Contribution of nitric
147. Liu YJ, Soumelis V, Watanabe N et al. TSLP: An epithelial cell oxide synthases 1, 2, and 3 to airway hyperresponsiveness and inam-
cytokine that regulates T cell dierentiation by conditioning dendritic mation in a murine model of asthma. J Exp Med 189(10): 162130,
cell maturation. Annu Rev Immunol 25: 193219, 2007. 1999.
148. Humbles AA, Lu B, Nilsson CA et al. A role for the C3a anaphylatoxin 171. Silko PE, Sylvester JT, Zamel N, Permutt S. Airway nitric oxide dif-
receptor in the eector phase of asthma. Nature 406(6799): 9981001, fusion in asthma: Role in pulmonary function and bronchial respon-
2000. siveness. Am J Respir Crit Care Med 161(4 Pt 1): 121828, 2000.
149. Karp CL, Grupe A, Schadt E et al. Identication of complement 172. Brindicci C, Ito K, Barnes PJ, Kharitonov SA. Dierential ow analy-
factor 5 as a susceptibility locus for experimental allergic asthma. sis of exhaled nitric oxide in patients with asthma of diering severity.
Nat Immunol 1(3): 22126, 2000. Chest 131(5): 135362, 2007.
150. Lambrecht BN. An unexpected role for the anaphylatoxin C5a recep- 173. Gustafsson LE, Leone AM, Persson MG, Wiklund NP, Moncada S.
tor in allergic sensitization. J Clin Invest 116(3): 62832, 2006. Endogenous nitric oxide is present in the exhaled air of rabbits, guinea
151. Kohl J, Wills-Karp M. A dual role for complement in allergic asthma. pigs and humans. Biochem Biophys Res Commun 181: 85257, 1991.
Curr Opin Pharmacol 7(3): 28389, 2007. 174. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R,
152. Kohl J, Baelder R, Lewkowich IP et al. A regulatory role for the C5a Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of
anaphylatoxin in type 2 immunity in asthma. J Clin Invest 116(3): asthmatic patients. Lancet 343: 13335, 1994.
78396, 2006. 175. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JM.
153. Walsh GM. Advances in the immunobiology of eosinophils and their Expired nitric oxide levels during treatment of acute asthma. Am J
role in disease. Crit Rev Clin Lab Sci 36(5): 45396, 1999. Respir Crit Care Med 152: 8003, 1995.
154. Cross CE, van der Vilet A, Eiserich JP. Peroxidases wheezing 176. Kharitonov SA, Gonio F, Kelly C, Meah S, Barnes PJ. Reproducibility
their way into asthma. Am J Respir Crit Care Med 164(7): 11023, of exhaled nitric oxide measurements in healthy and asthmatic adults
2001. and children. Eur Respir J 21(3): 43338, 2003.
155. Chambellan A, Cruickshank PJ, McKenzie P et al. Gene expression 177. Covar RA, Szeer SJ, Martin RJ et al. Relations between exhaled
prole of human airway epithelium induced by hyperoxia in vivo. Am nitric oxide and measures of disease activity among children with
J Respir Cell Mol Biol 35(4): 42435, 2006. mild-to-moderate asthma. J Pediatr 142(5): 46975, 2003.
156. Montuschi P, Corradi M, Ciabattoni G, Nightingale J, Kharitonov SA, 178. Hunt JF, Fang KZ, Malik R et al. Endogenous airway acidication:
Barnes PJ. Increased 8-isoprostane, a marker of oxidative stress, in Implications for asthma pathophysiology. Am J Respir Crit Care Med
exhaled condensate of asthma patients. Am J Respir Crit Care Med 161(3): 69499, 2000.
160(1): 21620, 1999. 179. Gaston B, Kelly R, Urban P et al. Buering airway acid decreases
157. Baraldi E, Carraro S, Alinovi R et al. Cysteinyl leukotrienes and exhaled nitric oxide in asthma. J Allergy Clin Immunol 118(4): 81722,
8-isoprostane in exhaled breath condensate of children with asthma 2006.
exacerbations. Thorax 58(6): 5059, 2003. 180. Ricciardolo FL, Gaston B, Hunt J. Acid stress in the pathology of
158. Baraldi E, Ghiro L, Piovan V et al. Increased exhaled 8-isoprostane in asthma. J Allergy Clin Immunol 113(4): 61019, 2004.
childhood asthma. Chest 124(1): 2531, 2003. 181. Lim S, Jatakanon A, Meah S, Oates T, Chung KF, Barnes PJ.
159. Comhair SA, Ricci KS, Arroliga M et al. Correlation of systemic Relationship between exhaled nitric oxide and mucosal eosinophilic
superoxide dismutase deciency to airow obstruction in asthma. inammation in mild to moderately severe asthma. Thorax 55(3):
Am J Respir Crit Care Med 172(3): 30613, 2005. 18488, 2000.
160. Comhair SA, Xu W, Ghosh S et al. Superoxide dismutase inactivation 182. Kharitonov SA, Barnes PJ. Exhaled biomarkers. Chest 130(5):
in pathophysiology of asthmatic airway remodeling and reactivity. 154146, 2006.
Am J Pathol 166(3): 66374, 2005. 183. Murphy MP, Packer MA, Scarlett JL, Martin SW. Peroxynitrite:
161. Cosio BG, Mann B, Ito K et al. Histone acetylase and deacetylase A biologically signicant oxidant. Gen Pharmacol 31(2): 17986, 1998.
activity in alveolar macrophages and blood mononocytes in asthma.
Am J Respir Crit Care Med 170(2): 14147, 2004.

420
Pathophysiology of Asthma 33
184. Robbins RA, Hadeli K, Nelson D, Sato E, Hoyt JC. Nitric 206. Kumar SD, Emery MJ, Atkins ND, Danta I, Wanner A. Airway
oxide, peroxynitrite, and lower respiratory tract inammation. mucosal blood ow in bronchial asthma. Am J Respir Crit Care Med
Immunopharmacology 48(3): 21721, 2000. 158(1): 15356, 1998.
185. Smith AD, Cowan JO, Brassett KP, Herbison GP, Taylor DR. Use of 207. Brieva JL, Danta I, Wanner A. Eect of an inhaled glucocorticoster-
exhaled nitric oxide measurements to guide treatment in chronic oid on airway mucosal blood ow in mild asthma. Am J Respir Crit
asthma. N Engl J Med 352(21): 216373, 2005. Care Med 161(1): 29396, 2000.
186. Menzies D, Nair A, Lipworth BJ. Portable exhaled nitric oxide 208. Paredi P, Kharitonov SA, Barnes PJ. Correlation of exhaled breath tem-
measurement: Comparison with the gold standard technique. Chest perature with bronchial blood ow in asthma. Respir Res 6: 15, 2005.
131(2): 41014, 2007. 209. Knox AJ, Deacon K, Cliord R. Blanching the airways: Steroid eects
187. Jeery PK, Godfrey RW, Adelroth E, Nelson F, Rogers A, in asthma. Thorax 62(4): 28385, 2007.
Johansson SA. Eects of treatment on airway inammation and thick- 210. McFadden ER Jr. Hypothesis: Exercise-induced asthma as a vascular
ening of basement membrane reticular collagen in asthma. A quantita- phenomenon. Lancet 335(8694): 88083, 1990.
tive light and electron microscopic study. Am Rev Respir Dis 145(4 Pt 211. Searles CD. Transcriptional and posttranscriptional regulation of
1): 89099, 1992. endothelial nitric oxide synthase expression. Am J Physiol Cell Physiol
188. Wilson JW, Li X. The measurement of reticular basement membrane 291(5): C80316, 2006.
and submusocal collagen in the asthmatic airway. Clin Exp Allergy 27: 212. Boo YC, Jo H. Flow-dependent regulation of endothelial nitric oxide
36371, 1997. synthase: Role of protein kinases. Am J Physiol Cell Physiol 285(3):
189. Saglani S, Molyneux C, Gong H et al. Ultrastructure of the reticular C499C508, 2003.
basement membrane in asthmatic adults, children and infants. Eur Respir 213. Abraham WM, Ahmed A, Serebriakov I et al. Whole-body periodic
J 28(3): 50512, 2006. acceleration modies experimental asthma in sheep. Am J Respir Crit
190. Jeery PK, Wardlaw AJ, Nelson FC, Collins JV, Kay AB. Bronchial Care Med 174(7): 74352, 2006.
biopsies in asthma. An ultrastructural, quantitative study and correla- 214. Bhandari V, Choo-Wing R, Chapoval SP et al. Essential role of nitric
tion with hyperreactivity. Am Rev Respir Dis 140(6): 174553, 1989. oxide in VEGF-induced, asthma-like angiogenic, inammatory, mucus,
191. Chetta A, Foresi A, Del Donno M et al. Bronchial responsiveness to and physiologic responses in the lung. Proc Natl Acad Sci USA 103(29):
distilled water and methacholine and its relationship to inammation 1102126, 2006.
and remodeling of the airways in asthma. Am J Respir Crit Care Med 215. Feltis BN, Wignarajah D, Zheng L et al. Increased vascular endothelial
153(3): 91017, 1996. growth factor and receptors: Relationship to angiogenesis in asthma.
192. Chetta A, Foresi A, Del Donno M, Bertorelli G, Pesci A, Olivieri D. Am J Respir Crit Care Med 173(11): 12017, 2006.
Airways remodeling is a distinctive feature of asthma and is related to 216. Siddiqui S, Sutclie A, Shikotra A et al. Vascular remodeling is a fea-
severity of disease. Chest 111(4): 85257, 1997. ture of asthma and nonasthmatic eosinophilic bronchitis. J Allergy
193. James AL, Maxwell PS, Pearcepinto G, Elliot JG, Carroll NG. The Clin Immunol 120(4): 81319, 2007.
relationship of reticular basement membrane thickness to airway wall 217. Persson CG. Plasma exudation and asthma. Lung 166: 123, 1988.
remodeling in asthma. Am J Respir Crit Care Med 166(12): 159095, 218. Chung KF, Rogers DF, Barnes PJ, Evans TW. The role of increased
2002. airway microvascular permeability and plasma exudation in asthma.
194. Chu EK, Foley J, Cheng J, Drazen JM, Tschumperlin DJ. Mechanical Eur Respir J 3(3): 32937, 1990.
regulation of the urokinase system as a potential mediator of remod- 219. Persson CGA, Andersson M, Grei L et al. Airway permeability. Clin
eling in a human airway cell culture model of asthma, Am J Respir Crit Exp Allergy 25: 80714, 1995.
Care Med 35(6): 62838, 2006. 220. Yager D, Shore S, Drazen JM. Airway luminal liquid. Sources and
195. Birring SS, Berry M, Brightling CE, Pavord ID. Eosinophilic bron- role as an amplier of bronchoconstriction. Am Rev Respir Dis 143:
chitis: Clinical features, management and pathogenesis. Am J Respir S52S54, 1991.
Med 2(2): 16973, 2003. 221. Yager D, Butler JP, Bastacky J, Israel E, Smith G, Drazen JM. Ampli-
196. Lazaar AL, Panettieri RA Jr. Airway smooth muscle: A modulator of air- cation of airway constriction due to liquid lling of airway interstices.
way remodeling in asthma. J Allergy Clin Immunol 116(3): 48895, 2005. J Appl Physiol 66: 287384, 1989.
197. Roth M, Johnson PRA, Borger P et al. Dysfunctional interaction 222. Dillard P, Wetsel RA, Drouin SM. Complement C3a regulates
of C/EBP and the glucocorticoid receptor in asthmatic bronchial Muc5ac expression by airway Clara cells independently of Th2
smooth-muscle cells. N Engl J Med 351(6): 56074, 2004. responses. Am J Respir Crit Care Med 175(12): 125058, 2007.
198. Sutclie A, Kaur D, Page S et al. Mast cell migration to Th2 stimulated 223. Song JS, Kang CM, Yoo MB et al. Nitric oxide induces MUC5AC
airway smooth muscle from asthmatics. Thorax 61(8): 65762, 2006. mucin in respiratory epithelial cells through PKC and ERK depend-
199. An SS, Bai TR, Bates JH et al. Airway smooth muscle dynamics: ent pathways. Respir Res 8: 28, 2007.
A common pathway of airway obstruction in asthma. Eur Respir J 224. Lora JM, Zhang DM, Liao SM et al. Tumor necrosis factor-alpha
29(5): 83460, 2007. triggers mucus production in airway epithelium through an IkappaB
200. Oliver BG, Black JL. Airway smooth muscle and asthma. Allergol Int kinase beta-dependent mechanism. J Biol Chem 280(43): 3651017, 2005.
55(3): 21523, 2006. 225. Perrais M, Pigny P, Copin MC, Aubert JP, Van SI. Induction of
201. Brightling CE, Ammit AJ, Kaur D et al. The CXCL10/CXCR3 axis MUC2 and MUC5AC mucins by factors of the epidermal growth
mediates human lung mast cell migration to asthmatic airway smooth factor (EGF) family is mediated by EGF receptor/Ras/Raf/extracel-
muscle. Am J Respir Crit Care Med 171(10): 11038, 2005. lular signal-regulated kinase cascade and Sp1. J Biol Chem 277(35):
202. Fernandes DJ, Mitchell RW, Lakser O, Dowell M, Stewart AG, 3225867, 2002.
Solway J. Invited review: Do inammatory mediators inuence the 226. Shao MXG, Nadel JA. Neutrophil elastase induces MUC5AC mucin
contribution of airway smooth muscle contraction to airway hyper- production in human airway epithelial cells via a cascade involving pro-
responsiveness in asthma? J Appl Physiol 95(2): 84453, 2003. tein kinase C, reactive oxygen species, and TNF--converting enzyme.
203. Cox G, Thomson NC, Rubin AS et al. Asthma control during the year J Immunol 175(6): 400916, 2005.
after bronchial thermoplasty. N Engl J Med 356(13): 132737, 2007. 227. Hallstrand TS, Debley JS, Farin FM, Henderson WR Jr.. Role of
204. Solway J, Irvin CG. Airway smooth muscle as a target for asthma MUC5AC in the pathogenesis of exercise-induced bronchoconstric-
therapy. N Engl J Med 356(13): 136769, 2007. tion. J Allergy Clin Immunol 119(5): 109298, 2007.
205. Huber HL, Koessler KK. The pathology of bronchial asthma. Arch 228. Barnes PJ. Is asthma a nervous disease? The Parker B. Francis lecture-
Intern Med 30: 689760, 1922. ship. Chest 107(3 Suppl): 119S25S, 1995.

421
Asthma and COPD: Basic Mechanisms and Clinical Management

229. Lou YP, Francocereceda A, Lundberg JM. Variable alpha2- 254. Avni O, Lee D, Macian F, Szabo SJ, Glimcher LH, Rao A. T(H) cell
adrenoceptor-mediated inhibition of bronchoconstriction and peptide dierentiation is accompanied by dynamic changes in histone acetyla-
release upon activation of pulmonary aerents. Eur J Pharmacol 210: tion of cytokine genes. Nat Immunol 3(7): 64351, 2002.
17381, 1992. 255. Du J, Huang C, Zhou B, Ziegler SF. Isoform-specic inhibition
230. Barnes PJ, Baraniuk JN, Belvisi MG. Neuropeptides in the respiratory of ROR-mediated transcriptional activation by human FOXP3.
tract, part I. Am Rev Respir Dis 144: 118798, 1991. J Immunol 180(7): 478592, 2008.
231. Joos GF, Germonpre PR, Pauwels R. Role of tachykinins in asthma. 256. Bonnans C, Levy BD. Lipid mediators as agonists for the resolu-
Allergy 55: 32137, 2000. tion of acute lung inammation and injury. Am J Respir Cell Mol Biol
232. Canning BJ, Undem BJ. Evidence that antidromically stimulated vagal 36(2): 2015, 2007.
aerents activate inhibitory neurones innervating guinea-pig trachea- 257. Schwab JM, Chiang N, Arita M, Serhan CN. Resolvin E1 and pro-
lis. J Physiol (Lond.) 480: 61325, 1994. tectin D1 activate inammation-resolution programmes. Nature
233. Barnes PJ. Modulation of neurotransmission in airways. Physiol Rev 447(7146): 86974, 2007.
72: 699729, 1992. 258. Bonnans C, Fukunaga K, Levy MA, Levy BD. Lipoxin A(4) regulates
234. Nockher WA, Renz H. Neurotrophins and asthma: Novel insight into bronchial epithelial cell responses to acid injury. Am J Pathol 168(4):
neuroimmune interaction. J Allergy Clin Immunol 117(1): 6771, 2006. 106472, 2006.
235. Lammers JW, Barnes PJ, Chung KF. Nonadrenergic, noncholinergic 259. Levy BD. Lipoxins and lipoxin analogs in asthma. Prostaglandins
airway inhibitory nerves. Eur Respir J 5(2): 23946, 1992. Leukot Essent Fatty Acids 73(34): 23137, 2005.
236. Belvisi MG, Stretton CD, Yacoub M, Barnes PJ. Nitric oxide is the 260. Levy BD, Bonnans C, Silverman ES, Palmer LJ, Marigowda G,
endogenous neurotransmitter of bronchodilator nerves in humans. Israel E. Diminished lipoxin biosynthesis in severe asthma. Am J
Eur J Pharmacol 210: 22122, 1992. Respir Crit Care Med 172(7): 82430, 2005.
237. Joos GF, De Swert KO, Schelfhout V, Pauwels RA. The role of neural 261. Levy BD, Desanctis GT, Devchand PR et al. Multi-pronged inhibi-
inammation in asthma and chronic obstructive pulmonary disease. tion of airway hyper-responsiveness and inammation by lipoxin
Ann NY Acad Sci 992: 21830, 2003. A(4). Nat Med 8(9): 101823, 2002.
238. Ollerenshaw SL, Jarvis D, Sullivan CE, Woolcock AJ. Substance P 262. Levy BD, Clish CB, Schmidt B, Gronert K, Serhan CN. Lipid
immunoreactive nerves in airways from asthmatics and nonasthmatics. mediator class switching during acute inammation: Signals in reso-
Eur Respir J 4: 67382, 1991. lution. Nat Immunol 2(7): 61219, 2001.
239. Howarth PH, Springall DR, Redington AE, Djukanovic R, 263. Sanak M, Levy BD, Clish CB et al. Aspirin-tolerant asthmatics gen-
Holgate ST, Polak JM. Neuropeptide-containing nerves in endobron- erate more lipoxins than aspirin-intolerant asthmatics. Eur Respir J
chial biopsies from asthmatic and nonasthmatic subjects. Am J Respir 16(1): 4449, 2000.
Cell Mol Biol 13(3): 28896, 1995. 264. Bonnans C, Fukunaga K, Keledjian R, Petasis NA, Levy BD.
240. Nadel JA. Neutral endopeptidase modulates neurogenic inammation. Regulation of phosphatidylinositol 3-kinase by polyisoprenyl phos-
Eur Respir J 4(6): 74554, 1991. phates in neutrophil-mediated tissue injury. J Exp Med 203(4):
241. Ahn KS, Aggarwal BB. Transcription factor NF-kappaB: A sensor for 85763, 2006.
smoke and stress signals. Ann NY Acad Sci 1056: 21833, 2005. 265. Aoki H, Hisada T, Ishizuka T et al. Resolvin E1 dampens airway
242. Wright JG, Christman JW. The role of nuclear factor kappa B in the inammation and hyperresponsiveness in a murine model of asthma.
pathogenesis of pulmonary diseases: Implications for therapy. Am J Biochem Biophys Res Commun 367(2): 50915, 2008.
Respir Med 2(3): 21119, 2003. 266. Levy BD, Kohli P, Gotlinger K et al. Protectin D1 is generated in
243. Ulevitch RJ. Therapeutics targeting the innate immune system. Nat asthma and dampens airway inammation and hyperresponsiveness.
Rev Immunol 4(7): 51220, 2004. J Immunol 178(1): 496502, 2007.
244. Hart LA, Krishnan VL, Adcock IM, Barnes PJ, Chung KF. Activation 267. Hawrylowicz CM. Regulatory T cells and IL-10 in allergic inam-
and localization of transcription factor, nuclear Factor-kappa B, in mation. J Exp Med 202(11): 145963, 2005.
asthma. Am J Respir Crit Care Med 158(5): 158592, 1998. 268. John M, Lim S, Seybold J et al. Inhaled corticosteroids increase inter-
245. Swartz MA, Tschumperlin DJ, Kamm RD, Drazen JM. Mechanical leukin-10 but reduce macrophage inammatory protein-1alpha,
stress is communicated between dierent cell types to elicit matrix granulocyte-macrophage colony-stimulating factor, and interferon-
remodeling. Proc Natl Acad Sci USA 98(11): 618085, 2001. gamma release from alveolar macrophages in asthma. Am J Respir Crit
246. Hilberg T. Etiology of exercise-induced asthma: Physical stress- Care Med 157(1): 25662, 1998.
induced transcription. Curr Allergy Asthma Rep 7(1): 2732, 2007. 269. Stelmach I, Jerzynska J, Kuna P. A randomized, double-blind trial of
247. Kumar A, Lnu S, Malya R et al. Mechanical stretch activates nuclear the eect of glucocorticoid, antileukotriene and beta-agonist treat-
factor-kappaB, activator protein-1, and mitogen-activated protein ment on IL-10 serum levels in children with asthma. Clin Exp Allergy
kinases in lung parenchyma: Implications in asthma. FASEB J 17(13): 32(2): 26469, 2002.
180011, 2003. 270. Martinez FD. Genes, environments, development and asthma: A reap-
248. Ho IC, Pai SY. GATA-3-not just for Th2 cells anymore. Cell Mol praisal. Eur Respir J 29(1): 17984, 2007.
Immunol 4(1): 1529, 2007. 271. Hersh CP, Raby BA, Soto-Quiros ME et al. Comprehensive testing
249. Caramori G, Lim S, Ito K et al. Expression of GATA family of tran- of positionally cloned asthma genes in two populations. Am J Respir
scription factors in T-cells, monocytes and bronchial biopsies. Eur Crit Care Med, 2007.
Respir J 18(3): 46673, 2001. 272. Van Eerdewegh P, Little RD, Dupuis J et al. Association of the
250. Maneechotesuwan K, Xin Y, Ito K et al. Regulation of Th2 cytokine ADAM33 gene with asthma and bronchial hyperresponsiveness.
genes by p38 MAPK-mediated phosphorylation of GATA-3. J Immunol Nature, 2002.
178(4): 249198, 2007. 273. Holgate ST, Yang Y, Haitchi HM et al. The genetics of asthma:
251. Finotto S, Neurath MF, Glickman JN et al. Development of sponta- ADAM33 as an example of a susceptibility gene. Proc Am Thorac Soc
neous airway changes consistent with human asthma in mice lacking 3(5): 44043, 2006.
T-bet. Science 295(5553): 33638, 2002. 274. Moatt MF, Kabesch M, Liang L et al. Genetic variants regulating
252. Hwang ES, Szabo SJ, Schwartzberg PL, Glimcher LH. T helper cell ORMDL3 expression contribute to the risk of childhood asthma.
fate specied by kinase-mediated interaction of T-bet with GATA-3. Nature 448(7152): 47073, 2007.
Science 307(5708): 43033, 2005. 275. Weiss ST, Litonjua AA, Lange C et al. Overview of the pharmacoge-
253. Usui T, Preiss JC, Kanno Y et al. T-bet regulates Th1 responses netics of asthma treatment. Pharmacogenomics J 6(5): 31126, 2006.
through essential eects on GATA-3 function rather than on IFNG 276. Liggett SB. Polymorphisms of the beta2-adrenergic receptor. N Engl J
gene acetylation and transcription. J Exp Med 203(3): 75566, 2006. Med 346: 53638, 2002.

422
Pathophysiology of Asthma 33
277. Martinez FD, Graves PE, Baldini M, Solomon S, Erickson R. longacting beta2-agonist therapy: A pharmacogenetic analysis of two
Association between genetic polymorphisms of the beta(2)- randomised studies. Lancet 370(9605): 211825, 2007.
adrenoceptor and response to albuterol in children with and without a 285. In KH, Asano K, Beier D et al. Naturally occurring mutations in the
history of wheezing. J Clin Invest 100(12): 318488, 1997. human 5-lipoxygenase gene promoter that modify transcription factor
278. Thakkinstian A, McEvoy M, Minelli C et al. Systematic review and binding and reporter gene transcription. J Clin Invest 99(5): 113037,
meta-analysis of the association between beta(2)-adrenoceptor poly- 1997.
morphisms and asthma: A HuGE review. Am J Epidemiol 162(3): 286. Drazen JM, Yandava C, Dube LM et al. Pharmacogenetic association
20111, 2005. between ALOX5 promoter genotype and the response to anti-asthma
279. Israel E, Drazen JM, Liggett SB et al. Eect of polymorphism of the treatment. Nat Genet 22: 16870, 1999.
beta(2)-adrenergic receptor on response to regular use of albuterol in 287. Lima JJ, Zhang S, Grant A et al. Inuence of leukotriene pathway
asthma. Int Arch Allergy Immunol 124(13): 18386, 2001. polymorphisms on response to montelukast in asthma. Am J Respir
280. Hall IP. Pharmacogenetics of asthma. Chest 130(6): 187378, 2006. Crit Care Med 173(4): 37985, 2006.
281. Israel E, Chinchilli VM, Ford JG et al. Use of regularly scheduled 288. Eder W, Ege MJ, Vonmutius E. Current concepts: The asthma epi-
albuterol treatment in asthma: Genotype-stratied, randomised, demic. N Engl J Med 355(21): 222635, 2006.
placebo-controlled cross-over trial. Lancet 364(9444): 150512, 2004. 289. Malo JL, Chanyeung M. Occupational asthma. J Allergy Clin Immunol
282. Wechsler ME, Lehman E, Lazarus SC et al. beta-Adrenergic receptor 108(3): 31728, 2001.
polymorphisms and response to salmeterol. Am J Respir Crit Care Med 290. Beckett WS. Current concepts: Occupational respiratory diseases. N
173(5): 51926, 2006. Engl J Med 342(6): 40613, 2000.
283. Bleecker ER, Yancey SW, Baitinger LA et al. Salmeterol response is 291. Niimi A, Matsumoto H, Takemura M, Ueda T, Chin K, Mishima
not aected by beta2-adrenergic receptor genotype in subjects with M. Relationship of airway wall thickness to airway sensitivity and
persistent asthma. J Allergy Clin Immunol 118(4): 80916, 2006. airway reactivity in asthma. Am J Respir Crit Care Med 168(8): 983
284. Bleecker ER, Postma DS, Lawrance RM, Meyers DA, Ambrose HJ, 88, 2003.
Goldman M. Eect of ADRB2 polymorphisms on response to

423
Pathophysiology of COPD
34
CHAPTER

INTRODUCTION commonest cause of COPD is chronic cigarette Peter J. Barnes1 and2


smoking, some patients, particularly in develop- Stephen I. Rennard
ing countries, develop the disease from inhala-
1
COPD is a complex inammatory disease that tion of wood smoke from biomass fuels or other National Heart and Lung Institute
involves multiple interacting cells and media- inhaled irritants [2]. However, only about 25% (NHLI), Clinical Studies Unit,
tors and various tissue destruction and repair of smokers develop COPD of at least moder- Imperial College, London, UK
mechanisms leading to structural changes that ate severity [3], although as many as half will 2
Pulmonary and Critical Care
result in progressive airow limitation with lit- develop abnormal lung function [4], suggest- Medicine, University of Nebraska
tle reversibility. The preceding chapters discuss ing that there may be genetic or host factors Medical Center, Omaha, NE, USA
details of the individual inammatory cells and that predispose to its development, although
mediators and the structural changes that are these have not yet been clearly identied (see
found in COPD. The links between the cellu- Chapter 4). The disease is thought to be relent-
lar and molecular mechanisms, the pathology, lessly progressive and, to date, only smoking ces-
the physiological abnormalities, and symptoms sation has been demonstrated to reduce the rate
are still not well understood. This is compli- of decline in lung function, although as the dis-
cated by the fact that there is heterogeneity of ease becomes more severe there is less eect of
the disease, with some patients showing a pre- smoking cessation and lung inammation per-
dominant emphysema pattern, whereas in oth- sists. However, it is not yet clear that all patients
ers small airway disease predominates, although with mild and moderate COPD show progres-
many patients have a mixed pattern, as ciga- sion. Some patients may have small lungs due to
rette smoking is a common causal mechanism. abnormalities in lung development due to fetal
There are dierences in the degree of mucus or early childhood inuences and develop symp-
hypersecretion between patients and in the toms of COPD as a result of the normal decline
frequency of infective exacerbations. Finally in lung function with aging but starting from a
it is nowrecognized that COPD aects extra- lower peak (Fig. 34.1). These patients may end
pulmonary organs, such as skeletal muscle, as up with airow obstruction without abnormal
well as increasing the prevalence of common co- inammatory changes.
morbid diseases, such as cardiovascular disease
and lung cancer, which together are the com-
monest cause of death in patients with COPD.
The denition of COPD adopted by the
Global initiative on Obstructive Lung Disease AIRFLOW LIMITATION
(GOLD) encompasses the idea that COPD
is a chronic inammatory disease and much of
Airow limitation in COPD may be the result
the recent research has focused on the nature
of three dierent pathological mechanisms
of this inammatory response [1]. COPD is
(Fig. 34.2):
an obstructive disease of the lungs which
slowly progresses over many decades leading to 1. Thickening and brosis of small airways
death from respiratory failure unless patients (chronic obstructive bronchiolitis) which is
die of co-morbidities such as heart disease and presumed to be due to the eect of chronic
lung cancer before this stage. Although the inammation.
425
Asthma and COPD: Basic Mechanisms and Clinical Management

3
Normal decline
FEV1 (l)

(~18 ml/yr) FIG. 34.1 Small airways in COPD patients. The airway
2 wall is thickened and infiltrated with inflammatory cells,
predominately macrophages and CD8 lymphocytes, with
Symptoms increased numbers of fibroblasts. In severe COPD there are
Normal decline
1 also lymphoid follicles, which consist of a central core of
with low initial lung volume
Accelerated decline B-lymphocytes, surrounded by T-lymphocytes and thought
to indicate chronic exposure to antigens (bacterial, viral, or
0 autoantigens). Similar changes are also reported in larger
30 40 50 60 70 airways. The lumen is often filled with an inflammatory
Age (years) exudate and mucus. There is peribronchial fibrosis, and,
Peak lung resulting in progressive and irreversible narrowing of the
function airway. Airway smooth muscle may be increased slightly.

Inflammatory exudate in lumen

Disrupted alveolar attachments

Thickened wall with inflammatory cells


macrophages, CD8 cells, fibroblasts
FIG. 34.2 Mechanisms of airflow limitation in
Peribronchial fibrosis COPD. Small airways are obstructed as a result
Lymphoid follicle
of loss of alveolar attachments, thickening
and distortion of the airway wall as a result
of inflammation and fibrosis and occlusion of
the airway lumen by mucus and inflammatory
exudate.

2. Emphysema results in expiratory airway collapse for ETIOLOGIC FACTORS


two reasons: Disruption of alveolar attachments permits
small airways to close on expiration due to lack of elastic
support; in addition, loss of lung elastic recoil decreases Inhalational exposures
the intralumenal pressure, which also predisposes to
small airway collapse, particularly with forced exhalation. The most important etiologic risk factors for the develop-
3. Luminal obstruction with mucus and plasma exudate as a ment of COPD are inhalational exposures and cigarette
result of inammation. smoke is by far the most important of these. Exposures
other than cigarette smoking, however, also contribute to
All three mechanisms may contribute to air trapping the risk for COPD. Such exposures can both cause COPD
and lung hyperination, resulting in dyspnea and exercise independently of cigarette smoking and can increase the
limitation [5] (Fig. 34.3) (see Chapter 5). The relative roles risk for COPD in the presence of concurrent cigarette
of emphysema and small airway disease in causing airow smoking. Exposures leading to COPD include a range of
limitation are debated, but recent studies with measurements both environmental fumes and dusts. Coal dust, for exam-
of emphysema by computerized tomography show that ple, alone can lead to airow limitation. Use of biomass for
varying severity of emphysema can be present with similar home cooking can result in signicant inhalational exposure
impairment of FEV1, although there may be abnormalities and a marked increased risk for the development of COPD
in diusion capacity and hypoxia [6]. The variable asso- [8]. Such exposure is a currently a major risk factor for
ciation of emphysema with airow limitation has focused COPD in parts of the developing world. Passive cigarette
increased attention on small airways in COPD, but it is dif- smoke exposure also is related to symptoms of cough and
cult to measure small airway function in patients and novel sputum production and is likely a risk factor for the devel-
imaging and physiological approaches are needed [7]. opment of airow limitation [9].

426
Pathophysiology of COPD 34
Normal Severe Respiratory Questionnaire. The suggestion has also been
Inspiration COPD made that individuals with frequent exacerbations experi-
ence a more rapid decline in lung function [10]. Damage
Small induced by infections and the ensuing inammatory proc-
airway ess may make the airways more susceptible to subsequent
infection [11].
Infections also have been related to increased risk for
Alveolar attachments Loss of alveolar the development of COPD. A history of severe childhood
attachments viral infection has been associated with reduced lung func-
Expiration tion and with increased respiratory symptoms in some stud-
ies. There are several, non-exclusive, possible explanations
Closure for these associations. First, there might be an increased
diagnosis of severe infections in children who have underly-
ing airways responsiveness which itself may be a risk fac-
tor. Second, viral infections may be related to another factor
such as socioeconomic status or birth weight which itself is
related to COPD. Viral infections, moreover, may directly
Health Dyspnea Air trapping
status Exercise capacity Hyperinflation contribute to the development of COPD. In this regard,
viral infections may result in incorporation of viral DNA
into airway cells which could alter the response to subse-
FIG. 34.3 Mechanism of dyspnea in COPD. The airway narrows on
expiration but in normal individuals the elasticated alveolar attachments
quent exposures increasing the risk for COPD. In contrast,
prevent closure so that the alveoli empty. In COPD there is a loss of elastin a more recent study failed to show an accelerated rate of
fibers as a result of elastases, which means that even in mild COPD small decline among adults age 3545 who had a history of child-
airways close to a greater extent. As COPD becomes more severe, the hood infections. However, another study did not demon-
thickness of small airways increases and alveolar attachments may be strate an accelerated rate of decline in adults aged 3545
disrupted so that peripheral airway may close during expiration. This results who had childhood infections [12].
in air trapping and hyperinflation, leading to dyspnea and reduced exercise Animal studies suggest the incorporation of adeno-
capacity. viral DNA can amplify the inammatory response induced
when airway epithelial cells are exposed to cigarette smoke
and increased levels of adenoviral DNA have been detected
Some exposures which represent risk factors for the in the lungs of patients with COPD compared to control
development of COPD, for example intravenous drug abuse patients [13]. In addition, HIV infection has also been
and cadmium may act through pathways distinct from the related to the development of emphysema [14]. HIV-
more common inhalational exposures. Intravenous drug induced pulmonary inammation may play a role, although
abuse may lead to the development of COPD by a vas- weight loss associated with HIV infection has also been
cular mechanism, and cadmium, which can cause emphy- suggested as a possible mechanism.
sema without inammation, may act by altering the repair
response. These rare causes of COPD are particularly
instructive as they indicate that a variety of mechanisms Nutrition
may lead to altered lung structure and function. To what
degree such mechanisms are involved in the more common Nutritional status, independent of concurrent viral infec-
etiologies of COPD remains to be determined. tion, is also likely a risk factor for the development of
It is likely that the varied inhalational exposures, COPD. The famine and resulting starvation in the Warsaw
which lead to COPD, share the ability to initiate some of ghetto in World War II was associated with the develop-
the same pathogenetic mechanisms. This may account for ment of emphysema. Reduced body weight, often expressed
some of the similarities of the disease induced by ciga- as body mass index, is a poor prognostic sign in individu-
rette smoke and other environmental exposures. Most als with established COPD. Underweight individuals with
exposures, however, are complex and contain many candi- COPD, moreover, appear to be at risk for the development
date toxins. Importantly, not all exposed individuals will of emphysema. The association of starvation and anabolic/
develop COPD. Genetic factors, most of which remain to catabolic status with the development of COPD, particu-
be dened, can aect risk. larly with emphysema, is also supported by experimental
studies in animals.
Infections
Infections play a key role in COPD exacerbations Birth weight, lung growth, and
(see Chapter 37). Up to two-thirds of exacerbations can development
be associated with a potential infectious pathogen, about
half being bacterial and half being viral. Individuals with Low birth weight has been associated with the development
frequent exacerbations have been demonstrated to have of COPD although this association remains controversial.
an inferior quality of life as assessed by the St. Georges Low birth weight has also been associated with increased

427
Asthma and COPD: Basic Mechanisms and Clinical Management

risk for asthma and with reduced lung function in both To date the only well-dened genetic risk factor is
childhood and young adulthood. In this regard, reduced deciency of alpha-1 protease inhibitor (PI). Severe de-
maximal attained lung function may identify individuals ciency in this major circulating inhibitor of serine proteases
who are at increased risk for the development of COPD. is associated with development of emphysema in non-
While some of these results are controversial it remains smokers, although not all decient individuals are aected.
possible that developmental problems associated with In smokers, alpha-1 PI deciency is associated with the
low birth weight represent a risk factor for the development accelerated development of emphysema and mortality.
of COPD. A number of other genetic factors, while not unequivocally
established, have been suggested to be related to risk for
COPD (see Chapter 4).
Aging
COPD is a disease of the elderly. While decrements in Tissue remodeling
lung function can be observed at any age and decline in
lung function can begin in young adulthood, clinically sig- Peribronchial brosis and narrowing contributes to air-
nicant COPD is unusual before the fth or sixth decade ow limitation in chronic bronchitis. The mechanisms
of life and is most common even later [15]. Lung function which lead to this process are unknown. However, injury
also declines with normal aging and compliance increases. of the small airways, either directly by inhaled toxins such
Associated with this process are changes in alveolar struc- as cigarette smoke or indirectly by the action of inamma-
ture including increased size and number of alveolar pores tory mediators likely initiates repair processes [22]. In this
together with enlargement in alveolar spaces. These changes, regard, the airway epithelium has considerable capacity to
which are sometimes termed senile emphysema, resemble, mediate repair. Following mechanical injury, for example,
in at least some features, clinical emphysema. This suggests airway epithelial cells from the edge of the wound rapidly
the concept that aging may lead to emphysema or may atten and migrate to cover the defect.
accelerate its development. Further supporting a connec- In vitro studies suggest that bronectin and trans-
tion between COPD and aging, circulating lymphocytes forming growth factor (TGF)- produced by the epithelial
in smokers have reduced telomere length [16] and chronic cells present in the wound may help direct these processes.
exposure to cigarette smoke can induce senescence in cells The newly recruited cells then replicate and undergo an
cultured in vitro [17]. In addition, broblasts cultured from orderly sequence of dierentiation. It is likely that these
the lungs of COPD patients proliferate more slowly and processes often can restore both anatomic structure and air-
also show markers of senescence [18, 19]. Finally, emphy- way function. In addition to the epithelial cells, mesenchy-
sema spontaneously develops in mice with genetic forms of mal cells (broblasts and myobroblasts) are also activated
accelerated aging [20, 21]. Taken together, these observa- in the repair response. The wave of accumulation and rep-
tions support the concept that COPD may represent a form lication of epithelial cells which occurs 24 h after mechani-
of accelerated senescence in the lung. cal injury is followed about 2 days later by the accumulation
and proliferation of mesenchymal cells. Under normal
circumstances, these cells disappear over the next few weeks.
However, as in many tissues, repair in the airways
Reactive airways can result in the excessive deposition of brotic extracellu-
lar matrix, and like most scars, these contract [22]. If this
Asthma and increased airways reactivity have been iden- were to happen circumferentially around an airway, airway
tied as risk factors for the development of COPD. This narrowing would result. In this context, airway epithelial
relationship was originally proposed by Orie and colleagues cells can also produce factors which drive broblast recruit-
and termed the Dutch hypothesis. Asthmatics, as a group, ment, matrix production and remodeling. Interestingly, the
experience accelerated loss of lung function as do smokers brotic process may be driven by some of the same media-
with increased airways reactivity. Whether airways reactiv- tors which may also lead to epithelial repair such as TGF-
ity results from an inammatory process, which, in turn, and bronectin. Consistent with a role for these mediators,
also leads to loss of lung function in COPD or whether both TGF- and bronectin have been reported in the air-
the airways reactivity directly causes COPD remains ways and bronchoalveolar lavage (BAL) uid in asthma
undetermined. and chronic bronchitis. While the processes which regulate
airway repair following injury are only partly delineated, it
seems likely that disordered repair processes can lead to tis-
Genetics sue remodeling with altered airway structure and function.
Altered repair mechanisms may contribute to the
It is likely that many genetic factors interact with and development of emphysema [22]. In this context, the net
increase (or decrease) the risk for developing COPD. tissue loss which characterizes emphysema may result from
Family studies have demonstrated an increased risk of inadequate repair in the face of injury. Several lines of evi-
COPD within families with COPD probands. Some of this dence support this concept. First, several animal models of
risk may be due to shared environmental factors, but several emphysema have been developed in which lung destruc-
studies in diverse populations also suggest shared genetic tion is induced either by exposure to large concentrations
risk (see Chapter 4). of neutrophil elastase (NE), to cigarette smoke or to other

428
Pathophysiology of COPD 34
similar insults. Such injury is characterized by a rapid loss in the lung periphery. Quantitative studies have shown
of lung connective tissue consistent with tissue destruction that the inammatory response in small airways and lung
taking place. These models, however, are also associated with parenchyma increases as the disease progresses [27] (see
a rapid onset of new connective tissue synthesis. In many Chapter 6). There is a specic pattern of inammation in
cases, total tissue matrix macromolecule concentration is COPD airways and lung parenchyma with increased num-
restored to normal or increased levels within a few weeks bers of macrophages, T-lymphocytes, with predominance
of tissue injury. Similarly, in mild human emphysema, lung of CD8 (cytotoxic) T-cells, and in more severe disease
collagen content has been reported to be increased consist- B-lymphocytes with increased numbers of neutrophils in
ent with initiation of matrix molecule production. the lumen [28] (Fig. 34.4). The inammatory response
The concept that repair processes initiated in model in COPD involves both innate and adaptive immune
systems of emphysema serve to mitigate the severity of the responses. Multiple inammatory mediators are increased in
resulting emphysema is supported by studies in which repair COPD and are derived from inammatory cells and struc-
processes are disrupted. For example, starvation can greatly tural cells of the airways and lungs [29]. A similar pattern
potentiate the development of emphysema following elastase of inammation is seen in smokers without airow limita-
exposure in rats [23]. Similarly, inhibition of matrix macro- tion, but in COPD this inammation is amplied and even
molecule cross-linking can also exacerbate the development further amplied during acute exacerbations of the disease
of emphysema. In this context, cigarette smoke can interfere which are usually precipitated by bacterial and viral infec-
with repair processes by a number of mechanisms. Smoke tions (Fig. 34.5). The molecular basis of this amplication
can inhibit parenchymal cell recruitment, proliferation, of inammation is not yet understood but may be, at least
matrix production, and tissue remodeling [22]. Smoke can in part, genetically determined. Cigarette smoke and other
also interfere with matrix macromolecule cross-linking [24]. irritants in the respiratory tract may activate surface macro-
It is possible, therefore, that smoke can lead to the develop- phages and airway epithelial cells to release chemotactic fac-
ment of emphysema by three interacting mechanisms: (1) tors that then attract circulating leukocytes into the lungs.
by initiating an inammatory response which causes tissue Amongst chemotactic factors, chemokines predominate
destruction; (2) by interfering with the defenses which nor- and therefore play a key role in orchestrating the chronic
mally protect tissues from injury during inammation; and inammation in COPD lungs and its further amplication
(3) by disrupting the repair processes which have the poten- during acute exacerbations [30]. These might be the initial
tial for restoring tissue architecture in the face of injury. inammatory events occurring in all smokers. However in
Several lines of evidence suggest that tissue repair or smokers who develop COPD this inammation progresses
maintenance may be abnormal in emphysema. Reduced into a more complicated inammatory pattern of adaptive
VEGF and VEGF receptors in the lungs of patients with immunity and involves T- and B-lymphocytes and possibly
COPD and, in an animal model, that interruption of dendritic cells along with a complicated interacting array of
VEGF signaling can lead to the development of emphy- cytokines and other mediators [31].
sema through apoptosis [25]. Fibroblasts from patients with
COPD proliferate more slowly [18] and are decient in
several in vitro measures of tissue repair [26]. Differences from asthma
Histopathological studies of COPD show a predominant
Pathogenesis: Integrating concepts involvement of peripheral airways (bronchioles) and lung
parenchyma, whereas asthma involves inammation in all
Taken together, the currently available data suggest that airways (particularly proximal airways) but usually without
the majority of COPD results from exposures to noxious involvement of the lung parenchyma [32]. In COPD there
agents. These, in turn, lead to activation of inammatory is narrowing of bronchioles, with brosis and inltration
processes within the lower respiratory tract. Damage to with macrophages and T-lymphocytes, along with destruc-
lung structures results both from the exposures and, more tion of lung parenchyma and an increased number of mac-
importantly, from the ensuing inammatory responses. rophages and T-lymphocytes, with a greater increase in
Inammation and injury also activate repair responses. CD8 than CD4 (helper) cells [27] (Fig. 34.6). Bronchial
Some individuals, either on a genetic or on a developmen- biopsies show similar changes with an inltration of mac-
tal basis, appear to be particularly sensitive to the injurious rophages and CD8 cells and an increased number of
eects of exposures and inammation. Similarly, individuals neutrophils in patients with severe COPD. BAL uid and
likely dier in their ability to repair lung injury, dierences induced sputum demonstrate a marked increase in macro-
which reect both genetic and acquired (e.g. nutritional) phages and neutrophils. In contrast to asthma, eosinophils
heterogeneity. The following sections will review briey the are not prominent except during exacerbations or when
cells and mediators believed to play a role in COPD. patients have concomitant asthma [32, 33]. In contrast to
the epithelial fragility of asthma, the airway epithelium in
patients with COPD may be metaplastic, as a result of the
COPD AS AN INFLAMMATORY DISEASE chronic release of epithelial growth factors such as epithelial
growth factor (EGF) from these cells. Another dierence
between COPD and asthma is the pattern of brosis, with
Both the small airway remodeling and narrowing and the subepithelial brosis (sometimes called basement mem-
emphysema are believed to be due to chronic inammation brane thickening) as a very characteristic feature of asthma,

429
Asthma and COPD: Basic Mechanisms and Clinical Management

Cigarette smoke
(and other irritants)

Epithelial cells Macrophage


CXCL9
CXCL10
TGF- CXCL11
FGF TNF- CXCL1 CCL2
IFN IL-1 CXCL8
Fibroblast IL-6 CCR2
CXCR3 CXCR2
Th1
Tc1 Monocyte
CTGF Neutrophil

Perforin Neutrophil elastase


Proteases
MMP-9

EGF
TGF-
Fibrosis Alveolar wall destruction
Mucus hypersecretion
(small airways) (emphysema)

FIG. 34.4 Inflammatory cells and mediators involved in COPD. Inhaled cigarette smoke and other irritants activate epithelial cells and macrophages to
release several chemotactic factors that attract inflammatory cells to the lungs, including CC-chemokine ligand 2 (CCL2), which acts on CC-chemokine
receptor 2 (CCR2) to attract monocytes, CXC-chemokine ligand 1 (CXCL1) and CXCL8, which act on CXCR2 to attract neutrophils and monocytes (which
differentiate into macrophages in the lungs) and CXCL9, CXCL10, and CXCL11, which act on CXCR3 to attract T helper 1 (Th1) cells and type 1 cytotoxic
T-cells (Tc1 cells), both of which release interferon- (IFN-). These inflammatory cells together with macrophages and epithelial cells release proteases, such
as matrix metalloproteinase 9 (MMP9), which cause elastin degradation and emphysema. Neutrophil elastase also causes mucus hypersecretion. Epithelial
cells and macrophages also release transforming growth factor- (TGF-) and fibroblast growth factors (FGF), which stimulate fibroblast proliferation,
resulting in fibrosis in the small airways as well as the proinflammatory cytokines tumor necrosis factor- (TNF-), IL-1 and IL-6 which amplify inflammation.
Mucus hypersecretion is stimulated by epithelial growth factor (EGF) and TGF-.




Neutrophils
Inflammation

Macrophages
 Cytokines
Mediators
Proteases


0
Non-smokers Normal Mild Severe Exacerbation
smokers COPD COPD

FIG. 34.5 Amplification of lung inflammation in COPD. Normal smokers have a mild inflammatory response, which represents the normal (probably
protective) reaction of the respiratory mucosa to chronic inhaled irritants. In COPD this same inflammatory response is markedly amplified, and this
amplification increases as the disease progresses. It is further increased during exacerbations triggered by infective organisms. The molecular mechanisms
of this amplification are currently unknown, but may be determined by genetic factors or possibly latent viral infection. Oxidative stress is an important
amplifying mechanism and may increase the expression of inflammatory genes through impairing the activity of histone deacetylase-2 (HDAC2), which is
needed to switch off inflammatory genes.

430
Pathophysiology of COPD 34
Asthma COPD
Cigarette smoke
Allergens

Y Y
Y

Ep cells Dendritic cell Mast cell Alv macrophage Ep cells


FIG. 34.6 Differences in inflammation between
COPD and asthma. Left panel shows that
inflammation in asthma is driven by allergens
resulting in activation of mast cells, dendritic cells
and epithelial cells and leading to the recruitment
CD4 cell Eosinophil CD8 cell Neutrophil of T helper 2 (Th2) cells and eosinophils. This
(Th2) (Tc1) results in airway hyperresponsiveness, the defining
physiologic defect in asthma. Right panel shows
Bronchoconstriction Small airway narrowing
Alveolar destruction that COPD is driven by inhaled irritants such as
AHR
cigarette smoke, which results in activation of
epithelial cells and macrophages, which recruit
neutrophils and T-lymphocytes into the lung,
Airflow limitation resulting in small airway narrowing (chronic
obstructive bronchiolitis) and alveolar destruction
Reversible Irreversible (emphysema).

whereas in COPD brosis is found mainly around small give misleading results. Nonetheless, there is a progressive
airways (peribronchiolar brosis). The airway smooth mus- increase in the numbers of inammatory cells in small air-
cle layer in asthma may be thickened as a result of hypertro- ways and lung parenchyma as COPD becomes more severe
phy and hyperplasia of airway smooth muscle cells, whereas even though the patients with most severe obstruction have
this layer is not very much increased in patients with stopped smoking for many years [27], indicating the exist-
COPD [34]. Mucus hypersecretion is a feature of asthma ence of mechanisms which perpetuate the inammatory
and COPD, with hyperplasia of goblet cells and submucosal reaction in COPD. This is in contrast to many other chronic
glands [35] (see Chapter 17). inammatory diseases, such as rheumatoid arthritis, where
Although the inammatory picture in asthma and the inammation tends to diminish in severe disease. The
COPD diers, in patients with severe asthma there are inammation of COPD lungs involves both innate immu-
greater similarities between the two diseases, with increased nity (neutrophils, macrophages, eosinophils, mast cells, NK
neutrophils, CD8 cells, and similar patterns of mediators cells, -T-cells, and dendritic cells) and adaptive immunity
in severe asthma as in COPD [31]. (T- and B-cells).
The inammation in the airways of COPD patients
does not seem to disappear after smoking cessation, even
after a period of several years [36, 37].
INFLAMMATORY CELLS
Epithelial cells
For many years, it was believed that the inammatory reac-
tion in the lungs of smokers consisted of neutrophils and Epithelial cells are activated by cigarette smoke to pro-
macrophages and those proteinases from these cells were duce inammatory mediators, including tumor necrosis
responsible for the lung destruction in COPD. More factor (TNF)-, interleukin (IL)-1, IL-6, granulocyte-
recently it has been recognized that there is a prominent T- macrophage colony-stimulating factor (GM-CSF) and
cell inltration in the lungs of patients with COPD, with CXCL8 (IL-8). Epithelial cells in small airways may be an
a predominance of CD8 T-cells, although CD4 (helper) important source of TGF-, which then induces local bro-
T-cells are also numerous. Although abnormal numbers of sis. Airway epithelial cells are also important in defense of
inammatory cells have been documented in COPD, the the airways, with mucus production from goblet cells, and
relationship between these cell types and the sequence of secretion of antioxidants, antiproteases, and defensins (see
their appearance and their persistence are not yet under- Chapter 16). It is possible that cigarette smoke and other
stood in detail [28]. Most studies have been cross-sectional, noxious agents impair these innate and adaptive immune
based on selection of patients with dierent stages of the responses of the airway epithelium, increasing susceptibil-
disease, and comparisons have been made between smokers ity to infection. The airway epithelium in chronic bronchitis
without airow limitation (normal smokers) and those with and COPD often shows squamous metaplasia, which may
COPD who have smoked a similar amount. There are no result from increased proliferation of basal airway epithe-
serial studies; selection biases (such as selecting tissue from lial cells but the nature of the growth factors involved in
patients suitable for lung volume reduction surgery) may epithelial cell proliferation, cell cycle, and dierentiation in

431
Asthma and COPD: Basic Mechanisms and Clinical Management

COPD are not yet known. Epidermal growth factor recep- LTB4, CXCL1, CXCL8
tors (EGFR) show increased expression in airway epithelial
cells of smokers and may contribute to basal cell prolifera- Neutrophil
tion, resulting in squamous metaplasia and an increased risk LTB4, CXCL8
.O2
of bronchial carcinoma [38].
MPO
Serine proteases
Mesenchymal cells MMPs

Mesenchymal cells, including broblasts, myobroblasts,


and smooth muscle cells, are the major structural cells Mucus
responsible for the production and maintenance of the Emphysema hypersecretion
interstitial connective tissue. As such, they play a major
role in the tissue remodeling that characterizes the struc-
tural alterations of COPD. In addition, mesenchymal cells FIG. 34.7 Neutrophils in COPD. Neutrophils are attracted to the lungs of
can also produce inammatory mediators and may play COPD patients by neutrophil chemotactic mediators, such as leukotriene
a role in persistent inammation in asthma and possibly B4 (LTB4) and the chemokines CXCL1 (also known as GRO-) and CXCL8
COPD. Fibroblasts from the lung parenchyma of patients (also known as IL-8). Activated neutrophils release more LTB4 and CXCL8
with COPD proliferate more slowly [18] and are less active to attract even more neutrophils, reactive oxygen species, such as
in chemotaxis and contraction of extracellular matrix [26]. superoxide anions (O2), and myeloperoxidase (MPO). They also release the
serine proteinases NE, cathpsin G, and proteinase-3, as well as the matrix
This suggests that decient repair could contribute to the
metalloproteinases (MMP)-8 and MMP-9. These proteinases stimulate
development of emphysema. Conversely, a pro-brotic mucus hypersecretion and elastolysis, resulting in emphysema.
phenotype has been reported in broblasts from various
brotic diseases [39, 40]. Whether such cells play a role in
the airways brosis in COPD is unknown.
Neutrophils secrete serine proteases, including NE, cathep-
sin G, and proteinase-3, as well as matrix metalloproteinase
Neutrophils (MMP)-8 and MMP-9, which may contribute to alveolar
destruction. Neutrophils have the capacity to induce tissue
Increased numbers of activated neutrophils are found in damage through the release of serine proteases and oxidants.
sputum and BAL uid of patients with COPD [41], yet are However, while neutrophils have the capacity to cause elas-
relatively little increased in the airways or lung parenchyma. tolysis, this is not a prominent feature of other pulmonary
This may reect their rapid transit through the airways and diseases where chronic airway neutrophilia is even more
parenchyma. The role of neutrophils in COPD is not yet prominent, including cystic brosis and bronchiectasis. This
clear, however, neutrophil numbers in induced sputum are suggests that other factors are involved in the generation of
correlated with COPD disease severity [41] and with the emphysema. Indeed neutrophils are not a prominent feature
rate of decline in lung function [42]. Smoking has a direct of parenchymal inammation in COPD. It is likely that
stimulatory eect on granulocyte production and release airway neutrophilia is more linked to mucus hypersecretion
from the bone marrow and survival in the respiratory in chronic bronchitis. Serine proteases from neutrophils,
tract, possibly mediated by GM-CSF and G-CSF released including NE, cathepsin G, and proteinase-3 are all potent
from lung macrophages [43]. Smoking may also increase stimulants of mucus secretion from submucosal glands and
intravascular neutrophil retention in the lung. Neutrophil goblet cells in the epithelium. There is a marked increase in
recruitment to the airways and parenchyma involves adhe- neutrophil numbers in the airways in acute exacerbations of
sion to endothelial cells and E-selectin which is up-regu- COPD accounting for the increased purulence of sputum.
lated on endothelial cells in the airways of COPD patients This may reect increased production of neutrophil chemo-
[44]. Adherent neutrophils then migrate into the respiratory tactic factors including LTB4 and CXCL8 [47, 48].
tract under the direction of neutrophil chemotactic factors.
There are several chemotactic signals that have the potential
for neutrophil recruitment in COPD, including leukotriene Macrophages
(LT)B4, CXCL8, and related CXC chemokines, includ-
ing CXCL1 (GRO-) and CXCL5 (ENA-78), which are Macrophages appear to play a pivotal role in the pathophysi-
increased in COPD airways [45] (Fig. 34.7). These media- ology of COPD and can account for most of the known
tors may be derived form alveolar macrophages T-cells and features of the disease [49] (Fig. 34.8). There is a marked
epithelial cells, but the neutrophil itself may be a major increase (5- to 10-fold) in the numbers of macrophages
source of CXCL8. Neutrophils from the circulation mar- in airways, lung parenchyma, BAL uid, and sputum in
ginate in the pulmonary circulation and adhere to endothe- patients with COPD. A careful morphometric analysis of
lial cells in the alveolar wall before passing into the alveolar macrophage numbers in the parenchyma of patients with
space. The neutrophils recruited to the airways of COPD emphysema showed a 15-fold increase in the numbers of
patients are activated as there is increased concentration macrophages in the tissue and alveolar space compared with
of granule proteins, such as myeloperoxidase (MPO) and normal smokers [50]. Furthermore, macrophages are local-
human neutrophil lipocalin, in the sputum supernatant [46]. ized to sites of alveolar wall destruction in patients with

432
Pathophysiology of COPD 34
Cigarette smoke
Wood smoke

Numbers
Secretion

ROS FIG. 34.8 Macrophages in COPD. Macrophages


Peroxynitrite CTGF
may play a pivotal role in COPD as they are activated
NO by cigarette smoke extract and secrete many
TGF-1 Fibrosis
inflammatory proteins that may orchestrate the
LTB4 inflammatory process in COPD. Neutrophils may
CXCL8 Elastolysis be attracted by CXCL8, CXCL1 and leukotriene B4
CXCL1 CCL2
CCL10 MMP-9, MMP-12 (LTB4), monocytes by CCL2, and CD8 lymphocytes
CXCL1 CCL11 by CXCL10 and CXCL11. Release of elastolytic
CXCR2 Cathepsins K,L,S
CXCR2 CCL12 CXCR3 enzymes including matrix metalloproteinases (MMP)
and cathepsins cause elastolysis, and release of
transforming growth factor (TGF)-1 and connective
tissue growth factor (CTGF). Macrophages also
Emphysema generate reactive oxygen species (ROS) and nitric
Neutrophils Monocytes CD8  cells oxide (NO) which together form peroxynitrite and
may contribute to steroid resistance.

emphysema and there is a correlation between macrophage The increased numbers of macrophages in COPD are
numbers in the parenchyma and severity of emphysema [51]. mainly be due to increased recruitment of monocytes, as
Macrophages may be activated by cigarette smoke extract to macrophages have a very low proliferation rate in the lungs.
release inammatory mediators including TNF-, CXCL8 Macrophages have a long survival time so this is dicult to
and other CXC chemokines, CCL2 (MCP-1), LTB4, and measure directly. However, in macrophages from smokers,
reactive oxygen species (ROS), providing a cellular mecha- there is markedly increased expression of the anti-apoptotic
nism that links smoking with inammation in COPD. protein Bcl-XL and increased expression of p21CIP/WAF1 in
Alveolar macrophages also secrete elastolytic enzymes, the cytoplasm [56]. This suggests that macrophages may have
including MMP-2, MMP-9, MMP-12, cathepsins K, L and a prolonged survival in smokers and patients with COPD.
S, and NE taken up from neutrophils [52]. Alveolar mac- Once activated, macrophages will increase production of
rophages from patients with COPD secrete more inam- ROS, nitric oxide, and lysosomal enzymes and will increase
matory proteins and have a greater elastolytic activity at secretion of many cytokines, including TNF-, IL-1, IL-6,
baseline than those from normal smokers and this is further CXCL8, and IL-18 among others. This activated macro-
increased by exposure to cigarette smoke [52]. Macrophages phage response increases the eciency of killing of organisms
demonstrate this dierence even when maintained in culture but may promote tissue damage and further inammation.
for 3 days and therefore appear to be intrinsically dierent Corticosteroids are ineective in suppressing inam-
from the macrophages of normal smokers and non- mation, including cytokines, chemokines, and proteases, in
smoking normal control subjects [52]. The predominant patients with COPD [57]. In vitro the release of CXCL8,
elastolytic enzyme secreted by alveolar macrophages in TNF-, and MMP-9 macrophages from normal sub-
COPD patients is MMP-9. Most of the inammatory jects and normal smokers are inhibited by corticosteroids,
proteins that are up-regulated in COPD macrophages are whereas corticosteroids are ineective in macrophages from
regulated by the transcription factor nuclear factor-B (NF- patients with COPD [58]. The reasons for resistance to cor-
B) which is activated in alveolar macrophages of COPD ticosteroids in COPD and to a lesser extent macrophages
patients, particularly during exacerbations [53]. from smokers may be the marked reduction in activity of
The increased numbers of macrophages in smokers histone deacetylase-2 (HDAC2) [59], which is recruited to
and COPD patients may be due to increased recruitment activated inammatory genes by glucocorticoid receptors
of monocytes from the circulation in response to the mono- (GRs) to switch o inammatory genes. The reduction in
cyte-selective chemokines CCL2 and CXCL1, which are HDAC activity in macrophages is correlated with increased
increased in sputum and BAL of patients with COPD [45]. secretion of cytokines like TNF- and CXCL8 and reduced
Monocytes from patients with COPD show a greater chem- response to corticosteroids. The reduction of HDAC activ-
otactic response to GRO- than cells from normal smokers ity in COPD patients may be mediated through oxidative
and non-smokers, but this is not explained by an increase stress and peroxynitrite formation [60].
in CXCR2 [54]. Interestingly, while all monocytes express
CCR2, the receptor for CCL2, only ~30% of monocytes
express CXCR2. It is possible that these CXCR2 express- Eosinophils
ing monocytes transform into macrophages that are more
inammatory. Macrophages also release the chemokines While eosinophils are the predominant leukocyte in asthma,
CXCL9, CXCL10, and CXCL11, which are chemotactic their role in COPD is much less certain. In some studies,
for CD8 Tc1 and CD4 Th1 cells, via interaction with the increased numbers of eosinophils have been described in the
chemokine receptor CXCR3 expressed on these cells [55]. airways and BAL of patients with stable COPD, whereas

433
Asthma and COPD: Basic Mechanisms and Clinical Management

others have not found increased numbers in airway biop-


IFN-
sies, BAL or induced sputum. The presence of eosinophils in
patients with COPD predicts a response to corticosteroids
and may indicate coexisting asthma [33]. Increased numbers
of eosinophils have been reported in bronchial biopsies and
BAL uid during acute exacerbations of chronic bronchi-
Epithelial cells Macrophage
tis [61]. Surprisingly the levels of eosinophil basic proteins
CXCL9, CXCL10, CXCL11
in induced sputum are as elevated in COPD, as in asthma,
despite the absence of eosinophils, suggesting the eosinophils
may have degranulated and are no longer recognizable by
microscopy [46]. Perhaps this is due to the high levels of NE CXCR3
that have been shown to cause degranulation of eosinophils. Th1 cell Tc1 cell

Dendritic cells
Emphysema
Dendritic cells play a central role in the initiation of the (apoptosis of type I
Perforin
innate and adaptive immune response and it is believed Granzyme B
pneumocytes)
that they provide a link between them [62]. The airways
and lungs contain a rich network of dendritic cells that are FIG. 34.9 T-lymphocytes in COPD. Epithelial cells and macrophages
localized near the surface, so that they are ideally located are stimulated by interferon- (IFN-) to release the chemokines CXC-
to signal the entry of foreign substances that are inhaled. chemokine ligand 9 (CXCL9), CXCL10, and CXCL11, which together act on
Dendritic cells can activate a variety of other inamma- CXC-chemokine receptor 3 (CXCR3) expressed on T helper 1 (Th1) cells
tory and immune cells, including macrophages, neutrophils, and type 1 cytotoxic T (Tc1) cells to attract them into the lungs. Tc1 cells,
through the release of perforin and granzyme B, induce apoptosis of type 1
T- and B-lymphocytes so dendritic cells may play an impor-
pneumocytes, thereby contributing to emphysema. IFN- released by Th1
tant role in the pulmonary response to cigarette smoke and and Tc1 cells then stimulates further release of CXCR3 ligands, resulting in a
other inhaled noxious agents. However, there does not persistent inflammatory activation.
appear to be an increase in dendritic cells in the airways of
COPD patients, in contrast to asthma patients [63].

a receptor activated by the chemokines CXCL9 (Mig),


T-lymphocytes CXCL10 (IP-10), and CXCL11 (I-TAC), all of which
are increased in COPD [66]. There is increased expression
There is an increase in the total numbers of T-lymphocytes of CXCL10 by bronchiolar epithelial cells and this could
in lung parenchyma, peripheral and central airways of contribute to the accumulation of CD4 and CD8 T-
patients with COPD, with the greater increase in CD8 cells, which preferentially express CXCR3 [67] (Fig. 34.9).
than CD4 cells [27, 55]. There is a correlation between the CD8 cells are typically increased in airway infections and
numbers of T-cells and the amount of alveolar destruction it is possible that the chronic colonization of the lower
and the severity of airow obstruction. Furthermore, the respiratory tract of COPD patients by bacterial and viral
only signicant dierence in the inammatory cell inltrate pathogens is responsible for this inammatory response. It
in asymptomatic smokers and smokers with COPD is an is possible that cigarette-induce lung injury may uncover
increase in T-cells, mainly CD8, in patients with COPD. previously sequestered autoantigens or cigarette smoke
There is also an increase in the absolute number of CD4 itself may damage lung interstitial and structural cells and
T-cells albeit in smaller numbers, in the airways of smok- make them antigenic [68]. The role of increased numbers of
ers with COPD and these cells express activated STAT-4, a CD4 cells in COPD, particularly in severe disease is also
transcription factor that is essential for activation and com- unknown [50]; however, it is now clear that T-cell help is
mitment of the Th1 lineage and IFN- [64]. required for the priming of cytotoxic T-cell responses, for
The ratio of CD4 and CD8 cells are reversed in maintaining CD8 T-cell memory and for ensuring CD8
COPD. The majority of T-cells in the lung in COPD are of T-cell survival. It is also possible that CD4 T-cells have
the Tc1 and Th1 subtypes [55]. There is a marked increase immunological memory and play a role in perpetuating the
in T-cells in the walls of small airways in patients with inammatory process in the absence of cigarette smoking.
severe COPD and the T-cells are formed into lymphoid In a mouse model of cigarette-induced emphysema there is
follicles, surrounding B-lymphocytes [27]. a predominance of T-cells which are directly related to the
The mechanisms by which CD8, and to a lesser severity of emphysema [69].
extent CD4 cells, accumulate in the airways and paren- The role of T-cells in the pathophysiology of COPD
chyma of patients with COPD is not yet understood [65]. is not yet certain, although they have the potential to pro-
However, homing of T-cells to the lung must depend upon duce extensive damage in the lung. CD8 cells have the
some initial activation (only activated T-cells can home to capacity cause cytolysis and apoptosis of alveolar epithelial
the organ source of antigenic products), then adhesion and cells through release of perforins, granzyme B, and TNF-
selective chemotaxis. CD4 and CD8 T-cells in the lung [70, 71]. There is an association between CD8 cells and
of COPD patients show increased expression of CXCR3, apoptosis of alveolar cells in emphysema [72]. Apoptotic

434
Pathophysiology of COPD 34
cells are potential sources of antigenic material that could harmful eects including damage to lipids, proteins, and
reach the DC and perpetrate the T-cell response. In addi- DNA. There is increasing evidence that oxidative stress is
tion CD8 T-cells also produce a number of cytokines of an important feature in COPD [81] (see Chapter 25).
the Tc1 phenotype including TNF-, lymphotoxin, and Inammatory and structural cells that are activated in the
IFN-, and there is evidence that CD8 in the lungs of airways of patients with COPD produce ROS, including,
COPD patients expresses IFN- [73]. All these cytokines neutrophils, eosinophils, macrophages, and epithelial cells.
would enhance the inammatory reaction in the lung Superoxide anions (O2) are generated by NADPH oxidase
besides the direct killing by CD8 cells. COPD has been and converted to hydrogen peroxide (H2O2) by superoxide
considered an autoimmune disease triggered by smoking, dismutases (SODs). H2O2 is then dismuted to water by
as previously suggested [68] and the presence of highly catalase. O2 and H2O2 may interact in the presence of free
activated oligoclonal T-cells in emphysema patients sup- iron to form the highly reactive hydroxyl radical (OH). O2
ports this [74]. There is evidence for anti-elastin antibodies may also combine with NO to form peroxynitrite, which
in experimental models of COPD and in COPD patients also generates OH. Oxidative stress leads to the oxidation
[75]. In addition to activated Th1 cells there is some evi- of arachidonic acid and the formation of a new series of
dence for an increase in Th2 cells that express IL-4 in BAL prostanoid mediators called isoprostanes, which may exert
uid of COPD patients in COPD patients [76]. signicant functional eects including bronchoconstriction
NK cells are also increased in COPD lungs and may and plasma exudation [82] (Fig. 34.10).
contribute to epithelial cell apoptosis in emphysema [77]. The normal production of oxidants is counteracted by
Invariant NK T-cells are CD4 cells that release IL-4 but several antioxidant mechanisms in the human respiratory
are not increased in COPD [78]. tract [81]. The major intracellular antioxidants in the airways
are catalase, SOD, and glutathione, formed by the enzyme -
glutamyl cysteine synthetase, and glutathione synthetase. In
the lung intracellular antioxidants are expressed at relatively
MEDIATORS OF INFLAMMATION low levels and are not induced by oxidative stress, whereas the
major antioxidants are extracellular. A transcription factor Nrf2
plays a key role in switching on several genes for antioxidants
Many inammatory mediators have now been implicated in in response to oxidative stress and there is evidence that Nrf2
COPD, including lipids, free radicals, cytokines, chemok- knock-out mice are more susceptible to developing emphy-
ines, and growth factors [29]. These mediators are derived sema after cigarette smoke exposure [83, 84]. Extracellular
from inammatory and structural cells in the lung and antioxidants, particularly glutathione peroxidase, are mark-
interact with each other in a complex manner. edly up-regulated in response to cigarette smoke and oxida-
tive stress. Extracellular antioxidants also include the dietary
antioxidants vitamin C (ascorbic acid) and vitamin E
Lipid mediators (-tocopherol), uric acid, lactoferrin, and extracellular super-
oxide dismutase (SOD3), which is highly expressed in human
The prole of lipid mediators in exhaled breath condensates lung, but its role in COPD is not yet clear.
of patients with COPD shows an increase in prostaglandins ROS have several eects on the airways and paren-
and leukotrienes [79]. There is a signicant increase in chyma and increase the inammatory response. ROS
PGE2 and PGF2 and an increase in LTB4 but not cystei- activate NF-B, which switches on multiple inamma-
nyl leukotrienes. This is a dierent pattern to that seen in tory genes resulting in amplication of the inammatory
asthma, in which increases in thromboxane and cysteinyl response. The molecular pathways by which oxidative stress
leukotrienes have been shown. The increased production activates NF-B have not been fully elucidated, but there
of prostanoids in COPD is likely to be secondary to the are several redox-sensitive steps in the activation pathway.
induction of cyclo-oxygenase-2 (COX2) by inammatory Oxidative stress results in activation of histone acetyltrans-
cytokines and increased expression of COX2 is found in ferase activity which opens up the chromatin structure
alveolar macrophages of COPD patients. LTB4 concentra- and is associated with increased transcription of multiple
tions are also increased in induced sputum and are further inammatory genes [85]. Exogenous oxidants may also
increased in sputum and exhaled breath condensate during be important in worsening airway disease. There is con-
acute exacerbations [47]. LTB4 is a potent chemoattractant siderable evidence for increased oxidative stress in COPD
for neutrophils, acting through high anity BLT1- [81]. Cigarette smoke itself contains a high concentration
receptors. A BLT1-receptor antagonist reduces the neu- of ROS. Inammatory cells, such as activated macrophages
trophil chemotactic activity of sputum by ~25% [80]. and neutrophils, also generate ROS, as discussed earlier.
Recently BLT1-receptors have been identied on T-lym- There are several markers of oxidative stress that may be
phocytes and there is evidence that LTB4 is involved in detected in the breath and several studies have demon-
recruitment of T-cells. strated increased production of oxidants, such as H2O2,
8-isoprostane, and ethane, in exhaled air or breath conden-
sates, particularly during exacerbations [47].
Oxidative stress The increased oxidative stress in the lung epithelium
of COPD patient may play an important pathophysi-
Oxidative stress occurs when ROS are produced in excess ological role in the disease by amplifying the inammatory
of the antioxidant defense mechanisms and result in response in COPD. This may reect the activation of

435
Asthma and COPD: Basic Mechanisms and Clinical Management

Cigarette smoke
Inflammatory cells

Anti-proteases
SLPI 1-AT NF-B FIG. 34.10 Oxidative stress in COPD. Oxidative
stress plays a key role in the pathophysiology
of COPD and amplifies the inflammatory and
Proteolysis destructive process. Reactive oxygen species
CXCL8 TNF-
from cigarette smoke or from inflammatory cells
O2, H2O2 (particularly macrophages and neutrophils) result
OH., ONOO Neutrophil in several damaging effects in COPD, including
Steroid recruitment decreased anti-protease defenses, such as
resistance
1-antitrypsin (AT) and secretory leukoprotease
inhibitor (SLPI), activation of nuclear factor-B
(NF-B) resulting in increased secretion of the
cytokines CXCL8 and tumor necrosis factor-
Bronchoconstriction (TNF-), increased production of isoprostanes and
Isoprostanes Plasma leak direct effects on airway function. In addition recent
Mucus secretion evidence suggests that oxidative stress induces
steroid resistance.

Normal COPD

Cigarette smoke
Stimuli
Corticosteroids
Alveolar Oxidative stress
macrophage
Peroxynitrite

GR
NF-B NF-B

HDAC2 HDAC2
Histone Histone
acetylation acetylation

TNF- Histone acetylation


IL-8 TNF-
MMP-9 CXCL8
MMP-9

FIG. 34.11 Mechanism of corticosteroid resistance in COPD. Stimulation of normal alveolar macrophages activates nuclear factor-B (NF-B) and other
transcription factors to switch on histone acetyltransferase leading to histone acetylation and subsequently to transcription of genes encoding inflammatory
proteins, such as tumor necrosis factor- (TNF-) and CXCL8 (IL-8). Corticosteroids reverse this by binding to glucocorticoid receptors (GRs) and recruiting
histone deacetylase-2 (HDAC2). This reverses the histone acetylation induced by NF-B and switches off the activated inflammatory genes. In COPD patients
cigarette smoke activates macrophages, as in normal subjects, but oxidative stress (acting in part through the formation of peroxynitrite) impairs the activity
of HDAC2. This amplifies the inflammatory response to NF-B activation, but also reduces the anti-inflammatory effect of corticosteroids as HDAC2 is now
unable to reverse histone acetylation.

NF-B and AP-1, which then induce a neutrophilic inam- the anti-inammatory eects of corticosteroids, compared
mation via increased expression of CXC chemokines, to cells from normal smokers and non-smokers [58]. In
TNF-, and MMP-9. Oxidative stress may also impair patients with COPD there is a marked reduction in activ-
the function of antiproteases such as 1-antitrypsin and ity of HDAC and reduced expression of HDAC2 in alveolar
SLPI, and thereby accelerates the breakdown of elastin in macrophages and peripheral lung tissue [59], which is cor-
lung parenchyma. Corticosteroids are much less eective in related with increased expression of inammatory cytokines
COPD than in asthma and do not reduce the progression or and a reduced response to corticosteroids. This may result
mortality of the disease. Alveolar macrophages from patients directly or indirectly from oxidative stress and is mimicked
with COPD show a marked reduction in responsiveness to by the eects of H2O2 in cell lines [86] (Fig. 34.11).

436
Pathophysiology of COPD 34
Nitrative stress of CXCR2 in monocytes of COPD patients [54]. CXCL5
shows a marked increase in expression in airway epithelial
The increase in exhaled nitric oxide (NO) is less marked in cells during exacerbations of COPD and this is accompa-
COPD than in asthma, partly because cigarette smoking nied by a marked up-regulation of epithelial CXCR2.
reduces exhaled NO. Recently exhaled NO has been par- CCL2 is increased in concentration in COPD spu-
titioned into central and peripheral portions and this shows tum and BAL uid [45] and plays a role in monocyte
reduced NO in the bronchial fraction but increased NO in chemotaxis via activation of CCR2. CCL2 appears to
the peripheral fraction, which includes lung parenchyma cooperate with CXCL1 in recruiting monocytes into the
and small airways [87] (see Chapter 30). The increased lungs. The chemokine CCL5 (RANTES) is also expressed
peripheral NO in COPD patients may reect increased in airways of COPD patients during exacerbations and acti-
expression of inducible NO synthase in epithelial cells and vates CCR5 on T-cells and CCR3 on eosinophils, which
macrophages of patients with COPD [88]. NO and super- may account for the increased eosinophils and T-cells in
oxide anions combine to form peroxynitrite which nitrates the wall of large airways that have been reported during
certain tyrosine residues in proteins and there is increased exacerbations of chronic bronchitis. As discussed earlier,
expression of 3-nitrotyrosine in peripheral lung and macro- CXCR3 are up-regulated on Tc1 and Th1 cells of COPD
phages of COPD patients [88]. There is tyrosine nitration patients with increased expression of their ligands CXCL9,
of HDAC2 which may lead to impaired activity and degra- CXCL10, and CXCL11.
dation of this enzyme, resulting in steroid resistance [86].
Growth factors
Inflammatory cytokines
Several growth factors have been implicated in COPD and
Cytokines are the mediators of chronic inammation and mediate the structural changes that are found in the airways
several have now been implicated in COPD [29, 89]. There (see Chapter 29). TGF-1 is expressed in alveolar macro-
is an increase in concentration of TNF- in induced spu- phages and airway epithelial cells of COPD patients and
tum in stable COPD with a further increase during exacer- is released from epithelial cells of small airways. TGF-
bations [41, 48]. TNF- production from peripheral blood is released in a latent form and is activated by various fac-
monocytes is also increased in COPD patients and has tors including MMP-9. TGF- may play an important
been implicated in the cachexia and skeletal muscle apop- role in the characteristic peribronchiolar brosis of small
tosis found in some patients with severe disease. TNF- airways, either directly or through the release of connective
is a potent activator of NF-B and this may amplify the tissue growth factor (Fig. 34.12). TGF- down-regulates
inammatory response. Unfortunately anti-TNF therapies 2-adrenergic receptors by inhibiting gene transcription
have not proved to be eective in COPD patients. IL-1 in human cell lines and may reduce the bronchodilator
and IL-6 are proinammatory cytokines that may amplify
the inammation in COPD and may be important for sys-
temic eects.
Bronchiolar
Macrophages
epithelial cells
Chemokines
Chemokines are small chemotactic cytokines that play a
key role in the recruitment and activation if inammatory
cells through specic chemokine receptors. Several chem- Latent TGF-
okines have now been implicated in COPD and have been
of particular interest since chemokine receptors are G- MMP-9
protein coupled receptors, for which small molecule antago- TGF-
nists have now been developed [30, 89]. CXCL8 concentra-
tions are increased in induced sputum of COPD patients CTGF 2-Adrenoceptors
and increase further during exacerbations [41, 48]. CXCL8
is secreted from macrophages, T-cells, epithelial cells, and
neutrophils. CXCL8 activates neutrophils via low an- Bronchiole
ity specic receptors CXCR1, and is chemotactic for neu- Airway
trophils via high anity receptors CXCR2, which are also smooth muscle
activated by related CXC chemokines such as CXCL1. Peribronchiolar fibrosis
CXCL1 concentrations are markedly elevated in sputum and (chronic obstructive bronchiolitis)
BAL uid of COPD patients and this chemokine may be
FIG. 34.12 Transforming growth factor (TGF)- in COPD. TGF- is released
more important as a chemoattractant than CXCL8, acting in a latent form that may be activated by matrix metalloproteinase-9
via CXCR2 which are expressed on neutrophils and mono- (MMP-9). It may then cause fibrosis directly through effects on fibroblasts
cytes [45]. CXCL1 induces signicantly more chemotaxis of or indirectly via the release of connective tissue growth factor (CTGF). TGF-
monocytes of COPD patient compared to those of normal may also down-regulate 2-adrenoceptors on cells such as airway smooth
smokers and this may reect increased turnover and recovery muscle to diminish the bronchodilator response to -agonists.

437
Asthma and COPD: Basic Mechanisms and Clinical Management

1-Antitrypsin
SLPI
Neutrophils Cigarette smoke Elafin
TIMPs
Oxidants TACE
Neutrophil elastase
Cathepsins
TGF- MMPs (1, 2, 9,12)
Others........
EGF
EGFR

MAP kinases

FIG. 34.14 Protease-anti-protease imbalance in COPD. In COPD the


balance appears to be tipped in favor of increased proteolysis, either
MUC5AC, MUC5B Goblet cell because of an increase in proteases, including NE, cathepsins, and matrix
Mucus hyperplasia metalloproteinases (MMP), or a deficiency in antiproteases, which may
MUC5AC
include 1-antitrypsin, elafin, secretory leukoprotease inhibitor (SLPI), and
MUC5B Submucosal gland tissue inhibitors of matrix metalloproteinases (TIMPs).

FIG. 34.13 Epidermal growth factor receptors (EGFR) in COPD. EGFR activated T-cells, which acts by increasing endothelial cell
play a key role in the regulation of mucus hypersecretion, with increased permeability, by inducing expression of endothelial adhesion
expression of mucin genes (MUC5AC, MUCB) and differentiation of goblet molecules and via its ability to act as a monocyte chemoat-
cells and hyperplasia of mucus-secreting cells. These effects are mediated tractant. VEGF also stimulates the expression of CXCL10
via the activation of mitogen-activated protein (MAP) kinases. EGFR and its receptor CXCR3. Thus VEGF is acts as an interme-
are activated by transforming growth factor- (TGF-), which is in turn diary between cell-mediated immune inammation and the
activated by tumor necrosis factor- converting enzyme (TACE), activated associated angiogenesis reaction.
via release of oxidants from cigarette smoke and neutrophils. EGFR may
also be activated by EGF.
Proteinases
response to -agonists in airway smooth muscle. TGF- An imbalance between proteinases that degrade connec-
is also a potent anti-inammatory molecule and TGF-1 tive tissue and anti-proteinases has long been postulated
decient mice die early from overwhelming inammation, in COPD (Fig. 34.14). There has been debate about which
which occurs even in germ-free animals [90]. In contrast, are the most important proteinases involved in emphysema.
mice decient in Smad3, a TGF signal transducing mol- While previous attention focused on NE, more recently
ecule are resistant the development of brotic lung disease a role of MMP has been recognized (see Chapter 28).
but spontaneously develop emphysema [91]. MMP-9 appears to be particularly important in degrad-
Alveolar macrophages produce TGF- in greater ing elastin bers in lung parenchyma and is secreted in
amounts than TGF- and this may be a major endogenous large amounts by alveolar macrophages of COPD patients
activator of epithelial growth factor receptors (EGFR) that [52, 96]. MMP-9 also activates TGF- accounting for the
play a key role in regulating mucus secretion in response to simultaneous occurrence of lung destruction and brosis
many stimuli, including cigarette smoke. Cigarette smoke in COPD (Fig. 34.15). In the mouse, MMP-12 appears
activates TNF- converting enzyme on airway epithelial to play a key role in the development of cigarette smoke-
cells which results in the shedding of TGF- and the acti- induced emphysema as MMP-12 derived elastin fragments
vation of EGFR, resulting in increased mucus secretion [92, are major factors driving macrophage recruitment [97]. A
93] (Fig. 34.13). role for MMP-1 has also been suggested in both human
VEGF is a major regulator of vascular growth and is and animal studies [98].
likely to be involved in the pulmonary vascular remodeling
that occurs as a result of hypoxic pulmonary vasoconstriction
in severe COPD. There is increased expression of VEGF in
pulmonary vascular smooth muscle of patients with mild and
moderate COPD but paradoxically a reduction in expression
SYSTEMIC MANIFESTATIONS
in severe COPD with emphysema [94]. Inhibition of VEGF
receptors using a selective inhibitor induces apoptosis of It is now recognized that COPD involves organs out-
alveolar endothelial cells in rats, resulting in emphysema and side the lung, but the mechanism for these systemic
this appears to be driven by oxidative stress [25] and may eects of COPD are not well understood (see Chapter
be mediated via the sphingolipid ceramide [95]. In addition, 44). Inammatory cytokines, such as TNF- and IL-6,
VEGF is also an important proinammatory cytokine pro- may spill over from the lung periphery into the systemic
duced by epithelial and endothelial cells, macrophages, and circulation, where they may have detrimental eects on

438
Pathophysiology of COPD 34
Macrophage Chemotactic Neutrophils
peptides
Neutrophil
1-AT elastase

Pro-MMP-9 MMP-9
Emphysema
Elastolysis

Latent TGF- Active TGF-

Small airway fibrosis


(chronic obstructive bronchiolitis)

FIG. 34.15 Connective tissue destruction and fibrosis coexist in COPD. Matrix metalloproteinase-9 (MMP-9) is the major elastolytic enzyme released by
alveolar macrophages and is generated from an inactive precursor pro-MMP-9. MMP-9 causes elastolysis directly, but also releases chemotactic peptides
that attract neutrophils, which release neutrophil elastase. The elastolytic effect of neutrophil elastase is enhanced as MMP-9 inactivates its specific anti-
proteinase 1-antitrypsin, resulting in emphysema. MMP-9 also activates transforming growth factor- (TGF-) from a latent precursor and this leads to
fibrosis of small airways. TGF- also plays a role in activating MMP-9, so the interplay between these mediators leads to tissue destruction and fibrosis
simultaneously, as observed in COPD lungs.

CONCLUSIONS

Cigarette smoke exposure induces a orid inamma-


tory response in the lung involving structural and inam-
matory cells and a large array of inammatory mediators.
The interaction of these complex steps eventually leads to
IL-6 IL-6, TNF-, IL-1 airway remodeling and obstruction and emphysema, albeit
Liver variable among individual smokers. Of interest, the main
Circulation
CRP dierence between smokers who develop COPD and the
ones who do not seems to be the presence of an adaptive
Skeletal Other immune response with CD8, CD4, and B-cells, which
muscle inflammatory express obvious signs of being activated eector cells. It is
diseases likely that genetic and epigenetic factors (such as histone
Cardiovascular disease Muscle wasting acetylation) are involved in determining the progression
of the inammatory cascade, as this is supported by ani-
FIG. 34.16 Systemic features of COPD and co-morbidities. Inflammation mal models, where dierent strains appear to have dier-
in the lungs may spill over into the systemic circulation resulting in
ent sensitivities to cigarette smoke. COPD is a complex
systemic inflammatory effects and exacerbation of cardiovascular diseases
and skeletal muscle wasting. Increased concentrations of interleukin-6
inammatory disease and the interactions between dier-
(IL-6) and tumor necrosis factor- (TNF-) are found in systemic circulation ent inammatory cells and mediators are still uncertain. The
of patients with severe COPD and these may directly result in skeletal role played by structural cells in responding to inammatory
muscle atrophy. IL-6 releases the acute phase protein C-reactive protein cell-mediated injury and the degree to which it is decient
(CRP) which is associated with increased cardiovascular disease risk [101]. in COPD remains to be fully dened. More research into
Cigarette smoke also contributes to increased cardiovascular risk. these mechanisms is needed in order to identify novel tar-
gets that may lead to the discovery of more eective thera-
pies that are able to prevent disease progression and reduce
the high mortality of this common disease [102].
skeletal muscle function and endothelial function [99]
(Fig. 34.16). IL-6 causes the release of the acute phase pro- References
tein C-reactive protein, which is increased in the circulation
of patients with COPD and correlates with disease severity 1. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P et al.
[100]. Skeletal muscle wasting is a feature of severe COPD Global strategy for the diagnosis, management, and prevention of
and this may in part be due to systemic inammation but COPD-2006 update. Am J Respir Crit Care Med 176: 53255, 2007.
is more likely due to disuse atrophy as a result of impaired 2. Mannino DM, Buist AS. Global burden of COPD: Risk factors, preva-
exercise performance. lence, and future trends. Lancet 370: 76573, 2007.

439
Asthma and COPD: Basic Mechanisms and Clinical Management

3. Lokke A, Lange P, Scharling H, Fabricius P, Vestbo J. Developing 26. Togo S, Holz O, Liu X, Sugiura H, Kamio K, Wang X et al. Lung
COPD: A 25 year follow up study of the general population. Thorax broblast repair functions in COPD patients are altered by multiple
61: 93539, 2006. mechanisms. Am J Respir Crit Care Med, 2008.
4. Rennard SI, Vestbo J. COPD: The dangerous underestimate of 15%. 27. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L et al.
Lancet 367: 121619, 2006. The nature of small-airway obstruction in chronic obstructive pulmo-
5. ODonnell DE, Laveneziana P. Dyspnea and activity limitation in nary disease. N Engl J Med 350: 264553, 2004.
COPD: Mechanical factors. COPD 4: 22536, 2007. 28. Barnes PJ, Shapiro SD, Pauwels RA. Chronic obstructive pulmonary
6. Makita H, Nasuhara Y, Nagai K, Ito Y, Hasegawa M, Betsuyaku T disease: Molecular and cellular mechanisms. Eur Respir J 22: 67288,
et al. Characterisation of phenotypes based on severity of emphysema 2003.
in chronic obstructive pulmonary disease. Thorax 62: 93237, 2007. 29. Barnes PJ. Mediators of chronic obstructive pulmonary disease.
7. Sturton G, Persson C, Barnes PJ. Small airways: An important but Pharmacol Rev 56: 51548, 2004.
neglected target in the treatment of obstructive airway diseases. Trends 30. Donnelly LE, Barnes PJ. Chemokine receptors as therapeutic targets
Pharmacol Sci, 2008. in chronic obstructive pulmonary disease. Trends Pharmacol Sci 27:
8. Zhang JJ, Smith KR. Household air pollution from coal and biomass 54653, 2006.
fuels in China: Measurements, health impacts, and interventions. 31. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary
Environ Health Perspect 115: 84855, 2007. disease. Nat Rev Immunol 8: 18392, 2008.
9. Yin P, Jiang CQ, Cheng KK, Lam TH, Lam KH, Miller MR et al. 32. Fabbri LM, Romagnoli M, Corbetta L, Casoni G, Busljetic K, Turato G
Passive smoking exposure and risk of COPD among adults in China: et al. Dierences in airway inammation in patients with xed air-
The Guangzhou Biobank Cohort Study. Lancet 370: 75157, 2007. ow obstruction due to asthma or chronic obstructive pulmonary
10. Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA. disease. Am J Respir Crit Care Med 167: 41824, 2003.
Relationship between exacerbation frequency and lung function 33. Papi A, Romagnoli M, Baraldo S, Braccioni F, Guzzinati I, Saetta M
decline in chronic obstructive pulmonary disease. Thorax 57: 84752, et al. Partial reversibility of airow limitation and increased exhaled
2002. NO and sputum eosinophilia in chronic obstructive pulmonary disease.
11. Sethi S, Murphy TF. Bacterial infection in chronic obstructive pulmo- Am J Respir Crit Care Med 162: 177377, 2000.
nary disease in 2000: A state-of-the-art review. Clin Microbiol Rev 14: 34. Jeery PK. Comparison of the structural and inammatory features of
33663, 2001. COPD and asthma. Chest 117: 251S260S, 2000.
12. Marossy AE, Strachan DP, Rudnicka AR, Anderson HR. Childhood 35. Rogers DF. The role of airway secretions in COPD: Pathophysiology,
chest illness and the rate of decline of adult lung function between ages epidemiology and pharmacotherapeutic options. COPD 2: 34153,
35 and 45 years. Am J Respir Crit Care Med 175: 35559, 2007. 2005.
13. Higashimoto Y, Elliott WM, Behzad AR, Sedgwick EG, Takei T, 36. Willemse BW, ten Hacken NH, Rutgers B, Lesman-Leegte IG,
Hogg JC et al. Inammatory mediator mRNA expression by adeno- Postma DS, Timens W. Eect of 1-year smoking cessation on airway
virus E1A-transfected bronchial epithelial cells. Am J Respir Crit Care inammation in COPD and asymptomatic smokers. Eur Respir J 26:
Med 166: 2007, 2002. 83545, 2005.
14. Petrache I, Diab K, Knox KS, Twigg HL III, Stephens RS, Flores S 37. Gamble E, Grootendorst DC, Hattotuwa K, OShaughnessy T,
et al. HIV associated pulmonary emphysema: A review of the literature Ram FS, Qiu Y et al. Airway mucosal inammation in COPD is simi-
and inquiry into its mechanism. Thorax 63: 46369, 2008. lar in smokers and ex-smokers: A pooled analysis. Eur Respir J 30:
15. Fletcher C, Peto R. The natural history of chronic airow obstruction. 46771, 2007.
BMJ 1: 164548, 1977. 38. Franklin WA, Veve R, Hirsch FR, Helfrich BA, Bunn PA Jr. Epidermal
16. Morla M, Busquets X, Pons J, Sauleda J, Macnee W, Agusti AG. growth factor receptor family in lung cancer and premalignancy. Semin
Telomere shortening in smokers with and without COPD. Eur Respir J Oncol 29: 314, 2002.
27: 52528, 2006. 39. Mio T, Nagai S, Kitaichi M, Kawatani A, Izumi T. Proliferative charac-
17. Nyunoya T, Monick MM, Klingelhutz A, Yarovinsky TO, Cagley JR, teristics of broblast lines derived from open lung biopsy specimens of
Hunninghake GW. Cigarette smoke induces cellular senescence. Am J patients with IPF (UIP). Chest 102: 83237, 1992.
Respir Cell Mol Biol 35: 68188, 2006. 40. Miki H, Mio T, Nagai S, Hoshino Y, Nagao T, Kitaichi M et al.
18. Holz O, Zuhlke I, Jaksztat E, Muller KC, Welker L, Nakashima M Fibroblast contractility: Usual interstitial pneumonia and nonspecic
et al. Lung broblasts from patients with emphysema show a reduced interstitial pneumonia. Am J Respir Crit Care Med 162: 225964, 2000.
proliferation rate in culture. Eur Respir J 24: 57579, 2004. 41. Keatings VM, Collins PD, Scott DM, Barnes PJ. Dierences in
19. Muller KC, Welker L, Paasch K, Feindt B, Erpenbeck VJ, Hohlfeld JM interleukin-8 and tumor necrosis factor-a in induced sputum from
et al. Lung broblasts from patients with emphysema show markers of patients with chronic obstructive pulmonary disease or asthma. Am J
senescence in vitro. Respir Res 7: 32, 2006. Respir Crit Care Med 153: 53034, 1996.
20. Sato A, Hirai T, Imura A, Kita N, Iwano A, Muro S et al. 42. Stanescu D, Sanna A, Veriter C, Kostianev S, Callagni PG, Fabbri LM
Morphological mechanism of the development of pulmonary emphy- et al. Airways obstruction, chronic expectoration and rapid decline in
sema in klotho mice. Proc Natl Acad Sci USA 104: 236165, 2007. FEV1 in smokers are associated with increased levels of sputum neu-
21. Sato T, Seyama K, Sato Y, Mori H, Souma S, Akiyoshi T et al. trophils. Thorax 51: 26771, 1996.
Senescence marker protein-30 protects mice lungs from oxidative 43. Vlahos R, Bozinovski S, Hamilton JA, Anderson GP. Therapeutic
stress, aging and smoking. Am J Respir Crit Care Med 174: 5307, potential of treating chronic obstructive pulmonary disease (COPD) by
2006. neutralising granulocyte macrophage-colony stimulating factor (GM-
22. Rennard SI, Togo S, Holz O. Cigarette smoke inhibits alveolar repair: CSF). Pharmacol Ther 112: 10615, 2006.
A mechanism for the development of emphysema. Proc Am Thorac Soc 44. Di Stefano A, Maestrelli P, Roggeri A, Turato G, Calabro S, Potena A
3: 7038, 2006. et al. Upregulation of adhesion molecules in the bronchial mucosa of
23. Sahebjami H, Domino M. Eects of starvation and refeeding on subjects with chronic obstructive bronchitis. Am J Respir Crit Care Med
elastase-induced emphysema. J Appl Physiol 66: 261116, 1989. 149: 80310, 1994.
24. Laurent P, Jano A, Kagan HM. Cigarette smoke blocks cross-linking 45. Traves SL, Culpitt S, Russell REK, Barnes PJ, Donnelly LE. Elevated
of elastin in vitro. Chest 83: 63S65S, 1983. levels of the chemokines GRO-a and MCP-1 in sputum samples from
25. Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S, COPD patients. Thorax 57: 59095, 2002.
Hirth PK et al. Inhibition of VEGF receptors causes lung cell 46. Keatings VM, Barnes PJ. Granulocyte activation markers in induced
apoptosis and emphysema. J Clin Invest 106: 131119, 2006. sputum: Comparison between chronic obstructive pulmonary disease,

440
Pathophysiology of COPD 34
asthma and normal subjects. Am J Respir Crit Care Med 155: 44953, 67. Saetta M, Mariani M, Panina-Bordignon P, Turato G, Buonsanti C,
1997. Baraldo S et al. Increased expression of the chemokine receptor
47. Biernacki WA, Kharitonov SA, Barnes PJ. Increased leukotriene B4 CXCR3 and its ligand CXCL10 in peripheral airways of smokers with
and 8-isoprostane in exhaled breath condensate of patients with exac- chronic obstructive pulmonary disease. Am J Respir Crit Care Med 165:
erbations of COPD. Thorax 58: 29498, 2003. 14049, 2002.
48. Aaron SD, Angel JB, Lunau M, Wright K, Fex C, Le Saux N et al. 68. Cosio MG, Majo J, Cosio MG. Inammation of the airways and lung
Granulocyte inammatory markers and airway infection during acute parenchyma in COPD: Role of T cells. Chest 121: 160S65S, 2002.
exacerbation of chronic obstructive pulmonary disease. Am J Respir Crit 69. Takubo Y, Guerassimov A, Ghezzo H, Triantallopoulos A, Bates JH,
Care Med 163: 34955, 2001. Hoidal JR et al. Alpha1-antitrypsin determines the pattern of emphy-
49. Barnes PJ. Macrophages as orchestrators of COPD. COPD 1: 5970, sema and function in tobacco smoke-exposed mice: Parallels with
2004. human disease. Am J Respir Crit Care Med 166: 1596603, 2002.
50. Retamales I, Elliott WM, Meshi B, Coxson HO, Pare PD, Sciurba FC 70. Hashimoto S, Kobayashi A, Kooguchi K, Kitamura Y, Onodera H,
et al. Amplication of inammation in emphysema and its associa- Nakajima H. Upregulation of two death pathways of perforin/
tion with latent adenoviral infection. Am J Respir Crit Care Med 164: granzyme and FasL/Fas in septic acute respiratory distress syndrome.
46973, 2001. Am J Respir Crit Care Med 161: 23743, 2000.
51. Meshi B, Vitalis TZ, Ionescu D, Elliott WM, Liu C, Wang XD et al. 71. Chrysofakis G, Tzanakis N, Kyriakoy D, Tsoumakidou M, Tsiligianni I,
Emphysematous lung destruction by cigarette smoke. The eects of Klimathianaki M et al. Perforin expression and cytotoxic activity of
latent adenoviral infection on the lung inammatory response. Am J sputum CD8 lymphocytes in patients with COPD. Chest 125: 7176,
Respir Cell Mol Biol 26: 5257, 2002. 2004.
52. Russell RE, Thorley A, Culpitt SV, Dodd S, Donnelly LE, Demattos C 72. Majo J, Ghezzo H, Cosio MG. Lymphocyte population and apoptosis
et al. Alveolar macrophage-mediated elastolysis: Roles of matrix metal- in the lungs of smokers and their relation to emphysema. Eur Respir J
loproteinases, cysteine, and serine proteases. Am J Physiol Lung Cell Mol 17: 94653, 2001.
Physiol 283: L867lL873, 2002. 73. Cosio MG. T-lymphocytes. In: Barnes PJ (ed.), Chronic Obstructive
53. Caramori G, Romagnoli M, Casolari P, Bellettato C, Casoni G, Pulmonary disease: Cellular and Molecular mechanisms, pp. 32125. New
Boschetto P et al. Nuclear localisation of p65 in sputum macrophages York: Taylor & Francis Group, 2005.
but not in sputum neutrophils during COPD exacerbations. Thorax 58: 74. Sullivan AK, Simonian PL, Falta MT, Mitchell JD, Cosgrove GP,
34851, 2003. Brown KK et al. Oligoclonal CD4 T cells in the lungs of patients
54. Traves SL, Smith SJ, Barnes PJ, Donnelly LE. Specic CXC but not with severe emphysema. Am J Respir Crit Care Med 172: 59096, 2005.
CC chemokines cause elevated monocyte migration in COPD: A role 75. Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S
for CXCR2. J Leukoc Biol 76: 44150, 2004. et al. Antielastin autoimmunity in tobacco smoking-induced emphy-
55. Grumelli S, Corry DB, Song L-X, Song L, Green L, Huh J et al. An sema. Nat Med 13: 56769, 2007.
immune basis for lung parenchymal destruction in chronic obstructive 76. Barczyk A, Pierzchala W, Kon OM, Cosio B, Adcock IM,
pulmonary disease and emphysema. PLoS Med 1: 7583, 2004. Barnes PJ. Cytokine production by bronchoalveolar lavage T lym-
56. Tomita K, Caramori G, Lim S, Ito K, Hanazawa T, Oates T et al. phocytes in chronic obstructive pulmonary disease. J Allergy Clin
Increased p21CIP1/WAF1 and B cell lymphoma leukemia-xL expres- Immunol 117: 148492, 2006.
sion and reduced apoptosis in alveolar macrophages from smokers. Am 77. Fairclough L, Urbanowicz RA, Corne J, Lamb JR. Killer cells in
J Respir Crit Care Med 166: 72431, 2002. chronic obstructive pulmonary disease. Clin Sci (Lond) 114: 53341,
57. Keatings VM, Jatakanon A, Worsdell YM, Barnes PJ. Eects of 2008.
inhaled and oral glucocorticoids on inammatory indices in asthma 78. Vijayanand P, Seumois G, Pickard C, Powell RM, Angco G, Sammut
and COPD. Am J Respir Crit Care Med 155: 54248, 1997. D et al. Invariant natural killer T cells in asthma and chronic obstruc-
58. Culpitt SV, Rogers DF, Shah P, de Matos C, Russell RE, tive pulmonary disease. N Engl J Med 356: 141022, 2007.
Donnelly LE et al. Impaired inhibition by dexamethasone of cytokine 79. Montuschi P, Kharitonov SA, Ciabattoni G, Barnes PJ. Exhaled leuko-
release by alveolar macrophages from patients with chronic obstructive trienes and prostaglandins in COPD. Thorax 58: 58588, 2003.
pulmonary disease. Am J Respir Crit Care Med 167: 2431, 2003. 80. Beeh KM, Kornmann O, Buhl R, Culpitt SV, Giembycz MA,
59. Ito K, Ito M, Elliott WM, Cosio B, Caramori G, Kon OM et al. Barnes PJ. Neutrophil chemotactic activity of sputum from patients
Decreased histone deacetylase activity in chronic obstructive pulmo- with COPD: Role of interleukin 8 and leukotriene B4. Chest 123:
nary disease. N Engl J Med 352: 196776, 2005. 124047, 2003.
60. Barnes PJ. Reduced histone deacetylase in COPD: Clinical implica- 81. Rahman I. Oxidative stress in pathogenesis of chronic obstructive
tions. Chest 129: 15155, 2006. pulmonary disease: Cellular and molecular mechanisms. Cell Biochem
61. Zhu J, Qiu YS, Majumdar S, Gamble E, Matin D, Turato G et al. Biophys 43: 16788, 2005.
Exacerbations of Bronchitis: Bronchial eosinophilia and gene expres- 82. Montuschi P, Barnes PJ, Roberts LJ. Isoprostanes: Markers and media-
sion for interleukin-4, interleukin-5, and eosinophil chemoattractants. tors of oxidative stress. FASEB J 18: 1791800, 2004.
Am J Respir Crit Care Med 164: 10916, 2001. 83. Rangasamy T, Cho CY, Thimmulappa RK, Zhen L, Srisuma SS,
62. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: Kensler TW et al. Genetic ablation of Nrf2 enhances susceptibility to
Linking innate and adaptive immunity in asthma. Nat Rev Immunol 8: cigarette smoke-induced emphysema in mice. J Clin Invest 114: 1248
193204, 2008. 59, 2004.
63. Rogers AV, Adelroth E, Hattotuwa K, Dewar A, Jeery PK. Bronchial 84. Ishii Y, Itoh K, Morishima Y, Kimura T, Kiwamoto T, Iizuka T et al.
mucosal dendritic cells in smokers and ex-smokers with COPD: An Transcription factor Nrf2 plays a pivotal role in protection against
electron microscopic study. Thorax, 2007. elastase-induced pulmonary inammation and emphysema. J Immunol
64. Di Stefano A, Caramori G, Capelli A, Gnemmi I, Ricciardolo F, 175: 696875, 2005.
Oates T et al. STAT4 activation in smokers and patients with chronic 85. Tomita K, Barnes PJ, Adcock IM. The eect of oxidative stress on his-
obstructive pulmonary disease. Eur Respir J 24: 7885, 2004. tone acetylation and IL-8 release. Biochem Biophys Res Commun 301:
65. Barnes PJ, Cosio MG. Characterization of T lymphocytes in chronic 57277, 2003.
obstructive pulmonary disease. PLoS Med 1: 2527, 2004. 86. Ito K, Tomita T, Barnes PJ, Adcock IM. Oxidative stress reduces his-
66. Costa C, Runo R, Traves SL, Silva LE, Barnes PJ, Donnelly LE. tone deacetylase (HDAC)2 activity and enhances IL-8 gene expres-
CXCR3 and CCR5 chemokines in the induced sputum from patients sion: Role of tyrosine nitration. Biochem Biophys Res Commun 315:
with COPD. Chest 133: 2633, 2008. 24045, 2004.

441
Asthma and COPD: Basic Mechanisms and Clinical Management

87. Brindicci C, Ito K, Resta O, Pride NB, Barnes PJ, Kharitonov SA. 96. Russell RE, Culpitt SV, DeMatos C, Donnelly L, Smith M, Wiggins J
Exhaled nitric oxide from lung periphery is increased in COPD. Eur et al. Release and activity of matrix metalloproteinase-9 and tis-
Respir J 26: 5259, 2005. sue inhibitor of metalloproteinase-1 by alveolar macrophages from
88. Ricciardolo FL, Caramori G, Ito K, Capelli A, Brun P, Abatangelo G patients with chronic obstructive pulmonary disease. Am J Respir Cell
et al. Nitrosative stress in the bronchial mucosa of severe chronic obstruc- Mol Biol 26: 6029, 2002.
tive pulmonary disease. J Allergy Clin Immunol 116: 102835, 2005. 97. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD.
89. Barnes PJ. Cytokine networks in asthma and chronic obstructive pul- Requirement for macrophage metalloelastase for cigarette smoke-
monary disease. J Clin Invest, 2008. induced emphysema in mice. Science 277: 20024, 1997.
90. Boivin GP, Ormsby I, Jones-Carson J, OToole BA, Doetschman T. 98. Imai K, Dalal SS, Chen ES, Downey R, Schulman LL, Ginsburg M
Germ-free and barrier-raised TGF beta 1-decient mice have similar et al. Human collagenase (matrix metalloproteinase-1) expression in
inammatory lesions. Transgenic Res 6: 197202, 1997. the lungs of patients with emphysema. Am J Respir Crit Care Med
91. Bonniaud P, Kolb M, Galt T, Robertson J, Robbins C, Stampi M et al. 163: 691, 2001.
Smad3 null mice develop airspace enlargement and are resistant to TGF- 99. Agusti A. Chronic obstructive pulmonary disease: A systemic disease.
beta-mediated pulmonary brosis. J Immunol 173: 2099108, 2004. Proc Am Thorac Soc 3: 47881, 2006.
92. Shao MX, Nakanaga T, Nadel JA. Cigarette smoke Induces MUC5AC 100. van Eeden SF, Sin DD. Chronic obstructive pulmonary disease: A
mucin overproduction via tumor necrosis factor-a converting enzyme chronic systemic inammatory disease. Respiration 75: 22438, 2008.
in human airway epithelial (NCI-H292) cells. Am J Physiol Lung Cell 102. Barnes PJ, Hansel TT. Prospects for new drugs for chronic obstructive
Mol Physiol 287: L420L427, 2004. pulmonary disease. Lancet 364: 98596, 2004.
93. Burgel PR, Nadel JA. Roles of epidermal growth factor receptor activa- 101. Rabe KF, Beghe B, Luppi F, Fabbri LM. Update in chronic obstruc-
tion in epithelial cell repair and mucin production in airway epithe- tive pulmonary disease 2006. Am J Respir Crit Care Med 175: 1222
lium. Thorax 59: 99296, 2004. 32, 2007.
94. Rothenberg ME. VEGF obstructs the lungs. Nat Med 10: 104142,
2004.
95. Petrache I, Natarajan V, Zhen L, Medler TR, Richter AT, Cho C et al.
Ceramide upregulation causes pulmonary cell apoptosis and emphy-
sema-like disease in mice. Nat Med 11: 49198, 2005.

442
Triggers of 6
PART

Asthma and COPD


Allergens
35 CHAPTER

Donald W. Cockcroft
by Noon in 1911 [6, 7]. Prausnitz and Kustner
HISTORY disproved the toxin hypothesis by the demon- Division of Respirology, Critical Care
stration of passive transfer of allergic sensitivity and Sleep Medicine, Department of
using the injection of the serum of a sensitized Medicine, University of Saskatchewan,
Introduction
individual into the skin of a nonsensitized sub- Royal University Hospital, Saskatoon,
Allergy, that is type I IgE-mediated sensitization ject [8] the basis of the so-called PK test. Saskatchewan, Canada
and sequelae to predominantly inhaled allergens, Coca demonstrated that a heat labile serum fac-
has become increasingly recognized as important tor that could not be precipitated from serum
in the pathogenesis of asthma. The development by the usual methods was the case of this sen-
of this increased recognition is briey outlined sitivity and, in 1923, he coined the terms atopy
focusing on identication of allergy and its (strange disease) and atopic reagin as the name
mechanisms, on the relevance of allergen in the for this serum factor [9]. Reaginic antibody was
pathogenesis of asthma, and on increased preva- eventually identied as IgE by Ishizaka in 1967
lence of the condition(s). [10]. The complete allergen response, including
allergen presenting cells, lymphocytes (T-helper
2 or Th2 and B cells), mast cells, basophils, and
Allergy mediators, is still being elucidated.

Although rose catarrh (likely actually due to


grass pollen) had been known for some time, the Allergy and asthma
rst comprehensive clinical description of aller-
gic rhinitis is attributed to John Bostock in 1819 The recognition of allergens as potential causes
[1]. The disease was given the medical name of asthma parallels the recognition of allergy.
catarrhus aestivus (summer cold) but was rec- Classic asthma symptoms were a major compo-
ognized in the lay literature as hay fever, a label nent of Bostocks original description of catarrhus
which persists to this day. This disease was felt to aestivus [1]. Pollen, both ragweed [11] and grass
be limited to the middle and upper classes, and [2], was specically shown to provoke attacks of
a major prevailing hypothesis was that it was asthma in the 1870s. Cat, horse, and house-dust
due to the eects of summer heat and/or sun- mite were identied as relevant asthma produc-
shine [1]. Grass pollen had been suggested as a ing allergens in the early part of the 20th century
potential cause, but this was not conrmed until [12], and more recently fungal spore sensitivity
the elegant experiments of Charles Blackley in has been identied as a risk factor for asthma
1873 [2]. The term allergy (dierent from nor- [13]. Asthma became lumped with the atopic
mal) was coined by Von Pirquet in 1906 [3]. In diseases by many in the early part of the 20th
1912, allergy skin testing was rst used in the century. However, the very inuential Oslers
investigation of this type of hypersensitivity [4]. textbook had stated that asthma was a neurosis
With the mistaken belief that pollen contained [14], an opinion that continued to hold weight
a toxin, the concept of pollen immunization for many years. The opposing view, namely that
was introduced by Dunbar [5] and popularized asthma was primarily an atopic allergic disease,

445
Asthma and COPD: Basic Mechanisms and Clinical Management

was argued by Cooke and colleagues [15]. This controversy


persisted well into the 20th century. The identication of ATOPY
allergen-induced late asthmatic responses [16], allergen-
induced airway hyperresponsiveness1 [17], and allergen- Atopy is the tendency to develop IgE antibodies to com-
induced airway inammation [18], that is features that dene monly encountered environmental allergens by natural expo-
clinical asthma, has led to allergens becoming recognized as sure in which the route of entry of allergen is across intact
important position in the pathogenesis of asthma. Although mucosal surfaces [26]. The recognized familial nature of atopy
still a subject of some controversy [19], many investigators is due to complex (multiple gene) inheritance (genetic het-
now regard the majority, that is 7580%, of nonselected asth- erogeneity) [21, 27]. The remarkable relatively short-term
matics as being atopic [20, 21]. increase in atopic prevalence indicates that environmental
factors are also important [2225]. The pathophysiologic basis
for subjects developing atopy is uncertain and is the topic of
a recent symposium [28]. An old hypothesis favoring aller-
Prevalence gen handling perhaps at the mucosal surface [29] rather than
increased capacity to produce IgE has not been excluded.
There has been a striking increase in the prevalence of The prevalence of atopy in random populations,
allergic rhinitis from the beginning to the end of the 19th dened as the presence of positive(s) on prick skin testing
century [22]. Within this time, frame allergic disease has with a small battery of common relevant allergens, ranges
gone from rare in the early 1800s to a common disorder from 30% to almost 50% [3032]. The peak period of sen-
by 1900. A case can be made that this is at least in part sitization is in the third decade; thereafter the prevalence
related to earlier under-recognition of a condition whose falls [26]. Our experience with a random young population
symptoms (rhinitis, wheezing) resemble other conditions would suggest that about 50% of atopic (skin test positive)
believed to have been more common, and whose symp- subjects will have symptoms referable to atopy, which will
toms are often so mild as to potentially go unreported at include asthma in about 50% [32]. Thus, atopy is common,
a time when there were many more serious diseases to be aecting about one in three, with about one in six having
concerned with. As early as 1873, Blackley noted the ris- symptomatic atopy and about one in twelve having atopic
ing prevalence of allergic disease and felt that this could asthma; the prevalences will be proportionately higher in
not adequately be explained by increased recognition populations with a higher prevalence of atopy.
which he admitted did play a role [2]. The prevalence of
atopic disease and asthma in particular has continued to
increase throughout the 20th century with more objective
data to support these trends. The reasons for the remark-
able increase in the prevalence of atopy and asthma are not
ALLERGENS
completely understood. Genetics play a role in determining
the presence and severity of atopy, however, large changes Inhaled complete allergens that provoke asthma by IgE-
in prevalence over a short period cannot be due to genet- mediated mechanisms are soluble organic high molecular
ics and therefore must be due to one or more environmen- weight (20,00040,000 MW) protein or protein-containing
tal factors. One hypothesis is that increased exposure to molecules, which may be derived from any phylum of either
allergens, particularly indoor allergens in modern air-tight the plant or the animal kingdoms (including bacteria) [33,
homes, may lead to increased atopy and asthma [23]. This 34]. The structural characteristics that make a protein aller-
has been demonstrated prospectively in Papua New Guinea genic have been recently reviewed [35]. In clinical nonoc-
where blanket introduction was followed by house-dust cupational settings, the important inhalant allergens fall
mites, house-dust mite sensitivity, and an asthma epidemic into four groups; pollen, fungal spores, animal danders, and
[24]. Another intriguing hypothesis is the hygiene hypoth- household arthropods (mite and insects) [36].
esis [25]. This hypothesis suggests that early childhood Pollen allergens that trigger asthma are predominantly
infections drive the immune system toward the T helper from wind-pollinated plants, namely trees, grass, and weeds
1 (Th1) paradigm that suppresses or reduces the Th2 arm [36]. The relevant allergens and seasonal uctuations will vary
of the immune system. Control/prevention of infections with geography and climate, with tree pollens predominant in
will remove this Th2 suppression and lead to an increased spring months, grasses in summer, and weeds in late summer
prevalence of Th2-related atopic disease. The evidence for and autumn [34]. Although whole pollen grains may have
this, including family size studies and epidemiologic studies limited access to the lower respiratory tract [37], the relation-
related to infection, is stronger for atopy than for asthma ship of pollen to clinical asthma is convincing [36].
alone [25]. Other factors related to lifestyle issues including Atmospheric fungal spores of many groups of fungi are
diet and level of activity may be important [23]. smaller and more respirable than pollen, and are recognized
as causing atopic sensitization [13]. Their role in trigger-
ing asthma is less certain than pollen [34, 36]. Fungal spore
types and seasons will also vary with geographic and climatic
1
The term airway (hyper)responsiveness throughout this chapter, unless (temperature and humidity) conditions. Fungal spores are
otherwise stated, refers to the nonallergic (hyper)responsiveness to hista- associated with decaying vegetation resulting in a late sum-
mine, cholinergic agonists, exercise, etc., which is a characteristic feature of mer and autumn peak for common fungal spores, Alternaria,
symptomatic asthma. Cladosporium, Aspergillus, Sporobolomyces, etc. [34]. A spring

446
Allergens 35
peak for atmospheric fungal spores may be seen in some areas 4.5
Inhalation
especially where late melting of snow cover leads to so-called
snow mold [34]. Thus, atmospheric fungal spores may be
responsible for fall or springfall asthma symptoms. Fungi 4.0
may also be present inside living areas in moist basements,
food storage areas, and waste receptacles [34]. Aspergillus may
cause a distinct clinical syndrome, allergic bronchopulmonary
aspergillosis, which will be covered separately. 3.5
Household animals [34], particularly cats and dogs,
but also small mammals (gerbils, hamsters, rabbits, etc.) and
birds, may release allergens in secretions (e.g. saliva) or excre- 3.0
tions (e.g. urine, feces). Large animals, particularly horses, Diluent
may also provoke atopic sensitivity. House dust, due to its Ragweed
content of mite antigens from various Dermatophagoides spe-
2.5
cies or insect antigens such as cockroach [38], is an impor-

FEV1 (l)
tant source of atopic sensitization. Dermatophagoides spp., in
particular, are likely the most important cause of atopic sen- Inhalation
sitization worldwide. Again, climatic conditions are impor- 3.0
tant, since areas of low indoor relative humidity do not favor
growth of house-dust mites [34, 39].
Other allergens are encountered less frequently, often
2.5
in occupational settings, and include various plant parts
[castor bean, cocoa bean, tobacco leaf, psyllium (laxative),
vegetable gums, etc.], insect dusts, bacterial enzymes, and in
the very highly sensitized even atmospheric molecular levels 2.0
of foods (e.g. cooking sh) [34].
Diluent
1.5 Grass

INHALED ALLERGENS 1.0

0 1 2 3 4 5 6 7 8
Patterns of airway response H

Airway responses to inhaled allergens have been assessed FIG. 35.1 Early and dual asthmatic responses to allergen. The top graph
by the somewhat articial inhalation tests in the laboratory shows an isolated EAR following ragweed pollen inhalation and the
with aqueous allergen extracts [17, 40, 41]. Nevertheless, the bottom graph a dual asthmatic response (in another subject) following
results of such challenges, especially the late sequelae, appear grass pollen inhalation. Modified and reproduced from Ref. [49] with
to be clinically relevant [42, 43], and allergen inhalation permission.
tests allow study of both the pharmacology and the patho-
physiology of allergen-induced asthma. Airway responses
to allergen can be divided into early and late sequelae.
The early asthmatic response (EAR) is an episode of air- following both experimental [17, 41, 42, 47] and natural
ow obstruction which is maximal 1020 min after allergen [42, 50, 51] allergen exposure. This is correlated with the
inhalation and resolves spontaneously in 12 h [17, 40, 41, occurrence and severity of the late response, often appear-
44]. The late sequelae include the late asthmatic response ing with small, previously ignored, late responses (515%
(LAR) [16, 17, 40, 41, 4346], airway hyperresponsiveness FEV1 (1-s forced expiratory volume) fall) [17, 41]. Airway
[17, 41, 42], recurrent nocturnal asthma [46], and airway responsiveness develops between 2 [52] and 3 h [5355]
inammation [18, 47]. The LAR is an episode of airow after exposure, is present at 78 h [17, 41, 52], and may
obstruction which develops after spontaneous resolution persist for days, occasionally worsening despite the return
of the EAR between 3 and 5 h after exposure, occasionally of airway caliber to baseline [41] (Fig. 35.2). As expected
earlier, rarely later [17, 40, 41,43,44,45]. Resolution usually [56], the increased airway responsiveness is associated with
begins by 68 h but may require in excess of 12 h [41, 43, symptoms of asthma [17, 41], including recurrent nocturnal
44]. Modest late responses respond well to bronchodila- asthma [46]. Both the LAR [18, 47] and the increased air-
tors [48]; unpublished observations suggest bronchodila- way responsiveness [47] are associated with increases in air-
tors may be required often (e.g. up to 2 h). More severe late way inammation. The occurrence of seasonal increases in
airway obstruction is not always completely reversible by airway responsiveness [42, 50, 51], and airway inammation
bronchodilator [44]. Examples of early and late responses [57] provides support for both the relevance of the bron-
are shown in Fig. 35.1. Allergen-induced increase in air- choprovocation model and the importance of the inam-
way responsiveness (e.g. to histamine/methacholine) occurs mation in the pathogenesis of the late sequelae.

447
Asthma and COPD: Basic Mechanisms and Clinical Management

Ragweed uninuenced by anticholinergics [75]. The enhanced airway


3.7
responsiveness to histamine following allergen inhalation is
no more responsive to atropine than it was prior to allergen
inhalation [76].
FEV1 (l)

Ingested theophylline oers partial protection against


both the EAR and the LAR [7780] and variable protec-
tion against the induced airway hyperresponsiveness [78
80]. It is not clear whether this is a functional antagonist or
an anti-inammatory eect.
2.7 Inhaled sodium cromoglycate (SCG) given prior to
allergen exposure inhibits both the EAR and LAR [44, 45,
58, 61, 78, 81], as well as the allergen-induced increased
responsiveness to both histamine [61] and methacholine [78].
10
Nedocromil sodium appears to have similar eects on aller-
Histamine PC20 (mg/ml)

gen-induced asthmatic responses [82]. SCG given after the


EAR will slightly delay but not inhibit the LAR [67].
The leukotriene pathway can be altered in several ways.
Leukotriene receptor antagonists (LTRAs) are the most com-
monly used leukotriene modiers. LTRAs provide protection
against both EAR [8385] and LAR [83, 84]. In addition,
Smoke 5-lipoxygenase inhibitors [86] or 5-lipoxygenase activating
1.0 Symptoms Cold air protein inhibitors [87] provide some protection. Recently, the
Exercise combination of desloratadine (H1 blocker) and montelukast
(LTRA) has shown a synergistic eect regarding inhibition of
0 1 2 3 4 5 6 7 8 9 10
the EAR [85]; hence, studies on the LAR are required.
Days
A single dose of inhaled corticosteroid, given prior to
FIG. 35.2 Allergen-induced increase in nonallergic airway responsiveness allergen, has no inuence on the EAR but provides eective
to inhaled histamine. A dual asthmatic response, with spontaneous and often complete inhibition of the LAR [44, 45, 58, 61,
recovery, occurred after a single inhalation of ragweed pollen extract. 66, 67, 81, 82, 89, 90, 91, 92]. A single dose given after the
Airway responsiveness to inhaled histamine, expressed as the provocation EAR will inhibit the LAR [67]. Longer treatment periods
concentration causing a 20% FEV1 fall (PC20), increased after allergen with inhaled corticosteroids will partially inhibit the EAR
exposure, and was associated with asthma symptoms on exposure to as well [68, 91, 92]. Corticosteroid-induced improvement
nonallergic stimuli. Reproduced from Ref. [49] with permission.) in airway responsiveness [68, 93, 94] provides only partial
explanation [68]. Reduction in mucosal mast cells [9597]
is likely more important.
Pharmacology H1 blockers partially inhibit the early portion of the
EAR [45, 58, 98100]. Newer H1 blockers may also show
Pharmacologic inhibition of airway responses to inhaled some inhibition of the LAR [100]; hence, further studies
allergen has been studied for both its therapeutic relevance are necessary. Ingested antiallergic drugs such as ketotifen
and further understanding of the pathophysiology of the and repirinast have produced variable eects on allergen-
responses. induced asthma [101107]. Most studies have failed to
Inhaled 2-agonists are the best inhibitors of the EAR show any signicant protection [101, 102, 106, 107]. Non-
[45, 5861] due to a combination of eects on smooth mus- steroidal anti-inammatory agents (cyclooxygenase inhibi-
cle and on mediator release [62, 63]. Despite the latter, inter- tors), particularly indomethacin, appear to have no eect or
mediate acting inhaled 2-agonists (salbutamol, terbutaline, perhaps enhance the EAR [108]; there is conicting evi-
etc.) do not inhibit the LAR [45, 58, 59, 61] or the increased dence regarding the late response [88, 109, 110]. Allergen-
airway responsiveness [61]. The long acting inhaled 2- induced increase in airway responsiveness appears to be
agonists salmeterol and formoterol completely inhibit or partially inhibited by indomethacin [88]. A thromboxane
mask all aspects of the allergen-induced airway response [64, synthetase inhibitor had no eect on allergen-induced early
65], likely due to functional antagonism rather than anti- or late responses or increased airway responsiveness [111].
inammatory eect [65]. Regular use of inhaled 2-agonists A platelet-activating factor antagonist proved ineective
for a week or more increases the EAR [66, 68, 69], the LAR against allergen-induced asthma [112]. Inhaled furosemide
[69, 70, 184], mast cell mediator release [69], and allergen- provides inhibition of both EAR and LAR [113]. Allergen
induced airway inammation [69, 70]. A larger dose of aller- injection therapy has produced variable results in modulat-
gen can be administered after an inhaled 2-agonist and will ing the EAR [114, 115] but may be particularly eective
lead to a larger LAR [71]. These features, failure to inhibit versus the LAR [116]. A novel recombinant anti-IgE mole-
the LAR, enhanced airway responses, and ability to tolerate cule directed against the Fc component of IgE is very eec-
a larger dose of allergen, may be relevant in 2-agonist-wors- tive at inhibiting both the EAR [117] and the LAR [118].
ened asthma control [72]. Anti-IL-5 [119] and IL-12 [120] suppressed allergen-
Muscarinic blockers cause variable minor inhibition induced eosinophilia with little eect on allergen-induced
of the EAR [60, 7375] and no inhibition of the LAR [73, airways hyperresponsiveness (AHR) casting some doubt on
75]. Allergen-induced airway hyperresponsiveness appears the causal relationship between inammation and AHR.
448
Allergens 35
Mechanisms [20, 21], the correlation of both airway hyperresponsiveness
and asthma with atopy in epidemiologic population studies
The mechanisms of allergen-induced asthmatic responses [32,139144] (Fig. 35.4), the relationship of both seasonal
have been studied in humans by indirect means. Animal [42, 50, 51, 145, 146] and indoor [23, 24, 39] allergen expo-
studies, in vitro studies on excised human tracheobronchial sure to symptoms and airway hyperresponsiveness, and their
smooth muscle, drug-inhibition studies and, more recently,
bronchoalveolar lavage (BAL), and induced sputum, have
Allergen
all been used to assess mechanisms. The EAR is due to 
the allergen-IgE mast cell acute mediator release includ- IgE
ing histamine [121], prostaglandins [122], and leukotrienes
[123] and is primarily bronchospastic. Individual mediator
blockers are only partially eective in inhibiting the EAR
[83, 84, 85, 98, 99], however, an in vitro study on human
tracheal smooth muscle demonstrated complete inhibi-
Allergic
tion of the EAR by combined H1 blocker, cyclooxygenase reaction
inhibitor, and lipoxygenase inhibitor [124]. The pathogen-
esis of the LAR is not so clear. An outdated hypothesis that Early
late responses were type III precipitin-mediated responses asthmatic
[44], controversial at that time [58], has been disproved response

since both cutaneous [125, 126] and pulmonary [118, 127]
late responses are IgE mediated. Animal studies have docu-
mented the requirement for inammatory cells (eosinophils,
neutrophils) in the LAR [128, 129] and induced airway
Increased
hyperresponsiveness [130132]. This has been conrmed in non-allergic and Late
humans using BAL [18, 133] and induced sputum [47, 84]. asthmatic
bronchial
response
The precise role of the chemokines and their cellular ori- reactivity
gin in the recruitment of inammatory cells are a topic of
current research [134136].

Allergens as a cause of asthma Symptoms on


exposure to
The importance of allergens as a cause of asthma (i.e. sympto- non-allergic stimuli
(irritants, exercise, etc.)
matic airway hyperresponsiveness and airway inammation
[137], which was hypothesized several years ago [49] (Fig. FIG. 35.3 Diagram of hypothesis explaining development and
35.3), is now generally accepted [138]. The lines of reason- maintenance of perennial allergen-induced asthma. Reproduced from
ing include the high prevalence of atopy amongst asthmatics Ref. [49] with permission.

Cockcroft Witt Witt Peat Burney

60 n 500 1293 891 2363 511

age 20 29 years 18 18 88 8 11 18 84


% with airway hyperresponsiveness

50
PC20 8 mg/ml PD20 3.9 mol 3.9 mol 7.8 mol 8 mol

40

30

20

10

0 3 0 5 0 5 0 5 0 2
Degree of atopy

FIG. 35.4 Degree of atopy (various scales) from nonatopic (0) to highly atopic (highest number) on the horizontal axis versus prevalence of airway
hyperresponsiveness (%) on the vertical axis from four population studies [20, 140142]. Reproduced from Ref. [143] with permission.

449
Asthma and COPD: Basic Mechanisms and Clinical Management

reduction with allergen avoidance [147, 183]. It is speculated particularly ocular or cutaneous and, to a lesser extent, nasal
that, in sensitized individuals, the duration and magnitude symptoms will bear a much closer relationship to exposure
of airways allergic exposure, if not suppressed pharmacologi- and may provide an important historical clue to sensitization.
cally, may lead not only to transient but also to persistent Irritant- and exercise-induced bronchospasm are sympto-
airway hyperresponsiveness and clinical asthma [49]. This is matic of underlying airway hyperresponsiveness and, conse-
supported by parallel observations in animals [148] and in quently, can be due to (often unrecognized) allergen exposure
human occupational asthma [149, 150]. that may not be acutely temporally related.
Sudden severe symptoms can occur following inhaled
allergen exposure if the exposure is marked, if the individ-
ual is highly sensitized, or if previous asthma symptoms/
airow obstruction have been either under-perceived or
INGESTED/INJECTED ALLERGENS ignored. However, genuinely sudden asthma exacerbations
raise the possibility of ingested or injected allergen (foods,
Isolated asthma caused by allergens introduced via routes drugs, bites, stings, allergen shots); this clinical scenario
other than inhalation is uncommon but has been reported should prompt an aggressive historical review for relevant
[151, 152]. Ingested allergens [152] (foods, drugs) or injected clues. Non-IgE-mediated responses to ASA and NSAIDs,
allergens [153] (hyposensitization injections, intradermal -adrenergic blockers, food additives (e.g. metabisulphites),
allergen tests, drugs, insect bites, and stings) can produce IgE- or cholinesterase inhibiting insecticides are included in the
mediated hypersensitivity reactions. Most often, these produce dierential diagnosis of sudden severe asthma.
systemic allergen reactions [151, 153] (violent gastrointestinal
upset, urticaria, angioedema, laryngeal edema, anaphylactic
shock, with or without bronchospasm). However, occasionally Diagnosis
such exposures produce reactions that appear to be centered
primarily in the lung [152, 154]. It is likely that these rep- The diagnosis of allergic asthma rests predominantly on the
resent systemic allergen reactions in subjects with pre-exist- historical features noted above. The inquiry into possible
ing asthma and high levels of airway hyperresponsiveness allergens should not stop with the home but must include
who develop disproportionately severe bronchospasm. Some work, school, social, and recreational exposures. Because of
cases of food/bite/sting-induced asthma may actually repre- the frequently subtle exposuresymptom relationships, non-
sent laryngeal spasm or edema which has been misdiagnosed. respiratory symptoms related to allergen exposure should be
Although relatively uncommon, allergen (asthmatic or other- sought. Following completion of a complete history, allergen
wise) responses to both ingested and injected allergens occur prick-skin testing with a small series of relevant allergens
rapidly and can be exceedingly severe. We have seen subjects should be done.
with sudden severe asthma due to foods (nuts, shellsh) and
two subjects with sudden onset status asthmaticus circum-
stantially linked to unrecognized black y bites.
Treatment
The treatment of allergen-induced asthma is the same as for
asthma in general and is outlined elsewhere in this book.
The pathophysiologic and clinical features outlined above
CLINICAL FEATURES stress the importance of environmental control. The possibil-
ity that chronic allergen exposure might lead to permanent
pathology suggests that therapeutic strategies that are largely
Clinical presentation eective by preventing the allergic response and sequelae
(allergen avoidance, cromones, anti-IgE) will be less eective
Allergic asthma usually begins at a young age, between about if their introduction is delayed.
2 and 20, but can develop at any time. A positive family his-
tory of asthma or atopy is common. Other atopic symptoms
are often present and include food sensitivity (infancy), child- Allergic bronchopulmonary mycoses
hood eczema, urticaria, conjunctivitis, and allergic rhinitis.
Asthma or asthma exacerbations should correlate with aller- A distinctive clinical syndrome occurs when atopic indi-
gen exposure or seasons. Spring, summer, or fall exacerba- viduals have organisms, against which they have IgE anti-
tions suggest fungal spore or pollen sensitivity whereas winter bodies, growing in their airways. The prototype, by far the
exacerbations are typical of indoor allergen sensitivity but commonest of these allergic bronchopulmonary mycoses,
may also occur in nonatopic asthmatics. The polyallergic sub- is allergic bronchopulmonary aspergillosis (ABPA) usu-
ject may show little seasonal variability. Because of the grad- ally caused by Aspergillus fumigatus [155158]. Other fungi,
ual onset of airway inammation and hyperresponsiveness, including Helminthosporium [159, 160], Curvularia and
allergen-induced asthma exacerbations are usually of gradual Drechslera [160, 161], Stemphylium [162], Candida [163],
onset often lagging behind and persisting beyond allergen Fusarium [164], and rarely bacteria such as Pseudomonas
exposure. An acute exposuresymptom relationship may be [165], may cause a similar syndrome. The syndrome that
lacking. For these reasons, the patient and physician may miss these organisms can produce involves complex immuno-
the importance of allergens. When present, nonrespiratory, logic and mechanical pathogenesis.

450
Allergens 35
The pathogenesis of ABPA initially involves acquisi- Allergic bronchopulmonary infestations with
tion of IgE-mediated type I hypersensitivity to the fungus. Aspergillus or other fungal organisms are not true infec-
Aspergillus fungal spores released from decaying vegetation, tions; progression to invasive fungal infections or mycetoma
particularly in the autumn germinate and grow in the airway formation is uncommon. Treatment is directed against
provoking reduced airway caliber and mucous hypersecre- the asthma and the immunologic abnormalities; intensive
tion leading to mucous plugs containing fungal hyphae pro- asthma treatment with systemic corticosteroids in doses
ducing cast-like outlines of the bronchial tree. The chronic sucient to suppress clinical and laboratory features of the
high-level allergen exposure leads to very high levels of both disease is indicated [171, 179, 180]. Total serum IgE may
allergen-specic and total serum IgE, an intense peripheral be useful to predict exacerbations [180]. With such treat-
and bronchial eosinophilia, and in most, but not all, to the ment, the prognosis is favorable; however, under-treatment
development of IgG precipitating antibodies. Other immu- can lead to substantial, even severe, permanent bronchopul-
nologic responses including cell-mediated immunity may monary damage. Itraconazole has been reported to be eec-
be stimulated. The continuous airways allergic reaction will tive in reducing corticosteroid requirements in ABPA [181]
lead to exacerbation of asthma while the other immuno- and may be useful in cystic brosis [176]. However, a recent
logic reactions may be responsible for bronchial and paren- Cochrane Database report concludes there is insucient
chymal damage that includes (proximal) bronchiectasis information available to recommend the use of azoles (itra-
and (upper lobe) interstitial pulmonary brosis. Fleeting or conazole, ketoconazole) in this condition [182].
xed pulmonary inltrates may be produced by immuno-
logic reactions within the lung, by the mechanical eect of
obstruction of major bronchi by mucous plugs, or both.
ABPA is a fairly common condition in some areas;
the precise prevalence is not certain. Amongst asthmatics,
SUMMARY
the prevalence of type I (IgE) sensitivity to Aspergillus may
approximate 25% [166168], and the prevalence of type 1. The majority of subjects with asthma are atopic.
III (IgG) sensitivity to Aspergillus is approximately half of
2. Allergen-induced airway hyperresponsiveness and airway
this [169, 170]. By contrast, on the dry Canadian prairies,
inammation point to allergens as a cause of asthma
Aspergillus skin sensitivity is uncommon, and we have seen
(i.e. symptomatic airway hyperresponsiveness with
no new cases of ABPA in over 20 years. Other organisms
inammation).
(e.g. Helminthosporium) are involved only rarely. The clinical
picture is generally that of a subject with pre-existent atopy, 3. The remarkable rise in the prevalence of atopy (hygiene
and usually previous asthma, presenting with exacerbation hypothesis, airtight home/indoor allergen exposure
of asthma accompanied by the expectoration of characteris- hypothesis, other hypotheses or some combination) over
tic rm brown plugs [155]. Pulmonary inltrates with eosi- the past 200 years may explain in whole or in part the
nophils with or without fever may be seen. Chronic disease rising prevalence of asthma.
may manifest as bronchiectasis with chronic or recurrent 4. Chronic/recurrent allergen exposure may lead to
pulmonary infection or pulmonary brosis with progressive persistent asthma.
dyspnea or both [155, 156].
The two features that must be present in all subjects 5. This provides a plausible basis to suggest that early
with ABPA are type I hypersensitivity to the Aspergillus prophylactically or actively anti-inammatory therapeutic
and the presence of Aspergillus in the airways. However, it strategies should improve long-term outcomes in allergic
is not always possible to grow the organism [171]. Specic asthma.
IgG precipitating antibodies are found in about 90% of 6. This also provides a rationale to speculate re-primary
cases [157]. Other features that are commonly seen include prevention of (allergic) asthma.
intense peripheral and bronchial eosinophilia [171], marked
elevations of total serum IgE [172], and transient pul-
monary inltrates [155158]; these all tend to correlate
with activity of disease. Chronic changes in established or
recurrent disease include radiographic demonstration of ACKNOWLEDGEMENT
an unusual and essentially pathognomonic proximal bron-
chiectasis [173] and, in more severe cases, progressive upper
The author wishes to thank Jacquie Bramley for assisting in
lobe interstitial pulmonary brosis similar to tuberculosis
the preparation of this manuscript.
and other upper lobe scarring conditions [156]. The com-
puterized tomographic scan may be particularly helpful as
an adjunct to the diagnosis of ABPA [174, 175]. ABPA
is a common complication of cystic brosis [176]; the 2% References
prevalence in one observational cohort was felt to repre-
sent under recognition [177]. Atopic skin testing has been 1. Bostock J. Case of a periodical aection of the eyes and chest. Med Chir
suggested as a useful screen in this population [176], how- Trans 10: 16165, 1819.
ever, may not be reliable [178]; specic skin tests, allergen- 2. Blackley CH. Experimental Researches on the Cause and Nature of
specic IgE, total IgE, and precipitins are recommended in Catarrhus Aestivus (Hay-Fever or Hay-Asthma). London: Balliere
this population [178]. Tindall & Cox, 1873.

451
Asthma and COPD: Basic Mechanisms and Clinical Management

3. von Pirquet C. Allergie. Munch Med Wochenschr 1906;53:1457. 31. Brown WG, Halonen MJ, Kaltenborn WT et al. The relationship of
(Translated from the German by Prausnitz C). In: Gell PGH, Coombs respiratory allergy, skin test reactivity, and serum IgE in a community
RRA (eds). Clinical Aspects of Immunology. Philadelphia, PA: F.A. Davis population sample. J Allergy Clin Immunol 63: 32835, 1979.
Company, 1963. 32. Cockcroft DW, Murdock KY, Berscheid BA. Relationship between
4. Schloss OM. A case of allergen to common foods. Am J Dis Child 3: atopy and bronchial responsiveness to histamine in a random popula-
34162, 1912. tion. Ann Allergy 53: 2629, 1984.
5. Dunbar WP. The present state of our knowledge of hay fever. J Hygiene 33. King TP. Immunochemical properties of antigens that cause disease. In:
13: 10548, 1913. Weis EB, Stein M (eds). Bronchial Asthma: Mechanisms and Therapies,
6. Noon L. Prophylactic inoculation against hay fever. Lancet 1: 157273, pp. 4349. Boston, MA: Little, Brown and Co., 1993.
1911. 34. Platts-Mills TAE, Solomon WR. Aerobiology and inhalant allergens.
7. Freeman J. Vaccination against hay fever: Report of results during the In: Middleton E, Reed CE, Ellis EF, Adkinson NF, Yuninger JW,
last three years. Lancet 1: 117880, 1914. Busse WW (eds). Allergy Principles and Practice, 4th edn., pp. 469528.
8. Prausnitz C, Kustner H. Studien uber Uberempndlichkeit. Centralb St. Louis, MO: Mosby YearBook Inc., 1993.
Bakteriol 1 Abt Orig 1921;86:160. (Translated by Prausnitz C). 35. Aalberse RC. Structural biology of allergens. J Allergy Clin Immunol
In: Gell PGH, Coombs RRA (eds). Clinical Aspects of Immunology, 106: 22838, 2000.
pp. 80816. Oxford: Blackwell Scientic Publications, 1962. 36. Dolovich J, Zimmerman B, Hargreave FE. Allergy in asthma. In:
9. Coca AF, Cooke RA. On the classication of the phenomena of hyper- Clark TJH, Godfrey S (eds). Asthma, pp. 13257. London: Chapman
sensitiveness. J Immunol 8: 16382, 1923. and Hall, 1983.
10. Ishizaka K, Ishizaka T. Identication of gamma-E antibodies as a 37. Busse WW, Reed CE, Hoehne JH. Where is the allergic reaction in
carrier of reaginic activity. J Immunol 99: 118798, 1967. ragweed asthma?. J Allergy Clin Immunol 50: 28993, 1972.
11. Wyman M. Autumnal Catarrah. Cambridge: Hurd and Houston, 1872. 38. Pollart SM, Chapman MD, Fiocco GP et al. Epidemiology of acute
12. Simons FER. Ancestors of Allergy. New York: Global Medical asthma: IgE antibodies to common inhalant allergens as a risk factor
Communications, 1994. for emergency room visits. J Allergy Clin Immunol 83: 87582, 1989.
13. Herxheimer H, Hyde HA, Williams DA. Allergic asthma caused by 39. Murray AB, Ferguson AC, Morrison B. The seasonal variation of aller-
fungal spores. Lancet 1: 57273, 1966. gic respiratory symptoms induced by house dust mites. Ann Allergy 45:
14. Osler W. Principles and Practice of Medicine. pp. 628632, 3rd edn. 34750, 1980.
New York: D. Appleton and Company, 1896. 40. Robertson DG, Kerigan AT, Hargreave FE et al. Late asthmatic
15. Cooke RA. Asthma. In: Musser JH (ed.), Internal Medicine, Theory and responses induced by ragweed pollen allergen. J Allergy Clin Immunol
Practice, pp. 9941001. Philadelphia, PA: Lea & Febiger, 1932. 54: 24454, 1974.
16. Herxheimer H. The late bronchial reaction in induced asthma. Int Arch 41. Cartier A, Thomson NC, Frith PA et al. Allergen-induced increase in
Allergy Appl Immunol 3: 32333, 1952. bronchial responsiveness to histamine: Relationship to the late asth-
17. Cockcroft DW, Run RE, Dolovich J et al. Allergen-induced increase matic response and change in airway caliber. J Allergy Clin Immunol 70:
in nonallergic bronchial reactivity. Clin Allergy 7: 50313, 1977. 17077, 1982.
18. de Monchy JGR, Kauman HF, Venge P et al. Bronchoalveolar eosi- 42. Boulet LP, Cartier A, Thomson NC et al. Asthma and increases in
nophilia during allergen-induced late asthmatic reactions. Am Rev nonallergic bronchial responsiveness from seasonal pollen exposure.
Respir Dis 131: 37376, 1985. J Allergy Clin Immunol 71: 399406, 1983.
19. Pearce N, Pekkanen J, Beasley R. How much asthma is really attribut- 43. OByrne PM, Dolovich J, Hargreave FE. State of the art: Late asth-
able to atopy?. Thorax 51: 26872, 1999. matic responses. Am Rev Respir Dis 136: 74051, 1987.
20. Cockcroft DW, Berscheid BA, Murdock KY et al. Sensitivity and 44. Pepys J. Immunopathology of allergic lung disease. Clin Allergy 3: 122,
specicity of histamine PC20 measurements in a random selection of 1973.
young college students. J Allergy Clin Immunol 89: 2330, 1992. 45. Booij-Noord H, deVries K, Sluiter HJ et al. Late bronchial obstructive
21. Postma DS, Koppelman GH, Meyers DA. The genetics of atopy reaction to experimental inhalation of house dust extract. Clin Allergy
and airway hyperresponsiveness. Am J Respir Crit Care Med 162: 2: 4361, 1972.
S118S123, 2000. 46. Newman Taylor AJ, Davies RJ, Hendrick DJ et al. Recurrent noctur-
22. Emanuel MB. Hay fever, a post industrial revolution epidemic: A history nal asthmatic reactions to bronchial provocation tests. Clin Allergy 9:
of its growth during the 19th century. Clin Allergy 18: 295304, 1988. 21319, 1979.
23. Platts-Mills TA, Blumenthal K, Perzanowski M. Determinants of clin- 47. Pin I, Freitag AP, OByrne PM et al. Changes in the cellular prole
ical allergic disease. The relevance of indoor allergens to the increase in of induced sputum after allergen-induced asthmatic responses. Am Rev
asthma [in process citation]. Am J Respir Crit Care Med 162: S12833, Respir Dis 145: 126569, 1992.
2000. 48. Dorsch W, Baur X, Emslander HP et al. Zur pathogenese und thera-
24. Dowse GK, Turner KJ, Stewart GA et al. The association between pie der allergeninduzierten verzogerten bronchialostruktion. Prax Klin
Dermatophagoides mites and the increasing prevalence of asthma in vil- Pneumol 34: 46168, 1980.
lage communities within the Papua New Guinea highlands. J Allergy 49. Cockcroft DW. Mechanism of perennial allergic asthma. Lancet 2:
Clin Immunol 75: 7583, 1985. 25356, 1983.
25. Strachan DP. Family size, infection and atopy: The rst decade of the 50. Altounyan REC. Changes in histamine and atropine responsiveness as
hygiene hypothesis. Thorax 55: S2S10, 2000. a guide to diagnosis and evaluation of therapy in obstructive airways
26. Pepys J. Atopy. In: Gell PGH, Coombs RRA, Lachman PJ (eds). disease. In: Pepys J, Franklands AW (eds). Disodium Cromoglycate in
Clinical Aspects of Immunology, pp. 877902. Oxford: Blackwell Scientic Allergic Airways Disease, pp. 4753. London: Butterworths, 1970.
Publications, 1975. 51. Lowhagen O, Rak S. Modication of bronchial hyperreactivity after
27. Sandford A, Weir T, Pare P. The genetics of asthma. Am J Respir Crit treatment with sodium cromoglycate during pollen season. J Allergy
Care Med 153: 174965, 1996. Clin Immunol 75: 46067, 1985.
28. Aalberse RC, Sterk PJ. The roots of allergic sensitisation: Fourth 52. Cockcroft DW, Murdock KY. Changes in bronchial responsiveness
Lunteren conference. Am J Respir Crit Care Med 162: S61136, 2000. to histamine at intervals after allergen challenge. Thorax 42: 3028,
29. Leskowitz S, Salvaggio JE, Schwartz HJ. An hypothesis for the devel- 1987.
opment of atopic allergy in man. Clin Allergy 2: 23746, 1972. 53. Millilo G. Discussion. In: Nadel JA, Pauwels R (eds). International
30. Woolcock AJ, Colman MH, Jones MW. Atopy and bronchial reactivity Conference on Bronchial Hyperreactivity, p. 17. Oxford: The Medicine
in Australian and Melanesian populations. Clin Allergy 8: 15564, 1978. Publishing Foundation, 1982.

452
Allergens 35
54. Durham SR, Graneek BJ, Hawkins R et al. The temporal relationship 74. Orehek J, Gayrard P, Grimaud Ch et al. Bronchoconstriction provo-
between increases in airway responsiveness to histamine and late asth- quee par inhalation dallergene dans lasthme: Eet antagoniste
matic responses induced by occupational agents. J Allergy Clin Immunol dun anticholinergique de synthese. Bull Eur Physiopathol Respir 11:
79: 398406, 1987. 193201, 1975.
55. Thorpe J, Steinberg D, Bernstein D et al. Bronchial hyperreactivity 75. Cockcroft DW, Run RE, Hargreave FE. Eect of Sch1000 in aller-
occurs soon after the immediate asthmatic response in dual responders. gen-induced asthma. Clin Allergy 8: 36172, 1978.
Am Rev Respir Dis 133: A93, 1986. 76. Boulet LP, Latimer KM, Roberts RS et al. The eect of atropine on
56. Hargreave FE, Ryan G, Thomson NC et al. Bronchial responsiveness allergen-induced increases in bronchial responsiveness to histamine.
to histamine or methacholine in asthma: Measurement and clinical Am Rev Respir Dis 130: 36872, 1984.
signicance. J Allergy Clin Immunol 68: 34755, 1981. 77. Pauwels R, van Renterghem D, Van Der Straeten M et al. The eect of
57. Djukanovic R, Feather I, Gratziou C et al. Eect of natural allergen theophylline and enprofylline on allergen-induced bronchoconstriction.
exposure during the grass pollen season on airways inammatory cells J Allergy Clin Immunol 76: 58390, 1985.
and asthma symptoms. Thorax 51: 57581, 1996. 78. Cockcroft DW, Murdock KY, Gore BP et al. Theophylline does not
58. Orie NGM, Van Lookeren Campagne JG, Knol K, et al. Late reactions inhibit allergen-induced increase in airway responsiveness to metha-
in bronchial asthma. In: Pepys J, Yamamura I (eds.), Intal in Bronchial choline. J Allergy Clin Immunol 83: 91320, 1989.
Asthma, Proceedings of the 8th International Congress of Allergollogy, 79. Crescioli S, Spinazzi A, Plebani M et al. Theophylline inhibits early
pp. 1729. Tokyo, 1974. and late asthmatic reactions induced by allergens in asthmatic subjects.
59. Hegardt B, Pauwels R, Van Der Straeten M. Inhibitory eect of KWD Ann Allergy 66: 24551, 1991.
2131, terbutaline, and DSCG on the immediate and late allergen- 80. Hendeles L, Harman E, Huang D et al. Theophylline attenuation of
induced bronchoconstriction. Allergy 36: 11522, 1981. airway responses to allergen: Comparison with cromolyn metered-dose
60. Run RE, Cockcroft DW, Hargreave FE. A comparison of the pro- inhaler. J Allergy Clin Immunol 95: 50514, 1995.
tective eect of Sch1000 and fenoterol on allergen-induced asthma. 81. Pepys J, Chan M, Hargreave FE et al. Inhibitory eects of disodium
J Allergy Clin Immunol 61: 4247, 1978. cromoglycate on allergen-inhalation tests. Lancet 2: 13437, 1968.
61. Cockcroft DW, Murdock KY. Comparative eects of inhaled salb- 82. Dahl R, Pedersen B. Inuence of nedocromil sodium on the dual
utamol, sodium cromoglycate and beclomethasone dipropionate on asthmatic reaction after allergen challenge: A double-blind, placebo-
allergen-induced early asthmatic response, late asthmatic responses and controlled study. Eur J Respir Dis 69(Suppl 147): 26365, 1986.
increased bronchial responsiveness to histamine. J Allergy Clin Immunol 83. Taylor IK, OShaughnessy KM, Fuller RW et al. Eect of cysteinyl-
79: 73440, 1987. leukotriene receptor antagonist ICI 204.219 on allergen-induced bron-
62. Church MK, Young KD. The characteristics of inhibition of histamine choconstriction and airway hyperreactivity in atopic subjects. Lancet
release from human lung fragments by sodium cromoglycate, salbuta- 337: 69094, 1991.
mol, and chlorpromazine. Br J Pharmacol 78: 67179, 1983. 84. Diamant Z, Grootendorst DC, Veselic-Charvat M et al. The eect
63. Howarth PH, Durham SR, Lee TH et al. Inuence on albuterol, cro- of Montelukast (MK-0476), a cysteinyl leukotriene receptor antago-
molyn sodium, and ipratropium bromide on the airway and circulating nist, on allergen-induced airway responses and sputum cell counts in
mediator responses to allergen bronchial provocation in asthma. Am asthma. Clin Exp Allergy 29: 4251, 1999.
Rev Respir Dis 132: 98692, 1985. 85. Davis BE, Todd DC, Cockcroft DW. Eect of combined montelukast
64. Twentyman OP, Finnerty JP, Harris A et al. Protection against aller- and desloratadine on the early asthmatic response to inhaled allergen.
gen-induced asthma by salmeterol. Lancet 336: 133842, 1990. J Allergy Clin Immunol 116: 76872, 2005.
65. Wong BJ, Dolovich J, Ramsdale EH et al. Formoterol compared with 86. Hui KP, Taylor IK, Taylor GW et al. Eect of a 5-lipoxygenase inhibi-
beclomethasone and placebo on allergen-induced asthmatic responses. tor on leukotriene generation and airway responses after allergen chal-
Am Rev Respir Dis 146: 115860, 1992. lenge in asthmatic patients. Thorax 46: 18489, 1991.
66. Cockcroft DW, McParland CP, Britto SA et al. Regular inhaled salb- 87. Diamant Z, Timmers MC, van der Veen H et al. The eect of MK-
utamol and airway responsiveness to allergen. Lancet 342: 83337, 1993. 0591, a novel 5-lipoxygenase activating protein inhibitor, on leukot-
67. Cockcroft DW, McParland CP, OByrne PM et al. Beclomethasone riene biosynthesis and allergen-induced airway response in asthmatic
given after the early asthmatic response inhibits the late response and subjects in vivo. J Allergy Clin Immunol 95: 4251, 1995.
the increased methacholine responsiveness and cromolyn does not. 88. Kirby JG, Hargreave FE, Cockcroft DW et al. Indomethacin inhibits
J Allergy Clin Immunol 91: 116368, 1993. allergen-induced airway hyperresponsiveness but not allergen-induced
68. Cockcroft DW, Swystun VA, Bhagat R. Interaction of inhaled 2 ago- asthmatic responses. J Appl Physiol 66: 57883, 1989.
nist and inhaled corticosteroid on airway responsiveness to allergen and 89. Booij-Noord H, Orie NGM, deVries K. Immediate and late bronchial
methacholine. Am J Respir Crit Care Med 152: 148589, 1995. obstructive reactions to inhalation of house dust and protective eects
69. Swystun VA, Gordon JR, Davis EB et al. Mast cell tryptase release and of disodium cromoglycate and prednisolone. J Allergy Clin Immunol 48:
asthmatic responses to allergen increase with regular use of salbutamol. 34454, 1971.
J Allergy Clin Immunol 106: 5764, 2000. 90. Pepys J, Davies RJ, Breslin ABX et al. The eects of inhaled beclom-
70. Gauvreau GM, Watson RM, Jordana M et al. The eect of regular ethasone dipropionate (Becotide) and sodium cromoglycate on asth-
inhaled salbutamol on allergen-induced airway responses and inam- matic reactions to provocation tests. Clin Allergy 4: 1324, 1974.
matory cells in blood and induced sputum. Am J Respir Crit Care Med 91. Van Der Star JG, Berg WC, Steenhuis EJ et al. Invloed van beclometason-
151: A39, 1995. dipropionaat per aerosol op de obstructieve reactie in de bronchien na
71. Lai CKW, Twentyman OP, Holgate ST. The eect of an increase huisstonhalatie. Ned Tijdschr Geneeskd 120: 192832, 1976.
in inhaled allergen dose after rimiterol hydrobromide on the occur- 92. Burge PS, Efthimiou J, Turner-Warwick M et al. Double-blind trials
rence and magnitude of the late asthmatic response and the associated of inhaled beclomethasone dipropionate and uocortin butyl ester in
change in nonspecic bronchial responsiveness. Am Rev Respir Dis 140: allergen-induced immediate and late reactions. Clin Allergy 12: 52331,
91723, 1989. 1982.
72. van Schaych CP, Cloosterman SGM, Hoand ID et al. How detrimen- 93. Du Toit JI, Salome CM, Woolcock AJ. Inhaled corticosteroids reduce
tal is chronic use of bronchodilators in asthma and chronic obstructive the severity of bronchial hyperresponsiveness in asthma but oral theo-
pulmonary disease?. Am J Respir Crit Care Med 151: 131719, 1995. phylline does not. Am Rev Respir Dis 136: 117478, 1987.
73. Yu DYC, Galant SP, Gold WM. Inhibition of antigen-induced bron- 94. Woolcock AJ, Yan K, Salome CM. Eect of therapy on bronchial
choconstriction by atropine in asthmatic patients. J Appl Physiol 32: hyperresponsiveness in the long-term management of asthma. Clin
82328, 1972. Allergy 18: 16576, 1988.

453
Asthma and COPD: Basic Mechanisms and Clinical Management

95. Laitinen LA, Laitinen A, Haahtela T. A comparative study of the 115. Van Metre TE, Marsh DG, Adkinson NF et al. Immunotherapy for
eects of an inhaled corticosteroid, budesonide, and of a 2-agonist, cat asthma. J Allergy Clin Immunol 82: 105568, 1988.
terbutaline, on airway inammation in newly diagnosed asthma. 116. Warner JD, Soothill JF, Price JF et al. Controlled trial of hyposensiti-
J Allergy Clin Immunol 90: 3242, 1992. zation to Dermatographoides pteronyssinus in children with asthma.
96. Djukanovic R, Wilson JW, Britton YM et al. Eect of an inhaled cor- Lancet 2: 91215, 1978.
ticosteroid on airway inammation and symptoms of asthma. Am Rev 117. Boulet L-P, Chapman KR, Cote J et al. Inhibitory eects of an anti-
Respir Dis 145: 66974, 1992. IgE antibody E25 on allergen-induced early asthmatic response. Am J
97. Trigg CJ, Manolitsas ND, Wang J et al. Placebo-controlled immun- Respir Crit Care Med 155: 183540, 1997.
opathologic study of four months of inhaled corticosteroids in asthma. 118. Fahy JV, Fleming HE, Wong HH et al. The eect of an anti-IgE
Am J Respir Crit Care Med 150: 1722, 1994. monoclonal antibody on the early- and late-phase responses to aller-
98. Holgate ST, Emanuel MB, Howarth PH. Astemizole and other H1- gen inhalation in asthmatic subjects. Am J Respir Crit Care Med 155:
antihistaminic drug treatment of asthma. J Allergy Clin Immunol 76: 182834, 1997.
37580, 1985. 119. Leckie MJ, ten Brinke A, Khan J et al. Eects of an interleukin-
99. Raerty P, Beasley R, Holgate S. The contribution of histamine to 5 blocking monoclonal antibody on eosinophils, airway hyper-
immediate bronchoconstriction provoked by inhaled allergen and responsiveness, and the late asthmatic response. Lancet 356: 214448,
adenosine 5 monophosphate in atopic asthma. Am Rev Respir Dis 2000.
136: 36973, 1987. 120. Bryan SA, OConnor BJ, Matti S et al. Eects of recombinant human
100. Hamid M, Raerty P, Holgate ST. The inhibitory eect of terfenadine interleukin-12 on eosinophils, airway hyperresponsiveness, and the
and urbiprofen on early and late-phase bronchoconstriction follow- late asthmatic response. Lancet 356: 214953, 2000.
ing allergen challenge in atopic asthma. Clin Exp Allergy 20: 26167, 121. Lee TH, Brown MJ, Nagy L et al. Exercise-induced release of hista-
1990. mine and neutrophil chemotactic factor in atopic asthmatics. J Allergy
101. Wells A, Taylor B. A placebo-controlled trial of ketotifen (HC20-511, Clin Immunol 70: 7381, 1982.
Sandoz) in allergen induced asthma and comparison with disodium 122. Murray JJ, Tonnel AB, Brash AR et al. Release of prostaglandin D2
cromoglycate. Clin Allergy 9: 23740, 1979. into human airways during acute antigen challenge. New Engl J Med
102. Pauwels R, Lamont H, Van Der Straeten M. Comparison between keto- 315: 8004, 1986.
tifen and DSCG in bronchial challenge. Clin Allergy 8: 28993, 1978. 123. Lewis RA, Austen KF. The biologically active leukotrienes:
103. Craps L, Greenwood C, Radielovic P. Clinical investigation of agents Biosynthesis, metabolism, receptors, functions and pharmacology.
with prophylactic anti-allergic eects in bronchial asthma. Clin Allergy J Clin Invest 73: 88997, 1984.
8: 37382, 1978. 124. Schellenberg RR, Du MJ, Foster A. Human bronchial responses to
104. Klein G, Urbanek R, Matthys H. Long-term study of the protec- anti-IgE in vitro. Clin Invest Med 8: A41, 1985.
tive eect of ketotifen in children with allergic bronchial asthma. The 125. Dolovich J, Hargreave FE, Chalmers R et al. Late cutaneous allergic
value of a provocation test in assessment of treatment. Respiration 41: responses in isolated IgE-dependent reactions. J Allergy Clin Immunol
12832, 1981. 52: 3846, 1973.
105. Adachi M, Kobayashi H, Aoki N et al. A comparison of the inhibi- 126. Solley GO, Gleich GJ, Jordon RE et al. The late phase of the immedi-
tory eects of ketotifen and disodium cromoglycate on bronchial ate wheal and are skin reactions. J Clin Invest 58: 40820, 1976.
responses to house dust, with special reference to the late asthmatic 127. Kirby JG, Robertson DG, Hargreave FE et al. Asthmatic responses to
response. Pharmatherapeutics 4: 3642, 1984. inhalation of anti-human IgE. Clin Allergy 16: 19194, 1986.
106. Cockcroft DW, Keshmiri M, Murdock KY et al. Allergen-induced 128. Schampain MP, Behrens BL, Larsen GL et al. An animal model of
increase in airway responsiveness is not inhibited by acute treatment late pulmonary responses to Alternaria challenge. Am Rev Respir Dis
with ketotifen or clemastine. Ann Allergy 68: 24550, 1992. 126: 49398, 1982.
107. Patel PC, Rutherford BC, Lux J et al. The eect of repirinast on 129. Abraham WM, Delehunt JC, Yerger L et al. Characterization of a late
airway responsiveness to methacholine and allergen. J Allergy Clin phase pulmonary response after antigen challenge in allergic sheep.
Immunol 90: 78288, 1992. Am Rev Respir Dis 128: 83944, 1983.
108. Fish JE, Ankin MG, Adkinson NF et al. Indomethacin modication 130. Chung KF, Becker AB, Lazarus SC et al. Antigen-induced hyper-
of immediate-type immunologic airway responses in allergic asth- responsiveness and pulmonary inammation in allergic dogs. J Appl
matic and non-asthmatic subjects; evidence for altered arachidonic Physiol 58: 134753, 1985.
acid metabolism in asthma. Am Rev Respir Dis 123: 60914, 1981. 131. OByrne PM, Walters EH, Gold ED et al. Neutrophil depletion
109. Nakazawa T, Toyoda T, Furukawa M et al. Inhibitory eects of various inhibits airway hyperresponsiveness induced by ozone exposure. Am
drugs on dual asthmatic responses in wheat our-sensitive subjects. Rev Respir Dis 130: 21419, 1984.
J Allergy Clin Immunol 58: 19, 1976. 132. Murphy KR, Wilson MC, Irvin CG et al. The requirement for poly-
110. Fairfax AJ. Inhibition of the late asthmatic response to house dust morphonuclear leukocytes in the late asthmatic response and height-
mite by non-steroidal anti-inammatory drugs. Prostaglandins Leuko ened airways reactivity in an animal model. Am Rev Respir Dis 134:
Med 8: 23948, 1982. 6268, 1986.
111. Manning PJ, Stevens WH, Cockcroft DW et al. The role of throm- 133. Metzger WJ, Richerson WB, Worden K et al. Bronchoalveolar lavage
boxane in allergen-induced asthmatic responses. Eur J Respir Dis 4: of allergic asthmatic patients following allergen bronchoprovocation.
66772, 1991. Chest 89: 47783, 1986.
112. Wilkens H, Wilkens JH, Bosse S et al. Eects of an inhaled PAF- 134. OByrne P. Asthma pathogenesis and allergen-induced late responses.
antagonist (WEB 2086 BS) on allergen-induced early and late J Allergy Clin Immunol 102: S8589, 1998.
asthmatic responses and increased bronchial responsiveness to metha- 135. OByrne PM, Gauvreau GM, Wood LJ. Interaction between haemopoi-
choline. Am Rev Respir Dis 143: A812, 1991. etic regulation and airway inammation. Clin Exp Allergy 29(Suppl 2):
113. Bianco S, Pieroni MG, Reni RM et al. Protective eect of inhaled 2732, 1999.
furosemide on allergen-induced early and late asthmatic reactions. 136. Varga EM, Wachholz P, Nouri-Aria KT et al. T cells from human
New Engl J Med 321: 106973, 1989. allergen-induced late asthmatic responses express IL-12 receptor
114. Ortolani C, Pastorello E, Moss RB et al. Grass pollen immuno- beta 2 subunit mRNA and respond to IL-12 in vitro. J Immunol 165:
therapy: A single year double-blind, placebo-controlled study in 287785, 2000.
patients with grass pollen-induced asthma and rhinitis. J Allergy Clin 137. Boulet L-P, Becker A, Berube D, Beveridge R, Ernst P. Canadian
Immunol 73: 28390, 1984. Asthma Consensus Group. CMAJ 161: S161, 1999.

454
Allergens 35
138. Nelson HS. The importance of allergens in the development of 160. Matthiesson AM. Allergic bronchopulmonary disease caused by fungi
asthma and the persistence of symptoms. J Allergy Clin Immunol 105: other than Aspergillus. Thorax 36: 719, 1981.
S62832, 2000. 161. McAleer R, Kroenert DB, Elder JL et al. Allergic bronchopulmonary
139. Cookson WOCM, Musk AW, Ryan G. Association between asthma disease caused by Curvularia lunata and Drechslera hawaiiensis. Thorax
history, atopy, and non-specic bronchial responsiveness in young 36: 33844, 1981.
adults. Clin Allergy 16: 42532, 1984. 162. Benatar SR, Allan B, Hewitson RP et al. Allergic bronchopulmonary
140. Witt C, Stuckey MS, Woolcock AJ et al. Positive allergy prick skin stemphyliosis. Thorax 35: 51518, 1980.
tests associated with bronchial histamine responsiveness in an unse- 163. Voisin C, Tonnel AB, Jacob M et al. Inltrats pulminaires avec grande
lected population. J Allergy Clin Immunol 77: 698702, 1986. eosinophilie sanguine associes a une candidose bronchique. Rev Fr
141. Peat JK, Britton WJ, Salome CM et al. Bronchial hyperresponsiveness Allergie Immunol Clin 16: 27981, 1976.
in two populations of Australian school children: III. Eect of expo- 164. Saini SK, Boas SR, Jerath A et al. Allergic bronchopulmonary myco-
sure to environmental allergens. Clin Allergy 17: 291300, 1987. sis to Fusarium vasinfectum in a child. Ann Allergy Asthma Immunol
142. Burney PFJ, Britton JR, Chinn S et al. Descriptive epidemiology of 80: 37780, 1998.
bronchial reactivity in an adult population: Results from a community 165. Gordon DS, Hunter RG, OReilly RJ et al. Pseudomonas aeruginosa
study. Thorax 42: 3844, 1987. allergy and humoral antibody-mediated hypersensitivity pneumonia.
143. Cockcroft DW, Hargreave FE. Relationship between atopy and air- Am Rev Respir Dis 108: 12731, 1973.
way responsiveness. In: Sluiter HJ, van der Lende R (eds). Bronchitis 166. Longbottom JL, Pepys J. Pulmonary aspergillosis: Diagnostic and
IV, pp. 2332. The Netherlands: Royal Vangorcum, Assen, 1988. immunologic signicance of antigens and C-substance in Aspergillus
144. Peat JK, Salome CM, Woolcock AJ. Longitudinal changes in atopy fumigatus. J Pathol Bacteriol 88: 14151, 1964.
during a 4-year period: Relation to bronchial hyperresponsiveness and 167. Hendrick DJ, Davies RJ, DSouza MF et al. An analysis of prick skin
respiratory symptoms in a population sample of Australian school test reactions in 656 asthmatic patients. Thorax 30: 28, 1975.
children. J Allergy Clin Immunol 85: 6574, 1990. 168. Malo JL, Paquin R. Incidence of immediate sensitivity to Aspergillus
145. Sotomayor H, Badier M, Vervloet D et al. Seasonal increase of carba- fumigatus in a North American asthma population. Clin Allergy 9:
chol airway responsiveness in patients allergic to grass pollen. Am Rev 37784, 1979.
Respir Dis 130: 5658, 1984. 169. Hoehne JH, Reed CE, Dickie HA. Allergic bronchopulmonary
146. Dorward AJ, Roberts JA, Thomson NC. Eect of nedocromil sodium aspergillosis is not rare. Chest 63: 17781, 1973.
on histamine airway responsiveness in grass-pollen sensitive asthmat- 170. Malo JL, Paquin R, Longbottom JL. Prevalence of precipitating
ics during the pollen season. Clin Allergy 16: 30915, 1986. antibodies to dierent extracts of Aspergillus fumigatus in a North
147. Platts-Mills TAE, Mitchell EB, Nock P et al. Reduction of bron- American asthmatic population. Clin Allergy 11: 33341, 1981.
chial hyperreactivity during prolonged allergen avoidance. Lancet 2: 171. McCarthy DS, Pepys J. Allergic bronchopulmonary aspergillosis. Clin
67578, 1982. Allergy 1: 26186, 1971.
148. Becker AB, Hershkovich J, Simons FER et al. Development of 172. Patterson R, Fink JN, Pruzansky JJ et al. Serum immunoglobulin levels
chronic airway hyperresponsiveness in ragweed sensitized dogs. J Appl in pulmonary allergic aspergillosis and certain other lung disease, with
Physiol 66: 269197, 1989. special reference to immunoglobulin E. Am J Med 54: 1622, 1973.
149. Chan-Yeung M, Lam S, Koener S. Clinical features and natural his- 173. Scadding JG. The bronchi in allergic bronchopulmonary aspergillosis.
tory of occupational asthma due to Western Red Cedar (Thuja pli- Scand J Respir Dis 48: 37277, 1967.
cata). Am J Med 72: 41115, 1982. 174. Panchal N, Bhagat R, Pant C, Shah A. Allergic bronchopulmonary
150. Mapp CE, Corona PC, De Marzo N et al. Persistent asthma due to aspergillosis: The spectrum of computed tomography appearances.
isocyanates: A follow-up study of subjects with occupational asthma Respir Med 91: 21319, 1997.
due to toluene di-isocyanate (TDI). Am Rev Respir Dis 137: 132629, 175. Johkoh T, Muller NL, Akira M et al. Eosinophilic lung diseases:
1988. Diagnostic accuracy of thin-section CT in 111 patients. Radiology
151. Metcalfe DD. The diagnosis of food allergy: Theory and practice. In: 216: 77380, 2000.
Spector SL (ed.), Provocative Challenge Procedures: Bronchial, Oral, 176. Nepomuceno IB, Esrig S, Moss RB. Allergic bronchopulmonary
Nasal, and Exercise, pp. 11932. Boca Raton, FL: CRC Press Inc., aspergillosis in cystic brosis: Role of atopy and response to itracona-
1983. zole. Chest 115: 36470, 1999.
152. Bock SA, Lee W-Y, Remigio LK et al. Studies of hypersensitivity 177. Geller DE, Kaplowitz H, Light MJ et al. Allergic bronchopulmonary
reactions to foods in infants and children. J Allergy Clin Immunol 62: aspergillosis in cystic brosis: Reported prevalence, regional distribution,
32734, 1978. and patient characteristics. Scientic Advisory Group, Investigators and
153. Orange RP, Donsky GJ. Anaphylaxis. In: Middleton E Jr., Reed CE, Coordinators of the Epidemiologic Study of Cystic Fibrosis. Chest 116:
Ellis EF (eds). Allergy Principles and Practice, pp. 56373. St.Louis, 63946, 1999.
MO: The CV Mosby Company, 1978. 178. Skov M, Koch C, Reimert CM et al. Diagnosis of allergic bronchop-
154. Gluck JC, Pacin MP. Asthma from mosquito bites: A case report. ulmonary aspergillosis (ABPA) in cystic brosis. Allergy 55: 5058,
Ann Allergy 56: 49293, 1986. 2000.
155. Malo JL, Hawkins R, Pepys J. Studies in chronic allergic bronchopul- 179. Sarstein BH, DSouza MF, Simon G et al. Five year follow-up of
monary aspergillosis 1: Clinical and physiological ndings. Thorax 32: allergic bronchopulmonary aspergillosis. Am Rev Respir Dis 108: 450
25461, 1977. 59, 1973.
156. Malo JL, Pepys J, Simon G. Studies in chronic allergic bronchopul- 180. Wang JLF, Patterson R, Roberts M et al. The management of allergic
monary aspergillosis 2: Radiological ndings. Thorax 32: 26268, bronchopulmonary aspergillosis. Am Rev Respir Dis 120: 8792, 1979.
1977. 181. Salez F, Brichet A, Desurmont S et al. Eects of itraconazole therapy
157. Malo JL, Longbottom J, Mitchell J et al. Studies in chronic allergic in allergic bronchopulmonary aspergillosis. Chest 116: 166568, 1999.
bronchopulmonary aspergillosis 3: Immunological ndings. Thorax 182. Wark P, Wilson AW, Gibson PG. Azoles for allergic bronchopulmo-
32: 26974, 1977. nary aspergillosis. Cochrane Database Syst Rev 3: CD001108, 2000.
158. Malo JL, Inouye T, Hawkins R et al. Studies in chronic allergic bron- 183. Dorward AJ, Collo MJ, MacKay NS et al. Eect of house dust mite
chopulmonary aspergillosis 4: Comparison with a group of asthmat- avoidance measures on adult atopic asthma. Thorax 43: 98102, 1988.
ics. Thorax 32: 27580, 1977. 184. Cockcroft DW, OByrne PM, Swystun VA et al. Regular use of
159. Dolan CT, Weed LA, Dines DE. Bronchopulmonary helminthospo- inhaled albuterol and the allergen-induced late asthmatic response.
riosis. Am J Clin Pathol 53: 23542, 1970. J Allergy Clin Immunol 96: 4449, 1995.

455
Occupational Agents
36 CHAPTER

asthma (IIA) is a type of OA caused by an appar-


OCCUPATIONAL ASTHMA ently nonimmunological mechanism [3].
Jean-Luc Malo1, Moira
Chan-Yeung2 and Susan
Definition Kennedy3
Agents that induce OA by 1
Department of Chest Medicine, Sacr-
A proposed denition of occupational asthma immunological mechanisms
Coeur Hospital, Montreal, Canada
(OA) should take into account at least two essen- 2
Department of Medicine, Respiratory
tial aspects of the condition. First, it should reect Agents that induce asthma by
Division, University of British Columbia,
the fact that OA shares the clinical, functional, IgE-dependent mechanisms
and pathological features of asthma, that is vari- Vancouver, BC, Canada
The agents causing OA by IgE-dependent 3
School of Environmental Health,
able, spontaneously or as a result of treatment, mechanisms include both high molecular weight
symptomatic airway caliber, hyperresponsive- University of British Columbia,
compounds (10 kDa) and some low molecular
ness, and inammation of the airways. Second, Vancouver, BC, Canada
weight compounds (10 kDa). High molecular
it should state that OA is caused by the work- weight compounds are usually proteins or polysac-
place in which, in most cases, an agent has been charides and induce specic IgE antibodies. Some
identied. The denition of OA which has been low molecular weight agents, such as platinum [4]
retained in a textbook on OA reads as follows: and acid anhydrides [5], can also induce specic
OA is a disease characterized by variable airow IgE antibodies by acting as haptens, which com-
limitation and/or airway hyperresponsiveness bine with a body protein to form complete anti-
and/or inammation due to causes and conditions gens. There is evidence to support the fact that
attributable to a particular occupational environ- the asthmogenic potency of a compound is deter-
ment and not to stimuli encountered outside the mined to a certain extent by its chemical structure
workplace [1]. Within this denition, two types [6, 7]. Often, agents such as trimellitic anhydride
of OA are distinguished by whether they appear and isocyanates can modify body proteins, an
after or without a latency period. important step toward becoming a hapten, and
thus stimulate the immune system.
Agents that cause OA and the These agents often aect atopic subjects.
The inhaled occupational sensitizer can bind to
pathophysiological mechanisms specic IgE antibodies on the surface of mast
cells, basophils, and possibly macrophages and
Over 300 agents in the workplace have been eosinophils. The mechanism of asthma induction
implicated in causing asthma [2]. Table 36.1 by these sensitizers is similar to nonoccupational
shows a list of the more common agents respon- allergens [8].
sible for OA. The agents can be divided into two
groups according to the pathogenic mechanisms:
Agents that induce asthma by
those that give rise to asthma by immunologi-
cal mechanisms and those by nonimmunological IgE-independent mechanisms
mechanisms. Agents in the former group can cause The agents that give rise to OA through IgE-
asthma by immunoglobulin (Ig)E-dependent or independent mechanisms are mostly low molec-
IgE-independent mechanisms. Irritant-induced ular weight compounds such as isocyanates and

457
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 36.1 Agents that cause immunologically mediated occupational plicatic acid (the agent responsible for Western red cedar
asthma. asthma) [8]. In subjects with both isocyanate and Western
red cedar asthma, specic IgE antibodies have been found
Common sources of exposure in only a small proportion proven to have the disease [9]
although, if present in high concentrations, they appear
High molecular weight agents specic for the disease [10] though, apparently, not causal
for the disease [11]. It is still uncertain whether the pres-
Animal-derived material Animal, poultry and insect work, ence of specic IgG antibodies is a marker of the disease or
Dander veterinary medicine, fishing and merely of exposure. In Western red cedar asthma, the sig-
fish processing, laboratory work nicance of the presence of specic IgE antibodies is not
Excreta
Secretions
clear since anti-IgE antibodies failed to inhibit the release
of histamine by plicatic acid from granulocytes of patients
Serum
with the disease [12].
Studies of both isocyanate and Western red cedar
Plant-derived material
asthma have demonstrated that T-lymphocytes might play
Flour Bakery a direct role in mediating the inammatory response in the
Grain Grain elevator and terminal and airways [13, 14]. In patients with isocyanate-induced asthma,
feed mill the majority of T-cell clones derived from bronchial mucosa
Castor bean Oil manufacture of subjects were found to be CD8 T-lymphocytes that pro-
Coffee bean Food processing duced IL-5 [13]. Among patients with Western red cedar
Wood dust Sawmill, carpentry, furniture work
asthma, peripheral blood lymphocytes released IL-5 and
IFN- after stimulation with plicatic acid in sensitized subjects
Vegetable gum Printing
[14]. Animal studies have shown that diisocyanate-induced
Psyllium Health care asthma is driven primarily by CD4 T-cells and is depend-
Latex Latex ent upon the expression of Th2 cytokines. However, animal
models are not always reective of human responses [15].
Enzymes
A more recent study of bronchial biopsies taken after
-amylase Bakery specic inhalation challenge testing from patients suspected of
Papain Food processing diisocyanate-induced asthma showed a striking absence of C
Alcalase Pharmaceutical industry and IL-4 mRNA-positive cells in bronchial biopsy specimens
Bacillus subtilis-derived Detergent enzyme industry irrespective of whether the response was positive or negative
enzyme indicating the absence of IgE involvement at least in a pro-
portion of patients with diisocyanate-induced asthma [11].
Toluene diisocyanate was also found to induce pro-
Low molecular weight agents
duction of proinammatory cytokines and chemokines from
Spray paints bronchial epithelial cells through the epidermal growth fac-
Toluene diisocyanate Manufacture of plastic, foam tor receptor and p38 mitogen-activated protein kinase path-
ways [16].
Dimethylphenyl diisocyanate Insulation
Hexamethylene diisocyanate Automobile spray paint

Wood dust Agents that induce OA by


Western red cedar Sawmill worker, carpenter, nonimmunological mechanism
furniture maker
Irritant-induced asthma is apparently a nonimmunologically
Acid anhydride Users of plastics, epoxy resins induced type of asthma. There seems to be a dose-dependent
Biocides relationship between exposure and likelihood of permanent
Formaldehyde Health care workers
disability/impairment [3, 1719].
Glutaraldehyde

Chloramine T Pathology
Colophny fluxes Electronic workers
Immunologically mediated OA
Irritant agents
There is no dierence between the pathology of the airways
of patients with occupational and those with nonoccupational
Chlorine Pulp and paper mills asthma. Studies of bronchoalveolar lavage (BAL) uid have
shown inux of eosinophils and neutrophils and a marked
Acetic acid Hospital setting increase in albumin concentration during late asthmatic reac-
Isocyanates Spray paint
tion after inhalation challenge with isocyanates [20]. In patients
with Western red cedar asthma, increases in eosinophils and
For further information see Ref. [2]. epithelial cells, as well as in albumin, histamine, and LTE4,

458
Occupational Agents 36
have also been found in the BAL uid [21]. Irrespective of the mesothelioma) [29]. Estimates of incidence of OA have been
sensitizing agent, the pathological features in the airways are made using registers based on mandated or voluntary physi-
similar. There is subepithelial brosis, hypertrophy of airway cian reporting, medicolegal statistics, and various national or
smooth muscle, edema of the airway wall, accumulation of disability registers. There are considerable between-country
inammatory cells, mostly eosinophils, and obstruction of the dierences in the estimated incidence of OA ranging from
airway lumen by exudate and/or mucus in patients with OA 22 per million per year in the United Kingdom to 187 per
[22]. Cessation of exposure to the sensitizing agent is associ- million per year in Finland. The dierences are likely due in
ated with a decrease in the number of inammatory cells in part to the methods used to derive these estimates. However,
the airway mucosa [23]. In isocyanate-induced asthma, some community-based studies on population-attributable risk
reversal of the subepithelial brosis has been found [24] but (PAR) of occupational exposure for asthma carried out in
airway eosinophilia and neutrophilia may persist. Studies of countries involved in the European Community Respiratory
induced sputum of patients with OA after cessation of expo- Health Survey using similar methodology have shown con-
sure for a mean interval of 8.7 years indeed showed that some siderable variation from 5% in Spain to 41% in New Zealand
subjects still had increased levels of neutrophils and eosinophils depending on the local industries, options for employment,
in induced sputum [25]. and the populations susceptibility [3032]. Studies in the
United States of adult patients with new-onset asthma have
Nonimmunologically induced asthma shown that about 8% reported a history of exposure to sen-
The mechanism of IIA is not known. IIA is a big bang sitizers at work and 13% when irritants were included [33,
phenomenon. Exposure to a high level of the irritant leads to 34]. The generally accepted PAR of occupational exposure
acute sloughing of the epithelium [3]. There are some dier- for asthma is around 915% [35, 36].
ences between the pathological features of IIA and those of Table 36.2 summarizes the ndings from selected
immunologically mediated asthma. In general, subepithelial cross-sectional studies for various workplaces [3746]. There
brosis is more evident in IIA [26]. In the acute phase, the is considerable variation in the prevalence of OA in dierent
airway epithelium is extensively damaged and the submucosa industries, ranging from 2% in latex-exposed workers to 50%
inltrated by mononuclear cells [27]. One study reported among detergent-enzyme workers [37]. While the between-
fewer T-lymphocytes in the airway mucosa in IIA than in workforce dierences can be due to varying data collection
allergen-induced asthma [28]. methodology, dierent denitions used for OA, and intensity
of exposure, it is quite possible that the asthmogenic poten-
tial of the agents is dierent.
Epidemiology: Frequency and Few longitudinal studies have been carried out in high-
risk apprenticeship programs including baking [47, 48] and
determinants students (bakers, animal health technicians and veterinarians,
dental hygienists, welders, spray painters) exposed to various
Frequency high [49, 50] and low [51, 52] molecular weight agents. This
OA has become one of the two most common occu- model is particularly interesting because it assesses young indi-
pational lung diseases in developed countries (the other is viduals who have not been previously exposed to the relevant

TABLE 36.2 Prevalence and determinants of work-related asthma: results of selected studies.

Prevalence

Work-related Smoking Atopy


Exposure/Industry No. Latency (months) asthma (%) (%) (%)

High molecular weight compounds

Enzyme/detergent [37] 98 N/A 50 52 64


Clam/Shrimp [38] 59 N/A 26 (8) 49 21
Snow-crab processor [39] 303 number unknown 21 (16) 67 11
Laboratory animal worker [40] 238 26 6 30 40
Bakery [41] 344 26 6 57 34
Latex/Hospital workers [42] 289 120 2 (3) 22 25

Low molecular weight compounds

Platinum refinery [43] 91 1224 54 63 33


Anhydrides, TCPA[44] 329 Up to 24 3.2 50 22
Toluene diisocyanate [45] 241 Up to 36 9.5 51 35
Plicatic acid/Red cedar sawmills [46] 652 N/A 4 38 19

For further information see Ref. [1].

459
Asthma and COPD: Basic Mechanisms and Clinical Management

workplace allergens and can best evaluate the natural history anhydride [44], and colophony [72]. As a result, a permissi-
of sensitization, symptoms, and disease even after beginning ble exposure limit has been proposed for a few occupational
of exposure and even after removal of exposure. agents [67]. The minimum concentration of occupational aller-
gen that causes sensitization may be one to two orders of mag-
Host determinants nitude greater than the concentration for eliciting symptoms.
Once an individual is sensitized to an agent, a minute dose can
Atopy (dened as positive skin test to one or more common trigger an attack of asthma. Concomitant environmental expo-
allergens) has been shown to be associated with sensitiza- sures, such as low levels of irritants and cigarette smoke, may
tion to some high molecular weight agents but not others enhance sensitization to some occupational agents. Further
(Table 36.2). The positive predictive value of atopy in various studies are required in this area.
studies of OA is low: 34% in animal laboratory workers [53,
54] and 7% in psyllium workers [55]. Moreover, over 40%
of young adults have positive skin-test reactions to common Diagnosis
allergens. These ndings do not justify routine screening for
atopy in high-risk workplaces. All asthmatic subjects should be questioned regarding possible
The relationship between smoking and OA is com- exposure to causal agents in their current or previous work-
plex and ndings of studies are often contradictory [56]. It places. It has been estimated that approximately 5% of all asth-
appears that the eect of smoking is dependent on the type matics seen in a specialized asthma clinic may suer from OA.
of occupational agent. An interaction between smoking and Also, it has to be remembered that because aected subjects
atopy has been found in OA in animal laboratory handlers could be left with permanent asthma after removal from the
[54] and in workers exposed to tetrachlorophthalic anhy- workplace, their current asthma symptoms could result from
dride [54]; atopic smokers had the highest prevalence of previous exposure to occupational causal agents. Physicians
sensitization and nonatopic nonsmokers, the lowest. Among should be aware that certain workplaces have a high risk of
platinum workers, smoking, not atopy, is the most important exposing subjects to asthma-causing agents. Information on
risk factor for sensitization [43]. Cigarette smoke probably workplaces at risk and on all agents causing OA is available
acts as an adjuvant to promote IgE sensitization [57]. When on the web site ASMANET.COM as prepared and reviewed
agents cause OA by IgE-independent mechanisms, as in the by Henriette Dhivert-Donnadieu and on ASTHMA-
cases of isocyanate-induced asthma and Western red cedar WORKPLACE.COM. Safety data sheets (SDS) on all prod-
asthma, both atopy and smoking are unimportant [58]. ucts used in the workplace can be obtained from employers
Studies of genetics of OA has proven to be dicult and/or from safety agencies. However, these SDS do not nec-
because of the small numbers of patients with asthma due essarily report all products present in the workplace. Although
to one type of occupational agents and the diculties of get- our and isocyanates are still the most frequent causes of OA,
ting appropriate controls. Nevertheless, there have been some many other high and low molecular weight agents emerge
studies showing association between HLA class II genes every year on the list of agents that often cause OA.
and several types of OA such as isocyanate-[59], trimellitic Clinical questionnaires should be sensitive although,
anhydride-[60], Western red cedar-[61], and platinum- in general, they are not very specic tools [73, 74]. Exposure
induced asthma [62]. However, such ndings should be con- to a known causal agent at work and the presence of asthma
sidered as preliminary [63] and this type of testing cannot be should be sucient to alert the physician to the possibil-
used for screening of susceptible subjects. Exposure to diiso- ity of OA even though the temporal relationship between
cyanates increases reactive oxygen species which are taken up exposure and symptoms may seem discordant. The possibil-
by the glutathione S-transferase enzyme [64]. Individuals ity of the presence of nasal or conjunctival symptoms should
with the glutathione S-transferase M1 null gene had a two- be addressed. Ocular and nasal symptoms often accompany
fold increase risk of diisocyanate-induced asthma [65]. The or even precede, in the case of high molecular weight agents,
combination of GSTM1 null and GTM3 AA phenotype the occurrence of OA. Moreover, ocular and nasal symptoms
was strongly associated with lack of diisocyanate-specic are good predictors of OA in the case of high molecular
IgE antibodies and with late asthmatic reaction [65]. weight agents [74].
Skin-prick tests can be done with extracts derived from
Exposure factors specic high molecular weight agents although these extracts
Improved industrial hygiene techniques have resulted in are not generally standardized. The presence of immediate
the ability to measure several low molecular weight com- skin sensitivity only indicates the presence of sensitization
pounds such as isocyanates, formaldehyde, and amines [66]. and does not conrm the diagnosis of OA. The target organ,
Immunochemical techniques for quantitating aeroallergens in this case the bronchial, should be shown to be hyperre-
have been developed and used in a number of workforce- sponsive. This can be done through the assessment of non-
based studies [67, 68]. allergic airway responsiveness. The combination of positive
Several studies have shown that there is a dose-response skin-test reaction to a relevant occupational allergen and
relationship between the level of exposure to occupational nonallergic airway hyperresponsiveness in a subject means
agents and the prevalence of sensitization and/or nonallergic that there is an 80% likelihood that he/she has OA [55].
bronchial hyperresponsiveness and/or asthma. These include Nonallergic airway hyperresponsiveness can be shown either
exposure to high molecular weight allergens such as -amylase by assessing the response to bronchodilators if there is airway
[69], animal laboratory allergen [70], and low molecular obstruction, or by estimating the degree of bronchoconstric-
weight compounds such as Western red cedar [71], acid tive response to a pharmacological agent. If the worker is still

460
Occupational Agents 36
employed, nonallergic airway responsiveness should be evalu- are most likely associated with conrmed diagnosis, negative
ated on a working day after a minimum period of 2 weeks at graphs cannot exclude OA [80]. Adding markers of airway
work. The absence of nonallergic airway hyperresponsiveness inammation (exhaled NO, induced sputum) for a period at
in a subject when still working and exposed to the agent(s) work and o work improves the accuracy of diagnosis [81].
suspected of causing OA virtually excludes OA. Exposing individuals to the potential causal agent(s)
Serial measurement of peak expiratory ow rates (PEF) in a hospital laboratory, or at the workplace under careful
in the diagnosis and management of asthma [75, 76] has been supervision, is a good method to conrm OA. This method
used since the late 1970s in the investigation of OA [77]. was rst proposed in the 1970s by Professor Jack Pepys at the
Workers are asked to measure and register their PEF at least Brompton Hospital in London, UK [82]. However, such tests
four times a day and to record their medication, symptoms, require the expertise of highly trained personnel and can only
and whether they are at work or away from work. Although be done in specialized centers. They should be performed
monitoring of PEF has been found to be a useful tool in the in a dose-response manner, exposing subjects to increasing
investigation of OA [77], there are several pitfalls including concentrations but nonirritant levels of the agent with serial
generally unsatisfactory compliance [78] and tracing, analysis monitoring of FEV1 following exposure for at least 8 h [83].
and interpretation of results, for which computerized methods The investigation of OA is a stepwise procedure, as
have been suggested. [79]. In our hands, while positive tracings illustrated in Fig. 36.1.

Clinical Investigation of Occupational Asthma

Compatible clinical history


and exposure to possible causal agents

Skin and RAST tests


(if possible)

Assessment of bronchial responsiveness


to pharmacological agents

Normal Increased

Subject still at work Subject no longer Subject still at work


at work

Laboratory challenges
with the suspected occupational agent

Positive Negative

Consider return to work

Workplace or laboratory challenges


with the suspected occupational agent
and/or PEF monitoring

Positive Negative

No asthma Occupational asthma Nonoccupational asthma

FIG. 36.1 Flow chart for the investigation of OA.

461
Asthma and COPD: Basic Mechanisms and Clinical Management

Management and compensation impairment/disability for asthma are: (1) airway caliber; (2)
airway responsiveness to a bronchodilator if airway obstruc-
Physicians may be asked to screen for OA in subjects exposed tion is present or to a bronchoconstrictor if it is not the case;
to known causal agents or in a workplace where cases of (3) the need for medication, which is a reection of the clini-
OA have been identied. These surveillance programs may cal severity of asthma [95]. Adding evaluation of persistent
include preemployment testing and periodic assessment. A airway inammation may help in this assessment [96].
cost-eectiveness examination of such programs needs to be
done. Preemployment testing to document the baseline status
may include a questionnaire, spirometry with assessment of Conclusion
nonallergic airway responsiveness, and skin testing to work-
related allergens in the case of exposure to high molecular Asthma is a common respiratory occupational ailment.
weight allergens. Workers with preexisting asthma should Whereas improvement in diagnostic tools and pathophysi-
not be excluded as there is no reason to believe that asth- ological mechanisms still need to be considered, emphasis
matic subjects are more likely to develop OA than anyone should be put on prevention programs through identication
else. It is dicult to recommend the frequency for screen- and application of permissible respirable levels and of aected
ing assessments. It has been estimated that 40% of subjects subjects at an early stage of sensitization or disease to prevent
with OA due to low molecular weight agents develop their long-term sequelae of permanent asthma.
symptoms during the rst year of exposure, while the cor-
responding gure for high molecular weight agents is 20%
[84]. After that, there is a progressive reduction in the rate of
development of OA. COPD
Prevention is relevant in high-risk workplaces where
inhalational accidents leading to IIA can occur. Assessment In most patients, COPD is caused by chronic inhalation
of airway responsiveness should also be considered as a of polluted air, specically, air contaminated by proinam-
preemployment test so that if an accident occurs compari- matory agents. The air can be polluted by personal choice
sons are possible [85]. (e.g. cigarette smoking), or by exposure sources in the ambient
Once the diagnosis of OA is conrmed, subjects should or work environment. Many patients may have been exposed
be advised to avoid exposure to the causal agent. Whereas to multiple sources, and the relative contributions of each may
wearing cartridge masks does not apparently reduce symp- be impossible to disentangle. COPD is the symptomatic and
tomatology or functional abnormalities [86], helmet masks functional consequence of chronic exposure to these polluted
may eliminate exposure completely and may be considered in air sources. Therefore, it seems appropriate to pose a deni-
some instances. Treatment with inhaled steroids, while keep- tion for occupational COPD that allows it to coexist with
ing the subject at work, improves the asthma but not to the smoking-induced COPD or urban air pollution-induced
extent of removal from exposure complemented by inhaled COPD. For the purpose of this section, we will dene occu-
steroids [87], especially when steroids are given early after pational COPD as the existence of COPD in a patient with
removal [88]. Subjects with OA whose symptoms persist a history of chronic exposure to proinammatory agents in
after removal from exposure should be treated in the same workplace air. This denition is useful for clinical manage-
way as patients with non-OA. ment and public health prevention as it points to the poten-
As for OA with a latency period, there might be func- tial for eliminating or modifying occupational risk factors for
tional recovery in the 2 years following the inhalation acci- the disease. Additional challenges with dening occupational
dent leading to IIA [89]. Steroids may be benecial [90]. COPD for legal or compensation purposes will be discussed
Patients with OA should be oered suitable help in below.
nding another job where there is no further exposure to
the causal agent, either with the same employer or another
one. Subjects aged 55 or over should be oered early retire- Epidemiology: Frequency and
ment, and young subjects should be retrained for a new job, determinants
all with nancial compensation. Compensation boards or
similar medicolegal agencies should oer these programs to The general population prevalence of occupational COPD
workers and assess their cost-eectiveness. The time needed is dicult to estimate due to the lack of occupational expo-
to make a diagnosis of OA and to implement a program is sure information in most population-based studies. After
generally too long, causing hardship to the subjects [91, 92]. reviewing all available published evidence from general
It has been estimated that during the 1990s, a single case of population epidemiologic studies, an ad hoc committee of
OA in Qubec cost approximately C$50,000, a gure that the American Thoracic Society recently estimated the con-
is currently C$75,000 [91]. tribution of occupational exposure to the overall population
Because OA can lead to permanent impairment/dis- burden of COPD to be at least 15% [97]. A 2001 study of
ability, subjects should be reassessed periodically. The rst 517 lifetime nonsmokers referred to a hospital pulmonary
assessment should take place 2 years after removal from function laboratory estimated the PAR among nonsmokers
exposure, when a plateau of improvement can occur, as in the from occupational exposure to be 23.6% for bronchitis and
case of a high molecular weight agent such as snow crab [93], 29.6% for obstructive lung disease [98].
although improvement can also occur at a later stage though Occupational COPD is associated with chronic expo-
at a slower pace [94]. The three main criteria for assessing sure to mineral and/or metal particulate matter and fumes,

462
Occupational Agents 36
organic particulate matter, combustion products, irritant looked specically for airow obstruction in these popula-
gases, and various combinations of these exposures. Evidence tions. Among miners and millers of wollastonite (another
for this has emerged primarily from epidemiological studies, brous dust) signicant dose-response relationships for
some of which are listed in Fig. 36.2. cumulative dust and airow obstruction were seen in both
nonsmokers and smokers [119]. COPD is also linked to
Mineral particulate particulate exposure (with and without crystalline silica) in
Prevalence rates for COPD with airow obstruction among other industries, such as quarrying, carbon black manufac-
miners range from 6% to 20% among nonsmokers, and turing [120122], and cement manufacturing [123].
up to 60% among smokers [104107]. Exposure-response
relationships are also seen. Airow obstruction was seen Metal fumes, irritant gases, combustion products
among 10.5% of nonsmoking British coal miners exposed Exposure to metal fumes, irritant gases, and combustion
to low dust levels and 20.6% of those exposed to medium products appears to augment the eect of exposure to dust
and high dust levels [106]. Similarly, among nonsmoking alone on COPD. This has been seen in mining and smelter
US underground coal miners the corresponding rates were workers [124, 125], workers in rubber manufacturing [126,
7.4% in the lower dust exposure category and 14.3% in the 127], welders [128132], tunnel workers [133], and re
higher dust exposure category [108, 109]. Chronic bron- ghters [134137]. There is some evidence that the risk of
chitis symptoms related to mining exposures are persistent airow obstruction in welders and smelter workers may be
even after cessation of exposure, as was seen among former higher among atopic workers, raising the possibility that at
Swedish iron ore miners after an average of 16 years of not least some of the excess airow obstruction seen in these
being exposed [110]. groups may be related to asthma [130]. Indeed, as discussed
Mine dust with greater crystalline silica content above in the section on OA, several metal fumes and irritant
appears to produce even higher COPD rates. Among South gases are recognized risk factors for OA.
African gold miners the estimated eect of dust exposure on
airow obstruction was approximately ten times greater than
that seen among coal miners [107, 111]. COPD mortality Organic dusts
is signicantly increased (in a dose-related fashion) among Although organic dust exposure is also associated with asthma
workers exposed to silica, especially in the presence of silico- and hypersensitivity pneumonitis, there is increasing evidence
sis [112, 113]. COPD has also been clearly demonstrated in that chronic respiratory symptoms and nonasthmatic airow
association with asbestos exposure [114118]. From the evi- obstruction are caused in part by exposure to both aller-
dence to date, airow obstruction appears more pronounced genic and nonallergenic organic dusts. For example, a recent
among workers installing or handling asbestos products study of nonasthmatic cedar sawmill workers found that the
than among miners [116]; however, only a few studies have annual decline in FVC was signicantly related to cumulative

Airflow Obstruction in Nonsmokers  Age 50


(FEV1/FVC% below lower 95% confidence limit)
25%

20%

15%

10%

5%

0%
Expected Blue-collar Wood Grain Aluminum Asbestos
rate controls workers handlers smelter insulators
Industry

FIG. 36.2 Prevalence rates for airflow obstruction (defined as FEV1/FVC % below the lower 95% confidence limit, based on age) among nonsmokers, aged
50 and over, exposed to mixed dusts and fumes and organic dusts, compared with a blue-collar control population [100] and to expected rates [101]; from
studies conducted by the UBC Occupational Lung Diseases Research Unit [99, 102, 103].

463
Asthma and COPD: Basic Mechanisms and Clinical Management

dust exposure, even when the average dust exposure was well British [106], US [108], and Italian [164] coal miners, among
below the accepted limit [138]. Studies of sawmill and furni- workers exposed to asbestos [165, 166], and grain [151, 156].
ture workers exposed to wood dusts from species not currently
known to cause asthma also show exposure-related airow
obstruction [139144]. Generally, in these studies, the excess Assessment of exposure
airow obstruction linked to dust exposure is similar among
nonsmokers and smokers. The relevant exposure for COPD is that to which the patient
There is also considerable evidence conrming a link was exposed in the past. Therefore, the best clinical tool for
between grain dust and COPD in both smokers and non- assessing this is a detailed occupational history, augmented
smokers, with consistent dose-response relationships found by specic inquiry about exposures to dusts, gases, and
[145150]. Among retired grain workers, moderate to fumes. In fact, research has shown that a positive response to
severe airow obstruction was found in 40% of nonsmokers the simple question have you been exposed to dusts, gases,
and 50% of smokers [151]. or fumes at work? is linked to a more rapid decline in FEV1
Mixed exposure to grains, other animal feeds, and and increased prevalence of chronic bronchitis in population
animal by-products are found in animal connement build- studies [167, 168]. Ideally, the occupational history should
ings, and there is growing evidence that exposure to these be reviewed by a professional with knowledge of the typical
environments leads to COPD. There is clear evidence of occupational exposures in the region. However, evidence has
excess chronic bronchitis among farmers and workers in shown that the patient is also a reliable source of informa-
poultry and swine connement buildings, with prevalence tion about exposure duration and intensity. For each main
rates remarkably consistent across studies at about 2535% job, the patient should be asked what year the job began and
[99, 152157]. Evidence for nonasthmatic chronic airow ended (to estimate duration of exposure); whether or not
obstruction in this industry is mixed [99, 157] although the there was noticeable dust, fumes, or gas exposure; and if so,
majority of studies indicate reductions in airow rates asso- how often (to estimate intensity of exposure).
ciated with duration of exposure [155, 158, 159]. COPD was Although it is not possible to provide a clear answer
found among 17% of 105 European farmers who had never to the question, How much exposure is necessary before
smoked and who worked in animal connement buildings, one should suspect an occupational contribution to COPD?
with 10% having moderate or severe disease by GOLD most research indicates that the relevant exposure duration
criteria. The highest disease prevalence was seen in farmers is measured in years (or even decades), not months or days.
with the highest dust and endotoxin exposure levels [160]. Many patients will have held more than one job, and expo-
sure duration should be summed over all jobs with relevant
exposures. Time spent in irregular work environments should
Relationship between cigarette smoking also be considered (e.g. in the armed forces, in prison, dur-
and occupational exposures ing extended periods of casual or temporary employment) as
hazardous exposures are common in these situations. Dust
The research indicates that occupational exposure plays a and fume exposures are seldom present every day, all day, so
clinically important role in the development of COPD in patients should be asked whether exposure occurred most
exposed workers. Among aluminum smelter workers, the days, only a few times a month, or only seldom (i.e. a few days
impact of a 30-year working career with particulate expo- a year). Exposure that occurred at least a few times a month
sure at the permitted level was about the same as smoking, should be considered relevant.
75 g/week [161]. Among tunnel workers exposed to dust Clinicians should be cautious about assuming that
and diesel exhaust, the decline in FEV1 associated with each workplace exposures within safe limits were not high enough
year of tunnel work was twice that associated with each to produce disease. Many of the studies discussed above found
pack year of cigarette smoking [133]. Among re ghters, signicant airow obstruction among workers exposed at or
the cumulative impact of being a re ghter on longitudi- below regulated allowable limits. It is useful to consider the
nal decline in FEV1 was about half as strong as the impact example of grain dust, which in many countries (including
of cigarette smoking [162]. Canada and much of the United States) is still regulated with
Cigarette smoking and dust exposure appear to exert reference to the nuisance dust standard, despite overwhelm-
their eect on airways in a roughly additive fashion regard- ing evidence that this standard is unacceptable. The American
less of the dust type [106, 111, 138] although there is a sug- Conference of Governmental and Industrial Hygienists has
gestion that the eect is more than additive in patients with adopted the term particulate not otherwise classied rather
marked airow obstruction, and in the presence of high than nuisance dust, to indicate that all materials are poten-
crystalline silica [163]. tially toxic and to avoid the implication that these materials
are harmless and to emphasize that although these materials
may not cause brosis and systemic eects, they are not bio-
Natural history logically inert [169].

Most research suggests that the natural history of COPD is


similar regardless of the source of the polluted air. However, Occupational COPD among women
there is growing evidence that there may be a link between
early rapid decline in airow rates due to occupational There is growing evidence that COPD mortality and mor-
exposures and later airow obstruction. This has been seen in bidity is rising among women relative to men and it has

464
Occupational Agents 36
been hypothesized that this may be linked to an increase evaluation should be carried out without reference to the
in tobacco exposure or susceptibility among women [170, question of etiology, as the level of disability or impairment
171]. However, other evidence indicates that smoking rates is unaected by the cause. The decision to attach the label
among adult women have declined in parallel with men possibly occupational to the diagnosis should also be based
since the 1980s, at least in Western countries [172], sug- on the occupational history. Although requirements vary
gesting that other factors must also be contributing to the among jurisdictions, it should be the attending physicians
apparent COPD increase among women. Environmental role to raise the possibility of occupational etiology, not to
(or domestic occupational) exposure to biomass particulate make the nal determination in law. General guidance can
matter (from indoor cooking) has been strongly linked to be found in the following statement from the BC Workers
COPD among women in developing countries. The poten- Compensation Board policy manual: Since workers com-
tial impact on COPD of womens work and changing occu- pensation operates on an enquiry basis rather than on an
pational environments has only been explored in a few very adversarial basis, there is no onus on the worker to prove his
recent studies, but there is some evidence of possible dif- or her case. All that is needed is for the worker to describe
ferential susceptibility to biological dusts among women. In his or her experience of the disease and the reasons why they
a 20-year follow-up study of Chinese cotton textile work- suspect the disease has an occupational basis. Then it is the
ers, nonsmoking men demonstrated a small improvement responsibility of the Board to research the available scientic
in airow rates after cessation of exposure (reduced decline literature and carry out any other investigations into the ori-
in FEV1), whereas no such improvement was seen among gin of the workers condition which may be necessary.
women, despite higher dust and endotoxin exposures That said, unfortunately, physicians do nd them-
among men [173]. Among a random sample of Australians, selves called upon by legal tribunals, to quantify the relative
employment in a job with exposure to biological dust was contribution of occupational exposure to COPD. This can
a strong and a signicant risk factor for COPD among be extremely dicult, if not impossible, in the absence of
women (sevenfold increase in risk); this eect was not seen detailed exposure information and the expertise to interpret
in men (no increased risk) [174]. Jobs with biological dust it. A useful benchmark may be the research among smelter
exposure held by women in this study included health care workers, discussed earlier in this section, that found the
professions, food and textile workers, artists, and clean- eect of working for 30 years at todays accepted exposure
ers. An analysis of data from the US National Health and concentration had an impact on airow of about the same
Nutrition Examination Survey [175] indicated an increased magnitude as a similar duration of cigarette smoking.
risk for COPD, using GOLD criteria (compared to the
population as a whole) among women in personal services,
agriculture, textiles, rubber and plastics industries, and sales- Prevention
related jobs. These very preliminary ndings require further
investigation to understand and quantify the potential role Some evidence suggests that a rapid decline in FEV1 (but
of occupational exposures in contributing to the growing still in the normal range) in young exposed workers may be
prevalence of COPD among women. They do point, how- associated with a worse prognosis. Therefore, although these
ever, to the importance of considering occupational risks workers are unlikely to seek treatment, if such a patient is
even among female patients with jobs not traditionally con- encountered (e.g. as a result of routine screening or for other
sidered dusty or industrial. reasons) the index of concern should rise. The evidence
is not strong enough to suggest that such a worker should
be removed from exposure, but the patient should be made
Management and compensation aware of his or her excess decline in lung function and the
potential role of occupational exposure should be explored.
When the clinician sees a patient (male or female) with Global prevention of occupational COPD will require
chronic nonspecic airow limitation (regardless of whether increased recognition of the disease (by physicians, by regula-
or not that patient is a smoker), it is useful to consider the tors, and by employers), and a willingness by regulators and
possibility that occupational exposures may have contrib- employers to act to reduce exposure to particulates and irritant
uted to the disease. This is important as continued occupa- gases and fumes in the work environment. Primary care and
tional exposure may contribute to a worsening of the disease pulmonary physicians can play a major role in prevention by
(in the patient, or in others in that workplace), if no preven- increasing the recognition of the disease, by reporting it, even
tive action is taken. For the patient still exposed to particu- if just as suspect or possible occupational COPD, to the
late or irritant gases or fumes, a recommendation should be local agency responsible for occupational disease prevention.
made to reduce or eliminate the potential for the exposure.
For the patient no longer exposed (or no longer working), it
is important to record and report the disease as potentially References
occupational in order that public health ocials can assess
the role of the occupational exposure and use this informa- 1. Bernstein IL, Chan-Yeung M, Malo JL, Bernstein DI. Asthma in the
tion to direct prevention activities in the workplace. Workplace, 3rd edn. New York, NY: Taylor & Francis, 2006.
Whether or not a patient with occupational COPD is 2. Malo JL, Chan-Yeung M. Agents causing occupational asthma with
entitled to compensation or disability, benets will depend key references. Appendix. In: Bernstein IL, Chan-Yeung M, Malo JL,
on the specic requirements of the compensation or insur- Bernstein DI (eds). Asthma in the Workplace, 82566. New York: Taylor &
ance carrier. These requirements vary broadly. Disability Francis Inc, 2006.

465
Asthma and COPD: Basic Mechanisms and Clinical Management

3. Gautrin D, Bernstein IL, Brooks SM, Henneberger PK. Reactive airways removal from the causal agent in occupational asthma due to high and
dysfunction syndrome and irritant-induced asthma. In: Bernstein IL, low molecular weight agents. Eur Respir J 7: 156775, 1994.
Chan-Yeung M, Malo JL, Bernstein DI (eds). Asthma in the Workplace, 23. Chan-Yeung M, Leriche J, Maclean L, Lam S. Comparison of cellu-
3rd edn, 579627. New York, NY: Taylor & Francis, 2006. lar and protein changes in bronchial lavage uid of symptomatic and
4. Patterson R, Zeiss CR, Roberts M, Pruzansky JJ. Human antihapten asymptomatic patients with red cedar asthma on follow-up examina-
antibodies in trimellitic anhydride inhalation reactions. J Clin Invest tion. Clin Exp Allergy 18: 35965, 1988.
62: 97178, 1978. 24. Saetta M, Maestrelli P, Turato G, Mapp CE, Milani G, Pivirotto F,
5. Pepys J. Occupational allergy due to platinum complex salts. Clin Fabbri LM, DiStefano A. Airway wall remodeling after cessation of
Immunol Allergy 4(1): 13158, 1984. exposure to isocyanates in sensitized asthmatic subjects. Am J Respir
6. Graham C, Rosenkranz HS, Karol MH. Structure-activity model of Crit Care Med 151: 48994, 1995.
chemicals that cause human respiratory sensitization. Regul Toxicol 25. Maghni K, Lemire C, Ghezzo H, Yuquan W, Malo JL. Airway
Pharmacol 26: 296306, 1997. inammation after cessation of exposure to agents causing occupational
7. Jarvis J, Seed MJ, Elton R, Sawyer L, Agius R. Relationship between asthma. Am J Respir Crit Care Med 169: 36772, 2004.
chemical structure and the occupational asthma hazard of low molecu- 26. Gautrin D, Boulet LP, Boutet M, Dugas M, Bhrer L, LArchevque J,
lar weight organic compounds. Occup Environ Med 62: 24350, 2005. Laviolette M, Ct J, Malo JL. Is reactive airways dysfunction syn-
8. Maestrelli P, Fabbri LM, Mapp CE. Pathophysiology. In: Bernstein IL, drome a variant of occupational asthma? J Allergy Clin Immunol 93:
Chan-Yeung M, Malo JL, Bernstein DI (eds). Asthma in the Workplace, 1222, 1994.
3rd edn, 10940. New York: Taylor & Francis, 2006. 27. Lemire C, Malo JL, Boulet LP, Boutet M. Reactive airways dysfunc-
9. Cartier A, Grammer L, Malo JL, Lagier F, Ghezzo H, Harris K, tion syndrome induced by exposure to a mixture containing isocyanate:
Patterson R. Specic serum antibodies against isocyanates: Association Functional and histopathologic behaviour. Allergy 51: 26265, 1996.
with occupational asthma. J Allergy Clin Immunol 84: 50714, 1989. 28. Chan-Yeung M, Lam S, Kennedy SM, Frew AJ. Persistent asthma after
10. Tee RD, Cullinan P, Welch J, Burge PS, Newman-Taylor AJ. Specic repeated exposure to high concentrations of gases in pulpmills. Am J
IgE to isocyanates: A useful diagnostic role in occupational asthma. Respir Crit Care Med 149: 167680, 1994.
J Allergy Clin Immunol 101: 70915, 1998. 29. McDonald JC, Keynes HL, Meredith SK. Reported incidence of occu-
11. Jones MG, Floyd A, Nouri-Aria KT, Jacobson MR, Durham SR, pational asthma in the United Kingdom, 198997. Occup Environ Med
Taylor AN. Is occupational asthma to diisocyanates a non-IgE- 57: 82329, 2000.
mediated disease? J Allergy Clin Immunol 117: 66369, 2006. 30. Kogevinas M, Anto JM, Soriano JB, Tobias A, Burney P. The risk of
12. Frew A, Chan H, Dryden P, Salari H, Lam S, Chan-Yeung M. asthma attributable to occupational exposures. Am J Respir Crit Care
Immunologic studies of the mechanisms of occupational asthma caused Med 154: 13743, 1996.
by western red cedar. J Allergy Clin Immunol 92: 46678, 1993. 31. Kogevinas M, Anto JM, Sunyer J, Tobias A, Kromhout H, Burney P
13. Maestrelli P, DelPrete GF, DeCarli M, Saetta M, DiStefano A, Ricci M, Group and the European Community Respiratory Health Survey
Romagnani S, Fabbri LM. Activated CD8 T lymphocytes producing Study. Occupational asthma in Europe and other industrialised areas:
interferon-gamma (IFN-gamma) and interleukin-5 (IL-5) in bronchial A population-based study. Lancet 353: 175054, 1999.
mucosa of subjects sensitized to toluene diisocyanate (TDI). Scand 32. Fishwick D, Pearce N, DSouza W, Lewis S, Town I, Armstrong R,
J Work Environ Health 20: 37681, 1999. Kogevinas M, Crane J. Occupational asthma in New Zealanders: A
14. Frew A, Chang JH, Chan H, Quirce S, Noertjojo K, Keown P, Chan- population based study. Occup Environ Med 54: 3016, 1997.
Yeung M. T-lymphocyte responses to plicatic acid-human serum albu- 33. Blanc PD, Cisternas M, Smith S, Yelin E. Occupational asthma in a
min conjugate in occupational asthma caused by western red cedar. community-based survey of adult asthma. Chest 109: 56S57S, 1996.
J Allergy Clin Immunol 101: 84147, 1998. 34. Blanc PD, Eisner MD, Israel L, Yelin EH. The association between
15. Mapp CE, Boschetto P, Miotto D, de RE. Asthma induced by isocy- occupation and asthma in general medical practice. Chest 115: 1259
anates: A model of IgE independent asthma. Acta Biomed 76(Suppl 2): 64, 1999.
1519, 2005. 35. Blanc PD, Toren K. How much asthma can be attributed to occupa-
16. Ogawa H, Inoue S, Ogushi F, Ogura H, Nakamura Y. Toluene diisocy- tional factors? Am J Med 107: 58087, 1999.
anate (TDI) induces production of inammatory cytokines and chem- 36. American Thoracic Society. Occupational contribution to the burden of
okines by bronchial epithelial cells via the epidermal growth factor airway disease. Am J Respir Crit Care Med 167: 78797, 2003.
receptor and p38 mitogen-activated protein kinase pathways. Exp Lung 37. Mitchell CA, Gandevia B. Respiratory symptoms and skin reactivity in
Res 32: 24562, 2006. workers exposed to proteolytic enzymes in the detergent industry. Am
17. Kennedy SM, Enarson DA, Janssen RG, Chan-Yeung M. Lung health Rev Respir Dis 104: 112, 1971.
consequences of reported accidental chlorine gas exposures among 38. Desjardins A, Malo JL, LArchevque J, Cartier A, McCants M,
pulpmill workers. Am Rev Respir Dis 143: 7479, 1991. Lehrer SB. Occupational IgE-mediated sensitization and asthma due
18. Kern DG. Outbreak of the reactive airways dysfunction syndrome after to clam and shrimp. J Allergy Clin Immunol 96: 60817, 1995.
a spill of glacial acetic acid. Am Rev Respir Dis 144: 105864, 1991. 39. Cartier A, Malo JL, Forest F, Lafrance M, Pineau L, St-Aubin JJ,
19. Bhrer L, Cushman R, Courteau JP, Quvillon M, Ct G, Bourbeau J, Dubois JY. Occupational asthma in snow crab-processing workers.
LArchevque J, Cartier A, Malo JL. Survey of construction workers J Allergy Clin Immunol 74: 26169, 1984.
repeatedly exposed to chlorine over a three to six month period in a 40. Cullinan P, Lowson D, Nieuwenshuijsen MJ, Sandiford C, Tee RD,
pulpmill: II. Follow up of aected workers by questionnaire, spirom- Venables KM, McDonald JC, Newman Taylor AJ. Work related symp-
etry and assessment of bronchial responsiveness 18 to 24 months after toms, sensitisation, and estimated exposure in workers not previously
exposure. Occup Environ Med 51: 22528, 1994. exposed to our. Occup Environ Med 51: 57983, 1994.
20. Saetta M, Di Stefano A, Maestrelli P, De Marzo N, Milani GF, 41. Cullinan P, Lowson D, Nieuwenhuijsen MJ, Gordon S, Tee RD,
Pivirotto F, Mapp CE, Fabbri LM. Airway mucosal inammation in Venables KM, McDonald JC, Newman Taylor AJ. Work related
occupational asthma induced by toluene diisocyanate. Am Rev Respir symptoms, sensitisation, and estimated exposure in workers not
Dis 145: 16068, 1992. previously exposed to laboratory rats. Occup Environ Med 51: 58992,
21. Lam S, LeRiche J, Phillips D, Chan-Yeung M. Cellular and pro- 1994.
tein changes in bronchial lavage uid after late asthmatic reaction in 42. Vandenplas O, Delwich JP, Evrard G, Aimont P, van der Brempt X,
patients with red cedar asthma. J Allergy Clin Immunol 80: 4450, 1987. Jamart J, Delaunois L. Prevalence of occupational asthma due to latex
22. Boulet LP, Boutet M, Laviolette M, Dugas M, Milot J, Leblanc C, among hospital personnel. Am J Respir Crit Care Med 151: 5460,
Paquette L, Ct J, Cartier A, Malo JL. Airway inammation after 1995.

466
Occupational Agents 36
43. Venables KM, Dally MB, Nunn AJ, Stevens JF, Stephens R, Farrer N, 64. Lange RW, Day BW, Lemus R, Tyurin VA, Kagan VE, Karol MH.
Hunter JV, Stewart M, Hughes EG, Newman Taylor AJ. Smoking and Intracellular S-glutathionyl adducts in murine lung and human bron-
occupational allergy in workers in a platinum renery. BMJ 299: 939 choepithelial cells after exposure to diisocyanatotoluene. Chem Res
42, 1989. Toxicol 12: 93136, 1999.
44. Venables KM, Newman Taylor AJ. Exposure-response relationships in 65. Piirila P, Wikman H, Luukkonen R, Kaaria K, Rosenberg C,
asthma caused by tetrachlorophthalic anhydride. J Allergy Clin Immunol Nordman H, Norppa H, Vainio H, Hirvonen A. Glutathione S-
85: 5558, 1990. transferase genotypes and allergic responses to diisocyanate exposure.
45. Venables KM, Dally MB, Burge PS, Pickering CAC, Newman Pharmacogenetics 11: 43745, 2001.
Taylor AJ. Occupational asthma in a steel coating plant. Br J Ind Med 66. Lesage J, Perrault G. Environmental monitoring of chemical agents. In:
42: 51724, 1985. Bernstein IL, Chan-Yeung M, Malo JL, Bernstein DI (eds). Asthma in
46. Chan-Yeung M, Vedal S, Kus J, Maclean L, Enarson D, Tse KS. the Workplace, 3rd edition. 297318. New York: Taylor & Francis, 2006.
Symptoms, pulmonary function, and bronchial hyperreactivity in 67. Nieuwenhuijsen M, Baur X, Heederik D. Environmental monitoring:
Western Red Cedar workers compared with those in oce workers. General considerations, exposure-response relationships, and risk assess-
Am Rev Respir Dis 130: 103841, 1984. ment. In: Bernstein IL, Chan-Yeung M, Malo JL, Bernstein DI (eds).
47. De Zotti R, Bovenzi M. Prospective study of work related respiratory Asthma and the Workplace, 3rd edition. 25374. New York: Taylor &
symptoms in trainee bakers. Occup Environ Med 57: 5861, 2000. Francis, 2006.
48. Walusiak J, Hanke W, Gorski P, Palczynski C. Respiratory allergy in 68. Swanson MC, Heederik D. Quantication of bio-active protein aerosols.
apprentice bakers: Do occupational allergies follow the allergic march? In: Bernstein IL, Chan-Yeung M, Malo JL, Bernstein DI (eds). Asthma
Allergy 59: 44250, 2004. in the Workplace, 3rd edition. 27596. New York: Taylor & Francis, 2006.
49. Gautrin D, Ghezzo H, Infante-Rivard C, Malo JL. Incidence and 69. Houba R, Heederik DJJ, Doekes G, van Run PEM. Exposure-
determinants of IgE-mediated sensitization in apprentices: A prospec- sensitization relationship for -amylase allergens in the baking indus-
tive study. Am J Respir Crit Care Med 162: 122228, 2000. try. Am J Respir Crit Care Med 154: 13036, 1996.
50. Malo JL, Chan YM. Occupational asthma. J Allergy Clin Immunol 108: 70. Heederik D, Venables KM, Malmberg P, Hollander A, Karlsson AS,
31728, 2001. Renstrom A, Doekes G, Nieuwenhijsen M, Gordon S. Exposure-response
51. Dragos DMC, Malo JL, Gautrin D. Specic IgG and IgE to hexam- relationships for work-related sensitization in workers exposed to rat uri-
ethylene diisocyanate (HDI) among car-painting apprentices at risk of nary allergens: Results from a pooled study. J Allergy Clin Immunol 103:
developing occupational asthma. Am J Respir Crit Care Med 169: A647, 67884, 1999.
2004. 71. Vedal S, Chan-Yeung M, Enarson D, Fera T, Maclean L, Tse KS,
52. El-Zein M, Malo JL, Infante-Rivard C, Gautrin D. Incidence of prob- Langille R. Symptoms and pulmonary function in western red cedar
able occupational asthma and of changes in airway calibre and respon- workers related to duration of employment and dust exposure. Arch
siveness in apprentice welders. Eur Respir J 22: 51318, 2003. Environ Health 41: 17983, 1986.
53. Slovak AJ, Hill RN. Laboratory animal allergy: A clinical survey of an 72. Burge PS, Edge G, Hawkins R, White V, Taylor AN. Occupational
exposed population. Br J Ind Med 38: 3841, 1981. asthma in a factory making ux-cored solder containing colophony.
54. Venables KM, Upton JL, Hawkins ER, Tee RD, Longbottom JL, Thorax 36: 82834, 1981.
Newman-Taylor AJ. Smoking, atopy and laboratory animal allergy. Br J 73. Malo JL, Ghezzo H, LArchevque J, Lagier F, Perrin B, Cartier A.
Ind Med 45: 66771, 1988. Is the clinical history a satisfactory means of diagnosing occupational
55. Malo JL, Cartier A, LArchevque J, Ghezzo H, Lagier F, Trudeau C, asthma? Am Rev Respir Dis 143: 52832, 1991.
Dolovich J. Prevalence of occupational asthma and immunologic sensi- 74. Vandenplas O, Ghezzo H, Munoz X, Moscato G, Perfetti L,
tization to psyllium among health personnel in chronic care hospitals. Lemire C, Labrecque M, LArchevque J, Malo JL. What are the
Am Rev Respir Dis 142: 135966, 1990. questionnaire items most useful in identifying subjects with occupa-
56. Siracusa A, Marabini A, Folletti I, Moscato G. Smoking and occupa- tional asthma? Eur Respir J 26: 105663, 2005.
tional asthma. Clin Exp Allergy 36: 57784, 2006. 75. Epstein SW, Fletcher CM, Oppenheimer EA. Daily peak ow meas-
57. Nielsen GD, Olsen O, Larsen ST, Lovik M, Poulsen LK, Glue C, urements in the assessment of steroid therapy for airway obstruction.
Brandor NP, Nielsen PJ. IgE-mediated sensitisation, rhinitis and BMJ 1: 22325, 1969.
asthma from occupational exposures. Smoking as a model for airborne 76. Turner-Warwick M. On observing patterns of airow obstruction in
adjuvants?. Toxicology 216: 87105, 2005. chronic asthma. Br J Dis Chest 71: 7386, 1977.
58. Mapp CE, Saetta M, Maestrelli P, Ciacia A, Fabbri LM. Low molec- 77. Burge PS, Moscato G, Johnson A, Chan-Yeung M. Physiological
ular weight pollutants and asthma: Pathogenetic mechanisms and assessment: Serial measurements of lung function and bronchial
genetic factors. Eur Respir J 7: 155963, 1994. responsiveness. In: Bernstein IL, Chan-Yeung M, Malo JL, Bernstein
59. Bignon JS, Aron Y, Ju LY, Kopferschmitt MC, Garnier R, Mapp C, DI (eds). Asthma in the Workplace, 3rd edn, 199226. New York, NY:
Fabbri LM, Pauli G, Lockhart A, Charron D, Swierczewski E. HLA Francis & Taylor, 2006.
class II alleles in isocyanate-induced asthma. Am J Respir Crit Care 78. Malo JL, Trudeau C, Ghezzo H, LArchevque J, Cartier A. Do sub-
Med 149: 7175, 1994. jects investigated for occupational asthma through serial PEF measure-
60. Young RP, Barker RD, Pile KD, Cookson WOCM, Newman TAJ. The ments falsify their results? J Allergy Clin Immunol 96: 6017, 1995.
association of HLA DR3 with specic IgE to inhaled acid anhydrides. 79. Burge PS, Pantin CFA, Newton DT, Gannon PFG, Bright P, Belcher J,
Am J Respir Crit Care Med 151: 21921, 1995. McCoach J, Baldwin DR, Burge CBSG. Development of an expert
61. Horne C, Quintana PJE, Keown PA, Dimich-Ward H, Chan-Yeung M. system for the interpretation of serial peak expiratory ow measure-
Distribution of HLA class II DQB1 alleles in patients with occupational ments in the diagnosis of occupational asthma. Occup Environ Med 56:
asthma due to Western red cedar. Eur Respir J 15: 91114, 2000. 75864, 1999.
62. Newman Taylor AJ, Cullinan P, Lympany PA, Harris JM, Dowdeswell RJ, 80. Leroyer C, Perfetti L, Trudeau C, LArchevque J, Chan-Yeung M,
DuBois R. Interaction of HLA phenotype and exposure intensity in Malo JL. Comparison of serial monitoring of peak expiratory ow and
sensitization to complex platinum salts. Am J Respir Crit Care Med 160: FEV1 in the diagnosis of occupational asthma. Am J Respir Crit Care
43538, 1999. Med 158: 82732, 1998.
63. Newman Taylor AJ, Yucesov B. Genetics and occupational asthma. In: 81. Girard F, Chaboillez S, Cartier A, Ct J, Hargreave FE, Labrecque M,
Bernstein IL, Chan-Yeung M, Malo JL, Bernstein DI (eds). Asthma in Malo JL, Tarlo S, Lemire C. An eective strategy for diagnosing
the Workplace, 3rd edn, pp. 87108. New York, NY: Taylor & Francis, occupational asthma. Induced sputum. Am J Respir Crit Care Med 170:
2006. 84550, 2004.

467
Asthma and COPD: Basic Mechanisms and Clinical Management

82. Pepys J, Hutchcroft BJ. Bronchial provocation tests in etiologic diag- 102. Chan-Yeung M, Enarson DA, MacLean L, Irving D. Longitudinal
nosis and analysis of asthma. Am Rev Respir Dis 112: 82959, 1975. study of workers in an aluminum smelter. Arch Environ Health 44:
83. Vandenplas O, Malo JL. Inhalation challenges with agents causing 13439, 1989.
occupational asthma. Eur Respir J 10: 261229, 1997. 103. Kennedy SM, Vedal S, Muller N, Kassam A, Chan-Yeung M. Lung
84. Malo JL, Ghezzo H, DAquino C, LArchevque J, Cartier A, Chan- function and chest radiograph abnormalities among construction
Yeung M. Natural history of occupational asthma: Relevance of type insulators. Am J Ind Med 20: 67384, 1991.
of agent and other factors in the rate of development of symptoms in 104. Fairman RP, OBrien RJ, Swecker S, Amandus HE, Shoub EP.
aected subjects. J Allergy Clin Immunol 90: 93744, 1992. Respiratory status of surface coal miners in the United States. Arch
85. Leroyer C, Malo JL, Infante-Rivard C, Dufour JG, Gautrin D. Environ Health 32: 21115, 1977.
Changes in airway function and bronchial responsiveness after acute 105. Kibelstis JA, Morgan EJ, Reger R, Lapp NL, Seaton A, Morgan WKC.
occupational exposure to chlorine leading to treatment in a rst-aid Prevalence of bronchitis and airway obstruction in American bitumi-
unit. Occup Environ Med 55: 35659, 1998. nous coal miners. Am Rev Respir Dis 108: 88693, 1973.
86. Ct J, Kennedy S, Chan-Yeung M. Outcome of patients with cedar 106. Marine WM, Gurr D, Jacobsen M. Clinically important respiratory
asthma with continuous exposure. Am Rev Respir Dis 141: 37376, eects of dust exposure and smoking in British coal miners. Am Rev
1990. Respir Dis 137: 10612, 1988.
87. Moscato G, Dellabianca A, Perfetti L, Brame B, Galdi E, Niniano R, 107. Oxman AD, Muir DC, Shannon HS, Stock SR, Hnizdo E, Lange HJ.
Paggiaro P. Occupational asthma. A longitudinal study on the clinical Occupational dust exposure and chronic obstructive pulmonary dis-
and socioeconomic outcome after diagnosis. Chest 115: 24956, 1999. ease. A systematic overview of the evidence (see comments). Am Rev
88. Malo JL, Cartier A, Ct J, Milot J, Leblanc C, Paquette L, Respir Dis 148: 3848, 1993.
Ghezzo H, Boulet LP. Inuence of inhaled steroids on the recovery 108. Seixas NS, Robins TG, Atteld MD, Moulton LH. Exposure-
of occupational asthma after cessation of exposure: An 18-month response relationships for coal mine dust and obstructive lung disease
double-blind cross-over study. Am J Respir Crit Care Med 153: following enactment of the Federal Coal Mine Health and Safety Act
95360, 1996. of 1969. Am J Ind Med 21: 71534, 1992.
89. Malo JL, Cartier A, Boulet LP, LArchevque J, Saint-Denis F, 109. Seixas NS, Robins TG, Atteld MD, Moulton LH. Longitudinal and
Bhrer L, Courteau JP. Bronchial hyperresponsiveness can improve cross sectional analyses of exposure to coal mine dust and pulmonary
while spirometry plateaus two to three years after repeated exposure function in new miners. Br J Ind Med 50: 92937, 1993.
to chlorine causing respiratory symptoms. Am J Respir Crit Care Med 110. Hedlund U, Jarvholm B, Lundback B. Respiratory symptoms and
150: 114245, 1994. obstructive lung diseases in iron ore miners: Report from the obstruc-
90. Demnati R, Fraser R, Martin JG, Plaa G, Malo JL. Eects of dexam- tive lung disease in northern Sweden studies. Eur J Epidemiol 19:
ethasone on functional and pathological changes in rat bronchi caused 95358, 2004.
by high acute exposure to chlorine. Toxicol Sci 45: 24246, 1998. 111. Hnizdo E. Combined eect of silica dust and tobacco smoking on
91. Dewitte JD, Chan-Yeung M, Malo JL. Medicolegal and compensa- mortality from chronic obstructive lung disease in gold miners. Br J
tion aspects of occupational asthma. Eur Respir J 7: 96980, 1994. Ind Med 47: 65664, 1990.
92. Ameille J, Parion JC, Bayeux MC, Brochard P, Choudat D, Conso F, 112. Calvert GM, Rice FL, Boiano JM, Sheehy JW, Sanderson WT.
Devienne A, Garnier R, Iwatsubo Y. Consequences of occupational Occupational silica exposure and risk of various diseases: An analy-
asthma on employment and nancial status: A follow-up study. Eur sis using death certicates from 27 states of the United States. Occup
Respir J 10: 5558, 1997. Environ Med 60: 12229, 2003.
93. Malo JL, Cartier A, Ghezzo H, Lafrance M, Mccants M, Lehrer SB. 113. Tse LA, Yu IT, Leung CC, Tam W, Wong TW. Mortality from non-
Patterns of improvement of spirometry, bronchial hyperresponsive- malignant respiratory diseases among people with silicosis in Hong
ness, and specic IgE antibody levels after cessation of exposure in Kong: Exposure-response analyses for exposure to silica dust. Occup
occupational asthma caused by snow-crab processing. Am Rev Respir Environ Med 64: 8792, 2007.
Dis 138: 80712, 1988. 114. Rodriquez-Roisin R, Merchant JA, Cochrane GE, Hickey BPH.
94. Malo JL, Ghezzo H. Recovery of methacholine responsiveness after Maximal expiratory ow volume curves in workers exposed to asbes-
end of exposure in occupational asthma. Am J Respir Crit Care Med tos. Respiration 39: 15865, 1980.
169: 13047, 2004. 115. Kilburn KH, Warshaw RH, Einstein K, Bernstein J. Airway disease
95. American Medical Association. Guides to the Evaluation of Permanent in non-smoking asbestos workers. Arch Environ Health 40: 29395,
Impairment, 5th edn, Cocchiarella L, Andersson GBJ (eds). Chicago, 1985.
IL, 2001. 116. Bgin R, Boileau R, Peloquin S. Asbestos exposure, cigarette smok-
96. Yacoub MR, Lavoie K, Lacoste G, Daigle S, LArchevque J, ing, and airow limitation in long-term Canadian chrysotile miners
Ghezzo H, Lemire C, Malo JL. Assessment of impairment/disabil- and millers. Am J Ind Med 11: 5566, 1987.
ity due to occupational asthma through a multidimensional approach. 117. Kilburn KH, Warshaw RH. Abnormal pulmonary function associated
Eur Respir J 29:88996, 2007. with diaphragmatic pleural plaques due to exposure to asbestos. Br J
97. Balmes J, Becklake M, Blanc P et al. American thoracic society state- Ind Med 47: 61114, 1990.
ment on occupational contribution to the burden of airway disease. 118. Demers RY, Neale AV, Robins T, Herman SC. Asbestos-related
Am J Respir Crit Care Med 167: 78797, 2003. disease in boilermakers. Am J Ind Med 17: 32739, 1990.
98. Mak GK, Gould MK, Kuschner WG. Occupational inhalant exposure 119. Hanke W, Sepulveda MJ, Watson A, Jankovic J. Respiratory morbid-
and respiratory disorders among never-smokers referred to a hospital ity in wollastonite workers. Br J Ind Med 41: 47479, 1984.
pulmonary function laboratory. Am J Med Sci 322: 12126, 2001. 120. Malmberg P, Hendenstm H, Sundblad BM. Changes in lung
99. Holness DL, OBlenis EL, Sass-Kortsak AM, Pilger CW, Nethercott JR. function in granite crushers exposed to moderately high silica con-
Respiratory eects and dust exposures in hog connement farming. Am J centrations: A 12 year follow-up. Br J Ind Med 50: 72531, 1993.
Ind Med 11: 57180, 1987. 121. Gardiner K, Thethowan NW, Harrington JM, Rossiter CE, Glass
100. Kennedy SM, Chan-Yeung M, Marion S, Lea J, Teschke K. Maintenance DC. Occupational exposure to carbon monoxide and sulphur dioxide
of stellite and tungsten carbide saw tips: Respiratory health and exposure- during the manufacture of carbon black. Ann Occup Hyg 36: 36372,
response evaluations. Occup Environ Med 52: 18591, 1995. 1992.
101. Crapo RO, Morris AH, Gardner RM. Reference spirometric values 122. Gardiner K, Thethowan NW, Harrington JM, Rossiter CE, Calvert IA.
using techniques and equipment that meet ATS recommendations. Respiratory health eects of carbon black: A survey of European car-
Am Rev Respir Dis 123: 65964, 1981. bon black workers. Br J Ind Med 50: 108296, 1993.

468
Occupational Agents 36
123. Mwaiselage J, Bratveit M, Moen BE, Mashalla Y. Respiratory 146. Enarson DA, Vedal S, Chan-Yeung M. Rapid decline in FEV1 in
symptoms and chronic obstructive pulmonary disease among cement grain handlers. Relation to level of dust exposure. Am Rev Respir Dis
factory workers. Scand J Work Environ Health 31: 31623, 2005. 132: 81417, 1985.
124. Manfreda J, Cheang M, Warren CP. Chronic respiratory disorders 147. Huy T, De Schipper K, Chan-Yeung M, Kennedy SM. Grain dust
related to farming and exposure to grain dust in a rural adult com- and lung function. Dose-response relationships. Am Rev Respir Dis
munity. Am J Ind Med 15: 719, 1989. 144: 131421, 1991.
125. Kennedy SM, Wright JL, Mullen JB, Pare PD, Hogg JC. Pulmonary 148. Smid T, Heederik D, Houba R, Quanjer PH. Dust- and endotoxin-
function and peripheral airway disease in patients with mineral dust related respiratory eects in the animal feed industry. Am Rev Respir
or fume exposure. Am Rev Respir Dis 132: 129499, 1985. Dis 146: 147479, 1992.
126. Fine LJ, Peters JM. Respiratory morbidity in rubber workers: II. 149. Jorna THJM, Borm PJA, Valds J, Houba R, Wouters EFM.
Pulmonary function in curing workers. Arch Environ Health 31: 610, Respiratory symptoms and lung function in animal feed workers.
1976. Chest 106: 105055, 1994.
127. Fine LJ, Peters JM. Respiratory morbidity in rubber workers: III. 150. Peelen SJ, Heederik D, Dimich-Ward HD, Chan-Yeung M,
Respiratory morbidity in processing workers. Arch Environ Health 31: Kennedy SM. Comparison of dust related respiratory eects in Dutch
13640, 1976. and Canadian grain handling industries: A pooled analysis. Occup
128. Hayden SP, Pincock AC, Hayden J, Tyler LE, Cross KW, Bishop Environ Med 53: 55966, 1996.
JM. Respiratory symptoms and pulmonary function of welders in the 151. Kennedy SM, Dimich-Ward H, Desjardins A, Kassam A, Vedal S,
engineering industry. Thorax 39: 44247, 1984. Chan-Yeung M. Respiratory health among retired grain elevator
129. Cotes JE, Feinmann EL, Male VJ, Rennie FS, Wickham CAC. workers. Am J Respir Crit Care Med 150: 5965, 1994.
Respiratory symptoms and impairment in shipyard welders and 152. Iversen M, Dahl R, Korsgaard J, Hallas T, Jensen EJ. Respiratory
caulker/burners. Br J Ind Med 46: 292301, 1989. symptoms in Danish farmers: An epidemiological study of risk factors.
130. Chinn DJ, Stevenson IC, Cotes JE. Longitudinal respiratory survey Thorax 43: 87277, 1988.
of shipyard welders: Eects of trade and atopic status. Br J Ind Med 153. Leistikow B, Pettit W, Donham K, Merchant J, Popendorf W.
47: 8390, 1990. Respiratory risks in poultry farmers. In: Dosman J, Cockcroft D (eds).
131. Hjortsberg U, Orbaek P, Arborelius M. Small airways dysfunction Principles of Health and Safety in Agriculture, pp. 6265. New York:
among non-smoking shipyard arc welders. Br J Ind Med 49: 44144, Academic Press, 1989.
1992. 154. Donham KJ. Health eects from work in swine connement build-
132. Luo JC, Hsu KH, Shen WS. Pulmonary function abnormalities and ings. Am J Ind Med 17: 1725, 1990.
airway irritation symptoms of metal fumes exposure on automobile 155. Morris PD, Lenhart SW, Service WS. Respiratory symptoms and
spot welders. Am J Ind Med 49: 40716, 2006. pulmonary function in chicken catchers in poultry connement units.
133. Ulvestad B, Bakke B, Melbostad E, Fuglerud P, Kongerud J, Am J Ind Med 19: 195204, 1991.
Lund MB. Increased risk of obstructive pulmonary disease in tunnel 156. Zejda JE, Hurst TS, Rhodes CS, Barber EM, McDue HH,
workers. Thorax 55: 27782, 2000. Dosman JA. Respiratory health of swine producers. Focus on young
134. Musk AW, Peters JM, Bernstein L, Rubin C, Monroe CB. Pulmonary workers. Chest 103: 7029, 1993.
function in reghters: A six-year follow-up in the Boston re 157. Choudat D, Goehen M, Korobae M, Boulet A, Dewitte JD, Martin
department. Am J Ind Med 3: 39, 1982. MH. Respiratory symptoms and bronchial reactivity among pig and
135. Minty BD, Royston D, Jones JG, Smith DJ, Searing CSM, Beeley M. dairy farmers. Scand J Work Environ Health 20: 4854, 1994.
Changes in permeability of the alveolar-capillary barrier in reghters. 158. Zejda JE, Pahwa P, Dosman JA. Decline in spirometric variables in
Br J Ind Med 42: 63134, 1985. grain workers from start of employment: Dierential eect of dura-
136. Rosen G, Lundstrom S. Concurrent video lming and measuring for tion of follow up. Br J Ind Med 49: 57680, 1992.
visualization of exposure. Am Ind Hyg Assoc J 48: 68892, 1987. 159. Schwartz DA, Donham KJ, Olenchock SA et al. Determinants of lon-
137. Horseld K, Guyatt AR, Cooper FM, Buckman M, Cumming G. gitudinal changes in spirometric function among swine connement
Lung function in West Sussex remen: A four year study. Br J Ind operators and farmers. Am J Respir Crit Care Med 151: 4753, 1995.
Med 45: 11621, 1988. 160. Monso E, Riu E, Radon K, Magarolas R, Danuser B, Iversen M
138. Noertjojo HK, Dimich-Ward H, Peelen S, Dittrick M, Kennedy SM, et al. Chronic obstructive pulmonary disease in never-smoking animal
Chan-Yeung M. Western red cedar dust exposure and lung function: farmers working inside connement buildings. Am J Ind Med 46: 357
A dose-response relationship. Am J Respir Crit Care Med 154: 96873, 62, 2004.
1996. 161. Soyseth V, Boe J, Kongerud J. Relation between decline in FEV1 and
139. Chan-Yeung M, Wong R, MacLean L et al. Respiratory survey of exposure to dust and tobacco smoke in aluminium potroom workers.
workers in a pulp and paper mill in Powell River, British Columbia. Occup Environ Med 54: 2731, 1997.
Am Rev Respir Dis 122: 24957, 1980. 162. Sparrow D, Bosse R, Rosner B, Weiss ST. The eect of occupational
140. Brooks SM, Edwards JJ, Apol A, Edwards FH. An epidemiologic exposure on pulmonary function. Am Rev Respir Dis 125: 31922, 1982.
study of workers exposed to western red cedar and other wood dusts. 163. Holman CD, Psaila-Savona P, Roberts M, McNulty JC. Determinants
Chest 81(Suppl 79): 81S85S, 1981. of chronic bronchitis and lung dysfunction in Western Australian
141. Hessel PA, Herbert FA, Melenka LS, Yoshida K, Michaelchuk D, gold miners. Br J Ind Med 44: 81018, 1987.
Nakaza M. Lung health in sawmill workers exposed to pine and 164. Carta P, Aru G, Barbieri MT, Avataneo G, Casula D. Dust exposure,
spruce. Chest 108: 64246, 1995. respiratory symptoms, and longitudinal decline of lung function in
142. Whitehead LW, Ashkaga T, Vacek P. Pulmonary function status of young coal miners. Occup Environ Med 53: 31219, 1996.
workers exposed to hardwood or pine dust. Am Ind Hyg Assoc J 41: 165. Hall SK, Cissik JH. Eects of cigarette smoking on pulmonary func-
17886, 1981. tion in asymptomatic asbestos workers with normal chest radiographs.
143. Whitehead LW. Health eects of wood dust Relevance for an occu- Am Ind Hyg Assoc J 43: 38186, 1982.
pational standard. Am Ind Hyg Assoc J 43: 67478, 1982. 166. Copes R, Thomas D, Becklake MR. Temporal patterns of exposure
144. Goldsmith DF, Shy CM. Respiratory health eects from occupational and non-malignant pulmonary abnormality in Quebec chrysotile
exposure to wood dusts. Scand J Work Environ Health 14: 115, 1988. workers. Arch Environ Health 40: 8087, 1985.
145. Corey P, Hutcheon M, Broder I, Mintz S. Grain elevator workers 167. Kaumann F, Drouet D, Lellouch J, Brille D. Occupational exposure
show work-related pulmonary function changes and dose-eect rela- and 12-year spirometric changes among Paris area workers. Br J Ind
tionships with dust exposure. Br J Ind Med 39: 33037, 1982. Med 39: 22132, 1982.

469
Asthma and COPD: Basic Mechanisms and Clinical Management

168. Le Moual N, Bakke P, Orlowski E et al. Performance of population 172. U.S. Department of Health and Human Services. Women and smoking:
specic job exposure matrices ( JEMs): European collaborative analyses A report of the Surgeon General: U.S. Department of Health and Human
on occupational risk factors for chronic obstructive pulmonary disease Services, Centers for Disease Control and Prevention, Oce on
with job exposure matrices (ECOJEM). Occup Environ Med 157: 126 Smoking and Health; 2001.
32, 2000. 173. Wang XR, Zhang HX, Sun BX, Dai HL, Hang JQ, Eisen EA et al.
169. American Conference of Governmental and Industrial Hygienists. A 20-year follow-up study on chronic respiratory eects of exposure
Threshold Limit Values for Chemical Substances and Physical Agents to cotton dust. Eur Respir J 26: 88186, 2005.
and Biological Exposure Indices. Cincinnati, OH: ACGIH, 1998. 174. Matheson MC, Benke G, Raven J, Sim MR, Kromhout H,
170. Mannino D, Homa D, Akinbami L, Ford E, Redd S. Chronic Vermeulen R et al. Biological dust exposure in the workplace is a risk fac-
obstructive pulmonary disease surveillance United States, 1971 tor for chronic obstructive pulmonary disease. Thorax 60: 64551, 2005.
2000. MMWR CDC Surveill Summ 51: 116, 2002. 175. Hnizdo E, Sullivan PA, Bang KM, Wagner G. Association between
171. Prescott E, Bjerg AM, Andersen PK, Lange P, Vestbo J. Gender dif- chronic obstructive pulmonary disease and employment by industry
ference in smoking eects on lung function and risk of hospitalization and occupation in the US population: A study of data from the Third
for COPD: Results from a Danish longitudinal population study. Eur National Health and Nutrition Examination Survey. Am J Epidemiol
Respir J 10: 82227, 1997. 156: 73846, 2002.

470
Infections
37 CHAPTER

INTRODUCTION respiratory infection and asthma exacerba-


Simon D. Message and
tions is well established although incompletely
understood. In the 1950s this association was Sebastian L. Johnston
Asthma and chronic obstructive pulmonary attributed to bacterial allergy [2] but it is now Department of Respiratory Medicine,
disease (COPD) are common diseases and clear that the majority of exacerbations are due
both result in signicant morbidity and mortal- National Heart and Lung Institute,
to viral rather than bacterial infection.
ity. Although they share some clinical features Imperial College London, London, UK
and although they may coexist in the same
individual, they are distinct disease syndromes
with dierent pathogenetic mechanisms. In Epidemiology
each case much of the morbidity and mortal-
ity is associated with exacerbations of disease, Viral respiratory tract infections are a major
in response to a variety of trigger factors. A cause of wheezing in infants and in adult
common feature of asthma and COPD is the patients with asthma. Their role may have been
important role of infection in triggering exac- underestimated in early epidemiological stud-
erbations. Infections have also been implicated ies because of diculties in isolation and iden-
in the etiology of the two diseases. This chapter tication [3]. The introduction of PCR to such
will review the epidemiological evidence impli- studies has implicated viral infection in the
cating infectious pathogens as triggers and will majority of asthma exacerbations.
discuss the mechanisms of interaction between Indirect evidence from population studies
the hostpathogen response and preexisting has established a signicant correlation between
airway pathology that result in an exacerbation. the seasonal variation in wheezing episodes in
young children and peaks of virus identica-
tion [4]. Seasonal patterns of identication of
respiratory viruses are associated with peaks
in hospital admissions for both children and
ASTHMA adults with asthma indicating a role for such
infections in severe asthma attacks [5]. Direct
evidence implicating viral infection in asthma
Asthma aects 2033% of children in the exacerbations has been provided by studies
United Kingdom [1]. It is a multifaceted syn- showing an increased rate of virus detection in
drome involving atopy, bronchial hyperreac- individuals suering asthma attacks. Viruses
tivity, and IgE and non-IgE-mediated acute have been detected in 8085% of asthma exac-
and chronic immune responses. The asthmatic erbations in children [410] and in 7580% in
airway is characterized by an inltrate of eosi- adults [1114]. The highest rates of identica-
nophils and of T-lymphocytes expressing the tion are in those studies where subjects were
type 2 cytokines IL-4, IL-5, and IL-13. Trigger followed prospectively allowing collection of
factors associated with acute exacerbations clinical specimens early in the course of the ill-
of asthma include exposure to environmental ness, where PCR-based methods of diagnosis
allergens, especially animals, molds, pollens and were used instead of or in addition to serology
mites, cold, exercise, and drugs. The link between and culture, and where the methodology used

471
Asthma and COPD: Basic Mechanisms and Clinical Management

allowed for detection of rhinoviruses. The rate of detec- allergen exposure and may stimulate the production of anti-
tion of viruses between exacerbations when individuals are inammatory mediators such as IL-10 or antiviral cytokines
asymptomatic is only of the order of 312%. In contrast a such as IFN- or TNF-. In work by de Kluijver et al. the
study of transtracheal aspirates in adult asthmatics during eects of a 10-day period of low dose allergen exposure in
exacerbations [15] yielded sparse bacterial cultures with no house-dust mite sensitive and/or experimental RV16 infec-
correlation to clinical illness and no dierence from those tion were studied. No synergistic or additive eects were
of normal subjects. observed as regards lung function parameters [33]. Further
In almost all studies of asthmatics, the predominant development of models of experimental combined allergen
viruses are rhinoviruses (RV), inuenza, RSV, and parain- exposure and virus infection is clearly required.
uenza viruses. RV alone are detected in around 50% of We have recently adopted the approach of infect-
virus-induced asthma attacks. Adenoviruses, enteroviruses, ing asthmatic volunteers with RV and then sending them
metapneumoviruses, bocaviruses, and coronaviruses are also home to continue their normal allergen exposure in the
detected but less frequently. Inuenza is only found during natural environment [34]. We investigated physiologic,
annual epidemics. virologic, and immunopathologic responses to experimental
rhinovirus infection in blood, induced sputum, and bron-
chial lavage in 10 atopic mild asthmatic and 15 nonatopic
Experimental virus infection normal volunteers. Rhinovirus infection induced signi-
cantly greater lower respiratory symptoms, lung function
The eects of respiratory virus infection in the nasal mucosa impairment, increases in bronchial hyperreactivity, and
and upper respiratory tract have been extensively inves- eosinophilic lower airway inammation in asthmatic com-
tigated. The eects of such viruses in the lower respira- pared to normal subjects. We also saw trends to increased
tory tract have been studied but detailed knowledge of the neutrophils and lymphocytes in the lower airway in asth-
pathogenetic mechanisms involved in asthma exacerbations matics, and coincident reductions in blood lymphocytes,
remains limited. Experimental respiratory virus infection suggesting tracking to the airway. In asthmatic, but not
in human volunteers is limited to mild disease by concerns normal subjects, virus load was signicantly related to lower
of safety [16]. Most such studies have therefore focused respiratory symptoms, bronchial hyperreactivity, and reduc-
on the experimental innoculation of rhinovirus in allergic tions in blood total and CD8 lymphocytes and lung func-
rhinitic or mild asthmatic individuals and normal control tion impairment was signicantly related to neutrophilic
subjects [1728]. Such studies provide a useful model of and eosinophilic lower airway inammation. This study
natural virus infection in asthma and oer the advantages demonstrated increased rhinovirus-induced clinical illness
of patient selection and monitoring, under controlled con- severity in asthmatic compared to normal subjects, provided
ditions before, during, and after infection, of administra- evidence of strong relationships between virus load, lower
tion of active and placebo medication, of ability to sample airway virus-induced inammation, and asthma exacerba-
the lower airway with timing from onset of infection accu- tion severity and suggests that this approach could provide
rately dened and the study of RV-induced eects includ- a very good model in which to examine asthma exacerba-
ing asthma symptomatology, lung function, and airway tion pathogenesis as well as treatment interventions.
pathology/immunology.
Recent epidemiological evidence conrms a synergis-
tic interaction between virus infection and allergen expo- Rhinovirus infection of the lower airway
sure in precipitating hospital admissions for asthma [29,
30]. Other trigger factors that may interact with infection Whereas other respiratory viruses such as inuenza, parain-
include air pollution. A study of asthmatic children demon- uenza, RSV, and adenovirus are well recognized causes of
strated an increased risk of developing an asthmatic episode lower airway syndromes such as pneumonia and bronchioli-
within 7 days of an upper respiratory tract infection if the tis and are capable of replication in the lower airway, until
nitrogen dioxide level was greater than 28 g/m3 [9, 31]. recently there was uncertainty as to whether RV infection
Most studies of experimental virus infection in aller- occurred in the lower airway or solely in the upper respi-
gic subjects are performed outside the relevant season for ratory tract. Although the possibility of nasopharyngeal
allergen exposure. One attempt to provide a model combin- contamination cannot be ruled out, RV has been detected
ing allergen exposure and virus infection utilized RV infec- in lower airway clinical specimens such as sputum [35],
tion in subjects with allergic rhinitis. Individuals received tracheal brushings [26], and BAL [36] by both RT-PCR
three high dose allergen challenges in the week prior to and culture. RV has been cultured in cell lines of bronchial
innoculation to try to mimic combined allergen exposure epithelial cell origin [37] and replication has been dem-
and virus infection [32]. Interestingly, prior allergen chal- onstrated in primary cultures of bronchial epithelial cells
lenge in this model, somewhat unexpectedly, appeared to [3840]. The preference of RV for culture at 33C rather
protect against an RV cold with delayed nasal leukocytosis, than 37C has been used as an argument against lower air-
increased generation of the proinammatory cytokines IL-6 way infection but there is now evidence that replication
and IL-8 and a delayed, less severe clinical course. There was does occur at lower airway temperatures [41]. Finally the
an inverse correlation between nasal lavage eosinophilia and use of in situ hybridization has conclusively demonstrated
the severity of cold symptoms. The explanation proposed by RV replication in bronchial biopsies of subjects following
the authors of this study is that limited high dose allergen experimental infection [38] and recent immunochemistry
challenge may not reproduce the eects of chronic low dose data suggests a preference for basal cells [42]. These data

472
Infections 37
conrm that RV infection of the lower airway does occur increases asthma symptoms, coinciding with an increase in
and directly implicate lower airway infection in the patho- the maximal bronchoconstrictive response to methacholine
genesis of asthma exacerbations. up to 15 days after infection [20]. There is also a signicant
increase in sensitivity to histamine in asthmatic subjects
after RV16 infection, most pronounced in those with severe
A mouse model of rhinovirus-induced cold symptoms [25] and our recent study conrmed that
asthma exacerbation these reductions in lung function and increases in symp-
toms and airway hyperresponsiveness were observed only in
Investigation into the pathogenesis of rhinovirus infections asthmatic, but not in normal subjects [34].
and rhinovirus-induced asthma exacerbations has been
severely hampered for the 50 years since their discovery,
as it has been believed that rhinoviruses only infect humans Components of the antiviral immune
and chimpanzees. However, a mouse model of rhinovirus response
infection has recently been successfully developed for the
rst time. New methods of purication and concentration Current concepts of a typical antiviral immune response, as
of rhinoviruses, were used to show that for the minor group reviewed in detail elsewhere [46, 47], result from research in
of rhinoviruses (the 10% that use the LDL receptor as human volunteers and patients but also in experimental ani-
their mode of entry into cells), wild-type BALB/c mice mals, especially inbred mice. Results of animal studies may
can be successfully infected and that most of the disease- not be directly applicable to the outbreed human population
related outcomes observed in humans were reproduced in but ethical considerations often limit direct investigation of
this unique new model. These outcomes include induction the human immune system. All immune responses are a
of both innate and acquired immune responses, induction combination of nonspecic (innate) and specic (adaptive)
of mucin synthesis and secretion, induction of both acute immunity.
neutrophilic and prolonged lymphocytic airway inamma- Nonspecic or innate [48] elements include: phago-
tion, and induction of chemokines responsible for chemo- cytes such as neutrophils and macrophages that engulf and
attraction of neutrophils, lymphocytes and dendritic cells as destroy viruses; natural killer (NK) cells that recognize and
well as a range of proinammatory cytokines. Mice trans- destroy virus-infected cells on the basis of reduced HLA
genic for a chimera of ICAM-1, the receptor for the major class I expression; cells including NK cells, neutrophils,
group (90%) of rhinoviruses, in which the rhinovirus- macrophages, mast cells, basophils, epithelial cells that
binding domains were human, but the remainder of the release cytokines, such as interferons, with immunoregula-
molecule mouse were then developed. This transgenic mouse tory or antiviral actions; components of body uids such
was then able to be infected by major group strains, thus as complement, defensins, and surfactant proteins that are
generating mouse models capable of being infected by all capable of neutralizing viruses independently of, or in com-
rhinovirus serotypes. Finally an established mouse model of bination with, antibodies.
allergic airway inammation was used to demonstrate that
rhinovirus infection of this model resulted in rhinovirus- Complement
induced exacerbation of allergic airway inammation. The
asthma-related outcomes exacerbated by infection in this Some viruses may also cause complement-mediated dam-
model include exacerbation of airway hyperresponsiveness, age. Complement components bind to epithelial cells both
exacerbation of mucin synthesis and secretion (MUC5AC in vitro and in vivo during RSV infections. C3a and C5a
and MUC5B), exacerbation of neutrophilic, eosinophilic, are increased in human volunteers infected with inuenza
and lymphocytic airway inammation, and augmented A virus [49]. There is little information on the role of com-
induction of both Th1 (IFN-) and Th2 (IL-4 and -13) plement in immunity to RV. Recent data suggests that the
cytokines. The development of this novel mouse model of RV 3C protease cleaves the complement factors C3 and C5
rhinovirus-induced asthma exacerbations, should allow which may interfere with the destruction of virus-infected
mechanisms of disease to be investigated in vivo and true cells [50]. For other viruses, for example inuenza, the com-
causation be established in vivo. [43]. plement system forms an important link between the innate
and specic immune systems. Mice decient for the third
component of complement are highly susceptible to pri-
Physiological effects of experimental mary inuenza, showing reduced priming of T-helper cells
rhinovirus infection and cytotoxic T-cells in lung draining lymph nodes and
severely impaired recruitment into the lung of virus-specic
Subjects with asthma and/or allergic rhinitis exhibit CD4 and CD8 eector T-cells producing IFN- [51].
increased pathophysiological eects as a result of RV infec- Activation of the complement cascade may be necessary for
tion as compared to nonatopic, nonasthmatic controls. the function of other innate antiviral proteins such as serum
With detailed monitoring, it is possible to detect reductions mannose-binding protein [52]
in both peak ow [44] and home recordings of FEV1 [24]
in atopic asthmatic patients in the acute phase of experi- Defensins
mental RV16 infection. There is an enhanced sensitivity The and defensins are small cationic antimicrobial pep-
to histamine and allergen challenge after RV16 innocula- tides which have the capacity to kill bacteria, fungi, and
tion in nonasthmatic atopic rhinitic subjects [19, 45]. RV16 enveloped viruses by disruption of the microbial membrane.

473
Asthma and COPD: Basic Mechanisms and Clinical Management

In vivo they are probably most important in phagocytic vac- there is relatively less activation of nonspecic immunity
uoles and on the surface of skin and mucosal epithelia. In and it may be dicult to detect a CD8 T-cell response.
addition to their direct antibiotic role, defensins are increas- Following experimental infection of seronegative sub-
ingly being found to have immunomodulatory actions [53] jects with RV2 [54] serum-specic antibodies are detectable
and to play a role in cell recruitment through activation of at 12 weeks, reach a maximum at 5 weeks, persist for at
certain chemokine receptors, for example hBD3 and CCR6 least a year and may remain elevated many years after infec-
on dendritic cell (DC). tion. Local specic antibody levels may be lost more rap-
Specic immunity involves production of antibody idly. High levels of serum neutralizing antibody or specic
by B-lymphocytes and the activities of cytotoxic T-cells IgA protect against reinfection with the same rhinovirus
following processing and presentation of viral antigens by serotype. However, since it appears relatively late, recovery
additional cells of the immune system, the most impor- from illness for seronegative hosts which usually occurs at
tant of which are probably dendritic cells. Immunological 710 days must be due to other components of the immune
memory modies the overall response to reinfection by pre- response. In seropositive subjects preexisting serum neutral-
viously encountered virus and alters the timing and magni- izing antibodies to RV39 and to RV-Hanks modify experi-
tude of contributions due to dierent components. mental infections in human subjects [55, 56]. Local IgA and
IgG passing from the vasculature into the pulmonary inter-
stitium contribute to viral clearance. However, the 100 RV
serotypes mean that repeated infection with RV to which an
Time course of innate and adaptive individual lacks appropriate antibodies is common.
T-cell responses to RV demonstrate MHC class I
immunity in primary and secondary restricted cross-reactivity between serotypes due to speci-
infections city for conserved epitopes within the capsid proteins VP
13 [57]. RV16- and RV49-specic T-cell clones from
In primary infection, viruses replicate in the respiratory tract human peripheral blood demonstrate recognition of both
reaching peak levels at around days 24. At this time type I serotype specic and shared viral epitopes [58]. Vigorous
interferons are rst detected, peaking around days 23 and proliferation of and IFN- production by PBMC in
falling to become undetectable once active replication has response to RV16 in seronegative subjects is associated
ceased. Interferons activate NK cells, rst detectable around with reduced viral shedding after inoculation [59], thus
day 3 and peaking around day 4. In addition to destruction T-cells responses also appear protective.
of virally infected cells NK cells release cytokines includ-
ing IFN- that activate additional inammatory cells in the
airway including macrophages. Such nonspecic immune Interactions between virus infection and
mechanisms are essential in early defense against virus asthmatic airway inflammation
in the rst few days. In addition, the innate immune sys-
tem plays a role in stimulating specic immunity and may The interaction of respiratory virus infection and chronic
inuence the nature of the specic response, for example asthmatic airway inammation results in respiratory symp-
whether this is characterized by type 1 or type 2 cytokines. toms that are more severe than those suered by nonas-
Meanwhile, viral antigens are processed locally and thmatic individuals [34, 60] and case-control studies have
in regional lymph nodes by dendritic cells and presented demonstrated clear synergistic interactions between virus
to T-cells. CD4 and CD8 T-cells are detectable from infection and allergen exposure in increasing risk of exac-
around day 4 then generally decline as infection resolves to erbation [29, 30]. The detailed immunological mechanisms
become undetectable by day 14. However, memory CD4 underlying this interaction are currently being investigated,
and CD8 responses may persist for life. T-cell recruit- but recent data suggest decient production of type I (),
ment is dependent on the production of chemokines and type II (), and type III () IFNs, as well as Th1 cytokines
on alterations in the expression of adhesion molecules on (IL-12) and anti-inammatory cytokines (IL-10), are likely
the endothelium of inamed tissues. Time is also required to increase virus-induced lower airway inammation [34,
to generate B-cell responses. Mucosal IgA may be detected 61, 62]. These deciencies are also accompanied by aug-
around day 3, serum IgM from days 56 and IgG days 78, mented production of Th2 cytokines suggesting that per-
increasing in amount and avidity over the next 23 weeks. haps allergen-induced inammation is also increased in the
IgA falls normally to low or undetectable levels over 36 pathogenesis of virus-induced asthma exacerbations.
months. Serum IgG may remain detectable for life. Specic Bronchial inammation is likely therefore a central
immune mechanisms such as CD8 T-cells and immu- event for virus-induced asthma exacerbations. The processes
noglobulin are responsible for the eradication of infectious involved include interacting cascades from the complement,
virus usually by 710 days after infection. coagulation, brinolytic, and kinin systems of the plasma as
Secondary infection with the same virus results in well as cell-derived cytokines, chemokines, and arachidonic
rapid mobilization of B- and T-cell specic immunity with acid metabolites. Our understanding of the interaction of
an earlier T-cell peak coinciding with the NK cell peak viruses with these cascades in asthma is incomplete and it
around days 34. If reinfection is with the same serotype a is likely that dierent viruses interact with each system to
rapid increase in levels of preexisting neutralizing antibodies dierent extents. However, it is reasonable to believe that in
may limit viral replication to such an extent that infection is all cases the initial trigger of the inammatory reactions is
clinically silent. Because this results in fewer infected cells epithelial cellvirus interaction.

474
Infections 37
TABLE 37.1 Current hypotheses for the pathogenesis of virus-induced Table 37.1 summarizes some of the current hypoth-
asthma exacerbations. eses proposed to explain the mechanisms of exacerbation of
asthma following respiratory virus infection. The evidence
Epithelial disruption Reduced ciliary clearance supporting these hypotheses is reviewed in detail below.
Increased permeability
Loss of protective functions
Kinins

Mediator production Complement The role of the airway epithelial cell


Arachidonic acid metabolites
Nitric oxide The airway epithelium is an important component of anti-
Reactive oxygen products viral defense. In addition to its function as a physical bar-
Cytokines rier to the entry of viruses, the responses of epithelial cells
(EC) following viral infection, whether or not this results in
Induction of inflammation Chemokines destruction of the cell, contribute to both innate and adap-
Immune cell activation tive antiviral immune responses. Information regarding the
Adhesion molecule induction eects of RV on EC comes from in vivo studies and from
Impaired innate IFN production in vitro models using either cultured primary airway EC or
Immune dysregulation Impaired apoptosis cell lines of epithelial cell origin such as A549, BEAS-2B,
Impaired Th1 immunity and H292.
Impaired IL-10 production EC contribute to the immune response following
Augmented Th2 immunity virus infection through the production of cytokines and
Increased total IgE chemokines (Fig. 37.1). They may also act as antigen pre-
senting cells particularly during secondary respiratory viral
IgE dysregulation Antiviral IgE production infections. Epithelial cells express MHC class I and the
Airway smooth muscle costimulatory molecules B7-1 and B7-2 and this expression
is upregulated in vitro by RV16 [63] (Fig. 37.2).
Airway remodeling Fibroblasts
The extent of epithelial cell destruction observed in
Myofibroblasts
the airway varies according to virus type. Inuenza typically
Growth factors
causes extensive necrosis [64], whereas RV causes little or
Increased cholinergic sensitivity
only patchy damage. Destruction of epithelial cells results
Alterations of neural responses Neuropeptide metabolism in both an increase in epithelial permeability, and increased
modulation penetration of irritants and allergens, and exposure of the
-adrenergic receptor dysfunction extensive network of aerent nerve bers. Both eects may
contribute to increased bronchial hyperresponsiveness.

FIG. 37.1 Airway epithelial cells participate in the immune response to respiratory virus, producing a variety of cytokines and chemokines with actions on
other cells. In addition the migration of inflammatory cells is aided by the upregulation of adhesion molecules and interferons help to establish an antiviral
state in neighboring epithelial cells. Upregulation of MHC class I may facilitate presentation of viral antigens.

475
Asthma and COPD: Basic Mechanisms and Clinical Management

Virus

Macrophage Dendritic cell

IgE
IL-12,-18
IL-4 IL-4,13
Th0

 IL-4
IL-2
IFN- Eosinophilia
Cytotoxicity IFN-  IL-5

Th1 Th2
Effective early viral clearance Failure to clear virus

Effective termination of immune Persistent eosinophilia


response
Th1 Th2 Prolonged inappropriate immune Th1 Th2
response with immunopathology
and tissue damage

Amplification of allergic inflammation

FIG. 37.2 Preexisting asthmatic airway inflammation may modify a predominantly Th1 antiviral immune response, favoring a Th2 or mixed response which
may provide less efficient viral clearance and result in prolonged virus-induced inflammation, increased associated immunopathology and increased tissue
damage.

In vivo RV causes some shedding of infected ciliated patchy nature of infection. The upregulation of ICAM-1
EC [65], but the extent of viral infection of the epithelium in the asthmatic airway is one possible explanation for the
may be incomplete even in the nose [66]. In vitro stud- increased severity of RV infection in asthma. RV upregulates
ies exposing monolayer cultures of nasal epithelial cells to expression of its own receptor ICAM-1 both in vitro and
respiratory viruses at 103104 TCID50/ml demonstrate no in vivo. Following experimental infection with RV, ICAM-1
detectable CPE (carboxypeptidase E) with RV or corona- expression is upregulated in nasal epithelium within 24 h,
virus in contrast to the extensive destruction with inuenza declining by day 5 [71]. RV has similar eects on EC from
and adenovirus [67]. Ex vivo infection of cells from both the lower airway. RV has been shown to upregulate ICAM-1
the upper and the lower respiratory tract suggest that less in primary bronchial EC in vitro [72] and ICAM-1 is
than 10% of cells in the epithelium are infected by RV [68], upregulated in bronchial biopsies following experimental
however the extent of virus-induced epithelial damage may infection of asthmatic subjects with RV16 [73].
be considerably greater in asthmatic than in normal sub- There are two forms of ICAM-1: membrane bound
jects, as asthmatic epithelium has been shown to be much (mICAM-1) which favors viral infection by acting as a
more susceptible in vitro [61]. In vivo studies of degrees virus receptor and soluble (sICAM-1) which binds virus
of epithelial damage would be technically challenging, but outside the cell and can thereby inhibit virus infection. RV
could generate interesting ndings. infection of EC is reported to alter the balance in favor of
further infection by inducing mRNA for mICAM-1 whilst
suppressing that of sICAM-1 [74]. The LDL receptor is the
Receptors for entry of RV into host cells receptor for the minor group RV. RV2 infection of primary
human tracheal EC (PHTEC) is blocked by an antibody to
Viruses enter into and replicate within airway EC. Entry is the LDL receptor and is also reported to upregulate LDL-R
dependent on the interaction with host cell surface proteins expression [48].
which function as receptors. In the case of the major group ICAM-1 expression by human nasal EC is upregulated
RV this is ICAM-1 [69] and infection can be blocked by in vitro by exposure to a number of inammatory cytokines
antibodies to ICAM-1 or with soluble ICAM-1 [37]. There and mediators including IL-1, IL-8, IFN-, TNF-, and
is relatively limited expression of ICAM-1 in airway epi- the eosinophil-derived proteins MBP and ECP [75]. IL-1
thelium prior to RV infection [70] and this may explain the in particular may be important in RV-induced induction of

476
Infections 37
ICAM-1. Antibodies to IL-1 but not TNF- decreased cells are important components of the antiviral response but
viral replication and ICAM-1 expression by PHTEC [76]. may also contribute to airway inammation and dysfunc-
Not all respiratory epithelial cell lines behave in the same way tion in asthma.
as primary EC for example A549 cells express ICAM-1
at lower levels constitutively and show upregulation by Type 1 interferons
IFN- and TNF- but not by ECP or MBP. The eect of Interferons (IFN) play an important role in innate resist-
IFN- is complex. Whilst IFN- upregulates ICAM-1 in ance to viruses [89], acting on virus-infected cells and sur-
uninfected cells this cytokine inhibits ICAM-1 upregulation rounding cells to produce an antiviral state characterized
by RV14 in H292 cells and its presence results in reduced by the expression and antiviral activity of IFN-stimulated
viral titers [72, 77]. genes (ISGs). There are three main types of IFN, type 1
A preexisting elevation of ICAM-1 expression in (IFN-, IFN-, IFN-, IFN-), type 2 (IFN-) and the
the asthmatic airway may contribute to increased symptom recently discovered type 3 (IFN-s [90, 91]). EC can pro-
severity of RV infection. Type 2 cytokines (IL-4, IL-5, duce both type 1 and type 3 IFNs. There are 14 IFN-
IL-13) upregulate ICAM-1 in H-292 cells [78]. Allergen genes but only 1 IFN- gene. IFN- synthesis involves NF-
challenge results in upregulation of ICAM-1 on conjunc- B, ATF/JUN and the interferon regulatory factors (IRFs)
tival and nasal EC in atopics [79]. In nasal brushing EC (up to 10 of which are currently identied), activation of
from atopics, basal ICAM-1 levels were increased relative which occurs in response to virus-specic signals including
to nonatopics and elevated in the relevant allergen season. dsRNA, a product of the replication of ssRNA viruses such
Nasal EC from atopics showed further upregulation after as RV, RSV, and inuenza.
in vitro culture with allergen. The highest basal ICAM-1 IFN- and IFN-4 are expressed early through the
was found on nasal polyp EC and this was increased fur- action of IRF3. Activation of the IFN intracellular signaling
ther after RV14 infection. Viral titers after RV14 infection pathway is required for induction of IRF7 which is required
were signicantly higher for polyp EC than for nonatopic for transcription of the full range of IFNs. DNA microarray
and atopic nonpolyp EC [80]. analysis has shown that following binding to their receptors
Modication of EC ICAM-1 expression is therefore on target cells IFNs trigger a complex signaling pathway
of possible therapeutic benet. In vitro RV increases expres- (mainly JAK-STAT) resulting in the transcription of hun-
sion of ICAM-1 and VCAM-1 in primary bronchial EC dreds of ISGs [92].
(PBEC) cultures and in A549 cells via a mechanism involv- Several ISGs have been well studied. These include
ing NF-B [5, 81, 82]. One of the actions of corticoster- the dsRNA-activated serine/threonine protein kinase
oids is inhibition of NF-B [83]. In both A549 cells and (PKR) which reduces cellular mRNA translation and tran-
in PBEC pretreatment with three corticosteroids, hydro- scriptional events, two enzymes involved in mRNA degra-
cortisone, dexamethasone, and mometasone furoate inhib- dation, 2
5
oligoadenylate synthetase (OAS) and RNase L,
its RV16-induced increases in ICAM-1 surface expression, the myxovirus resistance (Mx) proteins and RNA-specic
mRNA, and promoter activation without alteration of adenosine deaminase (ADAR) which is involved in RNA
virus infectivity or replication. Dexamethasone suppresses editing. These ISGs inhibit virus replication at a number of
ICAM-1 in PHTEC and inhibits RV infections [84]. levels and not surprisingly, viruses have evolved mechanisms
Dexamethasone does not inhibit infection of PHTEC by to resist the actions of IFNs, for example blocking of PKR
minor group RV2 [85]. Disappointingly, a study of inhaled by the inuenza NS1 protein [93]. IFNs also upregulate
corticosteroids in asthmatics prior to experimental RV cellular expression of MHC class I and II molecules there-
infection failed to show reduced virus-induced ICAM-1 fore increasing antigen presentation to CD8 and CD4
expression in bronchial biopsies [73] but it is possible that T-cells and enhancing cellular immune responses.
a longer course and/or a higher dose of inhaled steroid or One recent study has examined type 1 interferon pro-
administration of oral steroids might have demonstrated a duction by primary bronchial epithelial cells from normal
signicant eect. and asthmatic subjects infected ex vivo with RV [40].
Other drugs which aect EC ICAM-1 include reduc- Asthmatic EC following infection released a higher titer of
ing agents [86], the H1 receptor antagonists desloratidine/ virus into culture supernatant and exhibited impaired apop-
loratidine which inhibit RV-induced ICAM-1 upregulation tosis and a greater degree of necrotic cell death, favoring
in HPBEC and in A549 cells [87] and erythromycin which release of virus from dying cells. This was accompanied by
inhibits infection of PHTEC by both major group RV14 lower concentrations of IFN- after infection. Addition of
and minor group RV2 through eects including ICAM-1 IFN- to asthmatic ECs inhibited virus replication to levels
reduction, blockage of RV RNA entry into endosomes and observed in ECs from normal subjects. This study suggests
small reductions in LDL receptor expression [88]. that the production of IFN- is decient in asthmatic ECs
and that replacement/augmentation of IFN- to boost the
innate immune response could be a novel approach to treat-
RV induction of EC production of cytokines ment of virus-induced asthma exacerbations [94].
and chemokines
Type III interferons
EC can activate and recruit a variety of other cell types such A new family of interferons, called type III IFN-s, and
as lymphocytes, eosinophils, and neutrophils through the characterized by three elements: 1, 2, and 3, also termed
production of chemokines and cytokines (Fig. 37.1). Such IL-29, IL-28A, and IL-28B, has recently been described

477
Asthma and COPD: Basic Mechanisms and Clinical Management

[90, 91]. The three highly homologous IFN- proteins number of cell types, by both cells of the immune system,
demonstrate limited (about 20%) homology to type I IFNs which may be increased in number and activation status,
[95]. Human IFN-s bind to a unique heterodimeric and by cells often considered to be structural but which in
receptor (IFN-R), composed of CRF2-12 (also designated fact contribute signicantly to the immune response such
IFN-R1), and CRF2-4 (also designated IL-10R2) shared as EC. Ecient orchestration of the immune response by
with other class II cytokine-receptor ligands including IL- cytokines is essential for eradication of virus. Modication
10, IL-22, and IL-26 [90]. of cytokine expression in the airway may contribute to the
Viral infection induces upregulation of IFN- increased severity of virus infection in asthma.
mRNA in epithelial cells, peripheral blood mononuclear In vitro studies of bronchial EC lines or macrophages
cells (PBMCs), and dendritic cells [90, 91, 9699]. IFN-s have demonstrated the production of a wide range of proin-
exhibit some similar biological properties to type I IFNs: ammatory cytokines such as IL-1, IL-6, IL-11, IFN-,
they induce Jak/STAT pathways that lead to the upregu- IFN-, TNF-, and granulocyte-macrophage colony stim-
lation of several antiviral proteins and enzymes including ulating factor (GM-CSF) and the chemokines IL-8, ENA-
2
,5
-OAS and MxA, have antiviral activity in vitro [90, 91, 78, RANTES, and IP-10 and macrophage inammatory
97] and have also exhibited antviral activity in an in vivo protein (MIP)-1 in response to RV and RSV [37, 101
model of vaccinia virus-infected mice [100]. Based on cur- 103]. In vivo these cytokines can be found in nasal lavage in
rent knowledge, it thus appears that both IFN-/- and association with RV infection [104].
IFN- ligand-receptor systems can independently induce The specic roles of individual cytokines in the human
an antiviral state by engaging similar participants of the lower airway during viral infection are not well understood,
antiviral response, though the signaling pathways involved but increasing information is becoming available. Such
in IFN- production are currently largely unknown. Recent cytokines and chemokines activate and recruit a variety of
data suggest that IFN- may be involved in antiviral other cells including lymphocytes, eosinophils, and neu-
responses against RV. In vitro RV infection of a bronchial trophils. IL-1, TNF-, and IL-6 share proinammatory
epithelial cell line (BEAS-2B) led to IFN- production and properties such as the induction of the acute phase response
this cytokine demonstrated a dose-dependent antiviral eect and the activation of both T- and B-lymphocytes. IL-1
against RV [62]. Moreover, IFN- production occurred enhances the adhesion of inammatory cells to endothe-
after in vitro infection of primary bronchial epithelial cells, lium, facilitating chemotaxis [105]. TNF- is a potent
macrophages, and BAL cells from healthy volunteers. antiviral cytokine but in vitro increases the susceptibility
We have recently investigated the production of IFN-s of cultured epithelial cells to infection by RV14 through
in response to RV in primary bronchial ECs and in BAL upregulation of ICAM-1 [37]. IL-6 has been shown to
cells (90% macrophages) from normal and asthmatic sub- stimulate IgA-mediated immune responses. IL-11 may
jects [62]. Production of IFN-1 and IFN-23 was de- also be important in virus-induced asthma [106]. It appears
cient in ECs and BAL cells from asthmatic subjects after to cause bronchoconstriction by a direct eect on bron-
in vitro RV infection and induction of IFN-s by RV infec- chial smooth muscle [102]. Production of this cytokine by
tion of ECs was strongly inversely related to RV replication. human stromal cells in vitro is increased by RV14, RSV, and
To determine whether IFN- production was important parainuenza type 3 but not by cytomegalovirus (CMV),
in determining responses to RV infection in vivo, the same herpes simplex virus (HSV)-2 or adenovirus. In vivo IL-11
volunteers were then experimentally infected with RV16, is elevated in nasal aspirates from children with colds, levels
the severity of symptoms and reductions in lung function correlating with the presence of wheezing.
were monitored and the virus load was determined in BAL. Similarly the chemokine MIP-1 is increased in
In vitro production of IFN-s by RV infection of BAL cells nasal secretions during natural viral exacerbations of asthma
was strongly inversely correlated with both common cold [107]. Studies in MIP-1 knock-out mice suggest that it
symptoms and in vivo virus load and strongly positively cor- mediates pneumonitis due to inuenza [108]. The other
related with severity of falls in lung function, in asthmatic chemokines IL-8 and ENA-78, will recruit and activate
and normal volunteers experimentally infected with RV16. neutrophils, while RANTES and IP-10 will do the same
Asthmatic patients, in whom in vitro IFN- production in for lymphocytes [103].
BAL cells was signicantly lower than in normal subjects, Viral upregulation of cytokines and chemokines
exhibited increased common cold symptoms and reductions may be mediated through certain key transcription factors.
in lung function and virus load after in vivo RV16 infec- Increases in IL-6 and IL-8 production by cultured epithe-
tion. In marked contrast normal subjects had robust IFN- lial cells due to RV was dependent on NF-B [82, 107, 109]
responses, less severe cold symptoms, lower virus load, and and further upstream, protein kinase R (PKR)-mediated
no signicant changes in lung function. These results docu- RV-induced RANTES, IL-8, and IL-6 [110]. Rhinovirus
ment the importance of IFN- in the host defense against induction of IL-8 was shown to require IkappaB kinase-
RV infection in vitro and in vivo and indicate that decient beta (IKK) and the transcription factor NF-IL-6 as well as
IFN- production is likely to be important in the patho- NF-B. Similar observations have been made with regard
genesis of virus-induced asthma exacerbations. to the induction of IL-1, -6, -8, -11, and TNF- by RSV
[111, 112], thus the potential role of inhibition of NF-B
in this context has generated considerable interest.
Proinflammatory cytokines and chemokines In addition to the induction of IL-1 and IL-1
Viral infection of the respiratory tract results in signi- RV infection results in substantial increases in IL-1ra both
cant changes in the pattern of cytokine expression by a in vivo and in vitro. This is a relatively late eect, occurring

478
Infections 37
4872 h after infection and may contribute to symptom res- that one mechanism for increased airway hyperresponsive-
olution [113]. ness during respiratory virus infection is through inhibition
of NOS enzymes and a loss of NO-related relaxation of
airway smooth muscle [137]. Studies of human asthmatics
Kinins and nitric oxide would also suggest that NO has a protective role in virus-
A multitude of inammatory mediators are generated or induced exacerbations. Following experimental RV16 infec-
act on the epithelial surface. Bradykinin, a-nine-amino acid tion patients with the greatest increase in exhaled NO had
peptide generated from plasma precursors as part of the smaller increases in histamine airway responsiveness [131].
inammatory process has been shown to be present in nasal In experimental animals parainuenza virus-induced
secretions of RV-infected individuals [114]. Bradykinin hyperreactivity correlates with a deciency in constitutive
given intranasally is able to reproduce some of the symp- NO production [137]. Increased levels of exhaled NO are
toms of the common cold such as sore throat and rhini- found in nonasthmatic volunteers following natural colds
tis [115]. Although the presence of kinins in the lungs of [138] as well as in asthmatic patients after experimental
virus-infected individuals has not been reported they are RV infection [131]. In the latter study, an inverse associa-
present in both the upper and lower airways in allergic reac- tion between NO increase and worsening of airway hyper-
tions [114118]. responsiveness was demonstrated arguing in favor of a
Nitric oxide (NO) is produced by diverse sources protective role for this substance. This is further supported
including epithelial, endothelial, and smooth muscle cells. by the observation that NO reduces cytokine production
In human airways NO appears to be important in relaxa- and viral replication in an in vitro model of RV infection
tion of the human airway smooth muscle [119]. Nitric [133]. Interestingly, studies of viral upper respiratory tract
oxide (NO) may be important in a range of respiratory dis- infections have failed to demonstrate an increase in nasal
eases [120] including asthma [121] and in virus infection NO after experimental RSV, RV, and inuenza infections
[122]. NO is produced both by the constitutive enzymes, [139]. In normal subjects experimental inuenza infection
nitric oxide synthase (NOS)1, and NOS3 and by the induc- increased oral NO 8 days postinfection but had no eect
ible, calcium-independent NOS2 expressed by airway EC on nasal NO [140]. This raises the possibility that during
[123] and macrophages. respiratory virus infection induction of NO is selective for
In asthma there is increased NOS2 expression and the lower respiratory tract.
an elevated level of exhaled NO [124] that falls with cor-
ticosteroid therapy; the level of exhaled NO correlates with Signaling pathways
sputum eosinophilia and methacholine responsiveness The responses of airway EC to virus infection are conse-
[125]. In contrast in stable COPD, exhaled NO levels are quences of the interactions between virus and the intracellu-
not dierent from normal subjects [126] although there lar signaling pathways of the host cell [141]. Knowledge of
may be an increase during exacerbations [127]. In fact the mechanisms involved for rhinoviruses is currently very
in vitro cigarette smoke reduces cytokine-induced NOS2 limited. Activation of signaling pathways may be depend-
mRNA expression in the LA-4 murine cell line, in A549 ent on cell surface receptor (ICAM-1, LDL-R) binding
cells and in PHBEC [128]. or may occur during viral replication within the cell. The
The relative importance of the benecial antimicrobial need for replicative virus is demonstrated by the inhibi-
activity of NO versus the potentially disadvantageous sup- tion of RV induction of EC cytokines after UV inactiva-
pression of IFN- may be dependent on the specic patho- tion. One product of replication, common to ssRNA viruses
gen. NOS2 knock-out mice show an increased susceptibility such as RV and also RSV and inuenza, is dsRNA, which
to infections [129], perhaps because release of NO may be has been shown to activate components of signaling path-
important for NK cell-mediated target cell killing [130]. ways including dsRNA-dependent protein kinase PKR,
However NO may also possess antiviral activity. In vitro IKK, NF-B, and p38 mitogen-activated protein kinase
RV induces NOS expression in HPBEC [131]. There is with resultant induction of IL-6, IL-8, and RANTES [110,
increased expression of NOS2 mRNA in cultured HPBEC 142]. Activation of EC by dsRNA may be direct or indirect
after RV16 infection [132]. NO inhibits RV-induced pro- through the interferon system as discussed above. It has also
duction of IL-6, IL-8, and GM-CSF and viral replication been reported that dsRNA and virus infections activate EC
in a human respiratory epithelial cell line [132, 133]. RSV through binding to TLR3 [143, 144].
also induces NOS2 and increases nitrite levels in superna-
tant from A549 cells, from HPBEC culture and in BAL
uid from RSV-infected BALB/c mice, eects opposed by
IL-4 and dexamethasone but unaected by IL-13 or IFN- Effects of viruses on airway smooth
[134]. Replication of RSV in Hep2 cells is inhibited follow- muscle cells
ing transfection with a retroviral construct containing NOS
and this inhibition is abolished by the NOS inhibitor, NG- Studies utilizing isolated rabbit tissues and human cultured
methyl-l-arginine [135]. Replication of inuenza A and B airway smooth muscle cells suggest that, for RV16, expo-
in Mabin Darby kidney cells is severely impaired by the NO sure to the virus may have a direct eect on smooth muscle
donor, S-nitroso-N-acetylpenicillamine [136]. cells, resulting in increased contractility to acetylcholine and
Overall there is evidence that in vivo increased lower impaired relaxation to isoproterenol. This eect is depend-
airway NO production may be of benet in virus-induced ent on ICAM-1 and appears to involve an autocrine sig-
asthma exacerbations. Work in a guinea pig model suggests naling mechanism including upregulation of production of

479
Asthma and COPD: Basic Mechanisms and Clinical Management

IL-5 and IL-1 by the airway smooth muscle itself [145]. Dendritic cells
A more recent study demonstrated that RV induction of
IL-6 and IL-8 was increased in smooth muscle cells from Dendritic cells are key cells in IFN production, as well as in
asthmatic compared to normal subjects [146]. Whether antigen presentation both of allergens and pathogens with
rhinovirus reaches airway smooth muscle cells in sucient a capacity to induce both primary and secondary immune
quantity to produce a signicant eect by this mechanism responses. They may also play a role in the regulation of the
in vivo is as yet unknown. The eects of other respiratory type of T-cell-mediated immune response [154]. RV infec-
viruses on smooth muscle require further investigation. tion has been shown to induce production of the dendritic
cell attracting chemokine MIP-3 [43] and to increase in
number in the lung during RSV infection [155], suggest-
The cellular immune response to virus ing they are recruited to the lung during respiratory virus
infections. However, they have also been shown to be pro-
infection in the lower airway duced from local precursors during RSV infection [156].
Plasmacytoid dendritic cells are likely protective against
A variety of leukocytes show changes in number, site of infection as they have been shown to limit virus replication
accumulation, and activation state in response to virus in RSV infections, as well as reducing airway inammation
infection. Since these cells are also implicated in asthmatic and airway hyperresponsiveness [157]. In contrast, others
inammation of the lower airway they provide potential have reported induction of the high anity IgE receptor on
sites of interaction between the immunopathologies of virus dendritic cells during Sendai virus infection, and linked this
infection and asthma. with induction of mucus cell metaplasia and airway hyper-
reactivity [158]. There is thus increasing knowledge of the
immunobiology of these cells during respiratory infections
Monocytes/macrophages but their role in the context of viral exacerbations of asthma
remains unclear and further studies are needed.
Alveolar macrophages are present in large numbers in the
lower airway. They make up around 90% of the cells seen in
BAL from normal volunteers [28]. They are ideally placed Lymphocytes
for early phagocytosis of virus particles and are likely to play
an important role in the immune response through anti- Bronchial biopsies demonstrate increases in cells positive for
gen presentation to T-cells and through the production of CD3, CD4, and CD8 within the epithelium and submucosa
cytokines and other mediators. RV has been shown to enter of both normal and asthmatic subjects following experimen-
human monocytes and macrophages which express high tal RV infection [21] and we have recently demonstrated a
levels of the major RV receptor ICAM-1. It has not been trend toward increased numbers of lymphocytes in BAL
possible to demonstrate RV replication within alveolar mac- from asthmatic compared to normal subjects (p 0.06)[34].
rophages although low grade productive infection has been Such increases coincided with peripheral blood lymphope-
shown in the monocyte cell line THP-1 [147]. Replication nia, and reductions in blood total lymphocytes and CD8
also occurred in THP-1-derived macrophages but was lim- T-cells correlated strongly with virus load only in asthmatic
ited in monocyte-derived macrophages which are relatively subjects[34] suggesting increased recruitment of T-cells to
resistant to viral replication at least partly because of higher the asthmatic airway may be important in the context of
levels of type I interferon production [148]. RV entry into asthma exacerbations. Since T-cells are believed to be key
monocytes results in activation and the production of both cells in the pathogenesis of asthma the eects of viruses on
IL-8 [147] and TNF- [149]. In monocyte and THP-1- T-cells are of particular importance.
derived macrophages RV induction of TNF- is NF-B T-cell recruitment into the airway is at least partly
dependent [148]. under the inuence of chemokines, including those whose
A recent study reported that infectious but not UV- production by EC is upregulated by viruses. The nature and
inactivated RV-increased TNF- and IL-8 release by mac- the eectiveness of the specic immune response may be
rophages derived from resected lung tissue. Interestingly, inuenced by the balance of chemokine production by air-
infectious rhinovirus-impaired LPS and lipoteichoic acid- way EC. This balance may in turn be inuenced by preexist-
induced TNF- and IL-8 secretion by macrophages as well ing chronic inammation as found in asthma.
as the macrophage phagocytic response to labeled bacterial Studies of cloned T-cells suggest that Th1 and Th2
particles [150]. This RV-induced impairment of cytokine cells show dierential expression of chemokine recep-
responses to bacterial LPS and lipoteichoic acid and of tors. There is increased expression of CXCR3 (receptor for
phagocytosis in alveolar macrophages could lead to impair- IP-10, I-TAC, and Mig) and CCR5 (MIP-1) in human
ment of antibacterial host defense may have important Th1 cells and increased expression of CCR4 (TARC and
implications in the pathogenesis of exacerbations of respi- MDC) and to a lesser extent CCR3 (eotaxin and MCP-3)
ratory diseases including both asthma [151] and COPD in Th2 cells, with selective migration of cells in response to
[152]. In contrast, infection of human monocytes in vitro the appropriate chemokines. CCR1 (RANTES, MIP-1,
with inuenza A causes alterations in structure and activa- MCP-3) and CCR2 (MCP-1, -2 ,-3, -4) were found on
tion status and the production of IL-1 IL-6, TNF- IFN- both Th1 and Th2 cells [159]. Bronchial biopsies from asth-
, and IFN- [153], eects dramatically potentiated by matics show high levels of expression of CCR4 and signi-
subsequent exposure to bacterial LPS. cant levels of CCR8 by T-cells [160].

480
Infections 37
Increased recruitment of T-cells to the airway as a while production of the type 2 cytokines IL-4, -5 and, -13
result of virus-induced chemokine production by EC could were all increased [34]. Importantly CD4 T-cell produc-
amplify preexisting allergic inammation. If the asthmatic tion of IFN- was strongly inversely correlated with virus
airway microenvironment inuences the pattern of chem- load and reductions in lung function in the asthmatic sub-
okine expression following virus infection then this could jects when they then underwent RV experimental infection,
alter the Th1/Th2 balance of the antiviral immune response. suggesting that CD4 T-cell production of IFN- is protec-
tive in the context of RV-induced asthma [34]. Conversely,
CD4 T-cell production of each of IL-4, -5, and -13 was
CD4 T-cells positively correlated with lower respiratory symptom sever-
ity, suggesting CD4 T-cell production of each of IL-4, -5,
The CD4 T-cell response to virus infection is thought and -13 are associated with more severe exacerbations. These
to be of the T-helper 1 (Th1) type. It is thought that an data are novel and important, but causal roles cannot be
eective antiviral immune response is characterized by the established in such human challenge studies. Investigation
production of type 1 cytokines such as IFN-. IFN-, in of the possible causal role of each these cytokines in vivo is
addition to IFN-, IFN-, and IFN- from monocytes now required using the newly developed mouse model [43].
and macrophages, plays a role in establishing an antiviral
state in neighboring cells. IFN- has a complex role in the
pathogenesis of asthma. It appears to increase basophil and CD8 T-cells
mast cell histamine release [161] but on the other hand
inhibits the expression of type 2 cytokines. Production of CD8 T-cells are important eector cells in specic cell-
IFN- is increased in PBMC [162] and in nasal secretions mediated antiviral immunity. They also demonstrate
[104] during RV colds and in human and animal models of polarization of cytokine production, the major Tc1 cytokine
inuenza, parainuenza, and RSV infection [119, 163, 164]. again being IFN- and are believed to regulate CD4 Th1/
There are exceptions where the antiviral response exhibits a Th2 balance [170]. In a murine asthma model induction of
Th2 character or a mixture of Th1/Th2. In animal models bystander CD4 Th2 responses to ovalbumin resulted in a
of RSV, dierent proteins of the virus may induce either switch of virus-peptide specic lung CD8 T-cells to pro-
Th1 or Th2 type responses and priming with such proteins duction of Tc2 cytokines including IL-5 with, after virus
prior to infection with whole virus can inuence the char- peptide challenge, induction of airway eosinophilia [171].
acter, eectiveness, and associated immunopathology of the If this occurs in man it suggests a means whereby CD8
immune response [165]. antiviral function could be inhibited at the same time as
Asthma is believed to be characterized by type 2 CD8 amplication of allergic inammation through IL-5
inammation. Many studies have demonstrated mutual induction of airway eosinophilia. The role of CD8 T-cell
inhibition of Th1 and Th2 cells [166, 167]. It is therefore production of type 1 and type 2 cytokines in virus-induced
possible within an airway with a preexisting type 2 allergic asthma exacerbations requires investigation.
asthmatic microenvironment that there may be inhibition
of the normal eective type 1 antiviral immune response or
that the system may be skewed toward type 2 responses. -TCR T-cells
Papadopoulos et al. have shown that type 1 responses
to RV are decient in individuals with asthma [168]. -TCR T-cells are a minor subset of T-cells expressing
PBMC taken from asthmatics and exposed in vitro to RV receptors distinct from the receptors found on the major-
show lower levels of IFN- and IL-12 and higher levels of ity of T-cells involved in adaptive immunity. There appear to
IL-4 and IL-10 in culture supernatants than cells from nor- be at least two types of -TCR T-cells. The rst type is
mal subjects. The IFN-/IL-4 ratio was three times lower in found in the lymphoid tissue of all vertebrates and displays
the asthmatic group [168]. highly diversied receptors. The second type, intraepithelial
In a study by Gern et al. of experimental RV16 infec- -TCR T-cells, display receptors of limited diversity. It
tion in subjects with allergic rhinitis or asthma, the balance of has been suggested that this second subset recognize mole-
airway Th1 and Th2 cytokines in induced sputum induced by cules expressed only by nearby infected cells. Candidate lig-
viral infection was found to be related to clinical symptoms ands are heat-shock proteins, MHC class IB molecules and
and viral clearance. Although protein could not be detected in unorthodox nucleotides and phospholipids. Antigen is rec-
sputum due to the presence of inhibitors of the ELISA assay ognized directly rather than as processed peptide presented
used, there were increases in mRNA, as determined by semi- by MHC. Recognition of molecules expressed as a conse-
quantitative RT-PCR, for both IL-5 and IFN-. An inverse quence of infection rather than pathogen-specic molecules
correlation was demonstrated between the ratio of IFN- themselves would place -TCR T-cells at the intersection
mRNA to IL-5 mRNA and peak cold symptoms. In addition of innate and adaptive immunity [172].
subjects with RV16 still detectable 14 days after inoculation However, exaggerated responses to various pathogens
had lower IFN-/IL-5 ratios during the acute phase of the and self tissues have been found in studies of mice decient
cold than those subjects who had cleared the virus [169]. in -TCR T-cells rather than deciencies in control
We have recently investigated the production of type 1 of pathogens. Such work has suggested that at least some
and type 2 cytokines from BAL cells in asthmatic and normal -TCR T-cells have a regulatory role in modulating
subjects. We found that production of the type 1 cytokines immune responses [173], a function consistent with their dem-
IL-12 and IFN- were suppressed in the asthmatics, onstrated ability to secrete regulatory cytokines when activated.

481
Asthma and COPD: Basic Mechanisms and Clinical Management

It has been reported that -TCR T-cells are more in a rat model of parainuenza infection. Several viruses can
numerous in the asthmatic airway [174]. A recent study enhance basophil IgE-mediated histamine release, but the
found a greater capacity for production of IL-5 and IL-13 role of this cell in human asthma is controversial.
in bronchoalveolar lavage -TCR T-cells from asthmatic Mast cells are also important sources of inammatory
subjects [175]. If virus infection results in the release of mol- mediators. Their function and localization suggest an early
ecules from epithelial cells that activate -TCR T-cells in interaction with viruses. Leukotriene (LT) C4 is among the
the respiratory mucosa, such cells could provide a source of mediators responsible for the late phase of bronchospasm
type 2 cytokines that inuence the nature of the subsequent in asthma. During RSV infection increased levels of LTC4
immune response. The role of -TCR T-cells in virus- were found in the nasopharyngeal secretions of infants [186].
induced asthma exacerbations requires investigation. Levels correlated well with the symptoms of the disease with
concentrations in infants presenting with bronchiolitis being
vefold higher than in those with only upper respiratory
Eosinophils tract symptomatology. Cultured alveolar macrophages can be
infected with parainuenza virus and respond with an increase
Eosinophils are increased in bronchial epithelium in biop- in arachidonic acid metabolism. Several of the products of this
sies taken from normal and asthmatic volunteers following pathway are known inducers of airway constriction, including
experimental RV infection; in a small study eosinophilic LTC4, LTD4, PGF2, and thromboxanes and/or stimulants of
inammation persisted for up to 6 weeks in asthmatic sub- mucous secretion such as PGF2, LTB4, and 5-hydroxyeicosa-
jects [21] and in our recent study eosinophil numbers in the tetraenoic acid [187]. RV infection has been shown to induce
BAL were signicantly (threefold) increased in asthmatic prostaglandin and LT synthetic enzymes in bronchial biopsies
compared to normal subjects during the acute RV infection in normal subjects, as well as trends for increased numbers of
and correlated signicantly with reductions in lung func- mast cells ( p 0.07) bronchoalveolar lavage uid cysteinyl-
tion only in the asthmatic subjects [34]. In allergic rhinitis leukotriene levels ( p 0.13), but these outcomes have not
experimental RV infection increases BAL eosinophils fol- been studied in asthmatic subjects[188].
lowing segmental allergen challenge, again persisting for 6
weeks [28], and increased levels of ECP are found in the
sputum of RV-infected subjects [23] and during naturally Neutrophils
occurring acute exacerbations of asthma [13]. Eosinophils
accumulate in the airway under the inuence of IL-5, Neutrophils are recruited early during respiratory viral
GM-CSF, IL-8, RANTES, and eotaxin [176]. Of these infection in response to the production of IL-8, Gro-, and
only IL-5 has not been shown to be produced by airway ENA-78 by EC and activated neutrophils are a prominent
EC in vitro after infection by RV. Expression of RANTES feature of severe asthma. Induced sputum IS in asthmatics
is increased in nasal secretions of children with natural virus- and nonasthmatics demonstrates a signicant increase in
induced asthma [107]. RANTES is upregulated in primary neutrophils at day 4 of a natural cold, correlating with spu-
nasal EC cultures by RSV [177] and RV [178]. GM-CSF tum IL-8 [189]. Similar results were obtained in IS taken 2
is important in bone marrow eosinophil production and in and 9 days after experimental RV16 infection in asthmat-
eosinophil survival [176] but levels are not increased dur- ics. Intracellular staining demonstrated an increase in cells
ing viral upper respiratory tract infections [107, 179, 180]. positive for IL-8 at day 2 attributable to increased IL-8
Levels of eotaxin in nasal lavage rise after experimental positive neutrophils [25]. The chemokine IL-8 is a potent
RV16 infection [181]. These data suggest a pathogenic role chemoattractant for neutrophils but also acts on lym-
for eosinophils in virus-induced asthma. However, a protec- phocytes, basophils, and primed eosinophils. Increased IL-
tive role is also possible. In allergic rhinitic subjects, infected 8 has been found in NL from children with natural colds
with RV after high dose allergen challenge, the severity and [104]. Experimental RV16 infection of asthmatics resulted
duration of cold symptoms were inversely related to the NL in elevated NL IL-8, correlating with cold/asthma symp-
eosinophil count prior to infection [32]. Eosinophils may tom scores and histamine PC20 [25]. IS from asthmatics
contribute to viral antigen presentation. Eosinophils pre- with exacerbations has both elevated IL-8 and neutrophilia
treated with GM-CSF bind RV16 via ICAM-1 and present [23, 190]. A study of experimental infection in asthmatic
viral antigen to RV16-specic T-cells, inducing prolifera- children also demonstrated elevated IL-8 and neutrophilia
tion and secretion of IFN- [182]. Eosinophils have anti- in NL during the acute infection and levels of neutrophil
viral actions in parainuenza-infected guinea pigs [183]. myeloperoxidase correlated with symptom severity [191].
EDN and ECP have ribonuclease activity and reduce RSV In asthma, exacerbations in asthmatic adults [13] those
infectivity [184]. The role of the eosinophil in the antiviral with virus infection had increased sputum neutrophils and
immune response thus requires further evaluation. increased neutrophil elastase and more severe clinical dis-
ease. Such studies suggest a prominent role for the neu-
trophil in tissue damage during virus-induced asthma.
Mast cells/basophils
These cells are important sources of inammatory mediators, Natural killer cells
characteristic of allergic inammation in asthma. Mast cell
basal and stimulated histamine release increases after virus Natural killer (NK) cells are an important part of the innate
infection [185]. Airway mast cell numbers are upregulated immune response, their function being the elimination of

482
Infections 37
a variety of target cells including virus-infected cells and suggested that there may be the production of local IgE to
the modulation of adaptive immunity toward viruses [192]. as yet unknown environmental allergens in intrinsic asthma.
Cell killing by NK cells may occur through natural killing, Upregulation of total IgE or virus-/allergen-specic
antibody-dependent cellular cytotoxicity (ADCC), or IgE locally or systemically during respiratory virus infection
apoptotic killing of Fas-positive target cells via membrane would be expected to contribute to the duration and sever-
bound FasL. The ability to directly kill virus-infected cells ity of symptoms of an asthma exacerbation.
is regulated by a balance between inhibitory and activating Intranasal challenge with RV39 results in an increase
receptors [193]. Killer inhibitory receptors (KIRs), Ig-like in total serum IgE in allergic rhinitic subjects but no
receptors that recognize HLA-A, -B, or -C molecules, and increase in preexisting allergen-specic IgE [200]. In chil-
the lectin-like CD94/NKG2A receptor that interacts with dren with asthma, during infection with inuenza A there
HLA-E allow NK cells to recognize cells expressing normal was no change in total IgE but increases were observed in
self MHC class I [194]. Loss of inhibition occurs if potential specic serum IgE to house dust mite and in ex vivo prolif-
target cells have lost class I expression following virus infec- erative and IL-2 responses of lymphocytes challenged with
tion or if they display abnormal class I/peptide complexes. house dust mite allergen [201]. In a study of RSV infec-
NK cells are rapid and ecient producers of cytokines tion in infants the development of serum RSV-specic IgE
such as IFN-, important both in early viral infection in the occurred more frequently in atopics and correlated with
antigen-independent activation of antigen presenting cells clinical wheezing, histamine levels in nasal secretions, and
such as macrophages, dendritic cells, and epithelial cells, hypoxia [202]. There is no information as yet on the pres-
and for biasing the development of CD4 Th1 and CD8 ence of local virus-specic IgE in the airway during asthma
Tc1 cells. Cytokines and chemokines shown to enhance the exacerbations.
activities of NK cells in vitro and in vivo include IFN-/-,
IFN-, TNF-, IL-2, IL-12, IL-15, IL-18, MIP-1 MIP-
1, MCP-1, 2, 3, and RANTES. Transforming growth fac-
tor (TGF)- and IL-10 inhibit NK cell activity [195]. Type
2 cytokines may also modulate NK function, increasing COPD
NK type 2 activities and decreasing NK type 1 activities.
Human NK cells cultured in medium supplemented with Increasing interest in the clinical features and pathogenesis
IL-4 dierentiated into NK type 2 cells, secreting IL-5 and of COPD reects the worldwide importance of the disease.
IL-13 and when cultured in the presence of IL-12, dier- More than 14 million patients are aected in the United
entiated into NK type 1 cells secreting IL-10 and IFN-. States alone. It is predicted to become the third leading
IL-4 and IL-13 have also been shown to suppress IL-2- cause of death worldwide by 2020 [203]. National and glo-
induced cytolytic and proliferative activities and IFN- pro- bal initiatives have been launched and management guide-
duction of human NK cells [196]. NK cell production of lines have been published [204, 205].
IL-5 is enhanced by IL-4 and reduced by IL-10 and IL-12 The frequency of exacerbations is a major factor in
[197]. In a mouse model of asthma, intracellular staining of the quality of life of patients with COPD [206]. The typical
NK cells has demonstrated IL-5 production and depletion clinical features of an exacerbation include increased dysp-
of NK cells resulted in reduced airway eosinophilia [198]. nea, wheezing, cough, sputum production, and worsened gas
The function of NK cells in the asthmatic airway is as exchange. Although noninfectious causes of exacerbations
yet unexplored. It may be that, in an airway environment rich such as allergy, air pollution, or inhaled irritants including
in type 2 cytokines, that NK type 1 function and eective cigarette smoke may be important, acute airway infections
antiviral activity are inhibited. If this is the case then a key are the major precipitants [207]. The infection and conse-
component of the early immune response would be decient quent host inammatory response result in increased airway
and viral clearance would be impaired. In addition, if NK obstruction.
type 2 function is favored by the asthmatic microenviron-
ment, production of type 2 cytokines by NK cells in response
to virus infection might be one mechanism for amplica- Epidemiology
tion of allergic inammation. These hypotheses are as yet
untested in human studies of experimental virus infection. It is likely that two-thirds to three-quarters of COPD exac-
erbations may be caused by viral infections. In a study of
186 patients rhinoviruses, inuenza virus, parainuenza
virus, and coronavirus were signicantly associated with
B-lymphocytes and interaction of viruses COPD exacerbations [208]. Between 60% and 70% of
with IgE-dependent mechanisms exacerbations are associated with preceding symptoms of
a common cold. The frequency of exacerbations requiring
An elevated serum total and allergen-specic IgE are fea- hospitalization is higher in the winter. One explanation for
tures of extrinsic or atopic asthma. IgE-mediated mecha- this could be the increased frequency of respiratory viruses
nisms are certainly important in the pathophysiology of at this time of the year. A recent study of 321 exacerbations
extrinsic asthma. Recent studies suggest a similar airway in 83 patients with moderate to severe COPD using new
pathology in both extrinsic and intrinsic nonatopic asthma diagnostic methods including RT-PCR shows a high inci-
[199] where there is an absence of specic serum IgE dence of viral infection [209]. Viruses were detected in nasal
and negative skin prick tests to aeroallergens. It has been aspirates at exacerbation in almost 40% of cases. Rhinovirus

483
Asthma and COPD: Basic Mechanisms and Clinical Management

was the most common, occurring in 58% of cases where a failures as dened by respiratory deterioration were nearly
virus was present. The presence of virus was associated with twice as likely in the placebo group. Benet from antibiotics
increased dyspnea, cold symptoms, and sore throat and was most evident for patients with most symptoms (dysp-
with prolonged recovery from exacerbation. Earlier stud- nea, increased sputum volume, and sputum purulence).
ies relying on serology and virus culture quote lower virus Guidelines for the use of antibiotics in acute exacer-
detection rates of 1520% [208, 210212]. Other studies of bations of COPD are unclear because of the diculties in
more severe exacerbations using more comprehensive PCR dening the role of bacterial infection in an individual case.
methods conrmed the importance of virus infection, with The American Thoracic Society statement on COPD [205]
viruses being detected in around 50% of exacerbations [152, suggests using antibiotics if there is evidence of infection
213]. Because these studies all involve sampling relatively (fever, leukocytosis, CXR changes) but not all patients with
late in the course of illness, it is likely that these detection bacterial bronchial infection have fever (this is more com-
rates underestimate the true importance of virus infections. mon in viral infection or pneumonia) and few have CXR
The role of bacteria in precipitating exacerbations is changes. The European Respiratory Society recommends
also somewhat controversial. Bacteria may have a primary antibiotics if the sputum is purulent, using standard antibi-
role in the development of an exacerbation and/or represent otics as rst line, and sputum culture if these fail [226].
a secondary superinfection of an initial viral process. Various
bacterial species are present in the airways of 2540% of
patients, even when the COPD is stable but increased fre-
quency of recovery of bacteria during exacerbations (55%) Evidence for a role for bacterial infection in
as well as higher bacterial loads during exacerbations both pathogenesis/progression of COPD
suggest that they play an important role in a signicant
number [152, 214, 215]. Signicant bacterial infection has Bacterial infection has a denite role in the pathogenesis
been suggested when there is an abundance of neutrophils of other chronic lung diseases such as cystic brosis and
in the sputum [216] and when the sputum is purulent and bronchiectasis where bacterial infection is chronic, causing
green (due to neutrophil myeloperoxidase) [217]. Bacteria not only acute exacerbations but also inuencing long-term
may contribute to the pathogenesis of an exacerbation due prognosis [207].
to increased bacterial loads of bacteria already colonizing In these diseases chronic bacterial infection occurs as
diseased airways, however in addition to this, acquisition of the host immune response is unable to clear the bacteria,
new bacterial strains has also been shown to be important, the continuous infection leads to continuous inammatory
increasing the risk of exacerbation over twofold [218]. responses and continuous tissue damage [207]. Host and
The major bacterial organisms associated with bacterial factors attract and activate neutrophils, which pro-
COPD exacerbations are nontypable Haemophilis inuen- duce proteinases and reactive oxygen species. Lung antipro-
zae, Streptococcus pneumoniae, and Moraxella (Branhamella) teinase defenses are overwhelmed. Both proteinase enzymes
catarrhalis [219, 220]. Mycoplasma pneumoniae and and reactive oxygen species cause damage to the epithe-
Chlamydia pneumoniae may play a part [221, 222]. Evidence lium, stimulating mucus production and impairing muco-
also suggests that in more severe patients with a baseline ciliary clearance. Neutrophil elastase stimulates epithelial
FEV1 of 35% predicted or less, gram-negative bacteria espe- cell production of the chemokine IL-8 which attracts fur-
cially enterobacteriaceae and pseudomonas play an important ther neutrophils and in addition impairs phagocytosis by
part in acute exacerbations [223]. destroying antibody and cleaving complement receptors
Recent studies have addressed the role of coinfection from neutrophils and complement components from bacte-
with both bacteria and viruses one study showed this to ria. Neutrophils are also stimulated by cigarette smoke.
occur in 25% of exacerbations, and that patients with dual Identication of bacteria during exacerbation is also
infection had more marked lung function impairment and associated with increased levels of inammatory media-
longer hospitalizations [152]. Another reported that exac- tors in BAL and/or sputum. These include reactive oxidant
erbations with both cold symptoms (a marker of putative species, IL-8, TNF-, neutrophil elastase, LTB4, and mye-
viral infection) and a bacterial pathogen, the FEV1 fall was loperoxidase and many others. These clearly have poten-
greater and symptom count was higher than those with a tial to cause considerable tissue damage, as well as further
bacterial pathogen alone [224]. Thus even in exacerbations recruitment and activation of inammatory cells. COPD
in which viruses are detected, bacteria can also contribute to patients, particularly those at the more severe end of the
exacerbation severity. disease spectrum, may also be chronically colonized by bac-
Although the results of placebo-controlled trials show teria between exacerbations, bacterial numbers then increas-
conicting results, overall the eects of antibiotic treatment ing during exacerbations. In a study using bronchoscopic
also support an etiological role for bacteria in exacerbations protected brush specimens [214] 10 of 40 COPD patients
in some patients. A meta-analysis of nine studies showed a were colonized with bacteria when stable. During exacer-
small overall benet when antibiotics were used for COPD bations 50% had bacteria present and when present, bacte-
exacerbations [225]. The largest study included 362 exacer- rial numbers were greater. When protected brush specimens
bations in 173 outpatients [216]. Compared with placebo, were taken during severe acute exacerbations of COPD
the rate of symptom resolution and improvement of peak requiring ventilation [227] bacteria were detected in 50%
expiratory ow during exacerbations was slightly but signi- but it was not possible to distinguish patients more likely
cantly faster when patients were treated with co-trimoxazole, to have bacteria on the basis of clinical features or other
amoxicillin, or doxycycline. More importantly, treatment investigations.

484
Infections 37
The major bacterial pathogens isolated during bron- increases in lower respiratory tract symptoms, and reduc-
chial infections all form part of the commensal ora in the tions in PEF and FEV1 typical of an acute exacerbation of
nasopharynx. Bronchial infections occur in patients with COPD [237]. Further studies are clearly needed in view of
abnormal airways with reduced host defenses. Persistence the increasing evidence for a major role for viruses in caus-
of bacteria within the bronchial tree may come about ing COPD exacerbations.
through toxins that impair mucociliary clearance, enzymes
that breakdown local immunoglobulin, products that alter
immune eector cell function, adherence to mucus and Therapy for infective exacerbations of
damaged epithelium, or other mechanisms of avoiding asthma and COPD
immune surveillance [207, 228].
Bacterial colonization in the stable state represents Currently much of the treatment of infective exacerba-
an equilibrium in which the number of bacteria present in tions of asthma and COPD is symptomatic, consisting of
the bronchial tree is contained by the host defenses but not increased bronchodilators, either short-acting 2-agonists
eliminated. During an exacerbation this equilibrium is upset in inhaled or intravenous form or anticholinergics or the-
and bacterial numbers increase, inciting an inammatory ophyllines, or supportive in the form of oxygen and in
response. Change will usually occur because of a change severe cases noninvasive or invasive ventilatory measures.
in the host rather than altered virulence of the bacteria, for Corticosteroids are widely used in inhaled or oral form for
example as a result of viral infection. their anti-inammatory actions. The eects of corticos-
teroids are the result of actions at many points in various
inammatory cascades. Whilst this undoubtedly contributes
Evidence for a role for viruses in to their benecial eects it also results in signicant local
pathogenesis/progression of COPD and systemic side eects, in particular if oral steroid treat-
ment is prolonged or frequent. In addition systemic steroids
Exacerbations associated with viral infections also have may interfere with the antiviral immune response resulting
increased levels of many inammatory mediators also found in reduced viral clearance [238].
in bacterial exacerbations including TNF-, IL-8, neutrophil In persistent asthma, control of disease is achieved
elastase, myeloperoxidase, and LTB4. In contrast to bacterial predominantly with inhaled corticosteroids. There is a role
exacerbations where neutrophils and neutrophil products pre- for additional drugs such as long-acting 2-agonists and
dominate, during a viral exacerbation both neutrophils and leukotriene antagonists. The long-acting 2-agonists in par-
eosinophils are present and eosinophil products such as ECP ticular appear to increase the eectiveness of inhaled cor-
are also increased [152]. Other mediators implicated include ticosteroids allowing the dose needed to achieve control to
ENA-78, RANTES, and endothelin1 [229]. be reduced [239]. There is also evidence that these drugs
It has also been suggested that persistent virus infec- in combination with inhaled corticosteroids may further
tion contributes to the progression of COPD. In particu- reduce exacerbation frequency [240]. The leukotriene antag-
lar, adenovirus appears to persist in a latent form in which onists appear to be most eective in treating or preventing
viral proteins are produced without replication of complete exacerbations in children [241, 242].
virus. Such latent infection may amplify lung inammation Regular corticosteroid treatment is however only par-
due to cigarette smoke [230]. Adenoviral E1A DNA per- tially eective at preventing exacerbations. In adult asthma
sists in human lungs from patients with COPD compared inhaled steroids reduce exacerbation frequency by only 40%
with patients of similar age, sex, and smoking history who [243]. In school age children inhaled steroids are ineec-
do not have COPD [231]. The E1A protein has been dem- tive at reducing exacerbation frequency, duration, or severity
onstrated in airway epithelial cells from smokers [232]. It is [244]. In preschool age children with virus-induced wheeze
able to amplify many host genes through attachment to the oral steroids are ineective even in those with primed eosi-
DNA-binding sites of transcription factors [233]. Airway nophils [245].
epithelial cells transfected with E1A produce excess inam- Specic antibiotic therapy is available for bacterial
matory cytokines such as IL-8 [234] and surface adhesion infections and is indicated where there is good evidence of
molecules such as ICAM-1 [235] after in vitro challenge by such infection or when the exacerbation is severe and bacte-
an NF-B-dependent mechanism [236]. rial involvement is a possibility.
RSV has been identied in induced sputum from However, as discussed above the majority of infective
patients with stable COPD. These individuals have a higher asthma exacerbations are of viral rather than bacterial origin
plasma brinogen and serum IL-6, a higher pCO2 and and viruses are also common in exacerbations of COPD.
increased frequency of exacerbations [209]. This suggests
either that low grade persistent RSV infection contributes
to COPD severity or that patients with more severe COPD Vaccination
are less able to clear RSV from the airway.
The immunology of virus infection in COPD is not The success of vaccination to prevent respiratory virus infec-
well understood. Less data is available than for virus infec- tions has been limited by signicant variation within the
tion in asthma since this has not been a major subject of major virus types causing disease. There are 102 serotyped
human experimental infection studies. In a small safety strains of rhinovirus and several more that have not been
study of four patients, inoculation with low dose RV16 serotyped and no eective vaccine has been introduced.
resulted in symptomatic colds, viral replication, signicant A decavalent vaccine [246] developed in the 1970s was

485
Asthma and COPD: Basic Mechanisms and Clinical Management

ultimately of limited ecacy. The inuenza viruses dis- during the inuenza season and, as treatment, they reduce
play antigenic shift and drift. Vaccines must be modied the duration of illness if started within 3648 h of the onset
every 23 years to cover the strains prevalent at the time. of illness. Zanamivir must be given by inhalation whereas
Vaccination against RSV experienced a major setback in oseltamivir can be given orally. Ribavirin is a nucleoside
the 1960s when the use of formalin-inactivated virus in analog active against RSV in vivo and also against inu-
young babies resulted in increased disease severity follow- enza in vitro. Nebulized ribavirin therapy is licensed for use
ing subsequent virus infection [247]. Eighty percent of in hospitalized infants and children in the rst 3 days of
vaccinated children required hospitalization when subse- RSV bronchiolitis. It is however expensive and of unproven
quently infected with RSV, as compared to 5% of controls. benet on clinical outcome. Because of its toxicity it is not
The lungs of two vaccinated children who died contained appropriate for asthma. RSV enriched immunoglobulin
eosinophilic inltrates. It has been suggested that formalin was eective as prophylaxis for infants at high risk of RSV
inactivation may have modied epitopes within the RSV G bronchiolitis [258] but has been superseded by RSV neu-
and F surface glycoproteins, resulting in a modied immune tralizing monoclonal antibodies [259].
response to subsequent infection with enhanced immun-
opathology [248]. Vaccinated individuals demonstrate a
number of dierences from individuals who have suered
natural RSV infection including a lack of specic mucosal
Antirhinoviral agents
antibodies and decient neutralizing and fusion-inhibiting
RV are a major target for drug treatment. It has been esti-
serum antibodies [249]. There are also dierences in the
mated that rhinoviruses result in 610 colds per year in
cell-mediated immune response with some vaccinated indi-
young children [260]. As yet no eective agent is avail-
viduals demonstrating peripheral eosinophilia and exagger-
able for clinical use. Capsid-binding/canyon inhibitors
ated lymphocytic proliferative responses to RSV [250]. To
block RV binding to host cell receptor (ICAM-1 in the
protect against RSV infection a successful vaccine would
case of the major group). One example in phase 3 clinical
need to provide more eective protection than natural
trials is pleconaril (Picovir). These drugs can be extremely
infection, which is itself frequently followed by reinfection
potent but their clinical usefulness is often limited by tox-
[251], and would have to be administered early in infancy
icity, the need for rapid initiation of therapy and the pos-
to have an eect on infant bronchiolitis.
sible development of resistance. Alternative targets include
soluble ICAM-1 which inhibits major rhinovirus infection
Treatment for virus-induced asthma and conserved viral enzymes such as protein 3D, the RNA-
dependent RNA transcriptase, protein 2C, the associated
exacerbations ATP-helicase, and the cysteine protease 3C.
Simple nonspecic treatments for the common cold do exist
although their ecacy is debated. Vitamin C and zinc glu-
conate [252] both may shorten the duration of a cold by 12 New approaches
days. The inhalation of humidied hot air provides sympto-
matic relief [253]. Nasal IFN- is an eective treatment for Alternative approaches to direct antiviral therapy are sup-
the common cold [254] but must be given either prior to or pression of virus-induced inammation, or strategies that
shortly after exposure to the virus. It is also expensive and is promote innate or type 1 immune responses in individuals
associated with signicant local side eects such as bleeding with excessive type 2 responses. Understanding the com-
and discharge. These problems have limited its clinical use. plexities of the antiviral immune response, in particular how
However IFN therapy for virus-induced asthma exacerba- it may be altered in the context of preexisting chronic air-
tions may be more useful in view of the deciencies identi- way diseases such as asthma is an essential rst step. Further
ed by recent studies. Further because of the large number work is needed to elucidate the important sites of interac-
of viruses producing similar clinical syndromes, the gen- tion between the immunological networks of asthma and of
eral antiviral properties of IFNs would provide signicant virus infection. Greater knowledge is required if we are to
advantage over the use of specic antiviral drugs. identify key targets for therapeutic intervention, the aim of
which will be to minimize immunopathology whilst main-
taining or enhancing the host anti-viral immune response.
Antivirals
Specic antiviral agents exist for inuenza. Amantidine
and rimantidine are eective against inuenza A. The use References
of amantidine has been limited by CNS side eects such as
dizziness and insomnia; fewer such side eects are seen with 1. Asher MI, Montefort S, Bjorksten B, Lai CK, Strachan DP, Weiland SK,
Williams H, and ISAAC Phase Three Study Group. Worldwide time
rimantidine. Both drugs are indicated during epidemics for
trends in the prevalence of symptoms of asthma, allergic rhinoconjunc-
treatment and prophylaxis in high risk groups including tivitis, and eczema in childhood: ISAAC Phases One and Three repeat
asthmatics. Neither is active against inuenza B. Two neu- multicountry cross-sectional surveys. Lancet 368: 73343, 2006.
raminidase inhibitors, zanamivir and oseltamivir, are active 2. Stevens FA. Acute asthmatic episodes in children caused by upper res-
against both inuenza A and B [255257]. These agents are piratory bacteria during colds, with and without bacterial sensitization. J
eective in preventing infection when used as prophylaxis Allergy 24: 22126, 1953.

486
Infections 37
3. Bardin PG, Johnston SL, Pattemore PK. Viruses as precipitants of 22. Grunberg K, Kuijpers EA, de Klerk EP, de Gouw HW, Kroes AC,
asthma symptoms. II. Physiology and mechanisms. Clin Exp Allergy Dick EC, Sterk PJ. Eects of experimental rhinovirus 16 infection on
22: 80922, 1992. airway hyperresponsiveness to bradykinin in asthmatic subjects in vivo.
4. McIntosh K, Ellis EF, Homan LS, Lybass TG, Eller JJ, Fulginiti VA. Am J Respir Crit Care Med 155: 83338, 1997.
The association of viral and bacterial respiratory infections with exacer- 23. Grunberg K, Smits HH, Timmers MC, de Klerk EP, Dolhain RJ,
bations of wheezing in young asthmatic children. J Pediatr 82: 57890, Dick EC, Hiemstra PS, Sterk PJ. Experimental rhinovirus 16 infection.
1973. Eects on cell dierentials and soluble markers in sputum in asthmatic
5. Johnston SL, Pattemore PK, Sanderson G, Smith S, Campbell MJ, subjects. Am J Respir Crit Care Med 156: 60916, 1997.
Josephs LK, Cunningham A, Robinson BS, Myint SH, Ward ME 24. Grunberg K, Timmers MC, de Klerk EP, Dick EC, Sterk PJ.
et al. The relationship between upper respiratory infections and hospi- Experimental rhinovirus 16 infection causes variable airway obstruc-
tal admissions for asthma: A time-trend analysis. Am J Respir Crit Care tion in subjects with atopic asthma. Am J Respir Crit Care Med 160:
Med 154: 65460, 1996. 137580, 1999.
6. Minor TE, Dick EC, DeMeo AN, Ouellette JJ, Cohen M, Reed CE. 25. Grunberg K, Timmers MC, Smits HH, de Klerk EP, Dick EC,
Viruses as precipitants of asthmatic attacks in children. JAMA 227: Spaan WJ, Hiemstra PS, Sterk PJ. Eect of experimental rhinovirus
29298, 1974. 16 colds on airway hyperresponsiveness to histamine and interleukin-8
7. Horn ME, Brain EA, Gregg I, Inglis JM, Yealland SJ, Taylor P. in nasal lavage in asthmatic subjects in vivo [see comments]. Clin Exp
Respiratory viral infection and wheezy bronchitis in childhood. Thorax Allergy 27: 3645, 1997.
34: 2328, 1979. 26. Halperin SA, Eggleston PA, Hendley JO, Suratt PM, Groschel DH,
8. Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, Gwaltney JM. Pathogenesis of lower respiratory tract symptoms in
Symington P, OToole S, Myint SH, Tyrrell DA. Community study of experimental rhinovirus infection. Am Rev Respir Dis 128: 80610,
role of viral infections in exacerbations of asthma in 911 year old chil- 1983.
dren. BMJ 310: 122529, 1995. 27. Calhoun WJ, Swenson CA, Dick EC, Schwartz LB, Lemanske RFJ,
9. Chauhan AJ, Inskip HM, Linaker CH, Smith S, Schreiber J, Busse WW. Experimental rhinovirus 16 infection potentiates hista-
Johnston SL, Holgate ST. Personal exposure to nitrogen dioxide (NO2) mine release after antigen bronchoprovocation in allergic subjects. Am
and the severity of virus-induced asthma in children. Lancet 361: 1939 Rev Respir Dis 144: 126773, 1991.
44, 2003. 28. Calhoun WJ, Dick EC, Schwartz LB, Busse WW. A common cold
10. Freymuth F, Vabret A, Brouard J, Toutain F, Verdon R, Petitjean J, virus, rhinovirus 16, potentiates airway inammation after segmen-
Gouarin S, Duhamel JF, Guillois B. Detection of viral, Chlamydia tal antigen bronchoprovocation in allergic subjects. J Clin Invest 94:
pneumoniae and Mycoplasma pneumoniae infections in exacerbations of 22008, 1994.
asthma in children. J Clin Virol 13: 13139, 1999. 29. Green RM, Custovic A, Sanderson G, Hunter J, Johnston SL,
11. Nicholson KG, Kent J, Ireland DC. Respiratory viruses and exacerba- Woodcock A. Synergism between allergens and viruses and risk of
tions of asthma in adults. BMJ 307: 98286, 1993. hospital admission with asthma: Case-control study. BMJ 324: 763,
12. Beasley R, Coleman ED, Hermon Y, Holst PE, ODonnell TV, 2002.
Tobias M. Viral respiratory tract infection and exacerbations of asthma 30. Murray CS, Poletti G, Kebadze T, Morris J, Woodcock A, Johnston SL,
in adult patients. Thorax 43: 67983, 1988. Custovic A. Study of modiable risk factors for asthma exacerbations:
13. Wark PA, Johnston SL, Moric I, Simpson JL, Hensley MJ, Gibson PG. Virus infection and allergen exposure increase the risk of asthma hos-
Neutrophil degranulation and cell lysis is associated with clinical sever- pital admissions in children. Thorax 61: 37682, 2006.
ity in virus-induced asthma. Eur Respir J 19: 6875, 2002. 31. Linaker CH, Coggon D, Holgate ST, Clough J, Josephs L, Chauhan AJ,
14. Grissell TV, Powell H, Shafren DR, Boyle MJ, Hensley MJ, Jones PD, Inskip HM. Personal exposure to nitrogen dioxide and risk of airow
Whitehead BF, Gibson PG. Interleukin-10 gene expression in obstruction in asthmatic children with upper respiratory infection.
acute virus-induced asthma. Am J Respir Crit Care Med 172: 43339, Thorax 55: 93033, 2000.
2005. 32. Avila PC, Abisheganaden JA, Wong H, Liu J, Yagi S, Schnurr D,
15. Berman SZ, Mathison DA, Stevenson DD, Tan EM, Vaughan JH. Kishiyama JL, Boushey HA. Eects of allergic inammation of the
Transtracheal aspiration studies in asthmatic patients in relapse with nasal mucosa on the severity of rhinovirus 16 cold. J Allergy Clin
infective asthma and in subjects without respiratory disease. J Allergy Immunol 105: 92332, 2000.
Clin Immunol 56: 20614, 1975. 33. de Kluijver J, Evertse CE, Sont JK, Schrumpf JA, van Zeijl-van der Ham
16. Gwaltney JMJ, Hendley O, Hayden FG, McIntosh K, Hollinger FB, CJ, Dick CR, Rabe KF, Hiemstra PS, Sterk PJ. Are rhinovirus-induced
Melnick JL, Turner RB. Updated recommendations for safety-testing airway responses in asthma aggravated by chronic allergen exposure?
of viral inocula used in volunteer experiments on rhinovirus colds. Prog [see comment]. Am J Respir Crit Care Med 168: 117480, 2003.
Med Virol 39: 25663, 1992. 34. Message SD, Laza-Stanca V, Mallia P, Parker HL, Zhu J, Kebadze T,
17. Halperin SA, Eggleston PA, Beasley P, Suratt P, Hendley JO, Contoli M, Sanderson G, Kon OM, Papi A, Jeery PK, Stanciu L,
Groschel DH, Gwaltney JM. Exacerbations of asthma in adults during Johnston SL. Rhinovirus induced lower respiratory illness is increased
experimental rhinovirus infection. Am Rev Respir Dis 132: 97680, 1985. in asthma and related to viral load and Th1/2 cytokine and IL-10 pro-
18. Bardin PG, Fraenkel DJ, Sanderson G, Dorward M, Lau LC, duction. ProcNatl AcadSci USA 24, 2008, (in press).
Johnston SL, Holgate ST. Amplied rhinovirus colds in atopic sub- 35. Horn ME, Reed SE, Taylor P. Role of viruses and bacteria in acute
jects. Clin Exp Allergy 24: 45764, 1994. wheezy bronchitis in childhood: A study of sputum. Arch Dis Childhood
19. Lemanske RFJ, Dick EC, Swenson CA, Vrtis RF, Busse WW. 54: 58792, 1979.
Rhinovirus upper respiratory infection increases airway hyperreactivity 36. Gern JE, Galagan DM, Jarjour NN, Dick EC, Busse WW. Detection
and late asthmatic reactions. J Clin Invest 83: 110, 1989. of rhinovirus RNA in lower airway cells during experimentally induced
20. Cheung D, Dick EC, Timmers MC, de Klerk EP, Spaan WJ, Sterk PJ. infection. Am J Respir Crit Care Med 155: 115961, 1997.
Rhinovirus inhalation causes long-lasting excessive airway narrowing 37. Subauste MC, Jacoby DB, Richards SM, Proud D. Infection of a
in response to methacholine in asthmatic subjects in vivo. Am J Respir human respiratory epithelial cell line with rhinovirus. Induction of
Crit Care Med 152: 149096, 1995. cytokine release and modulation of susceptibility to infection by
21. Fraenkel DJ, Bardin PG, Sanderson G, Lampe F, Johnston SL, cytokine exposure. J Clin Invest 96: 54957, 1995.
Holgate ST. Lower airways inammation during rhinovirus colds 38. Papadopoulos NG, Bates PJ, Bardin PG, Papi A, Leir SH, Fraenkel DJ,
in normal and in asthmatic subjects. Am J Respir Crit Care Med 151: Meyer J, Lackie PM, Sanderson G, Holgate ST et al. Rhinoviruses
87986, 1995. infect the lower airways. J Infect Dis 181: 187584, 2000.

487
Asthma and COPD: Basic Mechanisms and Clinical Management

39. Schroth MK, Grimm E, Frindt P, Galagan DM, Konno SI, Love R, 59. Parry DE, Busse WW, Sukow KA, Dick CR, Swenson C, Gern JE.
Gern JE. Rhinovirus replication causes RANTES production in pri- Rhinovirus-induced PBMC responses and outcome of experimental
mary bronchial epithelial cells. Am J Respir Cell Mol Biol 20: 122028, infection in allergic subjects. J Allergy Clin Immunol 105: 69298, 2000.
1999. 60. Corne JM, Marshall C, Smith S, Schreiber J, Sanderson G, Holgate ST,
40. Wark PA, Johnston SL, Bucchieri F, Powell R, Puddicombe S, Johnston SL. Frequency, severity, and duration of rhinovirus infections
Laza-Stanca V, Holgate ST, Davies DE. Asthmatic bronchial epithelial in asthmatic and non-asthmatic individuals: A longitudinal cohort
cells have a decient innate immune response to infection with rhino- study. Lancet 359: 83134, 2002.
virus. J Exp Med 201: 93747, 2005. 61. Wark PA, Johnston SL, Bucchieri F, Powell R, Puddicombe S,
41. Papadopoulos NG, Sanderson G, Hunter J, Johnston SL. Rhinoviruses Laza-Stanca V, Holgate ST, Davies DE. Asthmatic bronchial epithelial
replicate eectively at lower airway temperatures. J Med Virol 58: 100 cells have a decient innate immune response to infection with rhino-
4, 1999. virus. J Exp Med 201: 93747, 2005.
42. Jakiela B, Brockman-Schneider R, Amineva S, Lee WM, Gern JE. 62. Contoli M, Message SD, Laza-Stanca V, Edwards MR, Wark PA,
Basal cells of dierentiated bronchial epithelium are more susceptible Bartlett NW, Kebadze T, Mallia P, Stanciu LA, Parker HL et al. Role
to rhinovirus infection. Am J Respir Cell Mol Biol 38(5): 51723, 2008. of decient type III interferon-lambda production in asthma exacerba-
43. Bartlett NW, Walton RP, Edwards MR, Aniscenko J, Caramori G, tions. Nat Med 12: 102326, 2006.
Zhu J, Glanville N, Choy KJ, Jourdan P, Burnet J et al. Mouse mod- 63. Papi A, Stanciu LA, Papadopoulos NG, Teran LM, Holgate ST,
els of rhinovirus-induced disease and exacerbation of allergic airway Johnston SL. Rhinovirus infection induces major histocompatibility
inammation. Nat Med 14: 199204, 2008. complex class I and costimulatory molecule upregulation on respiratory
44. Bardin PG, Fraenkel DJ, Sanderson G, van Schalkwyk EM, Holgate ST, epithelial cells. J Infect Dis 181: 178084, 2000.
Johnston SL. Peak expiratory ow changes during experimental rhino- 64. Hers JF. Disturbances of the ciliated epithelium due to inuenza virus.
virus infection. Eur Respir J 16: 98085, 2000. Am Rev Respir Dis 93 (Suppl 77), 1966.
45. Gern JE, Calhoun W, Swenson C, Shen G, Busse WW. Rhinovirus 65. Turner RB, Hendley JO, Gwaltney JM. Shedding of infected ciliated
infection preferentially increases lower airway responsiveness in allergic epithelial cells in rhinovirus colds. J Infect Dis 145: 84953, 1982.
subjects. Am J Respir Crit Care Med 155: 187276, 1997. 66. Turner RB, Winther B, Hendley JO, Mygind N, Gwaltney JM. Sites
46. Whitton JL, Oldstone MBA. Immune response to viruses. In: Fields BN, of virus recovery and antigen detection in epithelial cells during experi-
Knipe DN, Howley PM. Fields Virology, 34574. Philadelphia, PA: mental rhinovirus infection. Acta Otolaryngol Suppl 413: 914, 1984.
Lippincott-Raven, 1996. 67. Winther B, Gwaltney JM, Hendley JO. Respiratory virus infection of
47. Yewdell JW, Bennink JR. Immune responses to viruses. In: Richman DR, monolayer cultures of human nasal epithelial cells. Am Rev Respir Dis
Whiteley RJ, Hayden FG. Clinical Virology, 271306. New York: 141: 83945, 1990.
Churchill Livingstone, 1997. 68. Mosser AG, Brockman-Schneider R, Amineva S, Burchell L,
48. Suzuki T, Yamaya M, Kamanaka M, Jia YX, Nakayama K, Hosoda M, Sedgwick JB, Busse WW, Gern JE. Similar frequency of rhinovirus-
Yamada N, Nishimura H, Sekizawa K, Sasaki H. Type 2 rhinovi- infectible cells in upper and lower airway epithelium. J Infect Dis 185:
rus infection of cultured human tracheal epithelial cells: Role of 73443, 2002.
LDL receptor. Am J Physiol Lung Cell Mol Physiol 280: L40920, 69. Greve JM, Davis G, Meyer AM, Forte CP, Yost SC, Marlor CW,
2001. Kamarck ME, McClelland A. The major human rhinovirus receptor is
49. Bjornson AB, Mellencamp MA, Schi GM. Complement is activated ICAM-1. Cell 56: 83947, 1989.
in the upper respiratory tract during inuenza virus infection. Am Rev 70. Winther B, Greve JM, Gwaltney JMJ, Innes DJ, Eastham JR,
Respir Dis 143: 106266, 1991. McClelland A, Hendley JO. Surface expression of intercellular adhe-
50. Amineva SP, Gern JE. Rhinovirus 3C protease cleaves the C3 and C5 sion molecule-1 on epithelial cells in the human adenoid. J Infect Dis
complement factors. Am J Respir Crit Care Med 167: A212, 2003. 176: 52325, 1997.
51. Kopf M, Abel B, Gallimore A, Carroll M, Bachmann MF. 71. Winther B, Arruda E, Witek TJ, Marlin SD, Tsianco MM, Innes DJ,
Complement component C3 promotes T-cell priming and lung migra- Hayden FG. Expression of ICAM-1 in nasal epithelium and levels of
tion to control acute inuenza virus infection. Nat Med 8: 37378, soluble ICAM-1 in nasal lavage uid during human experimental rhi-
2002. novirus infection. Arch OtolaryngolHead Neck Surg 128: 13136, 2002.
52. Anders EM, Hartley CA, Reading PC, Ezekowitz RA. Complement- 72. Bianco A, Spiteri MA. A biological model to explain the association
dependent neutralization of inuenza virus by a serum mannose- between human rhinovirus respiratory infections and bronchial asthma.
binding lectin. J Gen Virol 75: 61522, 1994. Monaldi Arch Chest Dis 53: 8387, 1998.
53. Yang D, Biragyn A, Kwak LW, Oppenheim JJ. Mammalian defensins 73. Grunberg K, Sharon RF, Hiltermann TJ, Brahim JJ, Dick EC, Sterk PJ,
in immunity: more than just microbicidal. Trends Immunol 23: 29196, van Krieken JH. Experimental rhinovirus 16 infection increases inter-
2002. cellular adhesion molecule-1 expression in bronchial epithelium of
54. Barclay WS, al-Nakib W, Higgins PG, Tyrrell DA. The time course of asthmatics regardless of inhaled steroid treatment. Clin Exp Allergy 30:
the humoral immune response to rhinovirus infection. Epidemiol Infect 101523, 2000.
103: 65969, 1989. 74. Whiteman SC, Bianco A, Knight RA, Spiteri MA. Human rhinovirus
55. Alper CM, Doyle WJ, Skoner DP, Buchman CA, Seroky JT, selectively modulates membranous and soluble forms of its intercellu-
Gwaltney JM, Cohen SA. Prechallenge antibodies: Moderators of lar adhesion molecule-1 (ICAM-1) receptor to promote epithelial cell
infection rate, signs, and symptoms in adults experimentally challenged infectivity. J Biol Chem 278: 1195461, 2003.
with rhinovirus type 39. Laryngoscope 106: 1298305, 1996. 75. Altman LC, Ayars GH, Baker C, Luchtel DL. Cytokines and eosinophil-
56. Alper CM, Doyle WJ, Skoner DP, Buchman CA, Cohen S, Gwaltney JM. derived cationic proteins upregulate intercellular adhesion molecule-1 on
Prechallenge antibodies moderate disease expression in adults experi- human nasal epithelial cells. J Allergy Clin Immunol 92: 52736, 1993.
mentally exposed to rhinovirus strain hanks. Clin Infect Dis 27: 11928, 76. Terajima M, Yamaya M, Sekizawa K, Okinaga S, Suzuki T, Yamada N,
1998. Nakayama K, Ohrui T, Oshima T, Numazaki Y et al. Rhinovirus infec-
57. Hastings GZ, Francis MJ, Rowlands DJ, Chain BM. Epitope analysis tion of primary cultures of human tracheal epithelium: Role of ICAM-
of the T cell response to a complex antigen: Proliferative responses to 1 and IL-1beta. Am J Physiol 273: L74959, 1997.
human rhinovirus capsids. Eur J Immunol 23: 23005, 1993. 77. Sethi SK, Bianco A, Allen JT, Knight RA, Spiteri MA. Interferon-
58. Gern JE, Dick EC, Kelly EA, Vrtis R, Klein B. Rhinovirus-specic gamma (IFN-gamma) down-regulates the rhinovirus-induced expres-
T cells recognize both shared and serotype-restricted viral epitopes. sion of intercellular adhesion molecule-1 (ICAM-1) on human airway
J Infect Dis 175: 110814, 1997. epithelial cells. Clin Exp Immunol 110: 36269, 1997.

488
Infections 37
78. Bianco A, Sethi SK, Allen JT, Knight RA, Spiteri MA. Th2 cytokines 97. Osterlund P, Veckman V, Siren J, Klucher KM, Hiscott J, Matikainen S,
exert a dominant inuence on epithelial cell expression of the major Julkunen I. Gene expression and antiviral activity of alpha/beta inter-
group human rhinovirus receptor, ICAM-1. Eur Respir J 12: 61926, ferons and interleukin-29 in virus-infected human myeloid dendritic
1998. cells. J Virol 79: 960817, 2005.
79. Canonica GW, Ciprandi G, Pesce GP, Buscaglia S, Paolieri F, 98. Coccia EM, Severa M, Giacomini E, Monneron D, Remoli ME,
Bagnasco M. ICAM-1 on epithelial cells in allergic subjects: A hallmark Julkunen I, Cella M, Lande R, Uze G. Viral infection and Toll-like
of allergic inammation. Int Arch Allergy Immunol 107: 99102, 1995. receptor agonists induce a dierential expression of type I and lambda
80. Bianco A, Whiteman SC, Sethi SK, Allen JT, Knight RA, Spiteri MA. interferons in human plasmacytoid and monocyte-derived dendritic
Expression of intercellular adhesion molecule-1 (ICAM-1) in nasal cells. Eur J Immunol 34: 796805, 2004.
epithelial cells of atopic subjects: A mechanism for increased rhinovi- 99. Spann KM, Tran KC, Chi B, Rabin RL, Collins PL. Suppression of
rus infection?. Clin Exp Immunol 121: 33945, 2000. the induction of alpha, beta, and lambda interferons by the NS1 and
81. Papi A, Johnston SL. Respiratory epithelial cell expression of vascu- NS2 proteins of human respiratory syncytial virus in human epithelial
lar cell adhesion molecule-1 and its up-regulation by rhinovirus infec- cells and macrophages. J Virol 78: 436369, 2004.
tion via NF-kappaB and GATA transcription factors. J Biol Chem 274: 100. Bartlett NW, Buttigieg K, Kotenko SV, Smith GL. Murine interferon
3004151, 1999a. lambdas (type III interferons) exhibit potent antiviral activity in vivo
82. Papi A, Johnston SL. Rhinovirus infection induces expression of in a poxvirus infection model. J GenVirol 86: 158996, 2005.
its own receptor intercellular adhesion molecule 1 (ICAM-1) via 101. Becker S, Quay J, Soukup J. Cytokine (tumor necrosis factor, IL-6,
increased NF-kappaB-mediated transcription. J Biol Chem 274: and IL-8) production by respiratory syncytial virus-infected human
970720, 1999b. alveolar macrophages. J Immunol 147: 430712, 1991.
83. Barnes PJ, Adcock IM. Transcription factors and asthma. Eur Respir J 102. Einarsson O, Geba GP, Zhu Z, Landry M, Elias JA. Interleukin-11:
12: 22134, 1998. Stimulation in vivo and in vitro by respiratory viruses and induction
84. Papi A, Papadopoulos NG, Degitz K, Holgate ST, Johnston SL. of airways hyperresponsiveness. J Clin Invest 97: 91524, 1996.
Corticosteroids inhibit rhinovirus-induced intercellular adhesion mol- 103. Edwards MR, Johnson MW, Johnston SL. Combination therapy:
ecule-1 up-regulation and promoter activation on respiratory epithelial Synergistic suppression of virus-induced chemokines in airway epi-
cells. J Allergy Clin Immunol 105: 31826, 2000. thelial cells. Am J Respir Cell Mol Biol 34: 61624, 2006.
85. Suzuki T, Yamaya M, Sekizawa K, Yamada N, Nakayama K, Ishizuka S, 104. Corne JM, Lau L, Scott SJ, Davies R, Johnston SL, Howarth PH.
Kamanaka M, Morimoto T, Numazaki Y, Sasaki H. Eects of dexam- The relationship between atopic status and IL-10 nasal lavage levels
ethasone on rhinovirus infection in cultured human tracheal epithelial in the acute and persistent inammatory response to upper respira-
cells. Am J Physiol Lung Cell Mol Physiol 278: L56071, 2000. tory tract infection. Am J Respir Crit Care Med 163: 11017, 2001.
86. Papi A, Papadopoulos NG, Stanciu LA, Bellettato CM, Pinamonti S, 105. Proud D, Gwaltney JMJ, Hendley JO, Dinarello CA, Gillis S,
Degitz K, Holgate ST, Johnston SL. Reducing agents inhibit rhino- Schleimer RP. Increased levels of interleukin-1 are detected in nasal
virus-induced up-regulation of the rhinovirus receptor intercellular secretions of volunteers during experimental rhinovirus colds. J Infect
adhesion molecule-1 (ICAM-1) in respiratory epithelial cells. FASEB Dis 169: 100713, 1994.
J 16: 193436, 2002. 106. Einarsson O, Geba GP, Zhou Z, Landry ML, Panettieri RAJ,
87. Papi A, Papadopoulos NG, Stanciu LA, Degitz K, Holgate ST, Tristram D, Welliver R, Metinko A, Elias JA. Interleukin-11 in respi-
Johnston SL. Eect of desloratadine and loratadine on rhinovirus- ratory inammation. AnnNY Acad Sci 762: 89100, 1995.
induced intercellular adhesion molecule 1 upregulation and promoter 107. Teran LM, Seminario MC, Shute JK, Papi A, Compton SJ, Low JL,
activation in respiratory epithelial cells. J Allergy Clin Immunol 108: Gleich GJ, Johnston SL. RANTES, macrophage-inhibitory protein
22128, 2001. 1alpha, and the eosinophil product major basic protein are released
88. Suzuki T, Yamaya M, Sekizawa K, Hosoda M, Yamada N, Ishizuka S, into upper respiratory secretions during virus-induced asthma exacer-
Yoshino A, Yasuda H, Takahashi H, Nishimura H et al. Erythromycin bations in children. J Infect Dis 179: 67781, 1999.
inhibits rhinovirus infection in cultured human tracheal epithelial cells. 108. Cook DN, Beck MA, Coman TM, Kirby SL, Sheridan JF, Pragnell IB,
Am J Respir Crit Care Med 165: 111318, 2002. Smithies O. Requirement of MIP-1 alpha for an inammatory
89. Samuel CE. Antiviral actions of interferons. Clinical Microbiology response to viral infection. Science 269: 158385, 1995.
Reviews 14:778809, 2001. 109. Johnston SL, Papi A, Bates PJ, Mastronarde JG, Monick MM,
90. Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, Hunninghake GW. Low grade rhinovirus infection induces a pro-
Shah NK, Langer JA, Sheikh F, Dickensheets H, Donnelly RP. longed release of IL-8 in pulmonary epithelium. J Immunol 160:
IFN-lambdas mediate antiviral protection through a distinct class II 617281, 1998.
cytokine receptor complex. Nat Immunol 4: 6977, 2003. 110. Edwards MR, Hewson CA, Laza-Stanca V, Lau HT, Mukaida N,
91. Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, Hershenson MB, Johnston SL. Protein kinase R, IkappaB kinase-
Whitmore TE, Kuestner R, Garrigues U, Birks C, Roraback J et al. IL- beta and NF-kappaB are required for human rhinovirus induced pro-
28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol 4: inammatory cytokine production in bronchial epithelial cells. Mol
6368, 2003. Immunol 44: 158797, 2007.
92. Katze MG, He Y, Gale M. Viruses and interferon: A ght for suprem- 111. Mastronarde JG, He B, Monick MM, Mukaida N, Matsushima K,
acy. Nat Rev Immunol 2: 67587, 2002. Hunninghake GW. Induction of interleukin (IL)-8 gene expression
93. Bergmann M, Garcia-Sastre A, Carnero E, Pehamberger H, Wol K, by respiratory syncytial virus involves activation of nuclear factor
Palese P, Muster T. Inuenza virus NS1 protein counteracts PKR- (NF)-kappa B and NF-IL-6. J Infect Dis 174: 26267, 1996.
mediated inhibition of replication. J Virol 74: 62036, 2000. 112. Bitko V, Velazquez A, Yang L, Yang YC, Barik S. Transcriptional
94. Contoli M, Stanciu L, Message SD, Papi A, Johnston SL. Susceptibility induction of multiple cytokines by human respiratory syncytial virus
to asthma exacerbations: Antiviral immunity and protection against requires activation of NF-kappa B and is inhibited by sodium sali-
asthma exacerbations. In: Johnston SL, OByrne PM. Exacerbations of cylate and aspirin. Virology 232: 36978, 1997.
asthma, 16785. UK: Informa, 2007. 113. Yoon HJ, Zhu Z, Gwaltney JM, Elias JA. Rhinovirus regulation of
95. Kotenko SV. The family of IL-10-related cytokines and their receptors: IL-1 receptor antagonist in vivo and in vitro: A potential mechanism
Related but to what extent?. Cytokine Growth Factor Rev 13: 22340, of symptom resolution. J Immunol 162: 746169, 1999.
2002. 114. Proud D, Naclerio RM, Gwaltney JM, Hendley JO. Kinins are gener-
96. Robek MD, Boyd BS, Chisari FV. Lambda interferon inhibits hepatitis ated in nasal secretions during natural rhinovirus colds. J Infect Dis
B and C virus replication. J Virol 79: 385154, 2005. 161: 12023, 1990.

489
Asthma and COPD: Basic Mechanisms and Clinical Management

115. Proud D, Reynolds CJ, Lacapra S, Kagey-Sobotka A, Lichtenstein LM, 137. Folkerts G, van der Linde HJ, Nijkamp FP. Virus-induced airway
Naclerio RM. Nasal provocation with bradykinin induces symptoms hyperresponsiveness in guinea pigs is related to a deciency in nitric
of rhinitis and a sore throat. Am Rev Respir Dis 137: 61316, 1988. oxide. J Clin Invest 95: 2630, 1995.
116. Christiansen SC, Proud D, Cochrane CG. Detection of tissue kal- 138. Kharitonov SA, Yates D, Barnes PJ. Increased nitric oxide in exhaled
likrein in the bronchoalveolar lavage uid of asthmatic subjects. J Clin air of normal human subjects with upper respiratory tract infections.
Invest 79: 18897, 1987. Eur Respir J 8: 29597, 1995.
117. Christiansen SC, Zuraw BL, Proud D, Cochrane CG. Inhibition of 139. Gentile DA, Doyle WJ, Belenky S, Ranck H, Angelini B, Skoner DP.
human bronchial kallikrein in asthma. Am Rev Respir Dis 13: 1125 Nasal and oral nitric oxide levels during experimental respiratory syn-
31, 1989. cytial virus infection of adults. Acta Otolaryngol 122: 6166, 2002.
118. Christiansen SC, Proud D, Sarno RB, Juergens U, Cochrane CG, 140. Murphy AW, Platts-Mills TA, Lobo M, Hayden F. Respiratory nitric
Zuraw BL. Elevation of tissue kallikrein and kinin in the airways of oxide levels in experimental human inuenza. Chest 114: 45256,
asthmatic subjects after endobronchial allergen challenge. Am Rev 1998.
Respir Dis 145: 9005, 1992. 141. Mogensen TH, Paludan SR. Molecular pathways in virus-induced
119. Nijkamp FP, Folkerts G. Nitric oxide and bronchial reactivity. Clin cytokine production. Microbiol Mol Biol Rev 65: 13150, 2001.
Exp Allergy 24: 90514, 1994. 142. Gern JE, French DA, Grindle KA, Brockman-Schneider RA, Konno S,
120. Nevin BJ, Broadley KJ. Nitric oxide in respiratory diseases. Pharmacol Busse WW. Double-stranded RNA induces the synthesis of specic
Ther 95: 25993, 2002. chemokines by bronchial epithelial cells. Am J Respir Cell Mol Biol 28:
121. Fischer A, Folkerts G, Geppetti P, Groneberg DA. Mediators of 73137, 2003.
asthma: Nitric oxide. Pulm Pharmacol Ther 15: 7381, 2002. 143. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA. Recognition
122. Akaike T, Maeda H. Nitric oxide and virus infection. Immunology of double-stranded RNA and activation of NF-kappaB by Toll-like
101: 3008, 2000. receptor 3. Nature 413: 73238, 2001.
123. Donnelly LE, Barnes PJ. Expression and regulation of inducible nitric 144. Hewson CA, Jardine A, Edwards MR, Laza-Stanca V, Johnston SL.
oxide synthase from human primary airway epithelial cells. Am J Toll-like receptor 3 is induced by and mediates antiviral activity
Respir Cell Mol Biol 26: 14451, 2002. against rhinovirus infection of human bronchial epithelial cells. J Virol
124. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R, 79: 1227379, 2005.
Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of 145. Grunstein MM, Hakonarson H, Maskeri N, Chuang S. Autocrine
asthmatic patients. Lancet 343: 13335, 1994. cytokine signaling mediates eects of rhinovirus on airway respon-
125. Jatakanon A, Lim S, Kharitonov SA, Chung KF, Barnes PJ. siveness. Am J Physiol Lung Cell Mol Physiol 278: L114653,
Correlation between exhaled nitric oxide, sputum eosinophils, and 2000.
methacholine responsiveness in patients with mild asthma. Thorax 53: 146. Oliver BG, Johnston SL, Baraket M, Burgess JK, King NJ, Roth M,
9195, 1998. Lim S, Black JL. Increased proinammatory responses from asth-
126. Rutgers SR, van der Mark TW, Coers W, Moshage H, Timens W, matic human airway smooth muscle cells in response to rhinovirus
Kauman HF, Koeter GH, Postma DS. Markers of nitric oxide infection. Respir Res 7: 71, 2006.
metabolism in sputum and exhaled air are not increased in chronic 147. Johnston SL, Papi A, Monick MM, Hunninghake GW. Rhinoviruses
obstructive pulmonary disease. Thorax 54: 57680, 1999. induce interleukin-8 mRNA and protein production in human mono-
127. Maziak W, Loukides S, Culpitt S, Sullivan P, Kharitonov SA, cytes. J Infect Dis 175: 32329, 1997.
Barnes PJ. Exhaled nitric oxide in chronic obstructive pulmonary 148. Laza-Stanca V, Stanciu L, Message SD, Edwards MR, Gern JE,
disease. Am J Respir Crit Care Med 157: 9981002, 1998. Johnston SL. Rhinovirus replication in human macrophages induces
128. Hoyt JC, Robbins RA, Habib M, Springall DR, Buttery LD, NFkB dependent tumour necrosis factor alpha production. J Virol
Polak JM, Barnes PJ. Cigarette smoke decreases inducible nitric oxide 80(16): 824858, 2006.
synthase in lung epithelial cells. Exp Lung Res 29: 1728, 2003. 149. Gern JE, Dick EC, Lee WM, Murray S, Meyer K, Handzel ZT,
129. Wei XQ, Charles IG, Smith A, Ure J, Feng GJ, Huang FP, Xu D, Busse WW. Rhinovirus enters but does not replicate inside mono-
Muller W, Moncada S, Liew FY. Altered immune responses in mice cytes and airway macrophages. J Immunol 156: 62127, 1996.
lacking inducible nitric oxide synthase. Nature 375: 40811, 1995. 150. Oliver BG, Lim S, Wark P, Laza-Stanca V, King NJ, Black JL,
130. Cifone MG, Ulisse S, Santoni A. Natural killer cells and nitric oxide. Burgess JK, Roth M, Johnston SL. Rhinovirus exposure impairs
Int Immunopharmacol 1: 151324, 2001, [Review] [103 refs]. immune responses to bacterial products in human alveolar macro-
131. de Gouw HW, Grunberg K, Schot R, Kroes AC, Dick EC, Sterk PJ. phages. Thorax 43, 2008.
Relationship between exhaled nitric oxide and airway hyperrespon- 151. Johnston SL. Macrolide antibiotics and asthma treatment. J Allergy
siveness following experimental rhinovirus infection in asthmatic sub- Clin Immunol 117: 123336, 2006.
jects. Eur Respir J 11: 12632, 1998. 152. Papi A, Bellettato CM, Braccioni F, Romagnoli M, Casolari P,
132. Sanders SP, Kim J, Connolly KR, Porter JD, Siekierski ES, Proud D. Caramori G, Fabbri LM, Johnston SL. Infections and airway inam-
Nitric oxide inhibits rhinovirus-induced granulocyte macrophage mation in chronic obstructive pulmonary disease severe exacerbations.
colony-stimulating factor production in bronchial epithelial cells. Am Am J Respir Crit Care Med 173: 111421, 2006.
J Respir Cell Mol Biol 24: 31725, 2001. 153. Peschke T, Bender A, Nain M, Gemsa D. Role of macrophage
133. Sanders SP, Siekierski ES, Porter JD, Richards SM, Proud D. Nitric cytokines in inuenza A virus infections.. Immunobiology 189: 34055,
oxide inhibits rhinovirus-induced cytokine production and viral 1993, [Review] [33 refs].
replication in a human respiratory epithelial cell line. J Virol 72: 934 154. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ,
42, 1998. Pulendran B, Palucka K. Immunobiology of dendritic cells. [Review]
134. Kao YJ, Piedra PA, Larsen GL, Colasurdo GN. Induction and regu- [289 refs]. Ann Rev Immunol 18: 767811, 2000.
lation of nitric oxide synthase in airway epithelial cells by respiratory 155. Beyer M, Bartz H, Horner K, Doths S, Koerner-Rettberg C,
syncytial virus. Am J Respir Crit Care Med 163: 53239, 2001. Schwarze J. Sustained increases in numbers of pulmonary dendritic
135. Ali-Ahmad D, Bonville CA, Rosenberg HF, Domachowske JB. cells after respiratory syncytial virus infection. J Allergy Clin Immunol
Replication of respiratory syncytial virus is inhibited in target cells 113(1): 12733, 2004.
generating nitric oxide in situ. Front Biosci 8: A4853, 2003. 156. Wang H, Peters N, Laza-Stanca V, Nawroly N, Johnston SL,
136. Rimmelzwaan GF, Baars MM, de Lijster P, Fouchier RA, Schwarze J. Local CD11c MHC class II- precursors generate lung
Osterhaus AD. Inhibition of inuenza virus replication by nitric dendritic cells during respiratory viral infection, but are depleted in
oxide. J Virol 73: 888083, 1999. the process. J Immunol 177: 253642, 2006.

490
Infections 37
157. Wang H, Peters N, Schwarze J. Plasmacytoid dendritic cells limit viral selective production of the chemokine RANTES by upper airway
replication, pulmonary inammation, and airway hyperresponsiveness in epithelial cells. J Infect Dis 175: 497504, 1997.
respiratory syncytial virus infection. J Immunol 177(9): 626370, 2006. 178. Schroth MK, Grimm E, Frindt P, Galagan DM, Konno SI, Love R,
158. Grayson MH, Cheung D, Rohlng MM, Kitchens R, Spiegel DE, Gern JE. Rhinovirus replication causes RANTES production in pri-
Tucker J, Battaile JT, Alevy Y, Yan L, Agapov E, Kim EY, mary bronchial epithelial cells. Am J Respir Cell Mol Biol 20: 122028,
Holtzman MJ. Induction of high-anity IgE receptor on lung den- 1999.
dritic cells during viral infection leads to mucous cell metaplasia. J 179. Noah TL, Henderson FW, Henry MM, Peden DB, Devlin RB. Nasal
Exp Med 204(11): 275969, 2007. lavage cytokines in normal, allergic, and asthmatic school-age chil-
159. Bonecchi R, Bianchi G, Bordignon PP, DAmbrosio D, Lang R, dren. Am J Respir Crit Care Med 152: 129096, 1995a.
Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A et al. 180. Noah TL, Henderson FW, Wortman IA, Devlin RB, Handy J,
Dierential expression of chemokine receptors and chemotactic Koren HS, Becker S. Nasal cytokine production in viral acute upper
responsiveness of type 1 T helper cells (Th1s) and Th2s. J Exp Med respiratory infection of childhood. J Infect Dis 171: 58492, 1995b.
187: 12934, 1998. 181. Grei L, Andersson M, Andersson E, Linden M, Myint S, Svensson C,
160. Panina-Bordignon P, Papi A, Mariani A, Di Lucia P, Casoni G, Persson CG. Experimental common cold increases mucosal output of
Bellettato C, Buonsanti C, Miotto D, Mapp C, Villa A, Arrigoni G, eotaxin in atopic individuals. Allergy 54: 12048, 1999.
Fabbri L, Sinigaglia F. The C-C chemokine receptors CCR4 and 182. Handzel ZT, Busse WW, Sedgwick JB, Vrtis R, Lee WM, Kelly EA,
CCR8 identify airway T cells of allergen-challenged atopic asthmat- Gern JE. Eosinophils bind rhinovirus and activate virus-specic T
ics. J Clin Invest 107(11): 135764, 2001. cells. J Immunol 160: 127984, 1998.
161. Huftel MA, Swensen CA, Borcherding WR, Dick EC, Hong R, Kita H, 183. Adamko DJ, Yost BL, Gleich GJ, Fryer AD, Jacoby DB. Ovalbumin
Gleich GJ, Busse WW. The eect of T-cell depletion on enhanced sensitization changes the inammatory response to subsequent
basophil histamine release after in vitro incubation with live inuenza parainuenza infection. Eosinophils mediate airway hyperresponsive-
A virus. Am J Respir Cell Mol Biol 7: 43440, 1992. ness, m(2) muscarinic receptor dysfunction, and antiviral eects. J Exp
162. Hsia J, Goldstein AL, Simon GL, Sztein M, Hayden FG. Peripheral Med 190: 146578, 1999.
blood mononuclear cell interleukin-2 and interferon-gamma produc- 184. Domachowske JB, Dyer KD, Adams AG, Leto TL, Rosenberg HF.
tion, cytotoxicity, and antigen-stimulated blastogenesis during experi- Eosinophil cationic protein/RNase 3 is another RNase A-family ribo-
mental rhinovirus infection. J Infect Dis 162: 59197, 1990. nuclease with direct antiviral activity. Nucleic Acids Res 26: 335863,
163. Corne JM, Holgate ST. Mechanisms of virus induced exacerbations 1998.
of asthma. Thorax 52: 38089, 1997. 185. Folkerts G, Busse WW, Nijkamp FP, Sorkness R, Gern JE. Virus-
164. Folkerts G, Nijkamp FP. Virus-induced airway hyperresponsiveness. induced airway hyperresponsiveness and asthma. Am J Respir Crit
Role of inammatory cells and mediators. Am J Respir Crit Care Med Care Med 157: 170820, 1998.
151: 166673, 1995. 186. Volovitz B, Faden H, Ogra PL. Release of leukotriene C4 in respira-
165. Alwan WH, Kozlowska WJ, Openshaw PJ. Distinct types of lung dis- tory tract during acute viral infection. J Pediatr 112: 21822, 1988.
ease caused by functional subsets of antiviral T cells. J Exp Med 179: 187. Barnes PJ, Chung KF, Page CP. Inammatory mediators of asthma:
8189, 1994. An update. Pharmacol Rev 50: 51596, 1998.
166. Mosmann TR, Sad S. The expanding universe of T-cell subsets: Th1, 188. Seymour ML, Gilby N, Bardin PG, Fraenkel DJ, Sanderson G,
Th2 and more. Immunol Today 17: 13846, 1996. Penrose JF, Holgate ST, Johnston SL, Sampson AP. Rhinovirus infec-
167. Romagnani S. The Th1/Th2 paradigm. Immunol Today 18: 26366, tion increases 5-lipoxygenase and cyclooxygenase-2 in bronchial
1997. biopsy specimens from nonatopic subjects. J Infect Dis 185: 54044,
168. Papadopoulos NG, Stanciu LA, Papi A, Holgate ST, Johnston SL. A 2002.
defective type 1 response to rhinovirus in atopic asthma. Thorax 57: 189. Pizzichini MM, Pizzichini E, Efthimiadis A, Chauhan AJ,
32832, 2002. Johnston SL, Hussack P, Mahony J, Dolovich J, Hargreave FE.
169. Gern JE, Vrtis R, Grindle KA, Swenson C, Busse WW. Relationship Asthma and natural colds. Inammatory indices in induced sputum:
of upper and lower airway cytokines to outcome of experimental rhi- A feasibility study. Am J Respir Crit Care Med 158: 117884, 1998.
novirus infection. Am J Respir Crit Care Med 162: 222631, 2000. 190. Fahy JV, Kim KW, Liu J, Boushey HA. Prominent neutrophilic
170. Hussell T, Spender LC, Georgiou A, OGarra A, Openshaw PJ. Th1 inammation in sputum from subjects with asthma exacerbation.
and Th2 cytokine induction in pulmonary T cells during infection J Allergy Clin Immunol 95: 84352, 1995.
with respiratory syncytial virus. J Gen Virol 77: 244755, 1996. 191. Teran LM, Johnston SL, Schroder JM, Church MK, Holgate ST.
171. Coyle AJ, Erard F, Bertrand C, Walti S, Pircher H, Le Gros G. Role of nasal interleukin-8 in neutrophil recruitment and activation
Virus-specic CD8 cells can switch to interleukin 5 production and in children with virus-induced asthma. Am J Respir Crit Care Med
induce airway eosinophilia. J Exp Med 181: 122933, 1995. 155: 136266, 1997.
172. Holtmeier W, Kabelitz D. gammadelta T cells link innate and adap- 192. Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP.
tive immune responses. Chem Immunol Allergy 86: 15183, 2005. Natural killer cells in antiviral defense: Function and regulation by
173. Holtmeier W. Compartmentalization gamma/delta T cells and their innate cytokines. Ann Rev Immunol 17: 189220, 1999, [Review]
putative role in mucosal immunity. Crit Rev Immunol 23: 47388, [253 refs].
2003. 193. Moretta L, Biassoni R, Bottino C, Mingari MC, Moretta A.
174. Spinozzi F, Agea E, Bistoni O, Forenza N, Bertotto A. gamma delta Human NK-cell receptors. Immunol Today 21: 42022, 2000.
T cells, allergen recognition and airway inammation. Immunol Today 194. Mingari MC, Ponte M, Bertone S, Schiavetti F, Vitale C, Bellomo R,
19: 2226, 1998. Moretta A, Moretta L. HLA class I-specic inhibitory receptors in
175. Krug N, Erpenbeck VJ, Balke K, Petschallies J, Tschernig T, human T lymphocytes: Interleukin 15-induced expression of CD94/
Hohlfeld JM, Fabel H. Cytokine prole of bronchoalveolar lavage- NKG2A in superantigen- or alloantigen-activated CD8 T cells.
derived CD4(), CD8(), and gammadelta T cells in people with Proc Natl Acad Sci USA 95: 117277, 1998.
asthma after segmental allergen challenge. Am J Respir Cell Mol Biol 195. Biron CA. Role of early cytokines, including alpha and beta interfer-
25: 12531, 2001. ons (IFN-alpha/beta), in innate and adaptive immune responses to
176. Gleich GJ. Mechanisms of eosinophil-associated inammation. viral infections. Semin Immunol 10: 38390, 1998, [Review] [76 refs].
J Allergy Clin Immunol 105: 65163, 2000. 196. Peritt D, Robertson S, Gri G, Showe L, Aste-Amezaga M, Trinchieri G.
177. Saito T, Deskin RW, Casola A, Haeberle H, Olszewska B, Ernst PB, Dierentiation of human NK cells into NK1 and NK2 subsets.
Alam R, Ogra PL, Garofalo R. Respiratory syncytial virus induces J Immunol 161: 582124, 1998.

491
Asthma and COPD: Basic Mechanisms and Clinical Management

197. Warren HS, Kinnear BF, Phillips JH, Lanier LL. Production of IL-5 217. Stockley RA, OBrien C, Pye A, Hill SL. Relationship of sputum
by human NK cells and regulation of IL-5 secretion by IL-4, IL-10, color to nature and outpatient management of acute exacerbations of
and IL-12. J Immunol 154: 514452, 1995. COPD. Chest 117: 163845, 2000.
198. Walker C, Checkel J, Cammisuli S, Leibson PJ, Gleich GJ. IL-5 pro- 218. Sethi S, Evans N, Grant BJ, Murphy TF. New strains of bacteria and
duction by NK cells contributes to eosinophil inltration in a mouse exacerbations of chronic obstructive pulmonary disease. N Engl J Med
model of allergic inammation. J Immunol 161: 196269, 1998. 347(7): 46571, 2002.
199. Humbert M, Menz G, Ying S, Corrigan CJ, Robinson DS, Durham SR, 219. Murphy TF, Sethi S. Bacterial infection in chronic obstructive pulmo-
Kay AB. The immunopathology of extrinsic (atopic) and intrinsic nary disease. Am Rev Respir Dis 146: 106783, 1992.
(non-atopic) asthma: More similarities than dierences. Immunol 220. Ball P, Tillotson G, Wilson R. Chemotherapy for chronic bronchitis.
Today 20: 52833, 1999. Controversies Presse Med 24: 18994, 1995.
200. Skoner DP, Doyle WJ, Tanner EP, Kiss J, Fireman P. Eect of rhinovi- 221. Blasi F, Cosentini R, Schoeller MC, Lupo A, Allegra L. Chlamydia
rus 39 (RV-39) infection on immune and inammatory parameters in pneumoniae seroprevalence in immunocompetent and immunocom-
allergic and non-allergic subjects. Clin Exp Allergy 25: 56167, 1995. promised populations in Milan. Thorax 48: 126163, 1993.
201. Lin CY, Kuo YC, Liu WT, Lin CC. Immunomodulation of inuenza 222. Blasi F, Legnani D, Lombardo VM, Negretto GG, Magliano E,
virus infection in the precipitating asthma attack. Chest 93: 123438, Pozzoli R, Chiodo F, Fasoli A, Allegra L. Chlamydia pneumoniae
1988. infection in acute exacerbations of COPD. Eur Res J 6: 1922, 1993.
202. Welliver RC, Wong DT, Sun M, Middleton EJ, Vaughan RS, Ogra PL. 223. Eller J, Ede A, Schaberg T, Niederman MS, Mauch H, Lode H.
The development of respiratory syncytial virus-specic IgE and the Infective exacerbations of chronic bronchitis: Relation between bacte-
release of histamine in nasopharyngeal secretions after infection. New riologic etiology and lung function. Chest 113: 154248, 1998.
Engl J Med 305: 84146, 1981. 224. Wilkinson TM, Hurst JR, Perera WR, Wilks M, Donaldson GC,
203. Murray CJ, Lopez AD. Alternative projections of mortality and dis- Wedzicha JA. Eect of interactions between lower airway bacterial
ability by cause 19902020: Global burden of disease study. Lancet and rhinoviral infection in exacerbations of COPD. Chest 129(2):
349: 1498504, 1997. 31724, 2006.
204. Anonymous. BTS guidelines for the management of chronic 225. Saint S, Bent S, Vittingho E, Grady D. Antibiotics in chronic
obstructive pulmonary disease. The COPD Guidelines Group of obstructive pulmonary disease exacerbations. A meta-analysis. JAMA
the Standards of Care Committee of the BTS. Thorax 52(Suppl 5): 273: 95760, 1995.
S128, 1997. 226. Siafakas NM, Vermeire P, Pride NB, Paoletti P, Gibson J, Howard P,
205. Anonymous. Standards for the diagnosis and care of patients with Yernault JC, Decramer M, Higenbottam T, Postma DS. Optimal
chronic obstructive pulmonary disease. American Thoracic Society. assessment and management of chronic obstructive pulmonary dis-
Am J Respir Crit Care Med 152: S77121, 1995. ease (COPD). The European Respiratory Society Task Force. Eur
206. Seemungal TA, Donaldson GC, Paul EA, Bestall JC, Je ries DJ, Respir J 8: 1398420, 1995.
Wedzicha JA. Eect of exacerbation on quality of life in patients with 227. Fagon JY, Chastre J, Trouillet JL, Domart Y, Dombret MC, Bornet M,
chronic obstructive pulmonary disease. Am J Respir Crit Care Med Gibert C. Characterization of distal bronchial microora during
157: 141822, 1998. acute exacerbation of chronic bronchitis. Use of the protected speci-
207. Wilson R. The role of infection in COPD. Chest 113: 242S48S, men brush technique in 54 mechanically ventilated patients. Am Rev
1998. Respir Dis 142: 10048, 1990.
208. Smith CB, Golden CA, Kanner RE, Renzetti AD. Association of 228. Wilson R, Dowling RB, Jackson AD. The biology of bacterial coloni-
viral and Mycoplasma pneumoniae infections with acute respiratory ill- zation and invasion of the respiratory mucosa. Eur Respir J 9: 1523
ness in patients with chronic obstructive pulmonary diseases. Am Rev 30, 1996.
Respir Dis 121: 22532, 1980. 229. Sykes A, Mallia P, Johnston SL. Diagnosis of pathogens in exacerba-
209. Seemungal TA, Harper-Owen R, Bhowmik A, Moric I, Sanderson G, tions of chronic obstructive pulmonary disease. Proc Am Thorac Soc 4:
Message SD, MacCallum P, Meade TW, Je ries DJ, Johnston SL, 64246, 2007.
Wedzicha JA. The role of respiratory viral infections in COPD. Am J 230. Hogg JC. Childhood viral infection and the pathogenesis of asthma
Respir Crit Care Med, 164(9): 161823, 2001. and chronic obstructive lung disease. Am J Respir Critl Care Med 160:
210. Gump DW, Phillips CA, Forsyth BR, McIntosh K, Lamborn KR, S2628, 1999.
Stouch WH. Role of infection in chronic bronchitis. Am Rev Respir 231. Matsuse T, Hayashi S, Kuwano K, Keunecke H, Jeeries WA, Hogg JC.
Dis 113: 46574, 1976. Latent adenoviral infection in the pathogenesis of chronic airways
211. Tager I, Speizer FE. Role of infection in chronic bronchitis. New Engl obstruction. Am Rev Respir Dis 146: 17784, 1992.
J Med 292: 56371, 1975. 232. Elliott WM, Hayashi S, Hogg JC. Immunodetection of adenoviral
212. Greenberg SB, Allen M, Wilson J, Atmar RL. Respiratory viral infec- E1A proteins in human lung tissue. Am J Respir Cell Mol Biol 12:
tions in adults with and without chronic obstructive pulmonary dis- 64248, 1995.
ease. Am J Respir Crit Care Med 162: 16773, 2000. 233. Liu F, Green MR. Promoter targeting by adenovirus E1a through
213. Rohde G, Wiethege A, Borg I, Kauth M, Bauer TT, Gillissen A, interaction with dierent cellular DNA-binding domains. Nature
Bufe A, Schultze-Werninghaus G. Respiratory viruses in exacerba- 368: 52025, 1994.
tions of chronic obstructive pulmonary disease requiring hospitalisa- 234. Keicho N, Elliott WM, Hogg JC, Hayashi S. Adenovirus E1A upreg-
tion: A case-control study. Thorax 58: 3742, 2003. ulates interleukin-8 expression induced by endotoxin in pulmonary
214. Monso E, Ruiz J, Rosell A, Manterola J, Fiz J, Morera J, Ausina V. epithelial cells. Am J Physiol 272: L104652, 1997.
Bacterial infection in chronic obstructive pulmonary disease. A study 235. Keicho N, Elliott WM, Hogg JC, Hayashi S. Adenovirus E1A gene
of stable and exacerbated outpatients using the protected specimen dysregulates ICAM-1 expression in transformed pulmonary epithelial
brush. Am J Respir Crit Care Med 152: 131620, 1995. cells. Am J Respir Cell Mol Biol 16: 2330, 1997.
215. Irwin RS, Erickson AD, Pratter MR, Corrao WM, Garrity FL, 236. Keicho N, Higashimoto Y, Bondy GP, Elliott WM, Hogg JC, Hayashi
Myers JR, Kaemmerlen JT. Prediction of tracheobronchial coloniza- S. Endotoxin-specic NF-kappaB activation in pulmonary epithelial
tion in current cigarette smokers with chronic obstructive bronchitis. cells harboring adenovirus E1A. Am J Physiol 277: L52332, 1999.
J Infect Dis 145: 23441, 1982. 237. Mallia P, Message SD, Kebadze T, Parker HL, Kon OM, Johnston SL.
216. Anthonisen NR, Manfreda J, Warren CP, Hersheld ES, Harding GK, An experimental model of rhinovirus induced chronic obstructive
Nelson NA. Antibiotic therapy in exacerbations of chronic obstructive pulmonary disease exacerbations: A pilot study. Respir Res 7(116),
pulmonary disease. Ann Int Med 106: 196204, 1987. 2006.

492
Infections 37
238. Gustafson LM, Proud D, Hendley JO, Hayden FG, Gwaltney JM. serum neutralizing and glycoprotein antibody responses of infants and
Oral prednisone therapy in experimental rhinovirus infections. children who received inactivated respiratory syncytial virus vaccine.
J Allergy Clin Immunol 97: 100914, 1996. J Clin Microbiol 24: 197202, 1986.
239. Eickelberg O, Roth M, Lorx R. Ligand-independent activation of the 250. Kim HW, Leikin SL, Arrobio J, Brandt CD, Chanock RM, Parrott RH.
glucocorticoid receptor by 2-adrenergic receptor agonists in primary Cell-mediated immunity to respiratory syncytial virus induced by
human lung broblasts and vascular smooth muscle cells. J Biol Chem inactivated vaccine or by infection. Pediatr Res 10: 7578, 1976.
274: 100510, 1999. 251. Glezen WP, Taber LH, Frank AL, Kasel JA. Risk of primary infec-
240. Pauwels RA, Lofdahl CG, Postma DS, Tatterseld AE, OByrne P, tion and reinfection with respiratory syncytial virus. Am J Dis Child
Barnes PJ, Ullman A. Eect of inhaled formoterol and budesonide on 140: 54346, 1986.
exacerbations of asthma. Formoterol and Corticosteroids Establishing 252. Marshall S. Zinc gluconate and the common cold. Review of rand-
Therapy (FACET) International Study Group. New Engl J Med 337: omized controlled trials. Can Fam Physician 44: 103742, 1998.
140511, 1997. 253. Singh M. Heated, humidied air for the common cold. Cochrane
241. Robertson CF, Price D, Henry R, Mellis C, Glasgow N, Fitzgerald D, Database of Syst Rev4: CD001728, 2004.
Lee AJ, Turner J, Sant M. Short-course montelukast for intermittent 254. Hayden FG, Gwaltney JM. Intranasal interferon-alpha 2 treatment of
asthma in children: A randomised controlled trial. Am J Respir Crit experimental rhinoviral colds. J Infect Dis 150: 17480, 1984.
Care Med 175(4): 32329, 2007. 255. Khare MD, Sharland M. Inuenza. Expert Opinion on Pharmaco-
242. Bisgaard H, Zielen S, Garcia-Garcia ML, Johnston SL, Gilles L, therapy 1: 36775, 2000.
Menten J, Tozzi CA, Polos P. Montelukast reduces asthma exacer- 256. Lalezari J, Campion K, Keene O, Silagy C. Zanamivir for the treat-
bations in 2- to 5-year-old children with intermittent asthma. Am J ment of inuenza A and B infection in high-risk patients: a pooled
Respir Crit Care Med 171: 31522, 2005. analysis of randomized controlled trials. Archives of Internal Medicine
243. Pauwels RA, Pedersen S, Busse WW, Tan WC, Chen YZ, Ohlsson SV, 161: 21217, 2001.
Ullman A, Lamm CJ, OByrne PM START Investigators Group. 257. Monto AS, Robinson DP, Herlocher ML, Hinson JMJ, Elliott MJ,
Early intervention with budesonide in mild persistent asthma: A ran- Crisp A. Zanamivir in the prevention of inuenza among healthy
domised, double-blind trial. Lancet 361: 107176, 2003. adults: a randomized controlled trial. JAMA 282: 3135, 1999.
244. Doull IJ, Lampe FC, Smith S, Schreiber J, Freezer NJ, Holgate ST. 258. Rodriguez WJ, Gruber WC, Welliver RC, Groothuis JR, Simoes EA,
Eect of inhaled corticosteroids on episodes of wheezing associated Meissner HC, Hemming VG, Hall CB, Lepow ML, Rosas AJ
with viral infection in school age children: Randomised double blind et al. Respiratory syncytial virus (RSV) immune globulin intravenous
placebo controlled trial. BMJ 315: 85862, 1997. therapy for RSV lower respiratory tract infection in infants and young
245. Oommen A, Lambert PC, Grigg J. Ecacy of a short course of children at high risk for severe RSV infections: Respiratory Syncytial
parent-initiated oral prednisolone for viral wheeze in children aged Virus Immune Globulin Study Group. Pediatrics 99: 45461, 1997,
15 years: Randomised controlled trial. Lancet 362: 143338, 2003. [see comments].
246. Hamory BH, Hamparian VV, Conant RM, Gwaltney JM. Human 259. Wu H, Pfarr DS, Johnson S, Brewah YA, Woods RM, Patel NK,
responses to two decavalent rhinovirus vaccines. J Infect Dis 132: White WI, Young JF, Kiener PA. Development of motavizumab, an
62329, 1975. ultra-potent antibody for the prevention of respiratory syncytial virus
247. Kim HW, Canchola JG, Brandt CD, Pyles G, Chanock RM, Jensen K, infection in the upper and lower respiratory tract. J Mol.Biol 368(3):
Parrott RH. Respiratory syncytial virus disease in infants despite prior 65265, 2008.
administration of antigenic inactivated vaccine. Am J Epidemiol 89: 260. Pattemore PK, Johnston SL, Bardin PG. Viruses as precipitants of
42234, 1969. asthma symptoms. I. Epidemiology. Clinical & Experimental Allergy
248. Hall CB. Respiratory syncytial virus and parainuenza virus. New 22: 32536, 1992.
Engl J Med 344: 191728, 2001.
249. Murphy BR, Prince GA, Walsh EE, Kim HW, Parrott RH,
Hemming VG, Rodriguez WJ, Chanock RM. Dissociation between

493
Exercise as a Stimulus
38 CHAPTER

OVERVIEW not allowed patients with COPD to achieve nor- Sandra D. Anderson1
mal exercise performance, as asthmatics have done,
exercise has become an important intervention in and Jennifer A. Alison2
There are substantial dierences in the pulmo- the management of patients with COPD.
1
nary response between those with asthma and Department of Respiratory Medicine,
those with chronic obstructive pulmonary disease Royal Prince Alfred Hospital,
(COPD) when exercise is used as a stimulus. For Camperdown, NSW, Australia
the person with asthma, exercise is a stimulus for EXERCISE AND THE PERSON 2
Discipline of Physiotherapy, Faculty of
bronchodilatation and for bronchoconstriction
and the airway response varies according to rest-
WITH ASTHMA Health Sciences, University of Sydney,
Lidcombe, NSW, Australia
ing lung function, duration and intensity of exer-
cise, and the temperature and water content of The cardiopulmonary response to exercise in
the air inspired. The stimulus for exercise-induced asthmatics is extremely variable. When expira-
bronchoconstriction (EIB) relates to the ther- tory ow rates and airways resistance are normal,
mal and osmotic consequences of water loss by at rest and during exercise, the cardiopulmonary
evaporation from the airway surface when large response to exercise is not signicantly dier-
volumes of air need to be conditioned in a short ent to healthy subjects [1]. Asthmatics however
time. For most asthmatics however, EIB can be commonly have a higher ventilatory equivalent
well controlled with treatment and the emphasis (ventilation per unit of oxygen consumption)
has moved from exercise as a stimulus for provok- compared with non-asthmatics although this
ing attacks of asthma to exercise for the potential can normalize following physical training [2].
benets to health and lifestyle. For those with Asthmatic children with moderate to severe
COPD, exercise does not usually provide a dehy- asthma have reduced aerobic capacity, endurance
drating stimulus sucient to provoke EIB simply time, and cardiac function [3], particularly at
because the subjects cannot ventilate at a rate suf- high altitude [4]. Expiratory ow limitation
cient to achieve signicant loss of water from the and hyperination are likely contributors to the
airway surface. Some people with COPD how- increased work of breathing and shortness of
ever do demonstrate the airway hyperresponsive- breath in some people with asthma.
ness of asthma in response to inhaling aerosols Physical inactivity is thought to contrib-
of hyperosmolar agents such as saline or man- ute to the development of asthma because
nitol. The more classic response exercise provides bronchial smooth muscle can become sti and
to the patient with COPD is dynamic hyper- hyperresponsive without the benet of periodic
ination that increases end-expiratory lung vol- stretch from the increased tidal volume of exer-
ume (EELV) and decreases inspiratory capacity cise [5]. There are many reports of asthmatic
(IC). This strategy serves to reduce expiratory ow children being physically inactive and unt, par-
limitation and increase tidal volume but at a cost of ticularly those who are obese or overweight, but
greater respiratory eort that contributes to exer- being unt is not a universal nding in asth-
cise limitation. As with treatment of asthma there matic children [1]. Neither body mass index nor
has been a marked improvement in treatment of baseline lung function predicted exercise limita-
COPD in recent years. Although treatment has tion in children with asthma [6].

495
Asthma and COPD: Basic Mechanisms and Clinical Management

LUNG FUNCTION AND GAS EXCHANGE ([79], Figs 38.1 and 38.2). There are three phases of changes
DURING AND AFTER EXERCISE in the airway resistance (Raw) to exercise. During exercise
of variable intensity, Raw decreases when exercise intensity
is increased and increases when exercise intensity is reduced
It is the propensity for airway resistance and arterial oxygen [10]. During intense steady-state exercise there is an early
tension to change rapidly in response to exercise that is of phase in which Raw falls rapidly. This is not due to release
primary physiological importance to a person with asthma of nitric oxide or due to prostaglandins (PGs) [11, 12] but
maybe due to tidal volume stretch reducing bronchial smooth
muscle tone [13]. This early fall in Raw during exercise is fol-
5
lowed by a slow and progressive rise in Raw after the rst few
minutes, then on the cessation of 68 min of vigorous exer-
4 cise there is a marked increase in Raw. It is common for the
10% normal increase in Raw to be associated with a fall in peak expiratory
ow (PEF) or FEV1 from pre-exercise level of 10% or more
FEV1 (l)

3 25% mild within the rst 5 min. The terms commonly used to describe
these changes in lung function following exercise are EIB or
25% moderate exercise-induced asthma (Fig. 38.1).
2 50% severe
In untrained asthmatics with moderate airow limita-
tion at rest (% predicted FEV1  75%), the fall in Raw or bron-
Exercise chodilatation of the rst few minutes of exercise is associated
1 with an increase in arterial oxygen tension (PaO2) [7, 8, 14]
0 8 14 20 (Fig. 38.2). After exercise ceases the PaO2 falls in concert with
Time (min) the reduction in lung function and as a result of ventilation
perfusion inequality [7, 15, 16]. Carbon dioxide levels in asth-
FIG. 38.1 Changes in forced expiratory volume during and after exercise.
The severity of the response is assessed by the fall in FEV1 after exercise
matics usually, but not always, remain within normal limits
expressed as a percentage of the pre-exercise value. A normal response (40 mmHg) both during exercise and after exercise [8, 15].
is usually regarded as a fall less than 10% of baseline. A value less than Recovery to pre-exercise lung function occurs spontaneously
1025% is mild, 25.149.9% as moderate, and 50% or more as severe. The over 3060 min, or usually within 10 min in response to a
values refer to the severity of EIB in those not taking an ICS. In those taking bronchodilator, with PaO2 increasing with the improvement
ICS a value greater than 30% would be generally accepted as severe. in lung function. The lung function and gas exchange changes

100
42
PEFR % pred

80
39
PCO2

60
36

40 33
7.44
100
PaO2

7.40
pH

80
7.36

98
3
Base excess

0
SaO2

96
3

n 9 6
94
Rest After 2 min End Lowest Rest After 2 min End Lowest
exercise exercise post ex exercise exercise post ex

FIG. 38.2 Mean values for PEF expresses as a percentage of predicted, arterial oxygen tension (PaO2 mmHg), arterial saturation (SaO2 %), arterial carbon
dioxide tension (PaCO2 mmHg), pH, and base excess (BE) at baseline, during, and after exercise after placebo (broken line) and 70 min later after pre-
medication with terbutaline sulfate (1250 g 20 min after exercise, and another 1250 g 50 min later). The time between exercise tests was 90 min.
The data are from the study of Ref. [8].

496
Exercise as a Stimulus 38
associated with EIB can sometimes be extreme, and deaths in The intervention that has the greatest inuence on
young athletes, though uncommon, have been reported [17]. these factors is the medication being taken by the asthmatic
Dierences from this classic pattern of gas exchange subject, either daily as with ICS, long acting 2-agonist
described above have been reported in some trained asth- (LABA), or leukotriene receptor antagonists (LTRA), or
matics with good lung function (i.e. FEV1 94% predicted) prophylactically before exercise as with short acting 2 ago-
exercising at higher workloads for longer periods than 8 min. nists (SABA), sodium cromoglycate (SCG), or nedocromil
In one study, 8 of 21 asthmatic subjects were unable to sustain sodium [8, 2226]. Treatment of airway inammation and
their SaO2 above 94% during 11 min of strenuous treadmill prevention of bronchoconstriction during and after exercise
exercise (VO2 maximum range from 39.9 to 61.8 ml/min/kg). can lead to improved gas exchange (Fig. 38.2) and normal
The low SaO2 was attributed to a widened alveolar-arterial exercise performance [8]. Diets that include reduction in
oxygen gradient (A-aO2) and ventilatory insuciency that sodium intake [27], and supplementation with omega 3
resulted in a signicant increase in PaCO2 in some subjects free fatty acids [28] have been reported to reduce severity of
[9]. It was not possible to predict from pre-exercise lung func- EIB but their eect on gas exchange has not been reported.
tion or blood gases which asthmatic subject would desaturate Many young people with self-reported exercise
in response to exercise. In these trained asthmatics, who were symptoms are diagnosed clinically as having EIB but fail
not taking inhaled corticosteroids (ICS), the arterial hypox- to demonstrate EIB on objective testing and may thus be
emia, expiratory ow limitation, and dynamic hyperination over treated [29, 30]. In contrast, many young t and clini-
during exercise occurred at lower metabolic rates than would cally well controlled asthmatics with normal lung func-
be sometimes documented in highly trained healthy subjects tion can have a signicant reduction in lung function and
[9]. The ndings have signicant implications for untreated gas exchange abnormalities after exercise. Some may have
asthmatics wishing to participate in competitive high endur- become tolerant to the protective eects of 2-agonists
ance sports and support the policy that athletes with asthma [23], and others may be receiving too low a dose of inhaled
would benet from having objective tests [18]. steroids [25] (Fig. 38.3). These are reasons why exercise is
and should remain a concern for people with asthma.

PHYSICAL TRAINING AND EIB


DETERMINANTS OF EIB
The aim of exercise training in an asthmatic who is unt
is to increase the exercise intensity and thus the threshold
The most important external determinants of the severity
of ventilation that will provoke EIB [2]. Exercise training
of the airway response to exercise in asthmatics are the level
in an unt asthmatic results in an increase in maximum
oxygen uptake per kilogram and a reduction in ventilatory
equivalent and a modest improvement in maximum heart
si e
Vi elin

rate [19]. Since ventilation determines severity of EIB for


Vi t 1
Vi t 2
t3
s

si
si
Ba

an individual, physical tness should be encouraged, and at 0


an early age.
It is important to know tness does not eliminate
potential for EIB [20], but becoming t will increase the
workload to provoke EIB. Physical training does not
Mean max fall FEV1 %  1 SE

improve any parameters of lung function including PEF, 10


FEV1, FVC, or maximum ventilation [19]. It is for this
reason that treatment of the airway inammation is recom-
mended so that an asthmatic has the best opportunity to
perform normally at any level of competition [21]. *
20
*
* *
FACTORS WITH POTENTIAL TO INFLUENCE * *
*

THE ASTHMATIC RESPONSE TO EXERCISE *


* *
30 *
There are many endogenous factors that have the potential
to impact on the airway response to exercise in asthmatics.
These include severity of the underlying airway inamma- 40 g 80 g 160 g 320 g
tion, the associated bronchial smooth muscle responsiveness FIG. 38.3 Maximum percent fall in FEV1 from baseline after 8 min of
and increased vascular permeability, the degree of atopy, running exercise breathing dry air exercise in two groups of young adults
and the pre-exercise lung function in relation to predicted with repeatable EIB, that is, 15% or more fall in FEV1 at baseline. The fall
normal. Other factors include age and tness of the subject, is shown at baseline and after treatment with an ICS in a dose of 40 and
and body mass index, particularly for its potential to impact 160 g in one group and 80 and 320 g in another for a duration of 1, 2,
on functional residual capacity. and 3 weeks. Reproduced from the study of Ref. [25].

497
Asthma and COPD: Basic Mechanisms and Clinical Management

of ventilation reached and sustained during exercise and the subjects repeated exposure to plasma may induce passive
water content of the inspired air [31]. Thus exercise load is very sensitization of the bronchial smooth muscle in vivo [34],
important [32]. Whilst temperature of the air inspired during increasing its sensitivity to mast cell mediators [37]. In
exercise can inuence severity of EIB, it is of less importance healthy subjects mast cells are more abundant in the smaller
than the absolute water content. Inspiring air close to body airways compared with the larger airway [38]. Thus they are
temperature and fully saturated with water prevents EIB. well positioned to release histamine and other mediators that
That hyperpnea of dry air at high ventilation rates increase the plasma exudation from post-capillary venules.
is required to provoke EIB suggests that severity of EIB
depends on the rate at which generations of airways are
recruited into the air conditioning process [33]. This con-
cept explains why, for an individual, EIB occurs more often
and more severely the drier and cooler the inspired air and
STIMULUS AND MECHANISM OF EIB
the more rapid the increase in ventilation. It may also explain
why EIB may be reported for the rst time in athletes when The stimulus for exercise to provoke the airways of asthmatics
they have reached their full ventilatory capacity in their to narrow is the loss of water from the airway surface that
early twenties and can achieve ventilation rates of 100 l/min occurs in conditioning large volumes of air in a short time
or more [34]. It also explains why patients with COPD (Fig. 38.4). That is why EIB occurs more often when the
are unlikely to have EIB provoked by exercise, that is, they exercise is intense [32, 39] and performed in a dry envi-
cannot reach high enough ventilation during exercise. ronment [40]. It is also the reason that eucapnic voluntary
At the very high ventilation chronically achieved by hyperpnea of dry air (EVH) is an excellent surrogate for
cross-country skiers, rowers, and cyclists, a large surface area exercise to provoke EIB [41]. The mechanism whereby the
of airways including very small airways (1 mm in diameter) loss of water causes airway narrowing relates to the thermal
will be used to condition the air. If very small airways become (cooling) and osmotic consequences of dehydration of the
dehydrated and sustain injury then the restorative process of airway surface and the underlying mucosa [42].
plasma exudation [35] that follows may explain pathogenesis The thermal hypothesis proposes that EIB is a vas-
of EIB [34]. If exposure to plasma-derived products occurs cular event. Thus cooling of the airway surface from condi-
regularly then the contractile properties of the bronchial tioning inspired air causes vasoconstriction of the bronchial
smooth muscle may be altered over time [36]. In allergic vessels and a reactive hyperemia follows immediately after

Water loss from humidifying inspired air Heat loss


Recruitment of small
airways (1 mm) into
Dehydration of the airway surface liquid (ASL) humidifying process

Airway cooling
Epithelial damage,
Increase in [Na] [CI] [Ca] [K] loss of protective
mediator PGE2

Increased in osmolarity of ASL Glandular secretion


Microvascular leak and
Sensory exudation of plasma
Water moves from all cells to restore ASL nerves Cell

Mucus
Repeated exposure to plasma
Cell shrinkage products alters properties
Cough 
Followed by release of mediators of airway smooth muscle
breathlessness
All
erg t
Su
bje
ic

e.g. Histamine, prostaglandins,


c

leukotrienes  peptides Symptoms


AHR to Sensitization of
pharmacological airway smooth muscle
agents
Airway smooth muscle contraction  edema
Amplification of Increased response to
normal FEV1 acute increase of
response to leukotrienes, prostaglandins, etc
Exercise-induced bronchoconstriction
exercise
Exercise-induced bronchoconstriction

FIG. 38.4 The events that lead to EIB in people with asthma and events proposed to contribute to the pathogenesis of EIB and airway hyperresponsiveness
in people exercising strenuously. Reproduced from Ref. [34].

498
Exercise as a Stimulus 38
exercise when ventilation falls and the airways re-warm rap- narrowing occurs as a result of the contraction of bronchial
idly [43]. The mechanical events of vascular engorgement, smooth muscle by mediators [31]. The same mediators may
amplied by edema, serve to narrow the airways. increase vascular permeability and exudation of plasma, an
The osmotic hypothesis of EIB proposes that a tran- event that would serve to normalize osmolarity.
sient increase in osmolarity of the airway surface liquid Healthy subjects have the same burden to condition
occurs in response to evaporative water loss, and that the the air inspired during exercise as asthmatics. That water
cooler and drier the air inspired the greater the surface area replacement to the airway surface is compromised at a high
that will become dehydrated and hyperosmotic [31, 44]. It ventilation (at least for the rst 56 min) in both healthy
is the hyperosmolarity of the airway surface liquid and the and asthmatic subjects has been demonstrated by an acute
underlying mucosa that provides the appropriate microenvi- reduction in mucociliary clearance during hyperpnea of dry,
ronment for release of mediators [45, 46]. In contrast to the but not warm humid air ([47], Fig. 38.5).
thermal hypothesis, the osmotic hypothesis proposes airway The dierence in the lung function response to exercise
between healthy subjects and asthmatic subjects is likely due
60 Right peripheral region
to the bronchial smooth muscle of asthmatics being hyper-
responsive and having a greater abundance of mast cells and
Baseline their mediators close by [48]. Thus treatment that reduces the
Mean mucociliary clearance (%)

50 Dry eucapnic voluntary hyperpnea


source (e.g. ICS), or production (sh oil diet), of mediators
WH eucapnic voluntary hyperpnea
or their release (2-agonists, SCG, nedocromil sodium), or
40 their contractile action on smooth muscle (2-agonists, leu-
kotriene antagonists, histamine antagonists), are eective in
30 inhibiting or preventing EIB [22-28] (Fig. 38.6).

20

10 OPTIMIZING PROTOCOLS TO IDENTIFY EIB

0 The benet of optimizing an exercise protocol as a pro-


Asthmatic Healthy vocative stimulus for inducing an attack of asthma is that it
n 10 n 8 provides a simple and inexpensive method to monitor treat-
ment of asthma and can be utilized by both the patient and
FIG. 38.5 Mean values for the percentage (%) mucociliary clearance of
a radionuclide from the airways at baseline and following 6 min of
their doctor [49].
hyperpnea in asthmatic and healthy subjects. Hyperpnea with dry air but An exercise load that increases heart rate to 95% pre-
not humid air caused a significant reduction in mean mucociliary clearance dicted maximum [32], and ventilation to 17.5 FEV1 in
in the asthmatics suggesting the loss of water from the airway surface l/min within a few minutes, and one that can be sustained
exceeded the rate of replacement and dehydration occurred. Redrawn for at least 68 min whilst breathing dry air, is more likely
data from Ref. [47]. to provoke EIB than protocols using a lower intensity of

Mast cell as the source of mediators


Evaporative water loss increases
osmolarity of the microenvironment

Rx with steroids
Reduces mast cell number
2 agonists inhibit
and source of mediators
Release of PGD2 and
histamine via 2 receptors
Rx with Omega 3 rich diet
Reduces production of mediators Eosinophils
PGD2 LTC4 LT antagonists
Histamine antagonists
Sodium cromoglycate Histamine and 2 agonists protect
Inhibits PGD2 release against effect of mediator

Bronchial smooth muscle

FIG. 38.6 Schematic diagram of the mast cell showing the stimulus for the release of mediators such as prostaglandin D2 (PGD2), histamine and LTC4 and
the mode of action of various pharmaceutical agents in preventing or inhibiting EIB.

499
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 38.1 Challenge tests that can be used as a surrogate for exercise might be to increase the rate of return of water to the airway
to identify EIB. surface, probably by increasing bronchial blood ow. In this
way the osmotic threshold for mediator release from mast
The surrogate stimuli (eucapnic voluntary hyperpnea with dry air,
cells may not be reached either during warm up exercise or
hyperosmolar aerosols of salt or mannitol) are easy to perform.
later when an optimal exercise stimulus is used. The capacity
These stimuli can be easily repeated for monitoring the response to of the epithelium to produce and release protective mediators
treatment. may also be related to the extent to which the epithelium is
These stimuli are more potent than exercise so false negative results injured in response to the hyperpnea of dry air [66]. It is also
are less likely. likely that the increase in tidal volume causes stretch on the
Some tests (e.g. mannitol) are portable and provide a common bronchial smooth muscle reducing the latch state and mak-
operating standard and can be performed at the point of care. ing it less responsive to a subsequent stimulus [13].
Less expensive in manpower than field testing.

exercise for a longer duration [50]. There are, however, dif-


culties in achieving optimal conditions to provoke EIB, INFLAMMATORY MARKERS AND EXERCISE
particularly in t young adults with normal lung function. AND EIB
For this reason many laboratories have abandoned exercise
in preference to using EVH or other surrogates to identify
EIB (Table 38.1). Exercise can enhance transcription of two genes coding for
The protocol for EVH requires the subject to ventilate 5-lipoxygenase (ALOX5) and ALOX5-activating pro-
dry air containing 5% CO2 for 6 min [51]. An untrained tein leading to increased levels of LTB4 and leukotriene C4
person can easily hyperventilate at 21 FEV1 l/min for (LTC4) in plasma [67]. Leukotrienes sustain bronchocon-
6 min without respiratory muscle fatigue and a trained striction following exercise in asthmatics [68] and they may
subject can usually achieve and maintain a ventilation of contribute to EIB in elite athletes [34]. A mast cell-derived
30 FEV1 l/min [52]. mediator of importance in EIB is prostaglandin D2 (PGD2),
Other surrogates used to identify EIB include inha- usually reported as its metabolite 9, 11-PGF2 [69].
lation of aerosols of 4.5% saline or dry powder mannitol Histamine is a less potent mediator of EIB, but unlike the
that increase osmolarity and cause release of mediators PGs and LTs it is preformed in mast cells and thus is likely
[5356]. Some patients with COPD have been shown to to contribute to the reduction in lung function that occurs
have asthmatic responses to hyperosmolar agents [57, 58] immediately after exercise [9, 70]. It is however, the LTs and
even though they occasionally asthmatic responses to dry PGDs that sustain the fall in lung function after exercise and
air challenge [59]. this is the reason that antihistamines are not useful as a single
therapy for controlling EIB in most asthmatics [71].
The role of neuropeptides in EIB has not been well stud-
ied although the data are supportive and uctuations in osmo-
larity of the airway surface could stimulate their release [72].
REFRACTORINESS TO THE BRONCHO- Recent data suggest an association between increase in neu-
CONSTRICTING EFFECTS OF EXERCISE rokinin A levels in induced sputum and cysteinyl LTs in spu-
tum following exercise in subjects with EIB [73].
Eosinophils are a potential source of LTs and increased
Protocols that use a progressive increase in workload (e.g. numbers in sputum [74] and blood [75] are associated with
10 or 20 W increments each minute) to maximum work- presence of EIB independently of the presence of atopy.
ing capacity are inappropriate to identify EIB likely due to An acute increase in the percentage of eosinophils has been
refractoriness developing at the lower workloads. This was reported after exercise [76, 77].
recognized many years ago when these protocols became The percentage of columnar epithelial cells in sputum
popular in laboratories for assessing both heart disease and relates to severity of EIB [66], and the concentration of
lung disease in children and adults. Similarly submaxi- these cells is higher in asthmatic subjects with EIB com-
mal exercise for 20 min [60], or multiple warm up sprints pared with asthmatics without EIB [78].
of 30 s duration [61], induce refractoriness to an optimal The severity of EIB is related to the degree of atopy.
stimulus. Even when an optimal stimulus is used to provoke This is unsurprising in that atopy is an indirect marker of
EIB, 50% of subjects become refractory for about 2 h [62]. IgE on mast cells and it is the mediators from these cells
Refractoriness also develops to the eects of EVH when that are implicated in EIB. The presence of IgE on mast
ventilation is increased progressively [63] so single stage cells renders them less stable to hyperosmotic stress [45].
protocols are recommended [51]. The severity of EIB is also related to concentration of
Prostaglandins and leukotrienes (LTs) are thought to be expired nitric oxide (eNO) although this appears to be true only
important in protecting the airways and for inducing a state for atopic subjects and thus may be an epiphenomenon [79].
of refractoriness [64]. The evidence for this is the absence of A value for eNO 25 ppb has a negative predictive value of
refractoriness following treatment for a few days with clini- 100% for EIB but a positive predictive value of only 28% [80].
cally recommended doses of non-steroidal anti-inammatory Severity of EIB is related to markers of vascular permeability
agents, such as indomethacin [65]. The role of these mediators and vascular endothelial growth factor [81].

500
Exercise as a Stimulus 38
EXERCISE AND THE PERSON WITH COPD 2.5

2.0
COPD is characterized by airway inammation and air- Exercise
ow limitation that is not fully reversible. Despite optimal 1.5
medical management, people with COPD have signicant Rest
abnormalities in exercise capacity and ability to participate 1.0

Flow (l/s)
in everyday life. Often, the presenting symptom is breath-
lessness on exertion. Breathlessness during activity is evi- 0.5
dent in everyday life and varies with disease severity, with
those with mild disease only experiencing breathlessness 0.0
at high levels of exertion, whereas those with moderate to
severe COPD becoming breathless during everyday tasks. In 0.5
terms of the World Health Organization descriptors [82],
the impairments (i.e. the physiological and anatomical 1.0
changes) resulting from COPD lead to activity limitation
(inability to perform daily tasks or activities), and partici- 1.5
pation restriction (inability to contribute fully in societal 0 0.5 1.0 1.5 2.0 2.5
roles such as work and recreation). Volume (l)
The impairments that lead to exercise intolerance and
activity limitation are related to the eects of COPD on the IC
Exercise
ventilatory, cardiovascular, and muscular systems. IC
Rest

FIG. 38.7 Tidal flow-volume loops at rest and at peak exercise in COPD.
Ventilatory limitations to exercise Dynamic hyperinflation is evident by the shift of the exercise flow-volume
loop towards TLC. The reduction in IC indicates an increase in EELV.
Flow limitation and altered mechanics
of breathing
COPD  exercise
The increased airway resistance and expiratory ow limita-
tion that occurs in COPD results in hyperination at rest.
This alters the lengthtension relationship of the respiratory
muscles (i.e. muscles are in a more shortened position) which Dynamic
O2 Cost breathing
hyperinflation
places them at a mechanical disadvantage. During exercise,
ventilation increases (via increases in tidal volume and inspira-
tory and expiratory ow rates) to meet the metabolic demands.
The need for increased ow rates in the presence of expiratory High VD /VT V/Q mismatch
PpI/VT V CO2
ow limitation results in further hyperination during exercise
(i.e. dynamic hyperination) which is measured as a decrease
in IC, the corollary of which is an increase in end-expiratory VE /V CO2 O2 desaturation
lung volume EELV (Fig. 38.7). Such dynamic hyperination
further impacts on respiratory muscle lengthtension relation-
ships as well as limiting the tidal volume response to exercise,
resulting in a greater respiratory eort to achieve a given tidal Respiratory effort
volume (VT) (Fig. 38.8).
At low levels of exercise, dynamic hyperination
may enable higher ow rates and greater tidal volume than
Dyspnea
would have been possible without dynamic hyperination, and
allowing exercise to continue. With increasing intensity exercise limitation
of exercise the inability to further increase ventilation due
to expiratory ow constraints becomes a limiting factor. A
FIG. 38.8 Ventilatory factors leading to dyspnea and exercise limitation
measure of this ventilatory limitation is the ratio of peak in COPD. Ppl: pleural pressure as a measure of inspiratory effort; V T: tidal
exercise ventilation to maximum voluntary ventilation (VE/ volume; VCO2: carbon dioxide production; O2: oxygen; V/Q: ventilation/
MVV). A VE/MVV ratio above 0.9, while heart rate and perfusion; VE: ventilation; VD: dead space.
other physiological functions are below maximal capac-
ity, suggests severe ventilatory constraints to exercise [83].
However, since the estimate of MVV is usually obtained with severe hyperination. Arm elevation reduces the ability
from resting measures this approach to the determination of the accessory muscles to contribute to breathing due to
of ventilatory limitation may be imprecise. involvement in the arm task [84]. This shifts more of the
Altered mechanics of breathing in COPD may also respiratory work to the diaphragm which is already short-
impact on the capacity for arm exercise, especially in those ened due to hyperination, and thus is less eective in

501
Asthma and COPD: Basic Mechanisms and Clinical Management

responding to the added respiratory load. Dynamic hyper- hypoxia may contribute to skeletal muscle dysfunction [93].
ination during arm exercise [8587] and a higher EELV The physiological abnormalities evident in skeletal muscle
with arms elevated [86] may also contribute to exercise lim- of people with COPD are reduced oxidative enzyme capac-
itation, although this has not been examined in detail. ity [94], reduced muscle ber capillarization [95], atrophy
of Type I [96] and Type IIa muscle bers. Combined, these
Increased work of breathing abnormalities result in lactic acidosis at lower levels of exer-
Compared to the healthy population, respiratory muscle work cise and a reduced exercise endurance capacity compared to
during exercise is greater in people with COPD to overcome the healthy people [97, 98]. Muscle mass and strength are also
increased airway resistance. In addition, due to dynamic hyper- reduced in people with COPD. Importantly, some of these
ination, elastic work increases as tidal volume (VT) nears total abnormalities can be partially reversed by exercise training.
lung capacity (TLC). It has been shown that the oxygen con- Interestingly, skeletal muscle dysfunction is less evi-
sumption of the respiratory system in COPD during exercise dent in arm muscles compared to leg muscles in COPD [99]
may be 3540% of total body oxygen consumption (VO2) [88]. and therefore limitation of upper limb exercise capacity
Such a large oxygen demand by the respiratory muscles may is more likely due to alterations in mechanics of breath-
lead to a higher carbon dioxide production (VCO2) for a given ing, described previously, than to skeletal muscle dysfunc-
level of lower limb exercise, further increasing ventilation, as tion. Maintenance of arm skeletal muscle function has
well as possible diversion of blood ow from the lower limb been attributed to the requirements to continue performing
muscles to meet respiratory muscle requirements. Indirect evi- upper limb tasks for daily functional activities, while lower
dence for blood ow diversion comes from studies in healthy limb activity can be more easily reduced, leading to decon-
subjects [89] and in COPD [90]. dition. However, further research is required to validate this.

Gas exchange abnormalities


Gas exchange abnormalities arise from alveolar and capil- INTERVENTIONS TO IMPROVE EXERCISE
lary bed destruction leading to increased physiological dead CAPACITY IN COPD
space (i.e. a high dead space to tidal volume ratio (VD/VT)
and ventilationperfusion (V/Q) mismatch. The high VD/VT
means that there is an increased requirement for ventilation Interventions that can either decrease ventilatory demand
for a given level of VCO2, while the V/Q mismatch results in or that can increase ventilatory capacity may improve the
oxygen desaturation which may further increase ventilatory ability of people with COPD to exercise.
demand [83]. An added consequence of hypoxia is increased
ventilatory drive resulting from early onset of lactic acidosis. Interventions that reduce ventilatory
The above anatomical and physiological changes (i.e.
the impairments) in the ventilatory system that result in demand
dyspnea and exercise limitation are depicted in Fig. 38.8.
Exercise training
Lower limb endurance exercise training is eective in improv-
Cardiovascular constraints to exercise ing exercise capacity and quality of life and reducing exertional
dyspnea in people with COPD [100], largely due to physi-
Besides ventilatory constraints to exercise, people with ological changes that decrease ventilatory demand at a given
COPD commonly have coexisting cardiovascular constraints. level of exercise. These physiological changes include increases
Cardiac output may be reduced due to a reduction in right in muscle ber capillarization [101], mitochondrial density,
ventricular stroke volume related to (1) increased pulmonary and muscle oxidative/metabolic capacity [102104]. Such
vascular resistance from hypoxic vasoconstriction and/or loss training adaptations improve the aerobic capacity of the mus-
of capillary bed and vascular restructuring; (2) a reduction cle, delaying the onset of lactic acidosis and, as such, reduce
in right heart pre-load from decreased venous return conse- the ventilatory requirements for exercise at equivalent pre-
quent to hyperination [91]. Such reductions in cardiac out- training work rates [92]. This is reected in the ability of peo-
put may limit blood supply, and thus oxygen availability, to ple with COPD to perform equivalent work rates for longer
the exercising muscles, with consequent early onset of lactic after training as well as achieving higher peak work rates [92]
acidosis [92] adding to the constraints to exercise in COPD. and translates into a reduction in activity limitation and
participation restriction and improved quality of life.
The endurance exercise prescription in terms of the
Skeletal muscle dysfunction and exercise mode of training (cycle, treadmill, etc.) the intensity (high
or low), duration of a training session, frequency of train-
Skeletal muscle dysfunction in COPD may occur as a con- ing, and the length of the program has generated a great
sequence of deconditioning related to inactivity caused by deal of research, much of which is outlined in the major
exertional dyspnea. In approximately 40% of patients with recent guidelines of on Pulmonary Rehabilitation [105
COPD the main symptom limiting exercise is leg fatigue. 109] and Chapter 58. Information on the practical applica-
Besides deconditioning, other factors such as malnutrition, tion of exercise training in COPD patients can be accessed
skeletal muscle myopathy, low levels of anabolic hormones, through the Pulmonary Rehabilitation Toolkit (http://www.
systemic inammation, corticosteroid use, aging, and pulmonaryrehab.com.au).

502
Exercise as a Stimulus 38
It should be noted that while exercise training is con- [119, 120]. However, the gains in endurance capacity from
sidered the key component to achieve changes in exercise strength training were small compared to those that could be
capacity and quality of life, exercise training often occurs as elicited from endurance training [119, 120].
part of a comprehensive pulmonary rehabilitation program
which includes education, anxiety and dyspnea manage-
ment, smoking cessation support, and nutritional advice. Interventions that increase ventilatory
These additional components of pulmonary rehabilitation capacity
may enhance the outcomes of training regimen.
In addition to lower limb training, endurance training Bronchodilator therapy
of the upper limb using either supported arm exercise (arm
cranking) or unsupported arm exercise (free weights) has In general, lung function is poorly correlated with exer-
been shown to reduce ventilatory demand and improve arm cise capacity in COPD. However, as one of the main
exercise capacity [110, 111]. Such improvements are task factors limiting exercise in COPD is expiratory ow limi-
specic [112], therefore exercise mimicking daily activities, tation, medications such as inhaled anticholinergic and 2-
which are mostly unsupported arm tasks, are recommended sympathomimetic agents, that reduce airway resistance,
for inclusion in exercise training programs [108, 109]. may improve exercise capacity in COPD. Bronchodilator
medication delivered prior to exercise has been shown to
decrease resting hyperination and dynamic hyperination
Supplemental oxygen during exercise, which correlated strongly with improved
During exercise gas exchange abnormalities result is oxygen exercise capacity in severe COPD despite minimal change
desaturation in COPD patients with severe disease, with in spirometry [121]. A recent study of bronchodilator
consequent increases in ventilation (through earlier pro- therapy prior to exercise combined with supplemental oxy-
duction of lactate) and higher dyspnea levels [83]. Acute gen during exercise has shown an additive eect on exer-
oxygen supplementation during exercise in COPD reduces cise endurance, due to the combined physiological eects of
ventilatory demand for an equivalent work rate as well as reduced hyperination and reduced ventilatory drive [122].
reducing EELV [113] and exercise-induced pulmonary
hypertension [114], and improving leg blood ow [90]. All Non-invasive ventilation
these factors contribute to an increased exercise capacity
and reduced dyspnea. However, no signicant relationship In people with severe COPD, positive pressure non-invasive
has been found between exercise desaturation on room air ventilation reduces respiratory muscle load and prolongs the
and improvement in exercise capacity with supplemental tolerance to lactic acid accumulation resulting in a reduc-
oxygen [114, 115], suggesting that responses to oxygen sup- tion in dyspnea and improved exercise duration [123].
plementation need to be assessed on an individual basis. Exercise training using non-invasive ventilation in such
The eect of oxygen supplementation during exercise patient groups, especially those with a ventilatory limita-
training in COPD subjects who desaturate during exercise has tion to exercise, enabled higher training intensities to be
been equivocal, with most studies showing no greater improve- achieved resulting in a reduction in lactate at equivalent
ment in exercise capacity compared with exercise training on workloads and greater improvements in exercise capacity in
room air [109]. However, one study of oxygen supplementa- the intervention group [124126]. The clinical signicance
tion during exercise training in nonhypoxaemic COPD sub- of this form of intervention in terms of improvements in
jects, demonstrated signicantly greater improvements in functional capacity and quality of life, as well as the cost-
endurance exercise time in the oxygen trained group, most eectiveness has yet to be determined.
likely related to the higher training intensities achieved [116].
Further studies are required to validate the eectiveness of Lung volume reduction surgery
similar exercise training protocols using supplemental oxygen Lung volume reduction surgery (LVRS), the removal of
in COPD subjects who desaturate during exercise. areas of emphysematous lung tissue, increases static lung
recoil, thus reducing ow limitation and dynamic hyper-
Strength training ination [127]. Combined with post-operative exercise
Although strength training does not results in reduced venti- training, LVRS improved exercise capacity for both lower
latory demand per se it had been included in this section, since limbs [128] and upper limbs [129]. The improvements
the short bouts of this form of exercise induce lower ventila- in exercise capacity were additional to those gained from
tory demand and dyspnea compared to endurance training pre-surgery exercise training. LVRS is mostly eective in
and are usually well tolerated by people with COPD [117]. selected patients with upper-lobe emphysema and persist-
Reductions in skeletal muscle strength are evident in ent low exercise capacity after an exercise training program.
people with COPD and may aect their ability to perform
functional activities as well as increase the risk of falls; how- Body positioning
ever, the latter has not been specically studied in the COPD The lean forward position with shoulder girdle xation
population. Whether improvements in muscle strength is often adopted by people with COPD when breathless.
gained by resistance training programs [118] translate into Leaning forward has been shown to reduce accessory mus-
improved exercise capacity is not clear. Signicant improve- cle activity, increase transdiaphragmatic pressure, increase
ments in cycle endurance capacity were demonstrated in maximum inspiratory pressures, and reduce dyspnea at rest in
studies comparing strength training with no intervention COPD subjects with severe disease [130, 131], presumably by

503
Asthma and COPD: Basic Mechanisms and Clinical Management

increased abdominal pressure improving the lengthtension 7. Anderson SD et al. Metabolic and ventilatory changes in asthmatic
relationship of the diaphragm in people who are hyperin- patients during and after exercise. Thorax 27: 71825, 1972.
ated. Shoulder girdle xation allows the accessory muscles 8. Bye PTP et al. Plasma cyclic AMP levels in response to exercise and
terbutaline sulphate aerosol in normal and asthmatic patients. Eur J
to function with reverse origin and insertion (i.e. rather than
Respir Dis 61: 28797, 1980.
acting to move the arm on a xed torso these muscles can act 9. Haverkamp HC et al. Gas exchange during exercise in habitually active
to move the ribs in an inspiratory direction). Shoulder girdle asthmatic subjects. J Appl Physiol 99: 193850, 2005.
xation has been shown to improve MVV, potentially reduc- 10. Beck KC et al. Bronchoconstriction occurring during exercise in
ing the ventilatory constraint to exercise [132]. Some evidence asthmatic subjects. Am J Respir Crit Care Med 149: 35257, 1994.
that the lean forward position with shoulder xation can 11. Suman OE et al. Airway function after cyclooxygenase inhibition
enhance exercise capacity in people with COPD comes from during hyperpnea-induced bronchoconstriction in guinea pigs. J Appl
studies of rollator walking which have shown improved func- Physiol 89: 197178, 2000.
tional exercise capacity and a reduction in dyspnea [132, 133]. 12. Suman OE, Beck KC. Role of airway endogenous nitric oxide on lung
function during and after exercise in mild asthma. J Appl Physiol 93:
Breathing strategies 193238, 2002.
13. Fredberg JJ et al. Airway smooth muscle, tidal stretches, and dynami-
Pursed-lip breathing is another strategy adopted spontane- cally determined contractile states. Am J Respir Crit Care Med 156:
ously by people with severe COPD to prolong active expi- 175259, 1997.
ration and limit airway collapse. Pursed-lip breathing at 14. Silverman M et al. Metabolic changes preceding exercise-induced
rest has been shown to reduce EELV [134] which could bronchoconstriction. BMJ 1: 2079, 1972.
potentially increase ventilatory capacity. However, volitional 15. Munoz P. Pulmonary gas exchange response to exercise and mannitol
pursed-lip breathing during exercise has not been shown to challenges in exercise-induced asthma patients. Am J Respir Crit Care
increase exercise capacity in people with COPD [135]. The Med ASM 3(15): A721, 2006.
16. Young IH et al. Pattern and time course of ventilation-perfusion ine-
variable response to volitional pursed-lip breathing during
quality in exercise-induced asthma. Am Rev Respir Dis 125: 30411,
exercise suggests that benet may only occur in those who 1982.
spontaneously adopt pursed-lip breathing. 17. Becker JM et al. Asthma deaths during sports: Report of a 7-year expe-
rience. J Allergy Clin Immunol 113: 26467, 2004.
Respiratory muscle training 18. Anderson SD et al. Responses to bronchial challenge submitted
Respiratory muscle weakness is evident in a subgroup for approval to use inhaled 2 agonists prior to an event at the 2002
of people with COPD. The impact of inspiratory mus- Winter Olympics. J Allergy Clin Immunol 111: 4449, 2003.
cle weakness on exercise capacity is unclear and may only 19. Ram FSF et al. Physical training for asthma. Cochrane Database Syst
Rev 19: 2005, CD001116.
be important if such weakness reduces ventilatory capac-
20. Thio BJ et al. Exercise-induced asthma and cardiovascular tness in
ity such that it is a factor limiting exercise. Improvements asthmatic children. Thorax 51: 2079, 1996.
in exercise capacity gained with inspiratory muscle train- 21. Anderson SD et al. Bronchial challenges in athletes applying to inhale
ing are signicantly less than those gained with lower limb a 2-agonist at the 2004 Summer Olympics. J Allergy Clin Immunol
endurance training [136]. However, a number of studies 117: 76773, 2006.
have shown that the addition of inspiratory muscle train- 22. Anderson SD. Single dose agents in the prevention of exercise-induced
ing to a lower limb training program improved functional asthma: A descriptive review. Treat Respir Med 3: 36579, 2004.
exercise capacity above that gained from lower limb train- 23. Anderson SD et al. 2-Agonists and exercise-induced asthma. Clin Rev
ing alone [137, 138]. Inspiratory muscle training for people Allergy Immunol 31: 16380, 2006.
with COPD may only be of benet in those with suspected 24. Storms W et al. A comparison of the eects of oral montelukast and
inhaled salmeterol on response to rescue bronchodilation after chal-
or proven respiratory muscle weakness [108] and current
lenge. Respir Med 98: 105162, 2004.
guidelines do not support routine inclusion of this type of 25. Subbarao P et al. Eect of ciclesonide dose and duration of therapy on
training for people with COPD [109]. exercise-induced bronchoconstriction in patients with asthma. J Allergy
Clin Immunol 117: 100813, 2006.
26. Weiler JM et al. Eect of uticasone/salmeterol administered via a sin-
References gle device on exercise-induced bronchospasm in patients with persist-
ent asthma. Ann Allergy Asthma Immunol 94: 6572, 2005.
1. Welsh L et al. Fitness and physical activity in children with asthma. 27. Mickleborough T, Gotshall R. Dietary components with demonstrated
Sports Med 34: 86170, 2004. eectiveness in decreasing the severity of exercise-induced asthma.
2. Hallstrand TS et al. Aerobic conditioning in mild asthma decreases the Sports Med 33: 67181, 2003.
hyperpnea of exercise and improves exercise and ventilatory capacity. 28. Mickleborough TD et al. Protective eect of sh oil supplementation on
Chest 118: 146069, 2000. exercise-induced bronchoconstriction in asthma. Chest 129: 3949, 2006.
3. Alioglu B et al. Cardiopulmonary responses of asthmatic children to 29. Hallstrand TS et al. Eectiveness of screening examinations to detect
exercise: Analysis of systolic and diastolic cardiac function. Pediatr unrecognised exercise-induced bronchoconstriction. J Pediatr 141:
Pulmonol 42: 28389, 2007. 34349, 2002.
4. Varray A et al. Cardiac role in exercise limitation in asthmatic subjects 30. Rundell KW et al. Self-reported symptoms and exercise-induced
with special reference to disease severity. Eur Respir J 6: 101117, 1993. asthma in the elite athlete. Med Sci Sports Exerc 33: 20813, 2001.
5. Lucas SR, Platts-Mills TA. Physical activity and exercise in asthma: 31. Anderson SD, Daviskas E. The mechanism of exercise-induced asthma
Relevance to etiology and treatment. J Allergy Clin Immunol 115: is . J Allergy Clin Immunol 106: 45359, 2000.
92834, 2005. 32. Carlsen KH et al. Exercise induced bronchoconstriction depends on
6. Joyner BL et al. Cardiopulmonary exercise testing in children and exercise load. Respir Med 94: 75055, 2000.
adolescents with asthma who report symptoms of exercise-induced 33. Anderson SD, Holzer K. Exercise-induced asthma: Is it the right diag-
bronchoconstriction. J Asthma 43: 67578, 2006. nosis in elite athletes?. J Allergy Clin Immunol 106: 41928, 2000.

504
Exercise as a Stimulus 38
34. Anderson SD, Kippelen P. Exercise-induced bronchoconstriction: 62. Anderson SD, Schoeel RE. Respiratory heat and water loss during
Pathogenesis. Curr Allergy Asthma Rep 5: 11622, 2005. exercise in patients with asthma: Eect of repeated exercise challenge.
35. Erjefalt JS et al. Epithelial pathways for luminal entry of bulk plasma. Eur J Respir Dis 63: 47280, 1982.
Clin Exp Allergy 25: 18795, 1995. 63. Argyros GJ et al. The refractory period after eucapnic voluntary hyper-
36. Johnson PR, Burgess JK. Airway smooth muscle and broblasts in the ventilation challenge and its eect on challenge technique. Chest 108:
pathogenesis of asthma. Curr Allergy Asthma Rep 4: 1028, 2004. 41924, 1995.
37. Schmidt D et al. Passive sensitization of human airway increases 64. Manning PJ et al. Exercise-induced refractoriness in asthmatic subjects
responsiveness to leukotriene C4. Eur Respir J 14: 31519, 1999. involves leukotriene and prostaglandin interdependent mechanisms.
38. Carroll NG et al. Distribution and degranulation of airway mast cells Am Rev Respir Dis 148: 95054, 1993.
in normal and asthmatic subjects. Eur Respir J 19: 87985, 2002. 65. Wilson BA et al. The eects of indomethacin on refractoriness follow-
39. Silverman M, Anderson SD. Standardization of exercise tests in asth- ing exercise both with and without bronchoconstriction. Eur Respir J
matic children. Arch Dis Child 47: 88289, 1972. 12: 217478, 1994.
40. Anderson SD et al. Sensitivity to heat and water loss at rest and during 66. Hallstrand TS et al. Inammatory basis of exercise-induced bronchoc-
exercise in asthmatic patients. Eur J Respir Dis 63: 45971, 1982. onstriction. Am J Respir Crit Care Med 172: 67986, 2005.
41. Rundell KW et al. Field exercise vs laboratory eucapnic voluntary 67. Hilberg T et al. Transcription in response to physical stress-clues to
hyperventilation to identify airway hyperresponsiveness in elite cold the molecular mechanisms of exercise-induced asthma. FASEB J 19:
weather athletes. Chest 125: 90915, 2004. 149294, 2005.
42. Anderson SD, Daviskas E. The airway microvasculature and exercise- 68. Reiss TF et al. Increased urinary excretion of LTE4 after exercise
induced asthma. Thorax 47: 74852, 1992. and attenuation of exercise-induced bronchospasm by montelukast,
43. McFadden ER et al. Postexertional airway rewarming and thermally a cysteinyl leukotriene receptor antagonist. Thorax 52: 103035, 1997.
induced asthma. J Clin Invest 78: 1825, 1986. 69. OSullivan S et al. Evidence for mast cell activation during exercise-
44. Anderson SD. Is there a unifying hypothesis for exercise-induced induced bronchoconstriction. Eur Respir J 12: 34550, 1998.
asthma? J Allergy Clin Immunol 73: 66065, 1984. 70. Anderson SD et al. Arterial plasma histamine levels at rest, during and
45. Gulliksson M et al. Release of prostaglandin D2 and leukotriene C after exercise in patients with asthma: Eects of terbutaline aerosol.
in response to hyperosmolar stimulation of mast cells. Allergy 61: Thorax 36: 25967, 1981.
147379, 2006. 71. Dahln B et al. Inuence of zarlukast and loratadine on exercise-
46. Moloney ED et al. Release of inammatory mediators from eosinophils induced bronchoconstriction. J Allergy Clin Immunol 109: 78993, 2002.
following a hyperosmolar stimulus. Respir Med 97: 15, 2003. 72. Ichinose M et al. A neurokinin 1-receptor antagonist improves exercise-
47. Daviskas E et al. Changes in mucociliary clearance during and after iso- induced airway narrowing in asthmatic patients. Am J Respir Crit Care
capnic hyperventilation in asthmatic and healthy subjects. Eur Respir J Med 153: 93641, 1996.
8: 74251, 1995. 73. Hallstrand TS et al. Role of MUC5AC in the pathogenesis of exercise-
48. Brightling CE et al. Mast-cell inltration of airway smooth muscle in induced bronchoconstriction. J Allergy Clin Immunol, 119: 1092-1098, 2007.
asthma. N Engl J Med 346: 1699705, 2002. 74. Yoshikawa T et al. Severity of exercise-induced bronchoconstriction is
49. Haby MM et al. An exercise challenge for epidemiological studies of related to airway eosinophilic inammation in patients with asthma.
childhood asthma: Validity and repeatability. Eur Respir J 8: 72936, Eur Respir J 12: 87984, 1998.
1995. 75. Lee SY et al. Eosinophils play a major role in the severity of exercise-
50. Anderson SD et al. Laboratory protocol for exercise asthma to evaluate induced bronchoconstriction in children with asthma. Pediatr Pulmonol
salbutamol given by two devices. Med Sci Sports Exerc 33: 893900, 2001. 41: 116166, 2006.
51. Anderson SD et al. Provocation by eucapnic voluntary hyperpnoea 76. Haverkamp HC et al. Repeat exercise normalizes the gas-exchange
to identify exercise induced bronchoconstriction. Br J Sports Med 35: impairment induced by a previous exercise bout in asthmatic subjects.
34447, 2001. J Appl Physiol 99: 184352, 2005.
52. Spiering BA et al. Standardized estimated ventilation for eucapnic volun- 77. Kivity S et al. Eosinophil inux into the airways in patients with exercise-
tary hyperventilation: An evaluation. Med Sci Sports Exerc 34: S52, 2002. induced asthma. Respir Med 94: 12005, 2000.
53. Brannan JD et al. Responsiveness to mannitol in asthmatic subjects 78. Hallstrand TS et al. Airway immunopathology of asthma with exercise-
with exercise- and hyperventilation-induced asthma. Am J Respir Crit induced bronchoconstriction. J Allergy Clin Immunol 116: 58693, 2005.
Care Med 158: 112026, 1998. 79. Franklin PJ et al. Measuring exhaled nitric oxide levels in adults: The
54. Brannan JD et al. Inhibition of mast cell PGD2 release protects against importance of atopy and airway responsiveness. Chest 126: 154045, 2004.
mannitol-induced airway narrowing. Eur Respir J 27: 94450, 2006. 80. Lex C et al. Value of surrogate tests to predict exercise-induced bronchoc-
55. Holzer K et al. Mannitol as a challenge test to identify exercise-induced onstriction in atopic childhood asthma. Pediatr Pulmonol 42: 22530, 2007.
bronchoconstriction in elite athletes. Am J Respir Crit Care Med 167: 81. Kanazawa H et al. Vascular involvement in exercise-induced airway
53447, 2003. narrowing in patients with bronchial asthma. Chest 122: 16670, 2002.
56. Smith CM, Anderson SD. Inhalational challenge using hypertonic 82. World Health Organization. Towards a Common Language for
saline in asthmatic subjects: A comparison with responses to hyperp- Functioning, Disability and Health. Geneva: ICF, 2002.
noea, methacholine and water. Eur Respir J 3: 14451, 1990. 83. ODonnell DE. Ventilatory limitations in chronic obstructive pulmo-
57. Leuppi JD et al. Prediction of treatment-response to inhaled corti- nary disease. Med Sci Sports Exerc 33: S647655, 2001.
costeroids by mannitol-challenge test in COPD. A proof of concept. 84. Epstein SK et al. Ventilatory response to arm elevation, its determi-
Pulm Pharmacol Ther 18: 8388, 2005. nants and use in patients with chronic obstructive pulmonary disease.
58. Taube C et al. Airway response to inhaled hypertonic saline in patients Am J Respir Crit Care Med 152: 21116, 1995.
with moderate to severe chronic obstructive pulmonary disease. Am 85. Gigliotti F et al. Arm exercise and hyperination in patients with
J Respir Crit Care Med 164: 181015, 2001. COPD: Eect of arm training. Chest 128: 122532, 2005.
59. Ramsdale EH et al. Bronchial responsiveness to methacholine in 86. McKeough ZJ et al. Arm positioning alters lung volumes in subjects
chronic bronchitis: Relationship to airow limitation and cold air with COPD and healthy subjects. Aust J Physiother 49: 13337, 2003.
responsiveness. Thorax 39: 91218, 1984. 87. McKeough ZJ et al. Arm exercise capacity and dyspnea ratings in sub-
60. Rei DB et al. The eect of prolonged submaximal warm-up exercise jects with chronic obstructive pulmonary disease. J Cardiopulm Rehabil
on exercise-induced asthma. Am Rev Respir Dis 139: 47984, 1989. 23: 21825, 2003.
61. Schnall RP, Landau LI. Protective eects of repeated short sprints in 88. Levison H, Cherniack RM. Ventilatory cost of exercise in chronic
exercise-induced asthma. Thorax 35: 82832, 1980. obstructive pulmonary disease. J Appl Physiol 25: 2127, 1968.

505
Asthma and COPD: Basic Mechanisms and Clinical Management

89. Harms CA et al. Respiratory muscle work compromises leg blood 114. Fujimoto K et al. Benets of oxygen on exercise performance and pul-
ow during maximal exercise. J Appl Physiol 82: 157383, 1997. monary hemodynamics in patients with COPD with mild hypoxemia.
90. Maltais F et al. Eects of oxygen on lower limb blood ow and O2 Chest 122: 45763, 2002.
uptake during exercise in COPD. Med Sci Sports Exerc 33: 91622, 2001. 115. Jolly EC et al. Eects of supplemental oxygen during activity in
91. Sietsema K. Cardiovascular limitations in chronic pulmonary disease. patients with advanced COPD without severe resting hypoxemia.
Med Sci Sports Exerc 33: S65661, 2001. Chest 120: 43743, 2001.
92. Casaburi R et al. Reductions in exercise lactic acidosis and ventilation 116. Emtner M et al. Benets of supplemental oxygen in exercise training
as a result of exercise training in patients with obstructive lung disease. in nonhypoxemic chronic obstructive pulmonary disease patients. Am
Am Rev Respir Dis 143: 918, 1991. J Respir Crit Care Med 168: 103442, 2003.
93. A statement of the American Thoracic Society and European 117. Probst VS et al. Cardiopulmonary stress during exercise training in
Respiratory Society. Skeletal muscle dysfunction in chronic obstruc- patients with COPD. Eur Respir J 27: 111018, 2006.
tive pulmonary disease. Am J Respir Crit Care Med 159: S140, 1999. 118. OShea SD et al. Peripheral muscle strength training in COPD: A
94. Maltais F et al. Oxidative capacity of the skeletal muscle and lactic systematic review. Chest 126: 90314, 2004.
acid kinetics during exercise in normal subjects and in patients with 119. Ortega F et al. Comparison of eects of strength and endurance
COPD. Am J Respir Crit Care Med 153: 28893, 1996. training in patients with chronic obstructive pulmonary disease. Am
95. Jobin J et al. Chronic obstructive pulmonary disease: Capillarity and J Respir Crit Care Med 166: 66974, 2002.
ber-type characteristics of skeletal muscle. J Cardiopulm Rehabil 18: 120. Spruit MA et al. Resistance versus endurance training in patients with
43237, 1998. COPD and peripheral muscle weakness. Eur Respir J 19: 107278, 2002.
96. Jakobsson P et al. Skeletal muscle metabolites and bre types in patients 121. ODonnell DE et al. Spirometric correlates of improvement in exer-
with advanced chronic obstructive pulmonary disease (COPD), with and cise performance after anticholinergic therapy in chronic obstructive
without chronic respiratory failure. Eur Respir J 3: 19296, 1990. pulmonary disease. Am J Respir Crit Care Med 160: 54249, 1999.
97. Casaburi R. Skeletal muscle dysfunction in chronic obstructive pul- 122. Peters MM et al. Combined physiological eects of bronchodilators
monary disease. Med Sci Sports Exerc 33: S66270, 2001. and hyperoxia on exertional dyspnoea in normoxic COPD. Thorax 61:
98. Polkey MI. Muscle metabolism and exercise tolerance in COPD. 55967, 2006.
Chest 121: 131S135S, 2002. 123. vant Hul A et al. The acute eects of noninvasive ventilatory sup-
99. Gea JG et al. Metabolic characteristics of the deltoid muscle in port during exercise on exercise endurance and dyspnea in patients
patients with chronic obstructive pulmonary disease. Eur Respir J 17: with chronic obstructive pulmonary disease: A systematic review.
93945, 2001. J Cardiopulm Rehabil 22: 29097, 2002.
100. Lacasse Y et al. Pulmonary rehabilitation for chronic obstructive pul- 124. Costes F et al. Noninvasive ventilation during exercise training
monary disease. Cochrane Database Syst Rev: CD003793, 2006. improves exercise tolerance in patients with chronic obstructive pul-
101. Whittom F et al. Histochemical and morphological characteristics of monary disease. J Cardiopulm Rehabil 23: 30713, 2003.
the vastus lateralis muscle in patients with chronic obstructive pulmo- 125. Hawkins P et al. Proportional assist ventilation as an aid to exercise
nary disease. Med Sci Sports Exerc 30: 146774, 1998. training in severe chronic obstructive pulmonary disease. Thorax 57:
102. Maltais F et al. Skeletal muscle adaptation to endurance training in 85359, 2002.
patients with chronic obstructive pulmonary disease. Am J Respir Crit 126. vant Hul A et al. Training with inspiratory pressure support in
Care Med 154: 44247, 1996. patients with severe COPD. Eur Respir J 27: 6572, 2006.
103. McKeough ZJ et al. Exercise capacity and quadriceps muscle metabo- 127. Martinez FJ et al. Lung-volume reduction improves dyspnea, dynamic
lism following training in subjects with COPD. Respir Med 100: hyperination, and respiratory muscle function. Am J Respir Crit Care
181725, 2006. Med 155: 198490, 1997.
104. Sala E et al. Eects of endurance training on skeletal muscle bioener- 128. Fishman A et al. A randomized trial comparing lung-volume-
getics in chronic obstructive pulmonary disease. Am J Respir Crit Care reduction surgery with medical therapy for severe emphysema. N Engl
Med 159: 172634, 1999. J Med 348: 205973, 2003.
105. ACCP/AACVPR Pulmonary Rehabilitation Guidelines Panel. 129. McKeough ZJ et al. Supported and usnsupported arm exercise capac-
Pulmonary rehabilitation: Joint ACCP/AACVPR evidence-based guide- ity following lung volume reduction surgery: A pilot study. Chron
lines. American College of Chest Physicians. American Association of Respir Dis 2: 5965, 2005.
Cardiovascular and Pulmonary Rehabilitation. Chest 112: 136396, 1997. 130. ONeill S, McCarthy DS. Postural relief of dyspnoea in severe chronic
106. American Thoracic Society. Pulmonary rehabilitation 1999. Am J airow limitation: Relationship to respiratory muscle strength. Thorax
Respir Crit Care Med 159: 166682, 1999. 38: 595600, 1983.
107. British Thoracic Society Standards of Care Subcommittee on Pulmonary 131. Sharp JT et al. Postural relief of dyspnea in severe chronic obstructive
Rehabilitation. Pulmonary rehabilitation. Thorax 56: 82734, 2001. pulmonary disease. Am Rev Respir Dis 122: 20111, 1980.
108. Nici L et al. American Thoracic Society/European Respiratory Society 132. Probst VS et al. Mechanisms of improvement in exercise capacity
statement on pulmonary rehabilitation. Am J Respir Crit Care Med using a rollator in patients with COPD. Chest 126: 11027, 2004.
173: 1390413, 2006. 133. Solway S et al. The short-term eect of a rollator on functional exercise
109. Ries AL et al. Pulmonary rehabilitation: Joint ACCP/AACVPR capacity among individuals with severe COPD. Chest 122: 5665, 2002.
evidence-based clinical practice guidelines. Chest 131: 4S42S, 2007. 134. Bianchi R et al. Chest wall kinematics and breathlessness during
110. Couser JI Jr. et al. Pulmonary rehabilitation that includes arm exercise pursed-lip breathing in patients with COPD. Chest 125: 45965, 2004.
reduces metabolic and ventilatory requirements for simple arm eleva- 135. Garrod R et al. An evaluation of the acute impact of pursed lips breath-
tion. Chest 103: 3741, 1993. ing on walking distance in nonspontaneous pursed lips breathing chronic
111. Epstein SK et al. Arm training reduces the VO2 and VE cost of obstructive pulmonary disease patients. Chron Respir Dis 2: 6772, 2005.
unsupported arm exercise and elevation in chronic obstructive pulmo- 136. Crowe J et al. Inspiratory muscle training compared with other rehabilita-
nary disease. J Cardiopulm Rehabil 17: 17177, 1997. tion interventions in adults with chronic obstructive pulmonary disease:
112. Martinez FJ et al. Supported arm exercise vs unsupported arm exercise A systematic literature review and meta-analysis. COPD 2: 31929, 2005.
in the rehabilitation of patients with severe chronic airow obstruc- 137. Dekhuijzen PN et al. Target-ow inspiratory muscle training during pul-
tion. Chest 103: 1397402, 1993. monary rehabilitation in patients with COPD. Chest 99: 12833, 1991.
113. Somfay A et al. Dose-response eect of oxygen on hyperination and 138. Wanke T et al. Eects of combined inspiratory muscle and cycle
exercise endurance in nonhypoxaemic COPD patients. Eur Respir J ergometer training on exercise performance in patients with COPD.
18: 7784, 2001. Eur Respir J 7: 220511, 1994.

506
Atmospheric Pollutants
39 CHAPTER

In the 1940s and 1950s, air pollution was a fact agencies (Table 39.1). These give an indication
of life in industrialized cities, but the true extent of the levels to which populations are currently
Jon Ayres
of its health risks was not appreciated until exposed, although levels exceeding these are Department of Environmental and
the London Fog Incident of 1952 [1], which commonplace in large cities. The time frame for Occupational Medicine, Liberty Safe
caused 4000 excess deaths largely from cardiac these standards varies between pollutants. For Work Research Centre, University of
disease, bronchitis, or pneumonia. At the time, example, the UK air quality standard for SO2 Aberdeen, Aberdeen, UK
it was believed that the combination of particles is 100 ppb over a 15-min sampling time frame,
and SO2 was the cause of such eects, but sub- because of its ability to cause acute bronchoc-
sequent reanalysis of these historical data sug- onstriction in asthma, whereas the standard for
gested that the acidity of the aerosol could have ozone is based on 8 h, being the timescale in
been the major contributor to mortality [2]. which ozone is formed during sunlight.
As the Clean Air Act of 1956 began to reduce
black smoke levels, day-to-day changes in pol-
lution had progressively less impact on symp- Sources of gaseous pollutants
toms in chronic bronchitis [3] and the advice
given to Government was that air pollution The main source of SO2 is the burning of fos-
would never again represent a risk to human sil fuel in coal-red power stations, domestic
health, even though studies of urban compared res, or industrial processes with a variable con-
with rural areas showed a higher prevalence of tribution from sulfur-containing engine fuels.
productive cough in urban areas [4]. However, NO2 is largely derived from vehicle emissions,
by the late 1970s such reassurance was being although the highest individual exposures are
questioned as vehicle-generated air pollution from gas appliances indoors. Ozone is a second-
was being shown to impact on human health. ary pollutant formed by the action of ultraviolet
light on components of vehicle exhaust.

CURRENT POLLUTANT EXPOSURES Sources of particulate pollutants


The main sources of particles are vehicle emis-
The air pollutants relevant to asthma and chronic sions. Particles are often measured by a reect-
obstructive pulmonary disease (COPD) are: ance method, black smoke (BS) which, while
sulfur dioxide (SO2) simple and cheap has been superceded by a
mass measure of particles either using a high
nitrogen dioxide (NO2) volume lter system or a TEOM (tapered ele-
ozone ment oscillating microbalance). This produces
particles measures such as PM10 (particles broadly
10 m in diameter) or PM2.5 (2.5 m in
Levels of these pollutants vary worldwide diameter), depending on the size of the air inlet.
and health-based air quality standards have There is some evidence that at least part of the
been produced by many individual countries or toxic fraction of particles resides in the ultrane

507
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 39.1 Air quality standards.

WHO (2005 revision) EC UK

Sulfur dioxide

10 min 500 g/m3


15 min 100 ppb (286 g/m3)
24 h 125 g/m3 (interim target 1) 20 g/m3 (AQS)
Annual Now not needed as adherence to the 24-h 48.8 ppb if smoke 60 g/m3
standard will control annual exposures
67.5 ppb if smoke 60 g/m3

Nitrogen dioxide

1h 200 g/m3 70.6 ppb (133 g/m3)a 150 ppb (282 g/m3)
Annual 40 g/m3 26.2 ppb (49 g/m3)b

Ozone

8 h moving average 100 g/m3 55 ppb (110 g/m3) 50 ppb (100 g/m3)

Particles

24-h mean PM10 50 g/m3 PM10 80 g/m3 PM10 50 g/m3 c


PM2.5 25 g/m3
Annual mean PM10 20 g/m3
PM2.5 10 g/m3

EC: European Community; UK: United Kingdom; WHO: World Health Organization.
a
98 percentile.
b
50 percentile.
c
Currently under revision to a PM2 5 standard.

fraction (100 nm). The relevance of the surface chemistry in the lung is now not held. Once penetrating to the distal
of particles in terms of health eects is now recognized and lung, some particles will deposit and will either be taken up
the need for a measure of particle surface area has arisen. by alveolar macrophages or become interstitialized.
This is dicult, but particle numbers can be used as a sur- Deposition of gases will depend on, amongst other
rogate measure, and there is some evidence that this metric things, gas solubility, minute ventilation, and the presence
relates to health eects. of airow obstruction. The endogenous antioxidant systems
(largely urate in the nose and glutathione nitrate/nitrite in
the lower respiratory tract) will, up to certain concentra-
Exposures tions, counteract the proinammatory eects of oxidant
gases (ozone and NO2) thus also altering the exposure of
Exposure of an individual to pollutants is only very crudely the epithelium to the inhaled gas.
estimated by levels obtained from sentinel monitoring sites,
which do not take into account indoor levels of pollutants and
factors, such as personal activity and preexisting lung disease,
which will aect the amount of a given pollutant inhaled.
Particles, once emitted, agglomerate to produce sec- ASSESSING THE HEALTH EFFECTS OF
ondary particles of varying size and shape and undergo AIR POLLUTANTS
chemical reactions depending on the emission source and
the atmosphere into which they are emitted. There are
marked dierences in the deposition of particles by size both Health effects of air pollution
regionally in the lung and with respect to the airway level at
which deposition occurs. Larger particles (10 m in diam- Exposure to air pollutants can result in:
eter) mostly impact in the upper airways with those in the acute, or day-to-day eects;
respirable range (7.2 m) penetrating deeper (Fig. 39.1).
The belief that ultrane particles (1 m) do not remain chronic eects;
in the lung and are simply breathed out again leaving none latent eects.

508
Atmospheric Pollutants 39
Macrophage overload
Interstitialization COPD Heart
Inflammation disease

Thrombogenesis

IgE-mediated responses
Asthma
Upper airway stimulation
100 Heart
Autonomic disturbance disease
Eosinophil/Neutrophil
recruitment Asthma and
COPD
Inflammation Extrathoracic
Deposition (%)

50

Bronchial
Alveolar
0
0.01 0.1 1.0 10.0
Particle diameter (m)

FIG. 39.1 Deposition of particles at various levels of the respiratory tract by particle size and likely mechanisms of health effect.

Acute eects occur in patients with preexisting dis- Animal studies can be used for the study of more
ease, while chronic eects may impact on patients who are prolonged exposures than is possible with man.
apparently disease-free prior to exposure. The major at-risk Experimental and animal studies both have the benet
groups for acute eects are patients with asthma or COPD, of being able to study specic, controlled exposures of
although both deaths and hospital admissions for patients single or combined pollutant exposures.
with cardiac disease increase on days of higher particulate Computer modeling can predict outcomes providing the
levels [5]. While the cardiac eects will not be covered here, specic exposure characteristics of the pollutant(s) and
the frequent coincidence of coronary heart disease and recognized coexposures (e.g. meteorological variables)
COPD means that patients with COPD may be aected and host factors (e.g. exercise) are known.
by air pollution more through their cardiac than their pul-
monary disease. Most health eects of air pollutants are respiratory
although in COPD these are limited to mortality and hos-
pital admissions as there are no contemporary panel stud-
ies of well-characterized patients with COPD and little
Methods of assessment of health effects human challenge evidence. In asthma, in contrast, there is
a substantial body of evidence ranging from hospital admis-
Epidemiological studies provided the rst evidence sions to challenge studies.
of and quantied, to an extent, the adverse health
eects of air pollution. Time series studies of routinely
collected data (mortality, hospital admissions) and
panel studies of putative susceptible groups (asthma, Responses to an air pollutant
COPD) have provided information across a range of
severities. Individual response to an air pollutant is aected by many
factors (Table 39.2). Where possible these need to be
Experimental studies in man have relied on controlled allowed for when assessing the eects of a specic pollutant,
laboratory exposure of volunteers (almost exclusively usually by complex statistical manipulation of the data [6].
normal subjects or patients with mild asthma) to specic The observation of an eect will depend on whether there is
pollutants, on in vitro work on cells obtained from nasal a threshold for an eect which will always be present at an
or bronchoalveolar lavage (BAL) and studies of cultured individual level but which may be more dicult to deter-
cell monolayers. mine for populations.

509
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 39.2 Factors affecting response to air pollutants. protected the airway against asthmagenic stimuli is debatable.
Any such eect may take longer to be expressed than we have
Pollutant Degree of exposure (concentration)
had time to observe to date, but the change in hay fever rates
Duration of exposure
does suggest that allergic conditions may be emerging as a
Coexposure Other pollutants (both indoor and outdoor) result of this diering exposure, although indoor exposures
Allergens and possible changes in diet may also be playing a part.
Viral infections The studies of Seventh Day Adventists [12] (a non-
Meteorological conditions (e.g. cold air) smoking population) suggest that chronic exposure to dif-
Degree of physical activity ferent components of the pollutant mix may result in an
Cigarette smoke (both active and passive) increased incidence of new cases of diagnosed asthma over
time, but these are isolated ndings in an unusual population
Host factors Atopy and it is dicult to extrapolate these ndings more widely.
Age (infants, perhaps the elderly)
The fetus
Preexisting disease state Chronic obstructive pulmonary disease
Use of treatment (e.g. bronchodilators)
Airflow obstruction The evidence for pollution contributing to the prevalence
Bronchial hyperresponsiveness of COPD is limited to studies from the early 1960s in the
United Kingdom [4]. The overwhelming eect of cigarette
smoking as the main cause of this condition means that con-
tributions from other environmental exposures to initiation of
COPD are dicult to quantify. The recent ndings that coal-
CHRONIC EFFECTS OF AIR POLLUTION dust exposure may contribute to chronic productive bronchitis
and airow obstruction independent of cigarette smoke [13]
adds some supportive evidence to the possibility that cur-
The importance of a possible chronic eect of exposure to
rent air pollutant exposure may contribute to the prevalence
air pollution is that it brings more members of the popula-
of COPD, although the character of the particulate exposures
tion into the subpopulation susceptible to triggering by air
is clearly greatly dierent both in amount and in type. There
pollutant exposure. At present, the evidence for air pollution
are more recent data from Germany which suggests that in
contributing to the prevalence of both COPD and asthma
women living near busy roads, COPD is more prevalent.
is not compelling. However, it is generally believed that the
chronic eects of air pollution may prove to be, at least in
public health terms, more important than the acute eects.
A chronic eect can best be identied either from prospec-
tive, longitudinal studies such as the US Six Cities Study [7]
and the American Cancer Society Study [8], from cross-
ACUTE EFFECTS OF AIR POLLUTION
sectional studies comparing prevalence between areas of dif-
fering pollutant exposure or from life-table analysis [9]. All Asthma
types of study are open to the criticism that early life pollut-
ant exposures could have contributed to later life symptoms Panel studies
and morbidity, but are the best currently available methods
for assessing chronic eects. Panel studies of children with asthma, mostly from North
America, have shown small day-to-day changes in peak ow
and symptoms [14], the most frequently reported pollutants
Asthma associated with eects being NO2, ozone, and particles. On
average, inhaler use and symptoms increase by around 3%
In some cases, serial cross-sectional studies have provided for every 10 g/m3 rise in PM10 with a less than 1% fall in
important insights into changes in respiratory disease in peak ow for the same change [14]. However, not all stud-
the face of alterations in pollutant exposures. The German ies show such eects and the PEACE study from Europe
pre- and post-reunication studies [10, 11] have demon- was essentially negative [15, 16]. Where they occur, eects
strated that, with marked reductions in black smoke and are most marked in children either with more preexisting
SO2 levels (although no change in NO2 levels) in what was symptoms or who are atopic or both. It is not clear whether
previously East Germany, the prevalence of episodes of bron- greater disease severity predicts greater eects [17]. There
chitis in children has fallen. Asthma prevalence has, however, is some evidence to suggest that personal exposure to NO2
remained unchanged at a lower level than in the old West (most of which is likely to be from indoor sources) may pre-
Germany, although both hay fever and eczema have begun to dispose asthmatic subjects to exacerbations in association
increase. The implications of these ndings are still unclear. It with respiratory tract infections [18].
is likely that the higher levels of SO2 and black smoke expe-
rienced in East Germany before reunication contributed
to sputum production and episodes of bronchitis, in parallel Hospital admissions
with the pattern of symptoms seen in industrialized cities in Hospital admissions for asthma have been shown to be
the Western world up to the 1960s. Whether these exposures related to air pollutant levels on a day-to-day basis both in

510
Atmospheric Pollutants 39
adults and in children [19, 20], although not consistently. -coefficient
This may appear to conict with the small eects seen in
0.1 0 0.1
the panel studies, although it can be argued that some indi-
London (13)
viduals may be more likely to be admitted on days when
other asthmagenic factors combine to produce an attack if
Barcelona (40)
they are concurrently exposed to higher levels of pollution.
Paris (26)
Mortality
No relationship has been shown between death from asthma Athens (73)
and day-to-day changes in air pollution although the num-
bers of asthma deaths occurring in a city on a given day are Cracow (73)
so few that a small eect might yet exist.
Lodz (57)
Episodes
Episodes of air pollution might in theory result in asthma Wroclaw (54)
deaths or hospitalization. The London smog of 1952 [1]
resulted in many deaths from bronchitis, but no clear Poznan (34)
evidence of an eect in asthma. The more recent 1991 [21]
episode in the same city (where the sources of pollutants were Pooled relative risk
quite dierent) showed no eect on any index of asthma,
suggesting that any eect that might have occurred would Pooled relative risk
(western cities)
have been in terms of small changes in symptoms rather than
severe attacks. Pooled relative risk
(eastern cities)
0.9 1 1.1
Relative risk
Chronic obstructive pulmonary disease FIG. 39.2 Relative risks for death by city from respiratory disease for a
50 g/m3 rise in black smoke for a range of European cities (the APHEA
Panel studies project [26]).
There is limited information on day-to-day changes in clini-
cal state in patients with COPD. There is some evidence for
changes in symptoms in relation to particles and treatment
use in relation to NO2 and for changes in lung function MECHANISMS OF THE EFFECTS OF AIR
[22]. These changes were modest (the lung function change POLLUTANTS
amounted to a 0.2% fall in FEV1 for a 10 g/m3 rise in PM10)
and changes were either same day eects or lagged by 24 h.
Mechanisms of damage by particles
Hospital admissions
Particles are likely to exert health eects via a number of
There is a consistent eect of air pollutant exposure and hos- mechanisms. One hypothesis is that macrophages become
pital admissions for COPD from studies across a wide range overloaded allowing particles to penetrate through the
of countries. In the United States, hospital admissions increase alveolar/bronchiolar wall and become interstitialized [28].
between 1% and 3% for a 10 g/m3 rise in PM10 [14], while This initiates an inammatory response which could both
in Europe, the APHEA studies have shown smaller overall aect gas exchange in COPD and also release prothrom-
eects of around 1% [23, 24]. In most instances particles seem bogenic cytokines, which, in an individual with a compro-
to be the most important pollutant, although the London mised coronary circulation, could lead to an acute cardiac
study [25] showed no eect of particles, but an eect of ozone event [29]. The inammation may be caused through release
in the warmer months only. There is a range of eects how- of free radicals, perhaps because of transition metals on the
ever, across countries, with eects being less marked in eastern particles [30, 31] although just the very small size of the
Europe and more marked in western countries (Fig. 39.2). particles themselves may be responsible [32]. Patients with
COPD have a condition which is almost exclusively caused
Mortality by cigarette smoking and many will thus have coexistent
Deaths from COPD are similarly associated with pollution atheromatous coronary disease. It is possible, therefore, that
and the eects are usually most strongly seen at lags of around increase in hospital admissions and deaths seen in patients
3 days [14] mediated largely through particles, although SO2 with COPD associated with days of higher air pollution
also appears to contribute in Europe. On average the increase might not simply be due to enhancement of airway inam-
in respiratory mortality for a 10 g/m3 rise in PM10 is 3.4% mation or facilitation of microbial pathogenicity, but due to
on a day-to-day basis [14], although in those parts of Europe cardiac decompensation.
where particle exposures are generally higher, this eect is less There have been few studies of particulate challenge in
marked suggesting a tolerance eect [26, 27]. asthma and none in COPD. One series of freshly generated

511
Asthma and COPD: Basic Mechanisms and Clinical Management

diesel challenges in healthy normal subjects [33] has revealed [45]. Ozone can also increase nonspecic airway responsive-
inltration of inammatory cells and increases in some ness in both normal and asthmatic subjects in a dose-related
proinammatory cytokines, notably IL-8, albeit at high pol- manner [46]. Repeated daily exposures of asthmatic subjects
lutant concentrations which may be of relevance in both the to ozone lead to progressive loss of the response suggest-
asthmatic and COPD settings. For asthma, there is also evi- ing tolerance to its eects [47]. Ozone also potentiates the
dence that diesel exhaust can potentiate the local production bronchoconstrictor eect of allergen in asthma either alone
of specic IgE in the nose with an associated shift in T-cell [48] or in combination with NO2 [49]. The inammatory
patterns compatible with this [34, 35]. response to ozone is very variable. Although most studies
have shown an increase in neutrophils in BAL some, but not
all, have shown increases in certain adhesion molecules and
Mechanisms of damage by gases cytokines (notably IL-8) [41, 50, 51]. There are, however,
marked interindividual dierences and any changes in air-
Sulfur dioxide ow so induced do not necessarily match changes in inam-
matory markers which are often present at elevated levels
SO2 is a highly soluble, irritant gas quickly absorbed by the some hours after exposure even though the cellular response
respiratory tract. It is a potent bronchoconstrictor in asthma has settled. There is suggestive evidence that patients with
particularly with exercise, but there is marked variability in mild COPD exposed to ozone (along with exercise) at 200
the response between individuals, individuals responding to 300 ppb may show small decrements in both lung function
between 200 ppb and over 1500 ppb [36]. An airway eect and oxygen saturations although this needs conrmation
is only seen in normal subjects at exposures of 45 ppm. In with adequately powered studies [52, 53].
asthma SO2 is thought to activate rapidly adapting recep-
tors (RAR) [37], thus leading directly to bronchocon-
striction. Lower doses inhaled at rest (200 ppb) can alter
autonomic balance in asthma invoking the possibility that
the gas initially activates neurogenic inammation through SUMMARY
a RAR initiated, vagally mediated reex which subsequently
results in an autonomic modulating response which diers
from that seen in normal subjects [38]. Air pollution can aect some patients with asthma, in terms
of symptoms and hospital admissions although the picture
varies between countries and perhaps according to severity.
Nitrogen dioxide There is evidence for all components of the pollutant mix to
Nitrogen dioxide is an oxidant gas which at high concen- be capable of playing a role, although the mechanisms are
trations can cause acute pulmonary edema (e.g. silo llers only just being unraveled. For patients with COPD, air pol-
disease). At levels usually experienced in ambient air (15 lution increases the risks of hospital admission and death,
30 ppb as urban background levels) eects are dicult to particles being the most important pollutant. Particles may
nd. At high concentrations (10003000 ppb usually with exacerbate inammation by generation of free radicals, per-
exercise) some changes in spirometric indices and bronchial haps through transition metals. Interstitialization of par-
hyperresponsiveness (BHR) have been recorded in asth- ticles at bronchioloalveolar level may exacerbate COPD
matic subjects, but these are of modest degree [39]. At these itself or induce cardiac events by release of prothrombotic
high concentrations some inammatory markers are raised cytokines. Figure 39.1 summarizes the possible mechanisms
in BAL uid after challenge in normal subjects, although of the health eects of particles in both COPD and asthma.
there is considerable inconsistency between studies [40, 41]. At present there is no specic approach for dealing
At lower exposures (400 ppb) NO2 has been shown to with any air pollution-induced changes in clinical state in
enhance the acute bronchoconstrictor eect of house dust either asthma or COPD, although increasing inhaled anti-
mite allergen exposure, both at high dose [42] or as repeated inammatory treatment makes intuitive sense, while avoid-
smaller doses of allergen [43]. This suggests that exposure ing exertion and other known triggers during periods of
to gaseous air pollutants may exert more subtle eects than higher air pollution may also be sensible.
can be identied by epidemiological studies and which con- The overall impact on public health of these triggering
sequently may prove very dicult to quantify at a popula- events of air pollution on patients with airway diseases is sig-
tion level. There is no evidence that any such potentiating nicant although may prove to be swamped by the size of the
eects are seen in COPD, for instance with respect to viral chronic eects once these are adequately quantied [54, 55].
infections.

Ozone References
Ozone is a highly reactive, oxidant gas and can cause inam-
1. Ministry of Health. Mortality and Morbidity During the London Fog
matory changes in the bronchial mucosa (largely in the ter- of December 1952. London: HMSO, 1954.
minal bronchioles) in animals at concentrations similar to 2. Lippmann M, Ito K. Separating the eects of temperature and season
those which can be experienced in ambient air (80120 ppb) on daily mortality from those of air pollution in London: 19651972.
[41, 44]. With exercise, it can produce similar degrees of Inhal Toxicol 7: 8597, 1995.
bronchoconstriction in both normal and asthmatic subjects 3. Lawther PJ, Waller RE, Henderson M. Air pollution and exacerbations
although there are large interindividual variations in response of bronchitis. Thorax 25: 52539, 1970.

512
Atmospheric Pollutants 39
4. Holland WW, Reid D. The urban factor in chronic bronchitis. Lancet i: 27. Sunyer J, Castellsagu J, Sez M, Tobias A, Ant JM. Air pollution
44548, 1965. and mortality in Barcelona. J Epidemiol Comm Health 50(Suppl 1):
5. Poloniecki JD, Atkinson RW, Ponce de Leon A, Anderson HR. Daily S7680, 1996.
time series for cardiovascular hospital admissions and previous days air 28. Oberdrster G, Ferin J, Lehnert BE. Correlation between particle size,
pollution in London. Occup Environ Med 54: 53440, 1997. in vivo particle persistence and lung injury. Environ Health Perspect
6. Schwartz J. Air pollution and hospital admissions for respiratory dis- 102(Suppl 5): 17379, 1994.
ease. Epidemiology 7: 2028, 1996. 29. Seaton A, MacNee W, Donaldson K, Godden D. Particulate air pollu-
7. Dockery DW, Pope CA, Xu X et al. An association between air pollu- tion and acute health eects. Lancet 345: 17678, 1995.
tion and mortality in six US cities. N Engl J Med 329: 175359, 1993. 30. Carter JD, Ghio AJ, Samet JM, Devlin RB. Cytokine production
8. Pope CA, Thun MJ, Namboodiri MM et al. Particulate air pollution as by human airway epithelial cells after exposure to an air pollution
a predictor of mortality in a prospective study of US adults. Am J Respir particle is metal dependant. Toxicol Appl Pharmacol 146: 18088,
Crit Care Med 151: 66974, 1995. 1997.
9. Hurley JF, Holland MR, Markandya A et al. Towards assessing the 31. Costa DL, Dreher K-L. Bioavailable transition metals in particulate
health impacts of ambient particulate air pollution in the UK. Final matter mediate cardiopulmonary injury in health and compromised
report for UK Department of Health, 2000. animal models. Environ Health Perspect 105(S5): 105360, 1997.
10. von Mutius E, Fritzsch C, Weiland SK, Roll G, Magnussen H. 32. Donaldson K, Li XY, MacNee W. Ultrane (nanometer) particle-
Prevalence of asthma and allergic disorders among children in united mediated lung injury. J Aerosol Sci 29: 55360, 1998.
Germany. BMJ 305: 139599, 1992. 33. Salvi SS, Nordenhall C, Blomberg A et al. Acute exposure to diesel
11. Weiland SK, von Mutius E, Hirsch T et al. Prevalence of respiratory exhaust increases IL-8 and GRO-alpha production in healthy human
and atopic disorders among children in the East and West of Germany airways. Am J Respir Crit Care Med 161: 55057, 2000.
ve years after unication. Eur Respir J 14: 86270, 1999. 34. Diaz-Sanchez D, Dotson AR, Takenaka H, Saxon A. Diesel exhaust
12. Abbey DE, Nishino N, McDonnell WF et al. Long-term inhalable particles induce local IgE production in vivo and alter the pattern of
particles and other air pollutants related to mortality in non-smokers. IgE messenger RNA isoform. J Clin Invest 94: 141725, 1994.
Am J Respir Crit Care Med 159: 37382, 1999. 35. Diaz-Sanchez D, Tsien A, Fleming J, Saxon A. Combined diesel
13. Henneberger PK, Atteld MD. Respiratory symptoms and spirometry exhaust particulate and ragweed allergen challenge markedly enhances
in experienced coal miners: Eects of both distant and recent coal mine human in vivo nasal ragweed specic IgE and skews cytokine produc-
dust exposures. Am J Ind Med 32: 26874, 1997. tion to a Th2 type phenotype. J Immunol 158: 240613, 1997.
14. Dockery DW, Pope CA. Acute respiratory eects of particulate air 36. Sheppard D, Saisho A, Nadel JA, Boushey HA. Exercise increases
pollution. Ann Rev Public Health 15: 10732, 1994. sulfur dioxide-induced bronchoconstriction in asthmatic subjects. Am
15. Roemer W, Hoek G, Brunekreef B, Haluszka J, Kalandidi A, Rev Respir Dis 123: 48691, 1981.
Pekkanen J. Daily variations in air pollution and respiratory health in a 37. Atzori L, Bannenberg G, Corriga AM et al. Sulfur dioxide-induced
multicentre study: The PEACE project. Eur Respir J 12: 135461, 1998. bronchoconstriction via ruthenium red-sensitive activation of sensory
16. Roemer W, Hoek G, Brunekreef B et al. The PEACE project: General nerves. Respiration 59: 27278, 1992.
discussion. Eur Respir Rev 8: 12530, 1998. 38. Tunniclie WS, Mark D, Harrison RM, Ayres JG. Eect of particle
17. Brunekreef B, Kinney PL, Ware JH et al. Sensitive subgroups and nor- and sulphur dioxide challenge on heart rate variability in normal and
mal variation in pulmonary function response to air pollution episodes. asthmatic subjects. Eur Respir J 17: 6048, 2001.
Environ Health Perspect 90: 18993, 1991. 39. Folinsbee L. Does nitrogen dioxide exposure increase airways respon-
18. Linaker CH, Coggon D, Holgate ST et al. Personal exposure to nitro- siveness? Toxicol Ind Health 8: 27383, 1992.
gen dioxide and risk of airow obstruction in asthmatic children with 40. Blomberg A, Krishna MT, Bocchino V et al. The inammatory eects
upper respiratory infection. Thorax 55: 93033, 2000. of 2 ppm NO2 on the airways of healthy subjects. Am J Respir Crit Care
19. Walters S, Griths RK, Ayres JG. Temporal association between hos- Med 156: 41824, 1997.
pital admissions for asthma in Birmingham and ambient levels of sul- 41. Blomberg A. Airway inammatory and antioxidant responses to oxida-
phur dioxide and smoke. Thorax 49: 13340, 1994. tive and particulate air pollutants experimental exposure studies in
20. Sunyer J, Spix C, Quenel P et al. Urban air pollution and emergency humans. Clin Exp Allergy 30: 31017, 2000.
admissions for asthma in four European cities: The APHEA project. 42. Tunniclie WS, Burge PS, Ayres JG. Eect of domestic concentrations
Thorax 52: 76065, 1997. in nitrogen dioxide on airway responses to inhaled allergen in asth-
21. Anderson HR, Limb ES, Bland JM, Ponce de Leon A, Strachan DP, matic patients. Lancet 344: 173336, 1994.
Bower JS. Health eects of an air pollution episode in London, December 43. Strand V, Svartengren M, Rak S, Barck C, Bylin G. Repeated expo-
1991. Thorax 50: 118893, 1995. sures to an ambient level of NO2 enhances asthmatic response to a
22. Harre ESM, Price PD, Ayrey RB, Toop LJ, Martin IR, Town GI. non-symptomatic allergen dose. Eur Respir J 12: 612, 1998.
Respiratory eects of air pollution in chronic obstructive pulmonary 44. Aris RM, Christian D, Hearne PQ, Kerr K, Finkbeiner WE,
disease: A three month prospective study. Thorax 52: 104044, 1997. Balmes JR. Ozone-induced airway inammation in human subjects
23. Spix C, Anderson HR, Schwartz J et al. Short-term eects of air as determined by airway lavage and biopsy. Am Rev Respir Dis 148:
pollution on hospital admissions of respiratory diseases in Europe: 136372, 1993.
A quantitative summary of APHEA study results. Air pollution and 45. Hazucha MJ. Relationship between ozone exposure and pulmonary
health: A European approach. Arch Environ Health 53: 5464, 1998. function changes. J Appl Physiol 62: 167180, 1987.
24. Anderson HR, Spix C, Medina S et al. Air pollution and daily 46. Horstman DH, Folinsbee LJ, Ives PJ, Abdul-Saleem S, McDonnell WF.
admissions for chronic obstructive pulmonary disease in 6 European cit- Ozone concentration and pulmonary response relationships for 6.6 hour
ies: Results from the APHEA project. Eur Respir J 10: 106471, 1997. exposures with ve hours of moderate exercise to 0.08, 0.10 and 0.12 ppm.
25. Ponce de Leon A, Anderson HR, Bland JM, Strachan DP, Bower J. Am Rev Respir Dis 142: 115863, 1990.
Eects of air pollution on daily hospital admissions for respiratory 47. Horvath SM, Gliner JA, Folinsbee LJ. Adaptation to ozone: Duration
disease in London between 198788 and 199192. J Epidemiol Comm of eect. Am Rev Respir Dis 123: 49699, 1981.
Health 50(Suppl. 1): S6370, 1996. 48. Jrres R, Nowak D, Magnussen H. The eect of ozone exposure on
26. Katsouyanni K, Touloumi G, Spix C et al. Short term eects of ambient allergen responsiveness in subjects with asthma or rhinitis. Am J Respir
sulphur dioxide and particulate matter on mortality in 12 European Crit Care Med 153: 5664, 1996.
cities: Results from time series data from the APHEA project. 49. Jenkins HS, Devalia JL, Mister RL, Bevan AM, Rusznak C, Davies RJ.
BMJ 314: 165863, 1997. The eect of exposure to ozone and nitrogen dioxide on the airway

513
Asthma and COPD: Basic Mechanisms and Clinical Management

response of atopic asthmatics to inhaled allergen. Am J Respir Crit Care 53. Kehrl HR, Hazucha MJ, Solic JJ, Bromberg PA. Responses of subjects
Med 160: 3339, 1999. with chronic obstructive pulmonary disease after exposure to 0.3 ppm
50. Holz O, Jrres R, Timm P et al. Ozone-induced airway inammatory ozone. Am Rev Respir Dis 131: 71924, 1985.
changes dier between individuals and are reproducible. Am J Respir 54. Committee on the Medical Eects of Air Pollution. Quantication of
Crit Care Med 159: 77684, 1999. the health eects of air pollution in the UK. Department of Health:
51. Jrres RA, Holz O, Zachgo W et al. The eect of repeated ozone The Stationery Oce, 1997.
exposures on inammatory markers in bronchoalveolar lavage uid and 55. Knzli N, Kaiser R, Medina S et al. Public health impact of outdoor
mucosal biopsies. Am J Respir Crit Care Med 161: 185561, 2000. and trac-related air pollution: A European assessment. Lancet 356:
52. Solic JJ, Hazucha MJ, Bromberg PA. The acute eects of 0.2 ppm 795801, 2000.
ozone in patients with chronic obstructive pulmonary disease. Am Rev
Respir Dis 125: 66469, 1982.

514
Drugs
40CHAPTER

minutes to several hours after the ingestion of


NONSTEROIDAL ANTI- aspirin or other NSAIDs [2, 3]. The asthmatic
Neil C. Thomson1 and
INFLAMMATORY DRUGS reaction can be associated with other symptoms, Peter J. Barnes2
including rhinorrhea, ushing, and loss of con- 1
Department of Respiratory Medicine,
sciousness, and very rarely the attack may be fatal.
Introduction Division of Immunology, Infection and
In the typical case, symptoms of chronic
rhinitis are present for many years before asthma Inflammation, University of Glasgow,
In the early part of the 20th century it was rst develops. The rhinitis starts as intermittent watery Glasgow, UK
recognized that aspirin could precipitate asthma 2
rhinorrhea, which develops during the second or National Heart and Lung Institute
in susceptible individuals [1]. Similar asthmatic third decade of life. The rhinorrhea becomes pro- (NHLI), Clinical Studies Unit,
reactions were later shown to occur with other gressively more severe and is complicated by nasal Imperial College, London, UK
nonsteroidal anti-inammatory drugs (NSAIDs) polyp formation and sinusitis. On average 2 years
[2] and patients with aspirin-induced asthma later symptoms of asthma appear associated with
were found to be sensitive to other cyclooxygenase the development of acute asthma after the inges-
(COX)-1 inhibitors. Various terms have been used tion of COX-1 inhibitors. Although in this group
to describe the sensitivity to NSAIDs, includ- drugs may precipitate asthma, these patients con-
ing aspirin-induced asthma, aspirin-sensitive tinue to have nasal and asthmatic symptoms in
asthma, aspirin hypersensitivity, idiosyncratic the absence of ingesting aspirin or other NSAIDs.
reaction to aspirin, aspirin intolerance, and non- Respiratory symptoms are often chronic, severe,
allergic hypersensitivity reactions to aspirin and and perennial in nature. Individuals with aspirin-
NSAIDs [3]. induced asthma are more commonly female. Skin
tests to common external allergens are positive
in a third to over a half of subjects. A small per-
Prevalence centage of patients with aspirin-induced asthma
also develop associated symptoms of urticaria and
The prevalence of hypersensitivity reactions to
angioedema after ingestion of aspirin.
aspirin and NSAIDs in individuals with asthma
is unclear, with estimates ranging from 4.3% to
21% [46]. The prevalence gures are higher
when determined by oral provocation testing Diagnosis
(e.g. 21% in adults and 5% in children [6]). Oral
aspirin challenges undertaken in subgroups of In most clinical circumstances, the diagnosis is
patients with asthma, nasal polyps, and chronic based on a history of bronchoconstriction fol-
sinusitis attending specialized allergy centers lowing the ingestion of a COX-1 inhibitor. It
provide high positive rates of 30% or greater. should be appreciated, however, that a history
of either the presence or the absence of aspirin-
induced asthma may result in both a false posi-
Clinical features tive and a false negative diagnosis. For example,
patients with a history of aspirin-induced asthma
Aspirin-induced asthma is characterized by have a positive oral aspirin challenge in approxi-
the development of bronchoconstriction within mately 60% to over 90% of cases. A negative oral

515
Asthma and COPD: Basic Mechanisms and Clinical Management

challenge may occur in patients with a history of aspirin- thromboxane. An abnormality in the arachidonic acid path-
induced asthma due to changing responsiveness to aspirin, a way in the respiratory tract of patients with aspirin-induced
high threshold to aspirin, or a misleading history. asthma seems to be central to the development of an asth-
The only reliable method of establishing a diagnosis matic attack after the ingestion of NSAIDs (Fig. 40.1). In
of aspirin-induced asthma is by aspirin challenge. Oral aspi- support of this hypothesis, the severity of bronchoconstric-
rin challenge is indicated in patients with adult asthma who tion induced by aspirin or other NSAIDs, such as indometh-
require treatment with an NSAID. The oral challenge pro- acin, ufenamic acid, and naproxen, is directly proportional
cedure has been reported to be safe, if a standardized proto- to their ability to inhibit COX in vitro. Furthermore, speci-
col is used [3, 7]. An alternative but less widely used method mens of nasal polyps removed from patients with aspirin-
of detecting aspirin-induced asthma involves administering induced asthma are more sensitive to the inhibition eects
aspirin-lysine by inhalation [7]. It is claimed that this method of aspirin on the COX-1 pathway.
is safe and when compared to oral challenge the procedure is The inuence of PG on bronchomotor tone may dif-
shorter and the reaction is localized to the respiratory tract. fer between patients with or without aspirin-induced asthma.
Aspirin-lysine administered as a nasal challenge has also been The control of airway caliber in the latter group could be more
used to diagnose aspirin-sensitive asthma and this technique dependent on the eects of PG, such as PGE2, either due to
does not induce bronchoconstriction [8]. No in vitro test is its direct bronchodilator activity or indirectly by suppression
currently available to detect aspirin-induced asthma. of the release of bronchoconstrictor mediators. Alternately,
Tartrazine is a yellow dye used for coloring foods and the production of PGE2 may be impaired in these individuals.
drugs such as confectionery, soft drinks, antibiotics, and In support of this later mechanism, peripheral blood mono-
antihistamines. A number of earlier reports suggested that nuclear cells release reduced amounts of PGE2 in patients
tartrazine caused bronchospasm in some patients with aspi- with aspirin-induced asthma [10], although a recent clinical
rin-induced asthma, but further research has failed to sup- challenge study with aspirin found that urinary metabolites
port any cross-reactivity between tartrazine and aspirin in of PGE2 were not altered in patients with aspirin-induced
patients with aspirin-induced asthma [9]. asthma [11]. The expression of the E-prostanoid (EP) 2
receptor for PGE2 is reduced in nasal tissue of patients with
aspirin-induced asthma [12] and polymorphisms in the pro-
Mechanisms moter region of the gene coding for the EP2 gene are associ-
ated with sensitivity to aspirin [13].
Alterations in the arachidonic acid pathway The lipoxygenase pathway also appears to be abnor-
A characteristic feature of patients with aspirin-induced mal in these individuals, since the synthesis of leukotriene
asthma is the occurrence of airway obstruction following (LT) E4 is frequently increased in aspirin-induced asthma
the ingestion of drugs which inhibit COX-1, the enzyme [14], the expression of cys LT1 receptors is increased in
that converts arachidonic acid into prostaglandins (PG) and nasal inammatory cells [15], and leukotriene C4 synthase

Membrane
phospholipids
Aspirin

Arachidonic acid

5-LO and FLAP Cox-1 and Cox-2

LTA4 PGH2
TBxA2 synthase
PG synthase, Isomerase and
LTA2 hydrolase reductase
LTC4 synthase

LTB4 LTC4, LTD4, LTE4 TBxA2 PGI2 PGD2 PGE2 PGF2

2 1

cys LT1 receptor EP2 receptor

Bronchoconstriction Bronchodilation

FIG. 40.1 Biosynthetic pathways leading to the production of LT and PG. Possible mechanisms by which aspirin and NSAIDs inhibit COX-1 to induce
bronchoconstriction: (1) Impaired PGE2 synthesis and/or reduced PGE2 receptor function. (2) Increased cys LT synthesis and/or increased cys LT1 receptor
function. 5-LO: 5-lipoxygenase; FLAP: 5-lipoxygenase-activating protein; TBxA2: thromboxane A2; PGI2: prostacyclin.

516
Drugs 40
(LTC4S), an enzyme involved in the synthesis of LTs, is or betamethasone. Although cys LTs have been implicated
often overexpressed in eosinophils and in bronchial tissue in the pathogenesis of aspirin-induced asthma, LT receptor
[16]. Furthermore, genetic polymorphisms of LTC4S pro- antagonists and a 5
-lipoxygenase inhibitor have not been
moter region have been found in some patients with aspirin- found to be any more eective than in nonsensitive asthmatic
sensitive asthma [17], but this has not been conrmed in patients [20].
other populations [18].
Taken together, these ndings point to abnormali- Desensitization
ties in the arachidonic acid pathway in patients sensitive to Following the administration of oral or inhaled aspirin to
NSAIDs in particular impaired PGE2 synthesis and actions patients with aspirin-induced asthma there is a refractory
and excessive LT synthesis and eects. There is, however, an period to further aspirin challenge, which lasts for 25 days.
overlap in some of the defects in the arachidonic acid path- Continued administration of aspirin on a daily basis will
way found in patients with aspirin-induced asthma when maintain this refractory state, and this eect has been termed
compared with individuals without sensitivity to NSAIDs, desensitization. The process of outpatient desensitization
suggesting that the exact mechanisms underlying aspirin- is safe [21] and results in desensitization not only to aspi-
induced asthma are not yet fully elucidated. rin but also to other NSAIDs. Those patients who react to
small doses of aspirin require several aspirin challenges before
Other mechanisms desensitization is accomplished, whereas less aspirin-sensitive
There is no convincing evidence that aspirin-induced asthma patients are more quickly desensitized. The process by which
is due to IgE-mediated mechanisms. For example, specic aspirin ingestion causes desensitization is unknown.
anti-aspiryl IgE antibodies are generally absent and skin
tests to aspiryl-polylysine are negative in these patients. Basal
and post-aspirin challenge complement levels are not altered
in patients with aspirin-induced asthma. Inammatory
mediators are released following aspirin challenge, such as -BLOCKERS
neutrophil chemotactic factor, but this is thought to be sec-
ondary to the primary biochemical events.
Nonselective and cardioselective -blockers

Management Nonselective -adrenergic receptor antagonists precipitate


acute bronchoconstriction in asthma, even in individuals with
Avoidance mild disease [22], and attacks can be fatal. The dose of -
blockers that causes bronchospasm may be low and there are
Patients with a history of acute bronchoconstrictor response case reports of severe asthma attacks induced by eye drops of
after the ingestion of NSAIDs must be warned to avoid timolol, a nonselective -blocker used to treat glaucoma [23].
these drugs. If they require a simple analgesic, then acetami- Compared to individuals with asthma, patients with chronic
nophen (paracetamol) is usually safe in doses up to 500 mg. obstructive airways disease (COPD) are less likely to develop
Cross-reactivity can occur with high doses of acetaminophen deterioration in lung function after a nonselective -adrenergic
(paracetamol), which is a weak inhibitor of COX-1, although receptor antagonist [24], although there are a few case reports
the respiratory reaction tends to be mild. There is consider- of acute bronchospasm induced by nonselective -blockers
able data to indicate that COX-2 inhibitors do not induce [25]. In patients with COPD who have acute exacerbations
bronchoconstriction in individuals sensitive to aspirin [19], there is no evidence that -blockers are detrimental and may
although a few case reports of reactions have been published. even reduce mortality [26].
If a patient with aspirin-induced asthma should A meta-analysis of 19 studies on single-dose treat-
require an NSAID for the treatment of another condi- ment and 10 studies on continued treatment with cardiose-
tion, such as arthritis, then the patient can be desensitized lective 1-blockers concluded that this group of drugs does
to aspirin. This procedure should be undertaken by doctors not precipitate bronchospasm in patients with mild to mod-
with experience in aspirin challenge. erate reactive airway disease [27]. Nevertheless there are
case reports of individuals with asthma who have developed
Treatment of respiratory disease bronchospasm due to cardioselective -blockers [28]. Thus
These patients often have severe chronic asthma and nasal despite the recommendation that cardioselective -blockers
disease, which should be treated along similar lines to that should not be withheld from patients with mild to moder-
of patients with chronic asthma who are not sensitive to ate asthma [27] it seems prudent to administer these drugs
NSAIDs. Pretreatment with a number of drugs, including with great care to patients with moderate persistent asthma
sodium cromoglycate, the H1 receptor antagonist clemastine, and to avoid their use in individuals with severe persist-
and ketotifen, can inhibit the bronchoconstrictor response ent asthma [28]. A recent systematic review of 20 studies
to aspirin ingestion in patients with aspirin-induced asthma. of cardioselective -blockers given to patients with COPD
A small percentage of patients with aspirin-induced asthma found no evidence of adverse eects on lung function or on
develop bronchoconstriction after an intravenous injection of the response to 2-agonists [25, 29].
hydrocortisone, and occasionally the bronchoconstriction can Acute bronchospasm induced by a nonselective
be severe. These patients do not react adversely to other intra- -blocker should be treated with an inhaled anticholiner-
venous steroids such as methylprednisolone, dexamethasone, gic bronchodilator such as ipratropium or oxitropium [30].

517
Asthma and COPD: Basic Mechanisms and Clinical Management

Any fall in lung function due to a cardioselective 1-blocker Adrenaline Normal


can be reversed by an inhaled short-acting 2-agonist.
-blocker
Cholinergic 2
Possible mechanisms nerve ACh No effect

M3
-blocker-induced bronchoconstriction occurs in individu-
als with asthma, but not in healthy subjects, which sug- M2
gests that endogenous activation of -receptors is important
in maintaining airway tone in asthma against neural and Adrenaline
inammatory bronchoconstrictor stimuli. Asthma
-blocker

Circulating catecholamines ACh Bronchoconstriction


-blockers could antagonize the bronchodilator eect of cir-
culating catecholamines in patients with asthma but not in
normal subjects. However, against this suggestion, circulating Autoreceptor Cholinergic
catecholamines are not elevated in asthmatic subjects, even dysfunction hyperresponsiveness
in those subjects who have demonstrable bronchoconstric- FIG. 40.2 Possible mechanisms of -blocker-induced asthma.
tion after propranolol and the concentrations of adrenaline Blockade of prejunctional 2-receptors on cholinergic nerves in normal
in plasma (0.3 nmol/l) are too low to have a direct eect individuals results in increased release of acetylcholine (ACh), but this is
on human airway smooth muscle tone [31, 32]. -Blockers compensated by stimulation of prejunctional muscarinic M2 receptors
may inhibit the action of catecholamines on some other to inhibit any increase in ACh. In patients with asthma, prejunctional M2
target cell, such as airway mast cells or cholinergic nerves. receptors are dysfunctional, so that there is a net release of ACh; ACh
Mediator release from human lung mast cells is potently also has a greater bronchoconstrictor effect on the airways due to airway
inhibited by -agonists [33]. The eect of -blockers may, hyperresponsiveness.
therefore, be an increase in mediator release, which may be
more marked in the leaky mast cells of asthmatic individu-
als. This idea is supported by the observation that cromolyn
sodium, a mast cell stabilizer, prevents the bronchocon- cause bronchoconstriction. 2-adrenergic receptors inhibit
striction produced by inhaled propranolol. However after the release of tachykinins from airway sensory nerves [37],
intravenous propranolol, no increase in plasma histamine has thus -blockers may increase the release of these neu-
been detected [34]. ropeptides, thereby increasing bronchoconstriction and
airway inammation. While this mechanism may not be
relevant in patients with mild asthma, in whom choliner-
Neural pathways gic mechanisms appear to account for the bronchoconstric-
A more likely explanation for -blocker-induced asthma is tor response to -blockers [30], it may be relevant in more
that there is an increase in neural bronchoconstrictor mech- severely aected asthmatic patients in whom cholinergic
anisms. 2-adrenergic receptors on cholinergic nerves in mechanisms do not appear to be as important.
human airways may be tonically activated by adrenaline to
modulate acetylcholine (ACh) release [35] and therefore to
dampen cholinergic tone. Blockage of these receptors would Inverse agonism
therefore increase the amount of ACh released tonically, but Another possible mechanism may be related to the recently
this would be compensated for by the increased stimulation recognized phenomenon of inverse agonism [38]. It has been
of prejunctional M2-autoreceptors, which would act home- found that some mutants of the -receptor have constitu-
ostatically to inhibit any increase in ACh release and there- tive activity and activate the coupling protein Gs, even in the
fore no increase in airway tone would occur, even with high absence of occupation by agonist. In this situation -blockers
doses of a -blocker. By contrast, in patients with asthma function as inverse agonists and have an inhibitory eect on
-blockers inhibit prejunctional -receptors in the same baseline function. It is possible that in asthmatic patients
way, increasing the release of ACh [36]; however, there may 2-receptors are constitutively active, so that -blockers result
be a defect in M2-receptor function in asthmatic airways, in adverse eects. Dierent -blockers have diering poten-
so that the increased release in ACh cannot be compen- cies as inverse agonists that are unrelated to their -blocking
sated. Thus increased ACh reaches M3 receptors on airway potency. Thus, propranolol is a potent inverse agonist whereas
smooth muscle. In addition, bronchoconstrictor responses pindolol is not and this may relate to the dierent tendency
to ACh are exaggerated in asthma, a manifestation of air- of these two agents to induce asthma.
way hyperresponsiveness, and thus two interacting amplify- It has recently been suggested that some -blockers
ing mechanisms may lead to marked bronchoconstriction with inverse agonist activity may even be benecial in
(Fig. 40.2). Evidence to support this hypothesis is provided asthma when used chronically [39]. In murine models
by the inhibitory eect of an inhaled anticholinergic drug -blockers with inverse agonist activity (nadolol and
oxitropium bromide on -blocker-induced asthma [30]. carvedilol) increased 2-receptor expression and caused
In patients with more severe asthma there may be bronchodilation [40]. In a pilot open study nadolol has been
an additional neural mechanism by which -blockers may reported to be safe in asthmatic patients and to reduce airway

518
Drugs 40
hyperresponsiveness [41]. Further controlled studies may sensitivity is relatively common and may aect 4% of asth-
therefore be indicated. matic individuals, especially children [9]. Ingestion of tartra-
zine may result in urticarial rashes and bronchoconstriction.
The mechanism may depend upon mediator release from
mast cells. Monosodium glutamate (MSG, E621) is added
to food as a avor enhancer. It is found in soy sauce, spices,
ANGIOTENSIN CONVERTING ENZYME stock cubes, hamburgers, and in Chinese restaurant food.
INHIBITORS Some people react with sweating, ushing, and numbness
of the chest; in patients with asthma this may be accom-
panied by wheezing, which may begin several hours after
A retrospective cohort study suggested that bronchospasm the ingestion (Chinese restaurant asthma syndrome) [47]
was twice as common in patients treated with angiotensin Precipitation of asthma symptoms by MSG is uncommon,
converting enzyme (ACE) inhibitors (5.5%) compared however.
to the reference group treated with lipid-lowering drugs
(2.3%) [42]. However, the prevalence of a past history
of bronchospasm in patients reporting ACE inhibitor-
induced bronchospasm was not signicantly dierent from
the prevalence in patients on ACE inhibitors without an LOCAL ANESTHETICS
adverse reaction [42]. In a controlled trial in asthmatic and
hypersensitive patients (with and without cough), there
was no change in lung function following administration Aerosols of the local anesthetics, such as bupivacaine and
of captopril and no increase in reactivity to histamine or lignocaine (lidocaine), cause bronchoconstriction in a propor-
bradykinin [43]. Similar ndings were obtained in a group tion of asthmatic patients [48, 49]. The degree of bronchial
of 21 patients with asthma given ACE inhibitors for 3 reactivity to histamine does not predict the development or
weeks, although one subject developed a slight wheeze [44]. extent of bronchoconstriction following lignocaine inhala-
Administration of a potent ACE inhibitor (ramipril) to a tion [49]. The mechanism of local anesthetic-induced bron-
group of individuals with mild asthma showed no change in choconstriction is unclear. Pretreatment with anticholinergic
lung function or bronchial reactivity to inhaled histamine, drugs partially attenuates the bronchoconstrictor response
nor was there any increase in bronchoconstrictor response to to aerosols of local anesthetics, suggesting that they may be
inhaled bradykinin [45]. Taken together these ndings sug- acting in part via a vagal reex pathway. Inhaled local anes-
gest that ACE inhibitors are unlikely to worsen asthma in thetics may selectively inhibit nonadrenergic noncholinergic
the majority of patients, although there may be occasional bronchodilator nerves and so allow unopposed vagal tone.
patients in whom this occurs. When there is a possibility Some evidence for this is provided by the demonstration
that asthma has been worsened or precipitated by an ACE that lignocaine inhalation blocks nonadrenergic nonchol-
inhibitor, the drug should be withdrawn and an alternative inergic reex bronchodilation in human subjects, leading to
agent selected. As many as 20% of hypertensive patients a reex bronchoconstrictor response [50]. It is important to
treated with ACE inhibitors may develop an irritant cough, be aware that some patients with asthma may develop bron-
although this is unrelated to the presence of underlying air- choconstriction with topical local anesthetics during ber
way disease or atopic status. optic bronchoscopy. All patients with asthma should receive
premedication with a bronchodilator prior to bronchoscopy.

ADDITIVES
OTHER DRUGS
Several chemicals used as additives in drug preparations
and food have been associated with worsening of asthma Many other drugs have been reported to lead to exacerba-
and should, where possible, be avoided. Bisulphites and tion of asthma in occasional patients. Bronchoconstriction
metabisulphites (E220, E221, E222, E226, and E227) are may constitute part of an anaphylactic reaction to a drug,
antioxidants used as preservatives in several foods, including such as penicillin or to intravenous dextran or to con-
wines (especially sparkling wines), beer, fruit juices, salads, trast media. Other drugs, such as opiates, may cause direct
and medications. Characteristically, they produce bron- degranulation of mast cells. Bronchodilator aerosols may
choconstriction within 30 min of ingestion and this may occasionally cause a paradoxical bronchoconstriction. This is
account for several cases of food allergy. The mechanism presumed to be due to the propellant (Freon) or other addi-
of metabisulphite-induced asthma is probably explained by tives (such as oleic acid, which is used as a surfactant). The
release of sulfur dioxide (SO2) after ingestion that is then mechanism of bronchoconstriction may be via a cholinergic
inhaled, since nebulized metabisulphite solutions generate reex. The treatment of paracetamol poisoning with intra-
SO2 in sucient quantities to provoke bronchoconstric- venous N-acetylcysteine has been reported to exacerbate
tion in asthmatic subjects [46]. Tartrazine (E102), a yellow asthma. Cholinergic agents such as pilocarpine, used for the
dye, is used as a coloring in many foods, beverages (such as treatment of dry mouth and pyridostigmine, used to treat
orange squash), and pharmaceutical preparations. Tartrazine myasthenia gravis can induce attacks of asthma.

519
Asthma and COPD: Basic Mechanisms and Clinical Management

References 21. Williams AN, Simon RA, Woessner KM, Stevenson DD. The relation-
ship between historical aspirin-induced asthma and severity of asthma
induced during oral aspirin challenges. J Allergy Clin Immunol 120(2):
1. Cooke R. Allergy in drug idiosyncrasy. JAMA 73: 75960, 1919. 27377, 2007.
2. Smith A. Response to aspirin-allergic patients to challenge by some 22. McNeill R. Eect of a beta-adrenergic-blocking agent, propranol, on
analgesics in common use. Br Med J 2: 49496, 1977. asthmatics. Lancet 2: 11012, 1964.
3. Stevenson DD, Szczeklik A. Clinical and pathologic perspectives on 23. Schoene R, Martin T, Charan N, French C. Timolol-induced bron-
aspirin sensitivity and asthma. J Allergy Clin Immunol 118(4): 77386, chospasm in asthmatic ronchitis. JAMA 245(14): 146061, 1981.
2006. 24. Lammers J, Folgering H, van Herwaarden C. Ventilatory eects of
4. Kasper L, Sladek K, Duplaga M, Bochenek G, Liebhart J, Gladysz U long-term treatment with pindolol and metoprolol in hypertensive
et al. Prevalence of asthma with aspirin hypersensitivity in the adult patients with chronic obstructive lung disease. Br J Clin Pharmacol
population of Poland. Allergy 58(10): 106466, 2003. 20(3): 20510, 1985.
5. Vally H, Taylor ML, Thompson PJ. The prevalence of aspirin intolerant 25. Salpeter S, Buckley N. Use of beta-blockers and beta-agonists in
asthma (AIA) in Australian asthmatic patients. Thorax 57(7): 56974, COPD: A review of clinical outcomes. COPD Update 2: 13339, 2007.
2002. 26. Dranseld MT, Rowe SM, Johnson JE, Bailey WC, Gerald LB. Beta-
6. Jenkins C, Costello J, Hodge L. Systematic review of prevalence blocker use and the risk of death in hospitalized patients with acute
of aspirin induced asthma and its implications for clinical practice. exacerbations of COPD. Thorax. 2008 63: 30105.
Br Med J 328(7437): 434, 2004. 27. Salpeter SR, Ormiston TM, Salpeter EE. Cardioselective {beta}-
7. Nizankowska E, Bestynska-Krypel A, Cmiel A, Szczeklik A. Oral blockers in patients with reactive airway disease: A meta-analysis. Ann
and bronchial provocation tests with aspirin for diagnosis of aspirin- Intern Med 137(9): 71525, 2002.
induced asthma. Eur Respir J 15(5): 86369, 2000. 28. Self T, Soberman JE, Bubla JM, Chan CC. Cardioselective beta-
8. Alonso-Llamazares A, Martinez-Cocera C, Dominguez-Ortega J, blockers in patients with asthma and concomitant heart failure or
Robledo-Echarren T, Cimarra-Alvarez M, Mesa del Castillo M. Nasal history of myocardial infarction: When do benets outweigh risks?.
provocation test (NPT) with aspirin: A sensitive and safe method to J Asthma 40(8): 83945, 2003.
diagnose aspirin-induced asthma (AIA). Allergy 57(7): 63235, 2002. 29. Salpeter S, Ormiston T, Salpeter E. Cardioselective beta-block-
9. Stevenson D, Simon R, Lumry W, Mathison D. Adverse reactions to ers for chronic obstructive pulmonary disease. Cochrane Database Syst
tartrazine. J Allergy Clin Immunol 78: 18291, 1986. Rev. 2005 (Issue 4): CD003566. DOI: 10.1002/14651858.CD003566.
10. Schafer D, Schmid M, Gode UC, Baenkler HW. Dynamics of eicosa- pub2.
noids in peripheral blood cells during bronchial provocation in aspirin- 30. Ind P, Dixon C, Fuller R, Barnes P. Anticholinergic blockade of beta-
intolerant asthmatics. Eur Respir J 13(3): 63846, 1999. blocker-induced bronchoconstriction. Am Rev Respir Dis 139(6):
11. Mastalerz L, Sanak M, Gawlewicz-Mroczka A, Gielicz A, Cmiel A, 139094, 1989.
Szczeklik A. Prostaglandin E2 systemic production in patients with 31. Barnes P. Neural control of human airways in health and disease. Am
asthma with and without aspirin hypersensitivity. Thorax 63(1): 2734, Rev Respir Dis 134(6): 1289314, 1986.
January 1, 2008. 32. Thomson N, Dagg K, Ramsay S. Humoral control of airway tone.
12. Ying S, Meng Q, Scadding G, Parikh A, Corrigan CJ, Lee TH. Thorax 51: 46164, 1996.
Aspirin-sensitive rhinosinusitis is associated with reduced E-prostanoid 33. Church M, Hiroi J. Inhibition of IgE-dependent histamine release
2 receptor expression on nasal mucosal inammatory cells. J Allergy from human dispersed lung mast cells by anti-allergic drugs and salb-
Clin Immunol 117(2): 31218, 2006. utamol. Br J Pharmacol 90(2): 42129, 1987.
13. Jinnai N, Sakagami T, Sekigawa T, Kakihara M, Nakajima T, Yoshida K 34. Ind P, Barnes P, Brown M, Dollery C. Plasma histamine concentration
et al. Polymorphisms in the prostaglandin E2 receptor subtype 2 gene during propranolol induced bronchoconstriction. Thorax 40(12): 9039,
confer susceptibility to aspirin-intolerant asthma: A candidate gene 1985.
approach. Hum Mol Genet 13(24): 320317, 2004. 35. Barnes P. Muscarinic receptor subtypes: Implications for lung disease.
14. Daern P, Muileburg D, Hugli T, Stevenson D. Association of urinary Thorax 44(3): 16167, 1989.
leukotriene E4 excretion during aspirin challenges with severity of res- 36. Barnes P. Modulation of neurotransmission in airways. Physiol Rev
piratory responses. J Allergy Clin Immunol 104: 55964, 1999. 72(3): 699729, 1992.
15. Sousa AR, Parikh A, Scadding G, Corrigan CJ, Lee TH. Leukotriene- 37. Verleden G, Belvisi M, Rabe K, Miura M, Barnes P. Beta 2-adrenoceptor
receptor expression on nasal mucosal inammatory cells in aspirin- agonists inhibit NANC neural bronchoconstrictor responses in vitro. J
sensitive rhinosinusitis. N Eng J Med 347(19): 149399, 2002. Appl Physiol 73(3): 119599, 1993.
16. Cowburn AS, Sladek K, Soja J, Adamek L, Nizankowska E, Szczeklik A 38. Le P. Inverse agonism: Theory and practice. Trends Pharmacol Sci
et al. Overexpression of leukotriene C4 synthase in bronchial biopsies 16(8): 25659, 1995.
from patients with aspirin-intolerant asthma. J Clin Invest 101(4): 39. Bond RA, Spina D, Parra S, Page CP. Getting to the heart of asthma:
83446, 1998. Can [beta] blockers be useful to treat asthma?. Pharmacol Ther 115(3):
17. Sanak M, Simon H-U. Leukotriene C4 synthase promoter polymor- 36074, 2007.
phism and risk of aspirin-induced asthma. Lancet 350(9091): 1599, 40. Callaerts-Vegh Z, SinghEvans KLJ, Dudekula N, Cuba D, Knoll BJ,
1997. Callaerts PFK et al. Eects of acute and chronic administration of
18. Kedda M-A, Shi J, Duy D, Phelps S, Yang I, OHara K et al. {beta}-adrenoceptor ligands on airway function in a murine model of
Characterization of two polymorphisms in the leukotriene C4 synthase asthma. Proc Nat Acad Sci 101(14): 494853, January 1, 2004.
gene in an Australian population of subjects with mild, moderate, and 41. Hanania NA, Singh S, El-Wali R, Flashner M, Franklin AE, Garner WJ
severe asthma. J Allergy Clin Immunol 113(5): 88995, 2004. et al. The safety and eects of the beta-blocker, nadolol, in mild asthma:
19. Stevenson DD, Simon RA. Lack of cross-reactivity between rofecoxib An open-label pilot study. Pulm Pharmacol Therapeut 21(1): 13441,
and aspirin in aspirin-sensitive patients with asthma. J Allergy Clin 2008.
Immunol 108(1): 4751, 2001. 42. Wood R. Bronchospasm and cough as adverse reactions to the ACE
20. Dahlen S-E, Malmstrom K, Nizankowska EWA, Dahlen B, Kuna P, inhibitors captopril, enalapril and lisinopril. A controlled retrospective
Kowalski M et al. Improvement of aspirin-intolerant asthma by cohort study. Br J Clin Pharmacol 39(3): 26570, 1995.
Montelukast, a leukotriene antagonist. A randomized, double-blind, 43. Overlack A, Mller B, Schmidt L, Scheid M, Mller M, Stumpe K.
placebo-controlled trial. Am J Respir Crit Care Med 165(1): 914, Airway responsiveness and cough induced by angiotensin converting
January 1, 2002. enzyme inhibition. J Hum Hypertens 6(5): 38792, 1992.

520
Drugs 40
44. Kaufman J, Schmitt S, Barnard J, Busse W. Angiotensin-converting 47. Allen D, Baker G. Chinese-restaurant asthma. N Eng J Med 305(19):
enzyme inhibitors in patients with bronchial responsiveness and asthma. 115455, 1981.
Chest 101(4): 92225, 1992. 48. Thomson N. The eect of dierent pharmacological agents on respiratory
45. Dixon C, Fuller RPJB. The eect of an angiotensin converting enzyme reexes in normal and asthmatic subjects. Clin Sci 56(3): 23541, 1979.
inhibitor, ramipril, on bronchial responses to inhaled histamine and 49. McAlpine LG, Thomson NC. Lidocaine-induced bronchoconstriction
bradykinin in asthmatic subjects. Br J Clin Pharmacol 23(1): 9193, in asthmatic patients. Relation to histamine airway responsiveness and
1987. eect of preservative. Chest 96(5): 101215, 1989.
46. Stevenson D, Simon R. Sultes and asthma. J Allergy Clin Immunol 74: 50. Lammers J, Barnes P, Chung K. Nonadrenergic, noncholinergic airway
46972, 1984. inhibitory nerves. Eur Respir J 5(2): 23946, 1992.

521
Clinical Assessment 7
PART

of Asthma and
COPD
Diagnosis of Asthma and COPD
41 CHAPTER

INTRODUCTION or elderly patient, a smoker, who may be atopic Fabrizio Luppi1,


or have a history of asthma, who complains
of chronic dyspnea but not wheezing, chronic Bianca Begh1,
Asthma and chronic obstructive pulmonary cough, or sputum, and who presents with poorly Lorenzo Corbetta2 and
disease (COPD) are dierent chronic inam- reversible airow limitation. It is also dicult Leonardo M. Fabbri1
matory respiratory disorders that may share a to make a diagnosis of asthma or COPD in a 1
common functional abnormality, that is, poorly middle-aged or elderly patient who has a clear Department of Respiratory Diseases,
reversible airow limitation [14]. According to history of atopy and asthma, bronchodilator University of Modena and Reggio Emilia,
current guidelines, airow limitation in asthma reversibility, and recurrent wheezing, but who also Modena, Italy
smokes and has chronic cough and sputum and 2
is reversible or partly reversible [1], whereas air- Department of Respiratory Diseases,
ow limitation in COPD is poorly reversible or dyspnea that are not suppressed by inhaled ster- University of Firenze, Firenze, Italy
not reversible at all [2]. oids. In such patients, dierential diagnosis might
In the pathogenesis of both asthma and become important from a clinical and therapeu-
COPD, individual genetic susceptibility and tic point of view. Inhaled glucocorticosteroids are
environmental exposures are relevant for dis- the rst choice of regular medication in asthma
ease expression. Cigarette smoking is the major but not in COPD, whereas regular long-acting
cause of COPD [2]. The causes of asthma are bronchodilators are the rst choice of regular
largely unknown, although atopy and allergen medication in COPD but not in asthma. Thus, in
exposure have major roles [1, 2]. Asthma is a patients with overlapping features, the dierential
phenotypically heterogeneous disorder that, diagnosis between asthma and COPD is impor-
over the years, has been divided into many dif- tant in making the decision to prescribe regular
ferent clinical subtypes. In particular, asthma treatment with either steroids or bronchodilators.
starting in adulthood, asthma in smokers, non-
eosinophilic asthma, and asthma in obese sub-
jects are important subtypes in the adult asthma
population that are still poorly characterized
and that may overlap with COPD [57].
DEFINITIONS OF ASTHMA AND COPD
The dierential diagnosis between asthma
and COPD is quite simple when the typical Asthma is a chronic inammatory disease of the
clinical and functional features of either disease airways clinically characterized by recurrent res-
are present. It is easy to recognize asthma in a piratory symptoms as follows: dyspnea, wheez-
young, atopic, nonsmoking subject with recur- ing, chest tightness, or cough associated with
rent dyspnea, wheezing, or chest tightness and reversible airow limitation. Other important
fully reversible airow limitation. Similarly, it is features of asthma are an exaggerated respon-
easy to diagnose COPD in a subject older than siveness of the airways to various stimuli, and
40, a smoker, who presents with chronic dysp- a specic chronic inammation of the airways
nea, cough, sputum, and xed airow limitation characterized by an increased number of CD4
and no history of asthma or allergic diseases. Th2 lymphocytes, eosinophils, and methacro-
The diculty comes when trying to make a matic cells in the airway mucosa, and increased
diagnosis of asthma or COPD in a middle-aged thickness of the reticular layer of the epithelial

525
Asthma and COPD: Basic Mechanisms and Clinical Management

basement membrane. Familial predisposition, atopy, and TABLE 41.1 Differential diagnosis of asthma.
exposure to allergens and sensitizing agents are important
risk factors for asthma, even though the causes of asthma Localized pathology Inhaled foreign body
the factors responsible for the development of asthma rather Endobronchial tumor
than its exacerbations remain largely undetermined [1]. Vocal cord dysfunction
COPD is a syndrome characterized by poorly revers-
Diffuse airway pathology COPD
ible airow limitation, usually progressive, and often asso-
Eosinophilic bronchitis
ciated with chronic respiratory symptoms, such as dyspnea
Postinfectious airway hyperresponsiveness
and/or chronic cough and sputum [2]. COPD is associated
Cystic fibrosis
with chronic inammation of the airways that is remark-
Bronchiectasis
ably dierent from asthmatic inammation, and that is
characterized by an increased number of CD8 Th1/Tc1 Left ventricular failure
lymphocytes in the airway mucosa and neutrophils in the Other pathologies Gastroesophageal reflux
lumen, with no increased thickness of the reticular layer of Pulmonary embolism
the epithelial basement membrane [813]. Even though Pulmonary eosinophilia
genetic and familial predisposition as well as occupational Drug-induced airway hyperresponsiveness
exposure are considered risk factors, cigarette smoking is by
far the most important risk factor for COPD [2].

Asthma clusters in families and its genetic determi-


nants appear to be linked to those of other allergic IgE-
MINIMUM REQUIREMENTS FOR THE mediated diseases [1921]. Thus, a personal or family
DIAGNOSIS OF ASTHMA OR COPD history of asthma and/or allergic rhinitis, atopic dermatitis,
or eczema increases the likelihood of a diagnosis of asthma.
Physical activity is an important cause of symptoms
The diagnosis of asthma or COPD is based on clinical his- (wheezing and/or cough) for most asthma patients, particu-
tory and lung function tests, particularly peak expiratory larly in children, and for some it is the only cause [15, 16].
ow (PEF) and spirometry, with assessment of spontane- Exercise-induced asthma usually develops not during exer-
ous or postbronchodilator reversibility of airow limitation. cise but 510 min afterward, and it resolves spontaneously
Allergy tests are also usually performed for the diagnosis within 3045 min. Prompt relief of symptoms after the use
of asthma, but not of COPD patients, to identify allergens of inhaled beta2-agonist, or their prevention by pretreat-
responsible for asthma exacerbations and to consider the ment with an inhaled beta2-agonist before exercise, sup-
opportunity to treat the patient with immunotherapy. ports a diagnosis of asthma. Important aspects of personal
history are exposure to agents known to worsen asthma
in the home (heating system, cooking system, house-dust
mites), workplace conditions, air-conditioning, pets, cock-
Symptoms and medical history roaches, environmental tobacco smoke [2226], or even the
general environment (e.g. diesel fumes in trac [27]).
Asthma Since respiratory symptoms of asthma are nonspecic,
Most patients who are diagnosed with asthma seek medical the dierential diagnosis is quite extensive, and the main
attention because of respiratory symptoms. A typical feature goal for the physician is to consider and exclude other pos-
of asthma symptoms is their variability. One or more of the sible diagnoses (Table 41.1). This is even more important if
following symptoms wheezing, chest tightness, cough, and the response to a trial of therapy (i.e. bronchodilators) has
episodic shortness of breath are reported by more than 90% been negative.
of patients with asthma [14]. However, the simple presence Asthma is often classied by severity, but asthma
of these symptoms is not diagnostic, because identical symp- severity changes over time. It also depends not only on the
toms may be triggered by dierent stimuli in nonasthmatics, severity of the underlying disease but also on its respon-
such as in children by acute viral infections [15]. In some siveness to treatment, which becomes the most important
asthmatics, wheezing and chest tightness are absent, and criterion in treated subjects. An asthmatic patient might
the only symptom the patient complains of may be chronic be completely asymptomatic, either because he or she has
cough or cough after exercise [16]. This clinical entity is also mild intermittent asthma and long periods without symp-
called cough-variant asthma; it is particularly common in toms even without treatment, or because he or she has
children and is often more problematic at night [17, 18]. severe asthma and is receiving full anti-asthmatic treatment,
Symptoms of asthma may be triggered or worsened by sev- including systemic steroids.
eral factors, such as exercise, exposure to allergens, viral infec- While respiratory symptoms suggest asthma, the sine
tions, and emotions. Recurrent exacerbations of respiratory qua non for the objective diagnosis of asthma is the pres-
symptoms, worsening of lung function requiring change of ence of reversible airow limitation in subjects with persist-
treatment, unscheduled requests for medical assistance, and ent airways obstruction, and/or airway hyperresponsiveness
sometimes hospitalization are also among the characteristic or increased PEF variability in subjects without airways
clinical features of asthma. obstruction [1].

526
Diagnosis of Asthma and COPD 41
COPD TABLE 41.2 Differential diagnosis of COPD.
A clinical diagnosis of COPD should be considered in any Other airway inflammatory diseases Asthma
patient who has dyspnea, chronic cough, or sputum produc- Bronchiectasis
tion, and/or a history of exposure to risk factors for the dis- Diffuse panbronchiolitis
ease [2]. Most patients who are diagnosed with COPD seek
medical attention because of respiratory symptoms, particu- Infectious airway diseases Tuberculosis
larly dyspnea [28]. Since the early stages COPD may mani-
Cardiac diseases Congestive heart failure
fest as chronic cough and sputum production, they may be
present even in smokers without airow limitation. Cough
and sputum may precede the development of airow limita-
tion: in fact, respiratory symptoms may be an important risk has made it possible to detect and quantify airway abnor-
factor for the development of COPD [29, 30]. Regular pro- malities [38, 39], but more work is required before the tech-
duction of sputum for 3 or more months in 2 consecutive nique can be used in clinical practice.
years is dened as chronic bronchitis [31]. In some subjects, Studies in COPD patients recruited by experienced
chronic cough may be unproductive [3235], and airow pulmonary specialists in hospitals [40] or by general prac-
limitation may develop in the absence of cough. In COPD, titioners in outpatient clinics [41] report that up to 40%
dyspnea is characteristically persistent, unlike in asthma of smokers/ex-smokers with chronic respiratory symptoms
where it is variable and progressive [36]. In the early stages and clinical ndings compatible with COPD do not t
of the disease, dyspnea is noted only during the patients the spirometric denition of COPD reported above. Thus,
usual eort; as lung function decreases, dyspnea becomes even when there is a clear history of COPD chronic res-
more serious and is present during everyday activities or at piratory symptoms, exacerbations, smoking, and age 50
rest. Dyspnea is not closely correlated with arterial blood a large proportion of such patients either have normal
gases; for example, the typical blue bloater with periph- spirometric values or present with reduced lung volumes
eral edema, hypoxemia, and hypercapnia has generally less (a restrictive pattern), and thus the diagnosis of COPD
dyspnea than the pink puer, who generally does not have cannot be conrmed [40, 41]. Clinical features of COPD
these blood gas abnormalities but is much more dyspneic. correlate poorly with airow limitation. Thus, although not
Wheezing and chest tightness are nonspecic symptoms of yet recommended by current guidelines, proper diagno-
COPD and may vary on dierent days or over the course of sis and assessment of the severity of COPD require more
a single day. Recurrent exacerbations of respiratory symp- than a comprehensive approach that includes imaging [38, 39]
toms requiring change of treatment, unscheduled requests assessment of exercise tolerance [42], body mass index [42, 43],
for medical assistance, and sometimes hospitalization are and chronic comorbidities (e.g. chronic heart failure (CHF),
also among the characteristic clinical features of COPD. arterial hypertension, metabolic syndrome) that are often asso-
A detailed medical history of a patient with symp- ciated with COPD [44, 45].
toms suggestive of COPD should include exposure to risk Because chronic respiratory symptoms (particularly
factors (e.g. smoking and occupational or environmental dyspnea), clinical features, and poorly reversible airow lim-
exposures), family history of COPD or other chronic res- itation may also be present in other pathological conditions,
piratory disease, pattern of symptoms, history of exacerba- a careful dierential diagnosis between COPD and these
tions, presence of comorbidities, medical treatment, and the conditions should always be performed (Table 41.2).
patients quality of life.
Diagnosis and assessment of the severity of COPD Physical examination
are mainly based on the degree of airow limitation during
spirometric measurement. Thus, according to current guide- In mild asthma, physical examination is usually normal
lines, the sine qua non for the diagnosis of COPD is the under stable conditions but becomes characteristically
presence of poorly reversible airow limitation, that is, the abnormal during asthma attacks. Typical physical signs of
presence of a postbronchodilator forced expiratory volume asthma attacks are wheezing on auscultation, cough, expira-
in 1 s/forced vital capacity ratio (FEV1/FVC) 0.70 and tory ronchi throughout the chest, and signs of acute hyper-
FEV1 80% predicted. These values conrm the presence ination (e.g. poor diaphragmatic excursion at percussion,
of airow limitation that is not fully reversible [2]. use of accessory muscles of respiration). Some patients, par-
Airow limitation in COPD is due to both small ticularly children, may present with a predominant nonpro-
airways disease (obstructive bronchiolitis) and parenchy- ductive cough (cough-variant asthma). In some asthmatics,
mal destruction (emphysema), the relative contributions of wheezing which usually reects airow limitation may
which vary among patients [1, 2]. In contrast, airow limi- be absent or detectable only on forced expiration, even in
tation in asthma is almost exclusively due to airways dis- the presence of signicant airow limitation; this may be
ease [10]. Imaging (see below), particularly thin-section due to hyperination or to very marked airow limitation.
computed tomography (CT), has been used to quantify In these patients, however, the severity of asthma is mostly
emphysema by detecting areas of low attenuation. However, indicated by other signs, such as cyanosis, drowsiness, dif-
airow limitation as assessed by FEV1 correlates poorly with culty in speaking, tachycardia, hyperinated chest, use of
the severity of emphysema as evaluated by CT [4], possibly accessory muscles, and intercostal recession.
because small airways disease contributes signicantly to In the early stages of COPD, physical examination is
airow limitation [37]. Recent progress in CT technology usually normal. Current smokers may have signs of active

527
Asthma and COPD: Basic Mechanisms and Clinical Management

smoking, including an odor of smoke or nicotine staining of expiratory ow obtained during a forced expiration starting
ngernails. In more severe COPD, prolonged expiration and immediately after a deep inspiration from total lung capac-
wheezing and signs of hyperination (e.g. barrel chest and ity. PEF is a simple, reproducible index and can be measured
poor diaphragmatic excursion at percussion, use of accessory with inexpensive and portable meters [56]. If spirometry
muscles of respiration) are usually present. Cyanosis of the does not reveal airow limitation, the home monitoring of
lips and nail beds and signs of cor pulmonale, such as edema PEF for 24 weeks may help to detect an increased variabil-
of the ankle or lower leg, are often present in patients with ity of airway caliber, and thus help to diagnose asthma [57].
reduced oxyhemoglobin percentage. Even though clubbing For most asthmatic patients, PEF correlates well with FEV1
of the digits may be present in patients with severe COPD [58]. Daily monitoring of PEF (at least in the morning at
and is considered a sign of COPD, it is nonspecic; in fact, awakening and in the evening hours, preferably after bron-
its presence should alert the physician to the possible pres- chodilator inhalation) [1, 3] is also useful to assess the sever-
ence of other diseases, particularly lung cancer. Patients ity of asthma and its response to treatment, and it can help
with COPD often have reduced breath sounds and wheez- patients to detect early signs of asthma deterioration [59].
ing during quiet breathing or after forced expiration. However, PEF measurements have some limitations. PEF
Physical examination is usually not very useful in is eort dependent and mainly reects the caliber of large
making the dierential diagnosis between asthma and airways and may therefore underestimate the degree of air-
COPD, but it can be useful in assessing the severity of ow limitation present in peripheral airways [60]. Diurnal
exacerbations of both asthma and COPD [46]. variability is calculated as follows:
PEFmax  PEFmin 100
Lung function tests PEFmax  PEFmin / 2
A diurnal variability of PEF of more than 20% is
Spirometry diagnostic of asthma, and the magnitude of the variability
Lung function tests play a crucial role in the diagno- is broadly proportional to disease severity. PEF monitoring
sis and follow-up of asthma and COPD. Spirometric may be of use not only in establishing a diagnosis of asthma
measurements FEV1 and slow vital capacity (VC) or and assessing its severity, but also in uncovering an occupa-
FVC are the standard means for assessing airow limita- tional cause for asthma. When used in this way, PEF should
tion. Spirometry is recommended at the time of diagnosis be measured more frequently than twice daily, and special
and for the assessment of the severity of both asthma and attention should be paid to changes occurring in and out of
COPD [1, 2, 47, 48] it should be repeated to monitor the the workplace [55, 6163].
disease and when there is a need for reassessment, such as Even though PEF is at least as important to progno-
during exacerbations [3]. sis as FEV1 in moderate to severe COPD [64], PEF moni-
Poorly reversible airow limitation is indicated by toring is not frequently used in COPD for various reasons.
the absolute reduction of postbronchodilator FEV1/VC or First, PEF reects the patency of central airways, and air-
FEV1/FVC ratios 0.7 but it should be conrmed with ow limitation in COPD starts from peripheral airways.
postbronchodilator FEV1/VC values below the lower limit Thus, the PEF value may underestimate airow limitation,
of normal [4, 49]. Measurements of residual volume and particularly if it occurs in peripheral airways. Second, by
total lung capacity may also be useful in determining the denition, airow limitation is poorly reversible in COPD,
degree of hyperination and/or enlargement of airspaces and thus PEF usually does not vary signicantly. Finally,
[1, 2, 47, 48]. there is only limited evidence to support a role for PEF in
In asthma, airow limitation is usually reversible, detecting COPD exacerbations [65, 66].
either spontaneously or after treatment, except for moderate/
severe asthma with xed airway obstruction [1, 3, 10]. In Reversibility to bronchodilators
COPD, airow limitation is by denition not revers-
The reversibility of airow limitation following broncho-
ible (i.e. FEV1/FVC does not reach 0.7 even after inhala-
dilator therapy is no longer an accepted criterion in support
tion of a bronchodilator or a short course of long-acting
of the diagnosis of asthma, or to establish the dierential
beta2-agonist (LABA) and inhaled steroids). However, up
diagnosis between asthma and COPD [13]; even though
to one-third of COPD patients show a signicant increase
there is a large increase in FEV1, and particularly its return
in FEV1 (15%) after receiving inhaled beta-adrenergic
is above the lower limit of normal values, normal expiratory
agonists [5054], which simulate the reversible airow limi-
ows after anti-asthma treatment strongly suggests asthma.
tation observed in asthmatics.
Subjects with moderate to severe asthma may develop
In conclusion, while the best spirometric values are
poorly reversible airow limitation and have a response
useful to dene whether airow obstruction is reversible or
to treatment but not a return to normal values. Similarly,
not (i.e. does not return within normal values), the degree of
COPD patients may show a signicant response to treat-
reversibility after treatment does not help to make the dif-
ment, even without a return to normal expiratory ows.
ferential diagnosis between asthma and COPD (see below).
In subjects with airow limitation, an improvement
in FEV1 of 1215% predicted and more than 200 ml after
Peak expiratory flow administration of a bronchodilator (e.g. 200 g of inhaled
An important tool for the diagnosis and subsequent treat- salbutamol from a metered dose inhaler) is no longer con-
ment of asthma is the PEF meter [55]. PEF is the highest sidered a pathognomonic hallmark of asthma [1], or a

528
Diagnosis of Asthma and COPD 41
criterion for dierential diagnosis between asthma and important information about the patients lifestyle and
COPD [3, 6769]. In fact, an incomplete response to a sin- occupation, both of which inuence exposure to allergens
gle administration of a bronchodilator does not exclude the and the time and factors possibly involved in onset and in
possibility of reversibility to longer treatment with bron- exacerbations of asthma [84, 85]. In asthmatics, the rela-
chodilators or steroids [70, 71]. Thus, more attention should tionship between exposure to one or more allergens and
be paid to the response to long-term treatment. the occurrence of asthma and/or ocular and nasal symp-
In COPD patients, bronchodilator reversibility test- toms should be established [86]. Also, the relationship of
ing should generally be performed at least once. Airow symptoms to the time of the year (seasonal pollen asthma)
limitation in COPD is usually not reversible, but the one- and to the presence of pets in the home should be assessed,
third of COPD patients who show a signicant response to together with a description of the patients living environ-
bronchodilator agents [50, 51, 53, 54, 72] are likely to ben- ment with special attention to carpets, pillows, and other
et from treatment with glucocorticosteroids [73, 74]. The dust collectors [87]. Identifying the presence of an allergic
absence of a response to a bronchodilator should never be a component in asthma adds little to the diagnosis, but it can
reason to withhold bronchodilator therapy, as the response help in identifying potential triggers and directing allergen
to bronchodilators in COPD is mainly symptomatic rather immunotherapy [1].
than functional [7579]. Bronchodilator responsiveness is a Skin tests with all relevant allergens are present in the
continuous and poorly reproducible variable; thus, classify- geographic area in which the patient lives are the primary
ing patients as responders or nonresponders can be mislead- diagnostic tool in determining allergic status. Deliberate
ing and does not predict disease progression [68]. provocation of the airways with a suspected allergen or sen-
As recommended by recent guidelines [1], the terms sitizing agent may also be helpful in establishing causality,
reversibility and variability should refer to changes in especially in the workplace [63, 88]. Measurement of specic
symptoms accompanied by changes in airow limitation, IgE is not usually more informative than a skin test, and
which occur spontaneously or in response to treatment. is more expensive. Measurement of total IgE in serum has
At present, however, these terms often refer only to rapid no value as a diagnostic test for atopy. The main limitation
improvements in FEV1 measured within minutes after of the allergy test is that a positive test does not necessarily
inhalation of a rapid-acting bronchodilator, for example, mean that the disease is allergic in nature or that it is caus-
after 200400 g salbutamol or salbutamol and ipratropium ing asthma, as some individuals have specic IgE antibodies
bromide [54, 80]. In contrast, a history of symptoms and/ without any symptoms. The costbenet ratio of perform-
or functional reversibility (spontaneous or after any kind of ing inhalation tests with allergens or other sensitizing agents
treatment) is the essential component in the diagnosis of should be carefully examined for each patient because of the
asthma. high cost and the potential risk involved [69].
The assessment of reversibility of both clinical features The assessment of atopy is not useful in COPD.
and functional abnormalities may be useful in obtaining Even though atopy may be a risk factor for both asthma
the best level of asthma control achievable and/or the best and COPD [89, 90], the demonstration of atopy in COPD
lung function for individual patients [81]. Achieving and patients does not help in the identication of potential trig-
maintaining lung function at the best possible level is one gers as in allergic asthma. Allergen immunotherapy has no
of the objectives of both asthma and COPD management role in COPD.
[1, 81, 82].
In summary, while the best spirometric values are use-
ful to dene whether airow obstruction is reversible or not
(i.e. does not return within normal values), the degree of
reversibility after treatment does not help to make the dif-
ADDITIONAL TESTS
ferential diagnosis between asthma and COPD (see below).
While the diagnosis and assessment of severity of asthma
and COPD can be fully established on the basis of clini-
Arterial blood gases cal history and lung function tests (including arterial blood
gases, see below), additional tests might be helpful to better
In severe asthma and COPD and, more importantly, during characterize individual patients.
acute exacerbations of both asthma and COPD, the meas-
urement of arterial blood gases while the patient is breath-
ing air and/or after oxygen administration is essential for Reversibility to corticosteroids
the diagnosis of chronic and/or acute respiratory failure.
This test should be performed in all patients with clinical In patients with airow limitation that is not reversed by a
signs of acute or chronic respiratory and/or heart failure [2]. single dose of a short-acting bronchodilator, a 2-week treat-
ment with oral or inhaled glucocorticosteroids and bron-
chodilators might be considered. Glucocorticosteroids can
Allergy tests be administered orally (e.g. 40 mg daily prednisone) by aero-
sol (e.g. 2 mg daily beclomethasone, or equivalent) or both
The presence of allergic disorders in a patients family his- [9193] for at least 14 days [94]. Unfortunately, patients
tory should be investigated in all patients in whom symp- with COPD cannot be separated into discrete groups of
toms are suggestive of asthma [83]. A history provides glucocorticosteroid responders and nonresponders, and

529
Asthma and COPD: Basic Mechanisms and Clinical Management

thus glucocorticosteroid testing is an unreliable predictor of of exercise-induced hypoxemia, and response to treatment in
the benet from inhaled glucocorticosteroids in individual individuals with COPD. Simple walking tests are increas-
patients. ingly used in the assessment of COPD patients [111].
Because of their ecacy and infrequent adverse Severe COPD might be better assessed by a composite
eects, inhaled glucocorticosteroids alone or in combina- score such as BODE (body mass index, degree of airway
tion with long-acting bronchodilators are increasingly used obstruction, severity of dyspnea, and exercise tolerance),
in practice as rst-choice therapy to investigate the revers- which has been shown to be a better predictor of subse-
ibility of airow limitation [74, 95, 96]. quent survival and is increasingly used in clinical assessment
In most patients with a clear history of asthma or of patients.
COPD, the reversibility to glucocorticosteroids conrms
the diagnosis, even if signicant overlap exists [10]. Asthma
is usually responsive to bronchodilators and/or glucocortico- Diffusion capacity
steroids, whereas COPD is usually less responsive or not
responsive at all. In asthma, a combination of sputum eosi- Measurement of the diusing capacity of the lung for car-
nophilia and increased nitric oxide (NO) levels may be useful bon monoxide (Dlco) has been recommended for distin-
in predicting the response to a trial of oral steroids [97, 98], guishing asthma from COPD [112]. In asthma, Dlco is
and sputum eosinophilia may also predict the response to usually normal or increased [10, 113, 114]. In contrast,
steroids in COPD [99, 100]. However, some COPD patients Dlco is usually reduced in COPD, possibly due to emphy-
may show a signicant improvement in function after gluco- sema [10, 114, 115], but it may also be reduced in smokers
corticosteroid treatment [73], particularly if they present with without airow limitation [116]. Dlco is lower in COPD
pathological abnormalities similar to those in asthma. patients than in asthmatics with incomplete reversible air-
The simplest and potentially the safest way of identi- ow limitation [10, 117, 118]. However, patients with
fying these COPD patients is by an ex juvantibus treatment severe alpha-1 antitrypsin deciency may present with nor-
trial with inhaled glucocorticosteroids [92] in combination mal Dlco, despite having a signicant component of xed
with long-acting bronchodilators for 6 weeks to 3 months, airway obstruction and prominent panacinar emphysema on
using the same criteria for reversibility as in the bronchodila- a high-resolution CT (HRCT) scan, suggesting the limita-
tor trial (FEV1 increase of 200 ml and 12%) [2, 73, 92, 101]. tions of measuring Dlco in these patients [119, 120].
The response to glucocorticosteroids alone or in combina-
tion with long-acting bronchodilators should be evaluated
with respect to the postbronchodilator FEV1 [2, 4]. Airway hyperresponsiveness
COPD patients with a response to glucocortico-
steroids present some pathological features of asthma, such In patients who have symptoms consistent with asthma
as a signicantly higher number of eosinophils and higher but have normal lung function, bronchial provocation tests
levels of eosinophil cationic protein in their bronchoalveo- (methacholine, histamine, adenosine 5
-monophosphate,
lar lavage uid, and a thicker reticular basement membrane mannitol, and exercise) are helpful in measuring airway
[73, 101]. hyperresponsiveness and thereby conrming or excluding
All long-term studies in COPD have demonstrated the diagnosis of active asthma [121, 122]. Methacholine is
the lack of any eect of inhaled glucocorticosteroids on the mainly used to identify bronchial hyperresponsiveness and
natural history of COPD, as evaluated by the decline in to guide treatment. Exercise is used as a bronchial provo-
FEV1 [2, 102106]. Therefore, given the documented risks cation test because demonstrating prevention of exercise-
of chronic glucocorticosteroid therapy in both asthma [107] induced asthma is an indication for use of a drug [121].
and COPD, such as osteoporosis [102, 108], the decision to These measurements are sensitive for a diagnosis of
start long-term treatment with inhaled glucocorticosteroids asthma, but they have low specicity [123]. This means that
must be made very carefully. a negative test can be used to exclude a diagnosis of active
In patients with poorly reversible airow limita- asthma, but a positive test does not always mean that a
tion due to asthma, the benecial eects of inhaled gluco- patient has asthma [123]. Airway hyperresponsiveness has
corticosteroids are likely to overcome the risks of negative been described in workers who are acutely exposed to irri-
systemic eects [74]. However, in patients with poorly tants [124, 125], allergic rhinitis [126128], and other dis-
reversible airow limitation due to smoking, this may not be eases with airow limitation, such as cystic brosis [129131]
the case, particularly considering that inhaled glucocorticos- and COPD [132134]. Indeed COPD, especially in current
teroid alone may be associated with increased mortality [109], smokers, is often accompanied by airway hyperresponsiveness
and that inhaled glucocorticosteroid both alone and in com- [135] that is no dierent from that in asthmatics with a sim-
bination with a long-acting beta2-adrenergic agonist may ilar degree of airow limitation [10, 136]. In patients with
slightly increase the risk of pneumonia in COPD patients xed airow limitation, a similar degree of airway hyperre-
[109, 110]. sponsiveness was observed in those with a history of COPD
and those with a history of asthma [10]. In these patients,
hyperresponsiveness might be largely due to the airow limi-
Exercise testing tation itself.
In conclusion, the measurement of airway hyper-
Exercise testing is useful for assessing the degree of disabil- responsiveness may be useful to conrm asthma in subjects
ity, the role of comorbidities, prognosis for survival, presence with normal baseline lung function, but it is not useful in

530
Diagnosis of Asthma and COPD 41
the dierential diagnosis between asthma and COPD, TABLE 41.3 HRCT features in asthma and COPD.
particularly when patients have a similar degree of poorly
reversible airow limitation. Mild-to-
persistent Severe Healthy
asthma asthma COPD subjects
Imaging
Bronchial wall    
thickening
While chest radiography may be useful to exclude diseases
that may mimic asthma and COPD, it is not required in the Emphysema   
conrmation of the diagnosis and management of asthma.
The utility of chest radiography is to exclude other condi- Bronchiectasis /   /
tions that may imitate or complicate asthma, particularly
acute asthma. Examples include pneumonia, cardiogenic
pulmonary edema, pulmonary thromboembolism, tumors
(especially those that result in airway obstruction with Scintigraphic approaches may be used to assess COPD
resulting peripheral atelectasis), and pneumothorax [137]. or emphysema and to provide functional imaging. Ultrane
A number of novel imaging methods for assessing 133
Xe gas particles are being used for ventilation scintig-
airway pathology in asthmatic patients have been proposed raphy, including single photon emission CT (SPECT).
[39, 138]. Both direct and indirect signs of airway pathology SPECT imaging has been shown to be more useful than
have been described using HRCT. Direct signs are obtained morphologic HRCT in the evaluation of small airways dis-
by measuring airway or bronchial wall thickness, evaluat- ease, including pulmonary emphysema [147]. Diusion-
ing the ratio of bronchial diameters to adjacent pulmo- weighted, hyperpolarized 3He magnetic resonance imaging
nary arteries, and identifying a lack of bronchial tapering. has been shown to correlate with pulmonary function tests,
Indirect signs include foci of mucoid impaction (including particularly Dlco [148]. Also, dynamic contrast-enhanced
the nding of a tree-in-bud conguration of small periph- magnetic resonance imaging may detect abnormali-
eral lung nodules indicative of bronchiolitis) and mosaic ties of the pulmonary peripheral microvasculature [149].
attenuation [139]. Mosaic attenuation is the presence of These techniques might be useful in the assessment of pul-
geographic zones of decreased lung density adjacent to monary emphysema.
areas of apparent increased lung density in the absence of The clinical application of HRCT is mandatory
architectural distortion or honeycombing. This nding may inCOPD patients who are candidates for volume reduction
also be seen in patients with diuse inltrative lung disease surgery and in whom the regional distribution of emphy-
or, more rarely, chronic embolic pulmonary hypertension. sema, particularly upper lobe emphysema, is critical for
In most cases, reliable identication of air trapping result- the outcome of the intervention [150152]. HRCT fea-
ing in mosaic lung attenuation requires expiratory imaging. tures of asthma and COPD [39, 153156] are reported in
During exhalation, areas of air trapping become accentu- Table 41.3.
ated, simplifying the dierential diagnosis. Each of these
methods has serious limitations, and in clinical practice they
have not yet proved suciently accurate to warrant their use Laboratory examinations
in diagnosing most cases in which extensive airway remod-
eling has occurred [140]. Circulating eosinophils: circulating eosinophilia is a
Airow limitation in COPD is due to both small air- feature of many dierent lung diseases. In some condi-
ways disease (obstructive bronchiolitis) and parenchymal tions, eosinophils are increased in the blood but not in the
destruction (emphysema), the relative contributions of which lung tissue; in other diseases, there may be signicant eosi-
vary among patients [141, 142]. Thin-section CT has been nophilia in the lung tissue but not in the peripheral blood.
used to quantify emphysema by detecting low-attenuation In others, there may be lung eosinophilia without any radi-
areas, and the role of CT in diagnosing emphysema is well ographic evidence of disease, as in asthma. ChurgStrauss
established. However, airow limitation evaluated by FEV1 syndrome (CSS) is characterized by peripheral and pul-
does not show a good correlation with the severity of emphy- monary eosinophilia with inltrates on chest radiograph.
sema as evaluated by CT [143, 144], because small airways However, the primary features that distinguish CSS from
disease appears to contribute signicantly to airow limita- other pulmonary eosinophilic syndromes are the presence
tion [145]. Recent progress in CT technology has made it of eosinophilic vasculitis in the setting of asthma and the
possible to detect and quantify airway abnormalities [146]. involvement of multiple end organs. Therefore, in these
Theoretically, thin-section CT can depict the dimensions of cases it is important to establish a dierential diagnosis
airways as small as 1 to 2 mm in inner diameter, suggest- between these diseases, which may occur without any radio-
ing that CT can be used to evaluate airway dimensions in graphic evidence of disease. Although perceived to be quite
a variety of diseases [141, 142]. Hasegawa and colleagues rare, the incidence of this disease seems to have increased
[38], who developed new software for measuring airway in the last few years, particularly in association with various
dimension using curved multiplanar reconstruction, demon- asthma therapies.
strated that airway luminal area and wall area signicantly Clinical presentation of COPD exacerbation includes
correlated with FEV1 (% predicted). The correlation coef- worsening of dyspnea, increased cough and sputum, and
cients improved as the airways became smaller. changes in the aspect of expectorations.

531
Asthma and COPD: Basic Mechanisms and Clinical Management

Biomarkers of respiratory bacterial function or troponin, and is proportional to the magnitude of


infections NT-proBNP release, with higher risk observed among those
with a more marked elevation of the marker. An elevated ini-
Although clinical criteria are still used to determine which tial NT-proBNP concentration should prompt consideration
patient should be treated with antibiotics [157], these cri- of an early invasive management approach. Consideration
teria are neither sensitive nor specic enough to exclude should be given to repeating the NT-proBNP measure-
other causes of exacerbation of respiratory symptoms in ment after 2472 h and again at 36 months because these
these patients. Novel biomarkers (e.g. pro-calcitonin) have follow-up measurements provide more long-term prognostic
been recommended for guiding antibiotic treatment both in information than single measures at presentation. In acute
exacerbations of COPD and pneumonia, but these studies ischemic heart disease, an NT-proBNP value 250 ng/l is
need conrmation [158160]. associated with an adverse prognosis. In patients with sta-
Other frequent clinical conditions may mimic the ble coronary artery disease, measurement may be performed
symptoms of COPD exacerbation, including congestive for prognostication purposes at 6- to 18-month intervals. In
heart failure, pneumonia, pneumothorax, pleural eusion the case of clinical suspicion of disease progression, a new
and pulmonary embolism [2]. However, COPD is often sample may be warranted.
cited among the risk factors for acute venous thrombo-
embolism and is an independent predictor of pulmonary Alpha-1 antitrypsin
embolism [161]. In a small series of patients, the prevalence
of deep vein thrombosis in patients admitted with acute Severe hereditary deciency of alpha-1 antitrypsin is usually
exacerbation of COPD was 31% [162, 163]. Similarly, on associated with early-onset panacinar emphysema [172].
the basis of ventilationperfusion lung scintigraphy, the Thus, in patients who develop COPD before the age of 45
prevalence of pulmonary embolism in patients admitted and/or who have a strong family history of COPD, alpha-1
with acute exacerbation of COPD was as high as 20% [164]. antitrypsin should be measured; if the serum concentra-
More recently, Tillie-Leblond et al. [165] explored the tion is 1520% of the normal value, the patient should be
prevalence of pulmonary embolism in a cohort of patients considered for alpha-1 antitrypsin augmentation therapy.
with COPD with unexplained dyspnea and found a rate Like other genetic tests, this test has no clinical value in
of 25% in this population. D-dimer is a product of lysis of asthma [21].
stabilized brin-clot that is considered an indirect marker
of coagulation activation. Measurement of plasma D-dimer
has a well established diagnostic role in acute pulmonary
Assessment of airway inflammation
embolism because of its high negative predictive value
Bronchopulmonary inammation is markedly dierent in
[166]. The usefulness of D-dimer testing remains contro-
asthma and COPD [9, 12, 173182] (Fig. 41.1).
versial in inpatients, in part due to a high percentage of
While airway biopsies and bronchoalveolar lavage
positive D-dimer values among them, which is consequent
clearly distinguish between asthma and COPD in subjects
to a broad spectrum of diseases (other than pulmonary
with overlapping features that may provide useful informa-
embolism) and procedures related to the hospitalization.
tion in research protocols, they are considered too invasive
Moreover, for inpatients, a negative D-dimer reduces sus-
for the diagnosis or staging of either asthma or COPD
picion but its sensitivity is only 89%, unsatisfactory to
[179, 183, 184]. In contrast, noninvasive markers of airway
exclude pulmonary embolism [167]. If the helical contrast
inammation have been increasingly used in research pro-
CT angiogram is negative and ultrasound is negative, there
tocols to dierentiate asthma from COPD [183, 185].
is still a 5% false-negative rate for inpatients [168]. There
are no convincing data regarding a negative CT alone for Sputum
inpatients. Accordingly, CT and D-dimer evidence may add
Sputum induction has been widely used in the study of
information but conventional pulmonary arteriography may
airway inammation in asthma and COPD because it is
still be required to make a secure diagnosis. In its absence,
a safe, reproducible, and noninvasive technique that can
a clinical decision to treat (and suspend treatment, if
be used repeatedly, even during exacerbations [186188].
contraindications supervene) may be required [169].
Sputum ndings mainly represent the bronchial compart-
ment. Induced sputum from asthmatic patients during
Troponin and/or N-BNP stable conditions is usually characterized by a higher per-
centage of eosinophils and metachromatic cells than that
COPD and CHF are common conditions. The diag- found in samples from healthy subjects [189, 190]. Sputum
nosis of CHF can remain unsuspected in patients with neutrophilia may also be present across the range of disease
COPD, because shortness of breath is attributed to COPD. severity; its identication is important, as it is associated
Measurement of plasma B-type natriuretic peptide (BNP) with a poor response to glucocorticosteroids [191193].
levels helps to uncover unsuspected CHF in patients with In stable conditions, ex- or current smokers with
COPD and clinical deterioration [170]. Amino-terminal COPD characteristically show an increased total cell number
pro-B-type natriuretic peptides (NT-proBNP) are strong in spontaneous or induced sputum, with a predominance of
and independent prognostic indicators, representing a par- neutrophils and a small percentage of eosinophils in some
ticularly strong predictor of heart failure or death [171]. This subjects [133, 194, 195]. In some smokers with chronic
risk is independent of all other variables, including renal bronchitis, with or without chronic airow limitation,

532
Diagnosis of Asthma and COPD 41

FIG. 41.1 Photomicrographs showing


bronchial biopsy specimens immunostained
with anti-EG-2 (eosinophil cationic protein)
from a patient with fixed airflow obstruction
and a history of COPD (A) and from a patient
with fixed airflow obstruction and a history
of asthma (B). The two patients had a similar
degree of fixed airflow obstruction. In (B),
there is prominent eosinophilia beneath the
destroyed epithelium that is not present in (A).
Photomicrographs showing bronchial biopsy
specimens stained with H&E from a patient
with fixed airflow obstruction and a history of
COPD (C) and from a patient with fixed airflow
obstruction and a history of asthma (D). The two
patients had a similar degree of fixed airflow
obstruction. In (D), there is a thicker reticular
layer of the epithelial basement membrane
compared with (C) (from Ref. [10]).

an excess proportion of eosinophils (3%) in lower respi- TABLE 41.4 History, symptoms, and results of pulmonary function tests in
ratory secretions (called eosinophilic bronchitis) can also the differential diagnosis between asthma and COPD.
occur [100, 186, 187]. A recent study has elegantly shown
that in patients with COPD the main cells in sputum are Asthma COPD
neutrophils and macrophages, with more macrophages Onset Mainly in childhood In mid to late adult life
and eosinophils than in a sub-phenotype of patients with
chronic bronchitis [196]. Smoking Usually non-smokers Almost invariably
Analysis of sputum from patients with exacerbations smokers
of asthma or COPD has provided interesting new informa-
tion. Mild exacerbations of asthma induced by tapering the Chronic cough and Absent Frequent (chronic
dose of inhaled steroids are associated with sputum eosi- sputum bronchitis)
nophilia [197, 198]. In contrast, mild exacerbations that are Dyspnoea on effort Variable and reversible Constant, poorly
spontaneous are associated with eosinophilia in about 50% to treatment reversible and
of subjects, but the other 50% do not have sputum eosi- progressive
nophilia [199]. In children, eosinophilic airway inamma-
tion is associated with deteriorating asthma over time. This is Nocturnal symptoms Relatively common Uncommon
consistent with the hypothesis that airway inammation has
Airflow limitation Increased diurnal Normal diurnal
an adverse eect on the prognosis of childhood asthma and
variability variability
suggests a role for monitoring inammation in asthma man-
agement [200]. Severe asthma exacerbations are associated Response to Good Poor
with more prominent sputum neutrophilia [201]. Bronchial bronchodilator
neutrophilia has also been observed in bronchial lavage uid
from asthmatics during status asthmaticus [202]. Airway In most patients, with In most patients with
Interestingly, exacerbations of chronic bronchitis or hyperresponsiveness or without airflow airflow limitation
COPD are associated with quite similar changes in spu- limitation
tum cell count. Mild exacerbations of chronic bronchitis or
COPD are associated with eosinophilia in sputum and in
biopsy specimens [203, 204], whereas severe exacerbations making a dierential diagnosis in those few cases in which
of COPD are associated with sputum neutrophilia [205]. clinical ndings are not denitive.
Thus, at least in sputum, the changes in inammatory Several biochemical markers have been studied in
cells during exacerbations may be no dierent in asthma induced sputum from both asthma and COPD patients
and COPD. Once again, this evidence underlines the [206210] (Table 41.4). Although some markers are
similarities between the two diseases and the diculty in markedly dierent in asthma and COPD, and studying

533
Asthma and COPD: Basic Mechanisms and Clinical Management

NO EBC Other
Exhaled
breath VOC
analysis E-BreathT
E-Nose
BBF
Tests

Standardization
Completed FENO FENO
In progress MEFENO, nasal NO
Not done

Measured at Clinical lab GP, home (hand-held) Clinical lab GP, home (portable) Lab

Ready to be used in

Clinical research
FENO, MEFENO
Nasal NO
Clinical practice
FENO
General practice Nasal NO (?)

Home (by patients)

FIG. 41.2 Exhaled breath analysis: current state of standardization, research, and clinical use (from Ref. [212]).

these markers may provide useful information in research TABLE 41.5 Ancillary tests in the differential diagnosis between stable
protocols, their use in clinical practice has not been shown asthma and COPD.
to be superior to simple cell counts.
Ancillary test Asthma COPD

Exhaled NO Reversibility to Usually present Usually absent


bronchodilator and/or
While endogenous NO may be involved in the pathophysi-
glucocorticosteroids
ology of asthma and COPD [211, 212] (Fig. 41.2), exhaled
NO is increased in atopic asthma [213217] but less at lesser Lung volumes
extent so in nonatopic asthma [218, 219]. Furthermore, Residual volume, total Usually normal Usually irreversibly
exhaled NO is reduced by glucocorticosteroids [220] but lung capacity or, if increased, increased
not by bronchodilators [221]. Conicting results have been reversible
obtained in COPD [222228]. Diffusion capacity Normal Decreased
In patients with stable COPD, a partial bronchodi-
lator response to inhaled salbutamol is associated with Airway hyperresponsiveness Increased Usually not
increased exhaled NO and sputum eosinophilia [101]. measurable due to
Taken together with previous ndings [73], this suggests airflow limitation
that there is a subset of patients with COPD who share Allergy tests Often positive Often negative
some characteristics of asthmatic inammation and who
may be responsive to steroids [100]. Imaging of the chest Usually normal Usually abnormal

Sputum Eosinophilia Neutrophilia

Exhaled NO Increased Usually normal

DIFFERENTIAL DIAGNOSIS BETWEEN ASTHMA


AND COPD a nearly complete reversibility of airow limitation may
help to conrm a diagnosis of asthma, and those that show
In most patients, the clinical presentation and particularly poorly reversible airow limitation may help to conrm the
the history provide the strongest diagnostic criteria to dis- diagnosis of COPD (Table 41.5).
tinguish asthma from COPD (Table 41.5). Results of pul- Dierential diagnosis between asthma and COPD
monary function tests, particularly spirometry, that show becomes more dicult in elderly patients, in whom some

534
Diagnosis of Asthma and COPD 41
features may overlap, such as smoking and atopy, and, more in patients with COPD is associated with increased in-
importantly, when the patient develops poorly reversible air- hospital mortality [239]. Comorbid conditions that have
ow limitation that responds only partially to treatment. In been associated in particular with an increased mortality risk
these cases, symptoms, lung function, airway responsiveness, in COPD patients include chronic renal failure, cor pulmo-
imaging, and even pathological ndings may overlap and nale [244], and pulmonary vascular disease [245]. Underlying
thus may not provide solid information for the dierential heart diseases have not been consistently associated with a
diagnosis. Because the dierential diagnosis mainly aims to higher mortality risk. However, since COPD is frequently
provide better treatment, it is important in these cases to underreported, it is dicult to make an accurate estimate
undertake an individual approach and to perform additional of how comorbid conditions inuence COPD mortal-
tests. Reversibility to corticosteroids alone or in combina- ity or, conversely, how COPD aects the outcome of other
tion with long-acting bronchodilators, measurements of diagnoses [242].
lung volumes and diusion capacity, analysis of sputum and The complexity of chronic comorbidities applies
exhaled NO, and imaging of the chest may demonstrate to acute exacerbations of asthma and COPD to a similar
whether asthma or COPD is the predominant cause of extent. Acute exacerbations of respiratory symptoms may be
airow limitation (Table 41.5). In contrast, reversibility to present in several other acute conditions that should always
bronchodilator and assessment of airway hyperresponsive- be carefully considered and excluded, such as acute left ven-
ness or skin testing may not be useful in these patients. tricular failure, pulmonary thromboembolism, pneumonia,
metabolic acidosis, and anemia.

COMORBIDITIES OF ASTHMA AND COPD


ACKNOWLEDGMENTS
The coexistence of chronic rhinitis, nasal polyposis, and
sinusitis may contribute to the severity of asthma [229, 230]. We thank M. McKenney for scientic assistance with the
There is broad evidence to show that adequate treatment manuscript and E. Veratelli for her scientic secretarial
of these upper airway diseases is benecial to asthma by assistance.
mechanisms that are not clearly understood. The one Supported by MURST (Grants 60% and 40%),
airway concept developed by the WHO ARIA Group Consorzio Ferrara Ricerche (CFR), Associazione per la
[231] has drawn attention to the importance of treat- Cura e la Ricerca dellAsma (ARCA), Associazione per lo
ing the whole respiratory tract while managing asthma. Studio dei Tumori e delle Malattie Polmonari (ASTMP).
Gastroesophageal reux is also occasionally associated with
asthma, both in adults and in children [232], but treatment
of reux usually has little overall eect on mild to moderate References
asthma [233]. Two recent studies [234, 235] indicate that
proton-pump inhibitors in patients with symptomatic reux 1. Global Strategy for Asthma Management and Prevention. Global
Initiative for Asthma (GINA), 2007; Available from: http://www.
improve asthma control in severe disease. A frequent and
ginasthma.org
quite important comorbidity of asthma in adults is COPD, 2. Global Strategy for the Diagnosis, Management and Prevention of
most likely due to smoking, which is quite common in asth- COPD. Global Initiative for Chronic Obstructive Lung Disease
matics. Smoking modies the airway pathology of asth- (GOLD), 2007; Available from: http://www.goldcopd.org
matics to a COPD-like pattern and reduces the response 3. National Heart, Lung, and Blood Institute. National Asthma
to treatment [7]. Comorbidities may become important in Education and Prevention Program. Expert Panel Report 3. Guidelines
severe asthmatics, whereas overall they play a less important for the Diagnosis and Management of Asthma, 2007, Publication
role overall in the clinical manifestations of mild to moder- No. 07-4051.
ate asthma [236, 237]. 4. Celli BR, MacNee W. Standards for the diagnosis and treatment of
Recent research suggests that inammation in COPD patients with COPD: A summary of the ATS/ERS position paper. Eur
Respir J 23(6): 93246, 2004.
is not conned to the lungs, because the main risk factor,
5. Bel EH. Clinical phenotypes of asthma. Curr Opin Pulm Med 10(1):
smoking, may simultaneously cause pulmonary and sys- 4450, 2004.
temic inammation. This may account for the observation 6. Beuther DA, Weiss ST, Sutherland ER. Obesity and asthma. Am J
that patients with COPD often present with one or more Respir Crit Care Med 174(2): 11219, 2006.
comorbid conditions. The most common comorbidities that 7. Thomson NC. Smokers with asthma: What are the management
have been described in association with COPD are hyper- options? Am J Respir Crit Care Med 175(8): 74950, 2007.
tension, diabetes, coronary artery disease [238, 239], chronic 8. Baraldo S, Lokar Oliani K, Turato G, Zuin R, Saetta M. The role of
heart failure [240], pulmonary infections, cancer [241], and lymphocytes in the pathogenesis of asthma and COPD. Curr Med
pulmonary vascular disease [242, 243]. Chem 14(21): 225056, 2007.
Comorbidities are highly likely to aect health out- 9. Curtis JL, Freeman CM, Hogg JC. The immunopathogenesis of
chronic obstructive pulmonary disease: Insights from recent research.
comes in COPD. Progressive respiratory failure accounts
Proc Am Thorac Soc 4(7): 51221, 2007.
for only about one-third of COPD-related deaths: COPD 10. Fabbri LM, Romagnoli M, Corbetta L, Casoni G, Busljetic K,
patients are more likely to die of cardiovascular complica- Turato G, Ligabue G, Ciaccia A, Saetta M, Papi A. Dierences in
tions or cancer than of respiratory failure [242]. Therefore, airway inammation in patients with xed airow obstruction due to
factors other than the progression of lung disease must play asthma or chronic obstructive pulmonary disease. Am J Respir Crit Care
a substantial role. The number of preexisting comorbidities Med 167(3): 41824, 2003.

535
Asthma and COPD: Basic Mechanisms and Clinical Management

11. Hogg JC. Pathophysiology of airow limitation in chronic obstructive 32. Burrows B, Knudson RJ, Cline MG, Lebowitz MD. Quantitative rela-
pulmonary disease. Lancet 364(9435): 70921, 2004. tionships between cigarette smoking and ventilatory function. Am Rev
12. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Respir Dis 115(2): 195205, 1977.
Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The 33. Choudry NB, Fuller RW. Sensitivity of the cough reex in patients
nature of small-airway obstruction in chronic obstructive pulmonary with chronic cough. Eur Respir J 5(3): 296300, 1992.
disease. N Engl J Med 350(26): 264553, 2004. 34. Clark KD, Wardrobe-Wong N, Elliott JJ, Gill PT, Tait NP, Snashall
13. MacNee W. Pathogenesis of chronic obstructive pulmonary disease. PD. Cigarette smoke inhalation and lung damage in smoking volun-
Proc Am Thorac Soc 2(4): 25866, discussion 29091, 2005. teers. Eur Respir J 12(2): 39599, 1998.
14. Elliott MW, Adams L, Cockcroft A, MacRae KD, Murphy K, Guz A. 35. Smith J, Woodcock A. Cough and its importance in COPD. Int J
The language of breathlessness. Use of verbal descriptors by patients Chron Obstruct Pulmon Dis 1(3): 30514, 2006.
with cardiopulmonary disease. Am Rev Respir Dis 144(4): 82632, 36. Buist AS. Similarities and dierences between asthma and chronic
1991. obstructive pulmonary disease: Treatment and early outcomes. Eur
15. Townshend J, Hails S, McKean M. Diagnosis of asthma in children. Respir J Suppl 39: 30s35s, 2003.
BMJ 335(7612): 198202, 2007. 37. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P,
16. McFadden ER. Exertional dyspnea and cough as preludes to acute Fukuchi Y, Jenkins C, Rodriguez-Roisin R, van Weel C, Zielinski J.
attacks of bronchial asthma. N Engl J Med 292(11): 55559, 1975. Global strategy for the diagnosis, management, and prevention of
17. Dicpinigaitis PV. Chronic cough due to asthma: ACCP evidence-based chronic obstructive pulmonary disease: GOLD executive summary. Am
clinical practice guidelines. Chest 129(1 Suppl): 75S79S, 2006. J Respir Crit Care Med 176(6): 53255, 2007.
18. Pavord ID. Cough and asthma. Pulm Pharmacol Ther 17(6): 399402, 38. Hasegawa M, Nasuhara Y, Onodera Y, Makita H, Nagai K, Fuke S,
2004. Ito Y, Betsuyaku T, Nishimura M. Airow limitation and airway
19. Burrows B, Martinez FD, Cline MG, Lebowitz MD. The relationship dimensions in chronic obstructive pulmonary disease. Am J Respir Crit
between parental and childrens serum IgE and asthma. Am J Respir Care Med 173(12): 130915, 2006.
Crit Care Med 152(5 Pt 1): 1497500, 1995. 39. Mitsunobu F, Tanizaki Y. The use of computed tomography to assess
20. Burrows B, Martinez FD, Halonen M, Barbee RA, Cline MG. asthma severity. Curr Opin Allergy Clin Immunol 5(1): 8590, 2005.
Association of asthma with serum IgE levels and skin-test reactivity to 40. Kohler D, Fischer J, Raschke F, Schonhofer B. Usefulness of GOLD
allergens. N Engl J Med 320(5): 27177, 1989. classication of COPD severity. Thorax 58(9): 825, 2003.
21. Zhang J, Pare PD, Sandford AJ. Recent advances in asthma genetics. 41. Rutten FH, Moons KG, Cramer MJ, Grobbee DE, Zuitho NP,
Respir Res 9: 4, 2008. Lammers JW, Hoes AW. Recognising heart failure in elderly patients
22. Chauhan AJ, Krishna MT, Frew AJ, Holgate ST. Exposure to nitro- with stable chronic obstructive pulmonary disease in primary care:
gen dioxide (NO2) and respiratory disease risk. Rev Environ Health Cross sectional diagnostic study. BMJ 331(7529): 1379, 2005.
13(12): 7390, 1998. 42. Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca M,
23. Garrett MH, Hooper BM, Hooper MA. Indoor environmental factors Mendez RA, Pinto Plata V, Cabral HJ. The body-mass index, airow
associated with house-dust-mite allergen (Der p 1) levels in south- obstruction, dyspnea, and exercise capacity index in chronic obstructive
eastern Australian houses. Allergy 53(11): 106065, 1998. pulmonary disease. N Engl J Med 350(10): 100512, 2004.
24. Garrett MH, Hooper MA, Hooper BM, Abramson MJ. Respiratory 43. Ischaki E, Papatheodorou G, Gaki E, Papa I, Koulouris N, Loukides S.
symptoms in children and indoor exposure to nitrogen dioxide and gas Body mass and fat-free mass indices in COPD: Relation with variables
stoves. Am J Respir Crit Care Med 158(3): 89195, 1998. expressing disease severity. Chest 132(1): 16469, 2007.
25. Garrett MH, Rayment PR, Hooper MA, Abramson MJ, Hooper BM. 44. Fabbri LM, Luppi F, Beghe B, Rabe KF. Complex chronic
Indoor airborne fungal spores, house dampness and associations with comorbidities of COPD. Eur Respir J 31(1): 20412, 2008.
environmental factors and respiratory health in children. Clin Exp 45. Fabbri LM, Rabe KF. From COPD to chronic systemic inammatory
Allergy 28(4): 45967, 1998. syndrome? Lancet 370(9589): 79799, 2007.
26. Jarvis D, Chinn S, Luczynska C, Burney P. Association of respiratory 46. Flaherty KR, Kazerooni EA, Martinez FJ. Dierential diagnosis of
symptoms and lung function in young adults with use of domestic gas chronic airow obstruction. J Asthma 37(3): 20123, 2000.
appliances. Lancet 347(8999): 42631, 1996. 47. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R,
27. McCreanor J, Cullinan P, Nieuwenhuijsen MJ, Stewart-Evans J, Coates A, Crapo R, Enright P, van der Grinten CP, Gustafsson P,
Malliarou E, Jarup L, Harrington R, Svartengren M, Han IK, Ohman- Jensen R, Johnson DC, MacIntyre N, McKay R, Navajas D,
Strickland P, Chung KF, Zhang J. Respiratory eects of exposure to Pedersen OF, Pellegrino R, Viegi G, Wanger J. Standardisation of
diesel trac in persons with asthma. N Engl J Med 357(23): 234858, spirometry. Eur Respir J 26(2): 31938, 2005.
2007. 48. Wanger J, Clausen JL, Coates A, Pedersen OF, Brusasco V, Burgos F,
28. ODonnell DE, Banzett RB, Carrieri-Kohlman V, Casaburi R, Casaburi R, Crapo R, Enright P, van der Grinten CP, Gustafsson P,
Davenport PW, Gandevia SC, Gelb AF, Mahler DA, Webb KA. Hankinson J, Jensen R, Johnson D, Macintyre N, McKay R, Miller
Pathophysiology of dyspnea in chronic obstructive pulmonary disease: MR, Navajas D, Pellegrino R, Viegi G. Standardisation of the meas-
A roundtable. Proc Am Thorac Soc 4(2): 14568, 2007. urement of lung volumes. Eur Respir J 26(3): 51122, 2005.
29. de Marco R, Accordini S, Cerveri I, Corsico A, Anto JM, Kunzli N, 49. Pellegrino R, Viegi G, Brusasco V, Crapo RO, Burgos F, Casaburi R,
Janson C, Sunyer J, Jarvis D, Chinn S, Vermeire P, Svanes C, Coates A, van der Grinten CP, Gustafsson P, Hankinson J, Jensen R,
Ackermann-Liebrich U, Gislason T, Heinrich J, Leynaert B, Neukirch Johnson DC, MacIntyre N, McKay R, Miller MR, Navajas D,
F, Schouten JP, Wjst M, Burney P. Incidence of chronic obstructive Pedersen OF, Wanger J. Interpretative strategies for lung function tests.
pulmonary disease in a cohort of young adults according to the pres- Eur Respir J 26(5): 94868, 2005.
ence of chronic cough and phlegm. Am J Respir Crit Care Med 175(1): 50. Anthonisen NR, Wright EC. Bronchodilator response in chronic
3239, 2007. obstructive pulmonary disease. Am Rev Respir Dis 133(5): 81419,
30. Viegi G, Pistelli F, Sherrill DL, Maio S, Baldacci S, Carrozzi L. 1986.
Denition, epidemiology and natural history of COPD. Eur Respir J 51. Calverley PM, Rennard SI. What have we learned from large drug
30(5): 9931013, 2007. treatment trials in COPD? Lancet 370(9589): 77485, 2007.
31. Denition and classication of chronic bronchitis for clinical and 52. Celli BR. The importance of spirometry in COPD and asthma: Eect
epidemiological purposes. A report to the Medical Research Council on approach to management. Chest 117(Suppl 2): 15S19S, 2000.
by their Committee on the Aetiology of Chronic Bronchitis. Lancet 53. Soriano JB, Mannino DM. Reversing concepts on COPD irreversibil-
1(7389): 7759, 1965. ity. Eur Respir J 31(4): 69596, 2008.

536
Diagnosis of Asthma and COPD 41
54. Tashkin DP, Celli B, Decramer M, Liu D, Burkhart D, Cassino C, corticosteroid therapy for obstructive airways disease. Dutch Chronic
Kesten S. Bronchodilator responsiveness in patients with COPD. Eur Non-Specic Lung Disease Study Group [see comments]. N Engl J
Respir J 31(4): 74250, 2008. Med 327(20): 141319, 1992.
55. Chiry S, Cartier A, Malo JL, Tarlo SM, Lemiere C. Comparison 75. Chrystyn H, Mulley BA, Peake MD. Dose response relation to oral
of peak expiratory ow variability between workers with work- theophylline in severe chronic obstructive airways disease. BMJ
exacerbated asthma and occupational asthma. Chest 132(2): 48388, 2007. 297(6662): 150610, 1988.
56. Reddel HK, Marks GB, Jenkins CR. When can personal best peak 76. Gross NJ, Petty TL, Friedman M, Skorodin MS, Silvers GW, Donohue
ow be determined for asthma action plans? Thorax 59(11): 92224, JF. Dose response to ipratropium as a nebulized solution in patients
2004. with chronic obstructive pulmonary disease. A three-center study. Am
57. Wensley D, Silverman M. Peak ow monitoring for guided self- Rev Respir Dis 139(5): 118891, 1989.
management in childhood asthma: A randomized controlled trial. Am J 77. Higgins BG, Powell RM, Cooper S, Tatterseld AE. Eect of
Respir Crit Care Med 170(6): 60612, 2004. salbutamol and ipratropium bromide on airway calibre and bronchial
58. Stahl E. Correlation between objective measures of airway calibre and reactivity in asthma and chronic bronchitis. Eur Respir J 4(4): 41520,
clinical symptoms in asthma: A systematic review of clinical studies. 1991.
Respir Med 94(8): 73541, 2000. 78. Vathenen AS, Britton JR, Ebden P, Cookson JB, Wharrad HJ,
59. Harrison TW, Oborne J, Wilding PJ, Tatterseld AE. Randomised pla- Tatterseld AE. High-dose inhaled albuterol in severe chronic airow
cebo controlled trial of beta agonist dose reduction in asthma. Thorax limitation. Am Rev Respir Dis 138(4): 85055, 1988.
54(2): 98102, 1999. 79. Tashkin D, Kesten S. Long-term treatment benets with tiotropium in
60. Sawyer G, Miles J, Lewis S, Fitzharris P, Pearce N, Beasley R. COPD patients with and without short-term bronchodilator responses.
Classication of asthma severity: Should the international guidelines Chest 123(5): 144149, 2003.
be changed? Clin Exp Allergy 28(12): 156570, 1998. 80. Decramer M, Celli B, Tashkin DP, Pauwels RA, Burkhart D,
61. Kennedy WA, Girard F, Chaboillez S, Cartier A, Cote J, Hargreave F, Cassino C, Kesten S. Clinical trial design considerations in assessing
Labrecque M, Malo JL, Tarlo SM, Redlich CA, Lemiere C. Cost- long-term functional impacts of tiotropium in COPD: The UPLIFT
eectiveness of various diagnostic approaches for occupational asthma. trial. COPD 1(2): 30312, 2004.
Can Respir J 14(5): 27680, 2007. 81. Bateman ED, Boushey HA, Bousquet J, Busse WW, Clark TJ,
62. Mapp CE, Boschetto P, Maestrelli P, Fabbri LM. Occupational asthma. Pauwels RA, Pedersen SE. Can guideline-dened asthma control be
Am J Respir Crit Care Med 172(3): 280305, 2005. achieved? The Gaining Optimal Asthma ControL study. Am J Respir
63. Venables KM, Chan-Yeung M. Occupational asthma. Lancet Crit Care Med 170(8): 83644, 2004.
349(9063): 146569, 1997. 82. Spencer S, Mayer B, Bendall KL, Bateman ED. Validation of a
64. Hansen EF, Vestbo J, Phanareth K, Kok-Jensen A, Dirksen A. Peak ow guideline-based composite outcome assessment tool for asthma con-
as predictor of overall mortality in asthma and chronic obstructive pul- trol. Respir Res 8: 26, 2007.
monary disease. Am J Respir Crit Care Med 163(3 Pt 1): 69093, 2001. 83. Hemminki K, Li X, Sundquist K, Sundquist J. Familial risks for asthma
65. Mallia P, Message SD, Kebadze T, Parker HL, Kon OM, Johnston SL. among twins and other siblings based on hospitalizations in Sweden.
An experimental model of rhinovirus induced chronic obstructive pul- Clin Exp Allergy 37(9): 132025, 2007.
monary disease exacerbations: A pilot study. Respir Res 7: 116, 2006. 84. Berger Z, Rom WN, Reibman J, Kim M, Zhang S, Luo L, Friedman-
66. Wilkinson TM, Donaldson GC, Johnston SL, Openshaw PJ, Jimenez G. Prevalence of workplace exacerbation of asthma symptoms
Wedzicha JA. Respiratory syncytial virus, airway inammation, and in an urban working population of asthmatics. J Occup Environ Med
FEV1 decline in patients with chronic obstructive pulmonary disease. 48(8): 83339, 2006.
Am J Respir Crit Care Med 173(8): 87176, 2006. 85. Kogevinas M, Zock JP, Jarvis D, Kromhout H, Lillienberg L, Plana E,
67. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson JA. Radon K, Toren K, Alliksoo A, Benke G, Blanc PD, Dahlman-
Prednisolone response in patients with chronic obstructive pulmonary Hoglund A, DErrico A, Hery M, Kennedy S, Kunzli N, Leynaert
disease: Results from the ISOLDE study. Thorax 58(8): 65458, 2003. B, Mirabelli MC, Muniozguren N, Norback D, Olivieri M, Payo F,
68. Calverley PM, Burge PS, Spencer S, Anderson JA, Jones PW. Villani S, van Sprundel M, Urrutia I, Wieslander G, Sunyer J,
Bronchodilator reversibility testing in chronic obstructive pulmonary Anto JM. Exposure to substances in the workplace and new-onset
disease. Thorax 58(8): 65964, 2003. asthma: An international prospective population-based study
69. Sterk PJ, Fabbri LM, Quanjer PH, Cockcroft DW, (ECRHS-II). Lancet 370(9584): 33641, 2007.
OByrne PM, Anderson SD, Juniper EF, Malo JL. Airway responsive- 86. Corren J. The connection between allergic rhinitis and bronchial
ness. Standardized challenge testing with pharmacological, physical and asthma. Curr Opin Pulm Med 13(1): 1318, 2007.
sensitizing stimuli in adults. Report Working Party Standardization 87. Apter AJ, Szeer SJ. Advances in adult and pediatric asthma. J Allergy
of Lung Function Tests, European Community for Steel and Coal. Clin Immunol 117(3): 51218, 2006.
Ocial Statement of the European Respiratory Society. Eur Respir J 88. Busse WW, Wanner A, Adams K, Reynolds HY, Castro M,
Suppl 16: 5383, 1993. Chowdhury B, Kraft M, Levine RJ, Peters SP, Sullivan EJ. Investigative
70. Hansen EF, Phanareth K, Laursen LC, Kok-Jensen A, Dirksen A. bronchoprovocation and bronchoscopy in airway diseases. Am J Respir
Reversible and irreversible airow obstruction as predictor of overall Crit Care Med 172(7): 80716, 2005.
mortality in asthma and chronic obstructive pulmonary disease. Am J 89. Pearce N, Pekkanen J, Beasley R. How much asthma is really attribut-
Respir Crit Care Med 159(4 Pt 1): 126771, 1999. able to atopy. Thorax 54(3): 26872, 1999.
71. Ulrik CS, Backer V. Nonreversible airow obstruction in life-long non- 90. Weiss ST. Atopy as a risk factor for chronic obstructive pulmonary dis-
smokers with moderate to severe asthma. Eur Respir J 14(4): 89296, ease: Epidemiological evidence. Am J Respir Crit Care Med 162(3 Pt 2):
1999. S13436, 2000.
72. Gross NJ. COPD: A disease of reversible air-ow obstruction. Am Rev 91. Weir DC, Burge PS. Assessment of reversibility of airway obstruction
Respir Dis 133(5): 72526, 1986. in patients with chronic obstructive airways disease. Thorax 45(11):
73. Chanez P, Vignola AM, OShaugnessy T, Enander I, Li D, Jeery PK, 9078, 1990.
Bousquet J. Corticosteroid reversibility in COPD is related to features 92. Weir DC, Burge PS. Eects of high dose inhaled beclomethasone
of asthma. Am J Respir Crit Care Med 155(5): 152934, 1997. dipropionate, 750 micrograms and 1500 micrograms twice daily,
74. Kerstjens HA, Brand PL, Hughes MD, Robinson NJ, Postma DS, and 40 mg per day oral prednisolone on lung function, symptoms, and
Sluiter HJ, Bleecker ER, Dekhuijzen PN, de Jong PM, Mengelers HJ bronchial hyperresponsiveness in patients with non-asthmatic chronic
et al. A comparison of bronchodilator therapy with or without inhaled airow obstruction. Thorax 48(4): 30916, 1993.

537
Asthma and COPD: Basic Mechanisms and Clinical Management

93. Weir DC, Gove RI, Robertson AS, Burge PS. Corticosteroid trials 109. Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C,
in non-asthmatic chronic airow obstruction: A comparison of oral Jones PW, Yates JC, Vestbo J. Salmeterol and uticasone propionate
prednisolone and inhaled beclomethasone dipropionate. Thorax 45(2): and survival in chronic obstructive pulmonary disease. N Engl J Med
11217, 1990. 356(8): 77589, 2007.
94. Weir DC, Robertson AS, Gove RI, Burge PS. Time course of 110. Nannini LJ, Cates CJ, Lasserson TJ, Poole P. Combined corticoster-
response to oral and inhaled corticosteroids in non-asthmatic chronic oid and long-acting beta-agonist in one inhaler versus inhaled ster-
airow obstruction. Thorax 45(2): 11821, 1990. oids for chronic obstructive pulmonary disease. Cochrane Database Syst
95. Pauwels RA, Lofdahl CG, Postma DS, Tatterseld AE, OByrne P, Rev (4): CD006826, 2007.
Barnes PJ, Ullman A. Eect of inhaled formoterol and budesonide on 111. Brown CD, Wise RA. Field tests of exercise in COPD: The six-
exacerbations of asthma. Formoterol and Corticosteroids Establishing minute walk test and the shuttle walk test. COPD 4(3): 21723, 2007.
Therapy (FACET) International Study Group [see comments] [pub- 112. Macintyre N, Crapo RO, Viegi G, Johnson DC, van der Grinten CP,
lished erratum appears in N Engl J Med 338(2): 139, January 1998]. Brusasco V, Burgos F, Casaburi R, Coates A, Enright P, Gustafsson P,
N Engl J Med 337(20): 140511, 1997. Hankinson J, Jensen R, McKay R, Miller MR, Navajas D,
96. van Grunsven PM, van Schayck CP, Derenne JP, Kerstjens HA, Pedersen OF, Pellegrino R, Wanger J. Standardisation of the single-
Renkema TE, Postma DS, Similowski T, Akkermans RP, Pasker-de breath determination of carbon monoxide uptake in the lung. Eur
Jong PC, Dekhuijzen PN, van Herwaarden CL, van Weel C. Long Respir J 26(4): 72035, 2005.
term eects of inhaled corticosteroids in chronic obstructive pul- 113. Collard P, Njinou B, Nejadnik B, Keyeux A, Frans A. Single breath
monary disease: A meta-analysis [see comments]. Thorax 54(1): diusing capacity for carbon monoxide in stable asthma. Chest 105(5):
714, 1999. 142629, 1994.
97. Little SA, Chalmers GW, MacLeod KJ, McSharry C, Thomson NC. 114. Sin BA, Akkoca O, Saryal S, Oner F, Misirligil Z. Dierences
Non-invasive markers of airway inammation as predictors of oral between asthma and COPD in the elderly. J Investig Allergol Clin
steroid responsiveness in asthma. Thorax 55(3): 23234, 2000. Immunol 16(1): 4450, 2006.
98. Smith AD, Taylor DR. Is exhaled nitric oxide measurement a useful 115. Clausen JL. The diagnosis of emphysema, chronic bronchitis, and
clinical test in asthma? Curr Opin Allergy Clin Immunol 5(1): 4956, asthma. Clin Chest Med 11(3): 40516, 1990.
2005. 116. Sansores RH, Pare P, Abboud RT. Eect of smoking cessation on
99. Brightling CE, McKenna S, Hargadon B, Birring S, Green R, pulmonary carbon monoxide diusing capacity and capillary blood
Siva R, Berry M, Parker D, Monteiro W, Pavord ID, Bradding P. volume. Am Rev Respir Dis 146(4): 95964, 1992.
Sputum eosinophilia and the short term response to inhaled mometa- 117. Boulet LP, Turcotte H, Hudon C, Carrier G, Maltais F. Clinical,
sone in chronic obstructive pulmonary disease. Thorax 60(3): 19398, physiological and radiological features of asthma with incomplete
2005. reversibility of airow obstruction compared with those of COPD.
100. Brightling CE, Monteiro W, Ward R, Parker D, Morgan MD, Can Respir J 5(4): 27077, 1998.
Wardlaw AJ, Pavord ID. Sputum eosinophilia and short-term 118. Papi A, Romagnoli M, Bellettato CM, Cogo AL, Zanin A, Turato G,
response to prednisolone in chronic obstructive pulmonary disease: Saetta M, Ciaccia A, Fabbri LM. Pulmonary function and pathology
A randomised controlled trial. Lancet 356(9240): 148085, 2000. in asthma and chronic obstructive pulmonary disease (COPD) with
101. Papi A, Romagnoli M, Baraldo S, Braccioni F, Guzzinati I, Saetta M, similar degree of irreversible airow limitation. Eur Respir J 16(Suppl
Ciaccia A, Fabbri LM. Partial reversibility of airow limitation and 31): 551s, 2000.
increased exhaled NO and sputum eosinophilia in chronic obstructive 119. Wilson JS, Galvin JR. Normal diusing capacity in patients with PiZ
pulmonary disease. Am J Respir Crit Care Med 162(5): 177377, 2000. alpha(1)-antitrypsin deciency, severe airow obstruction, and signi-
102. The Lung Health Study Research Group. Eect of inhaled triamci- cant radiographic emphysema. Chest 118(3): 86771, 2000.
nolone on the decline in pulmonary function in chronic obstructive 120. Holme J, Stockley RA. Radiologic and clinical features of COPD
pulmonary disease. N Engl J Med 343(26): 19029, 2000. patients with discordant pulmonary physiology: Lessons from alpha1-
103. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson JA, Maslen antitrypsin deciency. Chest 132(3): 90915, 2007.
TK. Randomised, double blind, placebo controlled study of uticasone 121. Anderson SD. Provocative challenges to help diagnose and monitor
propionate in patients with moderate to severe chronic obstructive asthma: Exercise, methacholine, adenosine, and mannitol. Curr Opin
pulmonary disease: The ISOLDE trial. BMJ 320(7245): 1297303, Pulm Med 14(1): 3945, 2008.
2000. 122. Cockcroft DW, Hargreave FE. Airway hyperresponsiveness.
104. Paggiaro PL, Dahle R, Bakran I, Frith L, Hollingworth K, Relevance of random population data to clinical usefulness. Am Rev
Efthimiou J. Multicentre randomised placebo-controlled trial of Respir Dis 142(3): 497500, 1990.
inhaled uticasone propionate in patients with chronic obstruc- 123. Birnbaum S, Barreiro TJ. Methacholine challenge testing: Identifying
tive pulmonary disease. International COPD Study Group. Lancet its diagnostic role, testing, coding, and reimbursement. Chest 131(6):
351(9105): 77380, 1998. 193235, 2007.
105. Pauwels RA, Lofdahl CG, Laitinen LA, Schouten JP, Postma DS, 124. Miedinger D, Chhajed PN, Tamm M, Stolz D, Surber C, Leuppi JD.
Pride NB, Ohlsson SV. Long-term treatment with inhaled budeso- Diagnostic tests for asthma in reghters. Chest 131(6): 176067,
nide in persons with mild chronic obstructive pulmonary disease who 2007.
continue smoking. European Respiratory Society Study on Chronic 125. Padoan M, Pozzato V, Simoni M, Zedda L, Milan G, Bononi I,
Obstructive Pulmonary Disease. N Engl J Med 340(25): 194853, Piola C, Maestrelli P, Boschetto P, Mapp CE. Long-term follow-up
1999. of toluene diisocyanate-induced asthma. Eur Respir J 21(4): 63740,
106. Vestbo J, Sorensen T, Lange P, Brix A, Torre P, Viskum K. Long-term 2003.
eect of inhaled budesonide in mild and moderate chronic obstructive 126. Allergic Rhinitis and Its Impact on Asthma (ARIA). World Health
pulmonary disease: A randomised controlled trial. Lancet 353(9167): Organization Initiative. Geneva, 2000.
181923, 1999. 127. Polosa R, Ciamarra I, Mangano G, Prosperini G, Pistorio MP,
107. Wong CA, Walsh LJ, Smith CJ, Wisniewski AF, Lewis SA, Vancheri C, Crimi N. Bronchial hyperresponsiveness and airway
Hubbard R, Cawte S, Green DJ, Pringle M, Tatterseld AE. Inhaled inammation markers in nonasthmatics with allergic rhinitis. Eur
corticosteroid use and bone-mineral density in patients with asthma. Respir J 15(1): 3035, 2000.
Lancet 355(9213): 1399403, 2000. 128. Ramsdale EH, Morris MM, Roberts RS, Hargreave FE.
108. Mapp CE. Inhaled glucocorticoids in chronic obstructive pulmonary Asymptomatic bronchial hyperresponsiveness in rhinitis. J Allergy
disease. N Engl J Med 343(26): 196061, 2000. Clin Immunol 75(5): 57377, 1985.

538
Diagnosis of Asthma and COPD 41
129. Ip M, Lauder IJ, Wong WY, Lam WK, So SY. Multivariate analysis 150. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R,
of factors aecting pulmonary function in bronchiectasis. Respiration Ries A, Weinmann G, Wood DE. A randomized trial comparing lung-
60(1): 4550, 1993. volume-reduction surgery with medical therapy for severe
130. Nielsen KG, Pressler T, Klug B, Koch C, Bisgaard H. Serial lung emphysema. N Engl J Med 348(21): 205973, 2003.
function and responsiveness in cystic brosis during early childhood. 151. Martinez FJ, Chang A. Surgical therapy for chronic obstructive pul-
Am J Respir Crit Care Med 169(11): 120916, 2004. monary disease. Semin Respir Crit Care Med 26(2): 16791, 2005.
131. van Haren EH, Lammers JW, Festen J, van Herwaarden CL. 152. Lenfant C. Will lung volume reduction surgery be widely applied?
Bronchial vagal tone and responsiveness to histamine, exercise and Ann Thorac Surg 82(2): 38587, 2006.
bronchodilators in adult patients with cystic brosis. Eur Respir J 5(9): 153. McLean AN, Sproule MW, Cowan MD, Thomson NC. High resolu-
108388, 1992. tion computed tomography in asthma. Thorax 53(4): 30814, 1998.
132. Grootendorst DC, Rabe KF. Mechanisms of bronchial hyperreactivity 154. Webb WR. Radiology of obstructive pulmonary disease. AJR Am J
in asthma and chronic obstructive pulmonary disease. Proc Am Thorac Roentgenol 169(3): 63747, 1997.
Soc 1(2): 7787, 2004. 155. Boschetto P, Quintavalle S, Zeni E, Leprotti S, Potena A,
133. Rutgers SR, Timens W, Tzanakis N, Kauman HF, van der Ballerin L, Papi A, Palladini G, Luisetti M, Annovazzi L, Iadarola P,
Mark TW, Koeter GH, Postma DS. Airway inammation and hyper- De Rosa E, Fabbri LM, Mapp CE. Association between markers of
responsiveness to adenosine 5
-monophosphate in chronic obstructive emphysema and more severe chronic obstructive pulmonary disease.
pulmonary disease. Clin Exp Allergy 30(5): 65762, 2000. Thorax 61(12): 103742, 2006.
134. Schafroth Torok S, Leuppi JD. Bronchial hyper-responsiveness and 156. Kurashima K, Takayanagi N, Sato N, Kanauchi T, Hoshi T,
exhaled nitric oxide in chronic obstructive pulmonary disease. Swiss Tokunaga D, Ubukata M, Yanagisawa T, Sugita Y, Kanazawa M.
Med Wkly 137(2728): 38591, 2007. High resolution CT and bronchial reversibility test for diagnosing
135. Scichilone N, Battaglia S, La Sala A, Bellia V. Clinical implications COPD. Respirology 10(3): 31622, 2005.
of airway hyperresponsiveness in COPD. Int J Chron Obstruct Pulmon 157. Anthonisen NR, Manfreda J, Warren CP, Hersheld ES,
Dis 1(1): 4960, 2006. Harding GK, Nelson NA. Antibiotic therapy in exacerbations of
136. James AL, Wenzel S. Clinical relevance of airway remodelling in air- chronic obstructive pulmonary disease. Ann Intern Med 106(2):
way diseases. Eur Respir J 30(1): 13455, 2007. 196204, 1987.
137. Sung A, Naidich D, Belinskaya I, Raoof S. The role of chest radio- 158. Stolz D, Christ-Crain M, Morgenthaler NG, Leuppi J, Miedinger D,
graphy and computed tomography in the diagnosis and management Bingisser R, Muller C, Struck J, Muller B, Tamm M. Copeptin,
of asthma. Curr Opin Pulm Med 13(1): 3136, 2007. C-reactive protein, and procalcitonin as prognostic biomarkers in
138. Nakano Y, Muller NL, King GG, Niimi A, Kalloger SE, Mishima M, acute exacerbation of COPD. Chest 131(4): 105867, 2007.
Pare PD. Quantitative assessment of airway remodeling using high- 159. Stolz D, Christ-Crain M, Bingisser R, Leuppi J, Miedinger D,
resolution CT. Chest 122(Suppl 6): 271S75S, 2002. Muller C, Huber P, Muller B, Tamm M. Antibiotic treatment of exac-
139. Sharma V, Shaaban AM, Berges G, Gosselin M. The radiological erbations of COPD: A randomized, controlled trial comparing pro-
spectrum of small-airway diseases. Semin Ultrasound CT MR 23(4): calcitonin-guidance with standard therapy. Chest 131(1): 919, 2007.
33951, 2002. 160. Stolz D, Christ-Crain M, Gencay MM, Bingisser R, Huber PR,
140. Niimi A, Matsumoto H, Takemura M, Ueda T, Nakano Y, Mishima M. Muller B, Tamm M. Diagnostic value of signs, symptoms and labo-
Clinical assessment of airway remodeling in asthma: Utility of com- ratory values in lower respiratory tract infection. Swiss Med Wkly
puted tomography. Clin Rev Allergy Immunol 27(1): 4558, 2004. 136(2728): 43440, 2006.
141. Coxson HO, Rogers RM. Quantitative computed tomography of 161. Poulsen SH, Noer I, Moller JE, Knudsen TE, Frandsen JL. Clinical
chronic obstructive pulmonary disease. Acad Radiol 12(11): 145763, outcome of patients with suspected pulmonary embolism. A
2005. follow-up study of 588 consecutive patients. J Intern Med 250(2):
142. Coxson HO, Rogers RM. New concepts in the radiological assess- 13743, 2001.
ment of COPD. Semin Respir Crit Care Med 26(2): 21120, 2005. 162. Schonhofer B, Kohler D. Prevalence of deep-vein thrombosis of the
143. Baldi S, Miniati M, Bellina CR, Battolla L, Catapano G, leg in patients with acute exacerbation of chronic obstructive pulmo-
Begliomini E, Giustini D, Giuntini C. Relationship between extent nary disease. Respiration 65(3): 17377, 1998.
of pulmonary emphysema by high-resolution computed tomography 163. Winter JH, Buckler PW, Bautista AP, Smith FW, Sharp PF,
and lung elastic recoil in patients with chronic obstructive pulmonary Bennett B, Douglas AS. Frequency of venous thrombosis in patients
disease. Am J Respir Crit Care Med 164(4): 58589, 2001. with an exacerbation of chronic obstructive lung disease. Thorax 38(8):
144. Omori H, Fujimoto K, Katoh T. Computed-tomography ndings 6058, 1983.
of emphysema: Correlation with spirometric values. Curr Opin Pulm 164. Erelel M, Cuhadaroglu C, Ece T, Arseven O. The frequency of deep
Med 14(2): 11014, 2008. venous thrombosis and pulmonary embolus in acute exacerbation of
145. Hogg JC. State of the art. Bronchiolitis in chronic obstructive pulmo- chronic obstructive pulmonary disease. Respir Med 96(7): 51518,
nary disease. Proc Am Thorac Soc 3(6): 48993, 2006. 2002.
146. de Jong PA, Muller NL, Pare PD, Coxson HO. Computed tomo- 165. Tillie-Leblond I, Marquette CH, Perez T, Scherpereel A, Zanetti C,
graphic imaging of the airways: Relationship to structure and func- Tonnel AB, Remy-Jardin M. Pulmonary embolism in patients with
tion. Eur Respir J 26(1): 14052, 2005. unexplained exacerbation of chronic obstructive pulmonary disease:
147. Magnant J, Vecellio L, de Monte M, Grimbert D, Valat C, Boissinot Prevalence and risk factors. Ann Intern Med 144(6): 39096, 2006.
E, Guilloteau D, Lemarie E, Diot P. Comparative analysis of dier- 166. Palareti G, Cosmi B, Legnani C. Diagnosis of deep vein thrombosis.
ent scintigraphic approaches to assess pulmonary ventilation. J Aerosol Semin Thromb Hemost 32(7): 65972, 2006.
Med 19(2): 14859, 2006. 167. Kuruvilla J, Wells PS, Morrow B, MacKinnon K, Keeney M,
148. Fain SB, Panth SR, Evans MD, Wentland AL, Holmes JH, Kovacs MJ. Prospective assessment of the natural history of posi-
Korosec FR, OBrien MJ, Fountaine H, Grist TM. Early emphyse- tive D-dimer results in persons with acute venous thromboembolism
matous changes in asymptomatic smokers: Detection with 3He MR (DVT or PE). Thromb Haemost 89(2): 28487, 2003.
imaging. Radiology 239(3): 87583, 2006. 168. Musset D, Parent F, Meyer G, Maitre S, Girard P, Leroyer C,
149. Morino S, Toba T, Araki M, Azuma T, Tsutsumi S, Tao H, Nakamura Revel MP, Carette MF, Laurent M, Charbonnier B, Laurent F,
T, Nagayasu T, Tagawa T. Noninvasive assessment of pulmonary Mal H, Nonent M, Lancar R, Grenier P, Simonneau G. Diagnostic
emphysema using dynamic contrast-enhanced magnetic resonance strategy for patients with suspected pulmonary embolism: A prospec-
imaging. Exp Lung Res 32(12): 5567, 2006. tive multicentre outcome study. Lancet 360(9349): 191420, 2002.

539
Asthma and COPD: Basic Mechanisms and Clinical Management

169. Davidson BL, Tomkowski WZ. Management of pulmonary embo- 192. Kikuchi S, Nagata M, Kikuchi I, Hagiwara K, Kanazawa M.
lism in 2005. Dis Mon 51(23): 11623, 2005. Association between neutrophilic and eosinophilic inammation
170. Le Jemtel TH, Padeletti M, Jelic S. Diagnostic and therapeutic chal- in patients with severe persistent asthma. Int Arch Allergy Immunol
lenges in patients with coexistent chronic obstructive pulmonary dis- 137(Suppl 1): 711, 2005.
ease and chronic heart failure. J Am Coll Cardiol 49(2): 17180, 2007. 193. Pavord ID, Brightling CE, Woltmann G, Wardlaw AJ. Non-
171. Omland T, de Lemos JA. Amino-terminal pro-B-type natriuretic eosinophilic corticosteroid unresponsive asthma. Lancet 353(9171):
peptides in stable and unstable ischemic heart disease. Am J Cardiol 221314, 1999.
101(3A): 6166, 2008. 194. Grootendorst DC, Gauw SA, Verhoosel RM, Sterk PJ, Hospers JJ,
172. Blanco I, de Serres FJ, Fernandez-Bustillo E, Lara B, Miravitlles M. Bredenbroker D, Bethke TD, Hiemstra PS, Rabe KF. Reduction in
Estimated numbers and prevalence of PI*S and PI*Z alleles of sputum neutrophil and eosinophil numbers by the PDE4 inhibitor
alpha1-antitrypsin deciency in European countries. Eur Respir J roumilast in patients with COPD. Thorax 62(12): 108187, 2007.
27(1): 7784, 2006. 195. Pizzichini E, Pizzichini MM, Gibson P, Parameswaran K, Gleich GJ,
173. Mauad T, Dolhniko M. Pathologic similarities and dierences Berman L, Dolovich J, Hargreave FE. Sputum eosinophilia predicts
between asthma and chronic obstructive pulmonary disease. Curr benet from prednisone in smokers with chronic obstructive bronchi-
Opin Pulm Med 14(1): 3138, 2008. tis. Am J Respir Crit Care Med 158(5 Pt 1): 151117, 1998.
174. Bochner BS, Busse WW. Allergy and asthma. J Allergy Clin Immunol 196. Snoeck-Stroband JB, Lapperre TS, Gosman MM, Boezen HM,
115(5): 95359, 2005. Timens W, ten Hacken NH, Sont JK, Sterk PJ, Hiemstra PS. Chronic
175. Busse WW, Lemanske RF Jr. Asthma. N Engl J Med 344(5): 35062, bronchitis sub-phenotype within COPD: Inammation in sputum
2001. and biopsies. Eur Respir J 31(1): 7077, 2008.
176. Fabbri LM, Romagnoli M, Luppi F, Corbetta L. Asthma ver- 197. int Veen JC, Smits HH, Hiemstra PS, Zwinderman AE, Sterk PJ, Bel
sus COPD: Cellular and molecular dierences. In: Barnes PJ (ed.), EH. Lung function and sputum characteristics of patients with severe
COPD Cellular and Molecular Mechanisms. Lung Biology in Health and asthma during an induced exacerbation by double-blind steroid with-
Disease Series, pp. 5581. Marcel Dekker, Inc, New York Basel, 2004. drawal. Am J Respir Crit Care Med 160(1): 9399, 1999.
177. Jeery PK. Structural and inammatory changes in COPD: A com- 198. Jatakanon A, Lim S, Barnes PJ. Changes in sputum eosinophils pre-
parison with asthma. Thorax 53(2): 12936, 1998. dict loss of asthma control. Am J Respir Crit Care Med 161(1): 6472,
178. Fabbri L, Beghe B, Caramori G, Papi A, Saetta M. Similarities and 2000.
discrepancies between exacerbations of asthma and chronic obstruc- 199. Turner MO, Hussack P, Sears MR, Dolovich J, Hargreave FE.
tive pulmonary disease. Thorax 53(9): 8038, 1998. Exacerbations of asthma without sputum eosinophilia. Thorax 50(10):
179. Fabbri LM, Durham S, Holgate ST, OByrne PM, Postma DS. 105761, 1995.
Assessment of airway inammation: An overview. Eur Respir 200. Lovett CJ, Whitehead BF, Gibson PG. Eosinophilic airway inam-
J(Suppl 26): 6S8S, 1998. mation and the prognosis of childhood asthma. Clin Exp Allergy
180. Saetta M. Airway pathology of COPD compared with asthma. Eur 37(11): 1594601, 2007.
Respir Rev 7: 2933, 1997. 201. Fahy JV, Boushey HA, Lazarus SC, Mauger EA, Cherniack RM,
181. Saetta M. Airway inammation in chronic obstructive pulmonary Chinchilli VM, Craig TJ, Drazen JM, Ford JG, Fish JE, Israel E,
disease. Am J Respir Crit Care Med 160(5 Pt 2): S1720, 1999. Kraft M, Lemanske RF, Martin RJ, McLean D, Peters SP,
182. Saetta M, Timens W, Je rey PK. Pathology. In: Postma DS, Siafakas Sorkness C, Szeer SJ. Safety and reproducibility of sputum induc-
NM (eds.), Management of Chronic Obstructive Pulmonary Disease. tion in asthmatic subjects in a multicenter study. Am J Respir Crit Care
Sheeld, UK: Eur Respir Mon ERS Journals Ltd; 92111,1998. Med 163(6): 147075, 2001.
183. Barnes PJ, Chowdhury B, Kharitonov SA, Magnussen H, Page CP, 202. Lamblin C, Gosset P, Tillie-Leblond I, Saulnier F, Marquette CH,
Postma D, Saetta M. Pulmonary biomarkers in chronic obstructive Wallaert B, Tonnel AB. Bronchial neutrophilia in patients with non-
pulmonary disease. Am J Respir Crit Care Med 174(1): 614, 2006. infectious status asthmaticus. Am J Respir Crit Care Med 157(2):
184. Zhang JY, Wenzel SE. Tissue and BAL based biomarkers in asthma. 394402, 1998.
Immunol Allergy Clin North Am 27(4): 62332, vi, 2007. 203. Saetta M, Di Stefano A, Maestrelli P, Turato G, Ruggieri MP,
185. Menzies D, Nair A, Lipworth BJ. Non-invasive measurement of air- Roggeri A, Calcagni P, Mapp CE, Ciaccia A, Fabbri LM. Airway
way inammation in asthma. J Asthma 43(6): 40715, 2006. eosinophilia in chronic bronchitis during exacerbations. Am J Respir
186. Brightling CE. Clinical applications of induced sputum. Chest 129(5): Crit Care Med 150(6 Pt 1): 164652, 1994.
134448, 2006. 204. Siva R, Green RH, Brightling CE, Shelley M, Hargadon B,
187. Hargreave FE, Leigh R. Induced sputum, eosinophilic bronchitis, McKenna S, Monteiro W, Berry M, Parker D, Wardlaw AJ,
and chronic obstructive pulmonary disease. Am J Respir Crit Care Med Pavord ID. Eosinophilic airway inammation and exacerbations of
160(5 Pt 2): S5357, 1999. COPD: A randomised controlled trial. Eur Respir J 29(5): 90613, 2007.
188. Pizzichini MM, Pizzichini E, Clelland L, Efthimiadis A, Mahony J, 205. Piattella M, Maestrelli P, Saetta M, Mapp C, Caramori G, Ciaccia A,
Dolovich J, Hargreave FE. Sputum in severe exacerbations of asthma: Fabbri LM. Sputum eosinophilia during mild exacerbations and spu-
Kinetics of inammatory indices after prednisone treatment. Am J tum neutrophilia during severe exacerbations of COPD. Am J Respir
Respir Crit Care Med 155(5): 15018, 1997. Crit Care Med 153(Suppl): A822, 1996.
189. Kips JC, Fahy JV, Hargreave FE, Ind PW, int Veen JC. Methods for 206. Hill AT, Bayley D, Stockley RA. The interrelationship of sputum
sputum induction and analysis of induced sputum: A method for inammatory markers in patients with chronic bronchitis. Am J Respir
assessing airway inammation in asthma. Eur Respir J Suppl 26: 9S Crit Care Med 160(3): 89398, 1999.
12S, 1998. 207. Keatings VM, Barnes PJ. Granulocyte activation markers in induced
190. Pin I, Gibson PG, Kolendowicz R, Girgis-Gabardo A, Denburg JA, sputum: Comparison between chronic obstructive pulmonary dis-
Hargreave FE, Dolovich J. Use of induced sputum cell counts to ease, asthma, and normal subjects. Am J Respir Crit Care Med 155(2):
investigate airway inammation in asthma. Thorax 47(1): 2529, 44953, 1997.
1992. 208. Keatings VM, Barnes PJ. Comparison of inammatory cytokines
191. Green RH, Brightling CE, Woltmann G, Parker D, Wardlaw AJ, in chronic obstructive pulmonary disease, asthma and controls. Eur
Pavord ID. Analysis of induced sputum in adults with asthma: Respir Rev 7: 14650, 1997.
Identication of subgroup with isolated sputum neutrophilia and poor 209. ODriscoll BR, Cromwell O, Kay AB. Sputum leukotrienes in
response to inhaled corticosteroids. Thorax 57(10): 87579, 2002. obstructive airways diseases. Clin Exp Immunol 55(2): 397404, 1984.

540
Diagnosis of Asthma and COPD 41
210. Pizzichini E, Pizzichini MM, Efthimiadis A, Evans S, Morris MM, 229. Bresciani M, Paradis L, Des Roches A, Vernhet H, Vachier I,
Squillace D, Gleich GJ, Dolovich J, Hargreave FE. Indices of airway Godard P, Bousquet J, Chanez P. Rhinosinusitis in severe asthma. J
inammation in induced sputum: Reproducibility and validity of cell Allergy Clin Immunol 107(1): 7380, 2001.
and uid-phase measurements. Am J Respir Crit Care Med 154(2 Pt 1): 230. ten Brinke A, Zwinderman AH, Sterk PJ, Rabe KF, Bel EH. Factors
30817, 1996. associated with persistent airow limitation in severe asthma. Am J
211. Barnes PJ. Nitric oxide and airway disease. Ann Med 27(3): 38993, Respir Crit Care Med 164(5): 74448, 2001.
1995. 231. Bachert C, van Cauwenberge P, Khaltaev N. Allergic rhinitis and
212. Kharitonov SA, Barnes PJ. Exhaled biomarkers. Chest 130(5): its impact on asthma. In collaboration with the World Health
154146, 2006. Organization. Executive summary of the workshop report. December
213. Alving K, Weitzberg E, Lundberg JM. Increased amount of nitric 710, 1999, Geneva, Switzerland. Allergy 57(9): 84155, 2002.
oxide in exhaled air of asthmatics. Eur Respir J 6(9): 136870, 1993. 232. Gibson PG, Henry RL, Coughlan JL. Gastro-oesophageal reux
214. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R, treatment for asthma in adults and children. Cochrane Database Syst
Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of Rev (2): CD001496, 2003.
asthmatic patients. Lancet 343(8890): 13335, 1994. 233. Coughlan JL, Gibson PG, Henry RL. Medical treatment for reux
215. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JM. oesophagitis does not consistently improve asthma control: A system-
Expired nitric oxide levels during treatment of acute asthma. Am J atic review. Thorax 56(3): 198204, 2001.
Respir Crit Care Med 152(2): 8003, 1995. 234. Kiljander TO, Harding SM, Field SK, Stein MR, Nelson HS,
216. Persson MG, Zetterstrom O, Agrenius V, Ihre E, Gustafsson LE. Ekelund J, Illueca M, Beckman O, Sostek MB. Eects of esomepra-
Single-breath nitric oxide measurements in asthmatic patients and zole 40 mg twice daily on asthma: A randomized placebo-controlled
smokers. Lancet 343(8890): 14647, 1994. trial. Am J Respir Crit Care Med 173(10): 109197, 2006.
217. Turner S. Exhaled nitric oxide in the diagnosis and management of 235. Littner MR, Leung FW, Ballard ED II, Huang B, Samra NK. Eects
asthma. Curr Opin Allergy Clin Immunol 8(1): 7076, 2008. of 24 weeks of lansoprazole therapy on asthma symptoms, exacer-
218. Gratziou C, Lignos M, Dassiou M, Roussos C. Inuence of atopy on bations, quality of life, and pulmonary function in adult asthmatic
exhaled nitric oxide in patients with stable asthma and rhinitis. Eur patients with acid reux symptoms. Chest 128(3): 112835, 2005.
Respir J 14(4): 897901, 1999. 236. Slavin RG. Medical management of rhinosinusitis comorbidities-
219. Henriksen AH, Lingaas-Holmen T, Sue-Chu M, Bjermer L. asthma, aspirin sensitivity, gastroesophageal reux, immune decien-
Combined use of exhaled nitric oxide and airway hyperresponsive- cies. Clin Allergy Immunol 20: 27386, 2007.
ness in characterizing asthma in a large population survey. Eur Respir 237. Peroni DG, Piacentini GL, Ceravolo R, Boner AL. Dicult asthma:
J 15(5): 84955, 2000. Possible association with rhinosinusitis. Pediatr Allergy Immunol
220. van Rensen EL, Straathof KC, Veselic-Charvat MA, Zwinderman AH, 18(Suppl 18): 2527, 2007.
Bel EH, Sterk PJ. Eect of inhaled steroids on airway hyperre- 238. Holguin F, Folch E, Redd SC, Mannino DM. Comorbidity and mor-
sponsiveness, sputum eosinophils, and exhaled nitric oxide levels in tality in COPD-related hospitalizations in the United States, 1979 to
patients with asthma. Thorax 54(5): 4038, 1999. 2001. Chest 128(4): 200511, 2005.
221. Yates DH, Kharitonov SA, Barnes PJ. Eect of short- and long- 239. Sidney S, Sorel M, Quesenberry CP Jr, DeLuise C, Lanes S,
acting inhaled beta2-agonists on exhaled nitric oxide in asthmatic Eisner MD. COPD and incident cardiovascular disease hospitaliza-
patients. Eur Respir J 10(7): 148388, 1997. tions and mortality: Kaiser Permanente Medical Care Program. Chest
222. Corradi M, Majori M, Cacciani GC, Consigli GF, deMunari E, 128(4): 206875, 2005.
Pesci A. Increased exhaled nitric oxide in patients with stable chronic 240. Sin DD, Wu L, Anderson JA, Anthonisen NR, Buist AS, Burge PS,
obstructive pulmonary disease. Thorax 54(7): 57275, 1999. Calverley PM, Connett JE, Lindmark B, Pauwels RA, Postma DS,
223. Kanazawa H, Shoji S, Yoshikawa T, Hirata K, Yoshikawa J. Increased Soriano JB, Szafranski W, Vestbo J. Inhaled corticosteroids and
production of endogenous nitric oxide in patients with bronchial mortality in chronic obstructive pulmonary disease. Thorax 60(12):
asthma and chronic obstructive pulmonary disease. Clin Exp Allergy 99297, 2005.
28(10): 124450, 1998. 241. Balkwill F, Mantovani A. Inammation and cancer: Back to Virchow?
224. Maziak W, Loukides S, Culpitt S, Sullivan P, Kharitonov SA, Lancet 357(9255): 53945, 2001.
Barnes PJ. Exhaled nitric oxide in chronic obstructive pulmonary dis- 242. Mannino DM, Watt G, Hole D, Gillis C, Hart C, McConnachie A,
ease. Am J Respir Crit Care Med 157(3 Pt 1): 9981002, 1998. Davey Smith G, Upton M, Hawthorne V, Sin DD, Man SF, Van
225. Robbins RA, Floreani AA, Von Essen SG, Sisson JH, Hill GE, Eeden S, Mapel DW, Vestbo J. The natural history of chronic obstruc-
Rubinstein I, Townley RG. Measurement of exhaled nitric oxide tive pulmonary disease. Eur Respir J 27(3): 62743, 2006.
by three dierent techniques. Am J Respir Crit Care Med 153(5): 243. Sevenoaks MJ, Stockley RA. Chronic Obstructive Pulmonary
163135, 1996. Disease, inammation and co-morbidity a common inammatory
226. Rutgers SR, Meijer RJ, Kerstjens HA, van der Mark TW, Koeter phenotype? Respir Res 7: 70, 2006.
GH, Postma DS. Nitric oxide measured with single-breath and tidal- 244. Naeije R. Pulmonary hypertension and right heart failure in chronic
breathing methods in asthma and COPD. Eur Respir J 12(4): 81619, obstructive pulmonary disease. Proc Am Thorac Soc 2(1): 2022, 2005.
1998. 245. Naeije R, Vizza D. Current perspectives modern hemodynamic evalu-
227. Rutgers SR, van der Mark TW, Coers W, Moshage H, Timens W, ation of the pulmonary circulation. Application to pulmonary arterial
Kauman HF, Koeter GH, Postma DS. Markers of nitric oxide hypertension and embolic pulmonary hypertension. Ital Heart J 6(10):
metabolism in sputum and exhaled air are not increased in chronic 78488, 2005.
obstructive pulmonary disease. Thorax 54(7): 57680, 1999.
228. Taylor DR, Pijnenburg MW, Smith AD, De Jongste JC. Exhaled
nitric oxide measurements: Clinical application and interpretation.
Thorax 61(9): 81727, 2006.

541
Non-invasive Assessment of
Airway Inflammation
42CHAPTER

INTRODUCTION nature of the inammatory response. The dier- Ian D. Pavord1 and
ent strengths and weaknesses of the techniques
suggests that they may nd dierent roles, with
Sergei A. Kharitonov2
Airway inammation is thought to be an impor- FENO being used mainly in primary care to facili- 1
Department of Respiratory Medicine
tant component of asthma, chronic obstructive tate diagnosis and to titrate corticosteroid therapy and Thoracic Surgery, Institute for Lung
pulmonary disease (COPD), and the various and induced sputum used in secondary and terti- Health, Glenfield Hospital, Leicester, UK
cough syndromes. However, it is not routinely ary care, where more detailed information on the 2
Section of Airway Disease, National
assessed in clinical practice and much of what we type of lower airway inammation is necessary.
Heart and Lung Institute, Imperial
know about the nature and importance of air- In this chapter we will review the method-
ology and validation of induced sputum and FENO College, London, UK
way inammation is derived from bronchoscopy
studies. These studies are limited by the invasive and outline other more experimental approaches
nature of bronchoscopy, bronchial biopsy, and to the assessment of airway inammation. We will
bronchoalveolar lavage so it has not been possi- nish by discussing how the widespread applica-
ble to study large heterogeneous populations nor tion of induced sputum to large and heterogene-
has not been standard practice to assess airway ous populations of patients with airway disease
inammation in clinical practice or clinical trials. has furthered our understanding of the complex
Over the last 1520 years there has been relationship between airway inammation and the
an explosion of interest in the assessment of air- clinical expression of airway disease and opened
way inammation using non-invasive means and the way to a new approach to the management
there are now a large number of dierent tech- of airway disease based on assessment of airway
niques to assess airway inammation, each with inammation (inammometry).
its own strengths and weaknesses (Table 42.1).
Two techniques are particularly well developed
and are already widely used in clinical trials and INDUCED SPUTUM
impacting on clinical practice: induced sputum,
where a sputum dierential and total cell count is
used to determine the characteristics and inten- Methodology
sity of the lower airway inammatory response;
and exhaled nitric oxide (FENO), where the con- Sputum induction using nebulized hypertonic
centration of nitric oxide (NO) in exhaled air is saline is used to collect respiratory secretions from
used to provide information about the presence the airways of patients who do not expectorate
of eosinophilic, corticosteroid responsive airway spontaneously. The precise mechanism leading to
inammation. Induced sputum has the advantage production of increased secretions is not known
of providing measurements of the type of airway but increased production of mucous by the sub-
inammation (eosinophilic versus neutrophilic) mucosal glands, increased vascular permeability of
as well as its severity. The main limitation is that the bronchial mucosa and release of pro-inam-
results are not available immediately, limiting the matory mediators may all contribute. Eighty to
application of the technique in asthma moni- ninety percent of patients with airway disease and
toring. FENO has the advantage of being simple a similar proportion of healthy controls are able
to measure and provides an immediate result to produce a satisfactory sputum sample follow-
but it provides only limited information on the ing hypertonic saline inhalation.

543
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 42.1 Comparison of methods measuring airway inflammation.

Ease of performing Ease of analyzing Influence on outcome


technique result Time to result Cost proved Potential use

Induced sputum Simple sputum Simple, but 34 h Moderate Yes Secondary care
sample requires
experience

Blood eosinophil Simple blood test Simple 30 min Inexpensive Suggests eosinophilic Secondary care
count airway inflammation
when raised in correct
context

Eosinophil Urine test/sputum test/ Simple 34 h Moderate Not proven Research


cationic protein blood test conclusively

Exhaled nitric Very easy Easy Immediate Expensive though Not proven Research
oxide breathing test could become Possibly primary
cheaper care

Exhaled breath Thermometer. Complex Easy Immediate Inexpensive Studies awaited Research
temperature currently but could be
simplified.

Breath condensate Cooling/freezing Simple Moderate Inexpensive Studies awaited Research


of exhaled air

BAL, bronchial Biopsy- invasive Complex 2 days Moderate Yes Secondary care
wash, and biopsy

BOX 42.1PROTOCOL FOR SPUTUM INDUCTION USING NEBULIZED HYPERTONIC SALINE.


Measure FEV1

Salbutamol 200g by MDI with spacer

Remeasure FEV1 after 20min

Administer 3% saline using an ultrasonic nebulizer for 5min

Blow nose, rinse mouth, and swallow water

Expectorate sputum

10%, 20% fall in FEV1

Remeasure FEV1

20% fall in FEV1 or troublesome sy

Discontinue
10% fall in FEV1

Repeat procedure with 4% and 5% saline

A variety of protocols for sputum induction have longer duration of saline inhalation and reduced frequency
been published and shown to be safe provided patients are and timing of safety assessment by forced expiratory volume
pre-treated with bronchodilators and monitored care- in 1 s or peak expiratory ow reduce safety. Higher output
fully [1, 2]. Risk factors for bronchoconstriction include a nebulizers have also been associated with the develop-
low baseline FEV1% predicted [3], overuse of short-acting ment of a sputum neutrophilia 24 h after sputum induction
2-agonists [4] and poor asthma control [4]. Symptomatic [7]. Whether this is seen with the low output nebuliz-
bronchoconstriction is unusual in patients with COPD, ers is unknown. A common and well-validated protocol is
even those with severe airow obstruction [5]; sputum to use a relatively low output ultrasonic nebulizer (output
induction has also been carried out repeatedly and safely 0.70.9 ml/min) delivering increasing concentrations of
in patients with severe asthma [6]. It is likely that higher hypertonic saline [5, 6, 8]. The method is summarized in
nebulizer output, higher concentration of inhaled saline, a Box 42.1.
544
Non-invasive Assessment of Airway Inflammation 42
Once expectorated sputum should be processed within method used to process induced sputum is outlined in Box
2 h. There is evidence that sputum can be stored for up to 9 h 42.2. The total cell count, cell viability and squamous cell con-
in a refrigerator at 4oC or that sputum can be snap frozen for tamination are assessed using a hemocytometer. Dierential
longer without aecting cell counts [9], although experience cell counts are determined by counting 400 leukocytes on an
with these techniques is limited. The whole expectorate or appropriately stained cytospin. Other biomarkers can also be
selected sputum plugs can be processed. The latter approach measured in the sputum supernatant. Some of the molecular
has the advantage of producing better quality cytospins and markers of airway inammation that have been successfully
more repeatable dierential cell counts [10]. A common measured in sputum are shown in Table 42.2.

BOX 42.2 PROTOCOL FOR PROCESSING OF SPUTUM.


Process at 4C within 2 h of expectoration

Select sputum (if necessary using inverted microscope)

Weight and incubate with 4 volume 0.1% dithiothreitol (DTT)

Gently aspirate with pasteur pipette, vortex for 15 s

Rock on bench rocker for 15 min on ice

Mix with equal volume (to DTT) of Dulbeccos phosphate buffered saline (DPBS)

Vortex for 15 s

Filter through 48 m nylon gauze (pre-wet with DPBS)

Centrifuge 790 g 10 min Store supernatant at 70C

Resuspend cell pellet in DPBS Measure fluid phase

Perform total cell count and viability by trypan blue exclusion


method in Neubauer hemocytometer

Adjust cell suspension to 0.50.75 106 cells/ml with DPBS

Prepare cytospins by placing 50 or 75 l cell suspension in cups of cytocentrifuge and


centrifuge at 450 rpm for 6 min

Air dry and stain

TABLE 42.2 Cell types and molecular markers that have been successfully measured in induced sputum.

Cytokines/
Cells Effector mediators Cellular markers chemokines

Eosinophils Leukotrienes Eosinophilic cationic Interleukin-8


Neutrophils C/D/E4 protein
Macrophages Prostaglandin D2 Neutrophil elastase
Lymphocytes Histamine
Epithelial
Mast cells

545
Asthma and COPD: Basic Mechanisms and Clinical Management

have similar cell and molecular characteristics and they can


It is generally agreed that the central airways are sam- be used interchangeably in asthma and COPD; there is
pled with induced sputum. This view is supported by stud- no evidence that pre-treatment with salbutamol or a prior
ies showing a greater proportion of granulocytes in both methacholine inhalation test inuences sputum cell counts.
sputum and bronchial samples compared with bronchoal- The sputum eosinophil count is responsive in that it
veolar lavage [1113] and by the demonstration that spu- increases when asthma worsens (e.g. after allergen challenge
tum induction results in greater clearance of radiolabeled and following relevant occupational exposures [24, 25]),
aerosol from the central airways than the peripheral air- and decreases when asthma improves with inhaled corti-
way [14]. There is evidence that increasing the duration of costeroid (ICS) treatment [26]. Sputum and bronchoscopy
sputum induction leads to sampling of more distal airways studies with corticosteroids [26, 27] and anti-IL-5 [28, 29]
although, as yet, the clinical utility of this technique has not suggest that the sputum eosinophil count is more responsive
been explored [15]. than tissue eosinophil counts. The sputum dierential neu-
trophil count is reduced following treatment with the xed
dose combination inhaler seretide in patients with COPD
Validation [30]. There are theoretical reasons to suggest that the total
neutrophil count may be a more responsive measure than
Sputum dierential cell counts have been shown to corre- the dierential count since the relationship between them
late closely with bronchial wash cell counts [16], less closely becomes relatively at over a dierential neutrophil count
with bronchoalveolar lavage counts [13, 16] and not at all of 80% [31].
with biopsy cell counts [17]. This may reect sampling of Based on what is known about the responsiveness
dierent airway compartments by the dierent techniques and repeatability of the sputum eosinophil count, a 2-fold
and the fact that some cell types (notably eosinophils and change is regarded as clinically signicant [32]. A reduc-
neutrophils) are not tissue dwelling. tion of the sputum neutrophil dierential cell count of 13%
Normal ranges have been published for a large adult or more has been regarded as signicant since longitudinal,
population [1820] (Table 42.3). Age has been shown observational studies suggest that a dierence of this mag-
to inuence dierential sputum neutrophil counts, with nitude is associated with a clinically important reduction
the higher values occurring in the older age groups [20]. in the rate of decline in lung function in patients with
Sputum dierential eosinophils, macrophage and neutrophil COPD [33].
counts and the sputum supernatant concentration of eosi-
nophilic cationic protein (ECP), cysteinylleukotrienes,
prostanoids, and IL-8 can be measured repeatably in asthma Findings in disease
[2123] and in COPD [5] (Table 42.3). The dierential
lymphocyte and epithelial cell count and the total cell count Clinically relevant patterns of airway inammation include
are less repeatable [22]. Spontaneous and induced sputum neutrophilic, eosinophilic, and mixed eosinophilic and

TABLE 42.3 Normal ranges and within subject repeatability of induced sputum cell counts in adults.

Normal ranges Repeatability

Author Belda [18] Spanevello [19] Thomas [20] Pizzichini [22] int Veen [21] Spanavello [23]

Number (male) 96 (54) 96 (46) 66 (24) 39 (20) 21 (10) 88 (44)

Characteristics Healthy Healthy Healthy Asthma (19) Asthma Asthma (53)


Healthy (10), Healthy (19)
Smokers (10) Rhinitis (16)

Mean age Not recorded Not recorded 46 (25) 39 24 (4) 38


6
TCC (10 /g) 4.1 (4.8) 2.7 (2.5) 2.1 (2.4) 0.35 0.44

Eosinophils (%) 0.4 (0.9) 0.6 (0.8) 0.3 (0.6) 0.94 (0.75a)b 0.85 (6.2) 0.84 (0.17a)

Neutrophils (%) 37.5 (20.1) 27.3 (13.0) 47.0 (27.0) 0.81 (14.0) 0.57 (15.5) 0.75

Macrophages (%) 58.8 (21.0) 69.2 (13.0) 49.0 (25.2) 0.71 0.64 (7.7) 0.76

Lymphocytes (%) 1.0 (1.1) 1.0 (1.2) 1.0 (1.4) 0.25 0.76 (3.9) 0.39

Epithelial cells (%) 1.6 (3.9) 1.5 (1.8) 2.5 (3.2) 0.64 (7.7) 0.56
a
Figures represent mean (SD). Repeatability expressed as intra-class correlation coefficient (within subject SD or log within subject SD). Repeatability was assessed over 2-days
(int Veen et al.), 6-days (Pizzichini et al.) and 1-week (Spanavello et al.). bWithin subject log standard deviation of sputum eosinophil count overly influenced by small absolute
differences in normals; in subjects with asthma it was 0.25.

546
Non-invasive Assessment of Airway Inflammation 42
Normal eosinophil count (1.9%) Raised eosinophil count

Paucigranulocytic Eosinophilic
Normal neutrophil
count (61%) Well controlled or intermittent asthma Asthma
Consider alternative diagnosis Eosinophilic bronchitis

Neutrophilic Mixed granulocytic


Acute infection (viral or bacterial) Exacerbations
Chronic infection (chlamydia, Refractory asthma
adenovirus) COPD
Raised neutrophil Smoking and COPD
count Environmental pollutants (ozone, NO2)
Occupational antigens
Endotoxin exposure FIG. 42.1 Classification based on induced
Obesity sputum patterns of cellular inflammation
Chronic cough syndromes in airway diseases.

neutrophilic (Fig. 42.1). The latter pattern is most com-


100
monly seen in COPD, refractory asthma, and in exacerba-
tions of airway disease. A sputum neutrophilia with a raised 80
total cell count suggests infective bronchitis, COPD, and
bronchiectasis; a neutrophilia with a normal total cell count 60
%
is more non-specic but is commonly seen in chronic cough
40
and non-eosinophilic asthma. A signicant minority of
patients with airway disease have paucigranulocytic sputum 20
characterized by normal sputum eosinophil and neutrophil
counts (Fig. 42.1).
A raised sputum eosinophil count is the most charac- Normal Cough COPD Asthma
teristic nding in patients with asthma. 6080% of patients
with symptomatic untreated asthma and around 50% of FIG. 42.2 Approximate prevalence of eosinophilic airway inflammation
(defined as a sputum eosinophil count 1.9%) in different patient
patients taking ICS have a sputum eosinophil count above
populations. Each dot represents an estimate of prevalence from a single
the normal range. A minority of patients with asthma study. Adapted from Gibson et al. [75].
of all severity grades have consistently non-eosinophilic
sputum; many have a sputum neutrophilia. A raised spu- amount of NO present in the gas sample. This technique
tum eosinophil count is not exclusive to asthma: 3040% is sensitive, allowing measurements down to 1 part per bil-
of patients with cough and a similar proportion of patients lion (ppb) [34]. FENO can be measured either oine or
with COPD have a sputum eosinophilia (Fig. 42.2). online. Online measurement involves the inhalation of
Roughly a half of patients with cough and a sputum eosi- NO free air immediately followed by exhalation at a steady
nophilia have evidence of variable airow obstruction and/ ow directly into the measuring apparatus, whereas in oine
or airway hyperresponsiveness and meet diagnostic crite- measurements the exhaled air is collected in a Mylar bal-
ria for cough variant asthma. The remainder consistently loon and then transported to the NO analyzer.
do not and are classied as having eosinophilic bronchitis. There is now a consensus that FENO is best meas-
Patients with COPD and a sputum eosinophilia may have ured before other spirometric maneuvers, at an exhaled rate
more asthma like features such as bronchodilator revers- of 50 ml/s maintained within 10% for 6 s, and with an
ibility, atopy, and airway hyperresponsiveness but there is oral pressure of 520 cmH2O to ensure velum closure [35].
too much overlap in the presence of these features to make The technique is simple and well within the scope of most
it possible to reliably predict pathology on the basis of patients in primary care, including children as young as 5
these tests. years of age [36]. Results are expressed as the NO concen-
tration in ppb (equivalent to nanoliters/liter) based on the
mean of two or three values within 10% [37]. NO output
can be calculated as the product of the exhaled NO con-
EXHALED NITRIC OXIDE centration in nl/l and the exhalation ow in l/min. Alveolar
NO concentration can be estimated from measurement
of NO output at multiple exhalation ows [38]. Alveolar
Methodology NO may reect distal lung inammation; it is increased in
patients with refractory asthma and is reduced by oral, but
FENO is usually measured by chemiluminescence. This uses not inhaled, corticosteroids [39]. The clinical role of this
ozone to react with NO and produce nitrogen dioxide which derived measurement has not been rmly established and it
then emits photon in a stoichiometric relationship with the will not be considered further.

547
Asthma and COPD: Basic Mechanisms and Clinical Management

Validation Findings in disease


Robust estimates of FENO measured using the above meth- Any form of airway inammation increases FENO levels,
ods are available in children [40] and, to a lesser extent, adults including that seen in bronchiectasis, viral infection, bro-
[37]. FENO tends to increase in healthy children to the age sing alveolitis, allergic rhinitis, pulmonary tuberculosis,
of 17; thereafter the values are similar to adults. There is no COPD, and pulmonary sarcoidosis, although the relation-
age eect in adults. There is conicting data on the eects of ship between inammation and FENO is complex as pneu-
gender [37, 40]. One study suggested uctuations in FENO monia [51], ciliary dyskinesia, and cystic brosis [52] have
in healthy pre-menopausal women related to the menstrual been shown to be associated with reduced FENO. Cross-
cycle, with peak values mid-cycle [41]. A reasonable esti- sectionally, the relationship between FENO levels and eosi-
mate of the normal ranges of FENO in adults is 30 ppb [37, nophilic airway inammation is much closer than the
40]; this is reduced to 24 ppb when outliers are removed relationship with other markers of airway inammation
from the analysis and 22.4 ppb when outliers and atopics [53] and it is not clear to what extent the nding of a raised
are removed. Various factors have been shown to inuence FENO are a true reection of the presence of eosinophilic
FENO and some do so independently of the presence of eosi- airway inammation in these conditions.
nophilic airway inammation. These factors and the likely
mechanism of the eect on FENO are summarized in Table
42.4. Genetic factors are known to aect the production of
NO: vans Gravesande et al. demonstrated a strong relation- OTHER METHODS TO MEASURE AIRWAY
ship between a known functional NOS3 missense sequence INFLAMMATION
variant in the endothelial nitric oxide gene (G894T) and
FENO level in a cohort of subjects with asthma [42], and the
number of alpha-1 antitrypsin (AAT) repeats in intron 20 of Exhaled condensate
this gene correlate with NO levels, with a higher number of
repeats associated with lower NO levels [43]. The collection of exhaled breath condensate (EBC) and
The between subject standard deviation of FENO is assay of inammatory markers is the most recent devel-
around 25 ppb in asthma and 8 ppb in normal controls [37]. opment in non-invasive asthma monitoring technologies.
The within subject standard deviation has been estimated at Methodological aspects of EBC collection and analysis have
1.62 ppb in a range of subjects [37, 40]. FENO is reduced been reviewed by a European Respiratory Society (ERS)
2- to 4-fold by corticosteroids in patients with asthma working party [54]. Water vapor and respiratory droplets and
[4446] and is increased (by about 60%) by during the late particles are cooled and collected and used for conventional
response to allergen in subjects with atopic asthma [47]. assay or inammatory markers such as leukotrienes [55],
There is a dose-related eect of low dose ICS on FENO but 8-isoprostane [56], pH [57], and hydrogen peroxide [5860].
no additional benet above a dose of budesonide of 400 g/ The current methods for the collection of EBC vary prima-
day [44, 48, 49]. Based on what is known about the eect of rily in the type of condensers that are employed. The physical
ICS on FENO and the predictive value of a change in FENO surface properties of each condenser system may inuence the
[50], a reasonable minimally important dierence is a 60% condensate that is collected. Therefore it is possible that there
change either way. may be variation between systems in terms of the particles

TABLE 42.4 Factors associated with increased and decreased FENO.

Factor Effect on FENO Likely mechanism

Age Increase until age 17, no effect thereafter Increased airway size

Gender Mixed evidence. If anything lower in females ? Increased airway size in males
independent of height but not a major effect

Height Small increase with increasing height Increased airway size

Atopy Increased in atopic patients Underlying eosinophilic airway inflammation, which may be
asymptomatic in some

Smoking Increased in non-smokers Unclear. May depend on time of last cigarette. Relationship between
FENO and sputum eosinophil count lost in smokers

Spirometry Transient reduction after spirometry Unclear. FENO should be done before other tests.

Rhinitis Increased, independent of atopy ? Increased eosinophilic airway inflammation

548
Non-invasive Assessment of Airway Inflammation 42
collected. The inuence of salivary contamination on EBC val- Serum ECP
ues may also be a problem as many of the mediators assayed
are found in high concentration in saliva [61]. The measure- ECP is a product of eosinophils and there has been inter-
ment characteristics of some of the mediators detectable in est in the use of serum, sputum or urinary ECP as a non-
EBC are summarized in Table 42.5. invasive marker of eosinophilic airway inammation. Serum
Assessment of airway inammation using EBC is ECP concentrations are higher than EDTA plasma concen-
non-invasive and feasible in almost all patients. It has the tration, probably because blood eosinophils continue to pro-
potential to provide information about dierent aspects duce ECP ex-vivo in the absence of additives. Serum ECP
of the airway inammatory response and this informa- concentrations are thought to be better at discriminating
tion may be clinically important. However, results are not health from disease and are preferred. A standardized collec-
available immediately, limiting the clinical utility of the tion, processing, and testing method has been described [65].
technique. Future technological advances may overcome Serum ECP increases with spontaneous allergen expo-
this problem. There is evidence of signicant variabil- sure or after a laboratory allergen challenge and decreases
ity in the concentration of some EBC markers and the following allergen avoidance and ICS therapy. However,
reported relationship between the concentration of EBC serum ECP is a less responsive measure than the sputum
markers and other inammatory or clinical measures is eosinophil count and FENO [66, 67]. Compared with eosi-
variable between studies raising the possibility of unre- nophil counts, ECP measurements in either induced spu-
solved methodological issues and a need for further valida- tum or serum failed to reect treatment-related changes in
tion work. chronic asthma [68], supporting the view that serum ECP
is not a sensitive or reliable means of evaluating eosinophilic
airway inammation. Moreover, serum ECP was found to
be insensitive marker in titrating and monitoring therapy
Exhaled breath temperature with ICS over a wide dose range in childhood asthma [69]
and it does not help predict a response to corticosteroid
Exhaled breath temperature is a potential marker of airway therapy [70]. Finally, a randomized trial which compared a
inammation as it is likely to reect vascular hyperperfusion serum ECP based algorithm with a conventional algorithm
secondary to tissue inammation and airway remodeling. for managing asthma found no improvement in symptom
Measurement of exhaled breath temperature and bronchial scores, in spite of increased doses of ICS [71]. These nd-
blood ow [62] is feasible in asthma [63] and COPD [64]. ings suggest that serum ECP is an imperfect and insensitive
Asthma is associated with increased exhaled breath temper- measure of eosinophilic airway inammation, which pro-
ature presumable reecting increased blood ow due to new vides no particular advantages over other techniques.
vessel formation and vasodilatation. Exhaled breath tem-
perature is not consistently increased in COPD, suggesting
dierences in bronchial blood ow and tissue remodeling.
Whether exhaled breath temperature provides clinically NEW INSIGHTS INTO THE IMPORTANCE OF
important information over and above other techniques
has not been established but if it does, then the simplicity
AIRWAY INFLAMMATION IN AIRWAY DISEASE
of the technique makes it an attractive option as a non-
invasive marker of airway inammation although the lack Our view of the importance of inammation in the patho-
of specic information on the pattern of airway inamma- genesis of asthma and other airway diseases has been heav-
tion may limit its clinical value. ily inuenced by bronchoscopy studies performed over the

TABLE 42.5 Measurement characteristics of markers assayed in exhaled breath condensate.

8-isoprostane Cysteinyl leukotrienes Hydrogen Peroxide pH

Methodology EIA, GCMS EIA De-aerated sample, pH


meter

Normal range ? ? ? 6.88.1

Repeatability ? ? Acceptable Acceptable

Result in asthma Increased 3-fold Increased 3-fold Increased Reduced

Effect of corticosteroid Inhaled: nil Nil Reduced pH normalises


Oral: reduced

Correlation with other FEV1: NS FEV1: NS Correlates with eosinophils Correlated with
markers eosinophils, lung
function

Notes: EIA: enzyme immunoassay; GCMS: gas chromatographymass spectrometry; NS: not significant.

549
Asthma and COPD: Basic Mechanisms and Clinical Management

last 20 years [72]. These, by necessity, were largely limited 0.25 **p  0.01
to young volunteers with mild disease. The development of
a non-invasive technique to assess airway inammation has 0.20
made it possible to relate the presence of airway inamma-
tion to objective measures of disordered airway function in
larger and more heterogeneous populations than was pos- 0.15

D FEV1 (I)
sible with bronchoscopy studies. These studies have sug-
gested a complex relationship between airway inammation 0.10
and the pattern and severity of airway dysfunction [8, 73,
74]. Clinicians and researchers interested in using measures
0.05
of airway inammation need to be aware of this relation-
ship in order to fully appreciate the added value such meas-
ures provide. A number of key observations are particularly 0.00
important. All have been primarily made using induced
sputum as the measure of airway inammation but they 0.05
are potentially relevant to the use of FENO given the 1.3 1.3 4.5 4.5
close relationship between FENO and eosinophilic airway Eosinophil count
inammation.
Firstly, the presence of pattern of airway inam- FIG. 42.3 Mean net change in FEV1 with after 2 weeks treatment with
mation is not closely related to either the pattern or the prednisone 30 mg od in 69 patients with moderate and severe COPD
severity of the airway dysfunction or symptoms. A raised participating in a randomized, double blind placebo controlled trial.
Patients are stratified into tertiles based on baseline sputum eosinophil
sputum eosinophil count is seen in 6080% of corticosteroid
count. Reproduced with permission from Brightling et al. [77].
naive patients with asthma, 50% of corticosteroid treated
patients with symptomatic asthma [75], 3040% of patients
with cough [76], and up to 40% of patients with COPD
[77]. Within diagnostic groups there is a weak correlation Thirdly, the sputum eosinophil count is a better marker
between the presence of eosinophilic airway inammation for titrating corticosteroid therapy than standard clinical
and the severity of symptoms or disordered airway function measures. Studies in asthma [6, 83] and COPD [84] have
[73]. Thus, little can be deduced about the presence, nature shown that management strategies where decisions about
and severity of airway inammation from a standard clinical corticosteroid use and dose are guided by the sputum eosi-
assessment. nophil count results in a lower frequency of exacerbations and
This observation is important as it strongly implies more economical use of corticosteroids than management
that there is not a close causal link between eosinophilic air- guided by traditional clinical measures (Fig. 42.4). The nd-
way inammation and the airway dysfunction that underlies ings of these studies, together with evidence that a sputum
many of the day-to-day clinical manifestations of asthma. eosinophilia is an important and independent predictor of the
Further support for this view is provided by studies show- occurrence of an asthma exacerbation following corticosteroid
ing that treatment with anti-IL-5 markedly reduced eosi- withdrawal [85, 86], suggest that eosinophilic airway inam-
nophilic airway inammation but does not aect asthma mation is a more valid surrogate marker of preventable exac-
symptoms, lung function or airway responsiveness [29, 78]. erbation risk than other currently available markers.
A comparative bronchial biopsy study of asthma and eosi- Fourthly, there is a clear and consistent positive cor-
nophilic bronchitis (a condition where one sees eosinophilic relation between a raised sputum neutrophil count and
airway inammation in the absence of the abnormalities of the presence of xed airow obstruction as reected by a
airway function that characterize asthma) has shown that reduced post-bronchodilator FEV1 and FEV1/FVC. This is
inltration of the airway smooth muscle with mast cell is seen in patients with asthma [87] and COPD [88] and in
only seen in asthma [79]. Moreover, the extent of inltra- COPD a raised sputum neutrophil count is associated with
tion is closely related to airway hyperresponsiveness imply- longitudinal decline in FEV1 [33]. These ndings raise the
ing that it is the interaction between mast cells and airway possibility of a common mechanism for the development
smooth muscle that is of fundamental importance in the of xed airow obstruction involving neutrophilic airway
genesis of airway hyperresponsiveness. inammation. Much more information is required before
Secondly, the presence of eosinophilic airway inam- we can be sure that there is a causal link between neu-
mation is more closely associated with a positive response trophilic airway inammation and xed airow obstruction.
to corticosteroids than other clinical measures. Moreover, In particular, there is a need for longitudinal studies evalu-
a positive response to corticosteroids is seen irrespective of ating the eect of reducing neutrophilic airway inamma-
the pattern of airway disease in which eosinophilic airway tion. Nevertheless, there is sucient evidence to speculate
inammation occurs (Fig. 42.3) [26, 77, 8082]. Thus, if the that a raised sputum neutrophil count is a useful biomarker
clinical questions were whether a patient with symptoms of risk of subsequent decline in lung function.
suggesting airway disease should receive corticosteroid treat- The new understanding of the importance of airway
ment (as it often is), then the identication of eosinophilic inammation in airway disease has opened the way for
airway inammation would be a better basis for making this a new approach to the management of airways disease in
decision than the ndings of other tests. clinical practice where assessment of airway inammation

550
Non-invasive Assessment of Airway Inflammation 42
Results: Severe exacerbations
*p 0.01
120

100 BTS guidelines


Severe exacerbations
(cumulative number)

Six asthma
80 admissions

60

40
Sputum guidelines
FIG. 42.4 Comparison of effects of two treatment
20
One asthma strategies on rates of severe exacerbations of asthma.
admission One strategy (British Thoracic Society (BTS) guidelines)
0
utilized standard guidelines of the BTS and the other
(sputum guidelines) adjusted the anti-inflammatory
0 1 2 3 4 5 6 7 8 9 10 11 12
treatment with corticosteroids based on the eosinophil
Time (months) counts. Reproduced from Green et al. [6].

is used to identify risk of future adverse outcome and likely for either high (50 ppb) or low (25 ppb) FENO levels
response to treatment. are highly signicant and may be used with condence to
guide decision-making [94, 95]. More needs to be learnt
about the optimum cut-points for FENO as a marker of
corticosteroid responsive airway disease and as a tool for
FENO AS A MEANS OF MONITORING asthma monitoring, but the evidence to date suggests that
the use of regular FENO measurements is a promising way
AIRWAY INFLAMMATION to achieve more economical and eective use of ICS [96].

The main limitations of induced sputum are that the tech-


nique is labor intensive, it requires a signicant amount of
training to get reliable results and results are not available
immediately, limiting the application of the technique in POTENTIAL CLINICAL ROLE OF
disease monitoring. FENO has the advantages of being sim- INFLAMMOMETRY
ple to measure and providing an immediate result making it
much more suitable for a monitoring tool.
FENO is a reasonably robust measure of the presence Role in diagnosis
of eosinophilic airway inammation across a wide range of
patients diering in atopic status, diagnosis, and medication None of the currently available diagnostic tests are su-
[53]. The association is lost in current smokers [53]. As for ciently sensitive to rule out asthma [97, 98] with the result
sputum eosinophils, it has been shown that a raised FENO that treatment trials are often instigated without good evi-
level is a reliable indicator of a positive response to corti- dence variable airow obstruction, airway hyperresponsive-
costeroids in a heterogeneous population of patients with ness or airway inammation. One study has shown that
symptoms suggesting airway disease [89]. This nding was out of 263 subjects referred to a tertiary referral center
independent of the clinical diagnosis at presentation, in with suspected asthma, 160 received an alternative diagno-
particular the label of asthma. One corticosteroid reduc- sis [99]. Many of these had received prolonged treatment
tion study has shown that a raised FENO is predictive of with potentially toxic therapy before the correct diagnosis
loss of asthma control [50] but two others have shown less was reached. Even in tertiary referral centers the diagnosis
convincing evidence for such an eect [86, 90]. The rela- of refractory asthma can be dicult to make with certainty
tionship between airway eosinophilia, FENO levels, steroid [100]. The presence of a sputum eosinophilia or raised
responsiveness, and exacerbation risk raises the possibil- FENO in asthma is suciently common to suggest that
ity that FENO can be used as a guide to corticosteroid dose these ndings may have a role in the diagnosis of asthma.
requirements. This has been investigated in a number of Hunter et al. [98] showed that the sensitivity of a sputum
small studies in dierent populations [9193]. The ndings eosinophil count outside the normal range in identifying
show a consistent trend to reduction in asthma exacerba- asthma (dened as consistent symptoms with objective evi-
tion of around 25%, in one study this was achieved with a dence of abnormal variable airow obstruction) was around
40% lower mean daily dose of ICS [91]. More denitively, 80%, signicantly better than PEF amplitude % mean and
studies have shown that FENO levels may be used to iden- the acute bronchodilator response and approached the sen-
tify which patients do and do not require long-term main- sitivity and specicity of measurement of airway respon-
tenance therapy with inhaled steroids: the predictive values siveness. Smith et al. [97] have reported similar ndings; in

551
Asthma and COPD: Basic Mechanisms and Clinical Management

this study a high FENO concentration achieved a similarly Role in monitoring asthma
high diagnostic accuracy. Similar results have been reported
by Berkman et al. [101] and Dupont et al. [102]. FENO Once a decision is made to start corticosteroid treat-
performed less well in a community study [103], possi- ment the next question is how best to titrate this therapy.
bly because the diagnosis of asthma was less rigorous and Traditionally dose titration is done by assessing the clini-
because subjects were tested whilst taking ICS. cal response to treatment and attempting to dene the low-
Arguably it matters more to patients what can be est dose of ICS that maintains this. Many patients do very
done to help them than the diagnostic label attached to well with this approach [108] but there is evidence that
them. Thus, testing strategies that identify patients who are the use of the induced sputum eosinophil count to titrate
going to respond well to corticosteroid and provide guid- therapy results in a lower exacerbation frequency with
ance on the dosing of corticosteroids might be particularly no overall increase in treatment, particularly in patients
helpful. There is now compelling evidence that the presence with more severe asthma (Fig. 42.4) [6, 83]. A number of
of a sputum eosinophilia or a raised FENO predicts corticos- researchers have investigated whether a similar eect is seen
teroid responsiveness. This was rst clearly demonstrated by using FENO as the monitoring tool (Table 42.6). Smith
Morrow-Brown in the 1950s who showed that patients with et al. [91] used FENO measurements at a ow of 250ml/s
airway disease and a sputum eosinophilia responded to cor- to titrate ICS in a single blind parallel group trial involv-
ticosteroid treatment whereas those without a sputum eosi- ing 92 subjects with asthma. Subjects were treated accord-
nophilia did not [104]. It has since been shown that patients ing to their FENO measurements or Global Initiative for
with non-eosinophilic asthma respond less well to inhaled Asthma (GINA) guidelines. Following a run in period the
budesonide than a group with more typical sputum features steroid dose was reduced in the FENO group if the FENO
[26, 105]. This is also the case with longer-term corticoster- was 15 ppb (equivalent to an FENO of about 45 ppb using
oid treatment in patients with more severe asthma [6]. A an expiratory ow of 50 ml/s). In the control group, ster-
sputum eosinophilia or raised FENO [91] is a predictor of a oid reduction was based on current GINA guidelines and
steroid response irrespective of the clinical context: patients only occurred when the subjects had, over the course of
with chronic cough respond well to inhaled corticosteroids the previous week, achieved all of the following: less than
if there is a sputum eosinophilia [80, 106] and patients with 2 night-time awakenings, a mean peak ow amplitude of
COPD with a sputum eosinophilia respond better to sys- 20%, bronchodilator use 4 times on 1 or 2 days, mini-
temic and ICS than those without [77, 81, 107]. mal asthma symptoms, and a FEV1  90% predicted.

TABLE 42.6 Summary of studies investigation FENO as a tool for monitoring corticosteroid treatment in asthma.

Reduction in
exacerbation Change in ICS
Study Design Population Duration Intervention frequency dose

Smith et al. [91] Single blind, 97 adults with mild- 12 months, after Control: conventional 0.49 moderate and 40% lower in
parallel group moderate asthma an optimization measures of asthma severe exacerbation/ intervention
phase of up to control Intervention: patient/year in group
12 months FENO  15* ppb intervention and 0.9 (p 0.003)
in control (p 0.27)

Pijnenburg Single blind, 85 children with 12 months Control: control of Eight courses of Cumulative
et al. [92] parallel group atopic asthma symptoms Intervention: prednisolone in dose over 5
Control of symptoms intervention and 18 visits of 4407 g
and FENO  30 ppb in control; p 0.6 in intervention,
4332 g in
control

Shaw et al. [93] Single blind, 118 adults with 12 months Control: control of 0.42 severe 12% higher in
parallel group a primary care symptoms Intervention: exacerbation/ intervention
diagnosis of asthma FENO  26 ppb patient/year in group (p 0.4)
control and 0.33
in intervention
(p 0.4)

Note: ICS: inhaled corticosteroid.


*
Measured at 250 ml/s.

552
Non-invasive Assessment of Airway Inflammation 42
In the FENO group the optimal dose was one dose above the year. The study was underpowered to show a clinically
the dose at which the subjects FENO was 15 ppb. In the important eect on exacerbation frequency although the
control group the optimal dose was one dose above the dose eect on severe exacerbation frequency was similar to that
at which a loss of control had occurred. These became the seen by Smith et al.
optimal doses at which subjects entered the nal year of the The results of corticosteroid dosetitration studies
study. During this stage the steroid dose was increased in with FENO are therefore mixed. There is a fairly consist-
the FENO group if the FENO was 15 ppb and in the con- ent nding of a small reduction in severe asthma exacerba-
trol group the optimal dose was increased if a loss of con- tions in patients managed with reference to FENO (Table
trol occurred. Although there were fewer exacerbations in 42.6) but a much larger study will be needed to deter-
the FENO group, this did not reach statistical signicance. mine whether such an eect is real, and whether it can be
However, over the year of the study the FENO managed achieved cost eectively and with no overall increase in cor-
group used 40% less ICS. ticosteroid therapy.
A similar study in a pediatric population showed a The induced sputum studies showing a reduction in
signicant improvement in airway responsiveness in chil- exacerbation frequency have generally included a more
dren whose FENO was maintained below 30 ppb when severe patient population than the FENO studies and it is
compared to a population whose corticosteroid dose was possible that more clear evidence of benet of FENO moni-
titrated using traditional measures [92]. Few exacerbations toring would be seen in this population. In support of this,
were seen with either management strategy and there was Jayaram et al. [83] noted that the benet of sputum eosi-
no overall reduction in corticosteroid dose. nophil directed management was much more clearly seen
Shaw et al. [93] took a dierent approach in a single in patients with more severe asthma. Patients taking long-
blind parallel group trial of 118 adults with asthma man- acting 2-agonists did particularly well and it is possible
aged in primary care. In the control group the goal of treat- that the increased dissociation between eosinophilic air-
ment was to maintain the Juniper asthma control score way inammation and symptoms seen after treatment with
below 1.57 [109]; in the intervention group the aim was long-acting 2-agonists [110] is an important determinate
to achieve this and to maintain the FENO between 16 and of this. FENO is measurable in most patients with severe
26 ppb. Exacerbation frequency assessed over 12 months asthma and it appears to be as good a marker of eosinophilic
was 21% lower (p 0.4) in the intervention group and airway inammation as it is in less severe asthma [111], so
there was no overall dierence in corticosteroid dose over future studies should evaluate its use in severe asthma. In a

Symptoms suggesting airway disease

Assess for possible eosinophilic


airway inflammation

Raised FENO Normal or low FENO or


(25 ppb) or sputum sputum eosinophils*** FIG. 42.5 Suggested algorithm for
eosinophils assessment of patients presenting with
untreated airway disease. Eosinophilic airway
inflammation can be assessed using induced
Identify and treat sputum eosinophils or FENO. *Potentially
Corticosteroid therapy aggravating factors*
treatable aggravating factors include
Symptomatic therapy**
rhinitis, anxietyhyperventilation syndrome,
vocal cord dysfunction, bronchiectasis,
and gastroesophageal reflux disease.
Normal or low FENO or **Symptomatic therapy includes short- and
Adequate response Inadequate response
sputum eosinophils long-acting bronchodilator therapy, oral
theophylline, and mucolytics as well as
specific treatments for the aggravating factors.
***The suggested normal range for sputum
Raised sputum eosinophils
eosinophils is 2% and FENO in adults is
25 ppb. FENO  50 ppb is very predictive of a
response to corticosteroid and a relapse when
Continue treatment Taking treatment correctly? treatment is withdrawn; the interpretation of
FENO in the range 2550 ppb varies between
No Yes individual patients, and in some individuals
different cut-points may apply. The induced
Address compliance issues Increase dose sputum eosinophil count is likely to be a more
Check inhaler technique Consider referral reliable indicator of corticosteroid response
and relapse risk when FENO is in this range.

553
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 42.7 An approach to monitoring ICS treatment in patients with airway disease using FENO.

FENO Low Normal Intermediate High

Ppb 5 525 2550 50

Symptoms controlled Decrease/stop ICS Decrease/stop ICS No change Increase ICS/refer

Symptoms uncontrolled Decrease ICS, increase Decrease/stop ICS, Assess compliance. Assess compliance.
bronchodilators. Investigate increase bronchodilators Increase ICS Increase ICS/refer
for PCD and CF

Notes: ppb: parts per billion; PCD: primary ciliary dyskinesia; CF: cystic fibrosis.

less severe population the main impact of inammometry There is an urgent need for a non-invasive technique
might be a reduction in corticosteroid dose without loss of capable of providing accurate information on the character-
ecacy. istics of the lower airway inammatory response from an
Trials evaluating FENO as a monitoring tool for easily accessible sample. The technique should be capable
asthma have used a wide variety of target ranges for FENO of providing an immediate result and be useable in acute as
(Table 42.6). Since than there has been an increase in our well as outpatient settings. Whether FENO or EBC analysis
understanding of the relationship between FENO and eosi- as it currently stands are ideal for this purpose is doubtful.
nophilic airway inammation an there is now a consensus Renements of breath analysis, or perhaps identication of
that an FENO of 25 ppb is a sensitive indicator of the novel biomarkers in urine or blood are perhaps most likely
absence of eosinophilic airway inammation [93, 96] and is to advance this area. Work already done with induced spu-
therefore an appropriate cut o for a reduction in ICS [112]. tum provide a strong scientic rationale for the use of these
Conversely, an FENO above 50 ppb is a specic marker for markers as a means of dening risk and likely treatment
the presence of eosinophilic airway inammation [93, 96] response.
and a good response to corticosteroids is consistently seen
above this value [89]. These therefore would appear to be the
most appropriate values for a reduction and an increase in References
corticosteroid dose. Values lying between 25 and 50 ppb are
indeterminate and clinical criteria should be used to guide 1. Paggiaro PL, Chanez P, Holz O, Ind PW, Djukanovic R, Maestrelli P
corticosteroid treatment. A suggested approach to interpre- et al. Sputum induction. Eur Respir J Suppl 37: 3ss8s, 2002.
tation of FENO values in the diagnosis and monitoring of 2. Hunter CJ, Ward R, Woltmann G, Wardlaw AJ, Pavord ID. The safety
asthma is outlined in Fig. 42.5 and Table 42.7. and success rate of sputum induction using a low output ultrasonic
nebuliser. Respir Med 93(5): 34548, 1999.
3. de la Fuente PT, Romagnoli M, Godard P, Bousquet J, Chanez P.
Safety of inducing sputum in patients with asthma of varying severity.
Am J Respir Crit Care Med 157(4 Pt 1): 112730, 1998.
FUTURE DIRECTIONS 4. ten Brinke A, de Lange C, Zwinderman AH, Rabe KF, Sterk PJ,
Bel EH. Sputum induction in severe asthma by a standardized proto-
col: Predictors of excessive bronchoconstriction. Am J Respir Crit Care
Recently it has become possible to investigate the contribution Med 164(5): 74953, 2001 September 1.
of the distal lung in the inammatory process in asthma and 5. Brightling CE, Monterio W, Green RH, Parker D, Morgan MD,
other airway diseases by measurement of FENO at dierent Wardlaw AJ et al. Induced sputum and other outcome measures in
chronic obstructive pulmonary disease: Safety and repeatability. Respir
expiratory ows [38]. Lehtimaki et al. have used this method
Med 95(12): 9991002, 2001.
to demonstrate that patients with nocturnal symptoms have 6. Green RH, Brightling CE, McKenna S, Hargadon B, Parker D,
elevated alveolar NO concentrations [113] and that alveo- Bradding P et al. Asthma exacerbations and sputum eosinophil counts:
lar NO concentration does not fall in response to treatment A randomised controlled trial. Lancet 360(9347): 171521, 2002
with ICS [114]. Patients with refractory asthma have a raised November 30.
alveolar NO concentration which is reduced by oral but not 7. Pavord ID. Sputum induction to assess airway inammation: Is it an
ICS [39] suggesting that it reects corticosteroid responsive inammatory stimulus?. Thorax 53(2): 7980, 1998 February 1.
inammation in a site that cant be accessed by ICS. Induced 8. Green RH, Brightling CE, Woltmann G, Parker D, Wardlaw AJ,
sputum may also have a role in investigating proximal and Pavord ID. Analysis of induced sputum in adults with asthma:
distal airway inammation as there is evidence that distal Identication of subgroup with isolated sputum neutrophilia and poor
response to inhaled corticosteroids. Thorax 57(10): 87579, 2002.
airways are sampled with increasing output and duration of
9. Efthimiadis A, Jayaram L, Weston S, Carruthers S, Hargreave FE.
nebulized hypertonic saline [15]. The clinical utility of these Induced sputum: Time from expectoration to processing. Eur Respir J
methods has not been explored. They could provide useful 19(4): 7068, 2002.
information on the need for a systemic rather than an inhaled 10. Ward R, Woltmann G, Wardlaw AJ, Pavord ID. Between-observer
approach to anti-inammatory treatment with agents such as repeatability of sputum dierential cell counts. Inuence of cell viability
prednisolone, anti-IgE, and perhaps anti-IL-5. and squamous cell contamination. Clin Exp Allergy 29(2): 24852, 1999.

554
Non-invasive Assessment of Airway Inflammation 42
11. Alexis N, Soukup J, Ghio A, Becker S. Sputum phagocytes from 29. Leckie MJ, ten BA, Khan J, Diamant Z, OConnor BJ, Walls CM
healthy individuals are functional and activated: A ow cytometric et al. Eects of an interleukin-5 blocking monoclonal antibody on eosi-
comparison with cells in bronchoalveolar lavage and peripheral blood. nophils, airway hyper-responsiveness, and the late asthmatic response.
Clin Immunol 97(1): 2132, 2000. Lancet 356(9248): 214448, 2000 December 23.
12. Moodley YP, Krishnan V, Lalloo UG. Neutrophils in induced sputum 30. Barnes NC, Qiu YS, Pavord ID, Parker D, Davis PA, Zhu J et al.
arise from central airways. Eur Respir J 15(1): 3640, 2000. Antiinammatory eects of salmeterol/uticasone propionate in
13. Pizzichini E, Pizzichini MM, Kidney JC, Efthimiadis A, Hussack P, chronic obstructive lung disease. Am J Respir Crit Care Med 173(7):
Popov T et al. Induced sputum, bronchoalveolar lavage and blood from 73643, 2006 April 1.
mild asthmatics: Inammatory cells, lymphocyte subsets and soluble 31. Neale N, Parker D, Barlow S, Green RH, Brightling CE, Pavord ID.
markers compared. Eur Respir J 11(4): 82834, 1998. The relationship between total and dierential cell counts in induced
14. Alexis NE, Hu SC, Zeman K, Alter T, Bennett WD. Induced spu- sputum. Eur Respir J 20: 274s, 2002.
tum derives from the central airways: Conrmation using a radiola- 32. Pavord ID, Sterk PJ, Hargreave FE, Kips JC, Inman MD, Louis R
beled aerosol bolus delivery technique. Am J Respir Crit Care Med 164 et al. Clinical applications of assessment of airway inammation using
(10 Pt 1): 196470, 2001. induced sputum. Eur Respir J 37(Suppl.): 40s43s, 2002.
15. Gershman NH, Liu H, Wong HH, Liu JT, Fahy JV. Fractional analy- 33. Stanescu D, Sanna A, Veriter C, Kostianev S, Calcagni PG, Fabbri LM
sis of sequential induced sputum samples during sputum induction: et al. Airways obstruction, chronic expectoration, and rapid decline of
Evidence that dierent lung compartments are sampled at dierent FEV1 in smokers are associated with increased levels of sputum neu-
time points. J Allergy Clin Immunol 104(2 Pt 1): 32228, 1999. trophils. Thorax 51(3): 26771, 1996.
16. Keatings VM, Evans DJ, OConnor BJ, Barnes PJ. Cellular proles in 34. Lundberg JO, Weitzberg E, Lundberg JM, Alving K. Nitric oxide in
asthmatic airways: A comparison of induced sputum, bronchial wash- exhaled air. Eur Respir J 9(12): 267180, 1996.
ings, and bronchoalveolar lavage uid. Thorax 52(4): 37274, 1997. 35. ATS/ERS Recommendations for Standardized Procedures for the
17. Maestrelli P, Saetta M, Di SA, Calcagni PG, Turato G, Ruggieri MP Online and Oine Measurement of Exhaled Lower Respiratory
et al. Comparison of leukocyte counts in sputum, bronchial biopsies, Nitric Oxide and Nasal Nitric Oxide, 2005. Am J Respir Crit Care Med
and bronchoalveolar lavage. Am J Respir Crit Care Med 152(6 Pt 1): 171(8): 91230, 15 April 2005.
192631, 1995. 36. Gruyd-Jones K, Ward S, Stonham C, Macfarlane TV, Thomas M. The
18. Belda Jose, Leigh Rich, Parameswaran Kris, OByrne PM, Sears MR, use of exhaled nitric oxide monitoring in primary care asthma clinics:
Hargreave FE. Induced sputum cell counts in healthy adults. Am J A pilot study. Prim Care Respir J 16: 34956, 2007.
Respir Crit Care Med 161(2): 47578, 2000 February 1. 37. Kharitonov SA, Gonio F, Kelly C, Meah S, Barnes PJ. Reproducibility
19. Spanevello A, Confalonieri M, Sulotto F, Romano F, Balzano G, of exhaled nitric oxide measurements in healthy and asthmatic adults
Migliori GB et al. Induced sputum cellularity. Reference values and and children. Eur Respir J 21(3): 43338, 2003.
distribution in normal volunteers. Am J Respir Crit Care Med 162 38. Tsoukias NM, George SC. A two-compartment model of pulmonary
(3 Pt 1): 117274, 2000. nitric oxide exchange dynamics. J Appl Physiol 85(2): 65366, 1998.
20. Thomas RA, Green RH, Brightling CE, Birring SS, Parker D, 39. Berry M, Hargadon B, Morgan A, Shelley M, Richter J, Shaw D
Wardlaw AJ et al. The inuence of age on induced sputum dierential et al. Alveolar nitric oxide in adults with asthma: Evidence of distal
cell counts in normal subjects. Chest 126(6): 181114, 2004. lung inammation in refractory asthma. Eur Respir J 25(6): 98691,
21. int Veen JC, de Gouw HW, Smits HH, Sont JK, Hiemstra PS, 2005.
Sterk PJ et al. Repeatability of cellular and soluble markers of inam- 40. Buchvald F, Baraldi E, Carraro S, Gaston B, De JJ, Pijnenburg MW
mation in induced sputum from patients with asthma. Eur Respir J et al. Measurements of exhaled nitric oxide in healthy subjects age 4 to
9(12): 244147, 1996. 17 years. J Allergy Clin Immunol 115(6): 113036, 2005.
22. Pizzichini E, Pizzichini MM, Efthimiadis A, Evans S, Morris MM, 41. Kharitonov SA, Logan-Sinclair RB, Busset CM, Shinebourne EA.
Squillace D et al. Indices of airway inammation in induced sputum: Peak expiratory nitric oxide dierences in men and women: Relation
Reproducibility and validity of cell and uid-phase measurements. to the menstrual cycle. Br Heart J 72(3): 24345, 1994.
Am J Respir Crit Care Med 154(2 Pt 1): 113844, 1996. 42. vans Gravesande KS, Wechsler ME, Grasemann H, Silverman ES,
23. Spanevello A, Migliori GB, Sharara A, Ballardini L, Bridge P, Pisati P Le L, Palmer LJ et al. Association of a missense mutation in the NOS3
et al. Induced sputum to assess airway inammation: a study of repro- gene with exhaled nitric oxide levels. Am J Respir Crit Care Med 168(2):
ducibility. Clin Exp Allergy 10: 113844, 1997. 22831, 2003 July 15.
24. Lemiere C, Pizzichini MM, Balkissoon R, Clelland L, Efthimiadis A, 43. Henriksen AH, Sue-Chu M, Lingaas HT, Langhammer A, Bjermer L.
OShaughnessy D et al. Diagnosing occupational asthma: Use of Exhaled and nasal NO levels in allergic rhinitis: Relation to sensiti-
induced sputum. Eur Respir J 13(3): 48288, 1999. zation, pollen season and bronchial hyperresponsiveness. Eur Respir J
25. Pizzichini MM, Kidney JC, Wong BJ, Morris MM, Efthimiadis A, 13(2): 3016, 1999.
Dolovich J et al. Eect of salmeterol compared with beclomethasone on 44. Jatakanon A, Kharitonov S, Lim S, Barnes PJ. Eect of diering doses
allergen-induced asthmatic and inammatory responses. Eur Respir J of inhaled budesonide on markers of airway inammation in patients
9(3): 44955, 1996. with mild asthma. Thorax 54(2): 10814, 1999.
26. Pavord ID, Brightling CE, Woltmann G, Wardlaw AJ. Non- 45. Massaro AF, Gaston B, Kita D, Fanta C, Stamler JS, Drazen JM.
eosinophilic corticosteroid unresponsive asthma. Lancet 353(9171): Expired nitric oxide levels during treatment of acute asthma. Am J
221314, 1999 June 26. Respir Crit Care Med 152(2): 8003, 1995.
27. Bentley AM, Hamid Q , Robinson DS, Schotman E, Meng Q , Assou B 46. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R, Shinebourne
et al. Prednisolone treatment in asthma. Reduction in the numbers EA, Barnes PJ. Increased nitric oxide in exhaled air of asthmatic
of eosinophils, T cells, tryptase-only positive mast cells, and modula- patients. Lancet 343(8890): 13335, 1994 January 15.
tion of IL-4, IL-5, and interferon-gamma cytokine gene expression 47. Kharitonov SA, OConnor BJ, Evans DJ, Barnes PJ. Allergen-induced
within the bronchial mucosa. Am J Respir Crit Care Med 153(2): late asthmatic reactions are associated with elevation of exhaled nitric
55156, 1996. oxide. Am J Respir Crit Care Med 151(6): 189499, 1995.
28. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig 48. Jones SL, Herbison P, Cowan JO, Flannery EM, Hancox RJ,
MS et al. Anti-IL-5 treatment reduces deposition of ECM proteins in McLachlan CR et al. Exhaled NO and assessment of anti-inamma-
the bronchial subepithelial basement membrane of mild atopic asth- tory eects of inhaled steroid: Doseresponse relationship. Eur Respir J
matics. J Clin Invest 112(7): 102936, 2003. 20(3): 6018, 2002.

555
Asthma and COPD: Basic Mechanisms and Clinical Management

49. Kharitonov SA, Donnelly LE, Montuschi P, Corradi M, Collins JV, 69. Visser MJ, Postma DS, Brand PL, Arends LR, Duiverman EJ,
Barnes PJ. Dose-dependent onset and cessation of action of inhaled Kauman HF. Inuence of dierent dosage schedules of inhaled u-
budesonide on exhaled nitric oxide and symptoms in mild asthma. ticasone propionate on peripheral blood cytokine concentrations in
Thorax 57(10): 88996, 2002. childhood asthma. Clin Exp Allergy 32(10): 1497503, 2002.
50. Jones SL, Kittelson J, Cowan JO, Flannery EM, Hancox RJ, 70. Meijer RJ, Postma DS, Kauman HF, Arends LR, Koeter GH,
McLachlan CR et al. The predictive value of exhaled nitric oxide meas- Kerstjens HA. Accuracy of eosinophils and eosinophil cationic pro-
urements in assessing changes in asthma control. Am J Respir Crit Care tein to predict steroid improvement in asthma. Clin Exp Allergy 32(7):
Med 164(5): 73843, 2001 September 1. 1096103, 2002.
51. Adrie C, Monchi M, Dinh-Xuan AT, DallAva-Santucci J, Dhainaut JF, 71. Lowhagen O, Wever AM, Lusuardi M, Moscato G, De Backer WA,
Pinsky MR. Exhaled and nasal nitric oxide as a marker of pneumonia in Gandola L et al. The inammatory marker serum eosinophil cationic
ventilated patients. Am J Respir Crit Care Med 163(5): 114349, 2001. protein (ECP) compared with PEF as a tool to decide inhaled corti-
52. Balfour-Lynn IM, Laverty A, Dinwiddie R. Reduced upper airway costeroid dose in asthmatic patients. Respir Med 96(2): 95101, 2002.
nitric oxide in cystic brosis. Arch Dis Child 75(4): 31922, 1996. 72. Djukanovic R, Roche WR, Wilson JW, Beasley CR, Twentyman OP,
53. Berry MA, Shaw DE, Green RH, Brightling CE, Wardlaw AJ, Howarth RH et al. Mucosal inammation in asthma. Am Rev Respir
Pavord ID. The use of exhaled nitric oxide concentration to identify Dis 142(2): 43457, 1990.
eosinophilic airway inammation: An observational study in adults with 73. Rosi E, Ronchi MC, Grazzini M, Duranti R, Scano G. Sputum analysis,
asthma. Clin Exp Allergy 35(9): 117579, 2005. bronchial hyperresponsiveness, and airway function in asthma: Results
54. Horvath I, Hunt J, Barnes PJ, Alving K, Antczak A, Baraldi E et al. of a factor analysis. J Allergy Clin Immunol 103(2 Pt 1): 23237, 1999.
Exhaled breath condensate: Methodological recommendations and 74. Crimi E, Spanevello A, Neri M, Ind PW, Rossi GA, Brusasco V.
unresolved questions. Eur Respir J 26(3): 52348, 2005. Dissociation between airway inammation and airway hyperrespon-
55. Montuschi P, Barnes PJ. Exhaled leukotrienes and prostaglandins in siveness in allergic asthma. Am J Respir Crit Care Med 157(1): 49,
asthma. J Allergy Clin Immunol 109(4): 61520, 2002. 1998.
56. Mondino C, Ciabattoni G, Koch P, Pistelli R, Trove A, Barnes PJ 75. Gibson PG, Fujimura M, Niimi A. Eosinophilic bronchitis: Clinical man-
et al. Eects of inhaled corticosteroids on exhaled leukotrienes and ifestations and implications for treatment. Thorax 57(2): 17882, 2002.
prostanoids in asthmatic children. J Allergy Clin Immunol 114(4): 761 76. Brightling CE, Ward R, Goh KL, Wardlaw AJ, Pavord ID.
67, 2004. Eosinophilic bronchitis is an important cause of chronic cough. Am J
57. Jobsis Q , Raatgeep HC, Schellekens SL, Hop WC, Hermans PW, Respir Crit Care Med 160(2): 40610, 1999.
de Jongste JC. Hydrogen peroxide in exhaled air of healthy children: 77. Brightling CE, Monteiro W, Ward R, Parker D, Morgan MD,
Reference values. Eur Respir J 12(2): 48385, 1998. Wardlaw AJ et al. Sputum eosinophilia and short-term response
58. Antczak A, Nowak D, Shariati B, Krol M, Piasecka G, Kurmanowska Z. to prednisolone in chronic obstructive pulmonary disease: A rando
Increased hydrogen peroxide and thiobarbituric acid-reactive products mised controlled trial. Lancet 356(9240): 148085, 2000.
in expired breath condensate of asthmatic patients. Eur Respir J 10(6): 78. Kips JC, OConnor BJ, Langley SJ, Woodcock A, Kerstjens HA,
123541, 1997. Postma DS et al. Eect of SCH55700, a humanized anti-human
59. Horvath I, Donnelly LE, Kiss A, Balint B, Kharitonov SA, Barnes PJ. interleukin-5 antibody, in severe persistent asthma: A pilot study. Am J
Exhaled nitric oxide and hydrogen peroxide concentrations in asth- Respir Crit Care Med 167(12): 165559, 2003.
matic smokers. Respiration 71(5): 46368, 2004. 79. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ,
60. Dohlman AW, Black HR, Royall JA. Expired breath hydrogen perox- Pavord ID. Mast-cell inltration of airway smooth muscle in asthma.
ide is a marker of acute airway inammation in pediatric patients with N Engl J Med 346(22): 1699705, 2002.
asthma. Am Rev Respir Dis 148(4 Pt 1): 95560, 1993. 80. Brightling CE, Ward R, Wardlaw AJ, Pavord ID. Airway inam-
61. Montuschi P, Corradi M, Ciabattoni G, Nightingale J, Kharitonov SA, mation, airway responsiveness and cough before and after inhaled
Barnes PJ. Increased 8-isoprostane, a marker of oxidative stress, in budesonide in patients with eosinophilic bronchitis. Eur Respir J 15(4):
exhaled condensate of asthma patients. Am J Respir Crit Care Med 68286, 2000.
160(1): 21620, 1999. 81. Pizzichini E, Pizzichini MM, Gibson P, Parameswaran K, Gleich GJ,
62. Paredi P, Ward S, Cramer D, Barnes PJ, Kharitonov SA. A new method Berman L et al. Sputum eosinophilia predicts benet from prednisone
for the non-invasive measurement of bronchial blood ow. Am J Respir in smokers with chronic obstructive bronchitis. Am J Respir Crit Care
Crit Care Med 167: A448, 2003. Med 158(5 Pt 1): 151117, 1998.
63. Paredi P, Kharitonov SA, Barnes PJ. Faster rise of exhaled breath tem- 82. Berry MA, Morgan A, Shaw DE, Parker D, Bradding P, Green RH
perature in asthma. A novel marker of airway inammation?. Am J et al. Pathological features and inhaled corticosteroid response of eosi-
Respir Crit Care Med 165(2): 18184, 2002 January 15. nophilic and non-eosinophilic asthma. Thorax, in press 2007.
64. Paredi P, Caramori G, Cramer D, Ward S, Ciaccia A, Papi A et al. 83. Jayaram L, Pizzichini MM, Cook RJ, Boulet LP, Lemiere C, Pizzichini E
Slower rise of exhaled breath temperature in chronic obstructive pul- et al. Determining asthma treatment by monitoring sputum cell counts:
monary disease. Eur Respir J 21(3): 43943, 2003. Eect on exacerbations. Eur Respir J 27(3): 48394, 2006.
65. Venge P, Bystrom J, Carlson M, Hakansson L, Karawacjzyk M, 84. Siva R, Green RH, Brightling CE, Shelley M, Hargadon B, McKenna S
Peterson C et al. Eosinophil cationic protein (ECP): Molecular and et al. Eosinophilic airway inammation and exacerbations of COPD:
biological properties and the use of ECP as a marker of eosinophil A randomised controlled trial. Eur Respir J 29(5): 90613, 2007.
activation in disease. Clin Exp Allergy 29(9): 117286, 1999. 85. Jatakanon A, Lim S, Barnes PJ. Changes in sputum eosinophils predict
66. Kips JC, Pauwels RA. Serum eosinophil cationic protein in asthma: loss of asthma control. Am J Respir Crit Care Med 161(1): 6472, 2000.
What does it mean?. Clin Exp Allergy 28(1): 13, 1998. 86. Leuppi JD, Salome CM, Jenkins CR, Anderson SD, Xuan W, Marks GB
67. Currie GP, Syme-Grant NJ, McFarlane LC, Carey FA, Lipworth BJ. et al. Predictive markers of asthma exacerbation during stepwise dose
Eects of low dose uticasone/salmeterol combination on surrogate reduction of inhaled corticosteroids. Am J Respir Crit Care Med 163(2):
inammatory markers in moderate persistent asthma. Allergy 58(7): 40612, 2001.
6027, 2003. 87. Shaw DE, Berry MA, Hargadon B, McKenna S, Shelley MJ, Green
68. Aldridge RE, Hancox RJ, Cowant JO, Frampton CM, Town GI, RH et al. Association between neutrophilic airway inammation and
Taylor DR. Eosinophils and eosinophilic cationic protein in induced airow limitation in adults with asthma. Chest 132(6): 187175, 2007.
sputum and blood: Eects of budesonide and terbutaline treatment. 88. Keatings VM, Collins PD, Scott DM, Barnes PJ. Dierences in inter-
Ann Allergy Asthma Immunol 89(5): 49297, 2002. leukin-8 and tumor necrosis factor-alpha in induced sputum from

556
Non-invasive Assessment of Airway Inflammation 42
patients with chronic obstructive pulmonary disease or asthma. Am J 102. Dupont LJ, Demedts MG, Verleden GM. Prospective evaluation of
Respir Crit Care Med 153(2): 53034, 1996. the validity of exhaled nitric oxide for the diagnosis of asthma. Chest
89. Smith AD, Cowan JO, Brassett KP, Filsell S, McLachlan C, Monti- 123(3): 75156, 2003.
Sheehan G et al. Exhaled nitric oxide: A predictor of steroid response. 103. Travers J, Marsh S, Aldington S, Williams M, Shirtclie P, Pritchard
Am J Respir Crit Care Med 172(4): 45359, 2005. A et al. Reference ranges for exhaled nitric oxide derived from a ran-
90. Deykin A, Lazarus SC, Fahy JV, Wechsler ME, Boushey HA, dom community survey of adults. Am J Respir Crit Care Med 176(3):
Chinchilli VM et al. Sputum eosinophil counts predict asthma control 23842, 2007.
after discontinuation of inhaled corticosteroids. J Allergy Clin Immunol 104. Brown HM. Treatment of chronic asthma with prednisolone; signi-
115(4): 72027, 2005. cance of eosinophils in the sputum. Lancet 2(7059): 124547, 1958.
91. Smith AD, Cowan JO, Brassett KP, Herbison GP, Taylor DR. Use 105. Berry M, Morgan A, Shaw DE, Parker D, Green R, Brightling C
of exhaled nitric oxide measurements to guide treatment in chronic et al. Pathological features and inhaled corticosteroid response of eosi-
asthma. N Engl J Med 352(21): 216373, 2005. nophilic and non-eosinophilic asthma. Thorax 62(12): 104349, 2007.
92. Pijnenburg MW, Bakker EM, Hop WC, de Jongste JC. Titrating ster- 106. Pizzichini MM, Pizzichini E, Parameswaran K, Clelland L,
oids on exhaled nitric oxide in children with asthma: A randomized Efthimiadis A, Dolovich J et al. Nonasthmatic chronic cough: No
controlled trial. Am J Respir Crit Care Med 172(7): 83136, 2005. eect of treatment with an inhaled corticosteroid in patients without
93. Shaw DE, Berry MA, Thomas M, Green RH, Brightling CE, sputum eosinophilia. Can Respir J 6(4): 32330, 1999.
Wardlaw AJ et al. The use of exhaled nitric oxide to guide asthma 107. Brightling CE, McKenna S, Hargadon B, Birring S, Green R, Siva R
management: A randomized controlled trial. Am J Respir Crit Care et al. Sputum eosinophilia and the short term response to inhaled
Med 176(3): 23137, 2007. mometasone in chronic obstructive pulmonary disease. Thorax 60(3):
94. Pijnenburg MW, Hofhuis W, Hop WC, de Jongste JC. Exhaled nitric 19398, 2005.
oxide predicts asthma relapse in children with clinical asthma remis- 108. Bateman ED, Boushey HA, Bousquet J, Busse WW, Clark TJ,
sion. Thorax 60(3): 21518, 2005. Pauwels RA et al. Can guideline-dened asthma control be achieved?
95. Zacharasiewicz A, Wilson N, Lex C, Erin EM, Li AM, Hansel T The Gaining Optimal Asthma Control study. Am J Respir Crit Care
et al. Clinical use of non-invasive measurements of airway inamma- Med 170(8): 83644, 2004.
tion in steriod reduction in children. Am J Respir Crit Care Med, 2005. 109. Juniper EF, Bousquet J, Abetz L, Bateman ED. Identifying well-
96. Taylor DR, Pijnenburg MW, Smith AD, de Jongste JC. Exhaled controlled and not well-controlled asthma using the Asthma Control
nitric oxide measurements: Clinical application and interpretation. Questionnaire. Respir Med 100(4): 61621, 2006.
Thorax 61(9): 81727, 2006. 110. Mcivor RA, Pizzichini E, Turner MO, Hussack P, Hargreave
97. Smith AD, Cowan JO, Filsell S, McLachlan C, Monti-Sheehan G, FE, Sears MR. Potential masking eects of salmeterol on airway
Jackson P et al. Diagnosing asthma: Comparisons between exhaled inammation in asthma. Am J Respir Crit Care Med 158(3): 92430,
nitric oxide measurements and conventional tests. Am J Respir Crit 1998.
Care Med 169(4): 47378, 2004. 111. Haldar P, Shaw D, Siva R, Berry M, Brightling C, Green R et al.
98. Hunter CJ, Brightling CE, Woltmann G, Wardlaw AJ, Pavord ID. A The correlation between exhaled nitric oxide and sputum eosinophil
comparison of the validity of dierent diagnostic tests in adults with counts in patients with airway disease. Thorax 62: A128, 2008.
asthma. Chest 121(4): 105157, 2002. 112. Zacharasiewicz A, Wilson N, Lex C, Erin EM, Li AM, Hansel T
99. Joyce DP, Chapman KR, Kesten S. Prior diagnosis and treatment of et al. Clinical use of noninvasive measurements of airway inam-
patients with normal results of methacholine challenge and unex- mation in steroid reduction in children. Am J Respir Crit Care Med
plained respiratory symptoms. Chest 109(3): 697701, 1996. 171(10): 107782, 2005.
100. Robinson DS, Campbell DA, Durham SR, Pfeer J, Barnes PJ, 113. Lehtimaki L, Kankaanranta H, Saarelainen S,Turjanmaa V, Moilanen E.
Chung KF. Systematic assessment of dicult-to-treat asthma. Eur Increased alveolar nitric oxide concentration in asthmatic patients
Respir J 22(3): 47883, 2003. with nocturnal symptoms. Eur Respir J 20(4): 84145, 2002.
101. Berkman N, Avital A, Breuer R, Bardach E, Springer C, Godfrey S. 114. Lehtimaki L, Kankaanranta H, Saarelainen S,Turjanmaa V, Moilanen E.
Exhaled nitric oxide in the diagnosis of asthma: Comparison with Inhaled uticasone decreases bronchial but not alveolar nitric oxide
bronchial provocation tests. Thorax 60(5): 38388, 2005. output in asthma. Eur Respir J 18(4): 63539, 2001.

557
Quantitative Imaging of the Lung
43 CHAPTER

INTRODUCTION that we look at organs today. As the CT scanner


developed and images became easier in terms of
Harvey O. Coxson
time and nancial considerations, it has become Department of Radiology, James Hogg
Imaging of the lung has always played a very one of the rst lines of clinical medicine. The iCAPTURE Centre for Cardiovascular
important role in pulmonary medicine. The CT technology is now so pervasive that nearly and Pulmonary Research, University of
advent of quantitative imaging allowed lung every hospital has at least one CT scanner and British Columbia, Vancouver, BC, Canada
researchers to move beyond descriptive studies the technology behind CT has changed so much
in the lung and actually apply measurements that now images of entire organs can be obtained
that could be applied to humans generally and in less time than it took to acquire one image in
disease conditions specically. Weibel was one the original CT scanner. Of course many other
of the rst researchers to meticulously measure techniques have also started to proliferate includ-
the lung and open up a whole new eld of path- ing magnetic resonance imaging, which provides
ologic research known as quantitative stereology not only anatomic information, but also some
[16]. These techniques have undergone some functional information as well. This chapter will
modication over the last 40 years, with the focus on these non-invasive quantitative measure-
proliferation of computers, but the basic prin- ments of lung structure specically as they apply
ciples remain unchanged today. Unfortunately, to lung structure and the changes involved in
pathologic techniques have some considerable chronic obstructive pulmonary disease (COPD).
drawbacks most notably that they require the This chapter will start with an examination of
tissue from either resected or autopsy speci- CT since it is the most popular both in terms of
mens. The second limitation is that they are his- market penetration of the CT scanner, and quan-
tologic examinations and because of their depth titative methodologies. The other major focus of
of scale require an extensive amount of work this chapter will be hyperpolarized magnetic res-
to cover a small region of the lung. However, onance (MR) imaging since it is a growing eld
pathology and histology are still considered and potentially has some valuable insights. This
the gold standard by which all other measuring chapter will also divide COPD into two separate
devices are judged and remains the cornerstone sections based on widely accepted phenotypic
of quantitative analysis. expressions of the disease, emphysema and small
Today the search for quantitative imaging airways disease because the exact pathogenesis
techniques that are not as invasive as pathology of COPD is still unknown and it varies widely
and allow the investigator to obtain multiple between individuals.
measurements over time is becoming very impor- It has been established that the root cause
tant. With the explosion worldwide of chronic of COPD is an exaggerated inammatory
lung diseases and, therefore, our need to accurately response that, in susceptible people, results in
measure the structure of the lung and possibly either a destruction of lung tissue (emphysema)
any changes due to the pathogenic mechanism or a remodeling of the airway wall structure.
or interventions is key. It is for these reasons that Both of these processes result in changes to the
medical imaging has become so popular and the function of the lung that termed COPD, but the
introduction of the computed tomography (CT) exact role and contribution of these two proc-
scanner by Hounseld has changed the way esses varies between individuals and is thought

559
Asthma and COPD: Basic Mechanisms and Clinical Management

to be under genetic control. Therefore, studies of COPD application and have led to numerous disagreements in the
must take into account the dierences in these pathogenic literature concerning their applicability.
pathways particularly as therapeutic interventions are devel- Since these initial studies CT scanners have undergone
oped because it is very likely that a specic intervention remarkable changes as described above. The initial descrip-
for the inammatory response in airway walls may be the tion of the density mask technique found that for CT scans
exact wrong intervention for the destructive components of that were 10 mm thick and reconstructed using a low spa-
emphysema. tial frequency reconstruction algorithm, emphysematous
holes greater than 5 mm in diameter correlated well with a
threshold value of 910 HU (Fig. 43.1). This means that the
percentage of lung voxels with attenuation values less than
910 HU was correlated with the percentage of the lung
QUANTITATIVE MEASUREMENTS occupied by holes greater than 5 mm in diameter [12, 13].
OF THE LUNG IN COPD Coxson and colleagues took this analysis a step further and
found that by estimating the average maximal lung ination
for a subject (based on a 6 l TLC and 1000 g lung mass) they
Emphysema analysis found that a cut-o value of 6.0 ml gas per gram of tissue
(856 HU) estimated the extent of emphysematous holes
In brief, CT images are obtained by passing a beam of X-rays between 2 and 5 mm in size [15].
through the body and measuring their absorbance which, in As CT scanner technology changed threshold values
biological tissues, is directly proportional to the density of have also had to change. Gevenois and co-workers re-examined
tissue. This apparent absorbance of X-rays is converted to the density mask technique using 1 mm thick CT slices
a relative scale known as the Hounseld Unit (HU) scale
which is a linear scale based on the attenuation water. In
the Hounseld scale the apparent attenuation of water is set
at 0 HU while the absorbance of air is 1000 HU. The CT
scan is a transaxial image so each of the pixels, or picture ele-
ments, in the image has an X and a Y dimension. However,
the X-ray beam and detector system of the scanner also has
at width which gives the CT scan a slice thickness making
the pixel of the CT image more appropriately known as a
voxel or volume element. In a classic conventional CT scan
the slice thickness was 10 mm which allowed adequate cover-
age of the lung using the shortest breath hold maneuver. The
X-ray detectors then became thinner so that images could be
acquired down to 1 mm in thickness, which combined with
a high spatial frequency reconstruction algorithm, became
known as the high-resolution CT scan (HRCT). CT scan-
ners have seen immense growth over the last 1015 years as
the introduction of slip ring technology allowed continuous
acquisition of images while the patient moved through the
gantry, the helical or spiral CT scan, to the sub-division of
the X-ray detector into multiple elements allowing for mul-
tiple slice acquisitions simultaneously, the multi-detector row
or multi-slice CT scanner.
The normal human lung is approximately 80 % airspace
and 20% tissue and blood [1] and CT densitometry studies
have shown that the normal human lung has a symmetrical
distribution of X-ray attenuation values around a mean of
approximately 800 HU [710]. This distribution of X-ray
attenuation values was the rst quantitative feature of the
CT scan that was exploited to quantify structural changes
in disease states because changes in the quantity of tissue
and gas in the lung causes changes in the shape of this dis-
tribution. Independently, investigators led by Hayhurst [11]
in Edinburgh and Mller [12] in Vancouver found that the
extent of emphysema quantied pathologically was directly
correlated to either the percentage of lung voxels with atten-
uation values less than a HU specic threshold value, density FIG. 43.1 (A) This figure shows a CT scan from a subject with relatively
mask [12, 13], or the actual HU value at the lowest 5th per- severe emphysema indicated by the arrows. (B) The voxels with attenuation
centile of the frequency distribution curve [14]. While both values less than 950 HU or below the threshold or density mask are
of these techniques are similar in idea they are dierent in highlighted on the CT scan.

560
Quantitative Imaging of the Lung 43
reconstructed using a high spatial frequency reconstruction algorithm could make up to a 15% (average 9.4%) dier-
algorithm and found that the best correlation between lung ence in the extent of emphysema measured using a den-
pathology and CT was obtained using the 950 HU thresh- sity mask technique [32]. Their data showed that some
old value [16, 17]. Furthermore, the lowest 5th percentile reconstruction algorithms, described as over-enhancing,
was also redened to be the lowest 15th percentile [18, 19]. improve spatial resolution by over-enhancing the dier-
Finally with the advent of multi-detector row CT scan- ence between lung voxels; thereby changing the frequency
ners that obtain multiple CT images at the same time and distribution of the apparent X-ray attenuation values within
acquire these images helically while the subject is moving the CT image and ultimately the number of voxels below a
through the CT gantry. Madani and colleagues found that certain cut-o value [32]. Other investigators have looked
the best correlations with pathology were obtained using at the eect of dose, and scanner manufacturer. Using a
the 960 HU threshold and all percentile points below phantom, Stoel found that there were signicant dierences
the lowest 18th percentile correlated with macroscopic and between single and multi-slice scanners [33]. In this study
microscopic measures of emphysema, although the strongest they reported that there were more errors in the volume
correlation with the lowest rst percentile [20]. measurements of single slice scanners than multi-slice and
Uses of the density mask and percentile method the higher degree of apparent random error in the density
have proliferated in the literature in recent years. The den- measurements led them to conclude that only multi-slice
sity mask technique has been used to measure the extent CT scanners should be used in clinical multi-center studies.
of emphysema in very severe subjects undergoing lung vol- Madanai et al. recently examined the eect of slice thick-
ume reduction surgery (LVRS) [15, 2123] or other thera- ness and radiation dose on measurements of emphysema. In
peutic interventions such as retinoic acid treatment [24]. this study the authors used one multi-slice CT scanner and
It has also been used to describe dierences in the extent acquired images using dierent radiation doses (mA) and
of disease in males and females and has reported that for then reconstructed the images using dierent slice thick-
a given level of obstruction males have more emphysema nesses [34]. They found that there were signicant correla-
than females [25]. It has also even been used to investigate tions between their metrics of emphysema, either threshold
emphysema like changes in lung structure in subjects with cut-os or percentile technique, however X-ray dose had an
Anorexia Nervosa and found that the subjects with the eect on the threshold cut-o measurements but not per-
lowest body mass index had the lowest diusing capacity centile method while slice thickness had a signicant eect
for carbon monoxide and also had X-ray attenuation values of both threshold and percentile techniques. In a similar
that were similar to heavy smokers and normally attributed study, Yuan and colleagues compared two multi-slice CT
to emphysema [26]. scanners as well as changes in radiation dose and showed
The percentile technique has also been applied to that there was no dierence between the techniques in
LVRS studies [27] and numerous studies of 1-antitrypsin terms of volume measurements if scanner or dose was
deciency [18, 19, 2730]. These investigators have shown changed [35]. However, there were dierences in mean lung
that the lowest 15th percentile point is a robust threshold density between scanners and, most importantly, large dif-
and allows subjects to be compared over time and is less ferences in the extent of emphysema measured using either
susceptible to minor changes that may be induced by the the threshold or the percentile technique when the X-ray
CT scanner or the size of the breath the subject takes dur- dose was changed [35]. Therefore, the conclusions of all of
ing the scan [18, 28, 30]. these studies are that if you intend to use CT as a measure-
Having a robust metric of lung change is an extremely ment of emphysema in a longitudinal and/or multi-center
important tool because with the proliferation of CT scan- study you must pay careful attention to the details. A multi-
ners and the continuous changing of the devices by man- slice scanner should be use for these studies and if you are
ufacturers it makes it very dicult come up with a metric to use a Toshiba scanner a correction factor must be applied
that changes with the disease process and not just the with to the X-ray attenuation values. Furthermore, the radiation
features of the scanner. Therefore, when conducting longi- dose must be carefully monitored because this factor alone
tudinal studies all aspects of the scan must be considered, can have large eects on the emphysema measurements. In
including patient parameters such as breath size and motion conclusion, the CT scanner can be used to measure emphy-
during the scan, but also calibration of the scanner, slice sema, but it must be treated like all other measuring tools
thickness, reconstruction algorithm, type of scanner and and all of the parameters must be carefully monitored.
radiation dose. In a longitudinal study of subjects with 1 An aspect of emphysema measurements that has been
deciency, Parr et al. noticed a drift in density mask meas- largely overlooked in the literature is some type of emphy-
urements which they managed to trace back to calibration sema hole size measurement. While it has been shown that
of the CT scanner [31]. They found that while the water there are decent correlations between the threshold cut-o
calibration of the scanner was performed routinely, there and the percentile techniques and pathologic measurements
was no attempt to calibrate the scanner to air and therefore of emphysema, these techniques do not provide any infor-
the change they were observing in lung density was due mation on the size or location of the emphysematous hole
entirely to the scanner and not the disease. For this rea- (Fig. 43.2). This is an area that needs more research because
son the authors had to develop a correction factor for their the number of publications using these types of techniques
CT scans to take into account this shift in X-ray attenu- is very small. One of the rst attempt to quantify hole size
ation values [31]. In another study, Boedeker et al. found used a technique known as the fractal or cluster analy-
that even when all other aspects, such as scanner type, size sis [36, 37] whereby the size of low attenuation areas, as
of breath, etc. were carefully controlled, the reconstruction dened by the number of connected voxels below a given

561
Asthma and COPD: Basic Mechanisms and Clinical Management

FIG. 43.2 This figure shows the voxels identified as below 950 HU in two different subjects, one with predominately smaller holes (A) and one with
predominately large holes (B). A cluster analysis can be used to separate the subjects with large holes from the one with smaller holes.

threshold, is correlated to the number of these low attenuat- indicated by the NETT study and the emphysematous hole
ing areas. Mishima validated this technique using an elastic size studies above [22, 37]. Nakano rst showed that emphy-
spring model and showed that as the emphysema become sema located in the upper-outer regions of the lung pre-
more extensive the holes become larger and they decrease in dicted better outcome following surgery, presumably because
number [36]. They also found that in subjects with COPD it is more accessible to surgical resection than those with
but similar percentage emphysema as control subjects the more centrally located or diuse disease [39]. The studies
fractal measurement was lower in the COPD subjects sug- of location of emphysema and the eect that it has on lung
gesting that while they had the same extent of emphysema function are sparse in the literature, but it is hoped that with
the emphysematous lesions tended to be larger in subjects the widespread use of multi-slice CT scanners that these
with COPD. This study was follow-up by another study types of studies will shed more light on the pathogenesis of
that examined the size and location of the lesion in sub- emphysema (Fig. 43.3).
jects undergoing lung volume reduction surgery. This study Finally, there is great interest in using CT scans in
showed that that subjects with large upper lung region multi-center studies as an end point for a therapeutic inter-
lesions have the best outcome following LVRS [37]. The vention. This has already been accomplished in studies such as
National Emphysema Treatment Trial (NETT) applied a the NETT trial but the work of Dirksen really adds impor-
similar technique to their study subjects and found that there tance to this approach. In a small study of 1-antitrypsin
was a good correlation between the radiologist assessed hole decient subjects, Dirksen found that there was no signicant
size and the computer assessed metric [22]. Furthermore, the change in the functional or structural (assessed using CT)
size and location of these lesions, large lesions located in the characteristics over the course of the study [28]. However,
apical regions of the lung, seemed to have a predictive eect when the authors performed a power calculation of their data
on survival following surgery [22]. More complex techniques they found that to show a dierence in FEV1 the authors
that quantify multiple features within the CT scan, such as would have had to study 550 subjects but to show a dierence
the adaptive multiple feature method, have reported decent using CT the authors would only have had to study 130 [28].
results in disease stratication including the ability to sepa- These data add further credence to the important role that
rate smokers from non-smokers in the absence of other dis- CT scans can play in clinical intervention studies.
ease [38]. However, the disadvantage of these new texture In conclusion, these studies all show that CT measure-
analysis techniques is that they often use complex math- ments of emphysema are correlated with pathologic measure-
ematical terminology to describe the lung rather than ana- ments of emphysema. Furthermore, these measurements can
tomic or physiologic terminology that are more commonly be used to follow disease progression, possibly with more sen-
used in medicine and have not been validated using patho- sitivity than the traditional metrics of FEV1, and that size and
logic assessment of the underlying lung structure. Finally, the location of the disease is important. Also, these techniques
location of the emphysema seems to be important as well as can be used to measure early or small changes in the lungs

562
Quantitative Imaging of the Lung 43
(A) airow limitation because it has long been recognized that
the site of major airow limitation is the small airways [40].
There have been numerous investigations of small airway
dimensions and COPD that have conrmed the impor-
tance of the remodeling in these structures, but once again,
these rely on histologic specimens [4045]. The introduction
of CT scanning peaked investigators interest in measur-
ing airways non-invasively so several methods were imple-
mented to manually trace the airway walls using the CT
images printed on lm, similar to how it was accomplished
using histology. However, one thing that became immedi-
ately apparent was that optimal measurement of airway
(B)
lumen using this technique required very specic display
settings. For example, it was found that an accurate meas-
urement of wall area required the use of a window level of
450 HU and a window width of 10001400 HU [4649].
Unfortunately, these display settings produce images that are
too dark for optimal visualization of the lung parenchyma
and so are never used clinically. Furthermore, because the
technique is cumbersome, time consuming, and associated
with considerable intra- and interobserver variability, this
approach was quickly abandoned [4749].
Since the CT scan is a digital image and contains
(C) gray scale information attention quickly turned to using
automated airway analysis techniques. McNitt-Gray et
al. found that the lumen area could be accurately meas-
ured using a threshold cut o of 500 HU [50] which was
further validated using excised formalin xed pig lungs
by King and co-workers who reported that a threshold of
577 HU produced the least error in the measurements
[51]. However, these techniques just measured the airway
lumen so attention turned to using the X-ray attenuation
values of the CT scan to measure both the airway lumen
and wall. One of the more common techniques is the Full-
Width-At-Half-Maximum (half-max) method (Fig.
FIG. 43.3 This figure shows a three dimension analysis of emphysema
43.4) where the distribution of apparent X-ray attenua-
in a subject with predominately upper lobe emphysema. The trans-axial
images (A) show more highlighted regions (arrows) in the upper lobes
tion values along a ray projected from a central point of the
while the coronal images (B) show quite clearly that there are more lumen to the parenchyma is measured. The distance between
highlighted voxels in the upper lobes (arrows) than the lower lobes (arrow the point at which the attenuation is half way between the
heads). A cluster analysis (C) that highlights the connected regions clearly local minimum in the lumen or parenchyma and the maxi-
shows larger clusters in the upper lobes. Images were created using the mum within the wall is considered to be wall thickness [52,
Pulmonary Workstation 2.0 software (VIDA Diagnostics Inc.). 53]. Unfortunately, the shape of this curve is dependant on
various parameters including: the reconstruction algorithm
used to create the image, partial volume averaging due to
of early disease subjects and can be used to compare dier- eld of view and orientation of the airway within the CT
ences in disease between genders. Overall, the assessment of image and the inevitable blurring of edges that occurs due
emphysema using CT has a long and well developed history to the point spread function of the CT scanner. Validation
and is now starting to be applied in more and more longitu- studies show that the CT scans over-estimate airway wall
dinal studies. However, it is important to keep in mind that area and underestimate lumen area and that these errors
CT, like any measuring device, must be used properly. The become very large in small airways [52, 54]. Furthermore,
CT scanners must be comparable and the CT image acquisi- this algorithm requires that the airway being analyzed on
tion techniques must be comparable. It has been shown that CT is cut in cross section. While it is relatively easy for
low dose studies can be performed in these groups of subjects, observers to nd airways cut in cross section, because the
but because dose eects the measurements of emphysema the CT slice has a thickness, typically on the order of 1 mm, the
dose must be kept constant throughout the study. airway observed on CT may or may not have actually been
cut in cross section (Fig. 43.5). This is another source of
error within the airway wall measurements and makes them
Airway analysis especially unreliable as the airways approach the resolu-
tion of the CT scanner. For these reasons investigators have
Of course assessing the emphysema is only one part of the developed numerous other algorithms to measure these air-
analysis of the structural changes that cause the chronic ways such as the maximum-likelihood method whereby

563
Asthma and COPD: Basic Mechanisms and Clinical Management

(A) (B)

FIG. 43.4 This figure shows a CT scan (A) from which an airway is chosen and displayed using a magnified view (B). The measurement of the airway wall
dimensions is performed by using the Full-Width-At-Half-Maximum method whereby the length of the rays is determined by choosing the halfway point
between the minimum X-ray attenuation value in the lumen or lung parenchyma and the maximum value within the airway wall. The lumen area and the
internal perimeter of the airway wall are measured using the internal boundary of the rays (shown by arrowhead) and the wall area and the outer perimeter
are measured by the external boundary of the rays (arrow).

(A) (B) (C)

FIG. 43.5 This figure shows the same airway from gross pathology (A), histology (B) and from the pre-operative CT scan (C). From this image some of the
measuring artifacts can be seen including the fact that the airway may not actually be cut in cross section in the CT scan because the airway is slightly
different shape in the CT and can be seen to not be in perfect cross section in the gross pathology section. Secondly, the resolution issues can clearly be
seen as the actual pixels are visible in the CT scan and the image appears very pixilated. All of the scale bars are 5 mm in length.

the attenuation threshold along each ray is matched to an the emphysema and airway wall dimension data with pul-
ideal calculated ray [54], the score-guided erosion algo- monary function data. The data from this study showed that
rithm [51] where airway wall edges are found using an edge the thickening in this large airway correlated with FEV1 per-
nding algorithm that assumes that airways are circular and cent predicted, FVC percent predicted and RV/TLC while
have a relatively high density compared to the surrounding the emphysema correlated with FEV1, FEV/FVC ratio and
parenchyma, and an algorithm where ellipses are t to the the DLCO. Furthermore, a multiple regression analysis sug-
airway lumen and wall [55]. gested that, for a given FEV1, subjects with more extensive
Because it is well recognized that airway wall remod- emphysema had less airway wall thickening than those with
eling plays a very important role in COPD there is great less extensive emphysema [53]. Obviously a segmental air-
interest in non-invasive measurement of airway wall dimen- way is unlikely to be the major source of airow limitation
sions. However, the applications of these techniques have so in subjects with COPD. Furthermore, it is questionable
far been very limited. One of the rst papers published in this whether any of the airways that can be observed using CT
eld was a study by Nakano and co-workers [53] who evalu- are the airways responsible for the airow limitation. Since
ated the right apical segmental bronchus of asymptomatic histology is considered the gold standard for small airway
smokers and COPD subjects. The authors chose this air- wall remodeling, Nakano compared the measurements of
way to measure because it is routinely cut in cross section on wall area obtained using CT (average diameter of 3.2 mm)
transaxial CT scans, and they could compare the same air- to those obtained using histology of small airways (1.27 mm
way across all subjects. The authors also measured the extent diameter) in resected specimens [56]. These data show a
of emphysema using a threshold technique and correlated signicant association (R2 0.57, p 0.001) between the

564
Quantitative Imaging of the Lung 43
(A) (B)

FIG. 43.6 This figure shows multi-planar reformatted reconstructions of the airway tree (A). Using these new software tools it is now possible to segment
the airway tree from the lung parenchyma and, using a pre-defined pathway, reconstruct it as a long straight tube and obtain measurements at a cross
section to the center line of the airway at any point along the pathway (B). Images were created using the Pulmonary Workstation 2.0 software
(VIDA Diagnostics Inc.).

dimensions of the smaller and larger airways. The authors larger fraction of the total airway. Therefore, you can poten-
conclude that, at least for COPD, measuring airway dimen- tially measure a statistical dierence in WA% between indi-
sions in the larger bronchi, which are more accurately viduals, because there is an inammatory process occurring
assessed by CT, can provide an estimate of small airway in the airway wall that is making the wall area bigger, or you
remodeling. It is likely that the same pathophysiologic proc- could simply be comparing big airways to small airways due
ess, which causes small airway obstruction, also takes place in to improper sampling. The new three-dimensional recon-
larger airways where it has less functional eect. struction technique can avoid that trap by allowing investi-
Recently focus has turned away from measurements gators to compare the exact airway between individuals and
of airways cut in cross section on transaxial CT images over time. That was one of the original goals of the Nakano
to what is thought to be a true cross section of the airway paper where they measured the right apical segmental bron-
obtained by multi-planar reformats of three-dimensional chus only, but it is now better achieved and in smaller air-
airway data. Multi-slice scanners can now obtain CT images ways using current CT and image analysis technology.
with resolution in the Z dimension that approaches that This approach was recently put to the test by Hasegawa
of the XY dimensions, although most studies still use CT and co-workers [57] who used multi-slice CT scans to meas-
slices with 11.25 mm slice thickness. Investigators can ure the right apical and basal segmental bronchus at dierent
now segment the airway tree in three dimensions starting branch points along its pathway. These authors were not able
in the trachea and projecting out to the fth or sixth gen- to verify the ndings of Nakano by comparing the right api-
eration (Fig. 43.6). Also, this technique allows the investi- cal segmental bronchus to FEV1 however, they did show that
gator to section the airway at any place and any angle that as they moved more distally, to the sixth generation of the
they choose. Of course this technique assumes that the voxel airway tree, the correlation with FEV1 improved. The authors
dimension in the Z dimension is small, that is 1 mm or less, concluded that the airow limitations are more strongly
or the assumptions inherent to the re-format become too associated with the dimensions of the distal airways than
large. Another important advance to this technique is that the proximal ones and demonstrate that these new three-
now investigators can know exactly where they are in the dimensional techniques can be used to obtain measurements
airway tree and if they wish to compare the airway size of a from a specic location within a specic airway [57]. Also, it
specic airway between individuals and over time they have once again shows the importance of airway measurements in
the landmarks to do so and no longer have to count on ran- understanding COPD.
dom selections of airways. While random airway selections
have their advantages in that you have an overall picture of
airways within a subject they also have their drawbacks. For Hyperpolarized noble gas imaging
example, investigators often normalize airway wall dimen- of the lung
sions between individuals by comparing the percent of the
total airway (lumen  wall) that is occupied by wall, the so Other techniques such as MR imaging of the lung are
called Wall Area Percent (WA%). This is a very good esti- becoming very attractive techniques particularly because
mate of wall area between subjects but it assumes that you they do not expose the subject to any ionizing radiation.
are comparing equal sized airways because in large airways This has always been a strong point of MR, but factors such
the WA% is always small because the wall area is such as the small nuclear spin polarization at thermal equilibrium
a small fraction of the total airway. On the other hand, in and the eld inhomogeneity problems associated with the
small airways, the WA% is always large because now the lung have greatly limited the application of this technique
lumen is very small and the wall starts to occupy a much [58]. However, the last few years have seen resurgence in

565
Asthma and COPD: Basic Mechanisms and Clinical Management

MR imaging research because of the introduction of hyper- and functional information. However, because of techni-
polarized noble gases. Research has shown that by exposing cal and nancial reasons CT is currently the only readily
noble gases such as 129Xenon or 3Helium to a laser you can accessible, relatively non-invasive technique that provides
hyperpolarize the gas by aligning more of their magnetic quantitative structural data in vivo. The proper application
moments into the same orientation. This allows the gas to of these techniques should produce valuable information on
be used as a contrast agent within the lungs during an MR the pathogenesis of COPD and the eects of new thera-
scan and has allowed investigators to measure the ventila- peutic interventions.
tion of the lung [59, 60], the size of the alveolus airspace
[61] and airway dimensions [62, 63]. The most well estab-
lished of these techniques is the measurement of alveolar
size by a technique known as the Apparent Diusion Co- References
ecient or ADC. This technique works on the principle
that gas molecules will move, or diuse, within a given vol- 1. Weibel ER. Morphometry of the Human Lung. Berlin: Springer-Verlag,
1963.
ume such as the alveolus of the lung. In the normal lung the
2. Weibel ER. Stereological Methods, Volume 1: Practical Methods for
airspaces are much smaller so that the movement of Helium Biological Morphometry, London: Academic Press, 1980.
is restricted but in emphysema where the airspaces become 3. Weibel ER. Stereological Methods, Volume 2: Theoretical Foundations,
much larger. Several studies have correlated the ADC with London: Academic Press, 1980.
spirometry measurements in animal models, normal volun- 4. Weibel ER. Functional morphology of lung parenchyma. In:
teers and in subjects with airow limitations emphysema Macklem PT, Mead J, (eds). Handbook of Physiology, Section 3:
[6062, 6469]. In a recent study Woods et al. showed The Respiratory System Volume III, Part I, pp. 89111. Bethesda:
that there was a very good correlation between the ADC American Physiological Society, 1986.
and alveolar size measured using either the surface area to 5. Weibel ER. Measuring through the microscope: Development and
volume ratio of the lung parenchyma or the mean linear evolution of stereological methods. J Microsc 155: 393403, 1989.
6. Weibel ER. The structural basis of lung function. In: West JB (ed.).
intercept and that the ADC was able to separate emphyse-
Respiratory Physiology People and Ideas, pp. 346. New York: Oxford
matous lung from normal lung with greater sensitivity than University Press, 1996.
histology [61]. 7. Wegener OH, Koeppe P, Oeser H. Measurement of lung density by
Other investigators have shown that it is possible computed tomography. J Comput Assist Tomogr 2: 26373, 1978.
to create three-dimensional reconstruction of the airway 8. Robinson PJ, Kreel L. Pulmonary tissue attenuation with computed
lumen to the seventh-generation of airways [62, 63]. While tomography: Comparison if Inspiration and expiration scans. J Comput
this is an interesting technique it does not provide meas- Assist Tomogr 3: 74048, 1979.
urements of airway wall thickness and so the applicability is 9. Wollmer P, Albrechtsson U, Brauer K, Eriksson L, Jonson B, Tyln U.
still uncertain. Measurement of pulmonary density by means of X-ray computerized
Finally, while it appears that hyperpolarized gas MR tomography. Chest 90: 38791, 1986.
10. Coxson HO, Mayo JR, Behzad H, Moore BJ, Verburgt LM, Staples CA,
has some very powerful applications in the study of COPD,
Pare PD, Hogg JC. Measurement of lung expansion with computed
the technique its self is very limited by availability of a tomography and comparison with quantitative histology. J Appl Physiol
hyperpolarized helium or xenon source and the extremely 79: 152530, 1995.
technical nature and cost of the procedure. While it is an 11. Hayhurst MD, Flenley DC, McLean A, Wightman AJA, MacNee W,
exciting and innovative technique, it is very likely that Wright D, Lamb D, Best J. Diagnosis of pulmonary emphysema by
hyperpolarized gas MR will remain a research tool for the computerized tomography. Lancet 2: 32022, 1984.
immediate future. 12. Mller NL, Staples CA, Miller RR, Abboud RT. Density mask. An
objective method to quantitate emphysema using computed tomogra-
phy. Chest 94: 78287, 1988.
13. Miller RR, Mller NL, Vedal S, Morrison NJ, Staples CA. Limitations
of computed tomography in the assessment of emphysema. Am Rev
CONCLUSION Respir Dis 139: 98083, 1989.
14. Gould GA, MacNee W, McLean A, Warren PM, Redpath A, Best JJ,
Lamb D. CT measurements of lung density in life can quantitate distal
In conclusion, the last 20 years have seen a large amount airspace enlargement an essential dening feature of human emphy-
of growth in both the interest and the application of non- sema. Am Rev Respir Dis 137: 38092, 1988.
invasive quantitative imaging of the lung. As long as the 15. Coxson HO, Rogers RM, Whittall KP, DYachkova Y, Pare PD,
scanners are treated properly it is now possible to obtain Sciurba FC, Hogg JC. A quantication of the lung surface area in
reliable quantitative information on the extent of emphy- emphysema using computed tomography. Am J Respir Crit Care Med
sema, the size of the emphysematous lesions and airway 159: 85156, 1999.
wall dimensions. The advent of multi-slice CT scanners has 16. Gevenois PA, de Maertelaer V, De Vuyst P, Zanen J, Yernault JC.
opened up new avenues of research into regional distribu- Comparison of computed density and macroscopic morphometry in
pulmonary emphysema. Am J Respir Crit Care Med 152: 65357, 1995.
tion of disease and specic tagging and measurements of
17. Gevenois PA, De Vuyst P, de Maertelaer V, Zanen J, Jacobovitz D,
airways. On the downside CT imaging has some inherent Cosio MG, Yernault JC. Comparison of computed density and micro-
limitations: particularly concerning radiation exposure to scopic morphometry in pulmonary emphysema. Am J Respir Crit Care
the subject and the standardization and quality control of Med 154: 18792, 1996.
the CT scan parameters. Hyperpolarized gas MR imaging 18. Dirksen A, Friis M, Olesen KP, Skovgaard LT, Sorensen K. Progress of
has come a long ways in the last several years and has made emphysema in severe 1-antitrypsin deciency as assessed by annual
some important steps to provide both quantitative structure CT. Acta Radiol 38: 82632, 1997.

566
Quantitative Imaging of the Lung 43
19. Stolk J, Ng WH, Bakker ME, Reiber JH, Rabe KF, Putter H, Stoel BC. 37. Coxson HO, Whittall KP, Nakano Y, Rogers RM, Sciurba FC, Keenan RJ,
Correlation between annual change in health status and computer Hogg JC. Selection of patients for lung volume reduction surgery using
tomography derived lung density in subjects with alpha1-antitrypsin a power law analysis of the computed tomographic scan. Thorax 58:
deciency. Thorax 58: 102730, 2003. 51014, 2003.
20. Madani A, Zanen J, de Maertelaer V, Gevenois PA. Pulmonary emphy- 38. Uppaluri R, Mitsa T, Sonka M, Homan EA, McLennan G.
sema: Objective quantication at multi-detector row CT Comparison Quantication of pulmonary emphysema from lung computed tomog-
with macroscopic and microscopic morphometry. Radiology 238: raphy images. Am J Respir Crit Care Med 156: 24854, 1997.
103643, 2006. 39. Nakano Y, Coxson HO, Bosan S, Rogers RM, Sciurba FC, Keenan RJ,
21. Bae KT, Slone RM, Gierada DS, Yusen RD, Cooper JD. Patients with Walley KR, Pare PD, Hogg JC. Core to rind distribution of severe
emphysema: Quantitative CT analysis before and after lung volume emphysema predicts outcome of lung volume reduction surgery. Am J
reduction surgery. Work in progress. Radiology 203: 70514, 1997. Respir Crit Care Med 164: 219599, 2001.
22. Martinez FJ, Foster G, Curtis JL, Criner G, Weinmann G, Fishman A, 40. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of airway
DeCamp MM, Benditt J, Sciurba F, Make B et al. Predictors of mor- obstruction in chronic obstructive lung disease. N Engl J Med 278:
tality in patients with emphysema and severe airow obstruction. Am J 135560, 1968.
Respir Crit Care Med 173: 132634, 2006. 41. Bosken CH, Hards J, Gatter K, Hogg JC. Characterization of the
23. Rogers RM, Coxson HO, Sciurba FC, Keenan RJ, Whittall KP, Hogg JC. inammatory reaction in the peripheral airways of cigarette smokers
Preoperative severity of emphysema predictive of improvement after using immunocytochemistry. Am Rev Respir Dis 145: 91117, 1992.
lung volume reduction surgery: Use of CT morphometry. Chest 118: 42. Bosken CH, Wiggs BR, Pare PD, Hogg JC. Small airway dimensions in
124047, 2000. smokers with obstruction to airow. Am Rev Respir Dis 142: 56370, 1990.
24. Mao JT, Goldin JG, Dermand J, Ibrahim G, Brown MS, Emerick A, 43. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
McNitt-Gray MF, Gjertson DW, Estrada F, Tashkin DP et al. A pilot Cherniack RM, Rogers RM, Sciurba FC, Coxson HO et al. The nature
study of all-trans-retinoic acid for the treatment of human emphysema. of small-airway obstruction in chronic obstructive pulmonary disease.
Am J Respir Crit Care Med 165: 71823, 2002. N Engl J Med 350: 264553, 2004.
25. Dranseld MT, Washko GR, Foreman MG, Estepar RS, Reilly J, 44. Hogg JC, Chu FS, Tan WC, Sin DD, Patel SA, Pare PD, Martinez FJ,
Bailey WC. Gender dierences in the severity of CT emphysema in Rogers RM, Make BJ, Criner GJ et al. Survival after lung volume
COPD. Chest 132: 46470, 2007. reduction in chronic obstructive pulmonary disease: Insights from
26. Coxson HO, Chan IH, Mayo JR, Hlynsky J, Nakano Y, Birmingham CL. small airway pathology. Am J Respir Crit Care Med 176: 45459, 2007.
Early emphysema in patients with anorexia nervosa. Am J Respir Crit 45. Kim WD, Ling SH, Coxson HO, English JC, Yee J, Levy RD, Pare PD,
Care Med 170: 74852, 2004. Hogg JC. The association between small airway obstruction and
27. Stolk J, Versteegh MIM, Montenij LJ, Bakker ME, Grebski E, emphysema phenotypes in COPD. Chest 131: 137278, 2007.
Tutic M, Wildermuth S, Weder W, el Bardiji M, Reiber JHC et al. 46. Bankier AA, Fleischmann D, Mallek R, Windisch A, Winkelbauer FW,
Densitometry for assessment of eect of lung volume reduction sur- Kontrus M, Havelec L, Herold CJ, Hubsch P. Bronchial wall thick-
gery for emphysema. Eur Respir J 29: 107577, 2007. ness: Appropriate window settings for thin-section CT and radiologic-
28. Dirksen A, Dijkman JH, Madsen IF, Stoel B, Hutchison DCS, anatomic correlation. Radiology 199: 83136, 1996.
Skovgaard LT, Kok-Jensen A, Rudolphus A, Seersholm N et al. A ran- 47. McNamara AE, Muller NL, Okazawa M, Arntorp J, Wiggs BR, Pare PD.
domized clinical trial of a-1 antitrypsin augmentation therapy. Am J Airway narrowing in excised canine lung measured by high-resolution
Respir Crit Care Med 160: 146872, 1999. computed tomography. J Appl Physiol 73: 30716, 1992.
29. Parr DG, Stoel BC, Stolk J, Stockley RA. Pattern of emphysema dis- 48. Okazawa M, Muller NL, McNamara AE, Child S, Verburgt L, Pare PD.
tribution in alpha1-antitrypsin deciency inuences lung function Human airway narrowing measured using high resolution computed
impairment. Am J Respir Crit Care Med 170: 117278, 2004. tomography. Am J Respir Crit Care Med 154: 155762, 1996.
30. Stolk J, Dirksen A, van der Lugt AA, Hutsebaut J, Mathieu J, de Ree J, 49. Webb WR, Gamsu G, Wall SD, Cann CE, Proctor E. CT of a bron-
Reiber JH, Stoel BC. Repeatability of lung density measurements with chial phantom: Factors aecting appearance and size measurements.
low-dose computed tomography in subjects with alpha-1-antitrypsin Invest Radiol 19: 39498, 1984.
deciency-associated emphysema. Invest Radiol 36: 64851, 2001. 50. McNitt-Gray MF, Goldin JG, Johnson TD, Tashkin DP, Aberle DR.
31. Parr DG, Stoel BC, Stolk J, Nightingale PG, Stockley RA. Inuence of Development and testing of image-processing methods for the quan-
calibration on densitometric studies of emphysema progression using titative assessment of airway hyperresponsiveness from high-resolution
computed tomography. Am J Respir Crit Care Med 170: 88390, 2004. CT images. J Comput Assist Tomogr 21: 93947, 1997.
32. Boedeker KL, McNitt-Gray MF, Rogers SR, Truong DA, Brown MS, 51. King GG, Muller NL, Whittall KP, Xiang QS, Pare PD. An analysis algo-
Gjertson DW, Goldin JG. Emphysema: Eect of reconstruction algo- rithm for measuring airway lumen and wall areas from high-resolution
rithm on CT imaging measures. Radiology 232: 295301, 2004. computed tomographic data. Am J Respir Crit Care Med 161: 57480,
33. Stoel BC, Bakker ME, Stolk J, Dirksen A, Stockley RA, Piitulainen E, 2000.
Russi EW, Reiber JH. Comparison of the sensitivities of 5 dierent 52. Nakano Y, Whittall KP, Kalloger SE, Coxson HO, Pare PD.
computed tomography scanners for the assessment of the progression Development and validation of human airway analysis algorithm using
of pulmonary emphysema: A phantom study. Invest Radiol 39: 17, multidetector row CT. Proc SPIE 4683: 46069, 2002.
2004. 53. Nakano Y, Muro S, Sakai H, Hirai T, Chin K, Tsukino M, Nishimura K,
34. Madani A, De Maertelaer V, Zanen J, Gevenois PA. Pulmonary Itoh H, Pare PD, Hogg JC et al. Computed tomographic measure-
emphysema: Radiation dose and section thickness at multidetector CT ments of airway dimensions and emphysema in smokers. Correlation
quantication Comparison with macroscopic and microscopic mor- with lung function. Am J Respir Crit Care Med 162: 11028, 2000.
phometry. Radiology 243: 25057, 2007. 54. Reinhardt JM, DSouza ND, Homan EA. Accurate measurement of
35. Yuan R, Mayo JR, Hogg JC, Pare PD, McWilliams AM, Lam S, intrathoracic airways. IEEE Trans Med Imaging 16: 82027, 1997.
Coxson HO. The eects of radiation dose and CT manufacturer on 55. Saba OI, Homan EA, Reinhardt JM. Maximizing quantitative accu-
measurements of lung densitometry. Chest 132: 61723, 2007. racy of lung airway lumen and wall measures obtained from X-ray CT
36. Mishima M, Hirai T, Itoh H, Nakano Y, Sakai H, Muro S, Nishimura K, imaging. J Appl Physiol 95: 106375, 2003.
Oku Y, Chin K, Ohi M et al. Complexity of terminal airspace geometry 56. Nakano Y, Wong JC, de Jong PA, Buzatu L, Nagao T, Coxson HO,
assessed by lung computed tomography in normal subjects and patients Elliott WM, Hogg JC, Par PD. The prediction of small airway dimen-
with chronic obstructive pulmonary disease. Proc Natl Acad Sci USA 96: sions using computed tomography. Am J Respir Crit Care Med 171:
882934, 1999. 14246, 2005.

567
Asthma and COPD: Basic Mechanisms and Clinical Management

57. Hasegawa M, Nasuhara Y, Onodera Y, Makita H, Nagai K, Fuke S, Ito Y, 64. Fain SB, Panth SR, Evans MD, Wentland AL, Holmes JH, Korosec FR,
Betsuyaku T, Nishimura M. Airow limitation and airway dimensions OBrien MJ, Fountaine H, Grist TM. Early emphysematous changes
in chronic obstructive pulmonary disease. Am J Respir Crit Care Med in asymptomatic smokers: Detection with 3He MR imaging. Radiology
173: 130915, 2006. 239: 87583, 2006.
58. Mayo JR. Magnetic resonance imaging of the chest. Where we stand. 65. Fichele S, Woodhouse N, Swift AJ, Said Z, Paley MN, Kasuboski L,
Radiol Clin North Am 32: 795809, 1994. Mills GH, van Beek EJ, Wild JM. MRI of helium-3 gas in healthy
59. Salerno M, Altes TA, Brookeman JR, de Lange EE, Mugler JP. lungs: Posture related variations of alveolar size. J Magn Reson Imaging
Dynamic spiral MRI of pulmonary gas ow using hyperpolarized 20: 33135, 2004.
(3)He: Preliminary studies in healthy and diseased lungs. Magn Reson 66. Ley S, Zaporozhan J, Morbach A, Eberle B, Gast KK, Heussel CP,
Med 46: 66777, 2001. Biedermann A, Mayer E, Schmiedeskamp J, Stepniak A et al. Functional
60. Swift AJ, Woodhouse N, Fichele S, Siedel J, Mills GH, van Beek EJ, evaluation of emphysema using diusion-weighted 3Helium-magnetic
Wild JM. Rapid lung volumetry using ultrafast dynamic magnetic resonance imaging, high-resolution computed tomography, and lung
resonance imaging during forced vital capacity maneuver: Correlation function tests. Invest Radiol 39: 42734, 2004.
with spirometry. Invest Radiol 42: 3741, 2007. 67. Salerno M, de Lange EE, Altes TA, Truwit JD, Brookeman JR,
61. Woods JC, Choong CK, Yablonskiy DA, Bentley J, Wong J, Pierce JA, Mugler JP III. Emphysema: Hyperpolarized helium 3 diusion MR
Cooper JD, Macklem PT, Conradi MS, Hogg JC. Hyperpolarized 3He imaging of the lungs compared with spirometric indexes initial expe-
diusion MRI and histology in pulmonary emphysema. Magn Reson rience. Radiology 222: 25260, 2002.
Med 56: 1293300, 2006. 68. Swift AJ, Wild JM, Fichele S, Woodhouse N, Fleming S, Waterhouse J,
62. Tooker AC, Hong KS, McKinstry EL, Costello P, Jolesz FA, Albert MS. Lawson RA, Paley MN, Van Beek EJ. Emphysematous changes and
Distal airways in humans: Dynamic hyperpolarized 3He MR imaging normal variation in smokers and COPD patients using diusion 3He
feasibility. Radiology 227: 57579, 2003. MRI. Eur J Radiol 54: 35258, 2005.
63. Lewis TA, Tzeng YS, McKinstry EL, Tooker AC, Hong K, Sun 69. Mata JF, Altes TA, Cai J, Ruppert K, Mitzner W, Hagspiel KD, Patel
Y, Mansour J, Handler Z, Albert MS. Quantication of airway B, Salerno M, Brookeman JR, de Lange EE et al. Evaluation of emphy-
diameters and 3D airway tree rendering from dynamic hyperpo- sema severity and progression in a rabbit model: Comparison of hyper-
larized 3He magnetic resonance imaging. Magn Reson Med 53: 47478, polarized 3He and 129Xe diusion MRI with lung morphometry.
2005. J Appl Physiol 102: 127380, 2007.

568
Systemic Manifestations
44 CHAPTER

INTRODUCTION are quoted in this update even before stating that


its pulmonary component is characterized by air-
Alvar Agusti
ow limitation that is not fully reversible [2]. This Hospital University Son Dureta,
Single organ diseases can have systemic mani- highlights the clinical relevance of these extra- Servi Pneumologia, Palma de Majorca,
festations. Acute diseases, such as urinary tract pulmonary eects because: (1) they contribute Spain
infections or pneumonia can cause septic shock, to disease progression independently of airow Fundacion Caubet-CIMERA
multiple organ failure, and eventually, death. obstruction. In fact, it is likely that important
CIBER Enfermedades Respiratorias
Likewise, chronic diseases like cancer, heart fail- clinical outcomes, such as mortality or health
ure, or chronic obstructive pulmonary disease status, among others, eventually result from
(COPD), to name only a few, are associated with the interplay between the intra-pulmonary and
systemic manifestations as well. In a much more extra-pulmonary eects that occur in COPD
relentless way than in acute conditions, systemic (Fig. 44.1) [3]; and (2) they constitute novel ther-
manifestations in chronic diseases inuence the apeutic targets [4]. Table 44.1 presents the sys-
course of the disease, and eventually, contribute temic manifestations of COPD identied so far.
to death. This chapter reviews the systemic man-
ifestations associated to two chronic respiratory
diseases, COPD, and asthma, and discusses their Nutritional abnormalities
potential implications for the management and
treatment of these patients. Unexplained weight loss occurs in about 50% of
patients with severe COPD, but it can also be
seen in patients with mild to moderate disease
(1015%) [5]. It is a poor prognostic indicator [6,
SYSTEMIC MANIFESTATIONS 7] and interestingly, it is independent of other tra-
OF COPD ditional indices, such as the volume of air exhaled
in the rst second of a forced spirometry maneu-
ver (FEV1) or the arterial partial pressure of oxy-
The COPD guidelines published jointly by the gen [6]. Therefore, weight loss identies a new
American Thoracic Society (ATS) and European systemic domain of COPD that needs to be taken
Respiratory Society (ERS) in 2004 were the into consideration in their clinical management.
rst to explicitly recognize that although chronic In this context, Celli et al. have recently pro-
obstructive pulmonary disease (COPD) aects the posed a composite index (the BODE index) that
lungs, it also produces signicant systemic conse- includes body weight (assessed by the body mass
quences [1]. More recently, the last update of the index, BMI), the degree of airow obstruction
Global Obstructive Lund Disease (GOLD) initi- (assessed by the FEV1 value expressed as percent-
ative published in 2006 incorporates and extends age of the reference value), the level of dyspnea
this concept by dening COPD as a preventable experienced by the patient (assessed by the modi-
and treatable disease with some signicant extra- ed Medical Research Council scale), and their
pulmonary eects that may contribute to the severity exercise capacity (assessed by the 6-min walk-
in individual patients [2]. It is interesting to note ing test), that predicts survival much more accu-
that the systemic manifestations of the disease rately than FEV1 alone [8]. This approach has the

569
Asthma and COPD: Basic Mechanisms and Clinical Management

Symptoms
Exercise tolerance
HRQL
ECOPD
Bronchitis Prognosis
Emphysema
and
bronchiolitis

Systemic effects
of COPD

Airway Weight loss (cachexia)


Airflow inflammation Skeletal muscle dysfunction
limitation Cardiovascular disease
Others
Osteoporosis
Depression and fatigue
Systemic
inflammation Cancer

FIG. 44.1 The interaction between the pulmonary and extra-pulmonary components of COPD contributes the clinical presentation of the disease.
Reproduced from Ref. [3].

TABLE 44.1 Systemic effects of COPD. In this context, systemic inammation is probably a key
contributor [15]. Several molecular abnormalities have been
Nutritional abnormalities
described. On the one hand, a number of cytokines, par-
Skeletal muscle dysfunction ticularly TNF-, activate the transcription factor NF-B,
upregulate the inducible form of the nitric oxide synthase
Cardiovascular disease (iNOS) and facilitate the degradation of myosin heavy
chains through the ubiquitinproteasome complex [16]. On
Depression
the other hand, these same cytokines have the potential to
Osteoporosis promote apoptosis (programmed cell death) in skeletal mus-
cle cells, and this has been shown to occur in patients with
Metabolic syndrome COPD and low body weight [17]. Finally, abnormalities of
Systemic inflammation
the sarcoplasmicendoplasmic reticulum calcium adenosine
triphosphatase 2 (SERCA2) have been described by Morl
et al. [18]. These investigators found that the expression of
SERCA2 was signicantly lower in COPD patients with
historical merit of being the rst to assess COPD multi- low BMI (another domain captured by the BODE index
dimensionally, but it is likely that the BODE index may be discussed earlier) and, interestingly, related to iNOS [18].
modied in the future by the inclusion of other relevant All these molecular abnormalities are likely to contribute to
domains of the disease, such as the degree of inammation [9], skeletal muscle atrophy and dysfunction in COPD [19, 20].
the number and severity of exacerbations episodes [10], and/or
the severity of lung hyperination [11], to name only a few.
Cardiovascular disease
Skeletal muscle dysfunction Cardiovascular disease (CVD) is another clinically relevant
systemic manifestation of COPD [21]. Despite the fact
Unexplained weight loss in COPD is mostly due to skel- that tobacco smoking is a major risk factor for both COPD
etal muscle atrophy [12, 13]. Further, the remaining muscle and CVD, recent investigations have shown that the risk of
mass is often dysfunctional [14]. This combination con- CVD is higher among those smokers who develop COPD as
tributes signicantly to reduce the exercise capacity (thus, compared to those who do not develop it [21]. For instance,
health status) of patients with COPD [14] and is likely to Sin et al. reported that when the lowest quintile of FEV1 is
contribute to the BODE index discussed earlier. The mech- compared with the highest quintile, the risk of cardiovascu-
anisms underlying skeletal muscle abnormalities in COPD lar mortality increases by approximately 75% both in men
are not precisely dened, but they are probably multiple and and in women, and that the presence of symptoms of chronic
interdependent, including sedentarism, malnutrition, sys- bronchitis increases the risk of coronary deaths by 50% [22].
temic inammation, and tissue hypoxia among others [13]. Further, the presence of ventricular arrhythmias increases the

570
Systemic Manifestations 44
risk of coronary events two-fold, suggesting that the cardio- For instance, its prevalence in patients with moderate to
vascular eects of COPD are amplied in those who have severe COPD (FEV1 43% of reference value) who par-
underlying cardiac rhythm disturbances [22]. Sin et al. calcu- ticipated in a cardiopulmonary rehabilitation program was
lated that, for every 10% decrease in FEV1, all-cause mortal- 47%, whereas it was 21% in controls [33].
ity increases by 14%, CVD mortality by 28%, and nonfatal Insulin resistance and diabetes mellitus are a key com-
coronary event increases by 20%, and concluded that COPD ponents of the metabolic syndrome [33]. In the Nurses
is a powerful, independent risk factor for CVD morbidity Health Study (a prospective cohort that included 103,614
and mortality [22]. These ndings were later conrmed in a female nurses) during 8 years of follow-up, a total of 2,959
large (n 11,493), retrospective cohort study in health-care new cases of type 2 diabetes were identied [35]. The risk of
databases maintained by the government of Saskatchewan type 2 diabetes was signicantly higher (multivariate relative
(Canada). Curkendall et al. found that persons diagnosed and risk 1.8) for patients with COPD, suggesting that COPD may
treated for COPD have a signicantly increased risk for hos- be a risk factor for developing type 2 diabetes [35]. Authors
pitalizations and deaths due to CVD [23]. The mechanism(s) speculated that systemic inammation in these patients might
for the synergy between COPD and CVD are still under contribute to the increased risk of type 2 diabetes [35].
investigation, but the low-grade persistent systemic inam-
mation that characterizes COPD is likely to contribute sig-
nicantly [21, 24] by causing endothelial dysfunction [25, 26]. Systemic inflammation
The systemic manifestations of COPD reviewed earlier
Depression have been often related to the presence of systemic inam-
mation in these patients [34]. Many dierent studies have
In patients with severe COPD (FEV1 50% reference now provided convincing evidence that COPD is associated
value), the prevalence of depression was 25.0% whereas it with increased levels of several pro-inammatory cytokines
was 19.6% in patients with mild to moderate COPD and (such as TNF- and its soluble receptors soluble (sTNF-
17.5% in controls [27]. When results were adjusted for R55 and sTNF-R75)), interleukins 6 (IL-6) and 8 (IL-8),
demographic variables and comorbidity, the risk for depres- acute phase reactants (such as C-reactive protein, CRP),
sion was 2.5 times greater for patients with severe COPD oxidative stress and activations of several inammatory cells
than for controls [27]. (such as neutrophils, monocytes, and lymphocytes) [13, 36].
Gan and associates conducted a meta-analysis encompass-
ing all these previous studies and showed beyond doubt
Osteoporosis that COPD is associated with systemic inammation [15].
Interestingly, other chronic conditions such as chronic heart
Osteoporosis in COPD might develop possibly due to a failure, asthma (reviewed later), obesity, diabetes, and the
number of factors related or not related to the disease, includ- normal process of aging are also associated with a similar
ing age, sedentarism, smoking, steroid use, and systemic low-grade systemic inammatory process [3739].
inammation [28, 29]. In a study by Jorgensen et al., 68% of Many questions related to systemic inammation
patients with severe COPD (FEV1 33% of reference value) in COPD remain unanswered [3]. For instance, we do
had osteoporosis or osteopenia and the use steroids could not not know if systemic inammation occurs in all patients
explain this high prevalence [30]. The occurrence of fractures, with COPD or only in a subgroup of them. Also, because
as a consequence of osteoporosis, can contribute to the dis- all studies carried out so far are cross-sectional, we do not
ability and mortality of patients with COPD and add to the know the quantitative and qualitative longitudinal variation
economic burden of the disease [31]. Randomized placebo- of systemic inammation in a given patient, although recent
controlled trials are required to assess the eect of interven- data by Pinto Plata et al. indicate that, in the absence of an
tion, such as bisphosphonates, hormone replacement, calcium episode of exacerbation, CRP values tend to be relatively
supplementation, on the prevention and treatment of oste- stable [40, 41]. In contrast, systemic inammation (like pul-
oporosis and fractures in these patients [31]. Interestingly, monary inammation) appears to burst during the episodes
very recently Sabit and coworkers have described a link of exacerbation of the disease [4246].
between CVD and osteoporosis in patients with COPD [32]. The origin of systemic inammation in COPD is
These authors found that arterial stiness and serum inam- unclear. It is possible that the pulmonary inammation that
matory mediators were particularly increased in patients with characterizes COPD spills-over into the systemic cir-
osteoporosis suggesting that age-related bone and vascular culation and/or contribute to activate the inammatory
changes occur prematurely in COPD [32]. cells during their transit through the pulmonary circula-
tion [13]. Yet, several observations appear to contradict this
hypothesis. For instance, Vernooy et al. could not nd any
Metabolic syndrome relationship between the levels of soluble TNF- (and its
receptors) and IL-8 measured in induced sputum and in
The metabolic syndrome is characterized by the presence of plasma of 18 COPD patients [47]. More recently, Hurst
abdominal obesity, atherogenic dyslipidemia, raised blood et al. were also unable to nd such relationships for individ-
pressure, presence of insulin resistance, and prothrom- ual cytokines, albeit overall, global pulmonary inammation
botic and inammatory states that predispose to CVD correlated with global systemic inammation [48]. Another
[33]. It is frequently present in patients with COPD [34]. mechanism of systemic inammation in COPD is smoking.

571
Asthma and COPD: Basic Mechanisms and Clinical Management

It can cause by itself, that is, in the absence of COPD, sys- (A) 15
temic inammation [49]. However, Vernooy et al. found
that former smoker patients with COPD also had systemic
inammation [47]. Interestingly, the persistence of inam-
mation after smoking cessation occurs also in the lungs 10
of patients with COPD [9, 50] and has actually raised the

RTL
possibility that the pathogenesis of COPD may include an
auto-immune component [51]. Another potential mecha- 5
nism of systemic inammation in COPD is the lung hyper-
ination that occurs as a consequence of the chronic airow
obstruction that characterizes the disease [52], because it can
stimulate the production of cytokines from the lungs [53 0
55]. Importantly, if this was the case, systemic inammation 35 40 45 50 55 60 65 70 75 80
in COPD should be responsive to bronchodilator therapy. Age (years)
This hypothesis is testable. In fact, in support of this possi-
bility is the very recent observation that patients undergoing (B) 15
lung volume reduction surgery (a surgical procedure aimed
at, basically, reduce lung hyperination) present a reduced
incidence of episodes of exacerbation of COPD, which are
characterized by a burst of inammation [56]. Other poten- 10
tial origin of systemic inammation in COPD is the skele-

RTL
tal muscle, which, as discussed above, is often abnormal and
dysfunctional in these patients. In support of this possibility,
Rabinovitch et al. have shown that, contrary to healthy sub- 5
jects, systemic inammation increases after muscular exercise
in patients with COPD [57]. It has to be considered also that
the bone marrow is the site of production of inammatory 0
cells and that smoking, as well as air pollution, can stimulate 35 40 45 50 55 60 65 70 75 80
their release from the bone marrow [5860]. Finally, the pos-
Age (years)
sibility that bone marrow abnormalities can contribute to
systemic inammation in COPD has not been studied sys-
tematically, but a recent study by Palange et al. showed that (C) 15
circulating hemopoietic progenitors (CD34 cells) were sig-
nicantly reduced in COPD patients as compared to controls
and that CD34[] cell counts correlated with exercise capac- 10
ity and severity of airow obstruction [61]. Two nal consid-
RTL

erations deserve comment when considering the origin of


systemic inammation in COPD. First, the normal aging pro-
cess is also associated with low-grade systemic inammation 5
[62, 63], and COPD is an age-related disease [39]. Further,
smoking has been shown to enhance telomeric loss (a marker
of cell aging) (Fig. 44.2) [64], and evidence for cell senes-
cence has been recently identied in the lung parenchyma of 0
patients with emphysema [65, 66]. Thus, a better understand- 0 20 40 60 80 100 120
ing of the mechanisms of lung aging and their relationship Packs (years)
with the pathogenesis of COPD in general, and to systemic
FIG. 44.2 Relationship between relative telomeric length (RTL), a marker
inammation in particular, may be very valuable in this set-
of biological ageing, and chronological age in never smokers ( panel A)
ting. Second, even in subjects with lung function values within and smokers with (closed symbols) and without COPD (open symbols)
the normal range (FEV1 values above 80% predicted), an ( panel B). Panel C shows the relationship between RTL and FEV1 in smokers
inverse linear relationship exists between CRP concentrations with (closed symbols) and without COPD (open symbols). For further
and FEV1 [67]. This relationship occurs also in never-smoker explanations, see text. Reproduced from Ref. [64].
individuals, suggesting that systemic inammation may be
linked to early perturbations of pulmonary function [67].
Whether systemic inammation precedes or follows lung although not proven, that systemic oxidative stress [68] and
function changes requires longitudinal studies. the increased plasma levels of pro-inammatory cytokines
Irrespective of its origin, an intriguing possibility (most notably TNF- [69]) can contribute to the pathogen-
about systemic inammation in COPD is that it may feed esis of lung damage in COPD because, rst, the inhibition
back to the lungs and contribute to enhance the chronic of the vascular endothelial growth factor (VEGF) receptors
destructive process that characterizes the disease. In other cause lung cell apoptosis and emphysema [70], second, oxi-
words, a systemic manifestation of COPD may contribute dative stress and apoptosis interact and cause emphysema
to the core pulmonary domain of the disease. It is possible, due to VEGF receptor blockade [71], and third, endothelial

572
Systemic Manifestations 44
cell death and decreased expression of VEGF and the VEGF TABLE 44.2 Systemic manifestations of asthma.
receptor KDR/FLK-1 occur in patients with smoking-
Systemic inflammation
induced emphysema [72].
Obesity

Therapeutic implications Diabetes mellitus

Cardiovascular disease
The systemic manifestations of COPD discussed earlier
oer new opportunities for therapy because they constitute Skeletal muscle abnormalities
novel targets beyond the traditional therapies in COPD
(broncho-dilators, inhaled steroids), which aimed at improv- Osteoporosis
ing lung function [2]. For instance, malnutrition and poor
Depression and panic disorders
skeletal muscle performance can be improved by diet sup-
plementation and adequate rehabilitation programs [2, 73]. Inflammatory bowel disease
Tissue hypoxia and the associated systemic manifestations
are potentially treatable by domiciliary oxygen therapy [74,
75]. Likewise, those systemic manifestations of COPD
related to systemic inammation may be responsive to anti-
inammatory therapy. Sin and coworkers showed that with-
SYSTEMIC MANIFESTATIONS OF ASTHMA
drawal of inhaled corticosteroids in patients with COPD
increased the plasma levels of CRP by about 30% and that Although often overlooked, asthma is frequently associated
2 weeks of treatment with inhaled uticasone (or oral pred- with systemic manifestations. The link between asthma and
nisolone) reduced them by about 50% [76]. If low-grade rhinitis, sinusitis, or allergic dermatitis is a good examples
chronic systemic inammation is really a relevant mecha- [83]. However, other potential systemic manifestations of
nism in the pathogenesis of many of the systemic eects of asthma (Table 44.2) have been less extensively studied than
COPD described earlier, then it is likely that the biological those of COPD discussed earlier. Several features can con-
eects described by Sin et al. may well be clinically relevant. tribute to explain this. First, asthma tends to be a disease of
In fact, a retrospective study has suggested that the risk of young (otherwise healthy) individuals [84], whereas COPD
acute myocardial infarction in COPD patients was reduced is rarely diagnosed before the sixth decade of age in indi-
by 32% in those receiving low doses of inhaled steroids viduals who often have other comorbidities [2, 85]. Second,
[77]. The recently published, study, TORCH has explored lung disease is reversible in asthma but mostly irreversible in
the potential benecial eect on survival of the combina- COPD [86]. It is plausible that the chronicity of the disease
tion of salmeterol and uticasone propionate (versus pla- may favor the development of abnormalities in other distant
cebo) in patients with moderate and severe COPD [78]. organs. Finally, available therapies (mostly inhaled steroids
Strictly speaking, the p value of the study did not reach with or without long-acting bronchodilators) are very eec-
the predetermined level of statistical signicance, but this tive in the vast majority of asthmatics [84] whereas, inter-
occurred only after adjusting for the potential eect of the estingly, the same therapy is considerably less eective in
interim analysis performed. In absolute terms, there was patients with COPD [2].
a reduction in the risk of death of 17.5% in the patients
receiving the combination of a salmeterol and utica-
sone propionate [78], a reduction that is in the same order Systemic inflammation
of magnitude as that provided by statins. Unfortunately,
no measurement of systemic inammation was obtained Airway inammation is a key pathologic mechanism in
in these subjects, so the interpretation of these results is asthma [84]. As in COPD, there may be cross-communication
still open [78]. Other therapeutic alternatives, such as the between the airways and the bone marrow in patients with
use of the antioxidant N-acetyl-cysteine [79, 80], anti- asthma through inammatory mediators that can eventu-
TNF- or other cytokine antibodies, NF-B blockers and ally lead to systemic inammation [83]. Sauleda et al. were
i-NOS inhibitors, among others, require large and rigorously the rst to report that the activity of cytochrome oxidase
conducted clinical trials before entering clinical practice but, (CytOx), the terminal enzyme of the mitochondrial respi-
as recently reviewed [81, 82], they may be interesting alter- ratory chain, was abnormally increased in circulating lym-
natives. Finally, other systemic manifestations of COPD may phocytes of patients with asthma (as compared to those with
deserve specic therapy. For instance, because patients with COPD, arthritis and/or healthy subjects) [87] (Fig. 44.3).
COPD are at increased risk of developing depression, par- This was a totally unexpected and serendipitous observa-
ticularly those with severe airow obstruction, living alone, tion because the goal of that study was to investigate if the
with airow reversibility, respiratory symptoms, and physi- observation made previously by these same investigators of
cal impairment [27], it is important to reduce symptoms and increased CytOx activity in skeletal muscle of patients with
improve physical functioning both (rehabilitation) as well as COPD [88] could be reproduced in other cell types and
use anti-depressants drugs if needed. Likewise, as the preva- other chronic inammatory diseases. As shown in Fig. 44.2,
lence of osteoporosis and osteopenia is increased in severe CytOx activity was higher in COPD than in healthy sub-
COPD, it is necessary to select the individuals at risk and to jects (as previously observed in skeletal muscle [88]), but
initiate prophylaxis or treatment for the disease [30]. its activity was one order of magnitude greater in patients

573
Asthma and COPD: Basic Mechanisms and Clinical Management

4000 First National Health and Nutrition Examination Survey


CytOx activity (nKat/g protein)
studied 14,407 subjects, aged 2574, followed-up dur-
3000 ing a mean of 10 years and found that, both at baseline
and during the follow-up, increasing BMI was associated
2000 with increased prevalence of asthma [95]. Other studies
have shown that obesity precedes and predicts the onset
of asthma, increased obesity leads to more severe asthma,
weight reduction improves asthmatic symptoms, and obes-
600 ity co-occurs with intermediate asthma phenotypes [93, 94].
400 These observations suggest that obesity and asthma may be
causally related through immunologic and inammatory,
200 hormonal, genetic, and/or nutritional mechanisms [93].
0
COPD Asthma Arthritis Healthy
Diabetes mellitus
FIG. 44.3 Cytochrome oxidase activity in circulating lymphocytes of
healthy subjects and patients with COPD, asthma and rheumatoid arthritis.
Reproduced from Ref. [87].
It is unclear whether or not asthma and type 1 diabetes mel-
litus are related (and the direction of such potential relation-
ship). Contrary to ndings in COPD, the Nurses Health
with asthma [87] (Fig. 44.3). The mechanisms and impli- Study could not detect an increased risk of type 2 diabetes
cations of these ndings were unclear, but authors specu- among asthmatic patients [35]. Yet, other authors have sug-
lated that they were likely to reect activation of circulating gested an inverse relationship between asthma and type 1
lymphocytes (thus systemic inammation) in these con- diabetes mellitus (DM1) [96]. It has been hypothesized
ditions [87]. Other studies that were published later, pro- that because, according to the hygiene hypothesis, the risk
vided further evidence of systemic inammation in asthma. of asthma decreases with infections early in childhood by
Jousilahti et al. found that the age-adjusted odds ratios shifting the Th2 prole, common at birth, to the Th1 phe-
of asthma increased in proportion to the levels of several notype, the latter may favor the development of DM1 [96].
inammatory markers in plasma (CRP, serum amyloid-A, Thus further research is required to nd out whether or not
and brinogen), and that this association was independ- asthma and DM1 are related.
ent of smoking, the waist-to-hip ratio (a marker of central
obesity), and symptoms of chronic bronchitis [89]. Silvestri
et al. reported that patients with severe asthma had higher Cardiovascular disease
circulating neutrophil and eosinophil number, as well as
increased serum levels of IL-8 and TNF-, than controls Several pieces of evidences indicate that the risk of CVD
and that there was a positive correlation between circulating is increased in asthmatics [97]. Schanen et al. reported that
neutrophil counts, plasma TNF- levels, and exhaled nitric asthma was an independent risk factor for incident stroke
oxide (a well-established marker of pulmonary inamma- (but not coronary heart disease) in a cohort of 13,501 adults
tion in asthma) as well as between TNF- and IL-8 plasma followed during 14 years [98]. In an even larger cohort
levels and the degree of airow obstruction (FEV1, % of (74,342 participants in the Canadian Community Health
reference) [90]. Higashimoto et al. found that the systemic Survey), Dogra et al. found that asthmatics were 43% more
inammatory prole of patients with asthma was similar to likely to have heart disease and 36% more likely to have
that of patients with COPD, including increases in serum high blood pressure than non-asthmatics [97]. Interestingly,
CRP, brinogen, TNF-, IL-6, and the tissue inhibitor of the age of onset of asthma did not inuence this relation-
matrix metalloproteinase TIMP-1 [91]; interestingly, how- ship signicantly [97]. Finally, Wu et al. reported increases
ever, serum TGF-1 levels were higher in asthma patients in multiple inammatory markers of atherosclerosis in
than in COPD patients [91]. Finally, the latest US National patients with asthma [86]. Thus, available evidence supports
Health and Nutrition Examination Survey in 8020 adults the concept that CVD risk is increased in patients with
found that those with asthma have higher levels of CRP in asthma.
plasma, suggesting that this may be a clinically useful marker
for asthma severity and control [92]. From all these studies,
therefore, it is clear that asthma is also associated with sys-
temic inammation. Like in COPD, thus, it is possible that
Skeletal muscle abnormalities
systemic inammation in asthma contributes to some (or
Skeletal muscle abnormalities are uncommon (or at least,
all) the systemic manifestations reviewed later.
seldom studied) in patients with asthma, at variance with
COPD (see above). In one of the rare studies that have
Obesity addressed this issue, Picado et al. could not nd signicant
dierences between oral steroid-dependent asthmatics and
At variance with COPD, where unexplained weight loss is age- and sex-matched steroid nave asthmatics in respira-
frequent (see above), in patients with asthma, a relationship tory and skeletal muscle strength, endurance time, or ber-
with obesity has been suggested [93, 94]. For instance, the type composition [99].

574
Systemic Manifestations 44
Osteoporosis References
A recent systematic review of the literature by Kearney et al. 1. Celli BR, MacNee W, Agusti AG, Anzueto A, Berg BR, Buist AS,
found that patients with asthma (and COPD, as discussed Calverley P, Chavannes N, Dillard T, Fahy B, Fein A, Hener J, Lareau
earlier) are at risk of osteoporosis, particularly those treated SC, Meek P, Martinez F, McNicholas WT, Muris J, Austergard E,
with high doses of corticosteroids [100]. Pauwels R, Rennard S, Rossi A, Siafakas N, Tiep B, Vestbo J, Wouters
E, ZuWallack R. Standards for the diagnosis and treatment of patients
with COPD: A summary of the ATS/ERS position paper. Eur Respir J
Depression and panic disorders 23: 93246, 2004.
2. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P,
Fukuchi Y, Jenkins C, Rodriguez-Roisin R, van WC, Zielinski J.
Depression and panic disorders are frequent in patients Global strategy for the diagnosis, management, and prevention of
with asthma. Schneider et al. found that about 4% of asth- chronic obstructive pulmonary disease: GOLD executive summary.
matics visited in primary care suered from major depres- Am J Respir Crit Care Med 176: 53255, 2007.
sive disorder, 23% from minor depressive disorder, and 8% 3. Agusti A. Systemic eects of chronic obstructive pulmonary disease:
from panic disorder [101]. Further, the presence of depres- What we know and what we dont know (but should). Proceedings of the
sion predicted hospitalization within the subsequent year American Thoracic Society, 4:52225, 2007.
(odds ratios, OR 6.1) and panic disorder predicted unsched- 4. Agusti A, Thomas A. Ne lecture. Chronic obstructive pulmonary dis-
uled emergency home visits (OR 4.8). Likewise, McCauley ease: A systemic disease. Proc Am Thorac Soc 3: 47881, 2006.
et al. found that 125 out of 767 adolescents with asthma 5. Schols AM, Soeters PB, Dingemans AM, Mostert R, Frantzen PJ,
Wouters EF. Prevalence and characteristics of nutritional depletion
(16.2%) reported signicant anxiety and depressive dis-
in patients with stable COPD eligible for pulmonary rehabilitation.
orders in the last 12 months, and this was related to func- Am Rev Respir Dis 147: 115156, 1993.
tional status [102]. Overall, these results indicate that the 6. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss is a revers-
impact of psychiatric comorbidity on health outcomes for ible factor in the prognosis of chronic obstructive pulmonary disease.
patients with asthma is substantial [103]. Interestingly, the Am J Respir Crit Care Med 157: 179197, 1998.
frequency of symptoms of anxiety and depression is greater 7. Landbo C, Prescott E, Lange P, Vestbo J, Almdal TP. Prognostic value
among asthma patients (particularly those with uncon- of nutritional status in chronic obstructive pulmonary disease. Am J
trolled asthma) than among patients with COPD [104]. Respir Crit Care Med 160: 185661, 1999.
8. Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca M,
Mendez RA, Pinto Plata V, Cabral HJ. The Body-mass index, airow
Inflammatory bowel disease obstruction, dyspnea, and exercise capacity index in chronic obstructive
pulmonary disease. N Engl J Med 350: 100512, 2004.
The prevalence of asthma is particularly increased in 9. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,
Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD. The
patients with inammatory bowel disease. Berstein et al.
nature of small-airway obstruction in chronic obstructive pulmonary
found that patients with ulcerative colitis (n 3879) and disease. N Engl J Med 350: 264553, 2004.
Crohns disease (n 4193) had a signicantly greater like- 10. Soler-Cataluna JJ, Martinez-Garcia MA, Roman Sanchez P, Salcedo E,
lihood of having arthritis, asthma, bronchitis, psoriasis, Navarro M, Ochando R. Severe acute exacerbations and mortality in
and pericarditis than population controls, and that asthma patients with chronic obstructive pulmonary disease. Thorax 60: 925
was the most common comorbidity in Crohns disease 31, 2005.
patients [105]. The reasons for these associations are 11. Casanova C, Cote C, de Torres JP, guirre-Jaime A, Marin JM, Pinto-
unknown but shared inammatory mechanisms have been Plata V, Celli BR. Inspiratory-to-total lung capacity ratio predicts
postulated [83]. mortality in patients with chronic obstructive pulmonary disease. Am J
Respir Crit Care Med 171: 59197, 2005.
12. Wouters EFM. A wasting disease. In: Voelkel NF, MacNee W (eds).
Therapeutic implications Chronic Obstructive Lung Disease, pp. 36476. Hamilton, ON, Canada:
B.C. Dekker, 2002.
13. Agusti AG, Noguera A, Sauleda J, Sala E, Pons J, Busquets X. Systemic
Inhaled steroids (with or without long-acting bronchodila- eects of chronic obstructive pulmonary disease. Eur Respir J 21: 347
tors) are very eective in patients with asthma [84]. They 60, 2003.
reduce local inammation and improve pulmonary function 14. American Thoracic Society and European Respiratory Society. Skeletal
but their eects upon the systemic manifestations of asthma muscle dysfunction in chronic obstructive pulmonary disease. Am J
are less well studied [83]. In the context of the systemic Respir Crit Care Med 159: S1S40, 1999.
manifestations of asthma, oral medications, such as leuko- 15. Gan WQ, Man SF, Senthilselvan A, Sin DD. Association between
triene receptor antagonists, can block the action of cystei- chronic obstructive pulmonary disease and systemic inammation: A
nyl leukotrienes and thus improve both asthma and rhinitis systematic review and a meta-analysis. Thorax 59: 57480, 2004.
and other conditions systemically linked with asthma [83]. 16. Agusti A, Morla M, Sauleda J, Saus C, Busquets X. NF-KB activation
and iNOS upregulation in skeletal muscle of patients with COPD and
Other potential treatments include receptor-blocking mol-
low body weight. Thorax 59: 48387, 2004.
ecules and synthesis inhibitors related to eicosanoid inam-
17. Agusti AGN, Sauleda J, Miralles C, Gomez C, Togores B, Sala E, Batle
mation [83]. Further, as depression and panic disorders are S, Busquets X. Skeletal muscle apoptosis and weight loss in chronic
frequent in patients with asthma and both are predictive of obstructive pulmonary disease. Am J Respir Crit Care Med 166: 48589,
health-care resources utilization, specic psychiatric inter- 2002.
ventions may reduce inappropriate health-care utilization 18. Morla M, Iglesias A, Sauleda J, Cosio B, Agusti A, Busquets X.
and avoid adverse outcomes [101]. [Reduced expression of the sarcoplasmic calcium pump SERCA2 in

575
Asthma and COPD: Basic Mechanisms and Clinical Management

skeletal muscle from patients with chronic obstructive pulmonary dis- 39. Fabbri LM, Ferrari R. Chronic disease in the elderly: Back to the
ease and low body weight]. Arch Bronconeumol 43: 48, 2007. future of internal medicine. Breathe 3: 4049, 2006.
19. Montes de Oca M, Loeb E, Torres SH, De Sanctis J, Hernandez N, 40. Pinto-Plata V, Toso J, Lee K, Parks D, Bilello J, Mullerova H, Desouza
Talamo C. Peripheral muscle alterations in non-COPD smokers. Chest M, Vessey RS, Celli B. Proling serum biomarkers in patients with
133: 1318, 2008. COPD: Associations with Clinical Parameters. Thorax 62: 595601,
20. Barreiro EA, Schols MWJ, Polkey MI, Galdiz JB, Gosker HR, 2007.
Swallow EB, Coronell C, Gea J, on behalf of the ENIGMA in COPD 41. Pinto-Plata VM, Mullerova H, Toso JF, Feudjo-Tepie M, Soriano JB,
project. Cytokine prole in quadriceps muscles of patients with severe Vessey RS, Celli BR. C-reactive protein in patients with COPD, con-
COPD. Thorax 63: 100107, 2008. trol smokers and non-smokers. Thorax 61: 2328, 2006.
21. Sin DD, Man SF. Why are patients with chronic obstructive pulmo- 42. Malo O, Sauleda J, Busquets X, Miralles C, Agusti AG, Noguera A.
nary disease at increased risk of cardiovascular diseases? The potential Inamacin sistmica durante las agudizaciones de la enfermedad pul-
role of systemic inammation in chronic obstructive pulmonary dis- monar obstructiva crnica. Arch Bronconeumol 38: 17276, 2002.
ease. Circulation 107: 151419, 2003. 43. Dentener MA, Creutzberg EC, Schols AM, Mantovani A, vant Veer C,
22. Sin DD, Man SF. Chronic obstructive pulmonary disease as a risk fac- Buurman WA, Wouters EF. Systemic anti-inammatory mediators in
tor for cardiovascular morbidity and mortality. Proc Am Thorac Soc 2: COPD: Increase in soluble interleukin 1 receptor II during treatment
811, 2005. of exacerbations. Thorax 56: 72126, 2001.
23. Curkendall SM, DeLuise C, Jones JK, Lanes S, Stang MR, Goehring 44. Drost EM, Skwarski KM, Sauleda J, Soler N, Roca J, Agusti A,
E Jr, She D. Cardiovascular disease in patients with chronic obstruc- MacNee W. Oxidative stress and airway inammation in severe exac-
tive pulmonary disease, Saskatchewan Canada cardiovascular disease in erbations of COPD. Thorax 60: 293300, 2005.
COPD patients. Ann Epidemiol 16: 6370, 2006. 45. Wedzicha JA, Seemungal TA, MacCallum PK, Paul EA, Donaldson
24. McAllister DA, Maclay JD, Mills NL, Mair G, Miller J, Anderson D, GC, Bhowmik A, Je ries DJ, Meade TW. Acute exacerbations of
Newby DE, Murchison JT, MacNee W. Arterial Stiness is chronic obstructive pulmonary disease are accompanied by eleva-
Independently associated with emphysema severity in patients with tions of plasma brinogen and serum IL-6 levels. Thromb Haemost 84:
COPD. Am J Respir Crit Care Med 176: 120814, 2007. 21015, 2000.
25. Dinh-Xuan AT, Higenbottam TW, Clelland CA, Pepke-Zaba J, 46. Pinto-Plata VM, Livnat G, Girish M, Cabral H, Masdin P, Linacre P,
Cremona G, Butt Y, Large SR, Wells FC, Wallwork J. Impairment Dew R, Kenney L, Celli BR. Systemic cytokines, clinical and physi-
of endothelium-dependent pulmonary-artery relaxation in chronic ological changes in patients hospitalized for exacerbation of COPD.
obstructive lung disease. N Engl J Med 324: 153947, 1991. Chest 131: 3743, 2007.
26. Saadjian A, Philip-Joet F, Levy S, Arnaud A. Vascular and cardiac 47. Vernooy JH, Kucukaycan M, Jacobs JA, Chavannes NH, Buurman
reactivity in pulmonary hypertension due to chronic obstructive lung WA, Dentener MA, Wouters EF. Local and systemic inammation
disease: Assessment with varios oxygen concentrations. Eur Respir J 5: in patients with chronic obstructive pulmonary disease: Soluble tumor
52530, 1992. necrosis factor receptors are increased in sputum. Am J Respir Crit Care
27. van Manen JG, Bindels PJ, Dekker FW, IJzermans CJ, van der Zee JS, Med 166: 121824, 2002.
Schade E. Risk of depression in patients with chronic obstructive pul- 48. Hurst JR, Wilkinson TM, Perera WR, Donaldson GC, Wedzicha JA.
monary disease and its determinants. Thorax 57: 41216, 2002. Relationships among bacteria, upper airway, lower airway, and systemic
28. Incalzi RA, Caradonna P, Ranieri P, Basso S, Fuso L, Pagano F, Ciappi inammation in COPD. Chest 127: 121926, 2005.
G, Pistelli R. Correlates of osteoporosis in chronic obstructive pulmo- 49. Yanbaeva DG, Dentener MA, Creutzberg EC, Wesseling G, Wouters EF.
nary disease. Respir Med 94: 107984, 2000. Systemic eects of smoking. Chest 131: 155766, 2007.
29. Gross NJ. Extrapulmonary eects of chronic obstructive pulmonary 50. Retamales I, Elliot MW, Meshi B, Coxson HO, Pare PD, Sciurba FC,
disease. Curr Opin Pulm Med 7: 8492, 2001. Rogers RM, Hayashi S, Hogg JC. Amplication of inammation in
30. Jorgensen NR, Schwarz P, Holme I, Henriksen BM, Petersen LJ, emphysema and its association with latent adenoviral infection. Am J
Backer V. The prevalence of osteoporosis in patients with chronic Respir Crit Care Med 164: 46973, 2001.
obstructive pulmonary disease: A cross sectional study. Respir Med 101: 51. Agusti A, MacNee W, Donaldson K, Cosio M. Hypothesis:
17785, 2007. Does COPD have an autoimmune component? Thorax 58: 83234,
31. Ionescu AA, Schoon E. Osteoporosis in chronic obstructive pulmonary 2003.
disease. Eur Respir J Suppl 46: 64s75s, 2003. 52. Parker CM, ODonnell DE. Dynamic hyperination, dyspnoea
32. Sabit R, Bolton CE, Edwards PH, Pettit RJ, Evans WD, McEniery and exercise intolerance in chronic obstructive pulmonary dis-
CM, Wilkinson IB, Cockcroft JR, Shale DJ. Arterial stiness and ease. In: Barnes PJ, Postma DS (eds). Essentials of COPD Van
osteoporosis in chronic obstructive pulmonary disease. Am J Respir Crit Zuiden Communications, pp. 230. The Netherlands: Van Zuiden
Care Med 175: 125965, 2007. Communications BV, 2005.
33. Marquis K, Maltais F, Duguay V, Bezeau AM, Leblanc P, Jobin J, 53. Vassilakopoulos T, Roussos C, Zakynthinos S. The immune response to
Poirier P. The metabolic syndrome in patients with chronic obstructive resistive breathing. Eur Respir J 24: 103343, 2004.
pulmonary disease. J Cardiopulm Rehabil 25: 22632, 2005. 54. Vassilakopoulos T, Katsaounou P, Karatza MH, Kollintza A,
34. Fabbri LM, Rabe KF. From COPD to chronic systemic inammatory Zakynthinos S, Roussos C. Strenuous resistive breathing induces
syndrome?. Lancet 370: 79799, 2007. plasma cytokines: Role of antioxidants and monocytes. Am J Respir Crit
35. Rana JS, Mittleman MA, Sheikh J, Hu FB, Manson JE, Colditz GA, Care Med 166: 157278, 2002.
Speizer FE, Barr RG, Camargo CA Jr. Chronic obstructive pulmonary 55. Vassilakopoulos T, Zakynthinos S, Roussos C. Strenuous resistive
disease, asthma, and risk of type 2 diabetes in women. Diabetes Care 27: breathing induces proinammatory cytokines and stimulates the HPA
247884, 2004. axis in humans. Am J Physiol 277: R101319, 1999.
36. Wouters EF, Creutzberg EC, Schols AM. Systemic eects in COPD. 56. Washko GR, Fan VS, Ramsey SD, Mohsenifar Z, Martinez F, Make
Chest 121: 127S130S, 2002. BJ, Sciurba FC, Criner GJ, Minai O, DeCamp MM, Reilly JJ, for the
37. Sevenoaks MJ, Stockley RA. Chronic Obstructive Pulmonary Disease, National Emphysema Treatment Trial Research Group. The Eect
inammation and co-morbiditya common inammatory phenotype? of Lung Volume Reduction Surgery on Chronic Obstructive
Respir Res 7: 70, 2006. Pulmonary Disease Exacerbations. Am J Respir Crit Care Med 177:
38. De MM, Franceschi C, Monti D, Ginaldi L. Inamm-ageing and life- 16469, 2008.
long antigenic load as major determinants of ageing rate and longevity. 57. Rabinovich RA, Figueras M, Ardite E, Carbo N, Troosters T, Filella X,
FEBS Lett 579: 203539, 2005. Barbera JA, Fernandez-Checa JC, Argiles JM, Roca J. Increased

576
Systemic Manifestations 44
tumour necrosis factor-alpha plasma levels during moderate-intensity 79. Koechlin C, Couillard A, Simar D, Cristol JP, Bellet H, Hayot M,
exercise in COPD patients. Eur Respir J 21: 78994, 2003. Prefaut C. Does oxidative stress alter quadriceps endurance in chronic
58. van Eeden SF, Yeung AC, Quinlam K, Hogg JC. Systemic response obstructive pulmonary disease? Am J Respir Crit Care Med 169: 1022
to ambient particulate matter. Relevance to chronic obstructive pulmo- 27, 2004.
nary disease. Proc Am Thorac Soc 2: 6167, 2005. 80. Decramer M, Rutten-Van MM, Dekhuijzen PN, Troosters T, van
59. Mukae H, Vincent R, Quinlan K, English D, Hards J, Hogg JC, van HC, Pellegrino R, Van Schayck CP, Olivieri D, Del DM, De BW,
Eeden SF. The eect of repeated exposure to particulate air pollution Lankhorst I, Ardia A. Eects of N-acetylcysteine on outcomes in
(pm(10)) on the bone marrow. Am J Respir Crit Care Med 163: 2019, chronic obstructive pulmonary disease (Bronchitis Randomized on
2001. NAC Cost-Utility Study, BRONCUS): A randomised placebo-
60. van Eeden SF, Hogg JC. The response of human bone marrow to controlled trial. Lancet 365: 155260, 2005.
chronic cigarette smoking. Eur Respir J 15: 91521, 2000. 81. Agusti AG. COPD, a multicomponent disease: Implications for man-
61. Palange P, Testa U, Huertas A, Calabro L, Antonucci R, Petrucci E, agement. Respir Med 99: 67082, 2005.
Pelosi E, Pasquini L, Satta A, Morici G, Vignola MA, Bonsignore MR. 82. Barnes PJ, Stockley RA. COPD: Current therapeutic interventions and
Circulating haemopoietic and endothelial progenitor cells are decreased future approaches. Eur Respir J 25: 1084106, 2005.
in COPD. Eur Respir J 27: 52941, 2006. 83. Bjermer L. Time for a paradigm shift in asthma treatment: From
62. De Martinis M, Franceschi C, Monti D, Ginaldi L. Inammation relieving bronchospasm to controlling systemic inammation. J Allergy
markers predicting frailty and mortality in the elderly. Exp Mol Pathol Clin Immunol 120: 126975, 2007.
80: 21927, 2006. 84. Bateman ED, Hurd SS, Barnes PJ, Bousquet J, Drazen JM, FitzGerald M,
63. Ginaldi L, De MM, Monti D, Franceschi C. Chronic antigenic load Gibson P, Ohta K, OByrne P, Pedersen SE, Pizzichini E, Sullivan SD,
and apoptosis in immunosenescence. Trends Immunol 26: 7984, 2005. Wenzel SE, Zar HJ. Global strategy for asthma management and pre-
64. Morla M, Busquets X, Pons J, Sauleda J, MacNee W, Agusti AG. vention: GINA executive summary. Eur Respir J 31: 14378, 2008.
Telomere shortening in smokers with and without COPD. Eur Respir 85. Fabbri LM, Luppi F, Beghe B, Rabe KF. Complex chronic comorbidi-
J 27: 52528, 2006. ties of COPD. Eur Respir J 31: 20412, 2008.
65. Muller KC, Welker L, Paasch K, Feindt B, Erpenbeck VJ, Hohlfeld JM, 86. Wu TL, Chang PY, Tsao KC, Sun CF, Wu LL, Wu JT. A panel of
Krug N, Nakashima M, Branscheid D, Magnussen H, Jorres RA, multiple markers associated with chronic systemic inammation and
Holz O. Lung broblasts from patients with emphysema show mark- the risk of atherogenesis is detectable in asthma and chronic obstruc-
ers of senescence in vitro. Respir Res 7: 32, 2006. tive pulmonary disease. J Clin Lab Anal 21: 36771, 2007.
66. Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence in patients with 87. Sauleda J, Garcia-Palmer FJ, Gonzalez G, Palou A, Agusti AG. The
pulmonary emphysema. Am J Respir Crit Care Med 174: 88693, 2006. activity of cytochrome oxidase is increased in circulating lymphocytes
67. Aronson D, Roterman I, Yigla M, Kerner A, Avizohar O, Sella R, of patients with chronic obstructive pulmonary disease, asthma, and
Bartha P, Levy Y, Markiewicz W. Inverse association between pulmo- chronic arthritis. Am J Respir Crit Care Med 161: 3235, 2000.
nary function and C-reactive protein in apparently Healthy Subjects. 88. Sauleda J, Garca-Palmer FJ, Wiesner R, Tarraga S, Harting I, Tomas P,
Am J Respir Crit Care Med 174: 62632, 2006. Gomez C, Saus C, Palou A, Agust AGN. Cytochrome oxidase activity
68. Rahman I, Morrison D, Donaldson K, MacNee W. Systemic oxidative and mitochondrial gene expression in skeletal muscle of patients with
stress in asthma, COPD, and smokers. Am J Respir Crit Care Med 154: chronic obstructive pulmonary disease. Am J Respir Crit Care Med 157:
105560, 1996. 141317, 1998.
69. Reid MB, Li YP. Cytokines and oxidative signalling in skeletal muscle. 89. Jousilahti P, Salomaa V, Hakala K, Rasi V, Vahtera E, Palosuo T. The
Acta Physiol Scand 171: 22532, 2001. association of sensitive systemic inammation markers with bronchial
70. Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S, asthma. Ann Allergy Asthma Immunol 89: 38185, 2002.
Hirth PK, Waltenberger J, Voelkel NF. Inhibition of VEGF receptors 90. Silvestri M, Bontempelli M, Giacomelli M, Malerba M, Rossi GA,
causes lung cell apoptosis and emphysema. J Clin Invest 106: 131119, Di SA, Rossi A, Ricciardolo FL. High serum levels of tumour necrosis
2000. factor-alpha and interleukin-8 in severe asthma: Markers of systemic
71. Tuder RM, Zhen L, Cho CY, Taraseviciene-Stewart L, Kasahara Y, inammation? Clin Exp Allergy 36: 137381, 2006.
Salvemini D, Voelkel NF, Flores SC. Oxidative stress and apoptosis 91. Higashimoto Y, Yamagata Y, Taya S, Iwata T, Okada M, Ishiguchi T,
interact and cause emphysema due to vascular endothelial growth fac- Sato H, Itoh H. Systemic inammation in chronic obstructive pulmo-
tor receptor blockade. Am J Respir Cell Mol Biol 29: 8897, 2003. nary disease and asthma: Similarities and dierences. Respirology 13:
72. Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF. 12833, 2008.
Endotelial cell death and decreased expression of vegf and KDR/FLK-1 92. Arif AA, Delclos GL, Colmer-Hamood J. Association between
in smoking-induced emphysema. Am J Respir Crit Care Med 161: asthma, asthma symptoms and C-reactive protein in US adults: Data
A583, 2000. from the National Health and Nutrition Examination Survey, 1999
73. Wilt TJ, Niewoehner D, MacDonald R, Kane RL. Management of 2002. Respirology 12: 67582, 2007.
stable chronic obstructive pulmonary disease: A Systematic Review for 93. Castro-Rodriguez JA. Relationship between obesity and asthma. Arch
a Clinical Practice Guideline. Ann Intern Med 147: 63953, 2007. Bronconeumol 43: 17175, 2007.
74. Nocturnal Oxygen Therapy Trial Group. Continuous or nocturnal oxy- 94. Brand C, Sundararajan V, Jones C, Hutchinson A, Campbell D.
gen therapy in hypoxemic chronic obstructive lung disease. A clinical Readmission patterns in patients with chronic obstructive pulmonary
trial. Ann Intern Med 93: 39198, 1980. disease, chronic heart failure and diabetes mellitus: An administrative
75. Report of the Medical Research Council Working Party. Long term dataset analysis. Intern Med J 35: 29699, 2005.
domiciliary oxygen therapy in chronic hypoxic cor pulmonale compli- 95. Stanley AH, Demissie K, Rhoads GG. Asthma development with
cating chronic bronchitis and emphysema. Lancet 1: 68185, 1981. obesity exposure: Observations from the cohort of the National Health
76. Sin DD, Lacy P, York E, Man SFP. Eects of uticasone on systemic and Nutrition Evaluation Survey Epidemiologic Follow-up Study
markers of inammation in chronic obstructive pulmonary disease. Am (NHEFS). J Asthma 42: 9799, 2005.
J Respir Crit Care Med 170: 76065, 2004. 96. Alves C, Diniz AB, Souza MB, Ponte EV, Araujo MI. Controversies
77. Huiart L, Ernst P, Ranouil X, Suissa S. Low-dose inhaled corticoster- in the association between type 1 diabetes and asthma. Arq Bras
oids and the risk of acute myocardial infarction in COPD. Eur Respir J Endocrinol Metabol 51: 93037, 2007.
25: 63439, 2005. 97. Dogra S, Ardern CI, Baker J. The relationship between age of asthma
78. Vestbo J. The TORCH (towards a revolution in COPD health) sur- onset and cardiovascular disease in Canadians. J Asthma 44: 84954,
vival study protocol. Eur Respir J 24: 20610, 2004. 2007.

577
Asthma and COPD: Basic Mechanisms and Clinical Management

98. Schanen JG, Iribarren C, Shahar E, Punjabi NM, Rich SS, 102. McCauley E, Katon W, Russo J, Richardson L, Lozano P. Impact of
Sorlie PD, Folsom AR. Asthma and incident cardiovascular disease: anxiety and depression on functional impairment in adolescents with
The Atherosclerosis Risk in Communities Study. Thorax 60: 63338, asthma. Gen Hosp Psychiatry 29: 21422, 2007.
2005. 103. Kullowatz A, Kanniess F, Dahme B, Magnussen H, Ritz T.
99. Picado C, Fiz JA, Montserrat JM, Grau JM, Fernandez-Sola J, Association of depression and anxiety with health care use and qual-
Luengo MT, Casademont J, gusti-Vidal A. Respiratory and skel- ity of life in asthma patients. Respir Med 101: 63844, 2007.
etal muscle function in steroid-dependent bronchial asthma. Am Rev 104. Carvalho NS, Ribeiro PR, Ribeiro M, Nunes MP, Cukier A, Stelmach R.
Respir Dis 141: 1420, 1990. Comparing asthma and chronic obstructive pulmonary disease in
100. Kearney DM, Lockey RF. Osteoporosis and asthma. Ann Allergy terms of symptoms of anxiety and depression. J Bras Pneumol 33: 16,
Asthma Immunol 96: 76974, 2006. 2007.
101. Schneider A, Lowe B, Meyer FJ, Biessecker K, Joos S, Szecsenyi J. 105. Bernstein CN, Wajda A, Blanchard JF. The clustering of other chronic
Depression and panic disorder as predictors of health outcomes for inammatory diseases in inammatory bowel disease: A population-
patients with asthma in primary care. Respir Med 102(3): 35966, 2007. based study. Gastroenterology 129: 82736, 2005.

578
Therapies for 8
PART

Asthma and COPD


Cardiovascular Effects
45 CHAPTER

FEV1 to FVC ratio. There have been many stud-


INTRODUCTION ies that have evaluated the relationship between
Don D. Sin
FEV1 and cardiovascular diseases (CVD) in var- The James Hogg iCAPTURE Center for
Chronic obstructive pulmonary disease ious populations. Almost universally, these stud- Cardiovascular and Pulmonary Research
(COPD) and asthma are inammatory disor- ies have shown that reduced FEV1, regardless of (St. Pauls Hospital), Vancouver, BC,
ders of the lung, characterized by airow limi- the cause, is a signicant risk factor for CVD. Canada
tation and symptoms of dyspnea and cough [1]. As an example, in the First National Health and
In both of these conditions spirometric param- Examination Survey Epidemiologic Follow-up
eters are usually abnormal with a forced expira- Study (n 1861), Sin and colleagues found that
tory volume in one second (FEV1) to forced individuals in the lowest quintile of FEV1 (i.e.
vital capacity (FVC) ratio of less than 80%. reduced lung function) had the highest risk for
In asthma, this ratio may normalize following cardiovascular mortality (relative risk, RR, 3.36;
administration of a bronchodilator, whereas in 95% condence interval, CI, 1.547.34), while
COPD, there is little or no reversibility of lung individuals in the highest FEV1 quintile had
function [1]. Although these disorders are dis- the lowest risk. The mortality risk was particu-
tinct and have dierent pathophysiologies, in larly notable for deaths related to ischemic heart
both conditions, cardiovascular complications disease (RR, 5.65; 95% CI, 2.2614.13). The
are frequently observed. For instance, in patients CVD risk was slightly lower in FEV1 quintiles
with COPD, the risk of cardiovascular hospi- 2 and 3 but still signicantly higher than that
talizations and mortality including those related for the lowest quintile group (RR, 2.00, 95%
to ischemic heart disease, stroke, arrhythmias, CI, 1.033.89 for quintile 2; and RR, 2.22; 95%
heart failure, and sudden death is two to three CI, 1.234.01 for quintile 3) suggesting that the
times higher than those without COPD [2]. relationship between reduced FEV1 and CVD is
In mild to moderate cases of COPD, the lead- linear [2]. Similar ndings were observed when
ing causes of hospitalization are cardiovascular the analysis was limited to nonsmokers suggest-
events, accounting for approximately 50% of all ing that the relationship between reduced FEV1
cases [3]. In this chapter, we will review the epi- and CVD is independent of the eects of ciga-
demiological evidence and explore the potential rette smoking.
mechanistic link(s) between obstructive airways In another population-based study, the
diseases and cardiovascular comorbidities. SALIA Cohort (Study on the inuence of
Air pollution on Lung function, Inammation
and Aging) investigators found that the RR of
cardiovascular mortality of women with FEV1
EPIDEMIOLOGY less than 80% of predicted was 3.79 (95% CI,
1.648.74) at 5 years and 1.35 (95% CI, 0.66 to
2.77) at 12 years of follow-up compared with
Reduced lung function and CVD women who had normal lung function [4]. In
a Norway study, the investigators found that
One of the dening features of asthma and the risk of CVD mortality increased by 7%
COPD is reduced FEV1 in addition to a reduced with every 10% decrease in FEV1 (as percent

581
582

TABLE 45.1 Baseline characteristics of included studies and their reported association between FEV1 and cardiovascular mortality (from Ref. [2]).

Mean FEV1 Current


Publication (liter or % of smokers FEV1 categorization Follow-up RR of cardiovascular
Author year Study population Sample size Age (year) Male (%) predicted) (%) (% predicted or liter) (years) mortality (95% CI)

Marcus [11] 1989 Honolulu Heart Program, 5924 54 100 2.71 (94%) 48 Quintiles (2.10 l versus 1518 1.93 (1.46, 2.54)
US (Japanese American) 3.28 l)

Higgins [7] 1970 Tecumseh, US 5140 1675 47.7 NA NA 2.0 l (1.4b) versus 26 5.03(3.07, 8.22)
(range) 2.0 l (1.4b)

Hole [10] 1996 Renfrew & Paisley, UK 15,411 4564 45.8 2.83a 36 Quintiles (7375% 15 1.56 (1.26, 1.92)a
(range) 1.99b versus 108113%) 1.88 (1.44, 2.47)b

Beaty [12] 1985 Baltimore Longitudinal 874 50.7 100 95.6% 21 80% versus 80% 24 1.58 (0.96, 2.60)
Study of Aging, US

Schunemann [9] 2000 BuffaloErie County, US 1195 46.8 46.4 2.8 58.3 Quintiles (80% versus 29 2.11 (1.20, 3.71)a
109114%) 1.96 (0.99, 3.88)b

Ebi-Kryston [13] 1988 Whitehall Civil Servants, UK 17,717 4064 100 NA NA 65% versus 65% 10 1.49 (1.24, 1.80)
(range)

Lange [14] 1991 Copenhagen City Study, 12,511 53.1 NA NA NA 60% versus 80% 6.5 1.8 (1.4, 2.4)
Denmark

Tockman [15] 1989 Washington County, 884 NA 100 NA 32.3 Quartiles (65% versus 10 3.66 (1.76, 7.61)
Maryland 100%)

Krzyzanowski [16] 1986 Cracow, Poland 3047 1970 59 NA 38.4 65% versus 76100% 13 2.56 (1.35, 4.85)a
(range) 1.99 (0.95, 4.18)b

Hospers [17] 1999 Vlagtwedde-Vlaardingen, 5382 36 54 98% 55 80% versus 100% 25 1.82 (1.42, 2.34)
Netherlands

Kuller [18] 1989 Multiple Risk Factor 7368c 46 100 3.38 64 Quintiles (2.83.0 l 7 2.33 (1.35, 4.03)d
Intervention Trial versus 3.84.0 l)

Speizer [8] 1989 Harvard Six Cities Study 8427 49 45 2.85 39.9 Quartiles (2.02.6L 12 1.42 (1.07, 1.90)a
versus 2.94.1 l) 2.74 (1.93, 3.90)b

Pooled summary 83,880 1.99 (1.71, 2.29)

Test for heterogeneity, p 0.001.


NA: not available; FEV1: forced expiratory volume in 1 s;RR: relative risk of cardiovascular mortality; CI: confidence interval.
a
Male values.
b
Female values.
c
Although 12,866 participants were originally enrolled in the study, only 7368 participants had an acceptable pulmonary function testing.
d
Although the smoking-adjusted RR was reported, its 95% CI was not provided; thus, the adjusted data could not be used for the meta-analysis.
Cardiovascular Effects 45
predicted) over a 26-year follow-up [5]. In the Honolulu to the highest quintile levels) produced a population attrib-
Heart Program, which prospectively followed 5924 healthy, utable risk of 21% in men and 25% in women for deaths
middle-aged men for up to 18 years, Curb and colleagues related to ischemic heart disease [10] (Fig. 45.1). In certain
found that the individuals in the lowest FEV1 quintile had populations, reduced FEV1 may be responsible for 2030%
a 1.93-fold increase in the risk of cardiovascular mortal- of deaths related to ischemic heart disease [10].
ity compared to those in the best FEV1 quintile (RR, 1.93; The risk of CVD in those with reduced FEV1 is
95% CI, 1.462.54) [6]. In the Tecumseh Cohort Study, amplied by the presence of another risk factor for CVD
Higgins and colleagues observed that individuals with such as hypertension. In the Malmo Born in 1914 Study,
FEV1 values less than 2.0 l had a vefold increase in the risk the risk of cardiac events dened as fatal or nonfatal myo-
for CVD mortality compared with those with FEV1 greater cardial infarction, was only 10% higher in individuals who
than this value (RR, 5.03; 95% CI, 3.078.22) [7]. In the had reduced FEV1 but without hypertension [19]. The
Harvard Six Cities Study, Speizer and colleagues reported a risk nearly tripled (RR, 2.9; 95% CI, 1.84.5) when sub-
RR of 2.74 (95% CI, 1.933.90) in women and 1.42 (95% jects had both reduced FEV1 and hypertension together.
CI, 1.071.90) in men, comparing the lowest FEV1 quar- The incidence of stroke was nearly fourfold higher (RR,
tile to the highest quartile [8]. Schunemann and colleagues 3.8; 95% CI, 1.97.6) when subjects had both reduced
reported a RR of 1.96 (95% CI, 0.993.88) in women and FEV1 and hypertension. In contrast, reduced FEV1 alone
2.11 (95% CI, 1.203.71) in men, once again comparing was not associated with increased risk of stroke (RR, 1.2;
the lowest FEV1 quintile to the highest quintile [9]. Hole 95% CI, 0.702.2) [19]. In the same cohort, Engstrom and
and colleagues reported a RR of 1.88 (95% CI, 1.442.47) colleagues found that individuals who had reduced FEV1
in women and 1.56 (95% CI, 1.261.92) in men, compar- and ventricular dysrhythmia on the baseline electrocardio-
ing the lowest to the highest quintile of FEV1 [10]. Overall, gram (ECG) experienced a twofold increase in the risk of
these large population-based studies showed that reduced coronary events (RR, 2.31; 95% CI, 1.284.20) [20]. In
FEV1 is an important, independent risk factor for CVD contrast, reduced FEV1 alone in the absence of ventricular
morbidity and mortality (pooled RR, 1.99; 95% CI, 1.71 ectopy was not signicantly associated with coronary events
2.29) even among nonsmokers where the RR is 1.67 (95% (RR, 1.24; 95% CI, 0.672.27). Similarly, ventricular ectopy
CI, 1.352.01) [2]. The relationship between reduced FEV1 by itself was not associated with coronary events (RR, 1.16;
and cardiovascular mortality appears to be similar between 95% CI, 0.582.33).
men (RR, 1.64; 95% CI, 1.481.84) and women (RR, 2.14;
95% CI, 1.752.59). In general, when the lowest quintile of
FEV1 is compared with the highest quintile, the risk of car- The relationship between rate of
diovascular mortality increases by ~75% in both men and FEV1 decline and CVD
women [2]. Taken together, these population-based studies
clearly support the notion that CVD risk increases as FEV1 Another important physiologic phenotype associated with
decreases (Table 45.1). COPD is accelerated decline in lung function over time
The population attributable risk of ischemic car- [21]. Individuals who experience rapid decline in FEV1 are
diac deaths imposed by reduced FEV1 was estimated by two times more likely to experience COPD-related hospi-
Hole et al. [10]. They showed that when the lowest quin- talizations and mortality [22]. They are also more likely to
tile of FEV1 was compared with the highest quintile, the experience CVD events. In the Malmo Men Born in 1914
population attributable risk for deaths related to ischemic Study, the cardiovascular event rate among smokers in the
heart disease was 26% (95% CI, 1934%) in men and 24% high, middle, and low thirds with regard to the decline in
(95% CI, 1434%) in women, independent of the eects FEV1 was 56.0, 41.0, and 22.7 events per 1000 person-
of cigarette smoking. The magnitude of the mortality bur- years, respectively (p for trend 0.01) [23]. The Baltimore
den attributed to reduced FEV1 in this model was similar Longitudinal Study of Aging showed that individuals who
to that imposed by hypercholesterolemia. In the same study, experienced the most rapid decline in FEV1 over 16 years
comparison of total serum cholesterol (between the lowest were three to ve times more likely to die from a cardiac

40

35
% of total attributable risk

30

25

20

15

10

0
Smoking Hypercholesterolemia Hypertension Reduced FEV1 FIG. 45.1 Population attributable risk of reduced FEV1 on
(diastolic) 73% versus 108% mortality from ischemic heart disease in men [10].

583
Asthma and COPD: Basic Mechanisms and Clinical Management

cause of death than those who had the slowest decline in study, the leading cause of hospitalization was CVD [3] and
FEV1 [24]. In lifetime nonsmokers, accelerated decline in the leading cause of mortality was lung cancer [28]. CVDs
FEV1 was associated with a 5- to 10-fold increase in the accounted for nearly half of all hospitalizations and 22% of
risk for cardiac deaths. In the US National Study of Coal all deaths, while lung cancer accounted for 33% of all deaths.
Workers Pneumoconiosis, a longitudinal health survey of In contrast, only 8% of the cohort died from respiratory fail-
male underground coal miners, Beeckman and colleagues ure [28]. These data are a sobering reminder of the impor-
found that workers who had an accelerated decline in FEV1 tance of CVD and cancer in patients with mild COPD.
(dened as FEV1 decline of 60 mlyear or greater) were Even in more advanced disease (GOLD Stages 3 and 4),
more likely to develop incident asthma or COPD compared CVD and lung cancer are important causes of morbid-
to individuals with a slower FEV1 decline. Importantly, the ity and mortality. In TORCH (Towards a Revolution in
former group had nearly 50% excess mortality than the lat- COPD Health) study, a 3-year randomized controlled trial
ter group (RR of all-cause mortality, 1.45), driven mostly that assessed the eects of salmeteroluticasone in severe
by excess CVD deaths (RR, 1.90) and pulmonary causes of COPD (FEV1 60% of predicted), CVDs accounted for
mortality (RR, 3.20) [25]. 26% and cancer accounted for 21% of all deaths [29]. In
an analysis of data from the Kaiser Permanente Medical
Group, Sidney and coworkers found that having COPD
The relationship between FEV1 to FVC increased the risk of hospitalization for cardiac arrest and
ratio and CVD ventricular brillation by 2.8 fold, atrial brillation by 2.1
fold, angina by 2 fold, myocardial infarction by 87%, conges-
Reduced FEV1FVC ratio is a more specic indicator of tive heart failure by nearly 4 fold, stroke by 39%, pulmonary
airways disease than is reduced FEV1 alone in the general embolism by 2.7 fold, and other CVD by 86%, independent
population. Using data from the Third National Health of age, sex, and history of hypertension, hyperlipidemia, and
and Nutrition Examination Survey in the United States diabetes [30]. Curkendall and colleagues reported similar
[26], Sin and Man found that individuals with severe air- ndings using a completely dierent cohort [31]. On aver-
ow obstruction (dened as FEV1 50% of predicted and age, for every 10% decrease in FEV1, all-cause mortality
FEV1FVC ratio 70%) were twice as likely to have ECG increases by 14%, cardiovascular mortality increases by 28%,
evidence of probable or possible myocardial infarction than and nonfatal coronary event increased by almost 20%, after
individuals with normal lung function. In the Men Born adjustments for relevant confounders such as age, sex, and
in 1914 Study, Engstrom et al. found that compared to smoking status [3].
subjects in the highest FEV1FVC quintile (ratio 77.3%), In COPD, there are two major phenotypes: emphysema
those in the lowest FEV1FVC (66.3%) quintile were 73% and small airways disease. Emphysema can be determined
more likely to experience coronary events (p 0.01) [20]. noninvasively using quantitative computerized tomography
The risk for frequent or complex ventricular arrhythmia was (CT) scanning [32]. McAllister and coworkers used this
83% higher in the lowest FEV1FVC quintile compared method and found a signicant relationship between emphy-
with the highest quintile. As with FEV1, reduced FEV1 sema severity as assessed by quantitative CT scanning of the
to FVC ratio may have a modifying eect on CVD. In the lungs and arterial stiness as measured by pulse wave velocity
study by Engstrom et al., reduced FEV1FVC ratio by itself [33]. Arterial stiness is a good surrogate of atherosclerosis
was a modest independent risk factor for coronary events and a robust predictor of future CVD risk [34].
(RR, 1.30). Presence of arrhythmias in those with normal
FEV1FVC was not associated with coronary events (RR,
1.01). However, the combination of reduced FEV1FVC The relationship between asthma and CVD
ratio and presence of arrhythmias increased the risk of coro-
nary events by over twofold (RR, 2.43; 95% CI, 1.364.32) Although most of the attention has been directed at the
[20]. These data suggest that airow obstruction impacts relationship between COPD and CVD, there is emerging
synergistically on the diseased heart to make it more vul- literature supporting the notion that asthma is also a risk
nerable to acute coronary events. factor for CVD. In one study from Sweden, Toren and
colleagues estimated the risk of mortality in patients with
asthma. Compared to the expected mortality, the mortality
COPD and CVD rate among patients with severe asthma, dened as one who
needed daily treatment of oral corticosteroids for at least a
The Lung Health Study (LHS) is one of the largest COPD year, was twofold higher (RR, 2.1, 95% CI, 1.82.5) [35].
cohorts that studied the eects of smoking cessation and The RR of ischemic cardiac deaths was also nearly two-
inhaled anticholinergic drugs on disease progression in fold higher (RR, 1.9, 95% CI, 1.42.4) with women hav-
patients with mild COPD (Global Initiative for Chronic ing a particularly high RR at 2.5 (95% CI, 1.73.3) [35]. In
Obstructive Lung Disease, GOLD, Stages 1 and 2) [27]. another population-based study, Musk and colleagues found
Although the initial study was designed as a 5-year trial, that patients with asthma had excess mortality compared
the follow-up was subsequently extended to 14.5 years. to the rest of the population (standardized mortality ratio,
The latter was called Lung Health Study-3 (LHS-3) [28]. SMR, for all causes of death 1.6 for men (p  001) and
In this study, the LHS investigators conrmed the benets 1.7 for women (p  0.001)). The risk of ischemic cardiac
of smoking cessation in retarding disease progression and death was also elevated (SMR, 1.3) but only in the older
attenuating morbidity and mortality [28]. Notably in this age group [36]. A Canadian study found that patients with

584
Cardiovascular Effects 45
self-reported asthma were 43% more likely to have heart process starts in the lungs and then spills into the sys-
disease and 36% more likely to have hypertension com- temic circulation through the rich network of capillary beds
pared to nonasthmatics [37]. Using the Northern California in the pulmonary microcirculation [45]. The rate of spill-
Kaiser Permanente database, a large Health Maintenance age is heavily inuenced by inammation (which generally
Organization providing to over 3 million people, Iribarren increases permeability of the vasculature) and by underly-
and colleagues found that patients with asthma dened ing parenchymal disease. It is believed (though not proven)
based on self-report or hospitalization for asthma had a 24% that in asthma and in COPD, proinammatory cytokines
increase in the risk for coronary heart disease hospitalization and chemokines, such as tumor necrosis factor- (TNF-),
or death adjusted for confounders such as age, raceethnicity, interleukin (IL)-1, IL-6, and others, escape into the sys-
education level, smoking status, white blood cell count, temic circulation from the lungs inducing a systemic
hypertension and diabetes, and occupational exposures. The inammatory response in the liver, bone marrow, and other
relationship was particularly strong in women [38]. organs [46]. In asthma, the inammatory process in the
However, there are some dissenting studies. For lungs is thought to reect an allergic or atopic response
instance, Belli and coworkers failed to nd a signicant asso- to environmental allergens, while in COPD, it is thought to
ciation between asthma and mortality in an elderly group of be an abnormal (exaggerated) response to cigarette smoke
individuals. In this study, the risk of cardiovascular mortal- and other irritants. Systemic inammation is believed to
ity was similar between those with and without asthma contribute to plaque build-up in coronary and cerebral vas-
[39]. Similarly, in the Atherosclerosis Risk In Communities culature and to their rupture during periods of stress [47].
(ARIC) Study, Zureik and colleagues found that compared Consistent with the systemic inammation theory,
with individuals without asthma, those with (ever) asthma CRP levels in COPD patients have been associated with
(dened by self-report) had similar RR of coronary heart CVD and all-cause morbidity and mortality [48]. In LHS,
disease (RR, 0.87; 95% CI, 0.661.14). The RR using a for instance, the risk of all-cause and CVD mortality over
time-dependent analysis was 0.88 (95% CI, 0.691.11). 7 years of follow-up increased as a function of CRP levels.
However, they found that individuals with asthma had Compared to the lowest CRP quintile group (mean level
increased risk of stroke (RR, 1.43; 95% CI, 1.031.98 using 0.21 mgl), the highest CRP quintile (mean level 7.1 mgl)
a time-dependent analysis) [40]. had a mortality risk that was 1.8 times higher, adjusted for
One of the characteristic physiologic signs in sex, race, age, body mass index, pack-years of smoking, bio-
asthma is bronchial hyperresponsiveness (BHR). Zureik chemically validated smoking status (i.e. continued smokers,
and colleagues used data from the European Community sustained quitters, or intermittent quitters), rate of descent
Respiratory Health Survey to determine the relationship in FEV1 (in quintiles), and predicted FEV1 (in quintiles).
between BHR and carotid intima-media thickness, which is The risk of fatal and nonfatal CVD also increased along the
a marker of atherosclerosis [41] and a risk factor for stroke CRP gradient. Compared to the lowest quintile, the highest
[42]. They found that in men but not in women who dem- CRP quintile group experienced a 1.5-fold increase in fatal
onstrated BHR (dened as a 20% fall in baseline FEV1 with and nonfatal cardiovascular events [49].
methacholine exposure of 4 mg or less) the mean carotid
intima-media thickness was greater compared to men who
did not have BHR (0.68  0.11 in men with BHR versus
0.62  0.09 mm in men without BHR, p 0.002) [43]. DISTURBANCE OF NEUROHUMORAL TONE
Although the mechanism for this observation is not com-
pletely known, a recent study indicates that BHR is associ-
ated with systemic inammation, as measured by circulating Patients with severe COPD have a perturbed neurohumoral
C-reactive protein (CRP) [44]. regulatory system, leading to excess sympathetic nerv-
ous activity and reduced vagal tone [50], which is inversely
related to their oxyhemoglobin saturation. Accordingly,
COPD patients have raised resting heart rate and have
POSSIBLE MECHANISMS an increased risk of rhythm disturbances and ectopic
beats [19], which may increase their risk for CVD events.
Metaiodobenzylguanidine (MIBG), an analog of guanethi-
While the epidemiological link between COPD (and to dine, has similar metabolism to norepinephrine in sys-
lesser extent with asthma) and cardiovascular events is solid, temic nervous tissues [51]. MIBG has been used to image
the mechanism(s) that underpin this relationship are far the heart for assessment of cardiac sympathetic activity.
from certain. Increased sympathetic nervous activity leads to reduced
cardiac to mediastinal activity ratio in the delayed images
[51]. In a group of 28 COPD patients and 7 control sub-
The inflammation theory jects, COPD patients demonstrated reduced cardiac accu-
mulation of MIBG and a higher washout rate from the
It is well recognized and accepted that both asthma and heart than control subjects, indicating excess activity of
COPD are inammatory conditions associated with both the sympathetic nervous system with increased norepine-
lung and systemic inammation, though the content and phrine turnover [52]. Interestingly, the MIBG accumulation
the nature of the inammatory process diers between the rate correlated signicantly with the intensity of dyspnea
two conditions [1]. It is postulated that the inammatory as reported by these patients (p  0.05). Consistent with

585
Asthma and COPD: Basic Mechanisms and Clinical Management

the MIBG data, COPD patients had higher plasma nore- cardiovascular events, which included ventricular tachy-
pinephrine levels than did the control subjects (449 versus cardia, myocardial infarction, and sudden deaths [56]. The
69 pgml; p  0.01). There is little information on whether exact mechanisms by which these medications may poten-
or not patients with mild or moderate COPD have dis- tially increase cardiovascular morbidity and mortality are
turbed neurohumoral regulatory system. However, there is unknown. Future studies are needed to validate these initial
emerging data to indicate that in mild disease, patients are epidemiologic observations and to determine the potential
frequently exercise limited by an abnormal cardiovascular mechanisms by which these medications may increase cardi-
response to exercise (e.g. reduced anaerobic threshold). In ovascular risk in susceptible COPD and asthmatic patients.
contrast, subjects without COPD are limited mainly from
leg muscle fatigue [53]. These data suggest that even in mild
COPD, cardiac performance may be perturbed.
Whether certain inhaled drugs commonly used in CAN HEART-PROTECTIVE DRUGS REDUCE
COPD and asthma worsen cardiac status is very contro-
versial. Pharmacoepidemiological studies have linked the COPD MORBIDITY?
use of short- or long-acting bronchodilators with increased
risk of cardiovascular events in COPD and asthma. In one In view of the intrinsic link between COPD and CVD,
study, the use of ipratropium bromide, a short-acting anti- some have suggested that cardioprotective drugs may have
cholinergic medication, was associated with a 60% increase benecial eects in COPD. There have been no randomized
in adjusted mortality compared with nonuse among 827 controlled studies that have carefully and prospectively
patients with COPD [54]. In the LHS in which one-third tested this hypothesis. There are, however, several epide-
of the participants were assigned in a random fashion to miological studies that have generated promising data. For
usual care; another one-third to special intervention (for instance, Mancini and coworkers conducted a time-matched
smoking cessation) in addition to ipratropium bromide; and nested case-control study using administrative health data in
the remaining subjects to special intervention (for smoking Quebec to determine whether or not statins (hydroxymeth-
cessation) in addition to placebo puers, ipratropium bro- ylglutaryl CoA reductase inhibitors), angiotensin-
mide was associated with a 26% increase in the risk of car- converting enzyme (ACE) inhibitors, andor angiotensin
diovascular events compared with placebo. The risk of fatal receptor blockers (ARB) could improve survival in COPD.
cardiovascular events was even higher (relative risk, 2.6). They found that the combined use of statins and ACE or
Although this study was underpowered to detect cardio- ARBs was associated with a 34% reduction in hospitaliza-
vascular events, these data, nonetheless, raised concerns that tion for COPD, a 61% reduction in myocardial infarction,
ipratropium bromide may increase cardiovascular morbidity and a 58% reduction in all-cause mortality [58]. Similar
and mortality in COPD [3]. ndings were noted by an independent group in Japan [59].
There is a scarcity of cardiovascular safety data on tio- One of the major limitations of the previous two stud-
tropium. In the 1-year clinical trials, the risk of heart rate ies was that the diagnosis of COPD was not conrmed by
and rhythm disturbance disorders, such as arrhythmias, spirometry. A Norweigan group using a retrospective cohort
atrial brillation, and tachycardia, were about twofold higher of 854 patients with spirometry conrmed COPD (mean
in those assigned to tiotropium (n 550 patients) com- FEV1, 48% of predicted) demonstrated that treatment with
pared with those assigned to placebo (n 371). However, statins following hospitalization for COPD was associated
these events were rare occurring in only 4.4% of this care- with a 43% reduction in all-cause mortality. Interestingly,
fully selected group of COPD patients [55]. Nevertheless, in the same study, inhaled corticosteroids, which are anti-
tiotropium should be used with caution in patients with inammatory drugs, were also associated with reduced
COPD who also have coexisting rhythm disorders. mortality [60], a nding that has been observed in other
A systematic review and meta-analysis showed that a epidemiological studies [61, 62 ] but not in a randomized
single dose of a 2-agonist caused an increase in heart rate controlled trial [63]. Low-dose inhaled corticosteroid ther-
of 9 beatsmin and reduced serum potassium by 0.36 mmoll apy has been associated with reduced risk of myocardial inf-
compared with placebo [56]. The use of short-acting arction, arrhythmias, and CVD mortality [6466]. Other
2-agonist has been associated with increased cardiovascular groups have shown that beta-blockers may also reduce
events. Au and colleagues, for example, studied 630 patients mortality in COPD patients but the overall magnitude of
with unstable angina or myocardial infarction and 10,486 the benet appears to be small [67]. Moreover, they (espe-
control subjects enrolled in 7 Veterans Administration cially noncardioselective beta-blockers) are associated with
Medical Centers, and found that compared with subjects a slight increase in the risk for COPD hospitalizations [68].
who did not ll a short-acting 2-agonist, patients who had Thus, for safety reasons, in general, only cardioselective beta-
lled one 2-agonist prescription in the 3 months prior to blockers in low doses should be employed in COPD [69].
their index date had 70% increase in the risk for an acute
coronary event [57]. Importantly, the excess risk was lim-
ited to those patients who had a prior history of CVD; their
risk was over threefold higher than those who did not use
2-agonists. Additionally, new users of 2-agonists had a sev-
SUMMARY
enfold increase in the risk of cardiovascular events. Overall,
treatment with short-acting 2-adrenoceptor agonists was Cardiovascular events are common complications in COPD
associated with a 2.5-fold increase in the risk of adverse and asthma. While the mechanisms linking these disorders

586
Cardiovascular Effects 45
have not been well established, the epidemiological evi- 16. Krzyzanowski M, Wysocki M. The relation of thirteen-year mortal-
dence is strong. Reduced lung function (regardless of the ity to ventilatory impairment and other respiratory symptoms: The
cause) increases the risk of cardiovascular hospitalization by Cracow Study. Int J Epidemiol 15(1): 5664, 1986.
17. Hospers JJ, Postma DS, Rijcken B, Weiss ST, Schouten JP. Histamine
twofold may contribute up to 25% of all ischemic cardiac
airway hyper-responsiveness and mortality from chronic obstructive
deaths. In mild to moderate COPD, cardiovascular events pulmonary disease: A cohort study. Lancet 356(9238): 131317, 2000.
are the leading cause of hospitalization and the second lead- 18. Kuller LH, Ockene JK, Townsend M, Browner W, Meilahn E,
ing cause of mortality (trailing only behind lung cancer). Wentworth DN. The epidemiology of pulmonary function and COPD
System inammation and neurohumoral disturbances are mortality in the multiple risk factor intervention trial. Am Rev Respir
the leading putative mechanisms for the association. There Dis 140(3 Pt 2): S7681, 1989.
are emerging data that cardioprotective drugs may also have 19. Engstrom G, Hedblad B, Valind S, Janzon L. Increased incidence of
salutary eects in COPD. However, these promising obser- myocardial infarction and stroke in hypertensive men with reduced
vations must be validated in clinical trials before they can be lung function. J Hypertens 19(2): 295301, 2001.
routinely recommended in patients with obstructive airways 20. Engstrom G, Wollmer P, Hedblad B, Juul-Moller S, Valind S, Janzon L.
Occurrence and prognostic signicance of ventricular arrhythmia
disease.
is related to pulmonary function: A study from men born in 1914,
Malmo, Sweden. Circulation 103(25): 308691, 2001.
21. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P
References et al. Global strategy for the diagnosis, management, and prevention of
chronic obstructive pulmonary disease: GOLD executive summary. Am
1. OByrne PM, Postma DS. The many faces of airway inammation. J Respir Crit Care Med 176(6): 53255, 2007.
Asthma and chronic obstructive pulmonary disease. Asthma Research 22. Mannino DM, Davis KJ. Lung function decline and outcomes in an
Group. Am J Respir Crit Care Med 159(5 Pt 2): S4163, 1999. elderly population. Thorax 61(6): 47277, 2006.
2. Sin DD, Wu L, Man SF. The relationship between reduced lung func- 23. Engstrom G, Hedblad B, Janzon L, Valind S. Respiratory decline in
tion and cardiovascular mortality: A population-based study and a sys- smokers and ex-smokers an independent risk factor for cardiovascu-
tematic review of the literature. Chest 127(6): 195259, 2005. lar disease and death. J Cardiovasc Risk 7(4): 26772, 2000.
3. Anthonisen NR, Connett JE, Enright PL, Manfreda J. Hospitalizations 24. Tockman MS, Pearson JD, Fleg JL, Metter EJ, Kao SY, Rampal KG et al.
and mortality in the Lung Health Study. Am J Respir Crit Care Med Rapid decline in FEV1. A new risk factor for coronary heart disease
166(3): 33339, 2002. mortalityIVI. Am J Respir Crit Care Med 151(2 Pt 1): 39098, 1995.
4. Stavem K, Aaser E, Sandvik L, Bjornholt JV, Erikssen G, Thaulow E 25. Beeckman LA, Wang ML, Petsonk EL, Wagner GR. Rapid declines in
et al. Lung function, smoking and mortality in a 26-year follow-up of FEV1 and subsequent respiratory symptoms, illnesses, and mortality in
healthy middle-aged males. Eur Respir J 25(4): 61825, 2005. coal miners in the United States. Am J Respir Crit Care Med 163176(3
5. Schikowski T, Sugiri D, Ranft U, Gehring U, Heinrich J, Wichmann Pt 112): 633120891214, 2001.
HE et al. Does respiratory health contribute to the eects of long-term 26. Sin DD, Man SF. Why are patients with chronic obstructive pulmo-
air pollution exposure on cardiovascular mortality? Respir Res 8(1): 20, nary disease at increased risk of cardiovascular diseases? The potential
2007. role of systemic inammation in chronic obstructive pulmonary dis-
6. Curb JD, Marcus EB, Reed DM, MacLean C, Yano K. Smoking, pul- ease. Circulation 107(11): 151419, 2003.
monary function, and mortality. Ann Epidemiol 1(1): 2532, 1990. 27. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey WC, Buist AS
7. Higgins MW, Keller JB. Predictors of mortality in the adult population et al. Eects of smoking intervention and the use of an inhaled anti-
of Tecumseh. Arch Environ Health 21(3): 41824, 1970. cholinergic bronchodilator on the rate of decline of FEV1. The Lung
8. Speizer FE, Fay ME, Dockery DW, Ferris BG Jr. Chronic obstructive Health Study. JAMA 272(19): 1497505, 1994.
pulmonary disease mortality in six U.S. cities. Am Rev Respir Dis 140(3 28. Anthonisen NR, Skeans MA, Wise RA, Manfreda J, Kanner RE,
Pt 2): S4955, 1989. Connett JE. The eects of a smoking cessation intervention on 14.5-
9. Schunemann HJ, Dorn J, Grant BJ, Winkelstein W Jr, Trevisan M. year mortality: A randomized clinical trial. Ann Intern Med 142(4):
Pulmonary function is a long-term predictor of mortality in the gen- 23339, 2005.
eral population: 29-year follow-up of the Bualo Health Study. Chest 29. McGarvey LP, John M, Anderson JA, Zvarich M, Wise RA.
118(3): 65664, 2000. Ascertainment of cause-specic mortality in COPD: Operations of the
10. Hole DJ, Watt GC, Davey-Smith G, Hart CL, Gillis CR, Hawthorne TORCH Clinical Endpoint Committee. Thorax 62(5): 41115, 2007.
VM. Impaired lung function and mortality risk in men and women: 30. Sidney S, Sorel M, Quesenberry CP Jr, DeLuise C, Lanes S, Eisner MD.
Findings from the Renfrew and Paisley prospective population study. COPD and incident cardiovascular disease hospitalizations and mor-
BMJ 313(7059): 71115, discussion 56, 1996. tality: Kaiser Permanente Medical Care Program. Chest 128(4): 2068
11. Marcus EB, Curb JD, MacLean CJ, Reed DM, Yano K. Pulmonary 75, 2005.
function as a predictor of coronary heart disease. Am J Epidemiol 31. Curkendall SM, DeLuise C, Jones JK, Lanes S, Stang MR, Goehring E
129(1): 97104, 1989. Jr et al. Cardiovascular disease in patients with chronic obstructive
12. Beaty TH, Newill CA, Cohen BH, Tockman MS, Bryant SH, pulmonary disease, Saskatchewan Canada cardiovascular disease in
Spurgeon HA. Eects of pulmonary function on mortality. J Chronic COPD patientsM. Ann Epidemiol 16(1): 6370, 2006.
Dis 38(8): 70310, 1985. 32. Muller NL, Coxson H. Chronic obstructive pulmonary disease. IV:
13. Ebi-Kryston KL. Respiratory symptoms and pulmonary function as Imaging the lungsT in patients with chronic obstructive pulmonary
predictors of 10-year mortality from respiratory disease, cardiovascular disease. Thorax 57(11): 98285, 2002.
disease, and all causes in the Whitehall Study. J Clin Epidemiol 41(3): 33. McAllister DA, Maclay JD, Mills NL, Mair G, Miller J, Anderson D
25160, 1988. et al. Arterial stiness is independently associated with emphysema
14. Lange P, Nyboe J, Jensen G, Schnohr P, Appleyard M. Ventilatory severity in patients with COPD. Am J Respir Crit Care Med 176(12):
function impairment and risk of cardiovascular death and of fatal or 120814, 2007.
non-fatal myocardial infarction. Eur Respir J 4(9): 108087, 1991. 34. Willum-Hansen T, Staessen JA, Torp-Pedersen C, Rasmussen S, Thijs L,
15. Tockman MS, Comstock GW. Respiratory risk factors and mortality: Ibsen H et al. Prognostic value of aortic pulse wave velocity as index of
Longitudinal studies in Washington County, Maryland. Am Rev Respir arterial stiness in the general population. Circulation 113(5): 66470,
Dis 140(3 Pt 2): S5663, 1989. 2006.

587
Asthma and COPD: Basic Mechanisms and Clinical Management

35. Toren K, Lindholm NB. Do patients with severe asthma run an 53. Or D, Laveneziana P, Webb KA, Lam YM, ODonnell DE.
increased risk from ischaemic heart disease? Int J Epidemiol 25(3): Mechanisms of dyspnea during cycle exercise in symptomatic patients
61720, 1996. with GOLD Stage I COPD. Am J Respir Crit Care Med 177: 62229,
36. Musk AW, Ryan GF, Perera DM, DSouza BP, Hockey RL, Hobbs 2007.
MS. Mortality from asthma in Western Australia. Med J Aust 147(9): 54. Ringbaek T, Viskum K. Is there any association between inhaled iprat-
42327, 1987. ropium and mortality in patients with COPD and asthma? Respir Med
37. Dogra S, Ardern CI, Baker J. The relationship between age of asthma 97(3): 26472, 2003.
onset and cardiovascular disease in Canadians. J Asthma 44(10): 849 55. Committee FaDAP-ADA. Spiriva (tiotropium bromide) Inhalation
54, 2007. Powder, NDA 21-395, Boehringer Ingelheim Pharmaceuticals.
38. Iribarren C, Tolstykh IV, Eisner MD. Are patients with asthma at September 6, 2002.
increased risk of coronary heart disease? Int J Epidemiol 33(4): 74348, 56. Salpeter SR, Ormiston TM, Salpeter EE. Cardiovascular eects of
2004. beta-agonists in patients with asthma and COPD: A meta-analysis.
39. Bellia V, Pedone C, Catalano F, Zito A, Davi E, Palange S et al. Chest 125(6): 230921, 2004.
Asthma in the elderly: Mortality rate and associated risk factors for 57. Au DH, Curtis JR, Every NR, McDonell MB, Fihn SD. Association
mortality. Chest 132(4): 117582, 2007. between inhaled beta-agonists and the risk of unstable angina and
40. Schanen JG, Iribarren C, Shahar E, Punjabi NM, Rich SS, Sorlie PD myocardial infarction. Chest 121(3): 84651, 2002.
et al. Asthma and incident cardiovascular disease: The Atherosclerosis 58. Mancini GB, Etminan M, Zhang B, Levesque LE, FitzGerald JM,
Risk in Communities Study. Thorax 60(8): 63338, 2005. Brophy JM. Reduction of morbidity and mortality by statins,
41. Touboul PJ, Labreuche J, Vicaut E, Amarenco P. Carotid intima-media angiotensin-converting enzyme inhibitors, and angiotensin receptor
thickness, plaques, and Framingham risk score as independent deter- blockers in patients with chronic obstructive pulmonary disease. J Am
minants of stroke risk. Stroke 36(8): 174145, 2005. Coll Cardiol 47(12): 255460, 2006.
42. Tsivgoulis G, Vemmos K, Papamichael C, Spengos K, Manios E, 59. Ishida W, Kajiwara T, Ishii M, Fujiwara F, Taneichi H, Takebe N
Stamatelopoulos K et al. Common carotid artery intima-media thick- et al. Decrease in mortality rate of chronic obstructive pulmonary dis-
ness and the risk of stroke recurrence. Stroke 37(7): 191316, 2006. ease (COPD) with statin use: A population-based analysis in Japan.
43. Zureik M, Kony S, Neukirch C, Courbon D, Leynaert B, Vervloet D Tohoku J Exp Med 212(3): 26573, 2007.
et al. Bronchial hyperresponsiveness to methacholine is associated with 60. Soyseth V, Brekke PH, Smith P, Omland T. Statin use is
increased common carotid intima-media thickness in men. Arterioscler associated with reduced mortality in COPD. Eur Respir J 29(2): 27983,
Thromb Vasc Biol 24(6): 1098103, 2004. 2007.
44. Kony S, Zureik M, Driss F, Neukirch C, Leynaert B, Neukirch F. 61. Sin DD, Tu JV. Inhaled corticosteroids and the risk of mortality and
Association of bronchial hyperresponsiveness and lung function with readmission in elderly patients with chronic obstructive pulmonary
C-reactive protein (CRP): A population based study. Thorax 59(10): disease. Am J Respir Crit Care Med 164(4): 58084, 2001.
89296, 2004. 62. Soriano JB, Kiri VA, Pride NB, Vestbo J. Inhaled corticosteroids
45. Sin DD, Man SF. Systemic inammation and mortality in chronic withwithout long-acting beta-agonists reduce the risk of rehospi-
obstructive pulmonary disease. Can J Physiol Pharmacol 85177(1): talization and death in COPD patients. Am J Respir Med 2(1): 6774,
1416227629, 2007. 2003.
46. Mannino DM, Watt G, Hole D, Gillis C, Hart C, McConnachie A 63. Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW
et al. The natural history of chronic obstructive pulmonary disease. et al. Salmeterol and uticasone propionate and survival in chronic
Eur Respir J 27(3): 62743, 2006. obstructive pulmonary disease. N Engl J Med 356(8): 77589, 2007.
47. Ross R. Atherosclerosis an inammatory disease. N Engl J Med 64. Huiart L, Ernst P, Ranouil X, Suissa S. Low-dose inhaled corticoster-
340(2): 11526, 1999. oids and the risk of acute myocardial infarction in COPD. Eur Respir J
48. Stork S, Feelders RA, van den Beld AW, Steyerberg EW, Savelkoul 25(4): 63439, 2005.
HF, Lamberts SW et al. Prediction of mortality risk in the elderlyA. 65. Lofdahl CG, Postma DS, Pride NB, Boe J, Thoren A. Possible protec-
Am J Med 119(6): 51925, 2006. tion by inhaled budesonide against ischaemic cardiac events in mild
49. Man SF, Connett JE, Anthonisen NR, Wise RA, Tashkin DP, Sin DD. COPD. Eur Respir J 29(6): 111519, 2007.
C-reactive protein and mortality in mild to moderate chronic obstruc- 66. Macie C, Wooldrage K, Manfreda J, Anthonisen NR. Inhaled corti-
tive pulmonary disease. Thorax 61(10): 84953, 2006. costeroids and mortalityL in COPD. Chest 130(3): 64046, 2006.
50. Heindl S, Lehnert M, Criee CP, Hasenfuss G, Andreas S. Marked 67. Chen J, Radford MJ, Wang Y, Marciniak TA, Krumholz HM.
sympathetic activation in patients with chronic respiratory failure. Am J Eectiveness of beta-blocker therapy after acute myocardial infarc-
Respir Crit Care Med 164(4): 597601, 2001. tion in elderly patients with chronic obstructive pulmonary disease or
51. Wieland DM, Wu J, Brown LE, Mangner TJ, Swanson DP, asthma. J Am Coll Cardiol 37(7): 195056, 2001.
Beierwaltes WH. Radiolabeled adrenergi neuron-blocking agents: 68. Brooks TW, Creekmore FM, Young DC, Asche CV, Oberg B,
Adrenomedullary imaging with [131I]iodobenzylguanidine. J Nucl Samuelson WM. Rates of hospitalizations and emergency department
Med 21(4): 34953, 1980. visits in patients with asthma and chronic obstructive pulmonary dis-
52. Sakamaki F, Satoh T, Nagaya N, Kyotani S, Nakanishi N, Ishida Y. ease taking beta-blockersA. Pharmacotherapy 27(5): 68490, 2007.
Abnormality of left ventricular sympathetic nervous function assessed 69. Salpeter S, Ormiston T, Salpeter E. Cardioselective beta-blockers for
by (123)I-metaiodobenzylguanidine imaging in patients with COPD. chronic obstructive pulmonary disease. Cochrane Database Syst Rev(4),
Chest 116(6): 157581, 1999. 2005. CD003566

588
Allergen Avoidance
46CHAPTER

levels of microbial products such as endotoxin


INTRODUCTION and glucan. This may have unanticipated conse- Ashley Woodcock and
quences and may alter the eectiveness of aller- Adnan Custovic
Senitization to inhalant allergens is a major risk fac- gen avoidance on allergic disease.
tor for asthma, rhinitis, and eczema [1, 2]. Exposure When designing eective methods to University of Manchester,
of individuals with established allergic disease to reduce personal exposure, the aerodynamic char- Manchester, UK
high levels of sensitizing allergens causes exacerba- acteristics and distribution of allergens have to
tion of symptoms and worsening of the underlying be taken into account [16]. Mite and cockroach
inammatory process [310]. Complete cessation allergens are contained within particles 10 m
of exposure leads to the improvement in disease diameter and become airborne after vigorous
control. For example, patients with seasonal allergic cleaning, whereas substantial cat and dog aller-
rhinitis have no symptoms in the absence of expo- gen circulates in the air on small particles (5 m
sure to pollen, and moving atopic asthmatics into diameter) [1719]. Consequently, pet allergic
low-allergen environments such as hospitals [11] or patients often develop wheezing, sneezing, or
high-altitude sanatoria [1214] improves markers itchy eyes immediately on entering a home with
of asthma severity. a pet, but mite and cockroach-sensitized asth-
In occupational asthma, cessation of matics are unaware of the relationship between
exposure to an allergen may be associated with a exposure and symptoms. Air ltration units have
dramatic improvement in symptoms, and some- little eect on personal exposure to mite or cock-
times cure, but only early in the natural history roach but may be useful in removing airborne pet
of the disease [15]. However, exposure over a allergens.
longer period can result in asthma becoming
persistent in spite of removal from exposure.
This early window of opportunity appears var- Mite allergen avoidance
iable between individuals and may explain why
environmental control is ineective in some cir- Reduction of mites and mite allergens in the
cumstances. It suggests that any allergen avoid- home can be achieved by a number of measures
ance strategy must achieve an early and major (Table 46.1; for review see Ref. [16]).
reduction in exposure and be targeted to indi-
viduals likely to benet. Bed and bedding
The greatest exposure to mite allergen is in bed at
night, and synthetic quilts provide much greater
risk than feather cover does. The most eec-
ALLERGEN AVOIDANCE MEASURES tive mite allergen avoidance measure is to cover
the mattress, duvet, and pillows with imperme-
able encasings (or get rid of the quilt if it is not
Allergen avoidance measures are usually not spe- covered). Finely woven fabrics are the most eec-
cic to any given allergen, or even environmen- tive covers [20]. Sheets should be washed weekly,
tal contaminant. For example, measures against and blankets should be washed regularly in a hot
house dust mite allergen will also impact on cycle (above 55C) to kill the mites [21].

589
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 46.1 Practical measures for reducing house dust mite, pet, fungal, 50
and cockroach allergen levels. p 0.01

House dust mite 40

Bed and bedding


Encase mattress, pillow, and quilt in allergen impermeable covers 30

Counts
(preferably finely woven fabric)
Wash all bedding weekly. Use hot cycle (5560C) if possible
Replace carpets with hard flooring (e.g., linoleum or wood) 20
Minimize upholstered furniture/replace with leather furniture
Replace curtains with blinds
10
Minimize dust accumulating objects; keep in closed cupboards
Remove soft toys (if impossible, hot wash/freeze soft toys)
Reduce indoor humidity if possible 0
Baseline 1 2 3
Cat/dog Time

Remove cat/dog from the home FIG. 46.1 Air filtration with HEPA units results in only moderate reduction
in personal exposure to cat allergen (assessed by intranasal air filters). From
Ref. [28] with permission.
Fungi

Reduce indoor humidity if possible


Use HEPA air filters in main living areas and bedrooms where outdoor humidity is high and homes are poorly insu-
Use Fungicides on heavily contaminated surfaces lated. Portable dehumidiers are generally inadequate [24].
Minimize upholstered furniture Major reductions in personal exposure are possible,
Replace carpets with hard flooring (e.g., linoleum or wood) but single measures are unlikely to work. A stringent envi-
Ensure regular inspection of heating and air conditioning units to ronmental control regime combining several measures can
prevent contamination achieve and maintain a low-allergen environment [25], but
this will be expensive and require long-term discipline
Cockroaches
this may be beyond many patients.

Remove food and water sources


Use suitable pesticide in bait form Pet allergen avoidance
Remove all dead carcasses and frass
Wash down all surfaces, floors, and walls with detergent Cat and dog allergen levels are low in homes without a
Seal cracks in walls and plaster work to reduce further access pet [16]. After permanent removal of a pet, allergen lev-
Wash all bedding, clothing, and curtains els remain high for months [26]. Pet-sensitized children
exposed to pets may lose lung function in early life which
may never be recovered [3]. Almost all pet owning and pet-
sensitized asthmatics continue keeping their pets, in spite of
Carpets and upholstered furnishings asthma which is hard to control.
Air cleaning units with high-eciency particulate
Fitted carpets should be removed and replaced with hard arrest (HEPA) lters can reduce the airborne concentra-
oor coverings (e.g., wooden or linoleum oor) since they tion of cat and dog allergens in homes with pets [27], but
are impossible to clean (vacuuming provides substantial per- eld studies demonstrated that the magnitude of reduction
sonal exposure). Exposing carpets to direct sunlight, steam in personal exposure is small (Fig. 46.1) [28]. Washing pets
cleaning, use of acaricides or tannic acid, freezing with liq- produces a modest reduction in airborne allergen [29, 30]
uid nitrogen, etc. are only partially eective [21]. Allergen but needs to be done more than twice a week [31] and is
reduction in upholstered furniture is impossible, without unlikely to translate into clinical benet.
impermeable (leather or PVC) covers. Fabric curtains can In experimental chambers, vacuum cleaners with built-
be replaced with venetian blinds. Soft toys can be frozen in HEPA lters and double thickness bags do not leak pet
then washed to kill mites and remove allergens. allergens. However, in real life (actually vacuuming a car-
pet for which they were designed!) intranasal air samplers
Controlling humidity have shown a vefold increase in the amount of cat allergen
Mites live in beds because of the high levels of humidity; inhaled while using a range of brand new high-eciency
but reducing relative humidity of indoor air may be insuf- vacuum cleaners (Fig. 46.2) [32]. Data from experimental
cient to reduce humidity in the middle of a mattress [22, chamber studies are not a relevant index of personal exposure
23]. This depends on the local climate, housing design, and in a real-life situation and thus cannot be used to recommend
eectiveness of dehumidication. Central mechanical ven- high-eciency vacuum cleaners to allergy suerers.
tilation heat recovery units are eective in areas where out- Pet removal is the only eective advice to patients
door humidity is low and home insulation is good, but not with pet allergy who experience symptoms on exposure.

590
Allergen Avoidance 46
(A) (B)
140 1.5
Median Median

120

100

1.0
80
Counts (ng/ml)

Counts (ng/ml)
60

40
0.5

20

0 2 2
10 10 10 10 10 10 10 10
0.0
Old,Off Old,On New,Off New,On Old,Off Old,On New,Off New,On

FIG. 46.2 Vacuum cleaning and cat allergen exposure: Fel d 1 bearing particle counts (halo units; (A) and Fel d 1 level (ELISA; (B). Vacuum cleaners are
grouped into either control (old) or unused (new). Each value is the median (range) of the observations in each cell (2 observers 10 homes). There is a
fivefold increase in the amount of cat allergen inhaled while using a range of brand new high-efficiency vacuum cleaners. From Ref. [32] with permission.

Cockroach allergen reduction methods of mite allergen avoidance should not be recom-
mended to mite-sensitive asthmatics [37, 38]. The authors
Physical and chemical procedures can control cockroach concluded that they were unable to demonstrate any clini-
populations in infested houses [3335]. Several pesticides cal benet to mite-sensitive patients with asthma of meas-
are available, in either a gel or a bait form. Elimination of ures designed to reduce mite exposure (Figs 46.3 and 46.4).
food and water sources, sealing holes in plasterwork and The most recent meta-analysis [37] included 3002 patients
oors, and household cleaning (of contaminated oors, included into 54 trials, which is more than doubling the
surfaces, and bedding) are essential. Successful treatments number of patients since the rst such review [39]. The
reduce exposure in 2 weeks, have maximal eect within authors suggested that allergen avoidance methods did not
1 month, and reduce populations for up to 6 months. adequately reduce mite antigen levels but also stressed that
Discipline in cleaning and repeat treatments are critical. mite-sensitive asthmatic patients are usually sensitized to
other allergens, so that successful elimination of only one
allergen may have limited benet, whatever its success [37].
Recent Cochrane review of dehumidication showed no
satisfactory studies.
ALLERGEN AVOIDANCE IN THE TREATMENT OF In adults with asthma, three older studies (98 study
ALLERGIC DISEASE participants in total) suggested improvements in bron-
chial reactivity with mite allergen avoidance in mite-
sensitive patients [4042]. In contrast, the largest randomized
Vulnerable patients are bombarded with advertisements for double-blind, placebo-controlled SMAC trial assessed the
allergen avoidance, often with spurious or exaggerated claims eectiveness of mite-impermeable bed covers as a single
for extremely expensive vacuum cleaners, dehumidiers, etc. intervention, in over 1000 adult asthmatics. There was no
We have a duty to only recommend clinically eective con- improvement in any of the outcome measures [morning
trols, which are relevant to specic patients, often relevant to peak expiratory ow rate, PEFR, during the rst 6 months
their geographical location. The questions that remain contro- or 12 months (Fig. 46.5), the proportion of patients who
versial are whether home environmental changes are ade- are able to discontinue inhaled steroids during the second
quate, can be sustained in real life sucient to impact disease, 6 months of the study, symptoms scores, quality of life, etc.]
and how to identify those patients who will benet [36]. [43]. Over two-thirds had positive mite-specic IgE and
one in four beds had mite allergen levels 10 g/g Der p 1.
In a post-hoc analysis of the subgroup of 130 patients with
Mite allergen avoidance high mite-specic IgE (10 kUA/l) and high mite allergen
exposure (10 g/g Der p 1 in mattress dust), there was
Recent updates of the Cochrane meta-analysis reported no dierence between active and placebo groups. This was
no eect of the interventions and concluded that current conrmed by a study in 55 adult patients with asthma who

591
Asthma and COPD: Basic Mechanisms and Clinical Management

Review: House dust mite control measures for asthma


Comparison: 01 House dust mite reduction versus control
Outcome: 02 Asthma symptoms score
Study Treatment Control Standardized mean difference (fixed) Weight Standardized mean difference
N Mean (SD) N Mean (SD) 95% CI (%) (fixed) 95% CI
01 Chemical methods
Bahir (1997) 13 1.60 (1.50) 17 1.40 (1.50) 2.2 0.13 (0.59, 0.85)
Chang (1996) 12 1.10 (1.70) 14 0.40 (0.50) 1.8 0.56 (0.23, 1.35)
Dietemann (1993) 11 1.40 (1.24) 12 1.18 (0.36) 1.7 0.24 (0.58, 1.06)
Reiser (1990) 23 5.50 (4.30) 23 3.30 (3.50) 3.3 0.55 (0.04, 1.14)
Subtotal (95% CI) 59 66 9.0 0.39 (0.04, 0.75)
2
Test for heterogeneity chi-square 1.10 df 3 p 0.78 I 0.0%
Test for overall effect z 2.16 p 0.03

02 Physical methods parallel group studies


Chen (1996) 20 0.50 (0.66) 15 0.82 (1.01) 2.5 0.38 (1.05, 0.30)
Cinti (1996) 10 0.30 (0.68) 10 1.00 (1.15) 1.4 0.71 (1.62, 0.20)
Dharmage (2006) 15 0.02 (0.15) 15 0.04 (0.17) 2.2 0.12 (0.60, 0.84)
Fang (2001) 22 10.00 (6.80) 21 17.30 (10.30) 2.9 0.83 (1.45, 0.20)
Huss (1992) 26 8.80 (10.70) 26 13.10 (11.20) 3.8 0.39 (0.94, 0.16)
Rijssenbeek (2002) 16 2.25 (2.24) 14 2.37 (3.17) 2.2 0.04 (0.76, 0.67)
Sheikh (2002) 23 3.40 (29.50) 20 18.10 (27.80) 3.1 0.50 (0.11, 1.11)
Thiam (1999) 18 0.80 (0.50) 6 1.80 (0.18) 0.9 2.16 (3.30, 1.02)
Woodcock (2003) 315 1.03 (0.70) 310 1.03 (0.73) 46.2 0.0 (0.16, 0.16)
de Vries (2007) 48 1.23 (0.86) 48 1.13 (0.83) 7.1 0.12 (0.28, 0.52)
Subtotal (95% CI) 513 485 72.2 0.07 (0.20, 0.05)
2
Test for heterogeneity chi-square 27.72 df 9 p 0.001 I 67.5%
Test for overall effect z 1.11 p 0.3

03 Pyhsical methods crossover studies


Antonicelli (1991) 9 0.16 (0.32) 9 0.26 (0.34) 1.3 0.29 (1.22, 0.64)
Warner (1993) 14 0.20 (0.26) 14 0.19 (0.34) 2.1 0.03 (0.71, 0.77)
Zwemer (1973) 12 0.70 (0.51) 12 1.40 (0.43) 1.4 1.43 (2.35, 0.52)
Subtotal (95% CI) 35 35 4.7 0.48 (0.97, 0.01)
2
Test for heterogeneity chi-square 6.17 df 2 p 0.05 I 67.6%
Test for overall effect z 1.91 p 0.06

04 Combination methods
Cloosterman (1999) 76 5.50 (6.10) 81 6.30 (6.70) 11.6 0.12 (0.44, 0.19)
Marks (1994) 17 0.98 (0.57) 18 0.67 (0.55) 2.5 0.54 (0.14, 1.22)
Subtotal (95% CI) 93 99 14.1 0.01 (0.29, 0.28)
Test for heterogeneity chi-square 3.06 df 1 p 0.08 I 2 67.3%
Test for overall effect z 0.05 p 1

Total (95% CI) 700 685 100.0 0.04 (0.15, 0.07)


2
Test for heterogeneity chi-square 47.02 df 18 p 0.0002 I 61.7%
Test for overall effect z 0.73 p 0.5

4.0 2.0 0 2.0 4.0


Favors treatment Favors control

FIG. 46.3 Cochrane meta-analysis reported no effect of the interventions. House dust mite reduction versus control, outcome: asthma symptoms score.

were sensitized to mite (mite-specic IgE  0.7 kUA/l) and seven inner city areas of the USA. Mattress and pillow encas-
exposed to high levels of dust mite allergen in their mat- ings and a high-ltration vacuum cleaner were supplied to all
tresses (Der p 1  2 g/g) [44]. Recent Dutch studies have homes, and products required for specic interventions (e.g., a
given opposing results [45]. In asthma in adults, a single HEPA air lter for the reduction in passive smoke exposure)
intervention with allergen-impermeable encasings for the were supplied free of charge (the control group had no pla-
mattress, duvet, and pillows is ineective in long-term man- cebo interventions). Children in the intervention group had
agement, even in individuals who are highly allergic to dust signicantly fewer days with asthma symptoms, and fewer
mite and exposed to high levels of mite allergens. emergency room visits [50] (Fig. 46.5). An additional period
In contrast, several studies in children with asthma have of 34 symptom-free days over 2 years was obtained at a cost of
indicated that environmental interventions may improve air- $2000/child (Fig. 46.6) [51].
way reactivity [46], lung function [47], reduce acute Emergency
Room visits [48], and substantially reduce maintenance inhaled Rhinitis
steroids [49]. The Inner-City Asthma Study assessed individu- A Cochrane systematic review of mite avoidance measures
ally tailored environmental interventions [50]. A total of 937 in the management of perennial allergic rhinitis published
children aged 511 years with uncontrolled moderate/severe in 2001 reported that the quality of studies was poor and
asthma and at least one positive skin test were recruited from found no evidence of benet [52]. In the very large SMAC

592
Allergen Avoidance 46
Review: House dust mite control measures for asthma
Comparison: 01 House dust mite reduction versus control
Outcome: 07 PC20 (provocative concentration for 20% fall in FEV1)
Study Treatment Control Standardized mean difference (fixed) Weight Standardized mean difference
N Mean (SD) N Mean (SD) 95% CI (%) (fixed) 95% CI
01 Chemical methods
Chang (1996) 12 0.87 (2.29) 14 0.82 (3.84) 5.4 0.02 (0.76, 0.79)
Ehnert (1992) 7 0.57 (0.51) 4 0.29 (0.55) 2.0 0.49 (1.74, 0.77)
Reiser (1990) 23 0.48 (1.13) 23 0.70 (1.60) 9.5 0.16 (0.74, 0.42)
Sette (1994) 14 0.50 (0.49) 10 0.47 (0.56) 4.8 0.06 (0.76, 0.87)
van der Heide (1997a) 21 1.75 (1.60) 19 1.95 (2.09) 8.3 0.11 (0.73, 0.51)
Subtotal (95% CI) 77 70 30.0 0.10 (0.43, 0.23)
2
Test for heterogeneity chi-square 0.63 df 4 p 0.96 I 0.0%
Test for overall effect z 0.60 p 0.5

02 Physical methods parallel group studies


Dharmage (2006) 15 1.70 (1.00) 15 2.10 (1.10) 6.1 0.37 (0.35, 1.09)
Halken (2003) 26 3.84 (0.70) 21 3.89 (0.64) 9.6 0.07 (0.65, 0.50)
Htut (2001) 15 1.20 (1.10) 8 1.60 (1.10) 4.3 0.35 (0.51, 1.22)
Rijssenbeek (2002) 16 0.28 (0.29) 14 0.33 (0.34) 6.2 0.15 (0.87, 0.56)
Subtotal (95% CI) 72 58 26.2 0.08 (0.27, 0.43)
2
Test for heterogeneity chi-square 1.68 df 3 p 0.64 I 0.0%
Test for overall effect z 0.45 p 0.7

03 Physical methods crossover studies


Antonicelli (1991) 9 2.04 (0.48) 9 2.09 (0.28) 3.7 0.12 (1.05, 0.80)
Subtotal (95% CI) 9 9 3.7 0.12 (1.05, 0.80)
Test for heterogeneity not applicable
Test for overall effect z 0.26 p 0.8

04 Combination methods
Cloosterman (1999) 63 0.08 (2.29) 72 0.37 (3.10) 27.8 0.16 (0.18, 0.50)
Dorward (1988) 9 0.36 (0.63) 9 0.08 (0.56) 3.6 0.45 (0.49, 1.39)
Ehnert (1992) 7 0.07 (0.26) 3 0.29 (0.55) 1.5 0.91 (0.53, 2.36)
Marks (1994) 17 0.16 (0.91) 18 0.03 (0.78) 7.2 0.22 (0.88, 0.45)
Subtotal (95% CI) 96 102 40.1 0.15 (0.13, 0.43)
2
Test for heterogeneity chi-square 2.65 df 3 p 0.45 I 0.0%
Test for overall effect z 1.03 p 0.3

Total (95% CI) 254 239 100.0 0.05 (0.13, 0.22)


2
Test for heterogeneity chi-square 6.40 df 13 p 0.93 I 0.0%
Test for overall effect z 0.50 p 0.6

4.0 2.0 0 2.0 4.0


Favors control Favors treatment

FIG. 46.4 Cochrane meta-analysis reported no effect of the interventions. House dust mite reduction versus control, outcome PC20 (provocative
concentration for 20% fall in FEV1).

(A) 450 All patients (B) 450 Mite-sensitive patients

440 440 Control


Control group
Peak expiratory flow rate
Peak expiratory flow rate

430 group 430

420 420 Active-


intervention
(l/min)
(l/min)

410 410 group


Active-
intervention
400 group 400

390 390

380 380

0 0
Baseline 6 month 12 month Baseline 6 month 12 month

FIG. 46.5 No effect of covering mattress, pillow, and quilt with allergen-impermeable covers amongst adults with asthma. Mean morning peak expiratory
flow rate in the activeintervention and control groups at base line, 6 months, and 12 months among all patients (A) and among mite-sensitive patients (B).
From Ref. [43] with permission.

593
Asthma and COPD: Basic Mechanisms and Clinical Management

7 treatment. Some controversy still exists, however, regard-


Maximal number of days with ing the role of exclusion diets in patients with combined
symptoms (number/2 weeks) 6
eczema and food sensitivity. The few small studies on this
5 issue, most investigating the impact of an elemental diet,
showed no or limited benet [60, 61].
4 Control
3
Summary on the use of allergen avoidance
Intervention
2 in the treatment of asthma
1
There is no evidence to support the use of simple single
0 allergen avoidance methods amongst adults with asthma;
with respect to the eects of a multifaceted intervention,
M lin e

M h2

M h4

M h6

on 8

on 0

on 2

on 4

on 6

on 8

on 0

on 2
24
M h1

M h1

M h1

M h1

M h1

M h2

M h2
M th
t

th
se
on

on

on

on

there are as yet no adequately powered studies based upon


t

t
Ba

which to make an informed decision in this age group. In


Intervention year Follow-up year contrast, the majority of studies in children suggest that
FIG. 46.6 Environmental control is effective amongst children with allergen avoidance may be of benet.
asthma. Mean maximal number of days with symptoms for every 2-week Until unequivocal evidence for all age groups is
period before a follow-up assessment during the 2 years of the study. The obtained, we suggest the following pragmatic approach:
difference between the environmental intervention and control group was
significant in both the intervention year (p  0.001) and the follow-up year
Single avoidance measures ineective
(p  0.001). From Ref. [50] with permission. use a comprehensive environmental control regime
aiming to achieve a complete cessation of exposure, or
as great a reduction in personal exposure as possible
study [40], there was no dierence in rhinitis symptoms
between active bedding and control groups (data unpub- Tailor the intervention to the patients sensitization and
lished). A further large randomized, double-blind, placebo- exposure status
controlled study has since conrmed this [53]. if unable to assess the exposure, use the level of
allergen-specic IgE antibodies or the size of skin test
Eczema wheal as an indicator
There are as yet no systematic reviews assessing the eect of Start the intervention as early in the natural history of
mite avoidance measures in patients with eczema. Two small the disease as possible.
studies [54, 55] compared mite-impermeable encasings /
acaricides with controls and failed to show any improvement
in eczema. In contrast, an earlier study in which half of the
study population were children, with a more comprehensive
environmental intervention which included a combination of ALLERGEN AVOIDANCE IN THE PREVENTION
bed covers, acaricides, and high-ltration vacuum cleaners for
6 months, demonstrated a signicantly greater improvement
OF ALLERGIC DISEASE
in the severity score and area aected by eczema in the active
compared to the control group [56]. The consistent nding of an association between allergic
sensitization and childhood asthma raises the question as
to whether reducing exposure to allergens early in life can
Pet allergen avoidance reduce the risk of subsequent development of sensitization
and/or symptoms of allergic disease.
While experience suggests that clinical improvement can
occur when a pet to whom a patient is sensitized is removed
[57], it rarely happens. There have been three small studies Secondary prevention
of cat and dog allergen avoidance with the pet still in home
with contrasting outcomes [58, 59], A Cochrane Airways Secondary preventive measures are designed to halt the
review emphasized the lack of evidence on the clinical progression of disease in individuals who are at high risk
eectiveness of pet allergen avoidance [59]. Larger studies for the development of allergic disease, but who have not
are needed before denitive recommendations can be made. yet developed specic symptoms (e.g., prevention of asthma
in individuals with eczema, rhinitis, or evidence of allergen
senitization). The evidence for eective secondary preven-
Food allergen avoidance tion measures is scarce and only a small number of second-
ary prevention trials using allergen avoidance measures have
It is dicult to get patients to comply with food allergen been conducted (most research to date has focused on pri-
avoidance. The benecial eects of food avoidance in food- mary and tertiary prevention).
allergic patients without other allergic disorders are unam- The multicentre Study on the Prevention of Allergy in
biguous, this approach being indeed the mainstay of their Children in Europe (SPACE) secondary prevention study

594
Allergen Avoidance 46
investigated the protective eect of mite avoidance in high- the active group. At age 7, physician-diagnosed asthma was
risk allergic children (family history of atopy asthma, rhini- reduced in the intervention group compared to the control
tis, eczema, but not sensitized to mite). In one study, 636 group (14.9% versus 23.0%) [71], but there was no dier-
young children aged 1.55 years were randomized to mite- ence in allergic rhinitis, eczema, atopy, and bronchial hyper-
impermeable mattress covers and specic mite avoidance responsiveness [71].
advice versus general advice only [62]. The rate of senitiza- In the Study on the Prevention of Allergy in Children
tion to mite after 1 year was reduced in the active group. A in Europe (SPACE), the multifaceted intervention included
similar study and intervention including 242 children aged both inhalant and food allergens, studied in new born chil-
57 years was reported by another group [63]. After 1 year, dren from Austria, Germany, and UK [72, 73]. Mattress
fewer children were newly sensitized to mite in the active covers were applied to the infants bed. Mothers were
group, with a trend for fewer wheezing symptoms. advised to breast-feed, and hypoallergenic formula was rec-
These two studies indicate that senitization to mite ommended as an alternative (Austria and Germany, but not
can be prevented by allergen avoidance in the short term; UK). Parents were advised to delay the introduction of aller-
however, further follow-up of these children is essential genic foods. At age 2, no dierences were found between
to show whether this eect can be sustained, as well as to the control and the intervention groups in the prevalence of
ascertain its impact on the natural history of allergic disease. mite sensitization or respiratory symptoms.
The Childhood Asthma Prevention Study (CAPS)
is a multicentre, parallel group, randomized controlled trial
Primary prevention in Sydney, Australia [7477]. Children from at-risk fami-
lies were randomized into four groups (mite avoidance with
So, does successful reduction in exposure to allergens early placebo dietary intervention, mite avoidance with active die-
in life prevent the development of sensitization and aller- tary intervention, active dietary intervention alone, no mite
gic disease? This topic has been a subject of several recent avoidance; placebo dietary intervention, no mite avoidance).
review articles [64, 65]. Seven primary prevention studies A signicant reduction in the very high baseline mite lev-
are ongoing, all focussed on children at high risk of devel- els was achieved in the mite intervention group; the dietary
oping allergic disease (e.g., both parents atopic, single parent intervention consisted of omega 3 fatty acids supplemen-
atopic, mother with asthma, etc.). The studies used dier- tation. At age 3 and 5 years, there was a signicant reduc-
ent environmental control approaches (e.g., four combined tion in sensitization to house dust mite in the active mite
dietary intervention with environmental control). Dierent allergen avoidance group, but no signicant dierences in
outcomes were assessed at dierent ages. While the results wheeze were found with either intervention [75]. At 5, the
are not directly comparable, they do provide interesting and prevalence of eczema was higher in the active mite avoid-
complementary data. ance group (26% versus 19%) [76].
Isle of Wight was the rst primary prevention study In the Primary Prevention of Asthma in Children
[6669]. A complex intervention involved avoidance of both Study (PREVACS) in the Netherlands, children were
food and dust mite allergens until age 9 months. Mothers recruited during the prenatal period and randomized to
who were breast-feeding their children were asked to avoid either a control group (receiving usual care) or an interven-
allergenic foods, and these foods were not introduced into tion group in which families received instruction from nurses
the infants diet until after 9 months of age. Infants who on how to reduce exposure of newborns to mite, pet and food
were not breast-fed were given a hydrolyzed formula. allergens, and passive smoking. This study was not eective
Environmental control comprised of the application of aca- in reducing asthma-like symptoms in high-risk children dur-
ricide to carpets and upholstered furniture. Infants in both ing the rst 2 years of life, and although a modest eect was
groups slept on polyvinyl-covered mattresses with vented observed at age 2 years, no signicant dierences in the total
head area. Mite allergen levels at age 9 months were lower and specic IgE were found between the groups [78].
in the active than in the control group but were still higher The Prevention and Incidence of Asthma and Mite
than levels seen in most studies. Allergy (PIAMA) is a multi-centre, population-based
By age 8 years in the active group, sensitization to mite cohort study of 4000 children in the Netherlands [79, 80].
was reduced by more than 50%, and current wheeze, noc- Nested within this cohort is an intervention study amongst
turnal cough, wheeze with bronchial hyper-responsiveness, 810 high-risk infants who were randomly allocated to
and atopy were lower in the multivariate analysis. [69]. receive active or placebo mite-proof encasings for the mat-
However, due to the study design it is impossible to deter- tress and pillows of the parental and infant beds. Advice was
mine which intervention is responsible. given to the active group to wash bedding regularly at more
The Canadian Primary Prevention Study (CaPPS) than 60C. Baseline mite allergens levels were low in both
exemplies the complexity of adherence to multifaceted groups, and even lower in the active group at 1 year, there
interventions [70, 71]. Compliance with mattress encas- was little change in the absolute value [79]. At age 2 and
ings and acaricides was excellent. Mite allergens were sig- 4 years, sensitization and allergic symptoms were similar in
nicantly reduced in the parental bed throughout the both groups [79, 80].
study, while childs mattress levels were low in both groups. The Manchester Asthma and Allergy Study (MAAS)
However, despite advice, there was no reduction in the use is a whole population birth cohort study of 1000 children,
of carpets and in pet ownership in the active group com- with a nested environmental intervention study in the high-
pared to controls. There was a signicant reduction in risk group. Children with two atopic parents who had no
probable asthma and rhinitis without colds at age 2 in pets in their home were randomly allocated before birth to

595
Asthma and COPD: Basic Mechanisms and Clinical Management

an extremely stringent environmental control (n 145) or REFERENCES


normal regime (n 146) [81, 82]. This included mite proof
encasings to the parental mattress, duvet and pillows by 1. Illi S, von Mutius E, Lau S, Niggemann B, Gruber C, Wahn U.
16th week of pregnancy; mite proof cot and carry cot mat- Perennial allergen sensitisation early in life and chronic asthma in chil-
tresses; advice to hot wash bedding weekly at over 55C, dren: A birth cohort study. Lancet 368(9537): 76370, 2006.
supply of a high-ltration vacuum cleaner, carpets removed 2. Simpson BM, Custovic A, Simpson A, Hallam CL, Walsh D, Marolia H
from the nursery and a vinyl cushion oor tted, acaricide et al. NAC Manchester Asthma and Allergy Study (NACMAAS):
for carpets with high mite allergen levels, hot washable toy. Risk factors for asthma and allergic disorders in adults. Clin Exp Allergy
No attempt was made to introduce placebo devices into the 31(3): 39199, 2001.
3. Lowe LA, Woodcock A, Murray CS, Morris J, Simpson A, Custovic
control homes. A substantial and sustained reduction in
A. Lung function at age 3 years: Eect of pet ownership and exposure
exposure to mite, cat and dog allergens was observed in the to indoor allergens. Arch Pediatr Adolesc Med 158(10): 9961001, 2004.
homes of children in the active group. 4. Langley SJ, Goldthorpe S, Craven M, Morris J, Woodcock A, Custovic A.
At age 3, children in the intervention group were Exposure and sensitization to indoor allergens: Association with lung
signicantly and more frequently sensitized to dust mite function, bronchial reactivity, and exhaled nitric oxide measures in
compared to controls (risk ratio 2.85) [82]. However, lung asthma. J Allergy Clin Immunol 112(2): 36268, 2003.
function (assessed by the measurement of specic airway 5. Langley SJ, Goldthorpe S, Craven M, Woodcock A, Custovic A.
resistance) was markedly and signicantly better amongst Relationship between exposure to domestic allergens and bronchial
children in the intervention group. Since there was no dif- hyperresponsiveness in non-sensitised, atopic asthmatic subjects. Thorax
ference between the groups in infant lung function at age 60(1): 1721, 2005.
6. Rosenstreich DL, Eggleston P, Kattan M, Baker D, Slavin RG,
4 weeks [82], the dierence had occurred between 4 weeks
Gergen P et al. The role of cockroach allergy and exposure to cockroach
and 3 years of age. Thus, in MAAS stringent environmental allergen in causing morbidity among inner-city children with asthma.
control was associated with increased risk of mite senitiza- N Engl J Med 336(19): 135663, 1997.
tion but better lung function at age 3 years. 7. Tunniclie WS, Fletcher TJ, Hammond K, Roberts K, Custovic A,
Simpson A et al. Sensitivity and exposure to indoor allergens in adults
with diering asthma severity. Eur Respir J 13(3): 65459, 1999.
8. Murray CS, Poletti G, Kebadze T, Morris J, Woodcock A, Johnston SL
CONCLUSIONS et al. Study of modiable risk factors for asthma exacerbations: Virus
infection and allergen exposure increase the risk of asthma hospital
admissions in children. Thorax 61(5): 37682, 2006.
Established adult asthma: there is little evidence to 9. Simpson A, Custovic A, Pipis S, Adisesh A, Faragher B, Woodcock A.
support the use of simple physical or chemical methods Exhaled nitric oxide, sensitization, and exposure to allergens in patients
as single interventions to control dust mite or pet with asthma who are not taking inhaled steroids. Am J Respir Crit Care
allergen levels (e.g., mattress encasings, acaricides, Med 160(1): 4549, 1999.
or HEPA lters). While it remains possible that a 10. Custovic A, Taggart SC, Francis HC, Chapman MD, Woodcock A.
multifaceted intervention in carefully selected patients Exposure to house dust mite allergens and the clinical activity of
could have some benet, this has not as yet been asthma. J Allergy Clin Immunol 98(1): 6472, 1996.
11. Platts-Mills TA, Tovey ER, Mitchell EB, Moszoro H, Nock P,
addressed in an adequately designed study in adults.
Wilkins SR. Reduction of bronchial hyperreactivity during prolonged
There are serious concerns about the eectiveness and allergen avoidance. Lancet 2(8300): 67578, 1982.
sustainability outside the context of a clinical trial. There 12. Peroni DG, Piacentini GL, Costella S, Pietrobelli A, Bodini A,
is inadequate evidence to advise allergen avoidance in Loiacono A et al. Mite avoidance can reduce air trapping and airway
adults. inammation in allergic asthmatic children. Clin Exp Allergy 32(6):
85055, 2002.
In childhood asthma, several smaller trials of allergen-
13. Milanese M, Peroni D, Costella S, Aralla R, Loiacono A, Barp C et al.
impermeable bed encasings as a single intervention in Improved bronchodilator eect of deep inhalation after allergen avoid-
asthmatic children reported benets, as did a large study ance in asthmatic children. J Allergy Clin Immunol 114(3): 50511,
of a more comprehensive environmental intervention in 2004.
children living in poor quality housing. 14. Sensi LG, Piacentini GL, Nobile E, Ghebregzabher M, Brunori R,
The reasons for the apparent dierences in response Zanolla L et al. Changes in nasal specic IgE to mites after periods of
allergen exposure-avoidance: A comparison with serum levels. Clin Exp
between adults and children are not clear, but the
Allergy 24(4): 37782, 1994.
situation may be analogous to that observed in the 15. Chan-Yeung M, Malo JL. Occupational asthma. N Engl J Med 333(2):
occupational asthma, where early intervention is 10712, 1995.
associated with a better long-term outcome. 16. Custovic A, Murray CS, Gore RB, Woodcock A. Controlling indoor
In the prevention of allergic disease, the clinical outcomes allergens. Ann Allergy Asthma Immunol 88(5): 43241, 2002. quiz
reported from dierent intervention studies appear 44243, 529
17. Custovic A, Green R, Fletcher A, Smith A, Pickering CA,
inconsistent and often confusing. Much longer follow-up
Chapman MD et al. Aerodynamic properties of the major dog aller-
is required before we can be sure that the interventions gen Can f 1: Distribution in homes, concentration, and particle size of
do not cause harm. There is very unlikely to be a one- allergen in the air. Am J Respir Crit Care Med 155(1): 9498, 1997.
size-ts-all strategy for primary prevention which will be 18. Custovic A, Simpson A, Pahdi H, Green RM, Chapman MD,
applicable to the whole population; Specic environmental Woodcock A. Distribution, aerodynamic characteristics, and removal
interventions will be designed and targeted to genetically of the major cat allergen Fel d 1 in British homes. Thorax 53(1): 3338,
susceptible individuals [83, 84]. 1998.

596
Allergen Avoidance 46
19. Custovic A, Simpson B, Simpson A, Hallam C, Craven M, Woodcock A. 40. Walshaw MJ, Evans CC. Allergen avoidance in house dust mite sensi-
Relationship between mite, cat, and dog allergens in reservoir dust and tive adult asthma. Q J Med 58(226): 199215, 1986.
ambient air. Allergy 54(6): 61216, 1999. 41. van der Heide S, Kauman HF, Dubois AE, de Monchy JG. Allergen
20. Mahakittikun V, Boitano JJ, Tovey E, Bunnag C, Ninsanit P, reduction measures in houses of allergic asthmatic patients: Eects of
Matsumoto T et al. Mite penetration of dierent types of material air-cleaners and allergen-impermeable mattress covers. Eur Respir J
claimed as mite proof by the Siriraj chamber method. J Allergy Clin 10(6): 121723, 1997.
Immunol 118(5): 116468, 2006. 42. Htut T, Higenbottam TW, Gill GW, Darwin R, Anderson PB, Syed N.
21. Tovey E, Marks G. Methods and eectiveness of environmental con- Eradication of house dust mite from homes of atopic asthmatic sub-
trol. J Allergy Clin Immunol 103(2 Pt 1): 17991, 1999. jects: A double-blind trial. J Allergy Clin Immunol 107(1): 5560, 2001.
22. Simpson A, Woodcock A, Custovic A. Housing characteristics and 43. Woodcock A, Forster L, Matthews E, Martin J, Letley L, Vickers M
mite allergen levels: To humidity and beyond. Clin Exp Allergy 31(6): et al. Control of exposure to mite allergen and allergen-impermeable
8035, 2001. bed covers for adults with asthma. N Engl J Med 349(3): 22536, 2003.
23. Fletcher AM, Pickering CA, Custovic A, Simpson J, Kennaugh J, 44. Luczynska C, Tredwell E, Smeeton N, Burney P. A randomized con-
Woodcock A. Reduction in humidity as a method of controlling mites trolled trial of mite allergen-impermeable bed covers in adult mite-
and mite allergens: The use of mechanical ventilation in British domes- sensitized asthmatics. Clin Exp Allergy 33(12): 164853, 2003.
tic dwellings. Clin Exp Allergy 26(9): 105156, 1996. 45. van den Bemt L, van Knapen L, de Vries MP, Jansen M, Cloosterman S,
24. Custovic A, Taggart SC, Kennaugh JH, Woodcock A. Portable dehu- van Schayck CP. Clinical eectiveness of a mite allergen-impermeable
midiers in the control of house dust mites and mite allergens. Clin bed-covering system in asthmatic mite-sensitive patients. J Allergy Clin
Exp Allergy 25(4): 31216, 1995. Immunol 114(4): 85862, 2004.
25. Custovic A, Simpson BM, Simpson A, Hallam C, Craven M, Brutsche M 46. Ehnert B, Lau-Schadendorf S, Weber A, Buettner P, Schou C, Wahn U.
et al. Manchester Asthma and Allergy Study: Low-allergen environ- Reducing domestic exposure to dust mite allergen reduces bron-
ment can be achieved and maintained during pregnancy and in early chial hyperreactivity in sensitive children with asthma. J Allergy Clin
life. J Allergy Clin Immunol 105(2 Pt 1): 25258, 2000. Immunol 90(1): 13538, 1992.
26. Wood RA, Chapman MD, Adkinson NF Jr, Eggleston PA. The eect 47. Carswell F, Birmingham K, Oliver J, Crewes A, Weeks J. The respi-
of cat removal on allergen content in household-dust samples. J Allergy ratory eects of reduction of mite allergen in the bedrooms of asth-
Clin Immunol 83(4): 73034, 1989. matic children a double-blind controlled trial. Clin Exp Allergy 26(4):
27. Green R, Simpson A, Custovic A, Faragher B, Chapman M, 38696, 1996.
Woodcock A. The eect of air ltration on airborne dog allergen. 48. Carter MC, Perzanowski MS, Raymond A, Platts-Mills TA. Home
Allergy 54(5): 48488, 1999. intervention in the treatment of asthma among inner-city children.
28. Gore RB, Bishop S, Durrell B, Curbishley L, Woodcock A, Custovic A. J Allergy Clin Immunol 108(5): 73237, 2001.
Air ltration units in homes with cats: Can they reduce personal expo- 49. Halken S, Host A, Niklassen U, Hansen LG, Nielsen F, Pedersen S
sure to cat allergen?. Clin Exp Allergy 33(6): 76569, 2003. et al. Eect of mattress and pillow encasings on children with asthma and
29. de Blay F, Chapman MD, Platts-Mills TA. Airborne cat allergen house dust mite allergy. J Allergy Clin Immunol 111(1): 16976, 2003.
(Fel d I). Environmental control with the cat in situ. Am Rev Respir Dis 50. Morgan WJ, Crain EF, Gruchalla RS, OConnor GT, Kattan M, Evans R
43(6): 133439, 1991. et al. Results of a home-based environmental intervention among urban
30. Avner DB, Perzanowski MS, Platts-Mills TA, Woodfolk JA. children with asthma. N Engl J Med 351(11): 106880, 2004.
Evaluation of dierent techniques for washing cats: Quantitation 51. Kattan M, Stearns SC, Crain EF, Stout JW, Gergen PJ, Evans R 3rd
of allergen removed from the cat and the eect on airborne Fel d 1. et al. Cost-eectiveness of a home-based environmental intervention
J Allergy Clin Immunol 100(3): 30712, 1997. for inner-city children with asthma. J Allergy Clin Immunol 116(5):
31. Hodson T, Custovic A, Simpson A, Chapman M, Woodcock A, 105863, 2005.
Green R. Washing the dog reduces dog allergen levels, but the dog needs 52. Sheikh A, Hurwitz B. House dust mite avoidance measures for peren-
to be washed twice a week. J Allergy Clin Immunol 103(4): 58185, 1999. nial allergic rhinitis. Cochrane Database Syst Rev 4: 2001, CD001563.
32. Gore RB, Durrell B, Bishop S, Curbishley L, Woodcock A, Custovic A. 53. Terreehorst I, Hak E, Oosting AJ, Tempels-Pavlica Z, de Monchy JG,
High-eciency particulate arrest-lter vacuum cleaners increase per- Bruijnzeel-Koomen CA et al. Evaluation of impermeable covers for
sonal cat allergen exposure in homes with cats. J Allergy Clin Immunol bedding in patients with allergic rhinitis. N Engl J Med 349(3): 23746,
111(4): 78487, 2003. 2003.
33. Eggleston PA. Cockroach allergen abatement in inner-city homes. 54. Oosting AJ, de Bruin-Weller MS, Terreehorst I, Tempels-Pavlica
Ann Allergy Asthma Immunol 91(6): 51214, 2003. Z, Aalberse RC, de Monchy JG et al. Eect of mattress encasings
34. Eggleston PA, Arruda LK. Ecology and elimination of cockroaches on atopic dermatitis outcome measures in a double-blind, placebo-
and allergens in the home. J Allergy Clin Immunol 107(3 Suppl): controlled study: The Dutch mite avoidance study. J Allergy Clin
S42229, 2001. Immunol 110(3): 5006, 2002.
35. Eggleston PA, Butz A, Rand C, Curtin-Brosnan J, Kanchanaraksa S, 55. Gutgesell C, Heise S, Seubert S, Seubert A, Domhof S, Brunner E
Swartz L et al. Home environmental intervention in inner-city asthma: et al. Double-blind placebo-controlled house dust mite control meas-
A randomized controlled clinical trial. Ann Allergy Asthma Immunol ures in adult patients with atopic dermatitis. Br J Dermatol 145(1):
95(6): 51824, 2005. 7074, 2001.
36. Custovic A, Wijk RG. The eectiveness of measures to change the 56. Tan BB, Weald D, Strickland I, Friedmann PS. Double-blind control-
indoor environment in the treatment of allergic rhinitis and asthma: led trial of eect of housedust-mite allergen avoidance on atopic der-
ARIA update (Collaboration with GA(2)LEN). Allergy 60(9): 111215, matitis. Lancet 347(8993): 1518, 1996.
2005. 57. Shirai T, Matsui T, Suzuki K, Chida K. Eect of pet removal on pet
37. Gotzsche P, Johansen H. House dust mite control measures for asthma. allergic asthma. Chest 127(5): 156571, 2005.
Cochrane Database Syst Rev 2: 2008, CD001187. 58. Francis H, Fletcher G, Anthony C, Pickering C, Oldham L, Hadley E
38. Gotzsche PC, Johansen HK, Schmidt LM, Burr ML. House dust et al. Clinical eects of air lters in homes of asthmatic adults sensi-
mite control measures for asthma. Cochrane Database Syst Rev 4: 2004, tized and exposed to pet allergens. Clin Exp Allergy 33(1): 1015, 2003.
CD001187. 59. van der Heide S, van Aalderen WM, Kauman HF, Dubois AE,
39. Gotzsche PC, Johansen HK, Hammarquist C, Burr ML. House dust de Monchy JG. Clinical eects of air cleaners in homes of asthmatic
mite control measures for asthma. Cochrane Database Syst Rev 2: 2001, children sensitized to pet allergens. J Allergy Clin Immunol 104(2 Pt 1):
CD001187. 44751, 1999.

597
Asthma and COPD: Basic Mechanisms and Clinical Management

60. Lever R, MacDonald C, Waugh P, Aitchison T. Randomised controlled birth-cohort study 24 months results of the Study of Prevention of Allergy
trial of advice on an egg exclusion diet in young children with atopic in Children in Europe. Clin Exp Allergy 34(8): 122025, 2004.
eczema and sensitivity to eggs. Pediatr Allergy Immunol 9(1): 1319, 74. Mihrshahi S, Peat JK, Marks GB, Mellis CM, Tovey ER, Webb K et al.
1998. Eighteen-month outcomes of house dust mite avoidance and dietary
61. Mabin DC, Sykes AE, David TJ. Controlled trial of a few foods diet in fatty acid modication in the Childhood Asthma Prevention Study
severe atopic dermatitis. Arch Dis Child 73(3): 2027, 1995. (CAPS). J Allergy Clin Immunol 111(1): 16268, 2003.
62. Arshad SH, Bojarskas J, Tsitoura S, Matthews S, Mealy B, Dean T 75. Peat JK, Mihrshahi S, Kemp AS, Marks GB, Tovey ER, Webb K et al.
et al. Prevention of sensitization to house dust mite by allergen avoid- Three-year outcomes of dietary fatty acid modication and house dust
ance in school age children: A randomized controlled study. Clin Exp mite reduction in the Childhood Asthma Prevention Study. J Allergy
Allergy 32(6): 84349, 2002. Clin Immunol 114(4): 80713, 2004.
63. Tsitoura S, Nestoridou K, Botis P, Karmaus W, Botezan C, Bojarskas J 76. Marks GB, Mihrshahi S, Kemp AS, Tovey ER, Webb K, Almqvist C
et al. Randomized trial to prevent sensitization to mite allergens in tod- et al. Prevention of asthma during the rst 5 years of life: A rand-
dlers and preschoolers by allergen reduction and education: One-year omized controlled trial. J Allergy Clin Immunol 118(1): 5361, 2006.
results. Arch Pediatr Adolesc Med 156(10): 102127, 2002. 77. Almqvist C, Li Q, Britton WJ, Kemp AS, Xuan W, Tovey ER et al.
64. Simpson A, Custovic A. Allergen avoidance in the primary prevention Early predictors for developing allergic disease and asthma: Examining
of asthma. Curr Opin Allergy Clin Immunol 4(1): 4551, 2004. separate steps in the allergic march. Clin Exp Allergy 37(9): 1296302,
65. Simpson A, Custovic A. The role of allergen avoidance in primary and 2007.
secondary prevention. Pediatr Pulmonol Suppl 26: 22528, 2004. 78. Schonberger HJ, Dompeling E, Knottnerus JA, Maas T, Muris JW,
66. Arshad SH, Matthews S, Gant C, Hide DW. Eect of allergen avoid- van Weel C et al. The PREVACS study: The clinical eect of a mul-
ance on development of allergic disorders in infancy. Lancet 339(8808): tifaceted educational intervention to prevent childhood asthma. Eur
149397, 1992. Respir J 25(4): 66070, 2005.
67. Hide DW, Matthews S, Matthews L, Stevens M, Ridout S, Twiselton R 79. Koopman LP, van Strien RT, Kerkhof M, Wijga A, Smit HA,
et al. Eect of allergen avoidance in infancy on allergic manifestations de Jongste JC et al. Placebo-controlled trial of house dust mite-
at age two years. J Allergy Clin Immunol 93(5): 84246, 1994. impermeable mattress covers: Eect on symptoms in early childhood.
68. Hide DW, Matthews S, Tariq S, Arshad SH. Allergen avoidance in Am J Respir Crit Care Med 166(3): 30713, 2002.
infancy and allergy at 4 years of age. Allergy 51(2): 8993, 1996. 80. Corver K, Kerkhof M, Brussee JE, Brunekreef B, van Strien RT, Vos AP
69. Arshad SH, Bateman B, Matthews SM. Primary prevention of asthma et al. House dust mite allergen reduction and allergy at 4 yr: Follow up
and atopy during childhood by allergen avoidance in infancy: A ran- of the PIAMA-study. Pediatr Allergy Immunol 17(5): 32936, 2006.
domised controlled study. Thorax 58(6): 48993, 2003. 81. Custovic A, Simpson BM, Simpson A, Kissen P, Woodcock A. Eect
70. Becker A, Watson W, Ferguson A, Dimich-Ward H, Chan-Yeung M. of environmental manipulation in pregnancy and early life on respira-
The Canadian Asthma Primary Prevention Study: Outcomes at 2 years tory symptoms and atopy during rst year of life: A randomised trial.
of age. J Allergy Clin Immunol 113(4): 65056, 2004. Lancet 358(9277): 18893, 2001.
71. Chan-Yeung M, Ferguson A, Watson W, Dimich-Ward H, Rousseau R, 82. Woodcock A, Lowe LA, Murray CS, Simpson BM, Pipis SD,
Lilley M et al. The Canadian Childhood Asthma Primary Prevention Kissen P et al. Early life environmental control: Eect on symptoms,
Study: Outcomes at 7 years of age. J Allergy Clin Immunol 116(1): 49 sensitization, and lung function at age 3 years. Am J Respir Crit Care
55, 2005. Med 170(4): 43339, 2004.
72. Halmerbauer G, Gartner C, Schierl M, Arshad H, Dean T, Koller DY 83. Simpson A, John SL, Jury F, Niven R, Woodcock A, Ollier WE
et al. Study on the Prevention of Allergy in Children in Europe (SPACE): et al. Endotoxin exposure, CD14, and allergic disease: An interac-
Allergic sensitization at 1 year of age in a controlled trial of allergen tion between genes and the environment. Am J Respir Crit Care Med
avoidance from birth. Pediatr Allergy Immunol 14(1): 1017, 2003. 174(4): 38692, 2006.
73. Horak F Jr, Matthews S, Ihorst G, Arshad SH, Frischer T, Kuehr J et al. 84. Custovic A, Simpson A. Environmental allergen exposure, sensitisa-
Eect of mite-impermeable mattress encasings and an educational package tion and asthma: From whole populations to individuals at risk. Thorax
on the development of allergies in a multinational randomized, controlled 59(10): 82527, 2004.

598
Smoking Cessation
47 CHAPTER

declines in smoking prevalence, smoking-related


INTRODUCTION mortality is trending downward in developed
Michael A. Chandler and
countries like the United States and United Stephen I. Rennard
Cigarette smoking is a chronic relapsing disorder Kingdom; however, other developing countries University of Nebraska Medical Center,
with secondary complications including athero- are experiencing dramatic increases in mortality
Omaha, NE, USA
sclerotic cardiovascular disease, chronic obstruc- [3]. In China, smoking is attributed to a million
tive pulmonary disease (COPD), and lung cancer. deaths annually, a gure that is still on the rise [4].
Smoking remains the leading cause of pre- India is expected to achieve this dubious milestone
ventable death in the world [1]. Tobacco control by 2010 [5]. Barring alterations in present tobacco
policies and smoking cessation therapies have use, the tobacco epidemic will lead to an estimated
been shown to be eective deterrents and treat- one billion deaths this century alone [1].
ments (Fig. 47.1). This chapter will outline the
epidemiology, pathophysiology, and treatment of
cigarette smoking with particular emphasis on
current pharmacotherapies including nortriptyl-
ine, clonidine, nicotine replacement, bupropion,
A HETEROGENEOUS DISEASE
and varenicline.
Heterogeneity in the susceptibility to and persist-
ence of cigarette smoking is governed by genes,
environment, and gene by environment interac-
THE GLOBAL TOBACCO EPIDEMIC tions [6]. Genes that modulate neurotransmit-
ter signaling, best characterized for dopamine
and opiod signaling, have been implicated [7, 8].
Cigarette smoking led to ve million premature Polymorphisms in the enzymes that metabolize
deaths worldwide in 2000 [2] (Fig. 47.2). Due to nicotine into inactive metabolites like cotinine
Forced expiratory volume in 1 second

Never smoked or not


100 susceptible to smoke
(% of value at age 25)

75 Smoked regularly
and susceptible Stopped
to its effects at 50
50
Disability Stopped at 65
25
Death
0 FIG. 47.1 The natural history of chronic
25 50 75 bronchitis and emphysema. Adapted from
Age (years) Fletcher M, Peto R. BMJ. 1, 164548, 1977.

599
Asthma and COPD: Basic Mechanisms and Clinical Management

Tobacco use is a risk factor for six of the eight


leading causes of death in the world
8

7 Hatched areas indicate proportions of deaths


that are related to tobacco use and are
coloured according to the column of the
6 respective cause of death.
Millions of deaths (2005)

*Includes mouth and oropharyngeal cancers,


5 oesophageal cancer, stomach cancer, liver
cancer, other cancers, as well as cardiovascular
diseases other than ischaemic heart disease
4 and cerebrovascular disease.

Source: Mathers CD, Loncar D. Projections of


3 global mortality and burden of disease from
2002 to 2030. PLoS Medicine, 2006, 3(11):
e442. Additional information obtained from
2 personal communication with C.D. Mathers.

Source of revised HIV/AIDS figure: AIDS


1 Other epidemic update. Geneva, Joint United Nations
tobacco- Programme on HIV/AIDS (UNAIDS) and World
caused Health Organization (WHO), 2007.
diseases*
0

Ischaemic Cerebro- Lower Chronic HIV/AIDS Diarrhoeal Tuberculosis Trachea, Tobacco


heart vascular respiratory obstructive diseases bronchus, use
disease disease infections pulmonary lung cancers
disease

FIG. 47.2 Individual and Composite Impact of Tobacco Use on Death Rate of the Eight Leading Causes of Death in the World. From World Health
Organization (WHO), report on the global tobacco epidemic, 2008: The MPOWER package, Geneva.

and 3-hydroxycotinine also play a role. For example, slow


metabolizers are less likely to become smokers. Nevertheless, Nicotine: Addiction and conditioning
they comprise about 20% of total smokers but are likely to
smoke fewer cigarettes, perhaps because they overdose
experiencing higher levels of nicotine for greater duration.
The opposite is true for fast metabolizers [9]. Environmental
cues, such as social prohibition or acceptance of smoking, also
impact smoking prevalence. These factors and their interplay
lead to a broad spectrum of smoking behaviors. Nicotine

NICOTINE ADDICTION Neurologic


adaptation
Psychologic
conditioning

Nicotine as an addictive euphoriant is comparable to ampheta-


mines, cocaine, or opiates [10]. Nicotinic acetylcholine receptors
in the central nervous system regulate downstream neurotrans- Withdrawal Persistent Anxiety
mitter release. For example, nicotine addiction appears to be smoking
mediated by nicotine-stimulated dopamine release in the mes-
olimbic system, a pathway involved in endogenous reward and
behavioral consolidation [11, 12]. Additionally, smokers habit- FIG. 47.3 Biopsychosocial model of nicotine addiction.
ually smoke during common situations, for example, when
driving or socializing or after eating or during stressful conver-
sations. This leads to classic or Pavlovian conditioning, which resembles a grief response. Insights such as these have led to
is potentiated by nicotine [13]. our current understanding of tobacco dependence as a chronic
The interplay between these neurophysiologic and psy- relapsing disorder requiring recurrent therapy.
chologic forces leads to addiction for roughly 85% of smokers
[14] (Fig. 47.3). The presence of addiction is further evidenced
by a well-dened withdrawal syndrome upon smoking cessa-
tion. Features of this syndrome include dysphoria, insomnia,
irritability, anxiety, diminished concentration, restlessness,
SPECTRUM OF COMPLICATIONS
increased appetite, decreased heart rate, and weight gain [15].
Craving for smoking often persists years following cessation Smoking leads to about one-third of deaths from coronary
and may wane in frequency but not in intensity, a feature that artery disease (CAD), the most common cause of morbidity

600
Smoking Cessation 47
TABLE 47.1 The 5 As: brief smoking cessation strategies for routine practice. The 5 Rs: generating or reinforcing motivation to quit smoking.

Step Action

Five As

Ask about smoking Systematically assess smoking status


Smoking should be a vital sign

Advise all smokers to stop Give a clear, strong, and personalized message to stop smoking

Assess willingness to stop Determine whether the smoker is ready to stop currently or soon. Provide assistance to motivated smokers;
motivate those unwilling to stop

Assist motivated smokers to stop Help smoker with a quit plan, counseling, pharmacotherapy and additional materials

Arrange follow-up Follow up soon after the quit date. Assess success and difficulties. Consider referring for a more intensive
intervention if there is relapse

Five Rs

Relevance Encourage the patient to indicate why quitting is personally relevant

Risks Ask the patient to identify potential negative consequences of tobacco use

Rewards Ask the patient to identify potential benefits of stopping tobacco use

Roadblocks Ask the patient to identify barriers to quitting

Repetition Apply 5 Rs whenever interacting with unmotivated or relapsed smokers

Adapted from Ref. [45] and http:ahrq.gov/clinic/tobacco/5rs.htm.

and mortality in the developed world [8, 16, 17]. Active and trends have been decreasing in the United States, recent esti-
passive smoke exposure leads to increases in oxidative stress, mates suggest this decrease has slowed and nearly stopped
vascular inammation, blood coagulability, platelet aggre- with a current smoker rate of 20.8% [34]. Public health ini-
gation, and thrombus formation as well as reduced oxygen tiatives and tobacco control programs in the United States
delivery and coronary vasoconstriction [16, 18]. Heart dis- remain underfunded and susceptible to having monies with-
ease risk drops o signicantly after 12 years of cessation drawn and allocated to support other programs or to cover
and normalizes at 35 years [18, 19]. budget decits. Despite ample evidence supporting the
COPD and cigarette smoking are closely linked. achievements and returns to society of such programs, they
Smoking directly damages the lung and triggers a cascade are transient xtures in public health [35]. Nevertheless,
of secondary inammation and impaired repair that leads many states have continued to push for total bans on smok-
to ongoing damage [20]. These eects lead to COPD in ing and 24 states have been successful. Internationally, sev-
roughly 50% of active smokers and 7% of passive smoke eral countries now too have similar laws prohibiting smoking
exposed individuals [2123]. Exposure in utero or in adoles- nationwide.
cence compromises subsequent lung growth and predisposes
to COPD [2427].
In countries where smoking is common, the risk of
lung cancer increases 20-fold leading to 90% of lung cancers
[28]. Smoking has been clearly linked to genetic and epige- SMOKING CESSATION: BEHAVIORAL
netic alterations that promote carcinogenesis [16, 29, 30]. INTERVENTION

Smoking abstinence rates at 612 months in motivated quit-


ters without behavioral or pharmacologic therapy approach
PUBLIC HEALTH RESPONSE 35% [36]. Even minimal behavioral therapy delivered
by a medical practitioner increases this abstinence rate to
5.57.5% [37].
Smoking prevention and cessation should reduce the preva- The 5As provides general guidelines to smoking ces-
lence of smoking and, thus, prevent millions of premature sation in clinical practice (Table 47.1) [38]. The stages of
deaths [3]. Public health measures (advertisement bans, change model predicts smoking abstinence at 1 year and
indoor smoking bans and measures to increase cigarette pur- has become a useful tool in tailoring behavioral tobacco
chase price) increase smoking cessation and reduce tobacco cessation eorts [40] (Fig. 47.4). In this model, smokers
use and passive smoke exposure [3133]. While smoking are regarded as precontemplators who are not thinking of

601
Asthma and COPD: Basic Mechanisms and Clinical Management

Tobacco cessation: Stages of change


accounted for in future cessation trials. All tobacco cessation
attempts should supplement behavioral interventions with
pharmacotherapy as described below.
Precontemplation
Does not want to quit in 6 months

SMOKING CESSATION: PHARMACOTHERAPY


Contemplation
Wants to quit in 6 months
Because the subject characteristics and the support oered
dier across clinical trials, the most conventional means to
assess the eect of pharmacotherapy is the quit rate relative
to placebo.
Preparation When compared to placebo, pharmacotherapy dou-
Sets a quit date
bles or triples quit rates achieved by nonpharmacological
means (Table 47.2). The relatively high placebo quit rates
seen in many clinical trials, which are higher than the rates
noted above for minimal advice, are likely due to selection
Action
Begins pharmacotherapy
of highly motivated individuals for entry.

Nicotine replacement therapy


Maintenance Relapse Smokers who quit not only lose the euphoric eects of nico-
Remains smoke-free Alters cessation strategy
tine but frequently experience symptoms related to nico-
tine withdrawal [4]. Nicotine replacement therapy (NRT),
the most widely used tobacco cessation agent, may par-
FIG. 47.4 Transtheoretical model of change. Adapted from Ref. [39]. tially ameliorate these ill eects and is central to American
and British guidelines [1, 57]. NRT is given at doses suf-
cient to temper withdrawal without providing the rein-
forcing eects of bolus nicotine. NRT almost doubles the
quitting, contemplators who are considering quitting, or untreated quit rate regardless of whether behavioral coun-
preparers who are ready to trial cessation. Precontemplators seling is applied [8, 9]. Smoking abstinence rates at 612
and contemplators may be advanced by discussing the 5Rs months with combined NRT and limited behavioral therapy
(Table 47.1) [38]. With physician guidance, preparers create approach 17% (range 1234%) [43, 4750]. However, a sin-
a smoking cessation plan including a quit date. At this stage, gle round of NRT does not prevent relapse. About one-third
smokers should be advised to inform others of the impend- of quitters will relapse by 4 years whether NRT is employed
ing quit attempt and counseled to avoid places or situations or not a result consistent with our current understanding of
with close associations to smoking. The quit date marks tobacco dependence as a chronic relapsing disorder requiring
their graduation into the action then maintenance stages, recurrent therapy [7, 8].
marked by initial and chronic tobacco cessation, respectively. Nicotine gums, lozenges, patches, nasal sprays, and
Attempts to bypass stages generally decrease the likelihood inhalers are equally ecacious whereas heavy smokers may
of success. Inpatient or outpatient episodes of acute illness benet from higher doses [9, 1519, 51]. The patch and nasal
are an exception to this and represent windows of oppor- spray are benecial for up to 2 and 3 months, respectively.
tunity when smokers are more liable to quit [41, 42]. The Other forms of NRT are used for 36 month courses [9].
chronic, relapsing nature of cigarette smoking means patients There is no benet to tapering of NRT versus abrupt with-
often will cycle through stages several times before achieving drawal [9]. Physician-prescribed NRT appears to be more
stable abstinence [39]. Additional counseling advice may be eective than voluntary over-the-counter NRT [9]. The cost
reviewed elsewhere [38, 39]. of 6 weeks of NRT is comparable to that of smoking one
More intensive behavioral therapies like group or pack per day and diminishes as the course progresses.
individual counseling sessions improve the odds of cessation Nicotine is metabolized by the liver so that NRT
when compared to minimal advice (odds ratio 1.44, 95% with gum or lozenge is dependent on absorption through
CI 1.241.67) [37]. Many smokers, however, are unwill- the buccal mucosa. Moreover, nicotine is poorly absorbed at
ing to participate in more intensive programs. Augmenting low oral pH; thus, gum or lozenge use should be deferred
behavioral support with additional healthcare personnel, during and 15 min after snacks or meals and should not be
follow-up visits, or even a brief phone call increases quit used with acidic beverages [5254]. Oral irritation, indiges-
rates but is not consistently covered by private or public tion, and diarrhea, which likely result from swallowed drug,
health insurance [37, 38]. are the most common side eects [52, 53].
The majority of patients will relapse in the rst Transdermal nicotine patches require no special tech-
612 months. Reasons for relapse should be reviewed and nique for application, have no restrictions regarding dosage

602
Smoking Cessation 47
TABLE 47.2 Pharmacotherapies for smoking cessation.

Time peak venous Common side Odds ratio 1-year smoking


Product nicotine blood level effects Dosage Duration abstinence versus placebo

Nicotine gum or 2530 min Oral irritation, 1 piece every 12 h as needed, 3 months 1.80
lozenge indigestion, diarrhea then taper
Daily maximum: 24 pieces gum or
20 lozenges

Nicotine nasal 515 min Nasal irritation 1 inhalation, each nostril, every 3 months 2.03
spray 12 h as needed, then taper

Daily maximum: 40 doses (80


sprays)

Nicotine inhaler 15 min Oropharyngeal Multiple inhalations over 20 min 3 months 2.14
irritation every 12 h as needed, then taper

Daily maximum: 16 cartridges

Nicotine patch 612 h Vivid nightmares 1 patch daily for 16 or 24 h 2 months 2.16

Bupropion NA Insomnia, lowered 150 mg daily for 3 d, then 150 mg 3 months* 1.83
seizure threshold twice daily

Varenicline NA Nausea 0.5 mg daily increasing to 1 mg 3 months* 3.22


twice daily

Derived from manufacturers product data sheets and Refs [4346].


*Efficacy may be increased by extending duration of therapy beyond 3 months.

timing with oral intake, and are associated with fewer Serious side eects are extremely rare despite short-
gastrointestinal complaints. They also have the lowest addiction and long-term NRT use by millions of smokers over the
potential, likely due to their slow pharmacokinetics [55]. past two decades [9, 59]. Patients suering acute cardio-
Patches are often used at night to treat morning withdrawal vascular events should defer NRT use; otherwise, NRT
symptoms; however, this may lead sleep disturbances, particu- appears safe in patients with known cardiovascular disease
larly vivid dreams or nightmares [56]. Skin irritation occurs in [9, 5865]. Though not recommended, concurrent smoking
up to 54% of patch users but is generally mild and self-limited and NRT use does not appear to enhance cardiac risk [66].
[57]. Rotating the site of patch application is recommended to Nicotine crosses the placenta and is excreted in breast milk.
minimize irritation. While animal studies link various fetal and reproductive
Nicotine nasal spray delivers an aqueous solution of toxicities to nicotine exposure, NRT is devoid of the haz-
nicotine to the nasal mucosa and produces the most rapid ardous compounds present in tobacco smoke [67]. Ongoing
rise in plasma nicotine concentration amongst various tobacco smoking is the single largest modiable risk for
forms of NRT [58]. This may lead to prolonged nicotine pregnancy-related morbidity and mortality and pregnant
dependence. For example, the rate of continued use after 12 smokers have additional motivation to quit [68]. Thus,
months in clinical trials ranges from 3% to 13% for nicotine NRT is a reasonable alternative to ongoing smoking, should
nasal spray versus 1% and 8% for nicotine gum [9, 59]. The behavioral intervention fail. Nicotine gum and lozenge are
most common side eect is nasal irritation that persists in considered safer in this population than other forms of
up to 81% of patients after 3 weeks of use [60]. NRT by the US Food and Drug Administration (USFDA)
The nicotine inhaler is composed of a mouthpiece and [28, 29, 60, 61, 6770].
a plastic, nicotine-containing cartridge that releases nicotine
upon inhalation. Mild mouth or throat irritation and cough
occurs with initial use in one-third of cases and wanes with Bupropion
time [61]. While named an inhaler nicotine does not
eectively reach the lower respiratory tract with this device Bupropion is an atypical antidepressant that has been used
and absorption is mostly through the mouth and pharynx. as a smoking cessation agent for over a decade [71]. It is
Nevertheless, patients with severe airway reactivity should believed to block the reuptake of dopamine, serotonin, and
consider alternate forms of NRT as inhaled nicotine may norepinephrine and thereby treat occult depression, with-
cause bronchospasm. drawal dysphoria, and addiction [44, 72]. Smoking cessation

603
Asthma and COPD: Basic Mechanisms and Clinical Management

rates with bupropion (19% versus placebo) are comparable 1.89 versus placebo. This second- or third-line drug may be
to rates with NRT (17% versus placebo) by indirect com- useful in quitters with intense withdrawal symptoms, but
parison [68]. clear benet beyond 3 months of smoking cessation has not
The standard dose of sustained-released bupropion been established. Dry mouth and sedation, the most com-
used for tobacco cessation is 150 mg twice daily. A meta- mon side eects, occur in two-thirds of cases [81]. Postural
analysis comparing standard-dose to low-dose bupropion hypotension is also common. Clonidine should be tapered o
(150 mg once daily) did not demonstrate a benet to stand- to avoid rebound hypertension.
ard therapy but was underpowered to establish equivalency
[71, 73, 74]. Nonetheless, low-dose therapy remains a rea-
sonable alternative to discontinuation in patients who expe- Varenicline
rience side eects at standard doses. Bupropion is taken
for up to 12 weeks though longer courses may be bene- Varenicline is the rst nicotine receptor partial agonist
cial. One randomized, placebo-controlled trial found that approved for smoking cessation by the USFDA [82]. As
ongoing use of bupropion decreased weight gain (3.8 versus mentioned above, neuronal nicotinic acetylcholine recep-
5.6 kg) and relapse rates in quitters at 1 year of treatment tors stimulate the mesolimbic dopamine release that leads to
[75]. This benet was lost 1 year after drug discontinuation. subsequent nicotine addiction. Varenicline is a partial agonist
Two subsequent studies found no benet in relapse rates in of one such nicotinic receptor, the 42 nicotinic receptor.
quitters 1 year after cessation with limited duration bupro- As a partial agonist, varenicline elicits less dopamine release
pion (36 months) at standard or low doses [76, 77]. than nicotine and blunts subsequent nicotine-induced
Insomnia, the most common side eect, occurs in dopamine release [83]. This leads to less intense craving in
roughly one-third of patients. Bupropion may cause seizures the absence of nicotine and less intense pleasure in the pres-
in 1 patient per 1000 at standard doses leading to its con- ence of nicotine [8487]. By eroding the reinforcing prop-
traindication in persons predisposed to or suering from
erties of smoking, varenicline continues to capture quitters
seizure disorders [44, 72, 78]. Bupropion, like other antide-
weeks after the onset of therapy, an eect that seems to dif-
pressants, increases the risk of suicidality in patients under
fer from other pharmacotherapies [85, 86]. Cessation rates
age 24 with psychiatric disorders, though this does not pre-
up to 44% have been documented in clinical trials [88, 89].
clude its use in carefully monitored settings if the benets are
In a meta-analysis of four randomized, placebo-controlled
felt to outweigh the risks [79]. The USFDA considers bupro-
trials encompassing 3300 participants, the number-needed-
pion in the same risk category as nicotine gum and lozenge
to-treat for ongoing smoking abstinence with varenicline
when used by pregnant women [28, 29, 78]. A higher rate
was 8 versus 15 and 20 for bupropion and NRT, respectively
of spontaneous abortions has been identied in prospective
[88]. Trials subsequent to this analysis continue to demon-
trials of bupropion users who were pregnant or planning a
strate the ecacy and safety of varenicline [86, 87, 90, 91].
pregnancy [78, 80].
Nausea occurs in up to one-third of patients [88]. This
is limited by the initial dose titration and in clinical trials led
Other antidepressants to discontinuation rates of 2.57.6% [88]. Dose should be
adjusted for renal disease (creatinine clearance 30 mlmin)
Nortriptyline, which is not approved by the USFDA for [89]. At doses 3650 times the recommended daily maxi-
smoking cessation, is suggested for consideration for o- mum, low birthweight and subsequent reproductive toxicity,
label use in United States and United Kingdom guidelines but not teratogenicity, were demonstrated. Thus, varenicline
[38]. Quit rates with nortriptyline are comparable to those is USFDA pregnancy category C, the same risk category as
of NRT and bupropion therapy per indirect meta-analysis nicotine gum, nicotine lozenge, and bupropion.
and direct head-to-head comparison [44]. It is worth noting In 2007, the FDA informed health care providers
that the most recent studies in the aforementioned meta- about suicidal thoughts and aggressive and erratic behavior
analysis failed to show benet. Side eects include nausea, in patients treated with varenicline, even non-quitters [92].
sedation, and anticholinergic symptoms. No benet has been This has lead to recommendations that physicians and car-
found with selective serotonin reuptake inhibitors including egivers monitor for neuropsychiatric symptoms such as agi-
uoxetine, sertraline, and paroxetine, nor with serotonin- tation, behavior change, depressed mood, suicidal ideation,
and suicidal behavior.
norepinephrine uptake inhibitors including venlafaxine, in
either individual studies or meta-analyses [44].

Combination therapy
Clonidine
Supplementing trans-dermal nicotine with short-acting
Clonidine, an -2 adrenergic and imidazoline agonist used NRT products, such as nicotine gum, nasal spray, or inhaler,
for the treatment of essential hypertension, has been sug- increases the likelihood of cessation by 40% compared to
gested for o-label use for smoking cessation [1]. Clonidine the use of the same products independently [43]. Using
reduces withdrawal symptoms; however, only one of six ran- bupropion or nortriptyline in combination with NRT has
domized, placebo-controlled studies selected for recent meta- not been shown to be benecial by meta-analysis [44]. The
analysis found a statistically signicant benet [81]. Inclusion ecacy of combination therapy with varenicline and either
of this study raised the pooled odds ratio at 3 months up to NRT or bupropion is unknown at present.

604
Smoking Cessation 47
Cost-effectiveness response to specic pharmacological agents. Research in
this area will help tailor maximally eective smoking cessa-
Consumer expense, lost productivity, and direct and indirect tion pharmacotherapy.
medical costs compound the burden of morbidity and mor-
tality inherent with cigarette smoking. Despite this under-
standing, many practitioners reserve pharmacotherapy for
refractory cases in an eort to minimize costs. Smoking ces- References
sation is cost-eective, regardless of cessation method used,
statistical analysis utilized, and geographic or socioeconomic 1. World Health Organization. WHO report on the global tobacco
strata examined [9395]. Thus, current guidelines suggest epidemic. Geneva: The mpower package, 2008.
that all individuals who are willing to make a serious quit 2. Ezzati M, Lopez AD. Estimates of global mortality attributable to
attempt should be given the best opportunity to achieve suc- smoking in 2000. Lancet 362(9387): 84752, 2003.
3. MMWR. Annual smoking-attributable mortality, years of potential life
cess, including pharmacological support [96].
lost and productivity losses United States 19972001. MMWR Morb
In the United States in 2000, the cost per life-year Mortal Wkly Rep 54(25): 625628.
saved with NRT and bupropion was cost $1914 to $11,217, 4. Liu BQ , Peto R, Chen ZM, Boreham J, Wu YP, Li JY, Campbell TC,
a relative bargain. During this time period, the cost per Chen JS. Emerging tobacco hazards in China: 1. Retrospective propor-
quality-adjusted life-year saved with lovastatin, a popular tional mortality study of one million deaths. BMJ 317(7170): 141122,
drug used for the primary prevention of cardiovascular dis- 1998.
ease, was $56,415 to $438,780 [93]. Subsequent studies sup- 5. Jha P, Jacob B, Gajalakshmi V, Gupta PC, Dhingra N, Kumar R,
port the cost-eectiveness of varenicline [94]. In the near Sinha DN, Dikshit RP, Parida DK, Kamadod R, Boreham J, Peto R.
future, gene testing to predict treatment responses may oer A nationally representative case-control study of smoking and death in
additional cost reduction [97]. India. New Engl J Med 358(11): 113747, 2008.
6. Hardie TL, Moss HB, Lynch KG. Genetic correlations between smok-
ing initiation and smoking behaviors in a twin sample. Addict Behav
31(11): 203037, 2006.
7. Morley KI, Medland SE, Ferreira MA, Lynskey MT, Montgomery GW,
HARM REDUCTION Heath AC, Madden PA, Martin NG. A possible smoking susceptibil-
ity locus on chromosome 11p12: Evidence from sex-limitation linkage
analyses in a sample of Australian twin families. Behav Genet 36(1):
Harm reduction is an alternative strategy for the smoker 8799, 2006.
who cannot, or will not, quit. It is designed to mitigate the 8. Arinami T, Ishiguro H, Onaivi ES. Polymorphisms in genes involved
medical complications of smoking without achieving full in neurotransmission in relation to smoking. Eur J Pharmacol 410(23):
abstinence. In theory, this could be achieved by pharmaco- 21526, 2000.
therapy with nicotine or other agents or by substitution of 9. Lerman CE, Schnoll RA, Munafo MR. Genetics and smoking cessation
improving outcomes in smokers at risk. Am J Prev Med 33(6 Suppl):
less toxic tobacco products [98]. A similar strategy may also
S398405, 2007.
reduce the exposure to secondhand smoke among children of 10. Henningeld JE, Miyasato K, Jasinski DR. Abuse liability and phar-
smokers. To date, however, there are no demonstrated health macodynamic characteristics of intravenous and inhaled nicotine.
benets of a harm reduction approach. Even if a harm reduc- J Pharmacol Exp Ther 234: 112, 1985.
tion approach were to benet individual smokers, the eect 11. Picciotto MR, Zoli M, Rimondini R, Lena C, Marubio LM, Pich EM,
of such interventions in a population is dicult to predict. Fuxe K, Changeux JP. Acetylcholine receptors containing the beta2
Some individuals may use reduction as a bridge to cessation. subunit are involved in the reinforcing properties of nicotine. Nature
Others may opt not to quit but to reduce. These uncertain- 391(6663): 17377, 1998.
ties have led to considerable debate and controversy. 12. Tapper AR, McKinney SL, Nashmi R, Schwarz J, Deshpande P,
Labarca C, Whiteaker P, Marks MJ, Collins AC, Lester HA. Nicotine
activation of 4* receptors: Sucient for reward, tolerance, and sensiti-
zation. Science 306(5698): 102932, 2004.
13. Olausson P, Jentsch JD, Taylor JR. Repeated nicotine exposure enhances
FUTURE DIRECTIONS responding with conditioned reinforcement. Psychopharmacology (Berl)
173(12): 98104, 2004.
14. Shiman S. Tobacco chippers individual dierences in tobacco
A number of novel smoking cessation pharmacotherapies dependence. Psychopharmacology (Berl) 97(4): 53947, 1989.
have followed in the wake of recent insights into nicotine 15. Diagnostic and Statistical Manual of Mental Disorders. American
addiction. Several cannabinoid receptor antagonists are in Psychiatric Association, Washington, D.C., 1994.
development. One such cannabinoid receptor antagonist, 16. The 2004 United States Surgeon Generals Report. The health conse-
rimonobant, demonstrated ecacy but did not lead to drug quences of smoking. NSW Public Health Bull 15(56): 107, 2004.
approval due to concerns of toxicity [45]. A nicotine vaccine 17. Yusuf S, Reddy S, Ounpuu S, Anand S. Global burden of cardiovascular
diseases: part I: General considerations, the epidemiologic transition,
that stimulates the production of antibodies against nico-
risk factors, and impact of urbanization. Circulation 104(22): 274653,
tine has demonstrated ecacy in Phase II trials [99]. Other 2001.
potential targets for smoking cessation drugs under investiga- 18. Reid RD, Quinlan B, Riley DL, Pipe AL. Smoking cessation: Lessons
tion include the -amino butyric acid receptor, the -opioid learned from clinical trial evidence. Curr Opin Cardiol 22(4): 28085,
receptor, and the 2 nicotinic receptor. 2007.
Finally, pharmacogenomics studies continue to cast 19. White WB. Smoking-related morbidity and mortality in the cardio-
light on the mechanisms behind variations in individual vascular setting. Prev Cardiol 10(2 Suppl 1): 14, 2007.

605
Asthma and COPD: Basic Mechanisms and Clinical Management

20. Berlin AA, Hogaboam CM, Lukacs NW. Inhibition of SCF attenu- transdermal nicotine delivery as an adjunct to low-intervention smok-
ates peribronchial remodeling in chronic cockroach allergen-induced ing cessation therapy. Arch Intern Med 151: 74952, 1991.
asthma. Lab Invest 86(6): 55765, 2006. 43. Silagy C, Lancaster T, Stead L, Mant D, Fowler G. Nicotine replace-
21. Lokke A, Lange P, Scharling H, Fabricius P, Vestbo J. Developing ment therapy for smoking cessation. Cochrane Database Syst Rev(3):
COPD: A 25 year follow up study of the general population. Thorax 2004, CD000146.
61(11): 93539, 2006. 44. Hughes JR, Stead LF, Lancaster T. Antidepressants for smoking cessa-
22. Yin P, Jiang CQ , Cheng KK, Lam TH, Lam KH, Miller MR, tion. Cochrane Database Syst Rev: 2007, CD000031.
Zhang WS, Thomas GN, Adab P. Passive smoking exposure and risk of 45. Cahilll K, Ussher M. Cannabinoid type 1 receptor antagonists (rimona-
COPD among adults in China: The Guangzhou Biobank Cohort Study. bant) for smoking cessation. Cochrane Database Syst Rev: 2007, CD005353.
Lancet 370(9589): 75157, 2007. 46. Etter JF, Stapleton JA. Nicotine replacement therapy for long-term
23. Rennard SI, Vestbo J. COPD: The dangerous underestimate of 15%. smoking cessation: A meta-analysis. Tob Control 15(4): 28085, 2006.
Lancet 367: 121619, 2006. 47. Ahluwalia JS, McNagny SE, Clark WS. Smoking cessation among
24. DiFranza JR, Aligne CA, Weitzman M. Prenatal and postnatal envi- inner-city African Americans using the nicotine transdermal patch.
ronmental tobacco smoke exposure and childrens health. Pediatrics J Gen Intern Med 13(1): 18, 1998.
113(4 Suppl): 100715, 2004. 48. Croghan GA, Sloan JA, Croghan IT, Novotny P, Hurt RD,
25. Gold DR, Wang X, Wypij D, Speizer FE, Ware JH, Dockery DW. DeKrey WL, Mailliard JA, Ebbert LP, Swan DK, Walsh DJ,
Eects of cigarette smoking on lung function in adolescent boys and Wiesenfeld M, Levitt R, Stella P, Johnson PA, Tschetter LK,
girls. New Engl J Med 335: 93137, 1996. Loprinzi C. Comparison of nicotine patch alone versus nicotine nasal
26. Wang X, Mensinga TT, Schouten JP, Rijcken B, Weiss ST. spray alone versus a combination for treating smokers: A minimal
Determinants of maximally attained level of pulmonary function. Am J intervention, randomized multicenter trial in a nonspecialized setting.
Respir Crit Care Med 169(8): 94149, 2004. Nicotine Tob Res 5(2): 18187, 2003.
27. Gilliland FD, Berhane K, McConnell R, Gauderman WJ, Vora H, 49. Fiore MC, Kenford SL, Jorenby DE, Wetter DW, Smith SS, Baker TB.
Rappaport EB, Avol E, Peters JM. Maternal smoking during pregnancy, Two studies of the clinical eectiveness of the nicotine patch with dif-
environmental tobacco smoke exposure and childhood lung function. ferent counseling treatments. Chest 105(2): 52433, 1994.
Thorax 55(4): 27176, 2000. 50. Tonnesen P, Paoletti P, Gustavsson G, Russell MA, Saracci R,
28. Alberg AJ, Ford JG, Samet JM. Epidemiology of lung cancer: ACCP Gulsvik A, Rijcken B, Sawe U. Higher dosage nicotine patches increase
evidence-based clinical practice guidelines (2nd edition). Chest 132(3 one-year smoking cessation rates: Results from the European CEASE
Suppl): 29S55S, 2007. trial. Collaborative European Anti-Smoking Evaluation. European
29. Peebles KA, Lee JM, Mao JT, Hazra S, Reckamp KL, Krysan K, Respiratory Society. Eur Respir J 13(2): 23846, 1999.
Dohadwala M, Heinrich EL, Walser TC, Cui X, Baratelli FE, 51. Yusuf S, Reddy S, Ounpuu S, Anand S. Global burden of cardiovascular
Garon E, Sharma S, Dubinett SM. Inammation and lung carcino- diseases: Part II: Variations in cardiovascular disease by specic ethnic
genesis: Applying ndings in prevention and treatment. Expert Rev groups and geographic regions and prevention strategies. Circulation
Anticancer Ther 7(10): 140521, 2007. 104(23): 285564, 2001.
30. Wu X, Zhao H, Suk R, Christiani DC. Genetic susceptibility to 52. GlaxoSmithKline Healthcare. Product Information: Commit Oral
tobacco-related cancer. Oncogene 23(38): 650023, 2004. Lozenge, Nicotine Polacrilex Oral Lozenge. Pittsburgh, PA, 2003.
31. MMWR. Reducing tobacco use. A report of the Surgeon General. 53. GlaxoSmithKline Healthcare. Product Information: Nicorette Gum,
Executive Summary. MMWR Recomm Rep 49: 127, 2000. Nicotine Polacrilex Gum. Moon Township, PA, 2005.
32. Gorini G, Chellini E, Galeone D. What happened in Italy? A brief 54. Henningeld JE, Radzius A, Cooper TM, Clayton RR. Drinking coee
summary of studies conducted in Italy to evaluate the impact of the and carbonated beverages blocks absorption of nicotine from nicotine
smoking ban. Ann Oncol 18(10): 162022, 2007. polacrilex gum. JAMA 264(12): 156064, 1990.
33. Quentin W, Neubauer S, Leidl R, Konig HH. Advertising bans as a 55. Mulligan SC, Masterson JG, Devane JG, Kelly JG. Clinical and phar-
means of tobacco control policy: A systematic literature review of time- macokinetic properties of a transdermal nicotine patch. Clin Pharmacol
series analyses. Int J Public Health 52(5): 295307, 2007. Ther 47(3): 33137, 1990.
34. MMWR. Cigarette smoking among adults United States, 2006. 56. Page F, Coleman G, Conduit R. The eect of transdermal nicotine
MMWR Morb Mortal Wkly Rep 56(44): 115761, 2006. patches on sleep and dreams. Physiol Behav 88(45): 42532, 2006.
35. Farrelly MC, Pechacek TF, Thomas KY, Nelson D. The impact of 57. Greenland S, Sattereld MH, Lanes SF. A meta-analysis to assess the
tobacco control programs on adult smoking. Am J Public Health 98(2): incidence of adverse eects associated with the transdermal nicotine
3049, 2008. patch. Drug Saf 18(4): 297308, 1998.
36. Hughes JR, Keely J, Naud S. Shape of the relapse curve and long-term 58. Hughes JR, Goldstein MG, Hurt RD, Shiman S. Recent advances in
abstinence among untreated smokers. Addiction 99(1): 2938, 2004. the pharmacotherapy of smoking [see comments]. JAMA 281(1): 72
37. Lancaster T, Stead L. Physician advice for smoking cessation. Cochrane 76, 1999.
Database Syst Rev: CD000165, 2004. 59. Hajek P, McRobbie H, Gillison F. Dependence potential of nicotine
38. Fiore MC. Treating tobacco use and dependence: An introduction to the replacement treatments: Eects of product type, patient characteristics,
US public health service clinical practice guideline. Respir Care 45(10): and cost to user. Prev Med 44(3): 23034, 2007.
119699, 2000. 60. Pzer Consumer Healthcare. Product Information: Nicotrol vs Nicotine
39. Mallin R. Smoking cessation: Integration of behavioral and drug ther- Nasal Spray. Morris Plains, NJ, 2005.
apies. Am Fam Physician 65(6): 110714, 2002. 61. Pzer Consumer Healthcare. Product Information: Nicotrol Inhalation
40. DiClemente CC, Prochaska JO, Fairhurst SK, Velicer WF, Velasquez Solution, Nicotine Inhalation Solution. Morris Plains, NJ, 2005.
MM, Rossi JS. The process of smoking cessation: An analysis of precon- 62. Joseph AM, Fu SS. Safety issues in pharmacotherapy for smoking in
templation, contemplation, and preparation stages of change. J Consult patients with cardiovascular disease. Prog Cardiovasc Dis 45(6): 42941,
Clin Psychol 59(2): 295304, 1991. 2003.
41. Rigotti NA, Munafo MR, Murphy MF, Stead LF. Interventions for 63. Joseph AM, Norma SM, Ferry LH. The safety of transdermal nicotine
smoking cessation in hospitalised patients. Cochrane Database Syst Rev(1): as an aid to smoking cessation in patients with cardiac disease. New
CD001837, 2003. Engl J Med 335: 179298, 1996.
42. Daughton DM, Heatley SA, Prendergast JJ, Causey D, Knowles M, 64. Tzivoni D, Keren A, Meyler S, Khoury Z, Lerer T, Brunel P.
Rolf CN, Cheney RA, Hatelid K, Thompson AB, Rennard SI. Eect of Cardiovascular safety of transdermal nicotine patches in patients with

606
Smoking Cessation 47
coronary artery disease who try to quit smoking. Cardiovasc Drugs Ther 84. Jorenby DE, Hays JT, Rigotti NA, Azoulay S, Watsky EJ,
12(3): 23944, 1998. Williams KE, Billing CB, Gong J, Reeves KR. Ecacy of varenicline,
65. Working Group for the Study of Transdermal Nicotine in Patients with an alpha4beta2 nicotinic acetylcholine receptor partial agonist, vs
Coronary Artery Disease. Nicotine replacement therapy for patients placebo or sustained-release bupropion for smoking cessation: A rand-
with coronary artery disease. Arch Intern Med 154: 98995, 1994. omized controlled trial. JAMA 296(1): 5663, 2006.
66. Ford CL, Zlabek JA. Nicotine replacement therapy and cardiovascular 85. Gonzales D, Rennard SI, Nides M, Oncken C, Azoulay S, Billing CB,
disease. Mayo Clin Proc 80(5): 65256, 2005. Watsky EJ, Gong J, Williams KE, Reeves KR. Varenicline, an alpha-
67. Dempsey DA, Benowitz NL. Risks and benets of nicotine to aid 4beta2 nicotinic acetylcholine receptor partial agonist, vs sustained-
smoking cessation in pregnancy. Drug Saf 24(4): 277322, 2001. release bupropion and placebo for smoking cessation: A randomized
68. Le Houezec J. What smoking cessation interventions are eective in controlled trial. JAMA 296(1): 4755, 2006.
pregnant women? J Gynecol Obstet Biol Reprod 1: 18293, 2005. 86. Nakamura M, Oshima A, Fujimoto Y, Maruyama N, Ishibashi T,
69. Drugdex evaluations retrieved January 18 from online micromedex data- Reeves KR. Ecacy and tolerability of varenicline, an alpha4beta2 nic-
base, Available from: URL: www.micromedex.com. Nicotine (2007). otinic acetylcholine receptor partial agonist, in a 12-week, randomized,
70. Dempsey D, Jacob P. III, Benowitz NL. Accelerated metabolism placebo-controlled, doseresponse study with 40-week follow-up for
of nicotine and cotinine in pregnant smokers. J Pharmacol Exp Ther smoking cessation in Japanese smokers. Clin Ther 29(6): 104056, 2007.
301(2): 59498, 2002. 87. Tsai ST, Cho HJ, Cheng HS, Kim CH, Hsueh KC, Billing CB Jr,
71. National Institute on Drug Abuse. Buproprion Helps People with Williams KE. A randomized, placebo-controlled trial of varenicline, a
Schizophrenia Quit Smoking. Research Findings 20(5), 2006. selective alpha4beta2 nicotinic acetylcholine receptor partial agonist, as
72. Stack NM. Smoking cessation: An overview of treatment options with a new therapy for smoking cessation in Asian smokers. Clin Ther 29(6):
a focus on varenicline. Pharmacotherapy 27(11): 155057, 2007. 102739, 2007.
73. Hurt RD, Sachs DP, Glover ED, Oord KP, Johnston JA, Dale LC, 88. Cahill K, Stead LF, Lancaster T. Nicotine receptor partial agonists for
Khayrallah MA, Schroeder DR, Glover PN, Sullivan CR, Croghan IT, smoking cessation. Cochrane Database Syst Rev(1): 2007, CD006103.
Sullivan PM. A comparison of sustained-release bupropion and pla- 89. Pzer Labs. Product Information: Chantix Varenicline Tablets. New York,
cebo for smoking cessation. New Engl J Med 337: 1195202, 1997. 2008.
74. Swan GE, McAfee T, Curry SJ, Jack LM, Javitz H, Dacey S, Bergman 90. Aubin HJ, Bobak A, Britton JR, Oncken C, Billing CB Jr, Gong J,
K. Eectiveness of bupropion sustained release for smoking cessation Williams KE, Reeves KR. Varenicline versus transdermal nicotine
in a health care setting: A randomized trial. Arch Intern Med 163(19): patch for smoking cessation: Results from a randomised, open-label
233744, 2003. trial. Thorax, 2008, doi:10.1136/thx.2007.090647
75. Hays JT, Hurt RD, Rigotti NA, Niaura R, Gonzales D, Durcan MJ, 91. Williams KE, Reeves KR, Billing CB Jr, Pennington AM, Gong J.
Sachs DP, Wolter TD, Buist AS, Johnston JA, White JD. Sustained- A double-blind study evaluating the long-term safety of varenicline for
release bupropion for pharmacologic relapse prevention after smoking smoking cessation. Curr Med Res Opin 23(4): 793801, 2007.
cessation. A randomized, controlled trial. Ann Intern Med 135(6): 423 92. Per Product Center Database, accessed 2/3/2008. Safety update to
33, 2001. chantix package insert; (2008). Available from: URL: https://www.
76. Killen JD, Fortmann SP, Murphy GM Jr, Hayward C, Arredondo C, Pzerpro.Com/product_info/chantix_isi_about_letter.Jsp
Cromp D, Celio M, Abe L, Wang Y, Schatzberg AF. Extended treat- 93. Cornuz J, Gilbert A, Pinget C, McDonald P, Slama K, Salto E,
ment with bupropion SR for cigarette smoking cessation. J Consult Clin Paccaud F. Cost-eectiveness of pharmacotherapies for nicotine depend-
Psychol 74(2): 28694, 2006. ence in primary care settings: A multinational comparison. Tob Control
77. Hurt RD, Krook JE, Croghan IT, Loprinzi CL, Sloan JA, Novotny PJ, 15(3): 15259, 2006.
Kardinal CG, Knost JA, Tirona MT, Addo F, Morton RF, 94. Hoogendoorn M, Welsing P, Rutten-van Molken MP. Cost-eectiveness
Michalak JC, Schaefer PL, Porter PA, Stella PJ. Nicotine patch therapy of varenicline compared with bupropion, NRT, and nortriptyline for
based on smoking rate followed by bupropion for prevention of relapse smoking cessation in the Netherlands. Curr Med Res Opin 24(1): 5161,
to smoking. J Clin Oncol 21(5): 91420, 2003. 2008.
78. GlaxoSmithKline Healthcare. Product Information: Zyban Buproprion 95. Faulkner MA, Lenz TL, Stading JA. Cost-eectiveness of smoking
Hydrochloride Sustained-release Tablets. Research Triangle Park, 2007. cessation and the implications for COPD. Int J Chron Obstruct Pulmon
79. Center for Drug Evaluation and Research US Food and Drug Dis 1(3): 27987, 2006.
Administration, 2008. Background information on the suicidality clas- 96. Fiore M, Bailey W, Cohen S, Dorfman S, Goldstein M, Gritz E,
sication project and buproprion; Available from: URL: http://www. Heyman R, Jaen C, Kottke T, Lando H, Mecklenburg R, Mullen P,
Fda.Gov/cder/drug/infopage/buproprion/default.Htm. Nett L, Robinson L, Sistzer M, Tommasello A, Villejo L, Wewers M.
80. Chun-Fai-Chan B, Koren G, Fayez I, Kalra S, Voyer-Lavigne S, Treating Tobacco Use and Dependence. Rockville, MD: U.S.
Boshier A, Shakir S, Einarson A. Pregnancy outcome of women Department Of Health and Human Services, 2000.
exposed to bupropion during pregnancy: A prospective comparative 97. Welton NJ, Johnstone EC, David SP, Munafo MR. A cost-eectiveness
study. Am J Obstet Gynecol 192(3): 93236, 2005. analysis of genetic testing of the DRD2 Taq1A polymorphism to aid
81. Gourlay SG, Stead LF, Benowitz NL. Clonidine for smoking cessation. treatment choice for smoking cessation. Nicotine Tob Res 10(1): 23140,
Cochrane Database Syst Rev(3): 2004, CD000058. 2008.
82. The FDA approves new drug for smoking cessation. FDA Consum 98. Clearing the Smoke. National Academy Press, Washington, D.C., 2001.
40:29, 2006. 99. Hatsukami DK, Rennard SI, Jorenby D, Fiore M, Koopmeiners J,
83. Coe JW, Brooks PR, Vetelino MG, Wirtz MC, Arnold EP, de Vos A, Horwith G, Pentel PR. Safety and immunogenicity of a
Huang J, Sands SB, Davis TI, Lebel LA, Fox CB, Shrikhande A, nicotine conjugate vaccine in current smokers. Clin Pharmacol Ther
Heym JH, Schaeer E, Rollema H, Lu Y, Mansbach RS, 78(5): 45667, 2005.
Chambers LK, Rovetti CC, Schulz DW, Tingley FD III, ONeill BT.
Varenicline: An alpha4beta2 nicotinic receptor partial agonist for
smoking cessation. J Med Chem 48(10): 347477, 2005.

607
2-Adrenoceptor Agonists
48 CHAPTER

2-adrenoceptor by stimulation of adenylyl Ian P. Hall


INTRODUCTION cyclase as a result of activation of the G pro-
tein coupled to the 2-adrenoceptor, Gs [4]. Division of Therapeutics and Molecular
2-Agonists have been the mainstay bronchodi- This exists as a heterotrimeric complex but fol- Medicine, University Hospital of
lator agents used for the treatment of asthma and lowing stimulation of the 2-adrenoceptor Gs Nottingham, Nottingham, UK
chronic obstructive pulmonary disease (COPD) dissociates releasing free -subunits that are
since the development of inhaled isoprenaline able to stimulate adenylyl cyclase. Adenylyl
(isoproterenol) preparations in the 1960s. While cyclase exists in a number of dierent isoforms
the initial preparations were marketed at rela- although there is at least some evidence suggest-
tively high doses and had little 2-selectivity, the ing that adenylyl cyclase VI is important in air-
side-eect prole of these agents was markedly way smooth muscle; however, most of the other
improved by the development of short-acting adenylyl cyclase isoforms are also present in this
2-selective agents such as salbutamol (albuterol) tissue [5]. Adenylyl cyclase catalyzes the forma-
and terbutaline. More recently, long-acting tion of cyclic AMP from adenosine triphos-
2-selective agents have assumed an increasingly phate (ATP). Cyclic AMP is able to convert
important role in the management of asthma protein kinase A from an inactive form to the
and COPD. active form in which the catalytic and regula-
tory subunits dissociate. The catalytic subunit of
protein kinase A then phosphorylates key tar-
gets within the cell bringing about the majority
of the physiological eects of 2-adrenoceptor
MECHANISM OF ACTION stimulation. However, there is at least some evi-
dence to suggest that cyclic AMP-independent
2-Agonists bind to the 2-adrenoceptor which actions may also occur; for example, direct stim-
is present in the cell membrane of a number ulation by Gs of the BKCa channel present in
of airway cells including airway smooth mus- the airway smooth muscle cell membrane has
cle, airway epithelial cells, inammatory cells
including mast cells, vascular and endothelium
and vascular smooth muscle [1]. However, the TABLE 48.1 Mechanisms underlying airway smooth
major site of action of 2-agonists in the airways muscle relaxation by 2 adrenoceptor agonists.
is the airway smooth muscle cell. Following Inhibition of spasmogen-induced inositol 1,4,5
binding of 2-agonist to the 2-adrenoceptor trisphosphate production
on airway smooth muscle, a signaling cascade is Inhibition of spasmogen-induced rises in intracellular
triggered which results in a number of events, free calcium
all of which contribute to relaxation of airway Activation of calcium-activated K channels
smooth muscle (Table 48.1) [2, 3]. Alteration of sensitivity of contractile apparatus
The majority of these events are depend- Increased extrusion/re-uptake of calcium from
ent on elevation of cell cyclic adenosine mono- cytoplasm
phosphate (AMP) content, which is brought Hyperpolarization of cell membrane
about following binding of 2-agonist to the

609
Asthma and COPD: Basic Mechanisms and Clinical Management

2 AGONIST
VII VI Phe 290

RECEPTOR
ACTIVATION
Ser 207
I V Ser 204
GDP

II III IV
Exocite
GDP binding

GTP Asp 73 Asp 113


Pi
FIG. 48.2 Cross-sectional view of transmembrane spanning domains of
the 2-adrenoreceptor core. The 2 agonist ligand is shown sitting in the
binding pocket in the receptor. Key amino acid residues for binding of
 ligand are shown. Amino acids in transmembrane domain IV are implicated
in salmeterol exocite binding.

GTP
TABLE 48.2 Frequently used 2-adrenoceptor agonists.

Short-acting 2-adrenoceptor agonists


DOWNSTREAM EFFECTS
Salbutamol (albuterol)*

FIG. 48.1 G-protein regulatory cycle of activation and deactivation for Terbutaline
transmission of the signal from receptor to effector. When GDP is bound,
the heterotrimeric G-protein is inactive, receptor stimulation causes Fenoterol
conformational change in both the receptor and the G-protein which
decreases GDP binding affinity. GTP is abundant in the cell and replaces Long-acting 2-adrenoceptor agonists
GDP; the active conformation of the G subunit dissociates from .
Salmeterol
This activated state remains until GTP is hydrolysed to GDP whereby the
subunits reassociate. Formoterol

* USA name.
been described [2]. This is relevant because the BKCa chan-
nel (the calcium activated potassium channel) is thought
to be important in modulating changes in cell membrane spanning domains (Fig. 48.2) [6, 7]. The binding site for 2-
potential following stimulation with 2-agonist, and thus agonists consists of residues in at least three of the -helices
can contribute to the relaxant response of 2-adrenoceptor that pass through the cell membrane. The prolonged duration
stimulation. of action of salmeterol is believed to be due to the binding
The intracellular eects of 2-adrenoceptor stimula- of the lipophilic tail to residues deep in the fourth trans-
tion are relatively short lived. Cyclic AMP is broken down membrane domain [8, 9]. This process is essentially irrevers-
by phosphodiesterase isoenzymes present in the cell, with ible. The explanation for the prolonged duration of action of
type 3 and type 4 phosphodiesterase activities believed to formoterol is less clear although it has been proposed that
be the most important in regulating cyclic AMP content because of its lipophilicity formoterol partitions in the cell
in airway smooth muscle and type 4 phosphodiesterase membrane which forms a reservoir allowing prolonged inter-
being physiologically important in many inammatory cells action with the receptor [10]. The most commonly used 2-
including eosinophils. Continued stimulation by Gs is agonists in clinical practice are listed in Table 48.2.
prevented by the free Gs rapidly reassociating with to All the clinically important 2-agonists consist of a
reconstitute the heterotrimeric Gs complex (Fig. 48.1). benzene ring with a chain of two carbon atoms and either
an amine head or a substituted amine head. If a hydroxyl
(OH) group is present at positions 3 or 4 on the benzene
ring, the structure is a catechol nucleus and hence the agent
STRUCTURE AND FUNCTIONAL a catecholamine. If these hydroxyl groups are substituted
RELATIONSHIPS OF 2-AGONISTS AND or repositioned, the drug is generally less potent than the
THE 2-ADRENOCEPTOR synthetic catecholamine isoprenaline, which is a full ago-
nist at 2-adrenoceptors. This potential disadvantage may
be outweighed by the relative resistance of substituted cat-
The 2-adrenoceptor is a member of the G protein coupled echolamines to metabolic degradation by the enzyme cat-
receptor superfamily with the typical seven transmembrane echol-O-methyltransferase (COMT). Examples of such

610
2-Adrenoceptor Agonists 48
agents are salbutamol and terbutaline with salbutamol only TABLE 48.3 2-agonists in asthma and COPD.
being a partial agonist. As mentioned above, the prolonged
Asthma COPD
duration of action of salmeterol is due to a long side chain
substitution which is believed to bind to an additional Bronchodilator response greater Bronchodilator response less
site in the fourth transmembrane spanning domain of the than 15% change in FEV1 than 15% change in FEV1
receptor. Substitutions on the carbon atom help block
oxidation by monoamine oxidase (MAO). Bronchodilator hyperreactivity Bronchial hyperreactivity usually
The eects of catecholamines such as adrenaline present absent
(epinephrine), noradrenaline (norepinephrine), and isopre-
2-agonist protects against
naline are terminated by uptake into either sympathetic
nonspecific airway challenge
nerve endings (uptake 1) or other innervated tissues such
as smooth muscle (uptake 2). The dominant enzyme present Marked symptomatic benefit with Small or moderate symptomatic
in innervated tissues is COMT whereas the dominant 2-agonist benefit with 2-agonist
metabolic degradation route in sympathetic nerve endings
is through oxidation by MAO. In addition to degradation, Effective in long-term management No disease-modifying effects
exogenously administered 2-agonists can be conjugated to
sulfates or glucuronides in the liver, or the lung. Following
ingestion the drugs are partially conjugated during rst past
metabolism which accounts for roughly 50% of the metabo- bronchiectasis, although the magnitude of these eects is
lism of the short-acting drug salbutamol. generally much smaller. In normal individuals bronchodi-
lator responses are generally only observable by measur-
ing specic airway conductance. This contrast between the
marked eects of 2-agonists in asthmatic patients and the
CLINICAL PHARMACOLOGY OF 2-AGONISTS minimal eects in normal individuals led early investigators
to hypothesize that a primary defect in the 2-adrenoceptor
signaling pathway was the actual cause of asthma. This
General pharmacology hypothesis would not explain many of the inammatory
features present in the disease, but interest in the potential
2-Adrenoceptor agonists are predominantly used in for a primary abnormality of 2-adrenoceptors to contribute
the treatment of airow obstruction because of their in part to the pathophysiology of asthma resurfaced fol-
bronchodilator properties. These dier markedly between lowing the description of polymorphic variation within the
asthma and COPD (Table 48.3); indeed, reversibility of 2-adrenoceptor (see below).
airow obstruction with inhaled 2-agonist is often used In addition to having a bronchodilator action,
as a diagnostic marker of asthma and helps distinguish 2-agonists in asthmatics protect against bronchoconstrictor
asthma from COPD in patients where the distinction is in stimuli [1417]. One feature demonstrated by most (but
doubt (e.g., chronic asthmatics who have smoked, or pre- not all) asthmatics is nonspecic airway hyperreactivity
vious smokers who develop symptoms of wheeze in later to inhaled irritant challenge including allergen. The most
life). However, in addition to reversing airow obstruction frequently used stimuli are histamine and methacholine
in asthmatic individuals 2-agonists also protect against although abnormal responses are also seen to exercise, aller-
bronchoconstrictor challenge [11]. Because of this latter gen, and other challenges. The inhaled dose of histamine
eect, 2-agonists have been considered to have potential and other agents which required to provoke a 20% fall in
anti-inammatory actions (see below). In vitro 2-agonists FEV1 (PC20) is increased markedly (usually by about three
prevent mediator release from inammatory cells includ- doubling doses) by previous treatment with a short-acting
ing mast cells, an eect which if present in human airways 2-agonist. While a degree of such bronchial hyperreactivity
would be expected to reduce airway inammation [12, 13]. can also be demonstrated in other diseases including COPD,
However, the concentrations of these agents required to bronchiectasis, cystic brosis, and left ventricular failure, this
demonstrate these eects are in general much higher than is usually far less marked than in asthmatic subjects.
those seen in the lungs in vivo.

SHORT-TERM EFFECTS OF 2-AGONISTS LONG-TERM EFFECTS OF 2-AGONISTS


IN ASTHMA AND COPD IN ASTHMA AND COPD
As mentioned above, the ability of 2-adrenoceptor ago- While regular treatment of inhaled or nebulized 2-agonist
nists to reverse airow obstruction is a hallmark of asthma, is frequently used in the management of moderate or severe
with reversibility 15% being considered diagnostic. 2- COPD, despite relatively small improvements in symptoms,
Agonists do produce a measurable bronchodilator eect controversy has reigned over the long-term use of regular
in normal individuals and in patients with other diseases 2-agonist in the management of asthma [18, 19]. This con-
characterized by airow obstruction such as COPD and troversy originated from discussions following epidemics of

611
Asthma and COPD: Basic Mechanisms and Clinical Management

asthma deaths in New Zealand in the late 1960s and late and symptomatic improvement have been demonstrated in
1970s which were linked to the prescribing of high-dose severe COPD, the overall eect on lung function has gen-
isoprenaline and fenoterol, respectively. Several studies sub- erally been small, which is hardly surprising given the xed
sequently concentrated on the possibility that tachyphylaxis nature of the airow obstruction in the majority of patients.
may develop as a result of inhaled 2-agonists in asthma. In There have been no data suggesting deterioration in lung
general, studies attempting to demonstrate tachyphylaxis function following chronic administration of high doses of
to the bronchodilator eects of 2-agonists have failed to 2-agonist in COPD (these would be dicult to observe
identify clinically important loss of responsiveness although in short-term studies in any case) although there have been
small eects have been observed when looked for carefully concerns about other eects of high doses of 2-agonist in
(see for example, Ref. [20]). However, in contrast, the bron- this setting. In particular, high-dose nebulized 2-agonists
choprotective eects of 2-agonists against nonspecic air- are known to cause hypokalaemia and both supraventricu-
way challenge show tachyphylaxis that generally develops lar and ventricular arrhythmias [27], and there have been
within 24 h [21, 22]. Thus, the magnitude of the protective concerns that these may occur in patients on regular high-
eect against exercise, histamine challenge, or methacholine dose nebulized bronchodilator therapy for either asthma or
challenge is reduced in magnitude compared with the level COPD at home.
of initial protection following administration of 2-agonist
for periods of over 24 h. However, there is still overall pro-
tection against bronchoconstrictor challenge even if this is
less than the protection seen in the initial hours after treat-
ment. Thus, while it seems clear that tachyphylaxis develops 2-ADRENOCEPTOR POLYMORPHISM
to the bronchoprotective eects of 2-agonists against non- IN ASTHMA AND COPD
specic airway challenge, this is only partial, and one would
presume that patients would still be better despite this tach-
yphylaxis than if they were not taking 2-agonist at all. The As mentioned above, the identication of polymorphic
main concern therefore has revolved around patients taking variation within the gene for the 2-adrenoceptor reawak-
intermittent treatment or in those discontinuing treatment. ened interest in the possibility that primary abnormalities
Small rebound increases in airway reactivity have been dem- of 2-adrenoceptor signaling pathways may be involved
onstrated following the cessation of 2-agonist therapy [23]. in the pathogenesis of these airway diseases. The gene for
Clinical studies comparing regular (e.g., four times a day) the 2-adrenoceptor is situated on chromosome 5q31-33
short-acting 2-agonist versus as required 2-agonist have in a region showing linkage to intermediate phenotypes
not shown clinically important dierences despite earlier for asthma and/or atopy [28]. The 2-adrenoceptor gene
reports that asthma control deteriorated following regular and its immediate controlling regions show a high degree
treatment [24, 25]. None the less, there is no reason to sup- of polymorphic variation with nine single nucleotide type
pose that regular treatment is better than as required usage polymorphisms (SNPs) having been identied within the
which remains the preferred way of prescribing 2-agonists coding region of the gene and a further eight in the imme-
in asthma. diate 5 prime untranslated region (reviewed in Ref. [29]).
The controversy regarding 2-agonists resurfaced Of the nine coding region polymorphisms, ve are degen-
with the introduction into the marketplace of long-act- erate (i.e. do not alter the amino acid code of the recep-
ing 2-agonists (LABAs). There were initial concerns that tor) [30]. However, the other four all result in single amino
LABAs might worsen asthma control in asthmatics in gen- acid substitutions. While the polymorphism at codon 34
eral, but most asthmatics respond well to LABAs. At least (Val34Met) is rare and appears to have no functional eects,
for salmeterol, this might initially seems surprising, given the other three nondegenerate polymorphisms appear to
the fact that salmeterol is a partial agonist and essentially produce functional alteration in receptor behavior. Thus, the
binds irreversibly with the 2-adrenoceptor. The issue of rare Thr164Ile polymorphism results in reduced anity for
LABA safety has recently resurfaced, following a large study catechol ligands and an altered receptor sequestration pro-
examining ecacy of salmeterol in asthma (the SMART le [31]. Interestingly, Thr164 is very close to the salmeterol
study [26]). In this study, there were a small number of binding site within the fourth transmembrane spanning
deaths and severe exacerbations in the salmeterol treatment domain of the receptor, and it seems likely that the isoleu-
group which led to premature termination of the study and cine 164 substitution may alter the binding characteristics
a subsequent black box warning from the FDA which was of salmeterol to the 2-adrenoceptor. However, the allelic
later extended to all LABAs. The explanation for these nd- frequency of this polymorphism is only around 23% in
ings is unclear, although it is possible that reduced compli- Caucasian populations, hence homozygous individuals are
ance with inhaled corticosteroids may have played a role. very rare and to date have not been adequately studied.
The other commonly used LABA, formoterol, has under- In contrast, the two N-terminal polymorphisms at
gone recent studies which has led to a license in the UK for codon 16 (Arg16Gly) and 27 (Gln27Glu) are common.
use as an as required agent (in a combination inhaler with While neither alter agonist binding properties of the receptor,
inhaled budesonide). both result in altered downregulation proles following long-
Although mild COPD is often treated with as term agonist exposure. Thus, the Gly16 and Gln27 forms of
required 2-agonist, regular high-dose inhaled or nebulized the receptor show increased receptor downregulation follow-
2-agonists have been much more widely used in COPD ing agonist exposure while the Glu 27 form of the recep-
than in asthma. While small improvements in lung function tor appears to be partially protected from downregulation

612
2-Adrenoceptor Agonists 48
[9]. These eects have been shown both in transformed cell References
systems and in primary cultures of human airways smooth
muscle. Of the 5 prime untranslated region polymorphisms, 1. Davis C, Conolly ME, Greenacre JK. Beta-adrenoceptors in human
the strongest evidence for possible functional eects is for lung, bronchus and lymphocytes. Br J Clin Pharm 10: 425, 1980.
the 47 T-C SNP which alters the terminal amino acid in 2. Kume H, Hall IP, Washabow RJ, Takagi K, Kotliko MI. -Adrenergic
a short open reading frame that codes for the -upstream agonists regulate KCa channels in airway smooth muscle cAMP
peptide (also known as the 2-adrenoceptor 5 prime leader dependent mechanisms. J Clin Invest 93: 371, 1994.
cistron); this peptide is believed to be important in maintain- 3. Torphy TJ, Hall IP. Cyclic AMP and the control of airways smooth
ing a brake on receptor expression, and the Cys19Arg poly- muscle tone. In: Raeburn D, Giembycz MA (eds). Airways Smooth
Muscle: Biochemical Control of Contraction and Relaxation, p. 215. Basel:
morphism in this peptide may possibly increase translational
Birkhauser Verlag, 1994.
inhibition of the 2-adrenoceptor [32, 33]. 4. Johnson M. The -adrenoceptor. Am J Respir Crit Care Med 158: S146,
The role of 2-adrenoceptor polymorphism has been 1999.
extensively studied in asthma but less so in COPD. A 5. Billington CK, Hall IP, Stuart J, Mundell JLP et al. Inammatory
recent major study in UK 1958 birth cohort [34] failed to and contractile agents sensitize specic adenylyl cyclase isoforms in
demonstrate an association between 2-adrenoceptor poly- human airway smooth muscle. Am J Respir Cell Mol Biol 21: 597606,
morphism and asthma per se, although weak association 1999.
with IgE and the degree of bronchial hyperresponsiveness 6. Dohlman HG, Bouvier M, Benovic JL, Caron MG, Lefkowitz RJ. The
have previously been described in some (but not in all) stud- multiple membrane spanning topography of the 2-adrenergic receptor.
ies [35]. These eects may, in part, be due to linkage dis- J Biol Chem 262: 14282, 1987.
7. Emorine J, Marullo S, Delavier-Klutchko C, Kaveri SV, Duriev-
equilibrium with other important genes on chromosome 5q
Trautmann O, Strosberg AD. Structure of the gene for human 2-
such as the nearby Th2 cytokine locus. The possibility that adrenergic receptor: Expression and promoter characterisation. Proc
these polymorphisms may contribute to the development of Natl Acad Sci USA 84: 6995, 1987.
COPD has not been reported upon to date, although stud- 8. Green SA, Spaso AP, Coleman RA, Johnson M, Liggett SB.
ies in this area are currently in progress. Sustained activation of a G protein-coupled receptor via anchored
The other potential importance of 2-adrenoceptor agonist binding: Molecular localisation of the salmeterol exocite within
polymorphism is in pharmacogenetic studies. The possibil- the 2-adrenergic receptor. J Biol Chem 271: 24029, 1996.
ity that 2-adrenoceptor polymorphism may predict treat- 9. Green SA, Turki J, Bejarano P, Hall IP, Liggett SB. Inuence of 2-
ment response, particularly following long-term exposure to adrenergic receptor genotypes on signal transduction in human airway
agonists due to the altered downregulation prole of indi- smooth muscle cells. Am J Respir Cell Mol Biol 13: 25, 1995.
10. Lofdahl CG, Svedmyr N. Formoterol fumarate, a new 2-adrenoceptor
viduals carrying particular genotypes (e.g., Gly16, Gln27),
agonist. Allergy 44: 264, 1989.
has been studied by a number of groups. Initial studies dem- 11. Tatterseld AE. Eect of beta agonists and anticholinergic drugs on
onstrated an association between Gly16 and subsensitivity bronchial reactivity. Am Rev Respir Dis 136: S64, 1987.
to the bronchodilator eects of formoterol following chronic 12. Assem ESK, Schild HO. Inhibition by sympathomimetic amines of
dosing and also a reduced bronchodilator response to salb- histamine release induced by antigen in passively sensitized human
utamol [20, 36]. Gly16 has also been shown to be associ- lung. Nature 224: 1028, 1969.
ated with nocturnal asthma. Two relatively large studies, 13. Howarth PH, Durham SR, Lee TH, Kay AB, Church MK, Holgate
one retrospective and one prospective, suggested that treat- ST. Inuence of albuterol, cromolyn sodium and ipratropium bromide
ment with regular (four times a day) salbutamol (albuterol) on the airway and circulating mediator responses to allergen bronchial
resulted in poorer outcome in Arg16Arg asthmatics (15% provocation in asthma. Am Rev Respir Dis 132: 986, 1985.
14. Cheung D, Timmers MC, Zwinderman AH, Bel EH, Dijkman JH,
of the Caucasian population) [37, 38]. However, salbutamol
Sterk PJ. Long-term eects of a long-acting 2-adrenoceptor agonist,
is not currently used in most countries in this way, and the salmeterol, on airway hyperresponsiveness with mild asthma. N Engl J
asthmatics studied were not on inhaled corticoseroids, so Med 327: 1198, 1992.
the clinical relevance of these observations is unclear. More 15. Twentyman OP, Finnerty JP, Harris A, Palmer J, Holgate ST.
recently, a small retrospective study suggested that these Protection against allergen-induced asthma by salmeterol. Lancet 336:
eects may extend to LABAs [39], although subsequently 1338, 1990.
a larger retrospective analysis of pharma-funded studies 16. Twentyman OP, Finnerty JP, Holgate ST. The inhibitory eect of neb-
suggests that this does not extend to patients on combined ulized albuterol on the early and late asthmatic reactions and increase
LABA/inhaled steroid preparations [40]. In general, there- in airway responsiveness provoked by inhaled allergen in asthma.
fore, it appears that the eects of these polymorphisms upon Am Rev Respir Dis 144: 782, 1991.
17. Wong BJ, Dolovich J, Ramsdale EH et al. Formoterol compared with
treatment response to these drugs used as recommended in
beclomethasone and placebo on allergen-induced asthmatic responses.
national guidelines are likely to be relatively small and of Am Rev Respir Dis 146: 1156, 1992.
doubtful clinical signicance overall. It is also possible that 18. Sears MR, Taylor DR, Print CG et al. Regular inhaled -agonist treat-
the combination of groups of polymorphisms around this ment in bronchial asthma. Lancet 336: 1391, 1990.
region (i.e. the haplotype of an individual at this locus) may 19. Taylor DR, Sears MR, Herbison GP et al. Regular inhaled -agonist
be the main determinant of functional eects rather than in asthma: Eects on exacerbations and lung function. Thorax 48: 134,
single polymorphisms in isolation [41]. It will be interesting 1993.
to determine whether these polymorphisms are relevant to 20. Tan S, Hall IP, Dewar J, Dow E, Lipworth B. Association between 2-
treatment response in COPD; intuitively, one might imag- adrenoceptor polymorphism and susceptibility to bronchdilator desen-
ine that this could be the case given the higher doses of sitisation in moderately severe stable asthmatics. Lancet 350: 995, 1997.
21. Connor BJ, Aikman SL, Barnes BJ. Tolerance to the nonbronchodilator
agents used in COPD although, to date, no published data
eects of inhaled 2-agonists in asthma. N Engl J Med 327: 1204, 1992.
are available on this issue.

613
Asthma and COPD: Basic Mechanisms and Clinical Management

22. Ramage L, Lipworth BJ, Ingram CG, Cree IA, Dhillon DP. Reduced 34. Hall IP, Blakey JD, Al Balushi KA, Wheatley A, Sayers I, Pembrey
protection against exercise-induced bronchoconstriction after chronic ME, Ring SM, McArdle WL, Strachan DP. Beta2-adrenoceptor poly-
dosing with salmeterol. Respir Med 88: 363, 1994. morphisms and asthma from childhood to middle age in the British
23. Wahedna I, Wong CS, Wisniewski AFZ, Pavord ID, Tatterseld AE. 1958 birth cohort: A genetic association study. Lancet 368: 77179,
Asthma control during and after cessation of regular 2-agonist treat- 2006.
ment. Am Rev Respir Dis 148: 707, 1993. 35. Dewar JC, Wilkinson J, Wheatley A et al. The glutamine 27
24. Drazen JM, Israel E, Boushey HA et al. The National Heart, Lung, 2-adrenoceptor polymorphism is associated with elevated IgE levels
and Blood Institutes Asthma Clinical Research Network Comparison in asthmatic families. J Allergy Clin Immunol 100: 261, 1997.
of regularly scheduled with as-needed use of albuterol in mild asthma. 36. Martinez FD, Graves PE, Baldini M, Solomon S, Erickson R.
N Engl J Med 335: 841, 1996. Association between genetic polymorphisms of the 2-adrenoceptor
25. van Schayck CP, Dompeling E, van Herwaarden CLA et al. and response to albuterol in children with and without a history of
Bronchodilator treatment in moderate asthma or chronic bronchitis: wheezing. J Clin Invest 100: 3184, 1997.
Continuous or on demand? A randomised controlled study. BMJ 303: 37. Israel E, Drazen JM, Liggett SB, Boushey HA, Cherniack RM,
1426, 1991. Chinchilli VM, Cooper DM, Fahy JV, Fish JE, Ford JG, Kraft M,
26. Nelson HS, Weiss ST, Bleecker ER, Yancey SW, Dorinsky PM Kunselman S, Lazarus SC, Lemanske RF, Martin RJ, McLean DE,
SMART Study Group. The Salmeterol Multicenter Asthma Research Peters SP, Silverman EK, Sorkness CA, Szeer SJ, Weiss ST, Yandava
Trial: A comparison of usual pharmacotherapy for asthma or usual CN. The eect of polymorphisms of the beta(2)-adrenergic receptor
pharmacotherapy plus salmeterol. Chest 129: 1526, 2006. on the response to regular use of albuterol in asthma. Am J Respir Crit
27. Wong CS, Pavord ID, Williams J, Britton JR, Tatterseld AE. Care Med 162: 7580, 2000.
Bronchodilator, cardiovascular, and hypokalaemic eects of fenoterol, 38. Israel E, Chinchilli VM, Ford JG, Boushey HA, Cherniack R, Craig
salbutamol, and terbutaline in asthma. Lancet 336: 1396, 1990. TJ, Deykin A, Fagan JK, Fahy JV, Fish J, Kraft M, Kunselman SJ,
28. Kobilka BK, Dixon RAF, Frielle T et al. cDNA for the human 2- Lazarus SC, Lemanske RF Jr., Liggett SB, Martin RJ, Mitra N,
adrenergic receptor: A protein with multiple membrane-spanning Peters SP, Silverman E, Sorkness CA, Szeer SJ, Wechsler ME,
domains and encoded by a gene whose chromosomal location is shared Weiss ST, Drazen JM National Heart, Lung, and Blood Institutes
with that of the receptor for platelet-derived growth factor. Proc Natl Asthma Clinical Research Network. Use of regularly scheduled
Acad Sci USA 84: 46, 1987. albuterol treatment in asthma: Genotype-stratied, randomised, pla-
29. Fenech A, Hall IP. Pharmacogenetics of asthma. Br J Clin Pharmacol cebo-controlled cross-over trial. Lancet 364: 150512, 2004.
53: 315, 2005. 39. Wechsler ME, Lehman E, Lazarus SC, Lemanske RF Jr., Boushey
30. Reihsaus E, Innis M, MacIntyre N, Ligghett SB. Mutations in the HA, Deykin A et al. Beta-Adrenergic receptor polymorphisms
gene encoding for 2-adrenergic receptor in normal and asthmatic sub- and response to salmeterol. Am J Respir Crit Care Med 173: 51926,
jects. Am J Respir Cell Mol Biol 8: 334, 1993. 2006.
31. Green SA, Cole G, Jacinto M, Innis M, Liggett SB. A polymorphism 40. Bleecker ER, Yancey SW, Baitinger LA, Edwards LD, Klotsman M,
of the human 2-adrenergic receptor within the fourth transmembrane Anderson WH, Dorinsky PM. Salmeterol response is not aected by
domain alters ligand binding and functional properties of the receptor. beta2-adrenergic receptor genotype in subjects with persistent asthma.
J Biol Chem 268: 23116, 1993. J Allergy Clin Immunol 118: 80916, 2006.
32. McGraw DW, Forbes SL, Kramer LA, Liggett SB. Polymorphisms 41. Drysdale CM, McGraw DW, Stack CB et al. Complex promoter and
of the 5
leader cistron of the human 2-adrenergic receptor regulate coding region 2-adrenergic receptor haplotypes alter receptor expres-
receptor expression. J Clin Invest 102: 1927, 1998. sions and predict in vivo responsiveness. Proc Natl Acad Sci USA 97:
33. Scott MG, Swan C, Wheatley AP, Hall IP. Identication of novel pol- 1048388, 2000.
ymorphisms within the promoter region of the human 2-adrenergic
receptor gene. Br J Pharmacol 126: 84144, 1999.

614
Anticholinergic Bronchodilators
49 CHAPTER

INTRODUCTION basis by the Global Obstructive Lung Disease


(GOLD) guidelines (2007) for moderate (Stage
Trevor T. Hansel, Andrew
II), severe (Stage III), and very severe (Stage J. Tan, Peter J. Barnes
The major prescribed long-acting muscarinic IV) COPD to prevent or reduce symptoms, and Onn Min Kon
antagonist (LAMA) is now tiotropium bromide improve exercise performance, and decrease
(Spiriva) that is a once daily inhaled therapy for exacerbations [1, 2] (Fig. 49.1). Tiotropium has National Heart and Lung Institute
symptomatic management of chronic obstruc- been the subject of a number of recent reviews (NHLI), Clinical Studies Unit, Imperial
tive pulmonary disease (COPD). Indeed, tio- that address the clinical development proc- College, London, UK
tropium is recommended on a regular daily ess from research to clinical practice, and their

Stage I II III IV

Post-bronchodilator
Mild Moderate Severe Very severe
FEV1
80% 5080% 3050% 30%
(% predicted)

Avoidance of risk factors SMOKING CESSATION


Influenza vaccination

Short-acting bronchodilator if needed

Add regular treatment with one or more long-acting


bronchodilators, including tiotropium
Pulmonary rehabilitation

Add regular treatment with inhaled


corticosteroids if repeated exacerbations

Based on GOLD 2007


www.goldcopd.com Long-term oxygen
therapy (LTOT) if
respiratory failure
Consider surgical
options

FIG. 49.1 Treatment of COPD at different GOLD stages.

615
Asthma and COPD: Basic Mechanisms and Clinical Management

place in the management of patients with COPD [37].


Certain articles have centered on review and meta-analy- PLANT ORIGINS OF ANTICHOLINERGICS
ses of experience in clinical studies [810]. This chapter
will consider the pharmacology, biochemistry, and potential Herbal remedies have been used since ancient times to treat
anti-inammatory actions of anticholinergic bronchodila- human disease; and the leaves, roots, and seeds of many plants
tors, including possible anti-inammatory actions, as well contain anticholinergic agents [1116] (Fig. 49.2). Ayurvedic
as clinical trial evidence for the safety and ecacy of these medicine sources from India at around 450 ad describe the use
therapies. Important outcome measures for LAMAs involve of Datura stramonium (thorn-apple or jimson weed) extract for
considerations of eects on symptoms, lung function, the relief of asthma. Atropine and scopolamine represent par-
dynamic hyperination and exercise responses, quality of ent anticholinergic agents, and are alkaloids found in Datura
life, health economics, exacerbations, survival, and the natu- species as well as Atropa belladonna (deadly nightshade). After
ral history of COPD. We shall then discuss clinical trials serving in India, General Gent in 1802 recommended the
that have assessed combined inhaled LAMAs and LABAs, smoking of Datura to relieve bronchial complaints.
as well as combinations of LAMAs, LABAs, and ICSs. In the last century, plant extracts have largely been
Finally, we shall look to the future with novel LAMAs such replaced by synthetic chemicals, and in the late 1970s
as glycopyrrolate and aclidinium bromide (Almirall).

(A) (B) (C)

FIG. 49.2 Botanical origin of anticholinergics. (A) The black berries of Atropa belladonna (deadly nightshade) contain the deadly plant alkaloid, atropine.
Atropine is a competitive antagonist to acetylcholine at muscarinic receptor sites, M1, M2, and M3. Atropine, when taken orally, causes blocked vagal
innervation of the heart (tachycardia), decreased mucus secretion and bronchial smooth muscle relaxation in the respiratory system, mydriasis (pupillary
dilation), decreased salivation with dry mouth, decreased gut motility and acid secretion, bladder wall relaxation, and decreased sweating. (B) Hyosyamus
niger is the plant source of scopolamine (hyosine) that was employed by the notorious Dr Crippen to murder his wife in London in 1910. (C) Datura
stramonium (thorn-apple or jimsonweed) is the source of stramonium which was inhaled to alleviate respiratory disorders. These images are taken from the
Wikimedia Commons, free public domain.

Br Br
CH3  
N CH3 N C3H7 CH3 N C3

H
O
O C C O C C
O
O CH2OH O
O CH2OH
Atropine Ipratropium bromide

O H
S

Tiotropium bromide S

The plant alkaloids are represented by atropine. Ipratropium and tiotropium are synthetic anticholinergic agents that contain a positively
charged quaternary nitrogen moiety (highlighted in green). This ensures local action following inhalation, without systemic absorption.

FIG. 49.3 Structure of anticholinergics (see text on figure).

616
Anticholinergic Bronchodilators 49
short-acting synthetic anticholinergic drugs were intro- this leads to an increase in acetylcholine release and this may
duced: ipratropium bromide (4 times a day) and oxitropium work against the post-junctional blockade of M3-receptors,
bromide (3 times a day). These have largely been superseded making these antagonists less ecient. It has been dicult
by tiotropium bromide (Spiriva) that is administered once to develop M3-selective antagonists [23].
daily from the HandiHaler and was licensed in most of the Tiotropium bromide is a quaternary ammonium
European Union in 2002, while a license in the USA was derivative like ipratropium bromide and is a long-acting
obtained in 2003. anticholinergic drug that has unique kinetic selectivity for
M1 and M3 muscarinic receptors [17, 24, 25]. Tiotropium
has similar anity for the ve types of muscarinic recep-
MOLECULAR STRUCTURES tors, M1 to M5. In the airways, tiotropium has a high anity
and dissociates very slowly from M1- and M3-receptors in
human lung and produces long-term blockade of cholinergic
The parent compounds for anticholinergic therapy are neural bronchoconstriction in human airway smooth muscle,
the plant alkaloids atropine and scopolamine (Fig. 49.3). but much more rapidly from M2-receptors giving a unique
Atropine methyl nitrate, ipratropium bromide, oxitropium kinetic selectivity for M1- and M3-receptors [26, 27].
bromide, and tiotropium bromide share a quaternary ammo-
nium structure [1719]. Quaternary ammonium compounds
are poorly absorbed from respiratory or pharyngeal mucosal
surfaces, and they have minimal oral bioavailability, ensuring
that systemic anticholinergic eects are kept to a minimum. ANTI-INFLAMMATORY ACTIONS

There has been major recent progress in understanding


MUSCARINIC RECEPTOR PHARMACOLOGY cholinergic systems mediated by muscarinic acetylcholine
receptors [2830] as well as nicotinic pathways [31] (Fig.
49.4). In particular, lymphocytes have been found to express
Anticholinergics are muscarinic receptor antagonists, inhib- a variety of cholinergic receptors [32], and acetylcho-
iting cholinergic reex bronchoconstriction and reducing line/muscarinic receptor signaling involving epithelial and
vagal cholinergic tone, which is the main reversible com- inammatory cells has been documented in both asthma and
ponent in COPD. Normal airways have a small degree of COPD. Interestingly, anticholinergic may benet asthmat-
vagal cholinergic tone, but because the airways are patent ics with COPD-like inammation in asthma, viral hyper-
this has no perceptible eect and does not reduce airow. reactivity [33], and emergency asthma [34]. In guinea pigs
When the airways are irreversibly narrowed in COPD, tiotropium inhibits allergen-induced airway remodeling [35,
vagal cholinergic tone has a much greater eect on airway 36], while muscarinic receptors mediate lung broblasts pro-
resistance for geometric reasons, so when this increased liferation [37]. Tiotropium has been shown to suppress ace-
constriction is relieved by an anticholinergic drug, there is a tylcholine-induced release of chemotactic mediators in vitro
perceptible improvement in airow. In addition, anticholin- with a special activity on LTB4 [38]. A recent clinical study
ergics may reduce mucus hypersecretion [20] (Fig. 49.4). did not show an anti-inammatory eect on sputum inam-
Anticholinergics have no apparent eect on pulmonary ves- matory markers during exacerbations of COPD [39], but
sels, and therefore do not cause a fall in Pao2, as may some- this study was awed by the methodology of detection of
times be seen with 2-agonists and theophylline. sputum IL-6 and IL-8. This was because the supernatants
The existence of several subtypes of muscarinic recep- were formed after liquefaction of sputum with dithiothreitol,
tor in human airways has suggested that more selective and this agent can denature cytokines and thus lower detect-
muscarinic antagonists may have advantages over non- able levels on immunoassay.
selective agents, such as ipratropium bromide and oxitro-
pium bromide [21, 22] (Fig. 49.4).
M1-receptors are localized to parasympathetic
ganglia, and their blockade results in reduced reex
bronchoconstriction. TIOTROPIUM DELIVERY
M2-receptors located at cholinergic nerve terminals
inhibit the release of acetylcholine, thus acting as Tiotropium is licensed in Europe as a bronchodilator for the
autoreceptors. maintenance treatment of COPD but may have a role in the
chronic treatment of severe asthma, as well as in acute severe
M3-receptors mediate the bronchoconstrictor and mucus
emergency room asthma. Tiotropium is present in capsules
secretion action of acetylcholine in human airways. The
containing 22.5 g tiotropium bromide monohydrate equiv-
most important eects of anticholinergics appear to be
alent to 18 g tiotropium. These capsules are administered in
mediated through blockage of M3-receptors.
the HandiHaler, a specialized dry powder inhalation system
Non-selective anticholinergics, such as atropine and [40]. As with all inhaled therapy, attention to eective drug
ipratropium bromide, act on M1-, M2-, and M3-receptors delivery and training in inhaler technique is essential. COPD
[17]. Blockage of M1- and M3-receptors leads to bronchodil- patients may have diculty in coordinating a Metered Dose
atation. However, by blocking pre-junctional M2-receptors Inhaler (MDI), and breath-activated Dry Powder Inhalers

617
Asthma and COPD: Basic Mechanisms and Clinical Management

Pre-ganglionic
Neural cholinergic nerve

muscarinic
receptors Parasympathetic
ganglion ACh
N ()
M1 ()

Post-ganglionic
cholinergic nerve
M2 ()


Neuromuscular
junction

Airway smooth ACh


muscle M3 ()

Non-neuronal
effector cells Central nervous system

Nodose
ganglion

Laryngeal
oesophageal
afferents Vagus nerve

Airway wall ACh


A-fibre Parasympathetic
C-fibre ganglion

Submucosal
? gland
C-fibre ACh
receptors ACh
Irritant ? ACh
receptors
Inflammatory cells Airway smooth muscle
and Fibroblasts
ACh

Irritants Airway epithelium


(e.g. cigarette smoke)

The mechanism of vagally mediated reflex bronchoconstriction is induced by irritants and


non-specific stimuli acting on irritant receptors. Pre- and postganglionic parasympathetic
nerves release acetylcholine (ACh). Preganglionic nervefibers from the vagus nerve relay
in parasympathetic ganglia in the airway wall, and short postganglionic pathways release
ACh to cause bronchoconstriction and mucus secretion. Cholinergic muscarinic receptors
are present on inflammatory cells, airway smooth muscle, fibroblasts, submucosal gland
cells, and epithelial cells. The role of cholinergic receptors on inflammatory cells and
inflammatory mediators remains controversial (?). FIG. 49.4 Cholinergic control of airways (see text
For details see: Gosens R et al. Respir Res 2006; 7:73 doi: 10.1186/1465-9921-7-73 on figure).

(DPI) have advantages in elderly subjects. HandiHaler with concomitant clinical eect at 20 l/min. The delivered
requires low inspiratory ow rates to evacuate the cap- dose that leaves the HandiHaler is 10 g. The contents of
sule and so can be used by patients with severe COPD. one capsule of tiotropium bromide should be inhaled once
The capsule begins to vibrate at 15 l/min and evacuates daily with the HandiHaler at the same time of the day.

618
Anticholinergic Bronchodilators 49
Patient performance with the HandiHaler was found in a In contrast, ipratropium and oxitropium bromide have
clinical study to be superior to that with a MDI [41]. a slower onset of action than 2-agonists, with a peak eect
being obtained in between 30 and 90 min. Ipratropium bro-
mide has a duration of 68 h, while oxitropium is claimed
to maintain bronchodilation for 8 h, although this could be
PHARMACOKINETICS due to a higher dose of oxitropium [17]. On the basis of the
duration of action, ipratropium bromide is given four times
daily, while oxitropium bromide is given three times daily.
Tiotropium bromide provides a bronchodilator eect for
over 24 h with a plasma concentration of ~2 pg/ml. After
inhalation of a 10-g dose there is a rapid absorption with
a peak plasma concentration within 5 min of 6 pg/ml, fol-
lowed by a rapid fall within 1 h to the steady state level of
CLINICAL STUDIES
2 pg/ml and a terminal half-life of 56 days that is inde-
pendent of the dose [24]. It has been calculated that this A comprehensive range of clinical studies have been per-
concentration would occupy 5% muscarinic receptors, formed with tiotropium up to registration with the drug
perhaps accounting for the very low incidence of systemic regulatory authorities, and other studies that investigate the
side eects. There is no evidence for accumulation of tiotro- eects of tiotropium on exacerbations and the natural his-
pium after repeated administration. The pharmacokinetics tory of COPD are ongoing (Table 49.1).
and tissue distribution of tiotropium in the rat and dog have Single-dose relationships of tiotropium bromide have
also been described [42]. Following dry powder inhalation, been determined with nebulized doses of up to 160 g in
the absolute bioavailability of 19.5% suggests that the lung patients with COPD [23, 44], as well as doses of up to
fraction is highly bioavailable. Since tiotropium is a qua- 80 g from a DPI in asthma [72] (Fig. 49.5). In the open
ternary ammonium compound, it is poorly absorbed from label rst study in patients prolonged bronchodilation with-
the gastrointestinal tract (1015%). Food is not expected to out signicant adverse events was noted in COPD [44],
inuence absorption. and in a randomized placebo-controlled crossover design a
Tiotropium at a level of 72% to plasma proteins clear dose response could be demonstrated between for tio-
shows a volume of distribution of 32 l/kg. At steady state, tropium at single doses of between 10 and 80 g [44]. In
tiotropium plasma levels in COPD patients at peak were addition, in the single dose study in patients with asthma,
1719 pg/ml when measured 5 min after inhalation of an tiotropium mediated bronchoprotection against the con-
18-g dose, and steady state trough plasma concentra- strictor eects of methacholine [72].
tions were 34 pg/ml. The extent of biotransformation is A 1-week study in patients with COPD demon-
small, with urinary excretion of 74% of unchanged sub- strated that 70% of the bronchodilator eect of tio-
stance after an intravenous dose to young healthy volun- tropium are obtained after two doses, and that the FEV1
teers. Tiotropium should be used with care in patients with steady state is reached within 48 h [45]. A 4 week study of
moderate-to-severe renal insuciency [43]. The termina- tiotropium at doses of up to 36 g given daily at noon found
tion half-life of tiotropium is between 5 and 6 days follow- comparable responses at doses from 9 to 36 g, represent-
ing inhalation. Tiotropium has linear pharmacokinetics in ing a at dose response curve, with little dose dependency
the therapeutic range after both intravenous administration in terms of spirometric outcomes [49]. On this basis, a dose
and dry powder inhalation. of 18 g was selected for use in long-term studies, since the
36 g dose increased the incidence of dry mouth without
signicantly increasing FEV1.
The Dutch Tiotropium Study Group has undertaken
ONSET AND DURATION OF ACTION a 13-week study with 288 patients with COPD, in which
tiotropium at 18 g from the HandiHaler administered
once daily was signicantly more eective than ipratropium
Tiotropium bromide is given as 18 g once daily, usually in administered four times daily [61]. The US Tiotropium
the morning or at noon. In studies of patients with COPD, Study Group also undertook a 13-week multicenter study,
single doses of tiotropium bromide give an almost immedi- but compared tiotropium with placebo, and found signi-
ate bronchodilator response that is preserved for over 24 h cant improvements in lung function (FEV1 and PEFR),
[44]. Approximately 70% of the improvement in FEV1 is symptoms, as-needed albuterol/salbutamol usage and phy-
seen after 48 h, which is judged to be the steady state, while sician global assessment [48].
continued improvements can be expected beyond the rst Long-term studies with tiotropium bromide in
week of therapy [45], while dyspnea and health-related moderate-to-severe COPD have demonstrated signicant
quality of life improve over the longer term. Interestingly improvement in lung function, symptoms, and health-related
in patients with and without short-term bronchodilator quality of life, as well as a reduction in exacerbations [50, 62]
responses, long-term benets of treatment with tiotropium (Fig. 49.6). These studies conrm that the positive eects
are found in COPD patients [46]. Tiotropium was found seen in 4- and 13-week studies are extended to 1 year and
to improve signicantly in lung functions in African- suggest that tolerance does not occur in this period [73]. In
American patients with COPD [47]. the study from the Dutch/Belgian Tiotropium Study Group,

619
TABLE 49.1 Summary of selected randomized controlled clinical studies on tiotropium bromide.

Duration Mean FEV1 at baseline Dyspnea/ Exacerbation


Reference n (weeks) (% predicted) FEV1 symptoms frequency HRQOL score

Tiotropium versus Placebo

Casaburi [48] 470 13 39 NA NA


Littner [49] 169 4 39 NA NA NA
Casburi [50] 921 52 39
Donohue [51] 623 26 40
Calverley [52] 121 6 41 NA NA
Brusasco [53] 1207 26 40
Celli [54] 81 4 43 NA NA NA
Hasani [55] 34 3 44 NA NA NA
ODonnell [56] 187 6 42 NA
McNicholas [57] 95 4 32 NA NA NA
Dusser [58] 1010 52 48 NA
Niehwoehner [59] 1829 26 36 NA NA
Verkindre [60] 100 12 36 NA

Tiotropium versus Ipratropium

Van Noord [61] 288 13 41 NA NA


Vincken [62] 535 52 41

Tiotropium versus Salmeterol

Donohue [51] 623 26 40


Brusasco [53] 1207 26 40 NA
Briggs [63] 653 12 38 NA NA

Tiotropium versus Formoterol

Van Noord [64] 71 20 37 NA NA NA


Cazzola [65] 20 Single doses 35 NA NA NA
Di Marco [66] 21 Single doses 38 NA NA NA

Tiotropium (TIO) combinations with Salmeterol (SALM)/Fluticasone (FLU)/Formoterol (FORM)

Cazzola [67] 90 12 3739 NA NA


TIO
SALMFLU
TIO  SALMFLU

Aaron [68] 449 52 3839


TIO
TIO  SALM
TIO  SALMFLU

Singh [69] 41 2 47 NA NA
TIO
SALMFLU
TIO  SALMFLU

Van Noord [70] 95 2 38 NA NA


TIO
TIO  FORM

Di Marco [66] 21 Single doses 38 NA NA NA


TIO
TIO  FORM

Tashkin [71] Nebulized 129 6 38 NA


TIO
TIO  FORM

This table is adapted with permission from a review by Ref. [8].


Anticholinergic Bronchodilators 49
Tiotropium: Single dose study in COPD

Inhalation
0.35
0.30
0.25
0.20
FEV1 (l)

0.15
0.10
0.05
0.00
0.05
0.10

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32
Time post-administration (h)

80 g Tiotropium bromide 20 g Tiotropium bromide Placebo

Increase in adjusted mean improvement in FEV1 from test day baseline FIG. 49.5 Single-dose study of tiotropium in
Adapted from Fig.1, Ref. [44] COPD (see text on figure).

One year studies with tiotropium: effects on lung function


(A) (B)
1.5
Mean weekly PEFR (l/min)

240

230 (n 507)
1.4
FEV1 (l)

220
1.3
210 (n 332)
1.2
200

1.1 190
60 0 60 120 180 0 4 8 12 16 20 24 28 32 36 40 44 48 52
Time after inhalation (min) Time (weeks)

Tiotropium Ipratropium Tiotropium Placebo


Baseline Baseline
8 days 8 days
364 days 364 days

(A) Effects on trough FEV1 following inhalation of tiotropium or ipratropium (Adapted from Ref. [62]).

(B) Effects on mean weekly morning peak expiratory flow rates (PEFR) following inhalation of tiotropium or
placebo (Adapted from Ref. [50]).

FIG. 49.6 One year studies with tiotropium (see text on Figure).

tiotropium was eective in improving lung function, dyspnea, study from Casaburi and colleagues, tiotropium also showed
exacerbations and health-related quality of life, and improved signicant eects on the Transition Dyspnea Index (TDI)
trough FEV1 at 1 year [62]. In this study tiotropium was as well as health status assessed by the disease-specic St.
found to be more eective than ipratropium inhaled four Georges Respiratory Questionnaire (SGRQ) and the generic
times a day. In a separate placebo-controlled multicenter Short Form (SF) 36 [50].

621
Asthma and COPD: Basic Mechanisms and Clinical Management

EFFECTS OF TIOTROPIUM ON OUTCOMES arm. In addition, a small signicant benecial eect was
found on health status with an unexpected nding in
IN COPD lower death in SF-treated patients [83].

Bronchodilatation, symptoms of dyspnea


Natural history of COPD
Bronchodilators lessen airway smooth muscle tone and reduce
dynamic hyperination, to cause relatively rapid relief of symp- There is little evidence that bronchodilators, including tio-
toms and improved exercise tolerance [7476]. Relaxation tropium, modify the rate of decline in lung function (FEV1)
of airway smooth muscle tone causes an increase in FEV1, in COPD [84]. It is very important to assess whether
but in COPD these changes are generally small (10%). chronic therapy with tiotropium can aect the natural his-
Bronchodilator ecacy in COPD is generally assessed as the tory of COPD, which can be dened by the annual rate of
FEV1 measured by spirometry. Symptomatic improvement decline in post-bronchodilatory FEV1. A rather unsatis-
may be greater than changes in FEV1, especially in moderate- factory attempt to assess eects of tiotropium on longitu-
to-severe COPD [62], and tiotropium causes improvement in dinal eects of lung function was made by pooled analysis
dyspnea [50, 77, 78]. of two 1-year tiotropium studies [85]. These studies have
the problem that lung function should be measured at the
end of chronic tiotropium therapy, with a suciently large
Dynamic hyperinflation time interval to have no lingering bronchodilatory or other
eects of tiotropium.
Improved exercise performance may be a conspicuous fea- The UPLIFT (Understanding the Potential Long-
ture of bronchodilator usage in COPD [54]. Reduced Term Impacts on Function with Tiotropium) study is cur-
dynamic hyperination with a reduction in residual volume rently evaluating the long-term eects of 4 years of therapy
(RV) and functional residual capacity (FRC) but improved with tiotropium on the rate of decline in lung function,
resting inspiratory capacity (IC) [54] contributes markedly health status, and frequency of exacerbations [86]. The
to symptomatic benets, since this makes breathing more UPLIFT study measures trough FEV1, as well as FEV1,
comfortable and dyspnea is lessened on exertion [56,79]. 90 min after maximal bronchodilator administration and
Improvement of exercise tolerance is aided by the combina- includes a spectrum of patient-centered outcomes [87].
tion of tiotropium and pulmonary rehabilitation [80]. Other
eects of anticholinergics are to increase mucociliary clear-
ance and decrease mucous production. However, improved Costs and health economics
respiratory muscle function is unlikely at the dose of tiotro- Health outcomes have been found to be positive following
pium used clinically. treatment of COPD patients with once daily tiotropium
for 6 months [53], and similar results have been found after
1 year of treatment with tiotropium [88], and in compari-
Exacerbations son with treatment with ipratropium over a year [89]. In a
probabilistic Markov model to assess the cost-eectiveness
The reader is recommended to consult a recent review on of bronchodilator therapy in COPD, tiotropium was associ-
the eect of tiotropium on exacerbations and hospitaliza- ated in terms of exacerbations with maximum expected net
tions [81]. benet for plausible values of the ceiling ratio [90].
The 1-year tiotropium registration studies demonstrated
that tiotropium causes a decreased incidence of Sleep quality
exacerbations when compared with placebo [46, 50, 62]. Sustained LAMA therapy with tiotropium improves sleep-
Following this, Niewoehner and colleagues prospectively ing oxygen saturation in COPD without aecting sleep
demonstrated signicant tiotropium eects on reducing quality [57]. Tiotropium does not appear to abolish circa-
COPD exacerbations, and may reduce related healthcare dian variation in airway caliber [52].
utilization [59].
The MISTRAL (Mesure de LInuence de Spiriva sur les
Troubles Aigus Long Terme) study studied 500 patients
with COPD treated with tiotropium for a 1-year period COMPARISON OF TIOTROPIUM WITH
in comparison with a placebo group [58]. SALMETEROL AND FORMOTEROL
The INSPIRE (Investigating New Standards for
Prophylaxis in Reduction of Exacerbations) study A 6-month clinical trial in 623 patients with COPD has
compares salmeterol in addition to uticasone (SF) in a demonstrated superior eects of tiotropium on bronchodila-
combination inhaler compared with tiotropium [82]. This tion, dyspnea, and health-related quality of life compared to
study found no dierence in exacerbation rates between salmeterol [51]. In other studies with tiotropium being given
the two arms, with more patients failing to complete once daily, while salmeterol was given twice daily, tiotropium
the study while receiving tiotropium, yet more cases of has been shown to be superior in terms of daytime spiromet-
pneumonia were reported in the salmeterol/uticasone ric ecacy [63] and exacerbations and health outcomes [53].

622
Anticholinergic Bronchodilators 49
In comparison with formoterol, tiotropium was better 1.5
in terms of lung function, although a combination of both
1.4
drugs was most eective [64]. The benets of tiotropium
over formoterol have also been demonstrated in single-dose 1.3

FEV1 (l)
studies [65].
1.2

1.1
COMBINATION STUDIES WITH TIOTROPIUM 1.0

0.9
Combination bronchodilator products have already been 2 0 2 4 6 8 10 12 14 16 18 20 22 24
demonstrated to have advantages over a single class of 3.0
bronchodilator, so that both short-acting and long-acting
2-agonists may be combined with anticholinergics, and 2.8
additional oral theophylline may have benet. Combination
bronchodilator inhalers with an anticholinergic and a 2.6
short-acting 2-agonist, such as Combivent (ipratropium

IC (l)
bromide  salbutamol), are a convenient a and more eec- 2.4
tive way of giving bronchodilators [9193]. Tiotropium 2.2
bromide has additive eects with both short- and long-
acting 2-agonists (LABAs) [94]. 2.0
An important study demonstrated that adding tiotro-
pium to formoterol causes improvements in airow obstruc- 1.8
tion, resting hyperination, and use of rescue salbutamol 2 0 2 4 6 8 10 12 14 16 18 20 22 24
[70] (Fig. 49.7). Concomitant treatment with nebulized 9 AM 3 PM 9 PM 3 AM 9 AM
formoterol and tiotropium provided benets in patients Time (h)
with COPD [71], and the combination of formoterol and Mean FEV1 and Inspiratory Capacity (IC) before (24 h baseline)
tiotropium is complementary in treating acute exacerbations and at the end of 2-week treatment periods, adjusted for period,
of COPD [66]. center, and patient within center.
Combined LABAs and ICS in a single inhaler are Tiotropium qd  formoterol bid Tiotropium qd
licensed for asthma and COPD, with the combination of Tiotropium qd  formoterol qd 24-h baseline
salmeterol and uticasone available as Seretide or Advair
(GlaxoSmithKline) [95], with formoterol and budesonide Reproduced with kind permission from Ref. [70]
as Symbicort (AstraZeneca) [96]. Recently, there have been
studies to show the benets of triple therapy with the com- FIG. 49.7 Bronchodilator effect of tiotropium with and without formoterol
in COPD.
bination of salmeterol/uticasone and tiotropium [67, 69].
However, addition of salmeterol/uticasone to tiotropium
did not statistically inuence rates of COPD exacerbation
in a study of 449 patients over 1 year of treatment [68].
SAFETY OF TIOTROPIUM
A pooled clinical trial analysis (meta-analysis) of tiotropium
safety has been carried out on a population of 4435 patients
TIOTROPIUM IN ASTHMA receiving inhaled tiotropium [99]. Dyspnea, dry mouth,
COPD exacerbation, and upper respiratory tract infections
were the most commonly reported adverse events. A careful
Anticholinergics are less useful in asthma than in COPD, as study demonstrated that tiotropium does not retard mucus
inhaled 2-agonists are generally more eective [5]. However, clearance from the lungs [55].
with regard to allergen-induced airway remodeling in guinea A cohort study using UK primary care databases
pigs, tiotropium and budesonide have similar inhibitory e- found that tiotropium and LABAs had similar risks of
cacy [35]. Patients with concomitant asthma and COPD total mortality and cardiovascular endpoints [100], while
have been shown to benet from tiotropium [97], and in the SPRUCE primary care study showed benets and not
patients with severe asthma adding salmeterol/tiotropium adverse eects when adding tiotropium to COPD care
to patients with uticasone has benets [98]. Tiotropium [101]. Detailed electrocardiography (ECG) monitoring has
could reduce chronic viral inammation and may be helpful been performed in COPD patients receiving tiotropium
in acute severe asthma [34]. Hence, tiotropium is likely to with ECGs performed up to six times during 12 months
be eective as an additional bronchodilator in patients with of treatment [102], and tiotropium was not associated with
severe disease, especially when there is an element of xed any cardiac safety concerns in a separate ECG analysis in
airow limitation. It is likely that patients with severe asthma COPD patients [103].
will benet from the addition of tiotropium to combination Side eects of anticholinergics are pharmacologi-
inhalers containing long-acting 2-agonists and corticoster- cally predictable and dose dependent, and inhaled delivery
oids, but clinical trials are needed to document this. means that systemic adverse eects are less likely. However,

623
Asthma and COPD: Basic Mechanisms and Clinical Management

COPD patients are older and more likely to have comor- References
bidities than patients with asthma, so they have a greater
risk of developing adverse reactions to therapy. Side eects 1. Rabe KF, Hurd S, Anzueto A et al. Global strategy for the diagnosis,
of tiotropium bromide are very uncommon as tiotropium management, and prevention of chronic obstructive pulmonary disease:
acts topically and there is virtually no systemic absorption. GOLD executive summary. Am J Respir Crit Care Med 176(6): 53255,
This limits the type of adverse anticholinergic eects seen 2007.
with atropine; and extensive experience has shown inhaled 2. National Institutes of Health (NIH), National Heart Lung and Blood
anticholinergics to be a remarkably safe class of drug. Institute (NHLBI), World Health Organisation (WHO). Global
Initiative for Chronic Obstructive Lung Disease (GOLD): Global Strategy
Dry mouth may be associated with dental caries for the Diagnosis, Management, and Prevention of Chronic Obstructive
Pulmonary Disease. Updated 2007.
Cough, pharyngitis, upper respiratory infections 3. Tashkin DP, Cooper CB. The role of long-acting bronchodilators in
Ocular eects: narrow angle glaucoma is a recognized the management of stable COPD. Chest 125(1): 24959, 2004.
adverse eect of nebulized anticholinergics and 4. Decramer M. Tiotropium as essential maintenance therapy in COPD.
Eur Respir Rev 15: 5157, 2006.
tiotropium may cause eye pain, blurred vision, visual
5. Gross NJ. Anticholinergic agents in asthma and COPD. Eur J
halos, and conjunctival and corneal congestion (redness). Pharmacol 533(13): 3639, 2006.
Taste disturbance: a bitter metallic taste has been 6. Koumis T, Samuel S. Tiotropium bromide: A new long-acting bron-
reported by some patients. chodilator for the treatment of chronic obstructive pulmonary disease.
Clin Ther 27(4): 37792, 2005.
Immediate hypersensitivity reactions may occur. 7. Restrepo RD. Use of inhaled anticholinergic agents in obstructive air-
CNS: headache, nervousness, irritation, dizziness. way disease. Respir Care 52(7): 83351, 2007.
8. Currie GP, Rossiter C, Miles SA, Lee DK, Dempsey OJ. Eects of
Theoretical systemic anticholinergic eects: prostatic tiotropium and other long acting bronchodilators in chronic obstruc-
hypertrophy, urinary retention, bladder disorders, tive pulmonary disease. Pulm Pharmacol Ther 19(2): 11219, 2006.
constipation, tachycardia, cardiac arrhythmias, 9. Voduc N. Tiotropium in the treatment of chronic pulmonary disease:
palpitations. Review of clinical trials. Todays Therapeutic Trends: 3747, 2006.
10. Barr RG, Bourbeau J, Camargo CA, Ram FS. Tiotropium for stable
Renal and hepatic impairment: patients can use chronic obstructive pulmonary disease: A meta-analysis. Thorax 61(10):
tiotropium bromide at the recommended dose. 85462, 2006.
11. Sakula A. A history of asthma: The FitzPatrick lecture 1987. J R Coll
Physicians Lond 22: 3644, 1988.
12. Rau JL. Respiratory Care Pharmacology, 6th edn. St Louis, USA: Mosby,
NEW ANTICHOLINERGICS 2003.
13. Gross NJ. Ipratropium bromide. N Engl J Med 319(8): 48694, 1988.
14. Chapman KR. History of anticholinergic treatment in airways dis-
There have been considerable eorts from the pharmaceuti- ease. In: Gross NJ (ed.), Anticholinergic Therapy in Obstructive Airways
Disease, pp. 917. London: Franklin Scientic Publications, 1993.
cal industry to develop novel bronchodilators, and especially
15. Herxheimer H. Atropine cigarettes in asthma emphysema. BMJ 2: 167,
LABAs and LAMAs, for COPD [104, 105]. Glycopyrrolate 1959.
has been used extensively systemically for decades to control 16. Gandevia B. Historical review of the use of parasympatholytic agents in
salivation as a pre-anesthetic agent and causes bronchodila- the treatment of respiratory disorders. Postgrad Med J 51: S1320, 1975.
tion on isolated guinea pig trachea [106, 107]. Interestingly, 17. Barnes PJ. Tiotropium bromide. Expert Opin Investig Drugs 10(4):
glycopyrrolate is a quaternary ammonium derivative which 73340, 2001.
has minimal mucosal absorption and systemic toxicity when 18. Hvizdos KM, Goa KL. Tiotropium bromide. Drugs 62: 1195203,
inhaled, causing prolonged bronchodilation in patients with 2002.
asthma [106] and a family of anticholinergics based on gly- 19. Hansel TT, Barnes PJ. Tiotropium bromide: A novel once-daily anti-
copyrrolate have been synthesized [108]. A variant on this cholinergic bronchodilator for the treatment of COPD. Drugs Today
38(9): 585600, 2002.
agent is racemically pure R,R-glycopyrrolate as a LAMA
20. Tamaoki J, Chiyotani A, Tagaya E, Sakai N, Konno K. Eect of long
[109], while an inhaled glycopyrrolate derivative, NVA- term treatment with oxitropium bromide on airway secretion in chronic
237, is currently being developed by Novartis (license from bronchitis and diuse panbronchiolitis. Thorax 49(6): 54548, 1994.
Arakis and Ventura). An oral selective M3-selective M3- 21. Barnes PJ. Muscarinic receptor subtypes in airways. Life Sci 52: 521
cholinergic receptor antagonist that was studied in patients 28, 1993.
with COPD (OrM3, Merck) has apparently been discon- 22. Eglen RM, Choppin A, Watson N. Therapeutic opportunities from
tinued in clinical development [110]. However, Phase 3 muscarinic receptor research. Trends Pharmacol Sci 22: 40914, 2001.
studies have been successfully carried with the inhaled 23. Maesen FPV, Smeets JJ, Costongs MAL, Wald FDM, Comelissen PJG.
LAMA, aclidinium bromide (LAS-34273, Almirall), that is Ba 679 Br, a new long-acting antimuscarinic bronchodilator: A pilot
likely to be licensed in the near future. Recently there have dose-escalation study in COPD. Eur Respir J 6: 103136, 1993.
24. Disse B, Speck GA, Rominger KL, Witek TJ, Hammer R. Tiotropium
been eorts to develop a combined muscarinic antagonist
(Spiriva) mechanistical considerations and clinical prole in obstruc-
and beta-agonist in a single molecule: Theravance describes tive lung disease. Life Sci 64: 45764, 1999.
this as a MABA. In the future we can expect LABAs and 25. Barnes PJ. The pharmacological properties of tiotropium. Chest 117(2
LAMAs to be combined in single inhalers (eg. indacaterol- Suppl): 63S66S, 2000.
glycopyrrolate) and there may be the possibility of 3 agents 26. Disse B, Reichl R, Speck G, Traunecker W, Rominger KL, Hammer R. A
in a single inhaler (LABA, LAMA, ICS). novel long-acting anticholinergic bronchodilator. Life Sci 52: 53744, 1993.

624
Anticholinergic Bronchodilators 49
27. Haddad EB, Mak JC, Barnes PJ. Characterization of [3H]Ba 679 48. Casaburi R, Serby W, Menjoge SS, Witek TJ. The spirometric ecacy
BR, a slowly dissociating muscarinic antagonist, in human lung: of once daily dosing with tiotropium in stable COPD. Am J Respir Crit
Radioligand binding and autoradiographic mapping. Mol Pharmacol Care Med 159: A524, 1999.
45(5): 899907, 1994. 49. Littner MR, Ilowite JS, Tashkin DP et al. Long-acting bronchodila-
28. Wess J, Eglen RM, Gautam D. Muscarinic acetylcholine receptors: tion with once-daily dosing of tiotropium (Spiriva) in stable chronic
Mutant mice provide new insights for drug development. Nat Rev obstructive pulmonary disease. Am J Respir Crit Care Med 161(4 Pt 1):
Drug Discov 6(9): 72133, 2007. 113642, 2000.
29. Grando SA, Kawashima K, Kirkpatrick CJ, Wessler I. Recent progress 50. Casaburi R, Mahler DA, Jones PW et al. A long-term evaluation of
in understanding the non-neuronal cholinergic system in humans. Life once-daily inhaled tiotropium in chronic obstructive pulmonary dis-
Sci 80(2425): 218185, 2007. ease. Eur Respir J 19(2): 21724, 2002.
30. Wessler I, Kilbinger H, Bittinger F, Unger R, Kirkpatrick CJ. The 51. Donohue JF, van Noord JA, Bateman ED et al. A 6-month, placebo-
non-neuronal cholinergic system in humans: Expression, function and controlled study comparing lung function and health status changes in
pathophysiology. Life Sci 72(1819): 205561, 2003. COPD patients treated with tiotropium or salmeterol. Chest 122(1):
31. Ulloa L. The vagus nerve and the nicotinic anti-inammatory pathway. 4755, 2002.
Nat Rev Drug Discov 4(8): 67384, 2005. 52. Calverley PM, Lee A, Towse L, van Noord J, Witek TJ, Kelsen S.
32. Kawashima K, Fujii T. The lymphocytic cholinergic system and its con- Eect of tiotropium bromide on circadian variation in airow limita-
tribution to the regulation of immune activity. Life Sci 74(6): 67596, tion in chronic obstructive pulmonary disease. Thorax 58(10): 85560,
2003. 2003.
33. Adamko DJ, Fryer AD, Bochner BS, Jacoby DB. CD8 T lymphocytes 53. Brusasco V, Hodder R, Miravitlles M, Korducki L, Towse L, Kesten S.
in viral hyperreactivity and M2 muscarinic receptor dysfunction. Am J Health outcomes following treatment for six months with once daily
Respir Crit Care Med 167(4): 55056, 2003. tiotropium compared with twice daily salmeterol in patients with
34. Kanazawa H. Anticholinergic agents in asthma: Chronic bronchodi- COPD. Thorax 58(5): 399404, 2003.
lator therapy, relief of acute severe asthma, reduction of chronic viral 54. Celli B, ZuWallack R, Wang S, Kesten S. Improvement in resting
inammation and prevention of airway remodeling. Curr Opin Pulm inspiratory capacity and hyperination with tiotropium in COPD
Med 12(1): 6067, 2006. patients with increased static lung volumes. Chest 124(5): 174348,
35. Bos IS, Gosens R, Zuidhof AB et al. Inhibition of allergen-induced 2003.
airway remodelling by tiotropium and budesonide: A comparison. Eur 55. Hasani A, Toms N, Agnew JE, Sarno M, Harrison AJ, Dilworth P. The
Respir J 30(4): 65361, 2007. eect of inhaled tiotropium bromide on lung mucociliary clearance in
36. Gosens R, Bos IS, Zaagsma J, Meurs H. Protective eects of tiotro- patients with COPD. Chest 125(5): 172634, 2004.
pium bromide in the progression of airway smooth muscle remodeling. 56. ODonnell DE, Fluge T, Gerken F et al. Eects of tiotropium on lung
Am J Respir Crit Care Med 171(10): 1096102, 2005. hyperination, dyspnoea and exercise tolerance in COPD. Eur Respir J
37. Matthiesen S, Bahulayan A, Kempkens S et al. Muscarinic receptors 23(6): 83240, 2004.
mediate stimulation of human lung broblast proliferation. Am J Respir 57. McNicholas WT, Calverley PM, Lee A, Edwards JC. Long-acting
Cell Mol Biol, 2006. inhaled anticholinergic therapy improves sleeping oxygen saturation in
38. Buhling F, Lieder N, Kuhlmann UC, Waldburg N, Welte T. COPD. Eur Respir J 23(6): 82531, 2004.
Tiotropium suppresses acetylcholine-induced release of chemotactic 58. Dusser D, Bravo ML, Iacono P. The eect of tiotropium on exacerbations
mediators in vitro. Respir Med 101(11): 238694, 2007. and airow in patients with COPD. Eur Respir J 27(3): 54755, 2006.
39. Powrie DJ, Wilkinson TM, Donaldson GC et al. Eect of tiotropium 59. Niewoehner DE, Rice K, Cote C et al. Prevention of exacerbations of
on sputum and serum inammatory markers and exacerbations in chronic obstructive pulmonary disease with tiotropium, a once-daily
COPD. Eur Respir J 30(3): 47278, 2007. inhaled anticholinergic bronchodilator: A randomized trial. Ann Intern
40. Chodosh S, Flanders JS, Kesten S, Serby CW, Hochrainer D, Witek Med 143(5): 31726, 2005.
TJ. Eective delivery of particles with the HandiHaler dry powder 60. Verkindre C, Bart F, Aguilaniu B et al. The eect of tiotropium on
inhalation system over a range of chronic obstructive pulmonary dis- hyperination and exercise capacity in chronic obstructive pulmonary
ease severity. J Aerosol Med 14: 30915, 2001. disease. Respiration 73(4): 42027, 2006.
41. Dahl R, Backer V, Ollgaard B, Gerken F, Kesten S. Assessment of patient 61. van Noord JA, Bantje TA, Eland ME, Korducki L, Cornelissen PJ. A
performance of the HandiHaler compared with the metered dose inhaler randomised controlled comparison of tiotropium and ipratropium in
four weeks after instruction. Respir Med 97(10): 112633, 2003. the treatment of chronic obstructive pulmonary disease. The Dutch
42. Leusch A, Eichhorn B, Muller G, Rominger KL. Pharmacokinetics Tiotropium Study Group. Thorax 55(4): 28994, 2000.
and tissue distribution of the anticholinergics tiotropium and ipratro- 62. Vincken W, van Noord JA, Greefhorst AP et al. Improved health out-
pium in the rat and dog. Biopharm Drug Dispos 22(5): 199212, 2001. comes in patients with COPD during 1 yrs treatment with tiotropium.
43. Turck D, Weber W, Sigmund R et al. Pharmacokinetics of intrave- Eur Respir J 19(2): 20916, 2002.
nous, single-dose tiotropium in subjects with dierent degrees of renal 63. Briggs DD Jr, Covelli H, Lapidus R, Bhattycharya S, Kesten S, Cassino C.
impairment. J Clin Pharmacol 44(2): 16372, 2004. Improved daytime spirometric ecacy of tiotropium compared with salm-
44. Maesen FPV, Smeets JJ, Sledsens TJH, Wald FDM, Cornelissen PJG. eterol in patients with COPD. Pulm Pharmacol Ther 18(6): 397404, 2005.
Tiotropium bromide, a new long-acting antimuscarinic bronchodilator: 64. van Noord JA, Aumann JL, Janssens E et al. Comparison of tiotropium
A pharmacodynamic study in patients with chronic obstructive disease once daily, formoterol twice daily and both combined once daily in
(COPD). Eur Respir J 8: 150613, 1995. patients with COPD. Eur Respir J 26(2): 21422, 2005.
45. van Noord JA, Smeets JJ, Custers FL, Korducki L, Cornelissen PJ. 65. Cazzola M, Di MF, Santus P et al. The pharmacodynamic eects of
Pharmacodynamic steady state of tiotropium in patients with chronic single inhaled doses of formoterol, tiotropium and their combination
obstructive pulmonary disease. Eur Respir J 19(4): 63944, 2002. in patients with COPDA. Pulm Pharmacol Ther 17(1): 3539, 2004.
46. Tashkin D, Kesten S. Long-term treatment benets with tiotropium in 66. Di MF, Verga M, Santus P, Morelli N, Cazzola M, Centanni S. Eect
COPD patients with and without short-term bronchodilator responses. of formoterol, tiotropium, and their combination in patients with acute
Chest 123: 144149, 2003. exacerbation of chronic obstructive pulmonary disease: A pilot study.
47. Criner GJ, Sharafkhaneh A, Player R et al. Ecacy of tiotropium inha- Respir Med 100(11): 192532, 2006.
lation powder in african-american patients with chronic obstructive 67. Cazzola M, Ando F, Santus P et al. A pilot study to assess the eects
pulmonary disease. COPD 5(1): 3541, 2008. of combining uticasone propionate/salmeterol and tiotropium on

625
Asthma and COPD: Basic Mechanisms and Clinical Management

the airow obstruction of patients with severe-to-very severe COPD. 90. Oostenbrink JB, Rutten-van Molken MP, Monz BU, Fitzgerald JM.
Pulm Pharmacol Ther 20(5): 55661, 2007. Probabilistic Markov model to assess the cost-eectiveness of bron-
68. Aaron SD, Vandemheen KL, Fergusson D et al. Tiotropium in com- chodilator therapy in COPD patients in dierent countries. Value
bination with placebo, salmeterol, or uticasonesalmeterol for treat- Health 8(1): 3246, 2005.
ment of chronic obstructive pulmonary disease: A randomized trial. 91. Fitzgerald JM, Gruneld A, Pare PD et al. The clinical ecacy of
Ann Intern Med 146(8): 54555, 2007. combination nebulized anticholinergic and adrenergic bronchodila-
69. Singh D, Brooks J, Hagan G, Cahn T, OConnor B. Triple therapy tors vs nebulized adrenergic bronchodilator alone in acute asthma.
with salmeterol/uticasone propionate and tiotropium bromide versus Chest 111: 31115, 1997.
individual components in moderate to severe COPD. Thorax, 2008. 92. Chin JE, Hateld CA, Winterrowd GE et al. Preclinical evaluation
70. van Noord JA, Aumann JL, Janssens E et al. Eects of tiotropium with of anti-inammatory activities of the novel pyrrolopyrimidine PNU-
and without formoterol on airow obstruction and resting hyperina- 142731A, a potential treatment for asthma. J Pharmacol Exp Ther 290:
tion in patients with COPD. Chest 129(3): 50917, 2006. 18895, 1999.
71. Tashkin DP, Littner M, Andrews CP, Tomlinson L, Rinehart M, 93. Wilson JD, Serby CW, Menjoge SS, Witek TJ. The ecacy and safety
Denis-Mize K. Concomitant treatment with nebulized formoterol and of combination bronchodilator therapy. Eur Respir Rev 6: 28689,
tiotropium in subjects with COPD: A placebo-controlled trial. Respir 1996.
Med, 2008. doi:10.1016/j.rmed.2007.12.019 94. Tennant RC, Erin EM, Barnes PJ, Hansel TT. Long-acting beta(2)-
72. OConnor BJ, Towse LJ, Barnes PJ. Prolonged eect of tiotropium adrenoceptor agonists or tiotropium bromide for patients with
bromide on methacholine-induced bronchoconstriction in asthma. COPD: Is combination therapy justied? Curr Opin Pharmacol 3(3):
Am J Respir Crit Care Med 154: 87680, 1996. 27076, 2003.
73. Rees PJ. Tiotropium in the management of chronic obstructive pulmo- 95. Nelson HS. Advair: Combination treatment with uticasone propi-
nary disease. Eur Respir J 19(2): 2056, 2002. onate/salmeterol in the treatment of asthma. J Allergy Clin Immunol
74. ODonnell DE. Impacting patient-centred outcomes in COPD: 107: 397416, 2001.
Breathlessness and exercise tolerance. Eur Respir Rev 15: 3744, 2006. 96. McGavin JK, Goa KL, Jarvis B. Inhaled budesonide/formoterol com-
75. ODonnell DE. Lung mechanics in COPD: The role of tiotropium. bination. Drugs 61(1): 7178, 2001.
Eur Respir Rev 13(89): 4044, 2004. 97. Magnussen H, Bugnas B, van NJ, Schmidt P, Gerken F, Kesten S.
76. Ferguson GT. The ins and outs of breathing: An overview of lung Improvements with tiotropium in COPD patients with concomitant
mechanics. Eur Respir Rev 13: 3034, 2004. asthma. Respir Med 102(1): 5056, 2008.
77. Magnussen H. Exercise limitation in COPD: Mechanisms, assessment 98. Fardon T, Haggart K, Lee DK, Lipworth BJ. A proof of concept study
and treatment. Eur Respir Rev 13: 3539, 2004. to evaluate stepping down the dose of uticasone in combination with
78. Maltais F, Hamilton A, Marciniuk D et al. Improvements in symptom- salmeterol and tiotropium in severe persistent asthma. Respir Med
limited exercise performance over 8 h with once-daily tiotropium in 101(6): 121828, 2007.
patients with COPD. Chest 128(3): 116878, 2005. 99. Kesten S, Jara M, Wentworth C, Lanes S. Pooled clinical trial analysis
79. ODonnell DE, Lam M, Webb KA. Measurement of symptoms, lung of tiotropium safety. Chest 130(6): 1695703, 2006.
hyperination, and endurance during exercise in chronic obstructive pul- 100. Jara M, Lanes SF, Wentworth C, May C, Kesten S. Comparative
monary disease. Am J Respir Crit Care Med 158(5 Pt 1): 155765, 1998. safety of long-acting inhaled bronchodilators: A cohort study Using
80. Casaburi R, Kukafka D, Cooper CB, Witek TJ Jr, Kesten S. the UK THIN Primary Care Database. Drug Saf 30(12): 115160, 2007.
Improvement in exercise tolerance with the combination of tiotropium 101. Freeman D, Lee A, Price D. Ecacy and safety of tiotropium
and pulmonary rehabilitation in patients with COPD. Chest 127(3): in COPD patients in primary care the SPiRiva Usual CarE
80917, 2005. (SPRUCE) study. Respir Res 8: 45, 2007.
81. Rodriguez-Roisin R. Impacting patient-centred outcomes in COPD: 102. Morganroth J, Golisch W, Kesten S. Electrocardiographic monitoring
Exacerbations and hospitalisations. Eur Respir Rev 15: 4750, 2006. in COPD patients receiving tiotropium. COPD 1(2): 18190, 2004.
82. Seemungal T, Stockley R, Calverley P, Hagan G, Wedzicha JA. 103. Covelli H, Bhattacharya S, Cassino C, Conoscenti C, Kesten S.
Investigating new standards for prophylaxis in reduction of exacerba- Absence of electrocardiographic ndings and improved function with
tions the INSPIRE study methodology. COPD 4(3): 17783, 2007. once-daily tiotropium in patients with chronic obstructive pulmonary
83. Wedzicha JA, Calverley PM, Seemungal TA, Hagan GW, Ansari Z, disease. Pharmacotherapy 25(12): 170818, 2005.
Stockley RA. The prevention of COPD exacerbations by salmeterol/ 104. Bailey WC, Tashkin DP. Pharmacologic therapy: Novel approaches
uticasone propionate or tiotropium bromide. Am J Respir Crit Care for chronic obstructive pulmonary disease. Proc Am Thorac Soc 4(7):
Med 177: 1926, 2007. 54348, 2007.
84. Calverley PM, Rennard SI. What have we learned from large drug 105. Hanania NA, Donohue JF. Pharmacologic interventions in chronic
treatment trials in COPD? Lancet 370(9589): 77485, 2007. obstructive pulmonary disease: Bronchodilators. Proc Am Thorac Soc
85. Anzueto A, Tashkin D, Menjoge S, Kesten S. One-year analysis of lon- 4(7): 52634, 2007.
gitudinal changes in spirometry in patients with COPD receiving tio- 106. Hansel TT, Neighbour H, Erin EM et al. Glycopyrrolate causes pro-
tropium. Pulm Pharmacol Ther 18(2): 7581, 2005. longed bronchoprotection and bronchodilatation in patients with
86. Decramer M et al. Clinical trial design considerations in assessing asthma. Chest 128(4): 197479, 2005.
long-term functional impacts of tiotropium in COPD: The Uplift trial. 107. Villetti G, Bergamaschi M, Bassani F et al. Pharmacological assess-
J Chronic Obstr Pulm Dis 1: 30312, 2004. ment of the duration of action of glycopyrrolate vs tiotropium and
87. Tashkin DP. The role of patient-centered outcomes in the course of ipratropium in guinea-pig and human airways. Br J Pharmacol 148(3):
chronic obstructive pulmonary disease: How long-term studies con- 29198, 2006.
tribute to our understanding. Am J Med 119(10 Suppl 1): 6372, 2006. 108. Ji F, Wu W, Dai X et al. Synthesis and pharmacological eects of new,
88. Friedman M, Menjoge SS, Anton SF, Kesten S. Healthcare costs with N-substituted soft anticholinergics based on glycopyrrolate. J Pharm
tiotropium plus usual care versus usual care alone following 1 year of Pharmacol 57: 142735, 2005.
treatment in patients with chronic obstructive pulmonary disorder 109. Pahl A, Bauhofer A, Petzold U et al. Synergistic eects of the anti-
(COPD). Pharmacoeconomics 22(11): 74149, 2004. cholinergic R,R-glycopyrrolate with anti-inammatory drugs. Biochem
89. Oostenbrink JB, Rutten-van Molken MP, Al MJ, van Noord JA, Pharmacol, 2006.
Vincken W. One-year cost-eectiveness of tiotropium versus ipratro- 110. Lu S, Parekh DD, Kuznetsova O, Green SA, Tozzi CA, Reiss TF.
pium to treat chronic obstructive pulmonary disease. Eur Respir J 23(2): An oral selective M3 cholinergic receptor antagonist in COPD.
24149, 2004. Eur Respir J 28(4): 77280, 2006.

626
Theophylline
50 CHAPTER

INTRODUCTION CHEMISTRY Peter J. Barnes


National Heart and Lung Institute
Theophylline remains one of the most widely Theophylline is a methylxanthine similar in
(NHLI), Clinical Studies Unit,
prescribed drugs for the treatment of asthma and structure to the common dietary xanthines
chronic obstructive pulmonary disease (COPD) caeine and theobromine. Several substituted Imperial College, London, UK
worldwide, since it is inexpensive and widely derivatives have been synthesized, but none
available. In many industrialized countries, has any advantage over theophylline [4]. The
however, theophylline has become a third-line 3-propyl derivative, enprofylline, is more potent
treatment that is only used in poorly control- as a bronchodilator and may have fewer toxic
led patients. This has been reinforced by various eects; however, its clinical development was
national and international guidelines on asthma halted because of hepatic toxicity problems
therapy. Some have even questioned whether [5]. Many salts of theophylline have also been
theophylline is indicated in any patients with marketed, the most common being aminophyl-
asthma [1], although others have emphasized the line, the ethylene diamine salt used to increase
special benecial eects of theophylline which solubility at neutral pH, so that intravenous
still give it an important place in management of administration is possible. Other salts, such as
asthma [2]. The frequency of side-eects at the choline theophyllinate, do not have any advan-
previously recommended doses and the relatively tage and others, such as acepifylline, are virtu-
low ecacy of theophylline have recently led to ally inactive [4].
reduced usage, since inhaled 2-agonists are far
more eective as bronchodilators and inhaled
corticosteroids have a greater anti-inammatory
eect. Despite the fact that theophylline has
been used in asthma therapy for over 70 years, MOLECULAR MECHANISMS
there is still considerable uncertainty about
its molecular mode of action in asthma and its
OF ACTION
logical place in therapy. Recently, novel mecha-
nisms of action that may account for the eec- Although theophylline has been in clinical use
tiveness of theophylline in severe asthma have for more than 70 years, its mechanism of action
been elucidated [3]. Because of problems with at a molecular level and its site of action remain
side eects, there have been attempts to improve uncertain, although there have been important
on theophylline, and recently there has been recent advances. Several molecular mecha-
increasing interest in selective phosphodieste- nisms of action have been proposed, many of
rase (PDE) inhibitors, which have the possibil- which appear to occur only at higher concentra-
ity of improving the benecial and reducing the tions of theophylline than are eective clinically
adverse eects of theophylline. (Table 50.1).

627
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 50.1 Proposed mechanisms of action of theophylline. Agonist Agonist

PDE inhibition (non-selective)

Adenosine receptor antagonism (A1-, A2A-, A2B-receptors) R GC


G AC
Inhibition of nuclear factor-B ( nuclear translocation)
Theophylline
Inhibition of phosphoinositide 3 kinase-

IL-10 secretion 
PDE3,4,7 PDE5,9
Apoptosis of inflammatory cells
ATP 3
,5
cAMP 3
,5
cGMP GTP
Poly(ADP-ribose)polymerase-1 (inhibits cell death) AMP GMP
 

Histone deacetylase activity ( efficacy of corticosteroids)

Bronchodilatation Inflammatory cells

PDE inhibition FIG. 50.1 Effect of PDE inhibitors in the breakdown of cyclic nucleotides
in airway smooth muscle and inflammatory cells. AC: adenylyl cyclase; ATP:
Theophylline is a weak and nonselective inhibitor of PDEs, adenosine triphosphate; cAMP: cyclic adenosine monophosphate; cGMP:
cyclic guanosine monophosphate; GC: guanylyl cyclase; Gs: stimulatory
which break down cyclic nucleotides in the cell, thereby
G-protein; GTP: guanosine triphosphate; PDE: phosphodiesterase; R:
leading to an increase in intracellular cyclic 3
5
adenosine receptor.
monophosphate (AMP) and cyclic 3
,5
guanosine mono-
phosphate (GMP) concentrations (Fig. 50.1); however, the
degree of inhibition is small at concentrations of theophyl-
line which are therapeutically relevant. Thus, total PDE Inhibition of PDEs is likely to account for some of
activity in human lung extracts is inhibited by only 510% the moist frequent side eects of theophylline, includ-
by therapeutic concentrations of theophylline [6]. There is ing nausea and vomiting (PDE4), palpitations and cardiac
convincing in vitro evidence that theophylline relaxes airway arrhythmias (PDE3), and headaches (PDE4).
smooth muscle by inhibition of PDE activity, but relatively
high concentrations are needed for maximal relaxation [7].
Similarly, the inhibitory eect of theophylline on media- Adenosine receptor antagonism
tor release from alveolar macrophages appears to be medi-
ated by inhibition of PDE activity in these cells [8]. There Theophylline is a potent inhibitor of adenosine receptors
is no evidence that airway smooth muscle or inammatory at therapeutic concentrations. Both A1- and A2-receptors
cells concentrate theophylline to achieve higher intracellular are inhibited, but theophylline is less eective against
than circulating concentrations. Inhibition of PDE should A3-receptors, suggesting that this could be the basis for its
lead to synergistic interaction with -agonists, but this has bronchodilator eects [13]. Although adenosine has lit-
not been convincingly demonstrated in vivo or in clini- tle eect on normal human airway smooth muscle in vitro,
cal studies; however, this might be because the relaxation it constricts airways of asthmatic patients via the release
of airway smooth muscle by -agonists may involve direct of histamine and leukotrienes, suggesting that adenos-
coupling of -receptors via a stimulatory G-protein to the ine releases mediators from mast cells [14]. The receptor
opening of potassium channels, without the involvement of involved appears to be an A3-receptor in rat mast cells
cyclic AMP [9]. [15, 16], but in humans there is evidence for the involve-
At least 11 isoenzyme families of PDE have now ment of an A2B-receptor [17, 18]. Adenosine causes bron-
been recognized, and some (PDE3, PDE4, PDE5) are choconstriction in asthmatic subjects when given by
important in smooth muscle relaxation [10]; however, inhalation [19]. The mechanism of bronchoconstriction is
there is no convincing evidence that theophylline has any indirect and involves release of histamine from airway mast
greater inhibitory eect on the PDE isoenzymes involved cells [14, 20]. The bronchoconstrictor eect of adenosine
in smooth muscle relaxation. It is possible that PDE isoen- is prevented by therapeutic concentrations of theophyl-
zymes may have an increased expression in asthmatic air- line [19]; however, this only conrms that theophylline is
ways, as a result of either the chronic inammatory process capable of antagonizing the eects of adenosine at thera-
or the therapy. Elevation of cyclic AMP by -agonists may peutic concentrations and does not necessarily indicate
result in increased PDE activity, thus limiting the eect of that this is important for its anti-asthma eect. Adenosine
-agonists. Indeed, alveolar macrophages from asthmatic antagonism is likely to account for some of the side eects
patients appear to have increased PDE activity [11]. This of theophylline, such as central nervous system stimulation,
would mean that theophylline might have a greater inhibi- cardiac arrhythmias (both via blockade of A1-receptors),
tory eect on PDE in asthmatic airways than in normal gastric hypersecretion, gastroesophageal reux, and diuresis.
airways. Support for this is provided by the lack of bron- A novel AMP receptor, P2Y15, has been identied which is
chodilator eect of theophylline in normal subjects, com- more potently inhibited by theophylline [21], although the
pared to a bronchodilator eect in asthmatic patients [12]. function of these receptors has been questioned.

628
Theophylline 50
IL-10 release of prostaglandin eects, and antagonism of tumor necrosis
factor-. These eects are generally seen only at high con-
IL-10 has a broad spectrum of anti-inammatory eects, centrations of theophylline which are above the therapeu-
and there is evidence that its secretion is reduced in COPD tic range and are therefore unlikely to contribute to the
[22]. IL-10 release is increased by theophylline, and this anti-inammatory actions of theophylline. Despite intense
eect may be mediated via PDE inhibition [23], although eorts, it has been dicult to nd any molecular mecha-
this has not been seen at the low doses that are eective in nisms that can account for the anti-inammatory eects
asthma [24]. of theophylline in asthma and COPD. Although several
potential mechanisms have been demonstrated in vitro,
there is little evidence that these occur at plasma con-
Effect on gene transcription centrations of 510 mg/l where clinical benet and anti-
inammatory eects are seen. A novel mechanism of action
Theophylline prevents the translocation of the proinam- involving activation of histone deacetylases (HDAC) has
matory transcription factor, nuclear factor-B (NF-B), been described which, in contrast to the proposed molecu-
into the nucleus, thus potentially reducing the expression of lar mechanisms discussed above, is seen at therapeutically
inammatory genes in asthma and COPD [25]. Inhibition relevant concentrations [3].
of NF-B appears to be due to a protective eect against
the degradation of the inhibitory protein I-B, so that
nuclear translocation of activated NF-B is prevented [26]. HDAC activation
However, these eects are seen at high concentrations and
may be mediated by inhibition of PDE. Expression of inammatory genes is regulated by the bal-
ance between histone acetylation and deacetylation [34]. In
asthma multiple inammatory genes are activated through
Effect on kinases proinammatory transcription factors, such as NF-B,
leading to histone acetylation and increased transcription.
Theophylline directly inhibits phosphoinositie-3-kinases, This process is reversed by the recruitment of HDAC to
with greatest potency for the PI3K (p110)- subtype (IC50 the activated inammatory gene promoter site within the
75 M) [27], a subtype of the enzyme that has been impli- nucleus. Corticosteroids suppress inammation by recruiting
cated in responses to oxidative stress [28]. However, it is HDAC2 to activated inammatory genes, thus switching o
a relatively weak eect against the PI3K- subtype (IC50 their expression [35] (see Chapter 53). This molecular mech-
800 M), which is involved in chemotactic responses of anism is defective in COPD patients as HDAC2 activity
neutrophils and monocytes. The inhibitory eect of theo- and expression are markedly reduced, thus accounting for
phylline on PI3K- may account for the ability of theophyl- the steroid resistance of COPD [36]. There is also a defect
line to reverse corticosteroid resistance, which may be of in HDAC2 function in patients with severe asthma and in
critical importance for its clinical eects in severe asthma asthmatic patients who smoke [37, 38]. We have shown that
and COPD. theophylline is an activator of HDACs and enhances the
anti-inammatory eect of corticosteroids, as well as revers-
ing steroid resistance in cells from COPD patients [39, 40]
Effects on apoptosis (Fig. 50.2). This action of theophylline is seen at low plasma
concentrations (optimally 5 mg/l) and is completely inde-
Prolonged survival of granulocytes due to a reduction in pendent of PDE inhibition and adenosine antagonism. The
apoptosis may be important in perpetuating chronic inam- eect of theophylline is completely blocked by an HDAC
mation in COPD. Theophylline promotes inhibits apoptosis inhibitor called trichostatin A and by knocking out HDAC2
in neutrophils in vitro [29]. This is associated with a reduc- using interference RNA [41]. The reason why theophyl-
tion in the anti-apoptotic protein Bcl-2 [30]. This eect is line selective activates HDAC activity is not yet known but
not mediated via PDE inhibition but in neutrophils may be appears to be indirect through the activation of kinase and
mediated by antagonism of adenosine A2A-receptors [31]. phosphatase pathways in the cell that are activated by oxida-
Theophylline also induces apoptosis of T-lymphocytes thus tive stress and involved in the regulation of HDAC2 activity.
reducing their survival, and this eect appears to be medi- This eect of theophylline is seen particularly in the presence
ated via PDE inhibition [32]. Theophylline also inhibits the of oxidative and nitrative stress, and this accounts for why
enzyme poly(ADP-ribose)polymerase-1 (PARP-1), which theophylline is eective particularly in severe asthma, where
is activated by oxidative stress and leads to a reduction in oxidative and nitrative stress are greatest. Increased reactive
nicotine adenine diamine levels resulting in an energy crisis oxygen species and nitric oxide from increased expression
that leads to cell death [33]. of inducible nitric oxide synthase result in the formation of
peroxynitrite radicals. Peroxynitrite is unstable and nitrates
tyrosine residues in proteins, which may result in altered
Other effects protein function. 3-Nitrotyrosine adducts are increased in
asthmatic airways [42]. Peroxynitrite is also increased in
Several other eects of theophylline have been described, COPD lungs [43] and is associated with tyrosine nitration
including an increase in circulating catecholamines, inhi- and inactivation of HDAC2 [44]. Theophylline also appears
bition of calcium inux into inammatory cells, inhibition to reduce the formation of peroxynitrite, and this provides

629
Asthma and COPD: Basic Mechanisms and Clinical Management

Inflammatory stimuli FIG. 50.2 Theophylline directly activates


(e.g. IL-1, TNF-) Corticosteroids Theophylline HDACs which deacetylate core histones
that have been acetylated by the histone
acetyltransferase (HAT) activity of co-
activators, such as CREB binding protein
(CBP). This results in suppression of
inflammatory genes and proteins, such
GR  as granulocyte-macrophage colony
p65
NF-B stimulating factor (GM-CSF) and IL-8 that
p50
Co-activators have been switched on by proinflammatory
Inflammatory protein transcription factors, such as nuclear factor-
(e.g. GM-CSF, IL-8) B (NF-B). Corticosteroids also activate
HDACs, but through a different mechanism
p65 CBP GR HDAC2 resulting in the recruitment of HDACs to
p50 the activated transcriptional complex via
HAT
Acetylation  Deacetylation activation of the glucocorticoid receptors
(GR) which function as a molecular bridge.
This predicts that theophylline and
corticosteroids may have a synergistic effect
in repressing inflammatory gene expression.
Inflammatory gene Gene Gene Inflammatory
transcription activation repression gene transcription

Inflammatory cells Structural cells


Eosinophil
Airway smooth muscle
Numbers
(apoptosis)
Bronchodilatation
T-Iymphocyte

Cytokines,
Traffic Endothelial cell

Mast cell Leak


Theophylline

Mediators

Macrophage

Cytokines
Respiratory muscles
Neutrophil

Recruitment Strength? FIG. 50.3 Multiple cellular effects of


theophylline.

a further mechanism for increasing HDAC2 function in shown, it is equally eective in large and small airways [46].
asthma [45]. In airways obtained at lung surgery 25% of preparations
fail to relax with a -agonist but all relax with theophyl-
line [47]. The molecular mechanism of bronchodilatation is
almost certainly related to PDE inhibition, resulting in an
CELLULAR EFFECTS increase in cyclic AMP [7]. The bronchodilator eect of the-
ophylline is reduced in human airways by the toxin charyb-
dotoxin, which inhibits large conductance Ca2-activated
Theophylline has several cellular eects that may contribute K channels (maxi-K channels), suggesting that theophyl-
to its clinical ecacy in the treatment of asthma (Fig. 50.3). line opens maxi-K channels via an increase in cyclic AMP
[48]. Theophylline acts as a functional antagonist and inhib-
its the contractile response of multiple spasmogens. In air-
Airway smooth muscle effects ways obtained at post-mortem from patients who have died
from asthma, the relaxant response to -agonists is reduced,
The primary eect of theophylline is assumed to be the whereas the bronchodilator response to theophylline is no
relaxation of airway smooth muscle; as in vitro studies have dierent from that seen in normal airways [49]. There is

630
Theophylline 50
evidence that -adrenoceptors in airway smooth muscle of In vivo theophylline inhibits mediator-induced airway
patients with fatal asthma become uncoupled [50], and the- microvascular leakage in rodents when given in high doses
ophylline may therefore have a theoretical advantage over [64], although this is not seen at therapeutically relevant
-agonists in severe asthma exacerbations. However, theo- concentrations [65]. Theophylline has an inhibitory eect
phylline is a weak bronchodilator at therapeutically relevant on plasma exudation in nasal secretions induced by allergen
concentrations, suggesting that some other target cell may in patients with allergic rhinitis, although this could be sec-
be more relevant for its anti-asthma eect. In human air- ondary to inhibition of mediator release [66].
ways the EC50 for airway smooth muscle relaxation by the- In allergen challenge studies, chronic oral treatment
ophylline is 1.5 104M, which is equivalent to 67 mg/l with theophylline reduces the late response to allergen
assuming 60% protein binding [47], that is above the thera- [67]. This has been interpreted as an eect on the chronic
peutic range. As discussed above, it is important to consider inammatory response and is supported by a reduced inl-
the possibility that PDE activity may be increased in asth- tration of eosinophils into the airways after allergen chal-
matic airways, so that theophylline may have a greater than lenge following low doses of theophylline [68]. In patients
expected eect. with nocturnal asthma low-dose theophylline inhibits the
In vivo intravenous aminophylline has an acute bron- inux of neutrophils and, to a lesser extent, eosinophils seen
chodilator eect in asthmatic patients, which is most likely in the early morning [69]. Chronic treatment with low-dose
to be due to a relaxant eect on airway smooth muscle [51]. theophylline reduces the numbers of eosinophils in bron-
The bronchodilator eect of theophylline in chronic asthma chial biopsies, bronchoalveolar lavage and induced sputum
is small in comparison with 2-agonists, however. Several of patients with mild asthma [62]. However, these eects
studies have demonstrated a small protective eect of theo- are less than those expected with low doses of inhaled corti-
phylline on histamine, methacholine, or exercise challenge costeroids, and there is no reduction of exhaled nitric oxide,
[5255]. This protective eect does not correlate well with indicating a lesser eect on suppression of inammation
any bronchodilator eect, and it is interesting that in some than corticosteroids.
studies the protective eect of theophylline is observed at In patients with COPD, theophylline reduces the
plasma concentrations of 10 mg/l. These clinical studies proportion of neutrophils in induced sputum and reduces
suggest that theophylline may have anti-asthma eects that the concentration of IL-8, suggesting that it may have an
are unrelated to any bronchodilator action. anti-inammatory eect unlike corticosteroids [70, 71].
Since patients with severe asthma may have increased neu-
trophils in the airways [72, 73], this may provide a mecha-
Anti-inflammatory effects nism whereby theophylline is eective as an add-on therapy
to high does of inhaled corticosteroids in these patients.
There is increasing evidence that theophylline has anti-
inammatory eects in asthma and that these are seen at
lower plasma concentrations than are needed for bron- Immunomodulatory effects
chodilatation. Theophylline inhibits mediator release from
chopped human lung [56], although high concentrations T-lymphocytes are now believed to play a central role in
are necessary and it is likely that this eect involves an coordinating the chronic inammatory response in asthma.
increase in cyclic AMP concentration due to PDE inhibi- Theophylline has been shown to have several actions on
tion. Theophylline also has an inhibitory eect on superox- T-lymphocyte function, suggesting that it might have an
ide anion release from human neutrophils [57] and inhibits immunomodulatory eect in asthma. Theophylline has a
the feedback stimulatory eect of adenosine on neutrophils stimulatory eect on suppressor (CD8) T-lymphocytes
in vivo [58]. At therapeutic concentrations in vitro, theo- which may be relevant to the control of chronic airway
phylline may increase superoxide release via an inhibitory inammation [74, 75], and has an inhibitory eect on graft
eect on adenosine receptors, since endogenous adenos- rejection [76]. In vitro theophylline inhibits IL-2 synthesis
ine may normally exert an inhibitory action on these cells in human T-lymphocytes, an eect that is secondary to a
[59]. Similar results are also seen in guinea pig and human rise in intracellular cyclic AMP concentration [77]. At high
eosinophils [60]. At therapeutic concentrations there is an concentrations theophylline inhibits proliferation in CD4
increased release of superoxide anions from eosinophils, and CD8 cells, an eect that is mediated via inhibition
which appears to be mediated via inhibition of adenosine of PDE4 [78]. Theophylline also inhibits the chemotactic
A2-receptors and is mimicked by the adenosine antagonist response of T-lymphocytes, an eect that is also mediated
8-phenyltheophylline. Inhibition of eosinophil superoxide through PDE inhibition [79]. In allergen-induced airway
generation occurs only at high concentrations of theophyl- inammation in guinea pigs, theophylline has a signicant
line (104M) which are likely to inhibit PDE. Similar inhibitory eect on eosinophil inltration [80], suggest-
results have also been obtained in human alveolar macro- ing that it may inhibit the T-cell-derived cytokines respon-
phages [8]. Macrophages in bronchoalveolar lavage uid sible for this eosinophilic response. Theophylline has been
from patients taking theophylline have been found to have reported to decrease circulating concentrations of IL-4 and
a reduced oxidative burst response [61], but there is no IL-5 in asthmatic patients [81]. In asthmatic patients low-
reduction in the release of the proinammatory cytokines dose theophylline treatment results in an increase in acti-
TNF- or GM-CSF [62]. Theophylline inhibits neutrophil vated circulating CD4 and CD8 T-cells, but a decrease in
chemotaxis via inhibition of adenosine A2A-receptors, and these cells in the airways, suggesting that it may reduce the
this may be relevant in severe asthma [63]. tracking of activated T-cells into the airways [82]. This is

631
Asthma and COPD: Basic Mechanisms and Clinical Management

supported by studies in allergen challenge, where low-dose TABLE 50.2 Factors affecting clearance of theophylline.
theophylline decreases the number of activated CD4 and
CD8 T-cells in bronchoalveolar lavage uid after allergen Increased clearance
challenge, and this is mirrored by an increase in these cells in Enzyme induction (rifampicin, phenobarbitone, ethanol)
peripheral blood [83]. These eects are seen even in patients
treated with high does of inhaled corticosteroids, indicating Smoking (tobacco, marijuana)
that the molecular eects of theophylline are likely to be dif-
High protein, low carbohydrate diet
ferent from those of corticosteroids. Theophylline induces
apoptosis of T-lymphocytes, thus reducing their survival Barbecued meat
[32]. This eect may be mediated via PDE4 inhibition so
may not be relevant to clinical doses of theophylline. The Childhood
therapeutic range of theophylline was based on measurement Decreased clearance
of immediate bronchodilatation in response to the acute
administration of theophylline [51]. However, it is possible Enzyme inhibition (cimetidine, erythromycin, ciprofloxacin,
that the nonbronchodilator eects of theophylline, which allopurinol, zileuton)
may reect some anti-inammatory or immunomodulatory
eect, may be exerted at lower plasma concentrations and Congestive heart failure
that dierent molecular mechanisms may be involved [84]. Liver disease

Pneumonia
Extrapulmonary effects
Viral infection
It has been suggested that theophylline may exert its eects Vaccination (immunization)
in asthma via some action outside the airways. It may be
relevant that theophylline is ineective when given by inha- High carbohydrate diet
lation until therapeutic plasma concentrations are achieved
Old age
[85]. This may indicate that theophylline has eects on cells
other than those in the airway.
An eect of theophylline which remains controversial
is its action on respiratory muscles. Aminophylline increases dierences in hepatic metabolism (Table 50.2). Theophylline
diaphragmatic contractility and reverses diaphragm fatigue is metabolized in the liver by the cytochrome P450 micro-
[86]. This eect has not been observed by all investigators, somal enzyme system, and a large number of factors may
and there are now doubts about the relevance of these obser- inuence hepatic metabolism. Theophylline is predomi-
vations to the clinical benet provided by theophylline [87]. nantly metabolized by the CYP1A2 enzyme, while at higher
plasma concentrations CYP2E1 is also involved [89].

PHARMACOKINETICS Increased clearance


Increased clearance is seen in children (116 years), and in
There is a close relationship between the acute improvement cigarette and marijuana smokers. Concurrent administra-
in airway function and the serum theophylline concentra- tion of phenytoin and phenobarbitone increases activity of
tion. Below 10 mg/l therapeutic eects (at least in terms of P450, resulting in increased metabolic breakdown, so that
rapid improvement in airway function) are small, and above higher doses may be required.
25 mg/l additional benets are outweighed by side eects, so
that the therapeutic range was usually taken as 1020 mg/l
(55110 M) [4]. It is now apparent that nonbronchodilator Reduced clearance
eects of theophylline may be seen at plasma concentrations
of 10 mg/l and that clinical benet may be derived from Reduced clearance is found in liver disease, pneumonia,
these lower concentrations of theophylline. This suggests and heart failure and doses need to be reduced to half and
that it may be necessary to redene the therapeutic range plasma levels monitored carefully [90]. Decreased clearance
of theophylline based on anti-asthma eect, rather than the is also seen with certain drugs, including erythromycin, cer-
acute bronchodilator response that requires a higher plasma tain quinolone antibiotics (ciprooxacin, but not ooxacin),
concentration. The dose of theophylline required to give allopurinol, cimetidine (but not ranitidine), serotonin uptake
therapeutic concentrations varies among patients, largely inhibitors (uvoxamine), and the 5-lipoxygenase inhibitor
because of dierences in clearance. In addition, there may zileuton, which interfere with CYP1A2 function. Thus, if
be dierences in bronchodilator response to theophylline a patient on maintenance theophylline requires a course of
and, with acute bronchoconstriction, higher concentrations erythromycin, the dose of theophylline should be halved. Viral
may be required to produce bronchodilatation [88]. infections and vaccinations (immunizations) may also reduce
Theophylline is rapidly and completely absorbed, but clearance, and this may be particularly important in children.
there are large inter-individual variations in clearance, due to Because of these variations in clearance, individualization of

632
Theophylline 50
theophylline dosage is required and plasma concentrations has been questioned in view of the risk of adverse eects
should be measured 4 h after the last dose with slow-release compared with nebulized 2-agonists. In patients with
preparations, when steady state has usually been achieved. acute asthma, intravenous aminophylline is less eective
There is no signicant circadian variation in theophylline than nebulized 2-agonists [96] and should therefore be
metabolism [91], although there may be delayed absorption reserved for those who fail to respond to -agonists. In a
at night, which may relate to the supine posture [92]. meta-analysis of 27 studies which looked at addition of
intravenous aminophylline to nebulized 2-agonists, there
is no evidence for signicant benet in adults [97] or chil-
dren [98]. This indicates that aminophylline should not be
ROUTES OF ADMINISTRATION added routinely to nebulized -agonists. Indeed, addition of
aminophylline only increases adverse eects. Several deaths
have been reported after intravenous aminophylline. In one
Intravenous study of 43 asthma deaths in southern England, there was
a signicantly greater frequency of toxic theophylline con-
Intravenous aminophylline has been used for many years centrations (21%) compared with matched controls (7%)
in the treatment of acute severe asthma. The recommended [99]. These concerns have lead to the view that intravenous
dose is now 6 mg/kg given intravenously over 2030 min, aminophylline should be reserved for the few patients with
followed by a maintenance dose of 0.5 mg/kg/h. If the acute severe asthma who fail to show a satisfactory response
patient is already taking theophylline, or there are any fac- to nebulized 2-agonists. When intravenous aminophylline
tors that decrease clearance, these doses should be halved is used, it should be given as a slow intravenous infusion
and the plasma level checked more frequently. with careful monitoring of vital signs, and plasma theophyl-
line concentrations should be measured prior to and after
infusion.
Oral Aminophylline similarly has no place in the routine
management of COPD exacerbations [100, 101].
Plain theophylline tablets or elixir, which are rapidly
absorbed, give wide uctuations in plasma levels and are not
recommended. Several eective sustained-release prepara- Chronic asthma
tions now available are absorbed at a constant rate and pro-
vide steady plasma concentrations over a 1224 h period [93]. In most guidelines for asthma management theophylline
Although there are dierences between preparations, these is recommended as an additional bronchodilator if asthma
are relatively minor and of no clinical signicance. Both remains dicult to control after high doses of inhaled
slow-release aminophylline and theophylline are available and corticosteroids. The introduction of long-acting inhaled
are equally eective (although the ethylene diamine compo- 2-agonists has further threatened the position of theo-
nent of aminophylline has very occasionally been implicated phylline, since the side eects of these agents may be less
in allergic reactions). For continuous treatment, twice daily frequent than those associated with theophylline, and long-
therapy (~8 mg/kg b.i.d.) is needed, although some prepara- acting inhaled 2-agonists are more eective controllers
tions are designed for once daily administration. For noc- than theophylline [102]. Whether theophylline has some
turnal asthma, a single dose of slow-release theophylline at additional benet over its bronchodilator action is now an
night is eective [94, 95], and more eective than an oral important consideration. In chronic asthma oral theophyl-
slow-release -agonist preparation. Once optimal doses have line appears to be as eective as cromolyn sodium in control-
been determined, plasma concentrations usually remain sta- ling young allergic asthmatics [103] and provides additional
ble, providing no factors which alter clearance are introduced. control of asthma symptoms even in patients talking regu-
lar inhaled steroids [104]. In one study a group of adoles-
Other routes cent patients with severe asthma who were controlled with
oral and inhaled steroids, nebulized 2-agonists, inhaled
Aminophylline may be given as a suppository, but rectal anticholinergics, and cromolyn, in addition to regular oral
absorption is unreliable and proctitis may occur, so this route theophylline, withdrawal of the oral theophylline resulted
should be avoided. Inhalation of theophylline is irritating in a marked deterioration of asthma control which could
and ineective [85]. Intramuscular injections of theophylline not be controlled by further increase in steroids and only
are very painful and should never be given. responded to reintroduction of theophylline [105]. This sug-
gests that there may be a group of severe asthmatic patients
who particularly benet from theophylline. In a control-
led trial of theophylline withdrawal in patients with severe
CLINICAL USE asthma controlled only on high doses of inhaled corticoster-
oids, there was a signicant deterioration in symptoms and
lung function when placebo was substituted for the rela-
Acute exacerbations tively low maintenance dose of theophylline [82]. There is
also evidence that addition of theophylline improves asthma
Intravenous aminophylline has been used in the manage- control to a greater extent than 2-agonists in patients with
ment of acute severe asthma for over 50 years, but this use severe asthma treated with high-dose inhaled steroids [106].

633
Asthma and COPD: Basic Mechanisms and Clinical Management

This suggests that theophylline may have a useful place in thus reverses corticosteroid resistance [40]. It also reduces
the optimal management of moderate-to-severe asthma and nitrative stress in macrophages from patients with COPD,
appears to provide additional control above that provided by whereas high doses of an inhaled corticosteroid are without
high-dose inhaled steroids [107, 108]. eect [45]. This suggests that corticosteroids may be use-
Theophylline may be a useful treatment for noctur- ful in reversing corticosteroids resistance in patients with
nal asthma, and a single dose of a slow-release theophylline COPD, but long-term clinical trials are now needed to con-
preparation given at night may provide eective control of rm this [120, 121].
nocturnal asthma symptoms [94, 109]. There is evidence
that slow-release theophylline preparations are more eec-
tive than slow-release oral -agonists and inhaled -agonists Interaction with 2-agonists
in controlling nocturnal asthma [95, 110, 111]. Theophylline
has equal ecacy to salmeterol in controlling nocturnal If theophylline exerts its eects by PDE inhibition, a syner-
asthma, but the quality of sleep is better with salmeterol gistic interaction with -agonists would be expected. Many
compared to theophylline [112]. The mechanism of action studies have investigated this possibility, but while there is
of theophylline in nocturnal asthma may involve more than good evidence that theophylline and -agonists have addi-
long-lasting bronchodilatation and could involve inhibition tive eects, true synergy is not seen [122]. This can now be
of some components of the inammatory response, which understood in terms of the molecular mechanisms of action
may increase at night [69]. of -agonists and theophylline. -Agonists may cause
relaxation of airway smooth muscle via several mechanisms.
Classically, they increase intracellular cyclic AMP concen-
Add-on therapy trations, which were believed to be an essential event in
the relaxation response. It has recently become clear that
Several studies have demonstrated that adding low-dose -agonists may cause bronchodilatation, at least in part, by
theophylline to inhaled corticosteroids in patients who are opening maxi-K channels in airway smooth muscle cells
not controlled gives better asthma control than doubling the [9, 48]. Maxi-K channels are opened by low concentrations
dose of inhaled corticosteroids. This has been demonstrated of 2-agonists which are likely to be therapeutically rele-
in patients with moderate-to-severe and mild asthma vant. There is now evidence that -receptors may be coupled
[113115]. Interestingly, there is a greater degree of improve- directly to maxi-K channels via the -subunit of Gs [123],
ment in forced vital capacity than in FEV1, possibly indicat- and therefore may induce relaxation without any increase in
ing an eect on peripheral airways. Since the improvement cyclic AMP, thus accounting for a lack of synergy.
in lung function was relatively slow, this suggests that the Repeated administration of 2-agonists may result
eect of the added theophylline may be having an anti- in tolerance; however, this may be explained by down-
inammatory rather than a bronchodilator eect, particularly regulation of 2-receptors, an additional mechanism may
as the plasma concentration of theophylline in these studies involve upregulation of PDE enzymes (especially PDE4D)
was 10 mg/ml. These studies suggest that low-dose theo- which then break down cyclic AMP more readily [124].
phylline may be preferable to increase the dose of inhaled Theophylline may therefore theoretically prevent the devel-
steroids when asthma is not controlled on moderate doses of opment of tolerance. However, in a clinical study theo-
inhaled steroids; such a therapeutic approach would be much phylline failed to prevent the development of tolerance to
less expensive than adding long-acting inhaled 2-agonists. the bronchoprotective eect of salmeterol in asthmatic
However, theophylline is a less eective option than adding patients [125].
a long-acting inhaled 2-agonist [102].

Chronic obstructive pulmonary disease SIDE EFFECTS


Theophylline may also benet patients with COPD and
increases exercise tolerance [116, 117]. Theophylline reduces There is no doubt that theophylline provides clinical ben-
air trapping, suggesting an eect on peripheral airways, et in obstructive airway disease, but the main limitation to
and this may explain why some patients with COPD may its use is the frequency of adverse eects [126]. Unwanted
obtain considerable symptomatic improvement without any eects of theophylline are usually related to plasma concen-
increase in spirometric values [118]. Although the eect of tration and tend to occur when plasma levels exceed 20 mg/l;
theophylline on respiratory muscle weakness was believed however, some patients develop side-eects even at low plasma
to be important in contributing to symptomatic improve- concentrations. To some extent side eects may be reduced by
ment in patients with COPD [86], this seems unlikely as gradually increasing the dose until therapeutic concentrations
several investigators have failed to conrm any eect on are achieved.
respiratory muscle function at therapeutic concentrations of The commonest side eects are headache, nausea and
theophylline [87]. The demonstration that low doses of the- vomiting, abdominal discomfort, and restlessness. There may
ophylline reduce neutrophils in induced sputum of patients also be increased acid secretion, gastroesophageal reux, and
with COPD suggests that theophylline may have some diuresis. There has recently been concern that theophylline,
anti-inammatory eect [71, 119]. In COPD macrophages even at therapeutic concentrations, may lead to behavioral
in vitro theophylline restores HDAC activity to normal and disturbance and learning diculties in school children [127],

634
Theophylline 50
although it is dicult to design adequate controls for such 4. Weinburger M. The pharmacology and therapeutic use of theophylline.
studies. At high concentrations convulsions and cardiac J Allergy Clin Immunol 73: 52540, 1984.
arrhythmias may occur and, as stated previously, there is 5. Persson CGA. Development of safer xanthine drugs for the treatment
of obstructive airways disease. J Allergy Clin Immunol 78: 81724, 1986.
concern that intravenous aminophylline administered in the
6. Poolson JB, Kazanowski JJ, Goldman AL, Szentivanyi A. Inhibition of
emergency room may be a contributory factor to the deaths human pulmonary phosphodiesterase activity by therapeutic levels of
of some patients with severe asthma [99]. theophylline. Clin Exp Pharmacol Physiol 5: 53539, 1978.
Some of the side-eects of theophylline (central stim- 7. Rabe KF, Magnussen H, Dent G. Theophylline and selective PDE
ulation, gastric secretion, diuresis, and arrhythmias) may be inhibitors as bronchodilators and smooth muscle relaxants. Eur Respir J
due to adenosine receptor antagonism (A1A receptors), and 8: 63742, 1995.
these may therefore be avoided by PDE inhibitors, as dis- 8. Dent G, Giembycz MA, Rabe KF, Wolf B, Barnes PJ, Magnussen H.
cussed above. The commonest side eects of theophylline Theophylline suppresses human alveolar macrophage respiratory burst
are nausea and headaches, which may be due to inhibition through phosphodiesterase inhibition. Am J Respir Cell Mol Biol 10:
of certain PDEs (e.g. PDE4 in the vomiting center) and 56572, 1994.
9. Kume H, Hall IP, Washabau RJ, Takagi K, Kotliko MI. Adrenergic
cardiac arrhythmias due to inhibition of PDE3 [128].
agonists regulate KCa channels in airway smooth muscle by cAMP-
dependent and -independent mechanisms. J Clin Invest 93:
37179, 1994.
10. Beavo JA. Cyclic nucleotide phosphodiesterases: Functional implica-
FUTURE OF THEOPHYLLINE tions of multiple isoforms. Physiol Rev 75: 72548, 1995.
11. Bachelet M, Vincent D, Havet N, Marrash-Chahla R, Pradalier A, Dry J
et al. Reduced responsiveness of adenylate cyclase in alveolar macrophages
Although theophylline has recently been used much less from patients with asthma. J Allergy Clin Immunol 88: 32228, 1991.
in developed countries, there are reasons for thinking that 12. Estenne M, Yernault J, De Troyer A. Eects of parenteral aminophyl-
line on lung mechanics in normal humans. Am Rev Respir Dis 121:
it may come back in fashion for the treatment of chronic
96771, 1980.
asthma and COPD, with the recognition that it may have 13. Pauwels RA, Joos GF. Characterization of the adenosine receptors in
an anti-inammatory and immunomodulatory eect when the airways. Arch Int Pharmacodyn Ther 329: 15156, 1995.
given in low doses (plasma concentration 510 mg/l) [3]. 14. Bjrk T, Gustafsson LE, Dahln S-E. Isolated bronchi from asthmat-
At these low doses the drug is easier to use, side eects are ics are hyperresponsive to adenosine, which apparently acts indirectly
uncommon, and the problems of drug interaction are less of by liberation of leukotrienes and histamine. Am Rev Respir Dis 145:
a problem, thus making the clinical use of theophylline less 108791, 1992.
complicated. Theophylline appears to have an eect that is 15. Fozard JR, Pfannkuche HJ, Schuurman HJ. Mast cell degranulation
dierent from those of corticosteroids, and it may there- following adenosine A3 receptor activation in rats. Eur J Pharmacol
fore be a useful drug to combine with low-dose inhaled 298: 29397, 1996.
16. Hannon JP, Tigani B, Williams I, Mazzoni L, Fozard JR. Mechanism
steroids. The molecular mechanism of anti-inammatory
of airway hyperresponsiveness to adenosine induced by allergen chal-
eects of theophylline is now becoming clearer, and it lenge in actively sensitized Brown Norway rats. Br J Pharmacol 132:
seems likely that there is a synergistic interaction with the 150923, 2001.
anti-inammatory mechanism of corticosteroids through 17. Feoktistov I, Biaggioni I. Pharmacological characterization of adenos-
restoration of HDAC activity. This interaction may under- ine A2B receptors: Studies in human mast cells co-expressing A2A and
lie the benecial eects of theophylline when added to A2B adenosine receptor subtypes. Biochem Pharmacol 55: 62733, 1998.
inhaled corticosteroids. This may be particularly appropriate 18. Holgate ST. The identication of the adenosine A2B receptor as a novel
in patients with more severe asthma in whom corticoster- therapeutic target in asthma. Br J Pharmacol 145: 100915, 2005.
oids are less eective as there may be a reduction in HDAC 19. Cushley MJ, Tatterseld AE, Holgate ST. Adenosine-induced bron-
activity in these patients [37] as well as in smoking asth- choconstriction in asthma: Antagonism by inhaled theophylline. Am
Rev Respir Dis 129: 38084, 1984.
matics patients and patients with COPD. As slow-release
20. Cushley MJ, Holgate ST. Adenosine induced bronchoconstriction in
theophylline preparations are cheaper than long-acting asthma: Role of mast cell mediator release. J Allergy Clin Immunol 75:
inhaled 2-agonists and leukotriene modiers, this may jus- 27278, 1985.
tify the choice of low-dose theophylline as the add-on ther- 21. Inbe H, Watanabe S, Miyawaki M, Tanabe E, Encinas JA.
apy for asthma control. In addition, compliance with oral Identication and characterization of a cell-surface receptor, P2Y15,
therapy is likely to be greater than with inhaled therapies for AMP and adenosine. J Biol Chem 279: 1979099, 2004.
[129]. This suggests that low-dose theophylline may nd an 22. Takanashi S, Hasegawa Y, Kanehira Y, Yamamoto K, Fujimoto K,
important place in modern asthma management in patients Satoh K et al. Interleukin-10 level in sputum is reduced in bronchial
with moderate asthma as well as with severe asthma. asthma, COPD and in smokers. Eur Respir J 14: 30914, 1999.
23. Mascali JJ, Cvietusa P, Negri J, Borish L. Anti-inammatory eects of
theophylline: Modulation of cytokine production. Ann Allergy Asthma
Immunol 77: 3438, 1996.
References 24. Oliver B, Tomita K, Keller A, Caramori G, Adcock I, Chung KF et al.
Low-dose theophylline does not exert its anti-inammatory eects
1. Lam A, Newhouse MT. Management of asthma and chronic airow in mild asthma through upregulation of interleukin-10 in alveolar
limitation. Are methylxanthines obsolete? Chest 98: 4452, 1990. macrophages. Allergy 56: 108790, 2001.
2. Weinberger M, Hendeles L. Theophylline in asthma. N Engl J Med 25. Tomita K, Chikumi H, Tokuyasu H, Yajima H, Hitsuda Y, Matsumoto Y
334: 138088, 1996. et al. Functional assay of NF-kappaB translocation into nuclei by laser scan-
3. Barnes PJ. Theophylline: New perspectives on an old drug. Am J Respir ning cytometry: Inhibitory eect by dexamethasone or theophylline. Naunyn
Crit Care Med 167: 81318, 2003. Schmiedebergs Arch Pharmacol 359: 24955, 1999.

635
Asthma and COPD: Basic Mechanisms and Clinical Management

26. Ichiyama T, Hasegawa S, Matsubara T, Hayashi T, Furukawa S. 46. Finney MJB, Karlson JA, Persson CGA. Eects of bronchoconstriction
Theophylline inhibits NF-kB activation and IkBa degradation in and bronchodilation on a novel human small airway preparation. Br J
human pulmonary epithelial cells. Naunyn Schmiedebergs Arch Pharmacol Pharmacol 85: 2936, 1985.
364: 55861, 2001. 47. Guillot C, Fornaris M, Badger M, Orehek J. Spontaneous and pro-
27. Foukas LC, Daniele N, Ktori C, Anderson KE, Jensen J, Shepherd PR. voked resistance to isoproterenol in isolated human bronchi. J Allergy
Direct eects of caeine and theophylline on p110 delta and other Clin Immunol 74: 71318, 1984.
phosphoinositide 3-kinases. Dierential eects on lipid kinase and 48. Miura M, Belvisi MG, Stretton CD, Yacoub MH, Barnes PJ. Role of
protein kinase activities. J Biol Chem 277: 3712430, 2002. potassium channels in bronchodilator responses in human airways. Am
28. Yamamori T, Inanami O, Nagahata H, Kuwabara M. Phosphoinositide Rev Respir Dis 146: 13236, 1992.
3-kinase regulates the phosphorylation of NADPH oxidase compo- 49. Goldie RG, Spina D, Henry PJ, Lulich KM, Paterson JW. In vitro
nent p47(phox) by controlling cPKC/PKCdelta but not Akt. Biochem responsiveness of human asthmatic bronchus to carbachol, histamine,
Biophys Res Commun 316: 72030, 2004. beta-adrenoceptor agonists and theophylline. Br J Clin Pharmacol 22:
29. Yasui K, Hu B, Nakazawa T, Agematsu K, Komiyama A. Theophylline 66976, 1986.
accelerates human granulocyte apoptosis not via phosphodiesterase 50. Bai TR, Mak JCW, Barnes PJ. A comparison of beta-adrenergic
inhibition. J Clin Invest 100: 167784, 1997. receptors and in vitro relaxant responses to isoproterenol in asthmatic
30. Chung IY, Nam-Kung EK, Lee NM, Chang HS, Kim DJ, Kim YH et al. airway smooth muscle. Am J Respir Cell Mol Biol 6: 64751, 1992.
The downregulation of bcl-2 expression is necessary for theophylline- 51. Mitenko PA, Ogilvie RI. Rational intravenous doses of theophylline. N
induced apoptosis of eosinophil. Cell Immunol 203: 95102, 2000. Engl J Med 289: 6003, 1973.
31. Yasui K, Agematsu K, Shinozaki K, Hokibara S, Nagumo H, 52. McWilliams BC, Menendez R, Kelly WH, Howick J. Eects of theo-
Nakazawa T et al. Theophylline induces neutrophil apoptosis through phylline on inhaled methacholine and histamine in asthmatic children.
adenosine A2A receptor antagonism. J Leukoc Biol 67: 52935, 2000. Am Rev Respir Dis 130: 19397, 1984.
32. Ohta K, Yamashita N. Apoptosis of eosinophils and lymphocytes in 53. Cartier A, Lemire I, LArcheveque J. Theophylline partially inhib-
allergic inammation. J Allergy Clin Immunol 104: 1421, 1999. its bronchoconstriction caused by inhaled histamine in subjects with
33. Moonen HJ, Geraets L, Vaarhorst A, Bast A, Wouters EF, Hageman GJ. asthma. J Allergy Clin Immunol 77: 57075, 1986.
Theophylline prevents NAD depletion via PARP-1 inhibition in 54. Magnusson H, Reuss G, Jorres R. Theophylline has a dose-related
human pulmonary epithelial cells. Biochem Biophys Res Commun 338: eect on the airway response to inhaled histamine and methacholine
180510, 2005. in asthmatics. Am Rev Respir Dis 136: 116367, 1987.
34. Barnes PJ, Adcock IM, Ito K. Histone acetylation and deacetyla- 55. Magnussen H, Reuss G, Jrres R. Methylxanthines inhibit exercise-
tion: Importance in inammatory lung diseases. Eur Respir J 25: induced bronchoconstriction at low serum theophylline concentra-
55263, 2005. tions and in a dose-dependent fashion. J Allergy Clin Immunol 81:
35. Barnes PJ. How corticosteroids control inammation. Br J Pharmacol 53137, 1988.
148: 24554, 2006. 56. Orange RP, Kaliner MA, Laraia PJ, Austen KF. Immunological
36. Ito K, Ito M, Elliott WM, Cosio B, Caramori G, Kon OM et al. release of histamine and slow reacting substance of anaphylaxis from
Decreased histone deacetylase activity in chronic obstructive pulmo- human lung. II. Inuence of cellular levels of cyclic AMP. Fed Proc 30:
nary disease. N Engl J Med 352: 196776, 2005. 172529, 1971.
37. Hew M, Bhavsar P, Torrego A, Meah S, Khorasani N, Barnes PJ et al. 57. Nielson CP, Crawley JJ, Morgan ME, Vestal RE. Polymorphonuclear
Relative corticosteroid insensitivity of peripheral blood mononuclear leukocyte inhibition by therapeutic concentrations of theophylline is
cells in severe asthma. Am J Respir Crit Care Med 174: 13441, 2006. mediated by cyclic 3
,5
adenosine aminophosphate. Am Rev Respir
38. Murahidy A, Ito M, Adcock IM, Barnes PJ, Ito K. Reduction is his- Dis 137: 2530, 1988.
tone deacetylase expression and activity in smoking asthmatics: A 58. Kraft M, Pak J, Borish L, Martin RJ. Theophyllines eect on neu-
mechanism of steroid resistance. Proc Am Thorac Soc 2: A889, 2005. trophil function and the late asthmatic response. J Allergy Clin Immunol
39. Ito K, Lim S, Caramori G, Cosio B, Chung KF, Adcock IM et al. A 98: 25157, 1996.
molecular mechanism of action of theophylline: Induction of histone 59. Schrier DJ, Imre RM. The eects of adenosine antagonists on human
deacetylase activity to decrease inammatory gene expression. Proc Natl neutrophil function. J Immunol 137: 328489, 1986.
Acad Sci USA 99: 892126, 2002. 60. Yukawa T, Kroegel C, Dent G, Chanez P, Ukena D, Barnes PJ. Eect
40. Cosio BG, Tsaprouni L, Ito K, Jazrawi E, Adcock IM, Barnes PJ. of theophylline and adenosine on eosinophil function. Am Rev Respir
Theophylline restores histone deacetylase activity and steroid responses Dis 140: 32733, 1989.
in COPD macrophages. J Exp Med 200: 68995, 2004. 61. ONeill SJ, Sitar DS, Kilass DJ. The pulmonary disposition of theo-
41. Ito K, Yamamura S, Essile-Quaye S, Cosio B, Ito M, Barnes PJ et al. phylline and its inuences on human alveolar macrophage bactericidal
Histone deacetylase 2-mediated deacetylation of the glucocorticoid function. Am Rev Respir Dis 134: 122528, 1988.
receptor enables NF-kB suppression. J Exp Med 203: 713, 2006. 62. Lim S, Tomita K, Carramori G, Jatakanon A, Oliver B, Keller A et al.
42. Saleh D, Ernst P, Lim S, Barnes PJ, Giaid A. Increased formation of Low-dose theophylline reduces eosinophilic inammation but not
the potent oxidant peroxynitrite in the airways of asthmatic patients exhaled nitric oxide in mild asthma. Am J Respir Crit Care Med 164:
is associated with induction of nitric oxide synthase: Eect of inhaled 27376, 2001.
glucocorticoid. FASEB J 12: 92937, 1998. 63. Yasui K, Agematsu K, Shinozaki K, Hokibara S, Nagumo H, Yamada S
43. Ricciardolo FL, Caramori G, Ito K, Capelli A, Brun P, Abatangelo G et al. Eects of theophylline on human eosinophil functions:
et al. Nitrosative stress in the bronchial mucosa of severe chronic Comparative study with neutrophil functions. J Leukoc Biol 68:
obstructive pulmonary disease. J Allergy Clin Immunol 116: 102835, 194200, 2000.
2005. 64. Erjefalt I, Persson CGA. Pharmacologic control of plasma exudation
44. Ito K, Tomita T, Barnes PJ, Adcock IM. Oxidative stress reduces histone into tracheobronchial airways. Am Rev Respir Dis 143: 100814, 1991.
deacetylase (HDAC)2 activity and enhances IL-8 gene expression: Role 65. Boschetto P, Roberts NM, Rogers DF, Barnes PJ. The eect of anti-
of tyrosine nitration. Biochem Biophys Res Commun 315: 24045, 2004. asthma drugs on microvascular leak in guinea pig airways. Am Rev
45. Hirano T, Yamagata T, Gohda M, Yamagata Y, Ichgikawa T, Respir Dis 139: 41621, 1989.
Yanagisawa S et al. Inhibition of reactive nitrogen species production in 66. Naclerio RM, Bartenfelder D, Proud D, Togias AG, Meyers DA,
COPD airways: Comparison between inhaled corticosteroid and oral Kagey Sobotka A et al. Theophylline reduces histamine release during
theophylline. Thorax 61: 76166, 2006. pollen-induced rhinitis. J Allergy Clin Immunol 78: 87476, 1986.

636
Theophylline 50
67. Ward AJM, McKenni M, Evans JM, Page CP, Costello JF. 89. Zhang ZY, Kaminsky LS. Characterization of human cytochromes
Theophylline an immunomodulatory role in asthma? Am Rev Respir P450 involved in theophylline 8-hydroxylation. Biochem Pharmacol 50:
Dis 147: 51823, 1993. 20511, 1995.
68. Sullivan P, Bekir S, Jaar Z, Page C, Jeery P, Costello J. Anti- 90. Jusko WJ, Gardner MJ, Mangiore A, Schentag JJ, Kopp JR, Vance JW.
inammatory eects of low-dose oral theophylline in atopic asthma. Factors aecting aminophylline clearance: Age, tobacco, marijuana,
Lancet 343: 10068, 1994. cirrhosis, congestive heart failure, obesity, oral contraceptives, benzo-
69. Kraft M, Torvik JA, Trudeau JB, Wenzel SE, Martin RJ. Theophylline: diazepines, barbiturates and ethanol. J Pharm Sci 68: 135866, 1979.
Potential antiinammatory eects in nocturnal asthma. J Allergy Clin 91. Taylor DR, Run D, Kinney CD, McDevitt DG. Investigation of
Immunol 97: 124246, 1996. diurnal changes in the disposition of theophylline. Br J Clin Pharmacol
70. Culpitt S, Maziak W, Loukides S, Keller A, Barnes PJ. Eect of theo- 16: 41316, 1983.
phylline on induced sputum inammatory indices in COPD patients. 92. Warren JB, Cuss F, Barnes PJ. Posture and theophylline kinetics. Br J
Am J Respir Crit Care Med 157: A797, 1997. Clin Pharmacol 19: 7079, 1985.
71. Kobayashi M, Nasuhara Y, Betsuyaku T, Shibuya E, Tanino Y, Tanino M 93. Weinberger M, Hendeles L. Slow-release theophylline: Rationale and
et al. Eect of low-dose theophylline on airway inammation in basis for product selection. N Engl J Med 308: 76063, 1983.
COPD. Respirology 9: 24954, 2004. 94. Barnes PJ, Greening AP, Neville L, Timmers J, Poole GW. Single
72. Wenzel SE, Szeer SJ, Leung DY, Sloan SI, Rex MD, Martin RJ. dose slow-release aminophylline at night prevents nocturnal asthma.
Bronchoscopic evaluation of severe asthma. Persistent inammation Lancet i: 299301, 1982.
associated with high dose glucocorticoids. Am J Respir Crit Care Med 95. Heins M, Kurtin L, Oellerich M, Maes R, Sybrecht GW. Nocturnal
156: 73743, 1997. asthma: Slow-release terbutaline versus slow-release theophylline
73. Jatakanon A, Uasaf C, Maziak W, Lim S, Chung KF, Barnes PJ. therapy. Eur Respir J 1: 30610, 1988.
Neutrophilic inammation in severe persistent asthma. Am J Respir 96. Bowler SD, Mitchell CA, Armstrong JG. Nebulised fenoterol and i.v.
Crit Care Med 160: 153239, 1999. aminophylline in acute severe asthma. Eur Resp J 70: 28083, 1987.
74. Shohat B, Volovitz B, Varsano I. Induction of suppressor T cells in asth- 97. Parameswaran K, Belda J, Rowe BH. Addition of intravenous ami-
matic children by theophylline treatment. Clin Allergy 13: 48793, 1983. nophylline to beta2-agonists in adults with acute asthma. Cochrane.
75. Fink G, Mittelman M, Shohat B, Spitzer SA. Theophylline-induced Database Syst Rev: 2000, CD002742.
alterations in cellular immunity in asthmatic patients. Clin Allergy 17: 98. Mitra A, Bassler D, Goodman K, Lasserson TJ, Ducharme FM.
31316, 1987. Intravenous aminophylline for acute severe asthma in children over
76. Guillou PJ, Ramsden C, Kerr M, Davison AM, Giles GR. A prospec- two years receiving inhaled bronchodilators. Cochrane Database Syst
tive controlled clinical trial of aminophylline as an adjunct immuno- Rev: 2005, CD001276.
suppressive agent. Transplant Proc 16: 121820, 1984. 99. Eason J, Makowe HLJ. Aminophylline toxicity how many hospital
77. Didier M, Aussel C, Ferrua B, Fehlman M. Regulation of interleukin 2 asthma deaths does it cause?. Respir Med 83: 21926, 1989.
synthesis by cAMP in human T cells. J Immunol 139: 117984, 1987. 100. Duy N, Walker P, Diamantea F, Calverley PM, Davies L.
78. Giembycz MA, Corrigan CJ, Seybold J, Newton R, Barnes PJ. Intravenous aminophylline in patients admitted to hospital with non-
Identication of cyclic AMP phosphodiesterases 3, 4 and 7 in human acidotic exacerbations of chronic obstructive pulmonary disease: A
CD4 and CD8 T-lymphocytes. Br J Pharmacol 118: 194558, 1996. prospective randomised controlled trial. Thorax 60: 71317, 2005.
79. Hidi R, Timmermans S, Liu E, Schudt C, Dent G, Holgate ST et al. 101. Barr RG, Rowe BH, Camargo CA Jr. Methylxanthines for exacerba-
Phosphodiesterase and cyclic adenosine monophosphate-dependent tions of chronic obstructive pulmonary disease: Meta-analysis of ran-
inhibition of T-lymphocyte chemotaxis. Eur Respir J 15: 34249, 2000. domised trials. BMJ 327: 643, 2003.
80. Sanjar S, Aoki S, Kristersson A, Smith D, Morley J. Antigen challenge 102. Wilson AJ, Gibson PG, Coughlan J. Long acting beta-agonists versus
induces pulmonary eosinophil accumulation and airway hyperreac- theophylline for maintenance treatment of asthma. Cochrane Database
tivity in sensitized guinea pigs: The eect of anti-asthma drugs. Br J Syst Rev(2), 2000, CD001281.
Pharmacol 99: 67986, 1990. 103. Furukawa CT, Shapiro SG, Bierman CW. A double-blind study com-
81. Kosmas EN, Michaelides SA, Polychronaki A, Roussou T, Toukmatzi S, paring the eectiveness of cromolyn sodium and sustained release
Polychronopoulos V et al. Theophylline induces a reduction in circulat- theophylline in childhood asthma. Pediatrics 74: 43539, 1984.
ing interleukin-4 and interleukin-5 in atopic asthmatics. Eur Respir J 104. Nassif EG, Weinburger M, Thompson R, Huntley W. The value of
13: 5358, 1999. maintenance theophylline in steroid-dependent asthma. N Engl J Med
82. Kidney J, Dominguez M, Taylor PM, Rose M, Chung KF, Barnes PJ. 304: 7175, 1981.
Immunomodulation by theophylline in asthma: Demonstration by 105. Brenner MR, Berkowitz R, Marshall N, Strunk RC. Need for theophyl-
withdrawal of therapy. Am J Respir Crit Care Med 151: 190714, 1995. line in severe steroid-requiring asthmatics. Clin Allergy 18: 14350, 1988.
83. Jaar ZH, Sullivan P, Page C, Costello J. Low-dose theophylline mod- 106. Rivington RN, Boulet LP, Cote J, Kreisman H, Small DI, Alexander M
ulates T-lymphocyte activation in allergen-challenged asthmatics. Eur et al. Ecacy of slow-release theophylline, inhaled salbutamol and
Respir J 9: 45662, 1996. their combination in asthmatic patients on high-dose inhaled steroids.
84. Barnes PJ, Pauwels RA. Theophylline in asthma: Time for reappraisal?. Am J Respir Crit Care Med 151: 32532, 1995.
Eur Respir J 7: 57991, 1994. 107. Barnes PJ. The role of theophylline in severe asthma. Eur Respir Rev
85. Cushley MJ, Holgate ST. Bronchodilator actions of xanthine deriva- 6(rev 34): 154S59S, 1996.
tives administered by inhalation in asthma. Thorax 40: 17679, 1985. 108. Markham A, Faulds D. Theophylline. A review of its potential steroid
86. Aubier M, De Troyer A, Sampson M, Macklem PT, Roussos C. sparing eects in asthma. Drugs 56: 108191, 1998.
Aminophylline improves diaphragmatic contractility. N Engl J Med 109. Martin RJ, Pak J. Overnight theophylline concentrations and eects
305: 24952, 1981. on sleep and lung function in chronic obstructive pulmonary disease.
87. Moxham J. Aminophylline and the respiratory muscles: An alterna- Am Rev Respir Dis 145: 54044, 1992.
tive view. In: Respiratory Muscles: Function in health and disease. Clin 110. Zwilli CW, Neagey SR, Cicutto L, White DP, Martin RJ. Nocturnal
Chest Med 2: 32540, 1988. asthma therapy: Inhaled bitolterol versus sustained-release
88. Vozeh S, Kewitz G, Perruchoud A, Tschan M, Koppe C, Heitz M theophylline. Am Rev Respir Dis 139: 47074, 1989.
et al. Theophylline serum concentration and therapeutic eect in severe 111. Fairfax AJ, Clarke R, Chatterjee SS, Connolly CK, Higenbottam T,
acute bronchial obstruction: The optimal use of intravenously adminis- Holgate ST et al. Controlled release theophylline in the treatment of
tered aminophylline. Am Rev Respir Dis 125: 18184, 1982. nocturnal asthma. J Int Med Res 18: 27381, 1990.

637
Asthma and COPD: Basic Mechanisms and Clinical Management

112. Selby C, Engleman HM, Fitzpatrick MF, Sime PM, Mackay TW, 120. Barnes PJ. Theophylline in chronic obstructive pulmonary disease:
Douglas NJ. Inhaled salmeterol or oral theophylline in nocturnal New horizons. Proc Am Thorac Soc 2: 33439, 2005.
asthma? Am J Respir Crit Care Med 155: 1048, 1997. 121. Barnes PJ. Theophylline for COPD. Thorax 61: 74243, 2006.
113. Evans DJ, Taylor DA, Zetterstrom O, Chung KF, OConnor BJ, 122. Jenne JW. Theophylline as a bronchodilator in COPD and its combi-
Barnes PJ. A comparison of low-dose inhaled budesonide plus theo- nation with inhaled beta-adrenergic drugs. Chest 92: 7S14S, 1987.
phylline and high-dose inhaled budesonide for moderate asthma. 123. Kume H, Graziano MP, Kotliko MI. Stimulatory and inhibitory
N Engl J Med 337: 141218, 1997. regulation of calcium-activated potassium channels by guanine nucle-
114. Ukena D, Harnest U, Sakalauskas R, Magyar P, Vetter N, Steen H otide binding proteins. Proc Natl Acad Sci USA 89: 1105155, 1992.
et al. Comparison of addition of theophylline to inhaled steroid with 124. Giembycz MA. Phosphodiesterase 4 and tolerance to beta 2-adreno-
doubling of the dose of inhaled steroid in asthma. Eur Respir J 10: ceptor agonists in asthma. Trends Pharmacol Sci 17: 33136, 1996.
275460, 1997. 125. Cheung D, Wever AM, de GOEIJ JA, de GRAAFF CS, Steen H,
115. Lim S, Groneberg D, Fischer A, Oates T, Caramori G, Mattos W et al. Sterk PJ. Eects of theophylline on tolerance to the bronchoprotec-
Expression of heme oxygenase isoenzymes 1 and 2 in normal and tive actions of salmeterol in asthmatics in vivo. Am J Respir Crit Care
asthmatic airways: eect of inhaled corticosteroids. Am J Respir Crit Med 158: 79296, 1998.
Care Med 162: 191218, 2000. 126. Williamson BH, Milligan C, Griths K, Sparta S, Tribe AC,
116. Taylor DR, Buick B, Kinney C, Lowry RC, McDevitt DG. The e- Thompson PJ. An assessment of major and minor side eects of
cacy of orally administered theophylline, inhaled salbutamol, and theophylline. Aust NZ J Med 19: 539, 1988.
a combination of the two as chronic therapy in the management of 127. Rachelefsky WOJ, Adelson J, Mickey MR, Spector SL, Katz RM,
chronic bronchitis with reversible airow obstruction. Am Rev Respir Siegel SC et al. Behaviour abnormalities and poor school performance
Dis 131: 74751, 1985. due to oral theophylline use. Pediatrics 78: 111338, 1986.
117. Murciano D, Avclair M-H, Parievte R, Aubier M. A randomized con- 128. Nicholson CD, Challiss RAJ, Shahid M. Dierential modulation
trolled trial of theophylline in patients with severe chronic obstructive of tissue function and therapeutic potential of selective inhibitors of
pulmonary disease. N Engl J Med 320: 152125, 1989. cyclic nucleotide phosphodiesterase isoenzymes. Trends Pharmacol Sci
118. Chrystyn H, Mulley BA, Peake MD. Dose response relation to oral 12: 1927, 1991.
theophylline in severe chronic obstructive airway disease. BMJ 297: 129. Kelloway JS, Wyatt RA, Adlis SA. Comparison of patients compliance
150610, 1988. with prescribed oral and inhaled asthma medications. Arch Intern Med
119. Culpitt SV, de Matos C, Russell RE, Donnelly LE, Rogers DF, 154: 134952, 1994.
Barnes PJ. Eect of theophylline on induced sputum inammatory
indices and neutrophil chemotaxis in COPD. Am J Respir Crit Care
Med 165: 137176, 2002.

638
Corticosteroids
51CHAPTER

Corticosteroids (also known as glucocortico- target for inhaled corticosteroids, which are the
steroids, glucocorticoids, steroids) are by far the mainstay of modern asthma management. Inhaled
Peter J. Barnes
most eective controllers used in the treatment corticosteroids suppress many activated inam- National Heart and Lung Institute
of asthma and the only drugs that can eectively matory genes in airway epithelial cells (Fig. 51.2). (NHLI), Clinical Studies Unit,
suppress the characteristic inammation in asth- Epithelial integrity is restored by regular inhaled Imperial College, London, UK
matic airways. By contrast, they are ineective in corticosteroids. The suppression of mucosal
suppressing pulmonary inammation in COPD. inammation is relatively rapid with a signicant
After the discussion of the mechanism of action reduction in eosinophils detectable within 6 h and
and pharmacology of corticosteroids their cur- associated with reduced airway hyperresponsive-
rent use in the treatment of asthma and COPD ness [3, 4]. Reversal of airway hyperresponsiveness
will be described.

TABLE 51.1 Effect of corticosteroids on gene transcription.

Increased transcription
MECHANISMS OF ACTION Lipocortin-1
2-Adrenergic receptors
There have been major advances in understand- Secretory leukocyte inhibitory protein
ing the molecular mechanisms whereby corticos- IB- (inhibitor of NF-B)
teroids suppress inammation, based on recent Anti-inflammatory or inhibitory cytokines
developments in understanding the fundamen- IL-10, IL-12, IL-1 receptor antagonist
tal mechanisms of gene transcription [1, 2]. Mitogen-activated protein kinase phosphatase-1
Corticosteroids activate and suppress many genes (MKP-1, inhibits MAP kinase pathways)
relevant to understanding their action in asthma Decreased transcription
and in other allergic diseases (Table 51.1).
Inflammatory cytokines
IL-2, IL-3, IL-4, IL-5, IL-6, IL-11, IL-13, IL-15, TNF-, GM-
CSF, SCF
Cellular effects Chemokines
IL-8, RANTES, MIP-1, eotaxin
At a cellular level, corticosteroids reduce the num-
Inflammatory enzymes
bers of inammatory cells in the airways, includ-
Inducible nitric oxide synthase (iNOS), inducible
ing eosinophils, T-lymphocytes, mast cells, and
cyclo-oxygenase (COX-2) inducible phospholipase
dendritic cells (Fig. 51.1). These eects of corti-
A2 (cPLA2)
costeroids are produced through inhibiting the
Inflammatory peptides
recruitment of inammatory cells into the airway
Endothelin-1
by suppressing the production of chemotactic
Mediator receptors
mediators and adhesion molecules and by inhib-
Neurokinin (NK1)-, bradykinin (B2)-receptors
iting the survival in the airways of inammatory
Adhesion molecules
cells, such as eosinophils, T-lymphocytes, and mast
ICAM-1,VCAM-1
cells. Epithelial cells may be the major cellular

639
Asthma and COPD: Basic Mechanisms and Clinical Management

Inflammatory cells
Structural cells
Eosinophil
Numbers Epithelial cell

(apoptosis)
Cytokines

Mediators
T-lymphocyte

Cytokines Endothelial cell


Leak
Mast cell Corticosteroids

Numbers Airway smooth muscle


2-Receptors
Macrophage Cytokines
Cytokines Mucus gland

Dendritic cell
Mucus

Numbers secretion
FIG. 51.1 Cellular effects of corticosteroids.

Inhaled corticosteroids GRs rapidly translocate to the nucleus where they produce
their molecular eects. A pair of GRs (GR homodimer) bind
to glucocorticoid response elements in the promoter region
Epithelial cells
of steroid-responsive genes and this interaction switches
on (and sometimes switches o ) gene transcription (Fig.
51.3). Examples of genes that are activated by corticosteroids
include genes encoding 2-adrenergic receptors and the anti-
Cytokines Enzymes Peptides Adhesion inammatory proteins secretory leukoprotease inhibitor and
IL-1 iNOS ET-1 molecules
ICAM-1
mitogen-activated protein kinase phosphatase-1 (MKP-1),
IL-6 COX-2
cPLA2 which inhibits MAP kinase pathways. These eects may con-
GM-CSF
RANTES tribute to the anti-inammatory actions of corticosteroids [8,
Eotaxin 9]. GR interaction with negative GREs may suppress gene
MIP-1 transcription and it is thought that this may be important
Inflammation in mediating many of the side eects of corticosteroids. For
example, corticosteroids inhibit the expression of osteocalcin
FIG. 51.2 Inhaled corticosteroids may inhibit the transcription of that is involved in bone synthesis [10].
several inflammatory genes in airway epithelial cells and thus reduce
inflammation in the airway wall. NF-B: nuclear factor B; AP-1: activator
protein-1; GM-CSF: granulocyte-macrophage colony stimulating factor; Switching off inflammation
IL-1: interleukin-1; iNOS: inducible nitric oxide synthase; NO: nitric oxide;
COX-2: inducible cyclooxygenase; cPLA2: cytoplasmic phospholipase A2; The major action of corticosteroids is to switch o multi-
PG: prostaglandin; ET: endothelin; ICAM: intercellular adhesion molecule. ple activated inammatory genes that encode for cytokines,
chemokines, adhesion molecules, inammatory enzymes,
and receptors [11]. These genes are switched on in the air-
may take several months to reach a plateau, probably reecting ways by proinammatory transcription factors, such as
recovery of structural changes in the airway [5]. nuclear factor-B (NF-B) and activator protein-1, both
of which are activated in asthmatic airways and switch on
inammatory genes by interacting with coactivator mol-
ecules, such as CREB-binding protein, that have intrin-
Glucocorticoid receptors sic histone acetyltransferase (HAT) activity, resulting in
acetylation of core histones, which opens up the chroma-
Corticosteroids diuse across the cell membrane and bind to tin structure so that gene transcription is facilitated [12]. In
glucocorticoid receptors (GR) in the cytoplasm [2]. There is articial overexpression systems, activated GR may directly
only one form of GR that binds corticosteroids termed GR-. interact with NF-B and AP-1 to inhibit their activity, but
GR- is an alternatively spliced form of GR that interacts with this does not appear to occur in asthmatic patients treated
DNA but not with corticosteroids, so may act as a dominant with inhaled corticosteroids [13]. Glucocorticoid-acti-
negative inhibitor of glucocorticoid action by interfering with vated GR also interact with coactivator molecules and this
the binding of GR to DNA [6]. Whether GR- is involved inhibits the interaction of NF-B with coactivators, thus
in steroid resistance in asthma is controversial [7]. Activated reducing histone acetylation [1, 14]. Reduction of histone

640
Corticosteroids 51
Glucocorticoid

FIG. 51.3 Corticosteroids may regulate gene expression


in several ways. Glucocorticoids enter the cell to bind
Glucocorticoid receptor to glucocorticoid receptors in the cytoplasm that
translocate to the nucleus. GR homodimers bind to
glucocorticoid-response elements (GRE) in the promoter
Trans-activation Cis-repression Trans-repression region of steroid-sensitive genes, which may encode anti-
inflammatory proteins. Less commonly, GR homodimers
interact with negative GREs to suppress genes, particularly
NF-B
those linked to side effects of corticosteroids. Nuclear
CBP GR also interact with coactivator molecules, such as
GRE Negative GRE CREB-binding protein (CBP), which is activated by
Anti-inflammatory Side effects proinflammatory transcription factors, such as nuclear
Inflammatory
factor-B (NF-B), thus switching off the inflammatory
Annexin-1 POMC Cytokines genes that are activated by these transcription factors.
SLPI CRF-1 Chemokines SLPI: secretory leukoprotease inhibitor; MKP-1: mitogen-
MKP-1 Osteocalcin Adhesion molecules activated kinase phosphatase-1; IB-: inhibitor of NF-B;
IB- Keratin Inflammatory enzymes GILZ: glucocorticoid-induced leucine zipper protein;
Inflammatory receptors POMC: proopiomelanocortin; CRH: corticotrophin
Inflammatory proteins releasing factor.

Inflammatory stimuli
e.g. IL-1, TNF- Glucocorticoid
(low dose) FIG. 51.4 Corticosteroid suppression of activated
inflammatory genes. Inflammatory genes are
activated by inflammatory stimuli, such as
interleukin-1 (IL-1) or tumor necrosis factor-
IKK-2 (TNF-), resulting in activation of IKK2 (inhibitor
of I-B kinase-2), which activates the transcription
factor, nuclear factor B (NF-B). A dimer of p50
p65 and p65 NF-B proteins translocates to the nucleus
NF-B GR
p50 and binds to specific B recognition sites and also
to coactivators, such as CREB-binding protein (CBP)
AF or p300/CBP-activating factor (pCAF), which have
pC
intrinsic histone acetyltransferase (HAT) activity. This
p65 results in acetylation of core histone H4, resulting in
CBP
B GR HDAC2 increased expression of genes encoding multiple
p50
HAT inflammatory proteins. Glucocorticoid receptors (GR)
Inflammatory genes Acetylation  Deacetylation after activation by glucocorticoids translocate to
cytokines, chemokines the nucleus and bind to coactivators to inhibit HAT
adhesion molecules activity directly and recruiting histone deacetylase-2
inflammatory receptors, (HDAC2), which reverses histone acetylation leading
enzymes, proteins
Gene Gene in suppression of these activated inflammatory
transcription repression genes.

acetylation also occurs through the recruitment of histone have an unstable mRNA that is rapidly degraded by cer-
deacetylase-2 (HDAC2) to the activated inammatory tain RNAses but stabilized when cells are stimulated by
gene complex by activated GR, thereby resulting in eec- inammatory mediators. Corticosteroids reverse this eect,
tive suppression of all activated inammatory genes within resulting in rapid degradation of mRNA and reduced
the nucleus (Fig. 51.4). This accounts for why corticoster- inammatory protein secretion [15]. This may be through
oids are so eective in the control of asthmatic inamma- the inhibition of proteins that stabilize mRNAs of inam-
tion, but also why they are safe, since other activated genes matory proteins, such as tristetraprolin [16].
are not aected.
There may be additional mechanisms that are also
important in the anti-inammatory actions of corticosteroids. Corticosteroid resistance
Corticosteroids have potent inhibitory eects on mitogen-
activated kinase signaling pathways through the induction Patients with severe asthma have a poor response to corticos-
of MKP-1 and this may inhibit the expression of multiple teroids, which necessitates the need for high doses and a few
inammatory genes [8, 9]. Some inammatory genes, for patients are completely resistant. All patients with COPD
example granulocyte-macrophage colony stimulating factor, show corticosteroid resistance. Asthmatics who smoke are also

641
Asthma and COPD: Basic Mechanisms and Clinical Management

COPD
Severe asthma FIG. 51.5 Mechanism of corticosteroid
Mild asthma Smoking asthma resistance in COPD, smoking asthma, and severe
asthma. Stimulation of mild asthmatic alveolar
Cigarette smoke macrophages activates nuclear factor-B (NF-B)
Stimuli Corticosteroids and other transcription factors to switch on histone
Oxidative stress acetyltransferase leading to histone acetylation
Alveolar and subsequently to transcription of genes
macrophage encoding inflammatory proteins, such as tumor
Peroxynitrite
necrosis factor- (TNF-), interleukin-8 (IL-8), and
GR granulocyte-macrophage colony stimulating factor
(GM-CSF). Corticosteroids reverse this by binding to
NF-B NF-B
GR and recruiting HDAC2. This reverses the histone
acetylation induced by NF-B and switches off the
HDAC2 HDAC2 activated inflammatory genes. In COPD patients
Histone Histone and smoking asthmatics cigarette smoke generates
acetylation acetylation
oxidative stress (acting through the formation of
peroxynitrite) and in severe asthma and COPD
TNF- Histone acetylation intense inflammation generates oxidative stress
IL-8 TNF- to impair the activity of HDAC2. This amplifies
GM-CSF Corticosteroid IL-8
resistance the inflammatory response to NF-B activation,
GM-CSF
but also reduces the anti-inflammatory effect of
corticosteroids, as HDAC2 is now unable to reverse
histone acetylation.

relatively corticosteroid-resistant and require increased doses mediators, such as IL-1 through a stimulatory eect on a
of corticosteroids for asthma control [17]. Several molecular G-protein-coupled receptor kinase [28].
mechanisms have now been identied to account for corticos- There is also evidence that 2-agonists may aect GR
teroid resistance in severe asthma and COPD [18]. In patients and thus enhance the anti-inammatory eects of corticos-
with COPD, smoking asthmatics, and severe asthma there is a teroids. 2-Agonists increase the translocation of GR from
reduction in HDAC2 activity and expression, which prevents cytoplasm to the nucleus after activation by corticosteroids
corticosteroids switching o activated inammatory genes (Fig. [29]. This eect has now been demonstrated in sputum
51.5) [1921]. In steroid-resistant asthma other mechanisms macrophages of asthmatic patients after an inhaled gluco-
may also contribute to corticosteroid insensitivity, including corticoid and inhaled long-acting 2-agonist (LABA) [30].
reduced translocation of GR as a result of phosphorylation by This suggests that 2-agonists and glucocorticoid enhance
p38 MAP kinase [22] and abnormal histone acetylation pat- each others benecial eects in asthma therapy.
terns [23]. A proposed mechanism is an increase in GR-,
which prevents GR binding to DNA, but there is little evi-
dence that this would be sucient to account for corticosteroid
insensitivity as the amounts of GR- are too low [7].
PHARMACOKINETICS
Prednisolone is readily and consistently absorbed after oral
INTERACTION WITH 2-ADRENERGIC administration with little interindividual variation. Prednisone
RECEPTORS is converted in the liver to the active prednisolosne. Enteric
coatings to reduce the incidence of dyspepsia delay absorp-
tion but not the total amount of drug absorbed. Prednisolone
Inhaled 2-agonists and corticosteroids are frequently used is metabolized in the liver and drugs such as rifampicin, phe-
together in the control of asthma and it is now recognized nobarbitone, or phenytoin, which induce CYP450 enzymes,
that there are important molecular interactions between these lower the plasma half-life of prednisolone [31]. The plasma
two classes of drugs (Fig. 51.6) [24]. As discussed above, cor- half-life is 23 h, although its biological half-life is approxi-
ticosteroids increase the gene transcription of 2-receptors, mately 24 h, so that it is suitable for daily dosing. There is no
resulting in increased expression of cell surface receptors. This evidence that previous exposure to steroids changes their sub-
has been demonstrated in human lung in vitro [25] and nasal sequent metabolism. Prednisolone is approximately 92% pro-
mucosa in vivo after topical application of a glucocorticoid tein bound, the majority to a specic protein transcortin and
[26]. In this way corticosteroids protect against the down- the remainder to albumin; it is the unbound fraction which
regulation of 2-receptors after long-term administration [27]. is biologically active. Corticosteroid-resistant asthma is not
This may be important for the non-bronchodilator eects of explained by impaired absorption or metabolism of steroids,
2-agonists, such as mast cell stabilization. Corticosteroids may but is due to reduced anti-inammatory actions of corticos-
also enhance the coupling of 2-receptors to G-proteins, thus teroids, as discussed above. Measurement of plasma concen-
enhancing 2-agonist eects and reversing the uncoupling trations of prednisolone are useful in monitoring compliance
of 2-receptors that may occur in response to inammatory with inhaled corticosteroids and in assessing whether a poor

642
Corticosteroids 51
Corticosteroid
LABA
2-adrenoceptor

Glucocorticoid 
receptor


Anti-inflammatory effect Bronchodilation
FIG. 51.6 Interaction between corticosteroids and LABA.
GR translocation 2-receptor expression Corticosteroids have anti-inflammatory effects but also increase
GRE binding 2-receptor coupling
the numbers of 2-receptors, whereas 2-agonists, as well as
Anti-inflammatory effect Downregulation of 2-receptors
inducing direct bronchodilation, act on GR to increase the anti-
Prevention of -agonist tolerance
inflammatory effects of corticosteroids.

therapeutic response to corticosteroids is due to poor absorp-


tion or increased metabolism. MDI
~1020% inhaled

Mouth and
Inhaled delivery pharynx Lungs Systemic
~8090% swallowed Absorption circulation
The pharmacokinetics of inhaled corticosteroids is important ( spacer/mouthwash) from GI tract
in relation to systemic eects [32, 34]. The fraction of ster-
oid which is inhaled into the lungs acts locally on the airway GI tract Liver
mucosa, but may be absorbed from the airway and alveolar
surface. This fraction therefore reaches the systemic circulation
Systemic
(Fig 51.7). The fraction of inhaled steroid which is deposited Inactivation
side effects
in the oropharynx is swallowed and absorbed from the gut. in liver
The absorbed fraction may be metabolized in the liver before first pass
reaching the systemic circulation (rst-pass metabolism). FIG. 51.7 Pharmacokinetics of inhaled glucocorticoids.GI: gastrointestinal.
Budesonide and uticasone propionate (FP) have a greater
rst-pass metabolism than beclomethasone dipropionate
(BDP) and are therefore less likely to produce systemic eects
at high inhaled doses. The use of a large volume spacer cham- has been questioned, others have found that they speed the
ber reduces oropharyngeal deposition and therefore reduces resolution of attacks. There is no apparent advantage in giv-
systemic absorption of corticosteroids, although this eect is ing very high doses of intravenous steroids (such as methyl-
minimal in corticosteroids with a high rst-pass metabolism prednisolone 1 g) as this only increases the risk of side eects,
[35]. Mouth rinsing and discarding the rinse has a similar such as hyperglycemia and increased susceptibility to infec-
eect and this procedure should be used with high dose dry tions. Intravenous steroids are indicated in acute asthma if lung
powder steroid inhalers, since spacer chambers cannot be used function is  30% predicted and in whom there is no signi-
with these devices. The ideal inhaled corticosteroid with opti- cant improvement with a nebulized 2-agonist. Intravenous
mal therapeutic index should have high lung bioavailability, therapy is usually given until a satisfactory response is obtained
negligible oral bioavailability, low systemic absorption, high and then oral prednisolone may be substituted. Oral pred-
systemic clearance, and high protein binding [36]. nisolone (4060 mg) has a similar eect to intravenous hydro-
A recently introduced corticosteroid ciclesonide is an cortisone and is easier to administer [38, 39]. High doses of
inactive prodrug that is activated by esterases in the lung to inhaled corticosteroids may also substitute for a course of oral
the active metabolite des-ciclesonide [37]. This may reduce steroids in controlling acute exacerbations of asthma. High
oropharyngeal side eects as esterases appear to be less dose FP (2000 g daily) was as eective as a course of oral
active in this site than in the lower airways. Ciclesonide is prednisolone in controlling acute exacerbations of asthma in a
also claimed to be eective as a once daily therapy. family practice setting and in children in an emergency room
setting, although this route of delivery is more expensive [40,
41]. Although doubling the dose of inhaled corticosteroids
was recommended for mild exacerbations of asthma, this does
SYSTEMIC STEROIDS not appear to be useful [42, 43], but a fourfold increase in dose
appears to be eective [44]. Inhaled steroids have no proven
eect in the management of severe acute asthma in a hospi-
Hydrocortisone is given intravenously in acute severe asthma. tal setting [45], but trials with nebulized steroids, which can
While the value of corticosteroids in acute severe asthma deliver large doses, are underway.

643
Asthma and COPD: Basic Mechanisms and Clinical Management

CH2 OH CH2OCOC2H5 CH2 OH


C O C O C O
CH3 OH OCOC2H5 O H
HO HO HO C
17 CH3 O CH2CH2C
CH3

Cl
O O O
Hydrocortisone Beclomethasone dipropionate Budesonide

SCH2F CH2 OH CH2 OH


C O C O C O
O CH3 O CH3
HO OCOC2H5 HO C HO C
O CH3 O CH3
CH3

F F
O O O

F F
Fluticasone propionate Flunisolide Triamcinolone acetonide

FIG. 51.8 Chemical structures of inhaled glucocorticoids.

Maintenance treatment with oral steroids are reserved USE IN ASTHMA


for patients who cannot be controlled on maximum doses
of other therapy, the dose being titrated to the lowest
which provides acceptable control of symptoms. For any Studies in adults
patient taking regular oral steroids objective evidence of
steroid responsiveness should be obtained before mainte- As experience has been gained with inhaled corticosteroids
nance therapy is instituted. Short courses of oral steroids they have been introduced in patients with milder asthma,
(3040 mg prednisolone daily for 12 weeks) are indicated with the recognition that inammation is present even in
for exacerbations of asthma, and the dose may be tailed o patients with mild asthma. Inhaled anti-inammatory drugs
over 1 week once the exacerbation is resolved (although the have now become rst-line therapy in any patient who
tail-o period is not strictly necessary, patients often nd it needs to use a 2-agonist inhaler more than two to three
reassuring). times a week and this is reected in national and interna-
tional guidelines for the management of chronic asthma. In
patients with newly diagnosed asthma an inhaled corticos-
teroid (budesonide 600 g twice daily) reduced symptoms
INHALED CORTICOSTEROIDS and 2-agonist inhaler usage and improved lung func-
tion. These eects persisted over the 2 years of the study,
whereas in a parallel group treated with inhaled 2-agonists
There is no doubt that the early use of inhaled corticoster- alone there was no signicant change in symptoms or lung
oids has revolutionized the management of asthma, with function [47]. In another study, patients with mild asthma
marked reductions in asthma morbidity and improvement treated with a low dose of inhaled corticosteroid (budeso-
in health status. Inhaled steroids are now recommended nide 400 g daily) showed less symptoms and a progres-
as rst-line therapy for all patients with persistent asthma sive improvement in lung function over several months and
[46]. Several topically acting corticosteroids are now avail- many patients became completely asymptomatic [48]. There
able for inhalation (Fig. 51.8). Inhaled corticosteroids are was also a signicant reduction in the number of exacer-
very eective in controlling asthma symptoms in asth- bations; in patients with mild asthma a low dose of corti-
matic patients of all ages and severity. Inhaled corticos- costeroids (budesonide 400 g daily) signicantly reduces
teroids improve the quality of life of patients with asthma exacerbation by around 40% over a 3-year period [49].
and allow many patients to lead normal lives, improve Although the eects of inhaled corticosteroids on AHR
lung function, reduce the frequency of exacerbations, and may take several months to reach a plateau, the reduction in
may prevent irreversible airway changes. They were rst asthma symptoms occurs much more rapidly and reduced
introduced to reduce the requirement for oral corticoster- inammation is seen within hours [3, 4].
oids in patients with severe asthma and many studies have High dose inhaled corticosteroids may be used for
conrmed that the majority of patients can be weaned o the control of more severe asthma. This markedly reduces the
oral corticosteroids [32]. need for maintenance oral corticosteroids [50]. With the use

644
Corticosteroids 51
of add-on therapies, particularly LABA, most patients can eects of inhaled corticosteroids. This is obviously important
now be controlled on much lower doses of inhaled corticos- for the interpretation of clinical comparisons between dier-
teroids so that high doses are needed in only a few patients ent inhaled corticosteroids or inhalers. It is also important
with severe disease. Inhaled corticosteroids are the treatment to consider the type of patient included in clinical studies.
of choice in nocturnal asthma, which is a manifestation of Patients with relatively mild asthma may have relatively little
inamed airways, reducing nocturnal awakening and reducing room for improvement with inhaled corticosteroids, so that
the diurnal variation in airway function. maximal improvement is obtained with relatively low doses.
Inhaled corticosteroids eectively control asthmatic Patients with more severe asthma or with unstable asthma
inammation but must be taken regularly. When inhaled cor- may have more room for improvement and may therefore
ticosteroids are discontinued there is usually a gradual increase show a greater response to increasing doses, but it is often
in symptoms and airway responsiveness back to pretreatment dicult to include such patients in controlled clinical trials.
values [51]. Reduction in the dose of inhaled corticosteroids is More studies are needed to assess whether other out-
associated with an increase in symptoms and this is preceded come measures such as AHR or more direct measurements
by an increase in exhaled NO and sputum eosinophils [52, 53]. of inammation, such as sputum eosinophils or exhaled NO,
may be more sensitive than traditional outcome measures
such as symptoms or lung function tests [6063]. Higher
Studies in children doses of inhaled corticosteroids are needed to control AHR
than to improve symptoms and lung function, and this may
Inhaled corticosteroids are equally eective in children. In an have a better long-term outcome in terms of reduction in
extensive study of children aged 717 years there was a sig- structural changes of the airways [64]. Measurement of
nicant improvement in symptoms, peak ow variability, and sputum eosinophils to adjust the dose of inhaled corticos-
lung function compared to a regular inhaled 2-agonist which teroids may reduce the overall dose requirement for inhaled
was maintained over the 22 months of the study [54], but corticosteroids and exacerbations [65, 66]. Monitoring of
asthma deteriorated when the inhaled corticosteroids were exhaled nitric oxide also reduces the requirement for corti-
withdrawn [55]. There was a high proportion of dropouts costeroids but is not yet practical in clinical practice [67].
(45%) in the group treated with inhaled 2-agonist alone.
Inhaled corticosteroids are also eective in younger children.
Nebulized budesonide reduces the need for oral corticoster- Prevention of irreversible airway changes
oids and also improved lung function in children under the
age of three [56]. Inhaled corticosteroids given via a large Some patients with asthma develop an element of irrevers-
volume spacer improve asthma symptoms and reduce the ible airow obstruction, but the pathophysiological basis of
number of exacerbations in preschool children and in infants. these changes is not yet understood. It is likely that they are
the result of chronic airway inammation and that they may
be prevented by treatment with inhaled corticosteroids. There
Dose-response studies is some evidence that the annual decline in lung function may
be slowed by the introduction of inhaled corticosteroids [68]
Surprisingly, the dose-response curve for the clinical e- and this is supported by a 5-year study of low dose budeso-
cacy of inhaled corticosteroids is relatively at and, while all nide in patients with mild asthma [69, 70]. Increasing evidence
studies have demonstrated a clinical benet of inhaled cor- also suggests that delay in starting inhaled corticosteroids may
ticosteroids, it has been dicult to demonstrate dierences result in less overall improvement in lung function in both
between doses, with most benet obtained at the lowest doses adults and children [71, 73]. These studies suggest that intro-
used [57, 58]. This is in contrast to the steeper dose response duction of inhaled corticosteroids at the time of diagnosis is
for systemic eects, implying that while there is little clini- likely to have the greatest impact [72, 73]. So far there is no
cal benet from increasing doses of inhaled corticosteroids evidence that early use of inhaled corticosteroids is curative and
the risk of adverse eects is increased. However, the dose- even when inhaled corticosteroids are introduced at the time of
response eect of inhaled corticosteroids may depend on the diagnosis, symptoms and lung function reverts to pretreatment
parameters measured and, while it is dicult to discern a levels when corticosteroids are withdrawn [71].
dose response when traditional lung function parameters are
measured, there may be a dose-response eect in prevention
of asthma exacerbations. Thus, there is a signicantly greater Reduction in mortality
eect of budesonide 800 g daily compared to 200 g daily
in preventing severe and mild asthma exacerbations [59]. In a retrospective review of the risk of mortality and pre-
Normally, a fourfold or greater dierence in dose has been scribed antiasthma medication, there was a signicant pro-
required to detect a statistically signicant (but often small) tection provided by regular inhaled corticosteroid therapy
dierence in eect on commonly measured outcomes such [74]. By contrast, asthma mortality appears to increase with
as symptoms, PEF, use of rescue 2-agonist and lung func- increasing usage of short-acting 2-agonists, reecting the
tion and even such large dierences in dose are not always fact that increased rescue therapy is a marker of poor asthma
associated with signicant dierences in response. These control [75]. The increase in use of rescue therapy should
ndings suggest that pulmonary function tests or symptoms result in an increase in the maintenance dose of inhaled cor-
may have a rather low sensitivity in the assessment of the ticosteroids. The long-acting inhaled 2-agonist salmeterol is

645
Asthma and COPD: Basic Mechanisms and Clinical Management

associated with a small increase in asthma mortality, but the eects. Nebulized budesonide has been advocated in order
excess deaths appear to be related to underuse of inhaled cor- to give an increased dose of inhaled corticosteroid and to
ticosteroids [76]. reduce the requirement for oral corticosteroids [80], but this
treatment is expensive and may achieve its eects largely
via systemic absorption. The dose of inhaled corticosteroid
Comparison between inhaled should be the minimal dose that controls asthma and once
corticosteroids control is achieved the dose should be slowly reduced [81].

Several inhaled corticosteroids are currently prescribable in


asthma, although their availability varies between countries.
There have been relatively few studies comparing ecacy USE IN COPD
of the dierent inhaled corticosteroids, and it is important
to take into account the delivery system and the type of
patient under investigation when such comparisons are made. Patients with COPD have a poor response to corticosteroids
Because of the relatively at dose-response curve for the clin- in comparison to asthma with little improvement in lung func-
ical parameters normally used in comparing doses of inhaled tion. High doses of inhaled corticosteroids have consistently
corticosteroids, it may be dicult to see dierences in ecacy been shown a reduction (2025%) in exacerbations in patients
of inhaled corticosteroids. Most comparisons have concen- with severe disease and this is the main clinical indication for
trated on dierences in systemic eects at equally ecacious their use [82]. However, several large studies have shown that
doses, although it has often proved dicult to establish dose corticosteroids fail to reduce the progression in COPD (meas-
equivalence. There are few studies comparing dierent doses ured by annual fall in FEV1) [83] and they have not been
of inhaled corticosteroids in asthmatic patients. Budesonide found to reduce mortality in a large study [84]. These results
has been compared with BDP and in adults and children it are likely to reect the resistance of pulmonary inammation
appears to have comparable antiasthma eects at equal doses, to corticosteroids in COPD patients as a result of the reduc-
whereas FP appears to be approximately twice as potent as tion in HDAC2 [21].
BDP and budesonide [58]. Studies have consistently shown
that FP and budesonide have less systemic eects than BDP,
triamcinolone, and unisolide [33]. The new inhaled corti-
costeroids mometasone and ciclesonide are claimed to have
less systemic eects [77, 78].
ADD-ON THERAPY

Previously it was recommended to increase the dose of


Clinical application inhaled corticosteroids if asthma was not controlled, on the
assumption that there was residual inammation of the air-
Inhaled corticosteroids are now recommended as rst-line ways. However the dose-response eect of inhaled corticos-
therapy for all patients with persistent symptoms. Inhaled teroids is relatively at, so that there is little improvement
corticosteroids should be started in any patient who needs to in lung function after increasing the dose of inhaled corti-
use a 2-agonist inhaler for symptom control more than three costeroids. An alternative strategy is to add some other class
times weekly. It is conventional to start with a low dose of of controller drug and this is more eective than increasing
inhaled corticosteroid and to increase the dose until asthma the dose of inhaled corticosteroids for most patients [85].
control is achieved. However, this may take time and a pref-
erable approach is to start with a dose of corticosteroids in
the middle of the dose range (400 g twice daily) to establish LABA
asthma control. Once control is achieved (dened as normal
or best possible lung function and infrequent need to use an In patients who are not controlled on BDP 200 g twice
inhaled 2-agonist) the dose of inhaled corticosteroid should daily, addition of salmeterol 50 g twice daily was more
be reduced in a stepwise manner to the lowest dose needed eective than increasing the dose of inhaled corticosteroid
for optimal control. It may take as long as 3 months to reach a to 500 g twice daily, in terms of lung function improve-
plateau in response and any changes in dose should be made at ment, use of rescue 2-agonist and symptom control [86].
intervals of 3 months or more. When daily doses of 800 g This has been conrmed in several other studies [87].
daily are needed, a large volume spacer device should be used Similar results have been found with another long-acting
with a metered dose inhaler (MDI) and mouth washing with inhaled 2-agonist formoterol, which in addition reduced
a dry powder inhaler in order to reduce local and systemic side the frequency of mild and severe asthma exacerbations in
eects. Inhaled corticosteroids are usually given as a twice daily patients with mild, moderate, and severe persistent asthma
dose in order to increase compliance. When asthma is unstable [59, 88]. Analysis of several studies clearly show that add-
four times daily dosage is preferable [79]. ing a LABA is more eective than increasing the dose of
The dose of inhaled corticosteroid should be increased inhaled corticosteroids in terms of improving asthma con-
to 2000 g daily if necessary, but higher doses may result in trol and reducing exacerbations [89]. These studies showing
systemic eects. It may be preferable to add a low dose of the great ecacy of combined corticosteroids and LABA
oral corticosteroid, since higher doses of inhaled corticos- compared to increased doses of LABA have led to the
teroids are expensive and have a high incidence of local side development of xed combinations of corticosteroids and

646
Corticosteroids 51
LABA, such as FP/salmeterol and budesonide/formoterol, addition of LABA [106108]. There is no role of antileuko-
which may be more convenient for patients. These xed trienes in COPD patients.
combination inhalers also ensure that patients do not dis-
continue their inhaled corticosteroids when a long-acting
bronchodilator is used. For patients with mild persistent
asthma, combination inhalers are no more eective than the
inhaled corticosteroids alone in controlled trials [90], but SIDE EFFECTS
may have an advantage in the real world where adherence
to regular inhaled corticosteroids is very low. The ecacy of inhaled corticosteroids is now established
Recently, studies have demonstrated that when for- in short- and long-term studies in adults and in children,
moterol combined with budesonide is used as a reliever but there are still concerns about side eects, particularly in
therapy this gives better control of asthma compared to the children and when high inhaled doses are used. Several side
normally used short-acting 2-agonist as a rescue therapy eects have been recognized (Table 51.2).
with either the same dose of combination inhaler or a high
dose of inhaled corticosteroids as maintenance treatment [91,
92]. This advantage is particularly striking in terms of reduc- Local side effects
ing the number of severe exacerbations. When formoterol
was used as the reliever therapy this reduce exacerbations to Side eects due to the local deposition of the inhaled corticos-
a greater extent than the short-acting 2-agonist terbutaline teroid in the oropharynx may occur with inhaled corticoster-
but the combination was even more eective [93]. This sug- oids, but the frequency of complaints depends on the dose and
gests that the as required use of inhaled corticosteroids con- frequency of administration and on the delivery system used.
tributes to the marked reduction in acute exacerbations. The The commonest complaint is of hoarseness of the voice
mechanisms by which corticosteroids as required improve (dysphonia) and may occur in over 50% of patients using
asthma control and reduce exacerbations are not completely MDI. Dysphonia is not appreciably reduced by using spacers,
understood, but exacerbations of asthma evolve over several but may be less with dry powder devices. Dysphonia may be
days when patients take increasing amounts of rescue medi- due to myopathy of laryngeal muscles and is reversible when
cation [94]. During this time there is increasing inamma- treatment is withdrawn [109]. For most patients it is not
tion of the airways, as may be measured by exhaled nitric troublesome but may be disabling in singers and lecturers.
oxide and sputum eosinophils [52]. Taking the inhaled corti- Oropharyngeal candidiasis (thrush) may be a problem
costeroid at the same time as the formoterol to relieve symp- in some patients, particularly in the elderly, with concomi-
toms may suppress this evolving inammation, particularly tant oral corticosteroids and more than twice daily admin-
since corticosteroids appear to have a relatively rapid onset of istration [110]. Large volume spacer devices protect against
eect in suppressing airway inammation [95]. this local side eect by reducing the dose of inhaled corti-
LABA/corticosteroid inhalers are also more eective costeroid that deposits in the oropharynx.
in COPD patients than either treatment alone and reduce
exacerbations and improve symptoms [96, 97]. They are more
eective than LABA alone in reducing exacerbations [98]. Infections
This may be explained by molecular interactions between
LABA and corticosteroids as discussed above. However, there There is no evidence that inhaled corticosteroids, even in
is a reduction in COPD mortality although this fails to reach high doses, increase the frequency of infections, includ-
signicance in one large study (TORCH) [84], although this ing tuberculosis, in the lower respiratory tract in asthmatic
has been seen in another smaller study (INSPIRE) [99].
TABLE 51.2 Side effects of inhaled corticosteroids.
Theophylline Local side effects
Dysphonia
Addition of low doses of theophylline (giving plasma con- Oropharyngeal candidiasis
centrations of 10 mg/l) are more eective than doubling Cough
the dose of inhaled budesonide, either in mild or in severe
Pneumonia (COPD patients)
asthma [100102]. However, this is less eective than using
a long-acting inhaled 2-agonist as an add-on therapy Systemic side effects
[103]. Theophylline has not been examined as an add-on Adrenal suppression
therapy in COPD patients but there are theoretical reasons to Growth suppression
believe that low dose theophylline may reverse corticoster- Bruising
oid resistance in COPD (see Chapter 52).
Osteoporosis
Cataracts
Antileukotrienes Glaucoma
Metabolic abnormalities (glucose, insulin, triglycerides)
Antileukotrienes have also been used as an add-on therapy Psychiatric disturbances
in asthma [104, 105], although this is less eective than

647
Asthma and COPD: Basic Mechanisms and Clinical Management

patients. Recently several large controlled studies have Adrenal suppression


shown that high dose ICS increase physician-diagnosed Corticosteroids may cause hypothalamicpituitaryadrenal
pneumonias, either used alone or in combination with (HPA) axis suppression by reducing corticotrophin (ACTH)
a LABA [83, 98] and this has been conrmed in an epi- production, which reduces cortisol secretion by the adrenal
demiological study of hospital admissions for pneumonia gland. The degree of HPA suppression is dependent on dose,
amongst COPD patients [111]. duration, frequency, and timing of corticosteroid administra-
tion. Measurement of HPA axis function provides evidence
for systemic eects of an inhaled corticosteroid. Basal adrenal
Systemic side effects cortisol secretion may be measured by a morning plasma cor-
tisol, 24 h urinary cortisol, or by plasma cortisol prole over
The ecacy of inhaled corticosteroids in the control of 24 h. Other tests measure the HPA response following stimu-
asthma is undisputed, but there are concerns about sys- lation with tetracosactrin (which measures adrenal reserve) or
temic eects of inhaled corticosteroids, particularly as they stimulation with metyrapone and insulin (which measure the
are likely to be used over long periods and in children of response to stress). There are many studies of HPA axis func-
all ages [33, 112]. The safety of inhaled corticosteroids tion in asthmatic patients with inhaled corticosteroids, but
has been extensively investigated since their introduction the results are inconsistent as they have often been uncon-
30 years ago [32]. One of the major problems is to decide trolled and patients have also been taking courses of oral
whether a measurable systemic eect has any signicant corticosteroids (which may aect the HPA axis for weeks)
clinical consequence and this necessitates careful long-term [32]. BDP, budesonide, and FP at high doses by conventional
follow-up studies. As biochemical markers of systemic cor- MDI (1600 g daily) give a dose-related decrease in morn-
ticosteroid eects become more sensitive, then systemic ing serum cortisol levels and 24 h urinary cortisol, although
eects may be seen more often, but this does not mean that values still lie well within the normal range. However, when
these eects are clinically relevant. There are several case a large volume spacer is used doses of 2000 g daily of BDP
reports of adverse systemic eects of inhaled corticosteroids, or budesonide have little eect on 24 h urinary cortisol excre-
and these may be idiosyncratic reactions, which may be due tion. Stimulation tests of HPA axis function similarly show
to abnormal pharmacokinetic handing of the inhaled cor- no consistent eects of doses of 1500 g or less of inhaled
ticosteroid. The systemic eect of an inhaled corticosteroid corticosteroid. At high doses (1500 g daily) budesonide,
will depend on several factors, including the dose delivered and FP have less eect than BDP on HPA axis function. In
to the patient, the site of delivery (gastrointestinal tract and children no suppression of urinary cortisol is seen with doses
lung), the delivery system used, and the individual dier- of BDP of 800 g or less. In studies where plasma cortisol
ences in the patients response to the corticosteroid. Recent has been measured at frequent intervals there was a signi-
studies suggest that systemic eects of inhaled corticoster- cant reduction in cortisol peaks with doses of inhaled BDP as
oid are less in patients with more severe asthma, presumably low as 400 g daily, although this does not appear to be dose
as less drug reaches the lung periphery [113, 114]. related in the range 4001000 g . The clinical signicance of
The systemic eect of an inhaled corticosteroid is these eects is not certain, however.
dependent on the amount of drug absorbed into the sys-
temic circulation. Approximately 8090% of the inhaled
dose from an MDI deposits in the oropharynx and is swal- Bone metabolism
lowed and subsequently absorbed from the gastrointes- Corticosteroids lead to a reduction in bone mass by direct
tinal tract. Use of a large volume spacer device markedly eects on bone formation and resorption and indirectly by
reduces the oropharyngeal deposition, and therefore the suppression of the pituitarygonadal and HPA axes, eects
systemic eects of inhaled corticosteroids, although this is on intestinal calcium absorption, renal tubular calcium rea-
less important when oral bioavailability is minimal, as with bsorption, and secondary hyperparathyroidism [115]. The
FP. For dry powder inhalers similar reductions in systemic eects of oral corticosteroids on osteoporosis and increased
eects may be achieved with mouth washing and discarding risk of vertebral and rib fractures are well known, but there
the uid. All patients using a daily dose of 800 g of an are no reports suggesting that long-term treatment with
inhaled corticosteroid should therefore use either a spacer or inhaled corticosteroids is associated with an increased risk
a mouthwash to reduce systemic absorption. Approximately of fractures. Bone densitometry has been used to assess the
10% of an MDI enters the lung and this fraction (which eect of inhaled corticosteroids on bone mass. Although
presumably exerts the therapeutic eect) may be absorbed there is evidence that bone density is less in patients tak-
into the systemic circulation. As the fraction of inhaled cor- ing high dose inhaled corticosteroids, interpretation is
ticosteroid deposited in the oropharynx is reduced, the pro- confounded by the fact that these patients are also taking
portion of the inhaled dose entering the lungs is increased. intermittent courses of oral corticosteroids. Changes in bone
More ecient delivery to the lungs is therefore accompa- mass occur very slowly and several biochemical indices have
nied by increased systemic absorption, but this is oset by a been used to assess the short-term eects of inhaled corti-
reduction in the dose needed for optimal control of airway costeroids on bone metabolism. Bone formation has been
inammation. For example, a multiple dry powder deliv- measured by plasma concentrations of bone-specic alka-
ery system, the Turbuhaler, delivers approximately twice as line phosphatase, serum osteocalcin, or procollagen peptides.
much corticosteroid to the lungs as other devices, and there- Bone resorption may be assessed by urinary hydroxyproline
fore has increased systemic eects. However this is compen- after a 12-h fast, urinary calcium excretion, and pyridinium
sated for by the fact that only half the dose is required. cross-link excretion. Inhaled corticosteroids, even at doses

648
Corticosteroids 51
up to 2000 g daily, have no signicant eect on calcium factor. Longitudinal studies have demonstrated that there is no
excretion, but acute and reversible dose-related suppres- signicant eect of inhaled corticosteroids on statural growth
sion of serum osteocalcin has been reported with BDP and in doses of up to 800 g daily and for up to 5 years of treat-
budesonide when given by conventional MDI in several ment [32]. A meta-analysis of 21 studies, including over 800
studies. Budesonide consistently has less eect than BDP at children, showed no eect of inhaled BDP on statural height,
equivalent doses and only BDP increases urinary hydroxy- even with higher doses and long duration of therapy [120]
proline at high doses. With a large volume spacer even doses and in a large study of asthmatics treated with inhaled corti-
of 2000 g daily of either BDP or budesonide are with- costeroids during childhood there was no dierence in statural
out eect on plasma osteocalcin concentrations, however. height compared to normal children [121]. Another long-term
Urinary pyridinium and deoxypyridinoline cross-links, which follow-up study showed no eect of corticosteroids on nal
are a more accurate and stable measurement of bone and col- height in children treated over several years [122]. Short-term
lagen degradation, are not increased with inhaled corticoster- growth measurements (knemometry) have demonstrated
oids (BDP  1000 g daily), even with intermittent courses that even a low dose of an oral corticosteroid (prednisolone
of oral corticosteroids. It is important to monitor changes 2.5 mg) is sucient to give complete suppression of lower leg
in markers of bone formation as well as bone degradation, growth. However inhaled budesonide up to 400 g is without
as the net eect on bone turnover is important. There is no eect, although some suppression is seen with 800 g and with
evidence that inhaled corticosteroids increase the frequency 400 g BDP. The relationship between knemometry measure-
of fractures. Long-term treatment with high dose inhaled ments and nal height are uncertain since low doses of oral
corticosteroids has not been associated with any consistent corticosteroid that have no eect on nal height cause pro-
change in bone density. Indeed, in elderly patients there may found suppression.
be an increase in bone density due to increased mobility.
Metabolic effects
Connective tissue effects Several metabolic eects have been reported after inhaled
Oral and topical corticosteroids cause thinning of the skin, corticosteroids, but there is no evidence that these are clini-
telangiectasiae, and easy bruising, probably as a result of loss cally relevant at therapeutic doses. In adults, fasting glucose
of extracellular ground substance within the dermis, due to and insulin are unchanged after doses of BDP up to 2000 g
an inhibitory eect on dermal broblasts. There are reports daily and in children with inhaled budesonide up to 800 g
of increased skin bruising and purpura in patients using daily. In normal individuals high dose inhaled BDP may
high doses of inhaled BDP, but the amount of intermittent slightly increase resistance to insulin. However, in patients
oral corticosteroids in these patients is not known. Easy with poorly controlled asthma high doses of BDP and budes-
bruising in association with inhaled corticosteroids is more onide paradoxically decrease insulin resistance and improve
frequent in elderly patients [116] and there are no reports glucose tolerance, suggesting that the disease itself may lead
of this problem in children. Long-term prospective studies to abnormalities in carbohydrate metabolism. Neither BDP
with objective measurements of skin thickness are needed 2000 g daily in adults nor budesonide 800 g daily in chil-
with dierent inhaled corticosteroids. dren have any eect on plasma cholesterol or triglycerides.

Cataracts Psychiatric effects


Long-term treatment with oral corticosteroids increases the There are various reports of psychiatric disturbance, includ-
risk of posterior subcapsular cataracts and there are several ing emotional lability, euphoria, depression, aggressiveness
case reports describing cataracts in individual patients taking and insomnia, after inhaled corticosteroids. Only eight such
inhaled corticosteroids [32]. In a recent cross-sectional study in patients have so far been reported, suggesting that this is
patients aged 525 years taking either inhaled BDP or budeso- very infrequent and a causal link with inhaled corticoster-
nide no cataracts were found on slit-lamp examination, even in oids has usually not been established.
patients taking 2000 g daily for over 10 years [117]. A slight
increase in the risk of glaucoma in patients taking very high Pregnancy
doses of inhaled corticosteroids has also been identied [118]. Based on extensive clinical experience inhaled corticoster-
oids appear to be safe in pregnancy, although no controlled
Growth studies have been performed. There is no evidence for any
adverse eects of inhaled corticosteroids on the pregnancy,
There has been particular concern that inhaled corticoster- the delivery, or on the fetus [123]. It is important to recog-
oids may cause stunting of growth and several studies have nize that poorly controlled asthma may increase the inci-
addressed this issue. Asthma itself (as with other chronic dence of perinatal mortality and retard intrauterine growth,
diseases) may have an eect on the growth pattern and has so that more eective control of asthma with inhaled corti-
been associated with delayed onset of puberty and decelera- costeroids may reduce these problems.
tion of growth velocity that is more pronounced with more
severe disease [119]. However, asthmatic children appear to
grow for longer, so that their nal height is normal. The eect Side effects in COPD
of asthma on growth make it dicult to assess the eects of Patients with COPD are elderly and are likely to have
inhaled corticosteroids on growth in cross-sectional studies, increased systemic side eects from inhaled corticosteroids
particularly as courses of oral corticosteroids is a confounding as they have several additional risk factors. There have been

649
Asthma and COPD: Basic Mechanisms and Clinical Management

fewer studies of systemic side eects in COPD patients. Intramuscular triamcinolone acetonide (80 mg monthly)
However, a systematic review found no reduction in bone has been advocated in patients with severe asthma as an alter-
mineral density or increase in fractures in COPD patients native to oral corticosteroids [126, 127]. This may be consid-
treated for up to 3 years with inhaled corticosteroids [83]. ered in patients in whom compliance is a particular problem,
An epidemiological study showed an increase in cataracts but the major concern is the high frequency of proximal
which are more common in an elderly population [124]. myopathy associated with this uorinated corticosteroid. Some
Many patients with COPD suer from comorbidities, patients who do not respond well to prednisolone are reported
including hypertension, metabolic syndrome and diabetes, to respond to oral betamethasone, presumably because of phar-
and may therefore have a worsening of these conditions, but macokinetic handling problems with prednisolone.
this has not yet been systematically investigated.

Acute severe asthma


Intravenous hydrocortisone is given in acute severe asthma,
SYSTEMIC CORTICOSTEROIDS with a recommended dose of 200 mg intravenously. While
the value of corticosteroids in acute severe asthma has been
Oral or intravenous corticosteroids may be indicated in questioned, others have found that they speed the resolution
several situations. Prednisone is converted in the liver to of attacks [128]. There is no apparent advantage in giving very
the active prednisolone. In pregnant patients prednisone is high doses of intravenous corticosteroids (such as methylpred-
preferable as it is not converted to prednisolone in the fetal nisolone 1 g). Indeed, intravenous corticosteroids have occa-
liver, thus diminishing the exposure of the fetus to corti- sionally been associated with an acute severe myopathy [129].
costeroids. Enteric-coated preparations of prednisolone are No dierence in recovery from acute severe asthma was seen
used to reduce side eects (particularly gastric side eects) whether intravenous hydrocortisone in doses of 50, 200, or
and give delayed and reduced peak plasma concentrations, 500 mg 6 hourly were used [130], and another placebo-con-
although the bioavailability and therapeutic ecacy of these trolled study showed no benecial eect of intravenous cor-
preparations is similar to uncoated tablets. Prednisolone and ticosteroids [131]. Intravenous corticosteroids are indicated
prednisone are preferable to dexamethasone, betametha- in acute asthma if lung function is 30% predicted and in
sone, or triamcinolone, which have longer plasma half-lives whom there is no signicant improvement with nebulized
and therefore an increased frequency of adverse eects. 2-agonist. Intravenous therapy is usually given until a satisfac-
tory response is obtained and then oral prednisolone may be
substituted. Oral prednisolone (4060 mg) has a similar eect
Short courses to intravenous hydrocortisone and is easier to administer [38,
128]. Oral prednisolone is the preferred treatment for acute
Short courses of oral corticosteroids (3040 mg pred- severe asthma, providing there are no contraindications to oral
nisolone daily for 12 weeks or until the peak ow values therapy. There is some evidence that high doses of nebulized
return to best attainable) are indicated for exacerbations of corticosteroids may also be eective in acute exacerbations of
asthma, and the dose may be tailed o over 1 week once the asthma, with a more rapid onset of action [132].
exacerbation is resolved. The tail-o period is not strictly
necessary, but some patients nd it reassuring.
References
Maintenance corticosteroids 1. Barnes PJ. How corticosteroids control inammation. Br J Pharmacol
148: 24554, 2006.
Maintenance oral corticosteroids are only needed in a small 2. Rhen T, Cidlowski JA. Antiinammatory action of glucocorticoids
proportion of asthmatic patients (approximately 1%) with New mechanisms for old drugs. N Engl J Med 353: 171123, 2005.
3. Gibson PG, Saltos N, Fakes K. Acute anti-inammatory eects of
the most severe asthma that cannot be controlled with
inhaled budesonide in asthma: A randomized controlled trial. Am J
maximal doses of inhaled corticosteroids (2000 g daily) Respir Crit Care Med 163: 3236, 2001.
and additional bronchodilators. The minimal dose of oral 4. Ketchell RI, Jensen MW, Lumley P, Wright AM, Allenby MI,
corticosteroid needed for control should be used and reduc- OConnor BJ. Rapid eect of inhaled uticasone propionate on air-
tions in the dose should be made slowly in patients who way responsiveness to adenosine 5-monophosphate in mild asthma.
have been on oral corticosteroids for long periods (e.g. by J Allergy Clin Immunol 110: 6036, 2002.
2.5 mg per month for doses down to 10 mg daily and there- 5. Juniper EF, Kline PA, Yan Zieleshem MA, Ramsdale EH, OByrne
after by 1 mg per month). Oral corticosteroids are usually PM, Hargreave FE. Long-term eects of budesonide on airway
given as a single morning dose, as this reduces the risk of responsiveness and clinical asthma severity in inhaled steroid-depend-
adverse eects since it coincides with the peak diurnal con- ent asthmatics. Eur Respir J 3: 112227, 1990.
6. Lewis-Tun LJ, Cidlowski JA. The physiology of human glucocorti-
centrations. There is some evidence that administration in
coid receptor beta (hGRbeta) and glucocorticoid resistance. Ann NY
the afternoon may be optimal for some patients who have Acad Sci 1069: 19, 2006.
severe nocturnal asthma [125]. Alternate day administra- 7. Pujols L, Mullol J, Picado C. Alpha and beta glucocorticoid receptors:
tion may also reduce adverse eects, but control of asthma Relevance in airway diseases. Curr Allergy Asthma Rep 7: 9399, 2007.
may not be as good on the day when the oral dose is omit- 8. Barnes PJ. Corticosteroid eects on cell signalling. Eur Respir J 27:
ted in some patients. 41326, 2006.

650
Corticosteroids 51
9. Clark AR. MAP kinase phosphatase 1: A novel mediator of biological 31. Gambertoglio JG, Amend WJC, Benet LZ. Pharmacokinetics and bio-
eects of glucocorticoids?. J Endocrinol 178: 512, 2003. availability of prednisone and prednisolone in healthy volunteers and
10. Dostert A, Heinzel T. Negative glucocorticoid receptor response ele- patients: A review. J Pharmacokin Biopharm 8: 152, 1980.
ments and their role in glucocorticoid action. Curr Pharm Des 10: 32. Barnes PJ, Pedersen S, Busse WW. Ecacy and safety of inhaled corti-
280716, 2004. costeroids: An update. Am J Respir Crit Care Med 157: S153, 1998.
11. Barnes PJ, Adcock IM. How do corticosteroids work in asthma?. Ann 33. Lipworth BJ. Systemic adverse eects of inhaled corticosteroid therapy:
Intern Med 139: 35970, 2003. A systematic review and meta-analysis [see comments]. Arch Intern
12. Barnes PJ, Adcock IM, Ito K. Histone acetylation and deacetylation: Med 159: 94155, 1999.
Importance in inammatory lung diseases. Eur Respir J 25: 55263, 34. Derendorf H, Hochhaus G, Meibohm B, Mollmann H, Barth J.
2005. Pharmacokinetics and pharmacodynamics of inhaled corticosteroids.
13. Hart L, Lim S, Adcock I, Barnes PJ, Chung KF. Eects of inhaled cor- J Allergy Clin Immunol 101: S44046, 1998.
ticosteroid therapy on expression and DNA-binding activity of nuclear 35. Brown PH, Greening AP, Crompton GK. Large volume spacer devices
factor-kB in asthma. Am J Respir Crit Care Med 161: 22431, 2000. and the inuence of high dose beclomethasone diproprionate on
14. Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of hypothalamo-pituitary-adrenal axis function. Thorax 48: 23338, 1993.
histone deacetylase 2 inhibits IL-1b-induced histone H4 acetylation 36. Derendorf H. Corticosteroid pharmacokinetic/pharmacodynamic
on lysines 8 and 12. Mol Cell Biol 20: 6891903, 2000. parameters and their relationship to safety and ecacy. Allergy Asthma
15. Bergmann MW, Staples KJ, Smith SJ, Barnes PJ, Newton R. Proc 26: 32735, 2005.
Glucocorticoid inhibition of GM-CSF from T cells is independent of 37. Derendorf H. Pharmacokinetic and pharmacodynamic properties of
control by NF-B and CLE0. Am J Respir Cell Mol. Biol 30: 55563, 2004. inhaled ciclesonide. J Clin Pharmacol 47: 78289, 2007.
16. Brook M, Tchen CR, Santalucia T, McIlrath J, Arthur JS, Saklatvala J 38. Harrison BDN, Stokes TC, Hart GJ, Vaughan DA, Ali NJ, Robinson
et al. Posttranslational regulation of tristetraprolin subcellular locali- AA. Need for intravenous hydrocortisone in addition to oral pred-
zation and protein stability by p38 mitogen-activated protein kinase nisolone in patients admitted to hospital with severe asthma without
and extracellular signal-regulated kinase pathways. Mol Cell Biol 26: ventilatory failure. Lancet i: 18184, 1986.
240818, 2006. 39. Storr J, Barrell E, Barry W, Lenney W, Hatcher G. Eect of a single
17. Thomson NC, Spears M. The inuence of smoking on the treatment oral dose of prednisolone in acute childhood asthma. Lancet 1: 87982,
response in patients with asthma. Curr Opin Allergy Clin Immunol 5: 1987.
5763, 2005. 40. Levy ML, Stevenson C, Maslen T. Comparison of short courses of oral
18. Adcock IM, Lane SJ. Corticosteroid-insensitive asthma: Molecular prednisolone and uticasone propionate in the treatment of adults with
mechanisms. J Endocrinol 178: 34755, 2003. acute exacerbations of asthma in primary care. Thorax 51: 108792, 1996.
19. Ito K, Ito M, Elliott WM, Cosio B, Caramori G, Kon OM et al. 41. Manjra AI, Price J, Lenney W, Hughes S, Barnacle H. Ecacy of neb-
Decreased histone deacetylase activity in chronic obstructive pulmo- ulized uticasone propionate compared with oral prednisolone in children
nary disease. N Engl J Med 352: 196776, 2005. with an acute exacerbation of asthma. Respir Med 94: 120614, 2000.
20. Hew M, Bhavsar P, Torrego A, Meah S, Khorasani N, Barnes PJ et al. 42. Harrison TW, Oborne J, Newton S, Tatterseld AE. Doubling the dose
Relative corticosteroid insensitivity of peripheral blood mononuclear of inhaled corticosteroid to prevent asthma exacerbations: Randomised
cells in severe asthma. Am J Respir Crit Care Med 174: 13441, 2006. controlled trial. Lancet 363: 27175, 2004.
21. Barnes PJ. Reduced histone deacetylase in COPD: Clinical implica- 43. FitzGerald JM, Becker A, Sears MR, Mink S, Chung K, Lee J.
tions. Chest 129: 15155, 2006. Doubling the dose of budesonide versus maintenance treatment in
22. Irusen E, Matthews JG, Takahashi A, Barnes PJ, Chung KF, Adcock asthma exacerbations. Thorax 59: 55056, 2004.
IM. p38 Mitogen-activated protein kinase-induced glucocorticoid 44. Foresi A, Morelli MC, Catena E. Low-dose budesonide with the addition
receptor phosphorylation reduces its activity: Role in steroid-insensitive of an increased dose during exacerbations is eective in long-term asthma
asthma. J Allergy Clin Immunol 109: 64957, 2002. control. On behalf of the Italian Study Group. Chest 117: 44046, 2000.
23. Matthews JG, Ito K, Barnes PJ, Adcock IM. Defective glucocorticoid 45. Edmonds ML, Camargo CA Jr., Pollack CV Jr., Rowe BH. Early use
receptor nuclear translocation and altered histone acetylation pat- of inhaled corticosteroids in the emergency department treatment of
terns in glucocorticoid-resistant patients. J Allergy Clin Immunol 113: acute asthma. Cochrane Database Syst Rev, 2003, CD002308.
11008, 2004. 46. Global Initiative for Asthma. Global strategy for asthma management and
24. Barnes PJ. Scientic rationale for combination inhalers with a long- prevention. NHLBI/WHO Workshop Report. http://www.ginasthma.com
acting 2-agonists and corticosteroids. Eur Respir J 19: 18291, 2002. 2006.
25. Mak JCW, Nishikawa M, Barnes PJ. Glucocorticosteroids increase 47. Haahtela T, Jarvinen M, Kava T, Kiviranta K, Koskinen S, Lehtonen K
2-adrenergic receptor transcription in human lung. Am J Physiol 12: et al. Comparison of a 2-agonist terbutaline with an inhaled steroid in
L4146, 1995. newly detected asthma. N Engl J Med 325: 38892, 1991.
26. Baraniuk JN, Ali M, Brody D, Maniscalco J, Gaumond E, Fitzgerald T 48. Juniper EF, Kline PA, Vanzieleghem MA, Ramsdale EH, OByrne PM,
et al. Glucocorticoids induce 2-adrenergic receptor function in human Hargreave FE. Eect of long-term treatment with an inhaled corticos-
nasal mucosa. Am J Respir Crit Care Med 155: 70410, 1997. teroid (budesonide) on airway hyperresponsiveness and clinical asthma in
27. Mak JCW, Nishikawa M, Shirasaki H, Miyayasu K, Barnes PJ. nonsteroid-dependent asthmatics. Am Rev Respir Dis 142: 83236, 1990.
Protective eects of a glucocorticoid on down-regulation of pulmonary 49. Pauwels RA, Pedersen S, Busse WW, Tan WC, Chen YZ, Ohlsson SV
2-adrenergic receptors in vivo. J Clin Invest 96: 99106, 1995. et al. Early intervention with budesonide in mild persistent asthma: A
28. Mak JC, Chuang TT, Harris CA, Barnes PJ. Increased expression of G randomised, double-blind trial. Lancet 361: 107176, 2003.
protein-coupled receptor kinases in cystic brosis lung. Eur J Pharmacol 50. Mash B, Bheekie A, Jones PW. Inhaled vs oral steroids for adults
436: 16572, 2002. with chronic asthma. Cochrane Database Syst Rev 2, 2002, CD002160:
29. Roth M, Johnson PR, Rudiger JJ, King GG, Ge Q, Burgess JK et al. CD002160.
Interaction between glucocorticoids and 2 agonists on bronchial air- 51. Vathenen AS, Knox AJ, Wisniewski A, Tatterseld AE. Time course of
way smooth muscle cells through synchronised cellular signalling. change in bronchial reactivity with an inhaled corticosteroid in asthma.
Lancet 360: 129399, 2002. Am Rev Respir Dis 143: 131721, 1991.
30. Usmani OS, Ito K, Maneechotesuwan K, Ito M, Johnson M, Barnes 52. Jatakanon A, Lim S, Barnes PJ. Changes in sputum eosinophils predict
PJ et al. Glucocorticoid receptor nuclear translocation in airway cells loss of asthma control. Am J Respir Crit Care Med 161: 6472, 2000.
following inhaled combination therapy. Am J Respir Crit Care Med 172: 53. Leuppi JD, Salome CM, Jenkins CR, Anderson SD, Xuan W, Marks
70412, 2005. GB et al. Predictive markers of asthma exacerbation during stepwise

651
Asthma and COPD: Basic Mechanisms and Clinical Management

dose reduction of inhaled corticosteroids. Am J Respir Crit Care Med. 73. Selroos O, Pietinalcho A, Lofroos A-B, Riska A. Eect of early and
163: 40612, 2001. late intervention with inhaled corticosteroids in asthma. Chest 108:
54. van Essen-Zandvliet EE, Hughes MD, Waalkens HJ, Duiverman 122834, 1995.
EJ, Pocock SJ, Kerrebijn KF. Eects of 22 months of treatment with 74. Suissa S, Ernst P, Benayoun S, Baltzan M, Cai B. Low-dose inhaled
inhaled corticosteroids and/or 2-agonists on lung function, airway corticosteroids and the prevention of death from asthma. N Engl J Med
responsiveness and symptoms in children with asthma. Am Rev Respir 343: 33236, 2000.
Dis 146: 54754, 1992. 75. Ernst P, Habbick B, Suissa S, Hemmelgarn B, Cockcroft D, Buist
55. Waalkens HJ, van Essen-Zandvliet EE, Hughes MD, Gerritsen J, AS et al. Is the association between inhaled beta-agonist use and life-
Duiverman EJ, Knol K et al. Cessation of long-term treatment with threatening asthma because of confounding by severity? Am Rev Respir
inhaled corticosteroids (budesonide) in children with asthma results in Dis 148: 7579, 1993.
deterioration. Am Rev Respir Dis 148: 125257, 1993. 76. Nelson HS, Weiss ST, Bleecker ER, Yancey SW, Dorinsky PM. The
56. Berger WE. Budesonide inhalation suspension for the treatment of Salmeterol Multicenter Asthma Research Trial: A comparison of usual
asthma in infants and children. Drugs 65: 197389, 1973. pharmacotherapy for asthma or usual pharmacotherapy plus salmeterol.
57. Busse WW, Chervinsky P, Condemi J, Lumry WR, Petty TL, Rennard Chest 129: 1526, 2006.
S et al. Budesonide delivered by Turbuhaler is eective in a dose- 77. Nathan RA, Nayak AS, Graft DF, Lawrence M, Picone FJ, Ahmed
dependent fashion when used in the treatment of adult patients with T et al. Mometasone furoate: Ecacy and safety in moderate asthma
chronic asthma. J Allergy Clin Immunol 101: 45763, 1998. compared with beclomethasone dipropionate. Ann Allergy Asthma
58. Adams NP, Jones PW. The dose-response characteristics of inhaled Immunol 86: 20310, 2001.
corticosteroids when used to treat asthma: An overview of Cochrane 78. Reynolds NA, Scott LJ. Ciclesonide. Drugs 64: 51119, 2004.
systematic reviews. Respir Med 100: 1297306, 2006. 79. Malo J-L, Cartier A, Merland N, Ghezzo H, Burke A, Morris J et al.
59. Pauwels RA, Lofdahl C-G, Postma DS, Tatterseld AE, OByrne PM, Four-times-a-day dosing frequency is better than twice-a-day regimen
Barnes PJ et al. Eect of inhaled formoterol and budesonide on exacer- in subjects requiring a high-dose inhaled steroid, budesonide, to con-
bations of asthma. N Engl J Med 337: 141218, 1997. trol moderate to severe asthma. Am Rev Respir Dis 140: 62428, 1989.
60. Lim S, Jatakanon A, John M, Gilbey T, OConnor BJ, Chung KF et al. 80. Otulana BA, Varma N, Bullock A, Higenbottam T. High dose neb-
Eect of inhaled budesonide on lung function and airway inamma- ulized steroid in the treatment of chronic steroid-dependent asthma.
tion. Assessment by various inammatory markers in mild asthma. Resp Med 86: 1058, 1992.
Am J Respir Crit Care Med 159: 2230, 1999. 81. Hawkins G, McMahon AD, Twaddle S, Wood SF, Ford I, Thomson
61. Jatakanon A, Lim S, Chung KF, Barnes PJ. An inhaled steroid NC. Stepping down inhaled corticosteroids in asthma: Randomised
improves markers of inammation in asymptomatic steroid-naive asth- controlled trial. BMJ 326: 1115, 2003.
matic patients. Eur Respir J 12: 108488, 1998. 82. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P et al.
62. Jatakanon A, Kharitonov S, Lim S, Barnes PJ. Eect of diering doses Global strategy for the diagnosis, management, and prevention of
of inhaled budesonide on markers of airway inammation in patients COPD-2006 Update. Am J Respir Crit Care Med 176: 53255, 2007.
with mild asthma. Thorax 54: 10814, 1999. 83. Yang IA, Fong KM, Sim EH, Black PN, Lasserson TJ. Inhaled cor-
63. Kharitonov SA, Donnelly LE, Montuschi P, Corradi M, Collins JV, ticosteroids for stable chronic obstructive pulmonary disease. Cochrane
Barnes PJ. Dose-dependent onset and cessation of action of inhaled Database.Syst Rev, 2007, CD002991.
budesonide on exhaled nitric oxide and symptoms in mild asthma. 84. Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones
Thorax 57: 88996, 2002. PW et al. Salmeterol and uticasone propionate and survival in chronic
64. Sont JK, Willems LN, Bel EH, van Krieken JH, Vandenbroucke JP, obstructive pulmonary disease. N Engl J Med 356: 77589, 2007.
Sterk PJ. Clinical control and histopathologic outcome of asthma when 85. Kankaanranta H, Lahdensuo A, Moilanen E, Barnes PJ. Add-on ther-
using airway hyperresponsiveness as an additional guide to long-term apy options in asthma not adequately controlled by inhaled corticoster-
treatment. The AMPUL Study Group. Am J Respir Crit Care Med 159: oids: A comprehensive review. Respir Res 5: 17, 2004.
104351, 1999. 86. Greening AP, Ind PW, Northeld M, Shaw G. Added salmeterol ver-
65. Green RH, Brightling CE, McKenna S, Hargadon B, Parker D, sus higher-dose corticosteroid in asthma patients with symptoms on
Bradding P et al. Asthma exacerbations and sputum eosinophil counts: existing inhaled corticosteroid. Lancet 344: 21924, 1994.
A randomised controlled trial. Lancet 360: 171521, 2002. 87. Shrewsbury S, Pyke S, Britton M. Meta-analysis of increased dose
66. Jayaram L, Pizzichini MM, Cook RJ, Boulet LP, Lemiere C, Pizzichini of inhaled steroid or addition of salmeterol in symptomatic asthma
E et al. Determining asthma treatment by monitoring sputum cell (MIASMA). BMJ 320: 136873, 2000.
counts: Eect on exacerbations. Eur Respir J 27: 48394, 2006. 88. OByrne PM, Barnes PJ, Rodriguez-Roisin R, Runnerstrom E,
67. Smith AD, Cowan JO, Brassett KP, Herbison GP, Taylor DR. Use Sandstrom T, Svensson K et al. Low dose inhaled budesonide and for-
of exhaled nitric oxide measurements to guide treatment in chronic moterol in mild persistent asthma: The OPTIMA randomized trial.
asthma. N Engl J Med 352: 216373, 2005. Am J Respir Crit Care Med 164: 139297, 2001.
68. Dompeling E, Van Schayck CP, Molema J, Folgering H, van Grusven 89. Gibson PG, Powell H, Ducharme FM. Dierential eects of maintenance
PM, van Weel C. Inhaled beclomethasone improves the course of long-acting beta-agonist and inhaled corticosteroid on asthma control and
asthma and COPD. Eur Resp J 5: 94552, 1992. asthma exacerbations. J Allergy Clin Immunol 119: 34450, 2007.
69. OByrne PM, Pedersen S, Busse WW, Tan WC, Chen YZ, Ohlsson 90. Ni CM, Greenstone IR, Ducharme FM. Addition of inhaled long-
SV et al. Eects of early intervention with inhaled budesonide on lung acting beta2-agonists to inhaled steroids as rst line therapy for per-
function in newly diagnosed asthma. Chest 129: 147885, 2006. sistent asthma in steroid-naive adults. Cochrane Database Syst.Rev,
70. Lange P, Scharling H, Ulrik CS, Vestbo J. Inhaled corticosteroids and 2005, CD005307.
decline of lung function in community residents with asthma. Thorax 91. OByrne PM, Bisgaard H, Godard PP, Pistolesi M, Palmqvist M,
61: 1004, 2006. Zhu Y et al. Budesonide/formoterol combination therapy as both
71. Haahtela T, Jrvinsen M, Kava T, Kiviranta K, Koskinen S, Lemtonen maintenance and reliever medication in asthma. Am J Resp Crit Care
K et al. Eects of reducing or discontinuing inhaled budesonide in Med 171: 12936, 2005.
patients with mild asthma. N Engl J Med 331: 7005, 1994. 92. Rabe KF, Pizzichini E, Stallberg B, Romero S, Balanzat AM, Atienza
72. Agertoft L, Pedersen S. Eects of long-term treatment with an inhaled T et al. Budesonide/formoterol in a single inhaler for maintenance and
corticosteroid on growth and pulmonary function in asthmatic chil- relief in mild-to-moderate asthma: A randomized, double-blind trial.
dren. Resp Med 5: 36972, 1994. Chest 129: 24656, 2006.

652
Corticosteroids 51
93. Rabe KF, Atienza T, Magyar P, Larsson P, Jorup C, Lalloo UG. Eect 111. Ernst P, Gonzalez AV, Brassard P, Suissa S. Inhaled corticosteroid use
of budesonide in combination with formoterol for reliever therapy in in chronic obstructive pulmonary disease and the risk of hospitaliza-
asthma exacerbations: A randomised controlled, double-blind study. tion for pneumonia. Am J Respir Crit Care Med 176: 16266, 2007.
Lancet 368: 74453, 2006. 112. Kamada AK, Szeer SJ, Martin RJ, Boushey HA, Chinchilli VM,
94. Tatterseld AE, Postma DS, Barnes PJ, Svensson K, Bauer CA, Drazen JM et al. Issues in the use of inhaled steroids. Am J Respir Crit
OByrne PM et al. Exacerbations of asthma. A descriptive study of Care Med 153: 173948, 1996.
425 severe exacerbations. Am J Respir Crit Care Med 160: 59499, 113. Brutsche MH, Brutsche IC, Munawar M, Langley SJ, Masterson
1999. CM, Daley-Yates PT et al. Comparison of pharmacokinetics and
95. Barnes PJ. New therapies for asthma. Trends Mol Med 12: 51520, systemic eects of inhaled uticasone propionate in patients with
2006. asthma and healthy volunteers: A randomised crossover study. Lancet
96. Calverley P, Pauwels R, Vestbo J, Jones P, Pride N, Gulsvik A et al. 356: 55661, 2000.
Combined salmeterol and uticasone in the treatment of chronic 114. Harrison TW, Wisniewski A, Honour J, Tatterseld AE. Comparison
obstructive pulmonary disease: A randomoised controlled trial. Lancet of the systemic eects of uticasone propionate and budesonide given
361: 44956, 2003. by dry powder inhaler in healthy and asthmatic subjects. Thorax 56:
97. Szafranski W, Cukier A, Ramirez A, Menga G, Sansores R, 18691, 2001.
Nahabedian S et al. Ecacy and safety of budesonide/formoterol in 115. Efthimou J, Barnes PJ. Eect of inhaled corticosteroids on bone and
the management of chronic obstructive pulmonary disease. Eur Respir growth. Eur Respir J 11: 116777, 1998.
J 21: 7481, 2003. 116. Roy A, Leblanc C, Paquette L, Ghezzo H, Cote J, Cartier A et al.
98. Nannini L, Cates C, Lasserson T, Poole P. Combined corticosteroid Skin bruising in asthmatic subjects treated with high doses of inhaled
and long-acting beta-agonist in one inhaler versus long-acting beta- steroids: Frequency and association with adrenal function. Eur Respir
agonists for chronic obstructive pulmonary disease. Cochrane Database J 9: 22631, 1996.
Syst Rev, 2007, CD006829. 117. Simons FER, Persaud MP, Gillespie CA, Cheang M, Shuckett EP.
99. Wedzicha JA, Calverley PM, Seemungal TA, Hagan G, Ansari Z, Absence of posterior subcapsular cataracts in young patients treated
Stockley RA. The prevention of chronic obstructive pulmonary dis- with inhaled glucocorticoids. Lancet 342: 73638, 1993.
ease exacerbations by salmeterol/uticasone propionate or tiotropium 118. Garbe E, LeLorier J, Boivin J-F, Suissa S. Inhaled and nasal glucocor-
bromide. Am J Respir Crit Care Med 177: 1926, 2008. ticoids and the risks of ocular hypertension or open-angle glaucoma.
100. Evans DJ, Taylor DA, Zetterstrom O, Chung KF, OConnor BJ, JAMA 227: 72227, 1997.
Barnes PJ. A comparison of low-dose inhaled budesonide plus theo- 119. Pedersen S. Do inhaled corticosteroids inhibit growth in children?
phylline and high-dose inhaled budesonide for moderate asthma. Am J Respir Crit Care Med 164: 52135, 2001.
N Engl J Med 337: 141218, 1997. 120. Allen DB, Mullen M, Mullen B. A meta-analysis of the eects of
101. Ukena D, Harnest U, Sakalauskas R, Magyar P, Vetter N, Steen oral and inhaled corticosteroids on growth. J Allergy Clin Immunol 93:
H et al. Comparison of addition of theophylline to inhaled steroid 96776, 1994.
with doubling of the dose of inhaled steroid in asthma. Eur Respir J 121. Silverstein MD, Yunginger JW, Reed CE, Petterson T, Zimmerman
10: 275460, 1997. D, Li JT et al. Attained adult height after childhood asthma: Eect of
102. Lim S, Jatakanon A, Gordon D, Macdonald C, Chung KF, Barnes glucocorticoid therapy. J Allergy Clin Immunol 99: 46674, 1997.
PJ. Comparison of high dose inhaled steroids, low dose inhaled ster- 122. Agertoft L, Pedersen S. Eect of long-term treatment with inhaled
oids plus low dose theophylline, and low dose inhaled steroids alone budesonide on adult height in children with asthma. N Engl J Med
in chronic asthma in general practice. Thorax 55: 83741, 2000. 343: 106469, 2000.
103. Wilson AJ, Gibson PG, Coughlan J. Long acting beta-agonists versus 123. Schatz M. Asthma and pregnancy. Lancet 353: 12024, 1999.
theophylline for maintenance treatment of asthma. Cochrane Database 124. Ernst P, Baltzan M, Deschenes J, Suissa S. Low-dose inhaled
Syst Rev, 2000. , CD001281 and nasal corticosteroid use and the risk of cataracts. Eur Respir J
104. Laviolette M, Malmstrom K, Lu S, Chervinsky P, Pujet JC, Peszek I 27: 116874, 2006.
et al. Montelukast added to inhaled beclomethasone in treatment of 125. Beam WR, Ballard RD, Martin RJ. Spectrum of corticosteroid sensi-
asthma. Am J Respir Crit Care Med 160: 186268, 1999. tivity in nocturnal asthma. Am Rev Respir Dis 145: 108286, 1992.
105. Price DB, Hernandez D, Magyar P, Fiterman J, Beeh KM, James IG 126. McLeod DT, Capewell SJ, Law J, MacLaren W, Seaton A.
et al. Randomised controlled trial of montelukast plus inhaled budes- Intramuscular triamcinolone acetamide in chronic severe asthma.
onide versus double dose inhaled budesonide in adult patients with Thorax 40: 84045, 1985.
asthma. Thorax 58: 21116, 2003. 127. Ogirala RG, Aldrich TK, Prezant DJ, Sinnett MJ, Enden JB,
106. Nelson HS, Busse WW, Kerwin E, Church N, Emmett A, Rickard Williams MH. High dose intramuscular triamcinolone in severe
K et al. Fluticasone propionate/salmeterol combination provides more life-threatening asthma. N Engl J Med 329: 58589, 1991.
eective asthma control than low-dose inhaled corticosteroid plus 128. Engel T, Heinig JH. Glucocorticoid therapy in acute severe asthma
montelukast. J Allergy Clin Immunol 106: 108895, 2000. a critical review. Eur Respir J 4: 88189, 1991.
107. Ducharme F, Schwartz Z, Hicks G, Kakuma R. Addition of anti- 129. Decramer M, Lacquet LM, Fagard R, Rogiers P. Corticosteroids con-
leukotriene agents to inhaled corticosteroids for chronic asthma. tribute to muscle weakness in chronic airow obstruction. Am J Respir
Cochrane Database Syst Rev, 2004, CD003133. Crit Care Med 150: 1116, 1995.
108. Ducharme FM, Lasserson TJ, Cates CJ. Long-acting beta2-agonists 130. Bowler SD, Mitchell CA, Armstrong JG. Corticosteroids in acute
versus anti-leukotrienes as add-on therapy to inhaled corticosteroids severe asthma: Eectiveness of low doses. Thorax 47: 58487, 1992.
for chronic asthma. Cochrane Database Syst Rev, 2006., CD003137 131. Morell F, Orkiols R, de Gracia J, Curul V, Pujol A. Controlled trial of
109. Williamson IJ, Matusiewicz SP, Brown PH, Greening AP, Crompton intravenous corticosteroids in severe acute asthma. Thorax 47: 58891,
GK. Frequency of voice problems and cough in patients using pressu- 1992.
rised aersosol inhaled steroid preparations. Eur Resp J 8: 59092, 1995. 132. Devidayal, Singhi S, Kumar L, Jayshree M. Ecacy of nebulized
110. Toogood JA, Jennings B, Greenway RW, Chung L. Candidiasis budesonide compared to oral prednisolone in acute bronchial asthma.
and dysphonia complicating beclomethasone treatment of asthma. Acta Paediatr 88: 83540, 1999.
J Allergy Clin Immunol 65: 14553, 1980.

653
Mediator Antagonists
52CHAPTER

INTRODUCTION as LTB4 receptor antagonists, have no eect in


Kian Fan Chung and
asthma and are being considered for the treat-
ment of COPD. Peter J. Barnes
Inammatory mediators contribute to the
pathophysiology of asthma and chronic obstruc- National Heart and Lung Institute
tive pulmonary disease (COPD) (see Chapters (NHLI), Clinical Studies Unit,
2327) suggesting that antagonists of mediator 5
-Lipoxygenase inhibitors Imperial College, London, UK
receptors or inhibitors of their synthesis would
be benecial in treatment. However, because 5-LO is a critical enzyme involved in the gen-
a large number of mediators are involved and eration of leukotrienes. Inhibitors of 5-LO may
many mediators share similar eects on the air- be classied as direct inhibitors of the enzyme
ways, inhibitors of single mediators may have and indirect inhibitors that interfere with a
had little or no clinical benet to date. This nuclear membrane docking protein, 5-LO acti-
may underlie the fact that despite many years vating protein (FLAP), that is necessary for
of continuing eorts by pharmaceutical compa- enzyme activation. Many hydroxamates and
nies, only one class of mediator antagonists, the N-hydroxyureas are 5-LO inhibitors and act
anti-leukotrienes, has become established in the by interfering with the redox state of the active
treatment of asthma, representing the rst new binding site. Zileuton is the most extensively
class of therapy for asthma introduced in more investigated and is the only 5-LO inhibitor
than 40 years. So far, there has been no media- available for prescription in asthma (but only in
tor antagonists introduced for the treatment the USA) [1]. The eect of zileuton is similar
of COPD. In addition to reviewing the anti- to that of leukotriene receptor antagonists, and
leukotrienes, we will also review other classes of zileuton inhibits allergen- and exercise-induced
mediator antagonists that have failed to show asthma, as well as aspirin-induced asthma.
benecial eects or those currently under-inves- It decreases airway hyperresponsiveness and
tigation but whose potential has yet to be tested. inammatory cells in nocturnal exacerbations of
asthma [2]. In addition, zileuton inhibits eosi-
nophil inux induced by allergen challenge [3].
Zileuton has a short duration of action
and has to be taken four times daily; a twice
ANTI-LEUKOTRIENES daily controlled-release preparation is also
available [4]. Its side eects are mainly on the
liver, with frequent abnormalities of liver func-
Anti-leukotrienes can be divided into cysteinyl- tion tests. Other redox 5-LO inhibitors have
leukotriene (cys-LT) receptor antagonists that been developed but have not reached the mar-
antagonize the eects of cys-LTs, such as LTD4, ket. Non-redox 5-LO inhibitors and inhibitors
and leukotriene synthesis inhibitors that are of FLAP (e.g. MK-886, MK-591, Bay-x1005)
inhibitors of 5-lipoxygenase (5-LO) enzyme have been developed and are currently being
that generates cys-LTs and LTB4 (Fig. 52.1). developed further for clinical use in asthma and
Anti-leukotrienes are mainly indicated for the possibly COPD. One theoretical advantage of
treatment of asthma, and LTB4 inhibitors, such the 5-LO inhibitors on the receptor antagonists

655
Asthma and COPD: Basic Mechanisms and Clinical Management

Allergen
Exercise PAF

Aspirin MC Eos SO2

Arachidonic acid

5-LO inhibitors
5
-lipoxygenase 
zileuton
Plasma
exudation
Cysteinyl-leukotrienes
(LTC4, LTD4, LTE4)

LT-antagonists
pranlukast
Cys-LT1 receptors 
zafirlukast
montelukast

Mucus FIG. 52.1 Inhibition of effects of


secretion Bronchoconstriction Eos recruitment cysteinyl-leukotrienes (cys-LT) in
asthma.

is that they inhibit the formation of LTB4, and other 5-LO a single 40 mg dose of zarlukast produces a 100-fold shift
products such as eosxins, as well as cys-LTs. This may make of the LTD4 doseresponse curve, and signicant protec-
the drugs more applicable to other airway diseases where tion is present for 24 h [8]. Oral administration of leuko-
LTB4 may be involved, such as COPD and cystic brosis. triene receptor antagonist inhibits both the early and late
There is anecdotal evidence that zileuton was more eective responses to allergen, and exercise-induced asthma [9, 10].
in controlling severe asthma than cys-LT1 antagonists, sup- Leukotriene receptor antagonists are able to cause bron-
porting the view that other 5-LO products may be impor- chodilation, and their eect is additive to that of short-
tant, particularly in severe asthma. acting 2-agonists [11, 12].
In many studies of anti-leukotrienes such as zileuton,
zarlukast, pranlukast, and montelukast, their eective-
cys-LT antagonists ness has been compared with that of placebo in short-term
studies of 46 week duration [1316]. A greater increase in
The pathophysiological role of cys-LTs is discussed in FEV1, a reduction in asthma medication use and in asthma
Chapter 24. Cys-LTs cause airway obstruction through the symptoms, with an increase in morning peak ow has been
stimulation of specic receptors termed the cys-LT receptor demonstrated in mild-to-moderate severe asthma with
type 1 (Cyst-LT1) present on airway smooth muscle. This a some degree of airow obstruction and usually in cohorts
seven-transmembrane spanning, G-protein-coupled recep- not on inhaled corticosteroid therapy. These ndings have
tor, where the gene is mapped to the X chromosome [5]. been conrmed and extended in longer studies in patients
Signaling through cyst-LT1 occurs through stimulation of with mild-to-moderate chronic stable asthma with zileu-
phosphoinositide hydrolysis [6]. A second cys-LT receptor, ton, zarlukast, montelukast, all demonstrating clinical
cyst-LT2, is also expressed on vascular endothelium, blood benet [1719]. Their additive eect to the bronchodila-
eosinophils, mast cells, and lung macrophages, and could tion achieved with high doses of inhaled -agonist indicates
mediate activation of these cells [7] Zarlukast and monte- that they may have a place in the treatment of acute severe
lukast are potent leukotriene receptor antagonists available asthma. Intravenous administration of montelukast in acute
in most countries, while pranlukast is currently available severe asthma provided additional benet to the receovery
only in Japan and Korea. There is only one anti-leukotriene when given together with standard therapy [20]. Clinical
a 5-lipoxygenase inhibitor, zileuton, which is available in the benet has also been demonstrated with the addition of
USA. anti-leukotrienes to the patients with poor asthma control
who are already taking high doses of inhaled corticosteroids
Clinical studies in asthma [21]. Anti-leukotrienes may also reduce to a small extent
the doses of inhaled corticosteroids required for asthma
Leukotriene receptor antagonists in clinical use inhibit the control [22]. Anti-leukotrienes may reduce the risk of acute
bronchoconstrictor eects of inhaled cys-LTs. For example, severe asthma exacerbations [23, 24].

656
Mediator Antagonists 52
The eects of anti-leukotrienes have been compared Safety
with these inhaled corticosteroids [25]. In a recent meta-
analysis, inhaled corticosteroids (250400 g beclometha- At the recommended doses, all leukotriene receptor antag-
sone dipropionate equivalent per day) was found to provide onists have not resulted in nonrespiratory symptoms nor
better improvements in lung function and quality of life, as laboratory abnormalities when compared with placebo-
well as reduction in symptoms, night awakenings, and need treated groups. However, with zileuton, asymptomatic
for rescue -agonist. The rate of asthma exacerbations was 3-fold or greater increases in serum alanine-aminotrans-
similar when the anti-leukotrienes were compared with ferase levels were found in 4.6% of patients receiving zileu-
inhaled corticosteroids. The benet of adding leukotriene- ton at the standard dose of 600 mg four times per day,
receptor antagonist with inhaled corticosteroids has been compared with 1% of patients receiving standard asthma
evaluated. Addition of pranlukast to half the usual dose treatment together with placebo. Controlled-release zileu-
of inhaled corticosteroids in patients with moderate-to- ton at a dose of 1200 mg twice daily was shown to have e-
severe persistent asthma led to maintained control, while cacy within the range of zileuton 600 mg four times daily,
the placebo group demonstrated less improved asthma with similar side eect prole [40]. These elevations of
control [26]. Montelukast maintained control of asthma in liver enzymes usually occurred during the rst 3 months of
patients in whom removal of inhaled corticosteroids caused therapy, with sometimes normalization of the values despite
worsening of asthma [26]. When compared to long-acting continuation of treatment.
-agonists, montelukast was less superior for preventing A rare syndrome of ChurgStrauss, marked by circu-
exacerbations and for improving lung function, symptoms, lating eosinophilia and evidence of tissue or organ inltra-
and the use of rescue -agonists [27, 28]. Combination tion and vasculitis by eosinophils in association with heart
of a leukotriene receptor antagonists with long-acting failure, cutaneous or gastrointestinal involvement, and
-agonists is not advisable for the treatment of moderate peripheral neuropathy, has been associated with treatment
asthma [29]. with zarlukast and montelukast. Most patients devel-
Anti-leukotrienes are eective in blocking aspirin- oping ChurgStrauss syndrome have previously received
induced asthmatic responses [30] and may be particularly oral glucocorticoid therapy or high-dose inhaled corticos-
indicated in patients with aspirin-sensitive asthma. In addi- teroid therapy to control their asthma. This may be due to
tion, anti-leukotrienes are particularly eective in inhibit- unmasking of vasculitis of ChurgStrauss syndrome, as cor-
ing exercise-induced asthma, [10, 31, 32] without the loss ticosteroids are tapered with the introduction of leukotriene
of protection with prolonged usage. In addition, leukotriene receptor antagonist therapy [41].
inhibitors improve concomitant symptoms of allergic rhini-
tis [33, 34].
In United States, zileuton, zarlukast, and montelu- Role in COPD
kast have been approved as a rst-line choice for the trt-
eatment of asthma, while in Europe, montelukast and Small bronchodilator eects of zarlukast has been observed
zarlukast have been approved as an add-on therapy when in some patients with COPD [42, 43] Because LTB4 may
treatment with inhaled corticosteroids has failed, and for play a role in the recruitment of neutrophils in COPD, the
the prophylaxis of exercise-induced asthma. In the GINA use of 5-LO inhibitors may bring some additional eects.
2006 guidelines, anti-leukotrienes are listed as a less pre- In a small study of the leukotriene synthesis inhibitor, a
ferred initial treatment compared to inhaled corticosteroids 5-lipoxygenase activating protein inhibitor, BAYx1005,
and a less preferred option as an add-on therapy compared a small reduction in sputum LTB4 was observed with no
to long-acting 2-agonists [35]. clinical benet [44]. No studies of zileuton in COPD have
been reported yet.

Anti-inflammatory effects
Cys-LTs can induce airway eosinophilia in patients with
PROSTAGLANDIN AND CRTH2 INHIBITORS
asthma, and conversely, leukotriene-receptor antagonists or
synthesis inhibitors reduce blood and airway eosinophilia Prostaglandin D2 (PGD2) is produced by activated mast
associated with poorly controlled asthma, [23] and air- cells and is a relatively selective marker for mast cells, and
way inammation associated with allergen-induced airway two G-protein-coupled receptors, DP1 and DP2 (also
responses [36]. Therefore, anti-leukotrienes can be consid- known as CRTH2), mediate many of its eects. However,
ered as an anti-inammatory therapy for asthma, but this it is quite likely that the PGD2-bronchoconstrictor eect
remains to be conrmed in the clinical situation. In a study is mediated via the TP receptor, since the activation of
of the combination therapy of montelukast with inhaled DP1 receptor in airway smooth muscle cells leads to bron-
corticosteroid, there was no eect of montelukast on spu- chodilation. DP2-receptors play an important role in Th2
tum eosinophil counts [37]. Although there is cellular and lymphocyte recruitment and activation [45] (Fig. 52.2).
animal data to support a role for cys-LTs in airway smooth Ramatroban is an eective TP antagonist which has been
muscle hyperplasia and airway brosis [38, 39], there is as approved as a treatment for perennial rhinitis. It reduces
yet no data of the eect of leukotriene inhibitors on these bronchial hyperresponsiveness to methacholine in asth-
parameters in asthma. matics [46]. Ramatroban also has a partial action as an

657
Asthma and COPD: Basic Mechanisms and Clinical Management

Arachidonic acid

Cyclooxygenases (COX1, COX2)

PGD2

Receptors TP DP1 DP2/CRTH2


FIG. 52.2 Prostaglandin D2 (PGD2) effects. PGD2
acts on three distinct prostanoid receptiors:
the thromboxane receptor (TP) mediates
Bronchoconstriction Bronchodilatation Chemotaxis bronchonstriction, DP1-receptor mediates
Vasodilatation Th2 cells vasodilatation, and bronchodilatation and DP2-
Eosinophils receptor (also know as CRTH2) is chemotactic for
Basophils Th2 cells, eosinophils, and basophils.

antagonist at the DP2 receptor as shown by its inhibitory a small degree of bronchodilatation in asthmatic patients,
eect on PGD2-stimulated eosinophil chemotaxis [47]. indicating a certain degree of histamine tone, presum-
More selective and potent DP2 antagonists have now been ably due to the basal release of histamine from activated
developed and are undergoing trials in asthma and allergic mast cells [57]. Chronic administration of terfenadine has a
rhinitis [48]. small clinical eect in mild allergic asthmatic patients, [58]
but is far less eective that other anti-asthma therapies. H1-
receptor antagonists have not been found to be useful in
more severe asthmatic patients [48]. The new generation
THROMBOXANE INHIBITORS anti-histamines, cetirizine and astemizole, have some ben-
ecial eects in asthma, [59, 60] that may be unrelated to
their H1-antagonist eects [61].
Although thromboxane production is increased in asthma
H2-antagonists, such as cimetidine and ranitidine,
and thromboxane analogs are potent bronchoconstrictors
may be contraindicated in asthma on theoretical grounds,
in asthmatic patients [49], there is no convincing evidence
if H2-receptors are important in counteracting the bron-
that thromboxane receptor (TP) antagonists or thrombox-
choconstrictor eect of histamine. In clinical practice,
ane synthase inhibitors are eective in asthma [50]. A selec-
however, there is no evidence that H2-antagonists have
tive TP antagonist, GR32191, caused a modest inhibition
of early phase bronchoconstriction due to allergen but was any deleterious eect in asthma. H3-receptor agonists may
have some theoretical benet in asthma, since they may
not clinically eective in improving lung function or reduc-
modulate cholinergic bronchoconstriction and inhibit neu-
ing symptoms in mild asthma [51]. However, a thrombox-
rogenic inammation. Although (R)--methylhistamine
ane synthase inhibitor (ozagrel) and a receptor antagonist
relaxes rodent peripheral airways in vitro, it has no eect
(seratrodast) are used in the treatment of asthma in Japan,
when given by inhalation on airway caliber or metabisute-
although their eects in asthma are small [5254].
induced bronchoconstriction in asthmatic patients, indicat-
There is no evidence for a role for TP or DP receptors
ing that a useful clinical eect is unlikely [62].
in COPD.
Histamine H4-receptors are expressed on eosinophils,
T- cells, dendritic cells, basophils and mast cells, medi-
ate mast cell, eosinophil and dendritic cell chemotaxis, and
ANTI-HISTAMINES modulate cytokine production from dendritic cells and T-
cells, indicating that blockade of histamine H4-receptors
may lead to anti-allergic and anti-inammatory eects.
Histamine mediates most of its eects on airway function Several histamine H4-receptor antagonists are now avail-
via H1-receptors. Non-sedating potent H1-receptor antago- able but remain to be tested in allergic asthma or rhinitis
nists, such as terfenadine, fexafenadine, loratadine, deslorat- [63]. Antagonists that block both histamine H1- and H4-
adine, ebastine, and astemizole, have useful clinical eects receptors may be an eective combination. Anti-histamines
in allergic rhinitis, but they are far from eective in asth- have a useful eect in the treatment of rhinitis, and par-
matic patients [55]. The eects of anti-histamines are small ticularly the rhinorrhea. As a large proportion of patients
and clinically insignicant. Terfenadine causes about 50% with asthma have concomitant rhinitis, an H1-antagonist
inhibition of the immediate response to allergen, but has may help the overall management of asthma [64]. While
no eect on the late response [56]. Anti-histamines cause H1-receptor antagonists alone may be ineective, some

658
Mediator Antagonists 52
studies suggest that they may have some ecacy in combi-
nation with other antagonists. Thus, an H1-receptor antago-
BRADYKININ ANTAGONISTS
nist when added to an anti-leukotriene was able to inhibit
the early and late responses to allergen more eectively than There is evidence for the involvement of bradykinin in
the anti-leukotriene alone [65, 66], but as yet there has been asthma, in particular a role in sensitizing and activating air-
no studies of combination mediator antagonists in asthma. way sensory nerves. However, a peptide bradykinin antago-
There is no evidence that anti-histamines have any nist [D-Arg0,Hyp3,D-Phe7]bradykinin (NPC567) was
role in the treatment of COPD. unable to inhibit the eect of bradykinin on nasal secretions,
even when given at the same time as bradykinin, [76] pre-
sumably because of rapid local metabolism. There has been
other more stable antagonists developed [77, 78]. Icatibant
SEROTONIN ANTAGONISTS (HOE 140, D-Arg[Hyp3,Thi6,D-Tic7,Oic8] bradykinin is
a potent selective B2-receptor antagonist, [79] which has a
The evidence for involvement of serotonin in asthma is long duration of action in vivo since it is resistant to enzy-
weak. There is no evidence that serotonin is a direct con- matic degradation. This antagonist is potent in inhibiting
strictor in human airways, and it is not stored in and the bronchoconstrictor and microvascular leakage response
released from human mast cells, as in rodents. Serotonin to bradykinin [80] and the eect of bradykinin on airway
receptor antagonists have been studied experimentally in sensory nerves [81]. Nasal application of icatibant reduces
asthmatic patients. Ketanserin, which antagonizes 5HT2 the nasal blockage induced by allergen in patients with
receptors and blocks the bronchoconstrictor eects of allergic rhinitis [82]. In a clinical study of nebulized icati-
serotonin in animals, has no eect on airway function in bant in asthma, there was a small improvement in airway
asthmatic patients, but there is a small inhibitory eect on function after 4 weeks without an improvement in asthma
methacholine-induced bronchoconstriction [67]. Inhaled symptoms [83]. WIN 64338 is a nonpeptide B2-receptor
ketanserin has no eect on histamine-induced bronchoc- antagonist that blocked the bronchoconstrictor action of
onstriction but has a small inhibitory eect on adenosine- bradykinin in airway smooth muscle in vitro [84] and more
induced bronchoconstriction, indicating a possible action potent nonpeptide antagonists, such as FR167344 have now
on mast cells [68]. Tianeptine, which enhances serotonin been developed that have clinical potential [85].
uptake by platelets, lowers the elevated plasma serotonin
levels reported in patients with asthma and is associated
with a reduction in asthmatic symptoms [69].
TACHYKININ ANTAGONISTS

PAF ANTAGONISTS Tachykinins interacts with the tachykinin NK1-, NK2-,


and NK3-receptors which exist in the airways, particularly
on human bronchial glands, bronchial vessels, and bron-
Although PAF mimics many of the pathophysiological chial smooth muscle. Inammatory cells also express NK1-
features of asthma including induction of airway hyperre- receptors. NK1- and NK2-receptor antagonists have been
sponsiveness, PAF antagonists have proved to be very dis- shown to inhibit allergen-induced bronchial hyperrespon-
appointing in asthma therapy. siveness and inammation in various animal models. Potent
Apafant (WEB 2086) potently inhibits the eects of NK1- and NK2-receptor antagonists have been recently
inhaled PAF on airway function [70], but had no eect on available but have not shown any eects in asthma [86].
the early or late responses to allergen or on airway hyperre- Thus, an NK2-receptor antagonist, SR48968, that caused a
sponsiveness in patients with mild asthma [71]. A 3-month signicant inhibition of neurokinin-A bronchoconstriction
study of oral apafant in patients with symptomatic moder- by 35 fold shift did not aect baseline airway caliber or
ate asthma failed to show any eect on lung function, symp- bronchial responsiveness to adenosine in asthma [87]. Dual
toms or on rescue 2-agonist use [72]. Similarly, another NK1/NK2 and triple NK1/NK2/NK3 receptor antagonists
PAF antagonist, modipafant (UK80067) had no ben- are available with greater potencies, but it is unlikely that
ecial eects in moderately severe asthmatics [73]. Another there will be trials in asthma. There has been no studies of
potent and long-acting PAF receptor antagonist, foropafant tachykinin antagonists in COPD.
(SR27417A), was eective in inhibiting systemic, cellular,
and pulmonary eects of PAF challenge in patients with
mild bronchial asthma [74]. This antagonist produced a mod-
est but signicant reduction in the magnitude of the allergen-
induced late response, but no eect on the early response,
ADENOSINE AGONISTS AND ANTAGONISTS
allergen-induced hyperresponsiveness, or on baseline lung
function [75]. These clinical studies indicate that PAF plays Adenosine is released following allergen exposure and exer-
little or no part in human allergic asthma, despite convinc- cise of sensitized asthmatics and is a bronchoconstrictor in
ing data in animal models. There is no evidence that PAF is asthmatics through the activation of mast cells. Adenosine
involved in COPD, and no studies with PAF antagonists in may also be involved in allergic inammation and remod-
this disease have been reported. eling. It interacts with G-protein-coupled receptors,

659
Asthma and COPD: Basic Mechanisms and Clinical Management

adenosine A1-, A2A-, A2B- and A3-receptors that are 11. Hui KP, Barnes NC. Lung function improvement in asthma with a
present on inammatory and structural cells of the asth- cystemyl-leukotriene receptor antagonist. Lancet 337: 106263, 1991.
matic airway. Various agonists and antagonists of these 12. Reiss TF, Sorkness CA, Stricker W, Botto A, Busse WW, Kundu S
et al. Eects of montelukast (MK-0476); a potent cysteinyl leukotriene
receptors have been developed as potential therapies for
receptor antagonist, on bronchodilation in asthmatic subjects treated
a variety of cardiac and pulmonary conditions, includ- with and without inhaled corticosteroids. Thorax 52: 4548, 1997.
ing asthma and COPD [88]. Activation of A2A-receptors 13. Spector SL, Smith LJ, Glass M. Eects of six weeks of therapy with
has been shown to have anti-inammatory eects in ani- oral doses of ICI 204,219, a leukotriene D4 receptor antagonist, in
mal models, and a selective A2A- agonist has been shown subjects with bronchial asthma. Am J Respir Crit Care Med 150: 618
to reduce the number of eosinophils and neutrophils in 23, 1994.
allergen-induced inammation. However, two agonists of 14. Altman LC, Munk Z, Seltzer J, Noonan N, Shingo S, Zhang J et al.
the A2A-receptor, GW328267 and UK-432,097, have been A placebo-controlled, dose-ranging study of montelukast, a cysteinyl
tested in asthma. GW328267 had no eect on allergen- leukotriene-receptor antagonist. Montelukast Asthma Study Group.
induced early or late phase response, and UK-432097 had J Allergy Clin Immunol 102: 5056, 1998.
15. Barnes NC, Pujet JC. Pranlukast, a novel leukotriene receptor antago-
no eect on lung function or bronchial hyperreactivity of
nist: Results of the rst European, placebo controlled, multicentre clin-
mild asthma [89]. ical study in asthma. Thorax 52: 52327, 1997.
Adenosine has been shown to act through the A2B- 16. Israel E, Rubin P, Kemp JP, Grossman J, Pierson W, Siegel SC et al.
receptors to enhance various inammatory responses seen The eect of inhibition of 5-lipoxygenase by zileuton in mild- to-
in asthma. An antagonist of A2B-receptors, CVT-6883, has moderate asthma. Ann Intern Med 119: 105966, 1993.
been shown to inhibit the proinammatory cytokines and 17. Liu MC, Dube L, Lancaster J. and the Zileuton Study Group. Acute
probrotic eects found in adenosine deaminase-decient and chronic eects of a 5-lipoxygenase inhibitor in asthma: A 6 month
mice [90]. It is under development as an oral treatment randomized multicentre trial. J Allergy Clin Immunol 98: 85971, 1996.
for asthma. 18. Calhoun WJ. Summary of clinical trials with zarlukast. Am J Respir
Crit Care Med 157: S23845, 1998.
19. Blake KV. Montelukast: Data from clinical trials in the management of
asthma. Ann Pharmacother 33: 1299314, 1999.
20. Camargo CA Jr., Smithline HA, Malice MP, Green SA, Reiss TF.
References A randomized controlled trial of intravenous montelukast in acute
asthma. Am J Respir Crit Care Med 167: 52833, 2003.
1. Berger W, De Chandt MT, Cairns CB. Zileuton: Clinical implications 21. Virchow CJ, Prasse A, Naya I, Summerton L, harris A. Zarlukast
of 5-Lipoxygenase inhibition in severe airway disease. Int J Clin Pract improves asthma control in patients receiving high-dose inhaled corti-
61: 66376, 2007. costeroids. Am J Respir Crit Care Med 162: 57885, 2000.
2. Wenzel SE, Trudeau JB, Kaminsky DA, Cohn J, Martin RJ, Westcott JY. 22. Lofdahl C-G, Reiss T, Le JA, Israel E, Noonan M, Finn AF et al.
Eect of 5-lipoxygenase inhibition on bronchoconstriction and airway Randomized, placebo controlled trial of eect of a leukotriene receptor
inammation in nocturnal asthma. Am J Respir Crit Care Med 152: antagonist, montelukast, on tapering inhaled corticosteroids in asth-
897905, 1995. matic patients. B MJ 319: 8790, 1999.
3. Kane GC, Pollice M, Kim CJ, Cohn J, Dworski RT, Murray JJ et al. 23. Laviolette M, Malmstrom K, Lu S, Chervinsky P, Pujet JC, Peszek I
A controlled trial of the eect of the 5-lipoxygenase inhibitor, zileu- et al. Montelukast added to inhaled beclomethasone in treatment of
ton, on lung inammation produced by segmental antigen challenge in asthma. Montelukast/Beclomethasone Additivity Group. Am J Respir
human beings. J Allergy Clin Immunol 97: 64654, 1996. Crit Care Med 160: 186268, 1999.
4. Wenzel S, Busse W, Calhoun W, Panettieri R Jr., Peters-Golden M, 24. Barnes NC, Miller CJ. Eect of leukotriene receptor antagonist ther-
Dube L et al. The safety and ecacy of zileuton controlled-release tab- apy on the risk of asthma exacerbations in patients with mild to mod-
lets as adjunctive therapy to usual care in the treatment of moderate erate asthma: An integrated analysis of zarlukast trials. Thorax 55:
persistent asthma: A 6-month randomized controlled study. J Asthma 47883, 2000.
44: 30510, 2007. 25. Ducharme FM, Hicks GC. Anti-leukotriene agents compared to
5. Lynch KR, ONeill GP, Liu Q, Im DS, Sawyer N, Metters KM et al. inhaled corticosteroids in the management of recurrent and/or chronic
Characterization of the human cysteinyl leukotriene CysLT1 receptor. asthma (Cochrane review) [In Process Citation]. Cochrane Database
Nature 399: 78993, 1999. Syst Rev(3), 2000., CD002314.CD002314.
6. Crooke ST, Sarau HM, Saussy D, Winkler J, Foley J. Signal transduc- 26. Tamaoki J, Kondo M, Sakai N, Nakata J, Takemura H, Nagai A et al.
tion processes for the LTD4 receptor. Adv Prost Thromb Leuko Res 20: Leukotriene antagonist prevents exacerbations of asthma during reduc-
12737, 1990. tion of high-dose inhaled corticosteroid. Am J Respir Crit Care Med
7. Kanaoka Y, Boyce JA. Cysteinyl leukotrienes and their receptors: 155: 123540, 1997.
Cellular distribution and function in immune and inammatory 27. Ducharme FM, Lasserson TJ, Cates CJ. Long-acting beta2-agonists
responses. J Immunol 173: 150310, 2004. versus anti-leukotrienes as add-on therapy to inhaled corticosteroids
8. Smith LJ, Geller S, Ebright L, Glass M, Thyrum PT. Inhibition of for chronic asthma. Cochrane Database Syst Rev 43, 2006, CD003137.
leukotriene D4-induced bronchoconstriction in normal subjects by 28. Peters SP, Anthonisen N, Castro M, Holbrook JT, Irvin CG, Smith LJ
the oral LTD4 receptor antagonist ICI 204,2. Am Rev Respir Dis 141: et al. Randomized comparison of strategies for reducing treatment in
98892, 1990. mild persistent asthma. N Engl J Med 356: 202739, 2007.
9. Taylor IK, OShaughnessy KM, Fuller RW, Dollery CT. Eect of 29. Deykin A, Wechsler ME, Boushey HA, Chinchilli VM, Kunselman SJ,
cysteinyl-leukotriene receptor antagonist ICI 204-219 on allergen- Craig TJ et al. Combination therapy with a long-acting beta-agonist
induced bronchoconstriction and airway hyperreactivitiy in atopic sub- and a leukotriene antagonist in moderate asthma. Am J Respir Crit Care
jects. Lancet 337: 69094, 1991. Med 175: 22834, 2007.
10. Finnerty JP, Wood Baker R, Thomson H, Holgate ST. Role of leuko- 30. Israel E, Fischer AR, Rosenberg MA, Lilly CM, Callery JC, Shapiro J
trienes in exercise-induced asthma. Inhibitory eect of ICI 204219, et al. The pivotal role of 5-lipoxygenase products in the reaction of
a potent leukotriene D4 receptor antagonist. Am Rev Respir Dis 145: aspirin-sensitive asthmatics to aspirin. Am Rev Respir Dis 148: 1447
74649, 1992. 51, 1993.

660
Mediator Antagonists 52
31. Le JA, Busse WW, Pearlman D, Bronsky EA, Kemp J, Hendeles L 49. Saroea HG, Inman MD, OByrne PM. U46619-induced bronchocon-
et al. Montelukast, a leukotriene-receptor antagonist, for the treatment striction in asthmatic subjects is mediated by acetylcholine release. Am
of mild asthma and exercise-induced bronchoconstriction. N Engl J J Respir Crit Care Med 151: 32124, 1995.
Med 339: 14752, 1998, [see comments] 50. OByrne PM, Fuller RW. The role of thromboxane A2 in the patho-
32. Manning PJ, Watson RM, Margolskee DJ, Williams VC, Schwartz JI, genesis of airway hyperresponsiveness. Eur Respir J 2: 78286, 1989.
OByrne PM. Inhibition of exercise-induced bronchoconstriction by 51. Coleman RA. GR32191 and the role of thromboxane A2 in
MK-751, a potent leukotriene-D4 receptor antagonist. N Engl J Med asthmapreclinical and clinical ndings. Agents Actions 34(Suppl): 211
323: 173639, 1990. 20, 1991.
33. Donnelly AL, Glass M, Minkwitz MC, Casale TB. The leukotriene 52. Manning PJ, Stevens WH, Cockcroft DW, OByrne PM. The role of
D4-receptor antagonist, ICI 204,219, relieves symptoms of acute thromboxane in allergen-induced asthmatic responses. Eur Respir J 4:
seasonal allergic rhinitis. Am J Respir Crit Care Med 151: 173439, 66772, 1991.
1995. 53. Obase Y, Shimoda T, Matsuo N, Matsuse H, Asai S, Kohno S. Eects
34. Patel P, Philip G, Yang W, Call R, Horak F, Laforce C et al. of cysteinyl-leukotriene receptor antagonist, thromboxane A2 recep-
Randomized, double-blind, placebo-controlled study of montelukast tor antagonist, and thromboxane A2 synthetase inhibitor on anti-
for treating perennial allergic rhinitis. Ann Allergy Asthma Immunol 95: gen-induced bronchoconstriction in patients with asthma. Chest 114:
55157, 2005. 102832, 1998.
35. Bateman ED, Hurd SS, Barnes PJ, Bousquet J, Drazen JM, Fitzgerald M 54. Fukuoka T, Miyake S, Umino T, Inase N, Tojo N, Yoshizawa Y. The
et al. Global strategy for asthma management and prevention: GINA eect of seratrodast on eosinophil cationic protein and symptoms in
executive summary. Eur Respir J 31: 14378, 2008. asthmatics. J Asthma 40: 25764, 2003.
36. Calhoun WJ, Lavins BJ, Minkwitz MC, Evans R, Gleich GJ, 55. Van Ganse E, Kaufman L, Derde MP, Yernault JC, Delaunois L,
Cohn J. Eect of zarlukast (Accolate) on cellular mediators of Vincken W. Eects of antihistamines in adult asthma: A meta-analysis
inammation: Bronchoalveolar lavage uid ndings after segmen- of clinical trials. Eur Respir J 10: 221624, 1997.
tal antigen challenge. Am J Respir Crit Care Med 157: 138189, 56. Hamid M, Raerty P, Holgate ST. The inhibitory eect of terfenadine
1998. and urbiprofen on early and late-phase bronchoconstriction following
37. Barnes N, Laviolette M, Allen D, Flood-Page P, Hargreave F, Corris P allergen challenge in atopic asthma. Clin Exp Allergy 20: 26167, 1990.
et al. Eects of montelukast compared to double dose budesonide on 57. Eiser N, Mills J, Snashall P, Guz A. The role of histamine receptors in
airway inammation and asthma control. Respir Med 101: 165258, asthma. Clin Sci 60: 36370, 1981.
2007. 58. Taytard A, Beaumont D, Pujet JC, Sapene M, Lewis PJ. Treatment of
38. Salmon M, Walsh DA, Huang TJ, Barnes PJ, Leonard TB, Hay DW bronchial asthma with terfenadine: A randomized controlled trial. Br J
et al. Involvement of cysteinyl leukotrienes in airway smooth mus- Clin Pharmacol 24: 74346, 1987.
cle cell DNA synthesis after repeated allergen exposure in sensitized 59. Spector SL, Nicodemus CF, Corren J, Schanker HM, Rachelefsky GS,
Brown Norway rats. Br J Pharmacol 127: 115158, 1999. Katz RM et al. Comparison of the bronchodilatory eects of cetirizine,
39. Henderson WR, Chiang GK, Tien YT, Chi EY. Reversal of allergen- albuterol, and both together versus placebo in patients with mild-to-
induced airway remodeling by CysLT1 receptor blockade. Am J Respir moderate asthma. J Allergy Clin Immunol 96: 17481, 1995.
Crit Care Med 173: 71828, 2006. 60. Busse WW, Middleton E, Storms W, Dockhorn RJ, Chu TJ, Grossman J
40. Wenzel S, Busse W, Calhoun W, Panettieri R, Peters-Golden M, et al. Corticosteroid-sparing eect of azelastine in the management of
Dube L et al. The safety and ecacy of zileuton controlled-release tab- bronchial asthma. Am J Respir Crit Care Med 153: 12227, 1996.
lets as adjunctive therapy to usual care in the treatment of moderate 61. Walsh GM. The anti-inammatory eects of cetirizine. Clin Exp
persistent asthma: A 6-month randomized controlled study. J Asthma Allergy 24: 8185, 1994.
44: 30510, 2007. 62. OConnor BJ, Lecomte JM, Barnes PJ. Eect of an inhaled histamine
41. Wechsler ME, Garpestad E, Flier SR, Kocher O, Weiland DA, H3-receptor agonist on airway responses to sodium metabisulphite in
Polito AJ et al. Pulmonary inltrates, eosinophilia, and cardiomyopathy asthma. Br J Clin Pharmacol 35: 5557, 1993.
following corticosteroid withdrawal in patients with asthma receiving 63. Fung-Leung WP, Thurmond RL, Ling P, Karlsson L. Histamine H4
zarlukast. JAMA 279: 45557, 1998. receptor antagonists: The new antihistamines?. Curr Opin Investig
42. Cazzola M, Santus P, Di Marco F, Boveri B, Castagna F, Carlucci P Drugs 5: 117483, 2004.
et al. Bronchodilator eect of an inhaled combination therapy with sal- 64. Simons FE. Is antihistamine (H1-receptor antagonist) therapy useful
meterol  uticasone and formoterol  budesonide in patients with in clinical asthma? Clin Exp Allergy 29(Suppl 3): 98104, 1999.
COPD. Respir Med 97: 45357, 2003. 65. Roquet A, Dahlen B, Kumlin M, Ihre E, Anstren G, Binks S et al.
43. Nannini LJ, Flores DM. Bronchodilator eect of zarlukast in subjects Combined antagonism of leukotrienes and histamine produces pre-
with chronic obstructive pulmonary disease. Pulm Pharmacol Ther 16: dominant inhibition of allergen-induced early and late phase airway
30711, 2003. obstruction in asthmatics. Am J Respir Crit Care Med 155: 185663,
44. Gompertz S, Stockley RA. A randomized, placebo-controlled trial of 1997.
a leukotriene synthesis inhibitor in patients with COPD. Chest 122: 66. Meltzer EO, Malmstrom K, Lu S, Prenner BM, Wei LX, Weinstein SF
28994, 2002. et al. Concomitant montelukast and loratadine as treatment for sea-
45. Kostenis E, Ulven T. Emerging roles of DP and CRTH2 in allergic sonal allergic rhinitis: A randomized, placebo-controlled clinical trial.
inammation. Trends Mol Med 12: 14858, 2006. J Allergy Clin Immunol 105: 91722, 2000.
46. Aizawa H, Shigyo M, Nogami H, Hirose T, Hara N. BAY u3405, a 67. Cazzola M, Assogna G, Lucchetti G, Cicchitto G, DAmato G. Eect
thromboxane A2 antagonist, reduces bronchial hyperresponsiveness in of ketanserin, a new blocking agent of the 5-HT2 receptor, on airway
asthmatics. Chest 109: 33842, 1996. responsiveness in asthma. Allergy 45: 15153, 1990.
47. Mimura H, Ikemura T, Kotera O, Sawada M, Tashiro S, Fuse E et al. 68. Cazzola M, Matera MG, Santangelo G, Assogna G, DAmato G,
Inhibitory eect of the 4-aminotetrahydroquinoline derivatives, selec- Rossi F et al. Eect of the selective 5-HT2 antagonist ketanserin
tive chemoattractant receptor-homologous molecule expressed on T on adenosine-induced bronchoconstriction in asthmatic subjects.
helper 2 cell antagonists, on eosinophil migration induced by prostag- Immunopharmacology 23: 2128, 1992.
landin D2. J Pharmacol Exp Ther 314: 24451, 2005. 69. Lechin F, van der DB, Orozco B, Jara H, Rada I, Lechin ME et al.
48. Pettipher R, Hansel TT, Armer R. Antagonism of the prostaglandin The serotonin uptake-enhancing drug tianeptine suppresses asthmatic
D2 receptors DP1 and CRTH2 as an approach to treat allergic dis- symptoms in children: A double-blind, crossover, placebo-controlled
eases. Nat Rev Drug Discov 6: 31325, 2007. study. J Clin Pharmacol 38: 91825, 1998.

661
Asthma and COPD: Basic Mechanisms and Clinical Management

70. Adamus WS, Heuer H, Meade CJ, Frey G, Brecht HM. Inhibitory 80. Wirth KJ, Gehring D, Scholkens BA. Eect of Hoe 140 on
eect of oral WEB 2086, a novel selective PAF-acether antagonist, on bradykinin-induced bronchoconstriction in anesthetized guinea pigs.
ex vivo platelet aggregation. Eur J Clin Pharmacol 35: 23740, 1988. Am Rev Resp Dis 148: 7026, 1993.
71. Freitag A, Watson RM, Matsos G, Eastwood C, OByrne PM. The 81. Miura M, Belvisi MG, Barnes PJ. Eect of bradykinin on airway neu-
eect of an oral platelet activating factor antagonist, WEB 2086, on ral responses in vitro. J Appl Physiol 73: 153741, 1992.
allergen induced asthmatic responses. Thorax 48: 59498, 1993. 82. Austin CA, Foreman JC, Scadding GK. Reduction by HOE140, the
72. Spence DP, Johnston SL, Calverley PM, Dhillon P, Higgins C, B2 kinin receptor antagonist, of antigen-induced nasal blockage. Br J
Ramhamadany E et al. The eect of the orally active platelet- Pharmacol 111: 96971, 1994.
activating factor antagonist WEB 2086 in the treatment of asthma. Am 83. Akbary AM, Wirth KJ, Scholkens BA. Ecacy and tolerability of
J RespirCrit Care Med 149: 114248, 1994. Icatibant (Hoe 140) in patients with moderately severe chronic bron-
73. Kuitert LM, Hui KP, Uthayarkumar S, Burke W, Newland AC, Uden S chial asthma. Immunopharmacology 33: 23842, 1996.
et al. Eect of a platelet activating factor (PAF) antagonist UK-74, 505 84. Scherrer D, Daeer L, Trilie A, Gies JP. Eects of WIN 64338, a
on allergen-induced early and late response. Am Rev Respir Dis 147: nonpeptide bradykinin B2 receptor antagonist, on guinea-pig trachea.
8286, 1993. Br J Pharmacol 115: 112728, 1995.
74. Gomez FP, Marrades RM, Iglesia R, Roca J, Barbera JA, Chung KF 85. Inamura N, Asano M, Hatori C, Sawai H, Hirosumi J, Fujiwara T
et al. Gas exchange response to a PAF receptor antagonist, SR 27417A, et al. Pharmacological characterization of a novel, orally active, non-
in acute asthma: A pilot study. Eur Respir J 14: 62226, 1999. peptide bradykinin B2 receptor antagonist, FR1673. Eur J Pharmacol
75. Evans DJ, Barnes PJ, Cluzel M, OConnor BJ. Eects of a potent 333: 7986, 1997.
platelet-activating factor antagonist, SR27417A, on allergen-induced 86. De Swert KO, Joos GF. Extending the understanding of sensory neu-
asthmatic responses. Am J Respir Crit Care Med 156: 1116, 1997. ropeptides. Eur J Pharmacol 533: 17181, 2006.
76. Pongracic JA, Naclerio RM, Reynolds CJ, Proud D. A competitive 87. Kraan J, Vink-Klooster H, Postma DS. The NK-2 receptor antagonist
kinin receptor antagonist, [DArg0, Hyp3, DPhe7]- bradykinin, does SR 48968C does not improve adenosine hyperresponsiveness and air-
not aect the response to nasal provocation with bradykinin. Br J Clin way obstruction in allergic asthma. Clin Exp Allergy 31: 27478, 2001.
Pharmacol 31: 28794, 1991. 88. Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nat
77. Abraham WM, Scuri M, Farmer SG. Peptide and non-peptide Rev Drug Discov 5: 24764, 2006.
bradykinin receptor antagonists: Role in allergic airway disease. Eur J 89. Polosa R, Holgate ST. Adenosine receptors as promising therapeutic
Pharmacol 533: 21521, 2006. targets for drug development in chronic airway inammation. Curr
78. Abraham WM, Scuri M, Farmer SG. Peptide and non-peptide Drug Targets 7: 699706, 2006.
bradykinin receptor antagonists: Role in allergic airway disease. Eur J 90. Sun CX, Zhong H, Mohsenin A, Morschl E, Chunn JL, Molina JG et al.
Pharmacol 533: 21521, 2006. Role of A2B adenosine receptor signaling in adenosine-dependent pul-
79. Wirth K, Hock FJ, Albus V et al. HOE 140, a new potent and long- monary inammation and injury. J Clin Invest 116: 217382, 2006.
acting bradykinin antagonist; in vivo studies. Br J Pharmacol 102: 774
77, 1991.

662
Antibiotics
53 CHAPTER

and pathogenesis of exacerbations. In contrast,


INTRODUCTION such richness of data is unfortunately missing Sanjay Sethi
in the eld of clinical management of exacer-
Exacerbations of chronic obstructive pulmo- bations. Major modalities used to treat exac- Division of Pulmonary, Critical Care
nary disease (COPD) are a frequent cause for erbations include bronchodilators, systemic and Sleep Medicine, Department of
oce and emergency room visits as well as hos- corticosteroids, and antibiotics. Only for sys- Medicine, University of Buffalo, State
pitalizations. Exacerbations are intermittent temic corticosteroid use in severe exacerbations University of New York, and Veterans
episodes of increased respiratory symptoms and requiring emergency room care or hospitaliza- Affairs Western New York Health Care
worse pulmonary function that may be accom- tion is the adequate clinical trial data available. System, Buffalo, New York, USA
panied by fever and other constitutional symp- Adequately powered, well-designed clinical tri-
toms. These episodes contribute signicantly als studies for the other modalities used to treat
to the morbidity associated with COPD, and exacerbations are relatively few. Consequently,
in advanced disease, they are also the most fre- whether the use of antibiotics for exacerbations
quent cause of death [13]. The frequency of is appropriate is still debated and whether anti-
exacerbations varies widely between patients. biotic choice makes a dierence is even more
Though severity of airow obstruction is corre- controversial [9, 10].
lated with the frequency of exacerbations, other Principles that should guide appropriate
yet poorly understood factors predispose some use of antibiotics in exacerbations are listed in
patients to have more frequent exacerbations. Table 53.1. The comments in Table 53.1 illus-
Several investigators have reported that trate the several barriers that currently exist
exacerbations are important determinants of in fully applying these principles in everyday
the decline in health-related quality of life in practice. This chapter will describe the optimal
COPD [4, 5]. Following an exacerbation, acute approach to antibiotic treatment of exacerba-
symptoms usually subside over 23 weeks, how- tions recognizing these limitations. Such an
ever, quality of life indices takes several months approach relies upon an accurate diagnosis of an
to return to baseline. Furthermore, recurrent exacerbation, including judicious application of
exacerbations are clearly associated with a more diagnostic tests. This is followed by determining
rapid decline in quality of life in COPD [6]. the severity of an exacerbation, the probability
Based on studies performed by Fletcher and that it is bacterial and whether antibiotics are
Peto in the 1960s it was widely believed that indicated. If antibiotics are indicated, then a risk
exacerbations do not contribute to the decline stratication approach is described to choose an
in lung function (measured as FEV1). Recent appropriate antibiotic.
data has disputed these results and shown
that the frequency of exacerbations is associ-
ated with accelerated long-term decline in
lung function, in both mild COPD and more
advanced diseases [7, 8].
DIAGNOSIS OF ACUTE EXACERBATION
Undisputedly, exacerbations are a major
contributor to the morbidity, costs, and mortal- Clear, objective, universally accepted deni-
ity associated with COPD. Substantial progress tion of a disease or syndrome is a pre-requisite
has been made in understanding their etiology for its accurate diagnosis. Unfortunately, for

663
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 53.1 Principles of appropriate antibiotic use in exacerbations of COPD.

Principle Comment

Bacteria cause significant proportion of exacerbations 4050% of exacerbations are of bacterial origin

Bacterial exacerbations can be reliably distinguished from Sputum purulence is a useful marker but not always easily
nonbacterial episodes assessed or reliable

Placebo-controlled antibiotic trials have shown benefit of This is true for moderate-to-severe exacerbations, but not mild
antibiotics episodes

Antibiotics used are appropriate for the causative pathogen Sputum cultures are inaccurate and usually not performed,
therefore therapy is usually empiric

Antibiotic dose and duration are appropriate Shorter durations of 37 days appear to be as good as 1014
days

Antibiotic choice makes a difference in outcome Inadequate study methods have limited their ability to show
differences among antibiotics

Risk stratification is useful in choosing antibiotics Though not prospectively validated, it is widely advocated

exacerbations of COPD, the current denitions are impre- distinct etiology, pathogenesis and treatment, and therefore
cise, variable, and lack objective measures [11]. There are should be in the dierential diagnosis of an exacerbation
two widely used denitions of exacerbation. In 1987, in a rather than be included under the denition. In our clini-
large placebo-controlled trial in exacerbations of COPD, cal studies, we suspect an exacerbation when a patient with
Anthonisen and colleagues dened exacerbations based on COPD reports a minor increase (or new onset) of two or a
the presence of one or more of the three cardinal symp- major increase (or new onset) of one of the following res-
toms, including an increase or new onset of dyspnea, spu- piratory symptoms: dyspnea, cough, sputum production,
tum production, and sputum purulence [12]. When only sputum tenacity, sputum purulence [13]. The increase in
one cardinal symptom is present, then one or more support- symptoms should be of at least 24 h duration and should
ing symptoms or signs are required to make the diagnosis, be of greater intensity than their normal day-to-day vari-
including an upper respiratory tract infection in the past 5 ability. Furthermore, as described above, clinical evaluation
days, wheezing, cough, fever without an obvious source, or a should exclude other clinical entities that could present in a
20% increase in the respiratory rate or heart rate above base- similar manner.
line. Though simple and clinically useful, this denition does
have several limitations. It is narrow in its scope, and several
important symptoms of exacerbation such as cough, chest
congestion, chest tightness, fatigue, and sleep disturbance are
not included. It is subjective, however, that limitation applies
DETERMINATION OF SEVERITY
to all denitions of exacerbations as reliable objective meas-
ures of exacerbations are currently unavailable. The decision to treat exacerbations is often based on sever-
A more recent and also commonly used denition ity, with antibiotic treatment recommended for moderate
came from a consensus panel that dened an exacerbation to severe exacerbations. However, currently our determina-
as an acute sustained worsening of the patients condition tion of severity of exacerbations is imprecise, and like the
from stable state, beyond day-to-day variability and which diagnosis, is determined in a variable manner among stud-
requires additional treatment [11]. This denition though ies. The severity of an exacerbation is a complicated concept,
more inclusive of symptoms of an exacerbation, is not spe- determined by at least two factors, the severity of the under-
cic with regard to the nature and duration of symptoms. It lying COPD and the acute change induced by the exacerba-
lacks objective measures. Longitudinal cohort studies with tion. A severe exacerbation may therefore be assessed when
daily recording of symptoms have revealed that a signicant a patient with very severe underlying COPD has a relatively
proportion, up to 50%, of episodes of increased symptoms small change from his baseline, or when a patient with mild
are not reported by patients to their health care providers COPD has much larger acute change in his symptoms and
and therefore are not associated with additional treatment. lung function. The exacerbation etiology and pathophysiol-
Such episodes, which likely represent mild exacerbations ogy may dier considerably in these circumstances and may
would not meet this denition of exacerbation. warrant distinct treatment approaches.
Missing in both denitions is the clinical exclusion of Exacerbation severity has been variously dened
entities that could lead to increased respiratory symptoms among studies. Ideally, changes in lung function should be
in a manner similar to exacerbation, such as pneumonia, used to dene severity of exacerbations. However, spirome-
congestive heart failure, upper respiratory infection, non- try and lung volumes are dicult to measure accurately dur-
compliance with medications etc. These clinical entities have ing exacerbations, especially severe episodes. With simpler

664
Antibiotics 53
TABLE 53.2 Anthonisen classification of COPD exacerbations based on It is evident that we need a better denition and
cardinal symptoms. objective measures of severity of exacerbations. Recent work
in the development of patient reported outcomes to meas-
Severity of Type of
ure exacerbations has demonstrated that the experience of
exacerbation exacerbation Characteristics
an exacerbation from the patient perspective includes not
Severe Type 1 Increased dyspnea, sputum only respiratory symptoms but extra-respiratory manifesta-
volume, and sputum purulence tions including fatigue, anxiety, sleep disturbance etc. Future
denitions of exacerbation should therefore include such
Moderate Type 2 Any two of the above three symptoms. An expectation in the future is that a properly
cardinal symptoms developed patient reported outcome measure would become
Mild Type 3 Any one of the above three
universally applied to dene exacerbations, where a certain
cardinal symptoms and one or
change from baseline in this measure would constitute an
more of the following minor
exacerbation. Furthermore, the degree of change in such a
symptoms or signs
measure from baseline would represent an objective meas-
urement of severity.
Cough Biomarkers in exacerbations are being vigorously
Wheezing explored, as they hold the promise of being objective meas-
Fever without an obvious source ures to dene an exacerbation and determine its severity. A
Upper respiratory tract infection recent study explored 36 plasma biomarkers in 90 patients
in the past 5 days with exacerbations and found that none of them alone or
Respiratory rate increase 20% in combination were adequate to dene an exacerbation
over baseline [15]. In another study of multiple serum biomarkers in 20
Heart rate increase 20% over hospitalized patients with exacerbation, reduction in inter-
baseline leukin-6 (IL-6) and interleukin-8 (IL-8) correlated with
decrease in dyspnea during recovery from exacerbation,
Based on data from Ref. [12].
while decreases in IL-6 and tumor necrosis factor (TNF-)
were proportional to recovery in FEV1 [16]. Changes from
measures, such as peak ow, often the change with an exacer- baseline in sputum IL-8 and TNF- as well as sputum neu-
bation is of the same magnitude as its day-to-day variability. trophil elastase (NE) and serum C-reactive protein (CRP)
Severity has been also measured by site of care, with hospi- correlate with clinical severity of an exacerbation as assessed
talized exacerbations regarded as severe, outpatient exacerba- by a clinical score based on symptoms and signs [17, 18].
tions regarded as moderate and self medicated exacerbations It appears unlikely that a single biomarker will be capable
as mild [14]. Site of care is unreliable as a measure of exac- of dening an exacerbation, because of the heterogeneity
erbation severity. Though the major factor determining site of these episodes. However, biomarkers do hold promise in
of care is undoubtedly illness severity, it is also dependent on objectively determining severity of exacerbations and den-
dierences in hospital admission practices among countries ing etiology to guide appropriate treatment.
and health care systems, patient reporting of exacerbations,
physician preferences, etc. The intensity of recommended
treatment has also been used as a measure of severity of
exacerbations, with treatment with bronchodilators only
indicating mild exacerbations, while treatment with antibi-
PATHOGENESIS OF EXACERBATIONS
otics and steroids in addition to bronchodilators regarded as
indicating moderate or severe exacerbations. Such measure- An increase in airway inammation from the baseline level
ment of severity is beset with the same problems of prefer- in a patient appears central to the pathogenesis of most
ences and practice approach as discussed above. acute exacerbations [19, 20]. Airway inammation measured
Another widely used determination of severity of in induced or expectorated sputum, bronchoalveolar lavage
exacerbations is the Anthonisen classication [12]. This or bronchial biopsy has revealed that increased inamma-
classication relies on the number of cardinal symptoms and tion accompanies exacerbations and resolves with treatment
the presence of some supporting symptoms (Table 53.2). [2125]. Both neutrophilic and eosinophilic inammations
Though not developed as a severity classication, it has have been described. This acute increase in airway inam-
become so over time. Advantages of this determination of mation leads to increased bronchial tone, edema in the
severity are its simplicity and that it correlates with benet bronchial wall and mucus production. In a diseased lung,
with antibiotics, with such benet seen only in Type 1 and these processes worsen ventilation-perfusion mismatch and
2 exacerbations. Limitations include its lack of validation expiratory ow limitation. Clinically, these pathophysiologic
against objective measures of severity and that its ability to changes present as an increase in or new onset of dyspnea,
predict benet with antibiotics has not been consistent in cough, sputum production, tenacity and purulence along
other studies. Another limitation is the lack of gradation of with worsening gas exchange, which are the cardinal mani-
severity within each symptom, such that a Type 2 exacer- festations of an exacerbation. Inammation in exacerbations
bation with mild changes in two cardinal symptoms would extends beyond the lung, and increased plasma brinogen,
be regarded as the same severity in this classication as an interleukin 6 (IL-6) and CRP have been described during
exacerbation with a marked increase in both symptoms. exacerbations [18, 26, 27]. These and other mediators likely

665
Asthma and COPD: Basic Mechanisms and Clinical Management

cause the systemic manifestations of exacerbations, includ- described [2931]. Environmental pollutants, both particu-
ing fatigue and in some instances fever. late matter, such as PM-10 and diesel exhaust particles, and
The etiology of exacerbations appears to determine nonparticulate gases, such as ozone, nitrogen dioxide, sul-
the nature and degree of inammation in exacerbations. fur dioxide, are capable of inducing inammation in vitro
Neutrophilic inammation is characteristic of bacterial and in vivo [3234]. Infectious agents, including bacteria,
exacerbations, while both neutrophilic and eosinophilic viruses, and atypical pathogens are implicated as causes of
inammations have been described with viral infection [28]. up to 80% of acute exacerbations [35].
The intensity of neutrophilic inammation, when measured
as associated cytokines/chemokines (IL-8, TNF-) and
products of neutrophil degranulation (NE and myeloperox-
idase), is much greater in well-characterized bacterial exac- MICROBIAL PATHOGENS IN COPD
erbations than exacerbations of nonbacterial etiology [18].
Systemic inammation, measured as serum CRP is also
EXACERBATIONS
more intense in bacterial exacerbations [18]. These nd-
ings have important implications. They can form the basis Potential pathogens in COPD exacerbations includes typi-
for biomarkers to distinguish etiology of exacerbations in a cal respiratory bacterial pathogens, respiratory viruses, and
reliable and rapid manner, facilitating appropriate therapy. atypical bacteria (Table 53.3). Pneumocystis jiroveci, a fungus,
The heightened airway and systemic inammation with appears to cause chronic infection in COPD. Whether it
bacterial exacerbations can be potentially damaging to the induces exacerbations is being investigated. Among the typ-
lungs. Eective antibiotic therapy to eradicate the bacteria ical bacteria, Nontypeable Haemophilus inuenzae (NTHI)
responsible for exacerbations and reduce the inammation is the most common pathogen in COPD and is the best
to baseline levels becomes desirable and could have poten- understood [36]. Among the viruses, Rhinovirus and
tial long-term benets in COPD. Respiratory syncytial virus (RSV) have received considerable
A variety of noninfectious and infectious stimuli can attention in recent years and their importance in COPD is
induce an acute increase in airway inammation in COPD, now better appreciated [37, 38].
thereby causing an exacerbation. Increased respiratory The predilection of these pathogens for causing infec-
symptoms and respiratory mortality among patients with tions in COPD may be related to certain shared characteris-
COPD during periods of increased air pollution have been tics. NTHI, Streptococcus pneumoniae and Moraxella catarrhalis

TABLE 53.3 Microbial pathogens in exacerbations of COPD.

Proportion of
exacerbations (%) Specific species Proportion of class of pathogens (%)

Bacteria

4050 Nontypeable Haemophilus influenzae 3050


Streptococcus pneumoniae 1520
Moraxella catarrhalis 1520
Pseudomonas spp. and Enterobacteriaceae Isolated in very severe COPD, concomitant bronchiectasis,
and recurrent exacerbations
Haemophilus parainfluenzae Isolated frequently, pathogenic significance undefined
Haemophilus hemolyticus Isolated frequently, pathogenic significance undefined
Staphylococcus aureus Isolated infrequently, pathogenic significance undefined

Viruses

3040 Rhinovirus 4050


Parainfluenzae 1020
Influenza 1020
RSV 1020
Coronavirus 1020
Adenovirus 510

Atypical bacteria

510 Chlamydia pneumoniae 9095


Mycoplasma pneumoniae 510

666
Antibiotics 53
are the predominant bacterial causes of two other common to bacterial infection over the last few years. The current
respiratory mucosal infections, acute otitis media in chil- model of bacterial exacerbation pathogenesis involves
dren and acute sinusitis in children and adults. Pseudomonas both host and pathogen factors (Fig. 53.1). Acquisition of
aeruginosa is the dominant mucosal pathogen in cystic bro- strains of bacterial pathogens that are new to the host from
sis and noncystic brosis bronchiectasis [39]. These mucosal the environment is the primary event that puts the patient
infections have been related to anatomical abnormalities with with COPD at risk for an exacerbation [13]. Variation
impaired drainage of secretions, antecedent viral infections among strains of a species in the surface antigenic struc-
and defects in innate and adaptive immunity. All these pre- ture, as is seen with NTHI, S. pneumoniae, M. catarrhalis,
disposing factors likely exist in COPD. NTHI, S. pneumoniae and P. aeruginosa, is crucial to the development of recurrent
and M. catarrhalis are exclusively human pathogens. Their exacerbations with these pathogens. This variation allows
usual environmental niche in healthy humans is conned to these newly acquired strains to escape the pre-existing
the upper airway which they usually colonize without any host immune response that had developed following prior
clinical manifestations. Acquisition of these pathogens in exposure to other strains of the same species. These newly
COPD, because of compromised lung defense, allows estab- acquired strains can therefore proliferate in the lower air-
lishment of infection in the upper as well as the lower res- ways and induce acute inammation. The virulence of the
piratory tract (below the vocal cords). Infection of the lower strain and as yet unidentied host factors may determine if
respiratory tract in COPD can be with or without overt clin- the acute inammatory response to the pathogen reaches
ical manifestations, the former being addressed as exacerba- the threshold to cause symptoms that present as an exac-
tion while the latter as colonization. erbation [40]. In the majority of instances, mucosal and
Respiratory viruses implicated in COPD cause acute systemic antibodies develop to the pathogen [41, 42]. This
tracheobronchial infections in healthy hosts, clinically immune response, in combination with appropriate anti-
referred to as acute bronchitis. In the setting of COPD, biotics, eliminates or controls proliferation of the infecting
with diminished respiratory reserve, this acute bronchi- bacteria. However, because of antigenic variability among
tis has more profound manifestations and serious clinical strains of these bacterial species, these antibodies directed
consequences. at the infecting strain are usually strain-specic, and do not
protect the host from antigenically distinct strains of the
same species. This allows recurrent bacterial infection and
exacerbations in these patients.
The pathogenesis of acute viral exacerbations is less
PATHOGENESIS OF INFECTIOUS well understood, but may be similar to bacterial infec-
EXACERBATIONS tions. A common cause of exacerbations, the rhinovirus,
demonstrates considerable antigenic variation among its
more than 100 serotypes, allowing for recurrent infections.
Signicant progress has been made in our understanding The inuenza virus demonstrates drift in the antigenic make-
of acute exacerbation pathogenesis, especially in relation up of its major surface proteins, thereby leading to recurrent

Acquisition of new bacterial strain

Pathogen virulence
Host lung defense

Change in airway inflammation

Level of symptoms

Colonization Exacerbation

Strain-specific immune response


/ antibiotics

Tissue invasion
antigenic alteration
Elimination of
FIG. 53.1 Proposed model of bacterial exacerbation
infecting strain
pathogenesis in COPD. Reproduced with permission from
Persistent infection
Ref. [43].

667
Asthma and COPD: Basic Mechanisms and Clinical Management

infections. In vitro, viruses can damage airway epithelium, of clinical improvement as adequate rather than clinical
stimulate muscarinic receptors, and induce eosinophil and resolution to baseline has important implications. Clinical
neutrophil inux [44]. Whether these pro-inammatory improvement likely reects inadequate treatment, permit-
actions are enhanced in epithelial cells from patients with ting the inammatory process accompanying the exacer-
COPD is not known. Bronchial epithelial cells obtained bation to persist for prolonged periods of time, causing
from patients with asthma have diminished production of progressive airway damage [45]. Therefore, clinical resolu-
interferons and increased ICAM-1 expression, changes tion of symptoms to baseline is a more appropriate goal of
which could result in increased inammation, cell lysis, treatment of exacerbations.
and viral replication on viral infection. Increased ICAM-1 Additional important clinical goals of treatment
expression in COPD bronchial epithelium has been seen, include delaying the next exacerbation, prevention of early
however the other changes have not been described. relapse and more rapid resolution of symptoms [4648].
Lengthening the inter-exacerbation interval and preven-
tion of early relapse ultimately translate to a decrease in
the frequency of exacerbations, which is now a major focus
GOALS OF TREATMENT OF EXACERBATIONS of COPD treatment. Though most patients and physi-
cians would accept faster recovery to baseline as a desirable
goal of treatment, development of this parameter has been
The traditional aims of treatment of an exacerbation are hampered by lack of well-validated instruments to reliably
improvement in clinical status and the prevention of com- measure exacerbation resolution. Patient reported outcomes
plications. Though undoubtedly important, several new in development will address this need in the near future.
observations question the adequacy of these goals. These Biological goals of treatment are either still in their
include the importance of exacerbations in the course of infancy or, in the case of bacteriologic eradication, inad-
COPD, the role of infection in exacerbations, the high equately assessed in clinical studies to satisfy regulatory
rates of relapse with an adequate initial clinical response, requirements for approval of new antibiotics. Most exacer-
and the potential damaging eects of chronic colonization bations are inammatory events, therefore it is logical that
in COPD. To draw an analogy, conning our goal in the resolution of inammation to baseline should be an impor-
treatment of COPD exacerbations to short-term resolu- tant goal of treatment. Similarly, exacerbations are in many
tion of symptoms would be the equivalent of treating acute instances induced by infection, therefore eradication of the
myocardial infarction with the only aim being resolution of oending infectious pathogen should be a goal of treatment.
chest pain. Several other goals of treatment, both clinical Practical application of these biological goals of treatment of
and biological, should therefore be considered (Table 53.4). exacerbations awaits the development of biomarkers that
A good example of an inadequate goal in the treat- provide simple, rapid and reliable measurements of inam-
ment of exacerbations is clinical success, which is dened mation and infection.
as resolution or improvement of symptoms to a degree A multi-modality approach to treatment of exacerba-
that no further treatment is required in the opinion of the tions is common, that utilizes several modalities simultane-
treating physician. Recent observations have shown that ously, to relieve symptoms, to treat the underlying cause or
symptoms of an exacerbation are correlated with exagger- to provide support till recovery occurs [14, 49]. These thera-
ated airway and systemic inammation. Hence, acceptance pies include bronchodilators, corticosteroids, antimicrobials,

TABLE 53.4 Goals of treatment of COPD exacerbation.

Goals Comments

Clinical

Faster resolution of symptoms Needs validated symptom assessment tools


Clinical resolution to baseline Needs baseline assessment prior to exacerbation onset for comparison
Prevention of relapse Relapse within 30 days is quite frequent
Increasing exacerbation-free interval Needs long-term follow-up after treatment
Preservation of health-related quality of life Sustained decrements seen after exacerbations

Biological

Bacterial eradication Often presumed in usual antibiotic comparison studies


Resolution of airway inflammation Shown to be incomplete if bacteria persist
Resolution of systemic inflammation Persistence of systemic inflammation predicts early relapse
Restoration of lung function to baseline Incomplete recovery is seen in significant proportion
Preservation of lung function Needs long-term studies

668
Antibiotics 53
mucolytics and expectorants and, in the more severe cases, only three patients and the number needed to treat to pre-
oxygen supplementation and mechanical ventilation for vent one clinical failure was six patients. Diarrhea was the
acute respiratory failure. most frequently related adverse eect, with one episode per
seven patients treated. Antibiotic treatment was also bene-
cial in resolving sputum purulence. A benet on lung func-
tion and gas exchange was not observed, however, the data
ANTIBIOTICS IN THE TREATMENT examining this end point was scanty.
The reason that the two meta-analyses came up
OF EXACERBATIONS with dierent results is in large part inclusion in the later
analysis of a study performed by Nouira et al. published in
The role of antibiotics in the treatment of COPD exacerba- 2003. In this randomized double blind study, 93 patients
tions has been a matter of controversy. Even more conten- with exacerbations of severe underlying COPD requiring
tious has been the issue whether antibiotic choice is relevant ventilator support in an intensive care unit were randomly
to clinical outcome of exacerbations. Recommendations for assigned to receive a uoroquinolone antibiotic, ooxacin,
antibiotic use among published guidelines are inconsist- or placebo [57]. No systemic corticosteroids were admin-
ent [14, 5052]. There is a paucity of well-designed, large istered. Bacterial pathogens were isolated in tracheobron-
randomized controlled trials with adequate goals of treat- chial aspirates in 61% of patients. Ooxacin administration
ment comparing antibiotics to placebo or among antibiotic was associated with dramatic benets compared to placebo,
classes. This paucity of evidence upon which to base solid reducing mortality (4% versus 22%) and the need for addi-
recommendations has undoubtedly contributed to the con- tional antibiotics (6% versus 35%) by 17.5-fold and 28.4-
troversy and inconsistency of recommendations regarding fold, respectively.
antibiotic use [53]. Another important study, which for some reason
Recently, a few well-designed placebo control- has not been included in either meta-analysis, was per-
led and antibiotic comparison trials have been reported. formed in Italy by Allegra et al. and published in 1991.
Furthermore, epidemiologic studies have consistently iden- In this double blind randomized trial, amoxicillin/clavu-
tied certain clinical risk factors, which in the setting of lanate was compared with placebo in 414 exacerbations in
an exacerbation are predictive for failure of treatment or 369 patients with varying severity of underlying COPD
early relapse. The clinical outcomes of exacerbation in [58]. A unique feature of this study was the measurement
observational real-life studies are clearly sub-optimal, with of primary outcome at 5 days after the start of treatment,
as many as 2533% of patients experiencing treatment instead of the traditional 23 weeks. This earlier timing is
failure or early relapse. Considering the heterogeneity of clinically of greater relevance than the usual later one, as
COPD and of exacerbations, it is clear that the one size in clinical practice if patients are not improved within 35
ts all approach of using the same antibiotic in all episodes days, they are reassessed and their therapy altered. Clinical
is sub-optimal. It is likely that a proportion of treatment success (including resolution and improvement) was signi-
failures in exacerbations are related to ineective antibiot- cantly better with the antibiotic, seen in 86.4% of patients,
ics. Patients at risk for poor outcome are the logical can- compared with 50.6% in the placebo arm. In addition,
didates for aggressive initial antibiotic treatment, with the with increasing severity of underlying COPD, the benet with
expectation that such an approach would improve overall antibiotics as compared to placebo was larger.
exacerbation outcomes. This risk stratication approach Results of the meta-analyses, the Allegra study, and of
has also been advocated for other community-acquired the previous classic large placebo-controlled trial conducted
infections such as pneumonia and acute sinusitis [23, 54]. by Anthonisen et al., clearly demonstrate that antibiotics are
Though improved outcomes with such risk stratication has benecial in the treatment of moderate to severe exacerba-
not yet been demonstrated in prospective controlled trials, tions [12, 5658]. Furthermore, the benet with antibiot-
this approach takes into account concerns of disease hetero- ics is more marked early in the course of the exacerbation,
geneity, antibiotic resistance and judicious antibiotic use. suggesting that antibiotics hasten resolution of symptoms
[58, 59]. The benet with antibiotics is also greater as the
severity of underlying airow obstruction increases. This
Placebo-controlled antibiotic trials could be related to more frequent bacterial infections and/
or a decreased ability of the host in dealing with infections,
Exacerbations of COPD result in signicant antibiotic con- therefore requiring help with antibiotics to resolve them.
sumption, however, there are only a handful of placebo con- Important questions regarding the role of antibiot-
trolled trials in this disease. Two meta-analyses of placebo ics in exacerbations still remain. The benet of antibiotics
controlled trials in exacerbations have been published. The in mild exacerbations in the context of mild underlying
rst such analysis published in 1995 included nine trials and COPD is unproven and warrants a placebo controlled trial.
found a small but signicant benecial eect of antibiotics The eect of concomitant treatment with systemic corti-
over placebo [55]. In the second analysis published in 2006, costeroids on the benets of antibiotic therapy in exacerba-
11 trials were included, and a much larger benecial eect tions is not known. Placebo controlled trials of antibiotics
on mortality and prevention of clinical failure was demon- have not systematically regulated concomitant therapy, and
strated, especially in moderate to severe exacerbations [56]. all the placebo controlled trials of steroids had antibiot-
In this analysis, the number needed to treat in severe exac- ics administered to all patients. Because inammation and
erbations in hospitalized patients to prevent one death was infection are linked, it is likely that there would be additive

669
Asthma and COPD: Basic Mechanisms and Clinical Management

benets when both treatments are used over either treat- of regulatory requirements, these studies are conducted in
ment alone [56, 60]. a very similar manner and in similar patient populations.
This makes these trials very amenable to a meta-analytic
approach. Dimopoulos et al. used such an approach to
Antibiotic comparison trials determine whether there was any dierence in clinical
outcomes among rst-line antibiotics (amoxicillin, ampi-
Antibiotics are clearly useful in moderate to severe exac- cillin, pivampicillin, trimethoprim/sulfamethoxazole, and
erbations of COPD. However, there remains consider- doxycycline) and second-line antibiotics (amoxicillin/cla-
able controversy as to antibiotic choice, especially for initial vulanate, macrolides, second-generation or third-generation
empiric therapy of exacerbations [14, 5052, 61]. Most cephalosporins, and uoroquinolones) in the treatment of
exacerbations nowadays are treated without obtaining spu- exacerbations of chronic bronchitis [63]. They identied
tum bacteriology and with the trend to short course anti- 12 randomized controlled trials that had enrolled 2261
biotic therapy, this initial empiric choice often becomes the patients, 10 of these trials included the penicillins as the
only choice made of antibiotics in exacerbations. Results of rst-line antibiotic. Only a single trial each with trimetho-
antibiotic comparison trials should guide the recommenda- prim/sulfamethoxazole and doxycycline was included. In
tions for appropriate empiric antibiotics in exacerbations. the clinically evaluable patients, rst-line antibiotics were
However, though the literature is replete with such trials, only half as eective as second-line antibiotics with an
in the vast majority, antibiotic choice does not apparently odds ratio for clinical treatment success of 0.51 (95% CI,
aect the clinical outcome. However, dierences in bacte- 0.340.75). This result was consistent in several sensitivity
riological eradication rates among antibiotics are seen, with analyses, with the exception of trials published before 1991,
a dissociation between clinical and bacteriological outcomes where the dierence between rst line and second line anti-
[62]. These results are contrary to expectations that antibi- biotics was not seen. There was no dierence between the
otics with better in vitro and in vivo antimicrobial ecacy rst-line and second-line antibiotics in adverse eects.
and better pharmacodynamic and pharmacokinetic charac- This meta-analysis provides additional evidence that
teristics should show superior clinical outcomes. A closer antibiotic choice does make a dierence in the treatment
examination of the trial design of these studies reveals sev- of exacerbations. Similar treatment success for rst line and
eral shortcomings that oer potential explanations for this second line antibiotics in trials before 1991 but not after
paradox (Table 53.5) [53]. Many of these deciencies are 1991, suggests that resistance emergence in causative bacte-
related to the fact that these trials are performed for regula- rial pathogens (H. inuenzae, M. catarrhalis, S. pneumoniae)
tory approval of the drugs, therefore are designed for dem- is responsible for the ndings of this meta-analysis. Because
onstrating noninferiority rather than dierences between of the limited number of studies in which the rst line
the two antibiotics. In the face of this large body of data antibiotics were not penicillins, the results of this meta-analysis
showing clinical equivalence, it is not surprising that sev- mainly applies to the penicillins. Based on this meta-
eral guidelines do not dierentiate between antibiotics for analysis, recommendations to use amoxicillin and ampicillin
therapy of exacerbations. in the treatment of exacerbations cannot be supported.
Most antibiotic comparison trials are underpowered These investigators also performed a similar meta-
to detect dierences among antibiotics. However, because analysis where they compared second-line antibiotics,

TABLE 53.5 Limitations of published placebo-controlled antibiotic trials in acute exacerbations of COPD.

Limitation of study design Potential consequences

Small number of subjects Type 2 error

Subjects with mild or no underlying COPD included Diminished overall perceived efficacy of antibiotics

Nonbacterial exacerbations included Type 2 error

Endpoints compared at 3 weeks after onset Spontaneous resolution mitigates differences between arms
Clinically irrelevant as most decisions about antibiotic efficacy
are made earlier

Speed of resolution not measured Clinically relevant endpoint not assessed

Lack of long-term follow-up Time to next exacerbation not assessed

Antibiotic resistance to agents with limited in vitro Diminished overall perceived efficacy of antibiotics
antimicrobial efficacy

Poor penetration of antibiotics used in to respiratory tissues Diminished overall perceived efficacy of antibiotics

Concurrent therapy not controlled Undetected bias in use of concurrent therapy

Reproduced with permission from Ref. [53].

670
Antibiotics 53
the macrolides, the uoroquinolones and amoxicillin/clavu- to baseline) following treatment. A substantial proportion
lanate in a similar manner [64]. In this analysis of 19 rand- of the patients enrolled had one or more risk factors that
omized controlled trials that had enrolled 7405 patients, no would predispose to a poor outcome as discussed below.
dierences were found among these agents in clinical treat- Patients were followed up to 9 months after randomization
ment success dened in the conventional manner. to provide an estimate of recurrence of exacerbation.
In addition to these meta-analyses, welcome additions In line with usual antibiotic comparison trials, moxi-
to the literature on antibiotic treatment of exacerbations are oxacin and standard therapy were equivalent (88% versus
two recent antibiotic comparison trials that were designed 83%) for clinical success (resolution and improvement) at
as superiority studies. They also measured some uncon- 710 days after the end of therapy. However, moxioxacin
ventional but clinically relevant end-points. The GLOBE therapy was associated with a superior clinical cure rate
(Gemioxacin and Long term Outcome of Bronchitis (dened as resolution of symptoms to baseline, rather than
Exacerbations) trial, a double blind, randomized trial, com- simply improvement) than standard therapy (71% versus
pared a uoroquinolone, gemioxacin, with a macrolide, 63%), as well as with superior bacteriologic response (91.5%
clarithromycin [65]. End of therapy and long-term out- versus 81%). Several other a priori unconventional end-
come assessments were made at the conventional 1014 day points were examined. Moxioxacin treatment resulted in
and 28-day time intervals. These assessments, in line with signicantly fewer courses of additional antibiotic therapy
most antibiotic comparison trials, did not demonstrate sta- (8% versus 14%) and an extended time to the next exacer-
tistically signicant dierences in the two arms, with clini- bation (131 versus 104 days) [66]. A composite end-point
cal success rates of 85.4% and 84.6% for gemioxacin and of clinical failure, requirement of additional antibiotics and
clarithromycin respectively. Also in line with similar stud- recurrence of exacerbation demonstrated a clear dierence
ies, bacteriological success, measured as eradication and between the two arms, with moxioxacin being statistically
presumed eradication, was signicantly higher with gemi- superior to standard therapy for up to 5 months of follow-
oxacin (86.7%) compared to clarithromycin (73.1%). up. As with the GLOBE trial, if conventional clinical suc-
Patients with a successful clinical outcome at 28 cess would have been measured solely in this study, all the
days were enrolled in a follow-up period for a total of 26 other signicant dierences in the two arms would have not
weeks of observation. In this time period, the primary out- been discovered.
comes were the rate of repeat exacerbations, hospitaliza- The GLOBE and MOSAIC trials demonstrate that
tions for respiratory disease and health-related quality of in vitro microbiological superiority as well as the enhanced
life measures. A signicantly lower rate of repeat exacer- pharmacokinetic/pharmacodynamic properties in the res-
bations was observed with gemioxacin, with 71% of the piratory tract of the uoroquinolones does translate to
patients remaining exacerbation free at 26 weeks compared greater in vivo eectiveness in the treatment of exacerba-
to 58.5% in the clarithromycin arm. The relative risk reduc- tion. Antibiotics for exacerbation have very similar results
tion for recurrence of exacerbation was 30%. The rate of for the standard regulatory end-point of clinical success at
hospitalization for respiratory tract illness in the 26 weeks 714 days after the end of therapy. This standard end-point
was also lower in the gemioxacin treated than in the clari- not only lacks discriminatory power, it also has little clini-
thromycin treated patients (2.3% versus 6.3%, p 0.059) cal relevance. Most decisions about antibiotic benet in the
[65]. Patients who remained free of recurrence in the 26- clinical setting are made within the 1st week of therapy.
week period regained more of their health-related quality of Dierences among antibiotics are perceptible when clini-
life than those who had a recurrent exacerbation [6]. This cally relevant end-points such as speed of resolution, clini-
trial clearly demonstrates that conventional medium-term cal cure, need for additional antimicrobials and time to next
clinical outcomes are unsuitable for measuring dierences exacerbation are considered [65, 66].
among antibiotics in exacerbations. If the 26-week follow-
up period had not been included in the GLOBE study, sig-
nicant dierences in the two treatment arms in clinically
relevant outcomes of recurrence of exacerbations and respi-
ratory related hospitalization would have been missed.
RISK STRATIFICATION OF PATIENTS
The MOSAIC trial is another recent landmark
antibiotic comparison trial in exacerbations of COPD. Fluoroquinolones are excellent antimicrobials for exacer-
Patients in this study were randomized to a uoroqui- bations, and based on the MOSAIC and GLOBE studies,
nolone, moxioxacin or to standard therapy (which could be it is tempting to prescribe them for all moderate to severe
one of the following: amoxicillin, cefuroxime or clarithro- exacerbations. Such a strategy, though likely to be success-
mycin) [66]. Several unique design features of this trial are ful in the short-term, would foster antimicrobial resistance
noteworthy, which relate to observations made in this study to these valuable antibiotics in the long term. Therefore,
and set the standard for future antibiotic comparison trials it would be judicious to make an eort to identify those
in this disease. The number of patients enrolled was much patients and exacerbations that are most likely to benet
larger than previous studies, in order to provide adequate from these antibiotics and use them in those circumstances.
power to demonstrate superiority. Patients were enrolled Observational real life studies of the outcome of exac-
when stable to establish a baseline as a comparison to reli- erbations in the community have clearly demonstrated that
ably distinguish between clinical improvement (enough our current treatment approach is sub-optimal. In these
improvement that no additional antibiotic treatment is studies, treatment failure rates, either dened as failure
required) from clinical cure (improvement of symptoms to improve or relapse within 30 days of 2533% are seen

671
Asthma and COPD: Basic Mechanisms and Clinical Management

[67, 68]. These studies have also demonstrated that certain exacerbations based on the risk factors discussed above as
patient characteristics that antedate the onset of the exac- well as the in vitro and in vivo ecacy of antibiotics. Our
erbation impact the outcome of the exacerbation [48, 67 current treatment algorithm is shown in Fig. 53.2 [14, 51,
71]. Interestingly, several of these characteristics, such as 61]. Once an exacerbation is diagnosed, the initial step in
co-morbid cardiac disease and frequent exacerbations, were the algorithm is determination of the severity of the exac-
found to be relevant to outcome in more than one study. erbation. We use the Anthonisen criteria by dening single
These risk factors for poor outcome should be considered cardinal symptom exacerbations as mild, while the presence
in the decision regarding choice of empiric antibiotics when of two or all three of the cardinal symptoms denes moder-
treating exacerbations. In theory, patients at risk for poor ate and severe exacerbations.
outcome would have the greatest benet from early aggres- Mild exacerbations are initially managed with symp-
sive antibiotic therapy, such as with the uoroquinolones. tomatic treatment, including bronchodilators, antitussives
These are the patients in whom the consequences of treat- and expectorants and antibiotics are with held. However,
ment with an antibiotic ineective against the pathogen patients are counseled regarding the cardinal symptoms
causing the exacerbation are likely to be signicant, with and if additional symptoms appear, then antibiotics are
clinical failures, hospitalizations and early recurrences likely. prescribed. If a moderate to severe exacerbation is diag-
These at risk patients contribute substantially to the overall nosed, the next important step is the dierentiation of
poor clinical outcomes of exacerbations. Therefore, targeting uncomplicated patients from the complicated patients.
these patients with potentially more eective treatment is Uncomplicated patients do not have any of the risk factors
likely to have a signicant impact on the overall outcomes for poor outcome. Complicated patients have one or more
of exacerbations. of the following risk factors for poor outcome: Age 65
Risk factors for poor outcome identied in various years, FEV1  50%, co-morbid cardiac disease and three or
studies are increasing age, severity of underlying airway more exacerbations in the previous 12 months [14, 51, 61].
obstruction, presence of co-morbid illnesses (especially car- A threshold of four or more exacerbations in the previous
diac disease), a history of recurrent exacerbations, use of 12 months has been used to dene frequent exacerbations
home oxygen, use of chronic steroids, hypercapnia and acute in previous studies and guidelines. However, current ther-
bronchodilator use [48, 6771]. It is likely that home oxy- apy of COPD with long acting bronchodilators and inhaled
gen use, hypercapnia and chronic steroid use mainly reect steroids has reduced overall frequency of exacerbations in
increasing severity of underlying COPD. Acute bronchodi- this disease by about 25%. Therefore, we use a threshold
lator use could either be related to the severity of underly- of three or more exacerbations to dene the complicated
ing COPD or reect the wheezy phenotype of exacerbation patient on contemporary COPD treatment.
that would be less responsive to antibiotic treatment. Many In uncomplicated patients, antibiotic choices include
of these risk factors are continuous in severity, however, cer- an advanced macrolide (azithromycin, clarithromycin), a
tain thresholds have been dened in studies that are clini- cephalosporin (cefuroxime, cefpodoxime, or cefdinir), doxy-
cally useful and predictive of poor outcome and easier to use cycline or trimethoprim/sulfamethoxazole. Amoxicillin (or
clinically. These include an age of more than 65 years, forced ampicillin) is not an appropriate choice because of prevalent
expiratory volume in 1 s (FEV1) 50%, and 3 exacerba- antibiotic resistance and the results of the meta-analysis
tions in the previous 12 months. discussed above. In complicated patients, our usual antibi-
Another important consideration in choosing antibiot- otic choice is a respiratory uoroquinolone (moxioxacin,
ics for exacerbation is recent antibiotic use, especially within gemioxacin, levooxacin) with amoxicillin/clavulanate as
the past 3 months. Because of the recurrent nature of exac- an alternative.
erbation and the high prevalence of co-morbid conditions Other important considerations in choosing antibiot-
in COPD, such antibiotic exposure is likely to be prevalent ics include exposure to antibiotics within the past 3 months.
in COPD patients. In other respiratory infections such as This exposure history should be elicited not only for respi-
pneumonia, recent antibiotic use leads to increased risk for ratory infections, but includes antibiotics prescribed for any
harboring antibiotic resistant pathogens and therefore hav- indication. The antibiotic chosen to treat the exacerbation
ing a poor outcome following treatment. This phenomenon should be from a dierent class of agents from the one pre-
has been best described for S. pneumoniae among patients scribed within the past 3 months. For example, exposure to
with community acquired pneumococcal pneumonia and a macrolide in the past 3 months should lead to use of a
recently also described for this pathogen among patients cephalosporin in an uncomplicated patient. Similarly, prior
with COPD [72, 73]. Whether such selection for antibi- use of a uoroquinolone in a complicated patient should
otic resistant strains occurs among NTHI and M. catarrhalis lead to use of amoxicillin/clavulanate.
after antibiotic exposure is not known, but is possible. Another important consideration in the compli-
cated patients is a sub-group of these patients who are at
risk for infection by P. aeruginosa and Enterobacteriaceae
RISK STRATIFICATION APPROACH TO or have a documented infection by these pathogens [74].
ANTIBIOTIC THERAPY IN ACUTE These patients usually have very severe underlying COPD
(FEV1  35%), have developed bronchiectasis, are hospital-
EXACERBATION ized (often requiring intensive care), or have been recently
hospitalized or have received multiple courses of antibiot-
A risk stratication approach has been advocated by sev- ics. In such patients, empiric treatment with ciprooxacin
eral experts for the initial empiric antibiotic treatment of is appropriate. However, resistance among P. aeruginosa and

672
Antibiotics 53
Exacerbation

Mild Moderate or Severe


Only one of the three cardinal symptoms: At least two of the three cardinal symptoms:
Increased dyspnea Increased dyspnea
Increased sputum volume Increased sputum volume
Increased sputum purulence Increased sputum purulence

Uncomplicated COPD Complicated COPD


No antibiotics
no risk factors one or more risk factors
Increase bronchodilators
Age 65 years Age 65 years
Symptomatic therapy
FEV1 50% predicted FEV1 50% predicted
Instruct patient to report
3 exacerbations/year 3 exacerbations/year
additional cardinal symptoms
No cardiac disease Cardiac disease

Advanced macrolide (azithromycin, Fluoroquinolone (moxifloxacin,


clarithromycin), gemifloxacin, levofloxacin),
Cephalosporin (cefuroxime, Amoxicillin/clavunate
cefpodoxime, cefdinir), If at risk for Pseudomonas,
Doxycycline consider ciprofloxacin and obtain
Trimethoprim/sulfamethoxazole sputum culture
If recent (3 months) antibiotic If recent (3 months) antibiotic
exposure, use alternative class exposure, use alternative class

Worsening clinical status or inadequate response in 72 h

Re-evaluate
Consider sputum culture

FIG. 53.2 Algorithm for antibiotic treatment of acute exacerbations of COPD.

the Enterobacteriaceae to the uoroquinolones may compro- to treat pathogens and treat with an alternative agent with
mise their ecacy. Therefore, in this sub-group of patients, better in vitro microbiological ecacy.
we obtain a sputum (or tracheobronchial aspirate if intu-
bated) culture to allow adjustment of antibiotics based on
the in vitro susceptibility of pathogens isolated. Unless dic-
tated by poor clinical response and in vitro antimicrobial
susceptibility, we do not use combination or parenteral anti- ALTERNATIVE APPROACHES TO ANTIBIOTIC
biotic therapy for P. aeruginosa as this approach has never
been systematically examined and is of unproven benet in
THERAPY IN ACUTE EXACERBATION
exacerbations.
We instruct our patients to report to us any deterio- Patients with COPD exacerbation often experience a
ration or lack of improvement at 4872 h, because in this change in the color of sputum from white or gray (mucoid)
time frame clinical improvement should be apparent. In to yellow, green or brown (purulent). Purulence of sputum
these patients who are failing initial empiric antimicrobial is related to the presence of myeloperoxidase, a product of
therapy, we re-examine the patient to conrm the diagnosis, neutrophil degranulation. Neutrophil degranulation is asso-
consider sputum studies to ascertain for resistant or dicult ciated with bacterial infection, therefore sputum purulence

673
Asthma and COPD: Basic Mechanisms and Clinical Management

AUC 0.85 as biologic goals of bacterial eradication and inammation


1.0
reduction as discussed above were also not recorded.
0.9
Other biomarkers that have been explored in COPD
0.8 exacerbations include sputum TNF- and NE and CRP
0.7 (Fig. 53.3). Though individually they do not appear to dis-
criminate between exacerbations, a combination of these
Sensitivity

0.6
parameters reliably distinguishes well-characterized bacte-
0.5
rial exacerbations from others [18]. Additional studies are
0.4 required to conrm and extend these observations.
0.3 Other important considerations in antibiotic prescrib-
0.2 ing are safety and tolerability of the agent, drug interactions
0.1 and cost of treatment. It is important that cost of the anti-
biotic not be considered in isolation. Miravittles et al. have
0.0
0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8 0.9 1.0
shown that exacerbations in which initial empiric treatment
1  specificity
fails cost 10 times more than clinical successes [79]. The
investigators estimated that the overall cost of care could be
FIG. 53.3 Receiver operating characteristic (ROC) curve for distinguishing reduced by half with reduction in clinical failure rates by a
new bacterial strain exacerbations from exacerbations without new strains third. Though not shown as yet in properly designed stud-
with levels of sputum TNF-, sputum NE, and serum CRP at exacerbation ies, it is likely that an appropriate and logical approach to
included in the prediction model. The predictive values for each of the antibiotic choice, as discussed above, would reduce clinical
mediators in this model were sputum TNF-: 0.32 ng/ml; sputum NE: failure rates in exacerbations.
0.76nM; and serum CRP: 2.37 mg/l. AUC: Area under the ROC. Reproduced
with permission from Ref. [18].

References
at exacerbation is a marker of bacterial infection, dened
1. Burrows B, Earle RH. Course and prognosis of chronic obstructive
by quantitative cultures of sputum and bronchoscopic lung disease. A prospective study of 200 patients. N Engl J Med 280(8):
protected brush specimens [75, 76]. Presence of sputum 397404, 1969.
purulence has been advocated as the sole determinant for 2. Calverley PM et al. Salmeterol and uticasone propionate and survival
antibiotic treatment of exacerbations. However, its accuracy in chronic obstructive pulmonary disease. N Engl J Med 356(8): 775
and reproducibility as an indicator of bacterial infection is 89, 2007.
limited, and is likely to be even more so in clinical practice 3. Soler-Cataluna JJ et al. Severe acute exacerbations and mortality in
than in research studies [75, 76]. Often, patients have puru- patients with chronic obstructive pulmonary disease. Thorax 60(11):
lent sputum production even when stable, have intermittent 92531, 2005.
purulent sputum production during exacerbation, or have 4. Seemungal TAR et al. Eect of exacerbation on quality of life in
patients with chronic obstructive pulmonary disease. Am J Respir Crit
not have observed the sputum color. Sputum purulence is
Care Med 157: 141822, 1998.
one of the cardinal Anthonisen criteria, and we recommend 5. Spencer S et al. Health status deterioration in patients with chronic
it should be used in conjunction with other symptoms and obstructive pulmonary disease. Am J Respir Crit Care Med 163: 12228,
other measures of risk stratication in antibiotic treatment 2001.
of exacerbations (Fig 53.2). 6. Spencer S, Jones PW. Time course of recovery of health status follow-
Utilization of biomarkers to rapidly identify bacterial ing an infective exacerbation of chronic bronchitis. Thorax 58(7): 589
exacerbations and to guide antibiotic use is another poten- 93, 2003.
tial approach in exacerbations. Several potential biomark- 7. Kanner R et al. Lower respiratory illnesses promote FEV(1) decline in
ers have been identied in sputum and serum. The best current smokers but not ex-smokers with mild chronic obstructive pul-
studied biomarker of bacterial infection is serum procal- monary disease: Results from the lung health study. Am J Respir Crit
Care Med 164(3): 35864, 2001.
citonin level. In a recent study in patients hospitalized for
8. Donaldson GC et al. Relationship between exacerbation frequency and
exacerbations, antibiotic treatment was only recommended lung function decline in chronic obstructive pulmonary disease. Thorax
if the procalcitonin level was above a certain threshold. 57(10): 84752, 2002.
There was no dierence in outcomes in spite of reduction 9. Hirschmann JV. Do bacteria cause exacerbations of COPD? Chest 118:
in antibiotic use from 72% to 40% with procalcitonin guid- 193203, 2000.
ance [77]. However, this approach needs to be validated in 10. Murphy TF, Sethi S, Niederman MS. The role of bacteria in exacerba-
multi-center trials with varied populations before wide- tions of COPD. A constructive view. Chest 118(1): 2049, 2000.
spread application [78]. Furthermore, it needs to be tested 11. Rodriguez-Roisin R. Toward a consensus denition for COPD exacer-
in outpatients which represent the majority of exacerba- bations. Chest 117(5 Suppl 2): 398S401S, 2000.
tions treated, where patients are not as closely supervised 12. Anthonisen NR et al. Antibiotic therapy in exacerbations of
chronic obstructive pulmonary disease. Ann Intern Med 106: 196204,
and other supportive care is less rigorous. Only a minority
1987.
of patients received a uoroquinolone in this study, which 13. Sethi S et al. Acquisition of a new bacterial strain and occurrence of
should have been the antibiotics of choice in these compli- exacerbations of chronic obstructive pulmonary disease. N Engl J Med
cated patients. Therefore, the lack of eect of withholding 347(7): 46571, 2002.
antibiotics could have been related to their relative ine- 14. Globial Initiative for Obstructive Lung Disease; Available from: www.
cacy in the rst place. In this study, short-term goals as well goldcopd.com [cited].

674
Antibiotics 53
15. Hurst JR et al. Use of plasma biomarkers at exacerbation of chronic 38. Falsey AR et al. Respiratory syncytial virus infection in elderly and
obstructive pulmonary disease. Am J Respir Crit Care Med 174(8): 867 high-risk adults. N Engl J Med. 352(17): 174959 (2005).
74, 2006. 39. Aaron SD et al. Adult cystic brosis exacerbations and new strains of
16. Pinto-Plata VM et al. Systemic cytokines, clinical and physiologi- Pseudomonas aeruginosa. Am J Respir Crit Care Med 169(7): 81115,
cal changes in patients hospitalized for exacerbation of COPD. Chest 2004.
131(1): 3743, 2007. 40. Chin CL et al. Haemophilus inuenzae from patients with chronic
17. Sethi S et al. Airway inammation and etiology of acute exacerbations obstructive pulmonary disease exacerbation induce more inammation
of chronic bronchitis. Chest 118(6): 155765, 2000. than colonizers. Am J Respir Crit Care Med 172(1): 8591, 2005.
18. Sethi S et al. Inammatory prole of new bacterial strain exacerbations 41. Sethi S et al. Strain specic immune response to Haemophilus inuenzae
of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. in chronic obstructive pulmonary disease. Am J Respir Crit Care Med
177(5): 491-7 (2008). 169: 44853, 2004.
19. White AJ, Gompertz S, Stockley RA. Chronic obstructive pulmonary 42. Murphy TF et al. Moraxella catarrhalis in chronic obstructive pulmo-
disease. 6: The aetiology of exacerbations of chronic obstructive pulmo- nary disease: Burden of disease and immune response. Am J Respir Crit
nary disease. Thorax 58(1): 7380, 2003. Care Med 172(2): 19599, 2005.
20. Sethi S. New developments in the pathogenesis of acute exacerbations 43. Veeramachaneni SB, Sethi S. Pathogenesis of bacterial exacerbations of
of chronic obstructive pulmonary disease. Curr Opin Infect Dis 17(2): COPD. COPD: J Chronic Obstructive Pul Dis 3: 10915, 2006.
11319, 2004. 44. Johnston SL et al. Low grade rhinovirus infection induces a prolonged
21. Heckerling PS. The need for chest roentgenograms in adults with acute release of IL-8 in pulmonary epithelium. J Immunol 160(12): 617281,
respiratory illness. Arch Intern Med 146: 132124, 1986. 1998.
22. Bhushan R et al. Antigenic characterization and analysis of the human 45. White AJ et al. Resolution of bronchial inammation is related to bac-
immune response to outer membrane protein E of Branhamella catarrh- terial eradication following treatment of exacerbations of chronic bron-
alis. Infect Immun 65(7): 266875, 1997. chitis. Thorax 58(8): 68085, 2003.
23. Mandell LA et al. Update of practice guidelines for the management 46. Aaron SD et al. Outpatient oral prednisone after emergency treatment
of community-acquired pneumonia in immunocompetent adults. Clin of chronic obstructive pulmonary disease. N Engl J Med 348(26): 2618
Infect Dis 37(11): 140533, 2003. 25, 2003.
24. Kjaergard LL et al. Basophil-bound IgE and serum IgE directed 47. Anzueto A, Rizzo JA, Grossman RF. The infection-free interval: Its use
against Haemophilus inuenzae and Streptococcus pneumoniae in patients in evaluating antimicrobial treatment of acute exacerbation of chronic
with chronic bronchitis during acute exacerbations. APMIS 104: 6167, bronchitis. Clin Infect Dis 28(6): 134445, 1999.
1996. 48. Miravitlles M, Murio C, Guerrero T. Factors associated with relapse
25. Hiltke TJ, Sethi S, Murphy TF. Sequence stability of the gene encod- after ambulatory treatment of acute exacerbations of chronic bronchi-
ing outer membrane protein P2 of nontypeable Haemophilus inuenzae tis. DAFNE Study Group. Eur Respir J 17(5): 92833, 2001.
in the human respiratory tract. J Infect Dis 185(5): 62731, 2002. 49. Sethi S. Acute exacerbations of COPD: A multipronged approach. J
26. Wedzicha JA et al. Acute exacerbations of chronic obstructive pulmo- Respir Dis 23(4): 21755, 2002.
nary disease are accompanied by elevations of plasma brinogen and 50. Bach PB et al. Management of acute exacerbations of chronic obstruc-
serum IL-6 levels. Thromb Haemost 84(2): 21015, 2000. tive pulmonary disease: A summary and appraisal of published evi-
27. Dev D et al. Value of C-reactive protein measurements in exacerbations dence. Ann Intern Med 134(7): 60020, 2001.
of chronic obstructive pulmonary disease. Respir Med 92(4): 66467, 51. Balter MS et al. Canadian guidelines for the management of acute
1998. exacerbations of chronic bronchitis. Can Respir J 10(Suppl B): 3B32B,
28. Papi A et al. Infections and airway inammation in chronic obstruc- 2003.
tive pulmonary disease severe exacerbations. Am J Respir Crit Care Med 52. Celli BR, MacNee W. Standards for the diagnosis and treatment of
173(10): 111421, 2006. patients with COPD: A summary of the ATS/ERS position paper. Eur
29. Garcia-Aymerich J et al. Air pollution and mortality in a cohort of Respir J 23(6): 93246, 2004.
patients with chronic obstructive pulmonary disease: A time series 53. Sethi S. Bacteria in exacerbations of chronic obstructive pulmonary
analysis. J Epidemiol Community Health 54(1): 7374, 2000. disease. Phenomenon or epiphenomenon?. Proc Am Thorac Soc 1: 109
30. Sunyer J et al. Air pollution and emergency room admissions for 14, 2004.
chronic obstructive pulmonary disease: A 5-year study. Am J Epidemiol 54. Sinus A, Allergy Health P. Antimicrobial treatment guidelines for
137(7): 7015, 1993. acute bacterial rhinosinusitis 2004. Otolaryngol Head Neck Surg 130(1):
31. Sunyer J et al. Patients with chronic obstructive pulmonary disease are 145, 2004.
at increased risk of death associated with urban particle air pollution: 55. Saint S et al. Antibiotics in chronic obstructive pulmonary disease exac-
A case-crossover analysis. Am J Epidemiol 151(1): 5056, 2000. erbations. A meta-analysis. JAMA 273(12): 95796, 1995.
32. Ohtoshi T et al. Diesel exhaust particles stimulate human airway epi- 56. Ram FS et al. Antibiotics for exacerbations of chronic obstructive pul-
thelial cells to produce cytokines relevant to airway inammation monary disease. Cochrane Database Syst Rev(2), 2006. , CD004403
in vitro. J Allergy Clin Immunol 101(6 Pt 1): 77885, 1998. 57. Nouira S et al. Once daily oral ooxacin in chronic obstructive pul-
33. Devalia JL et al. Eect of nitrogen dioxide and sulphur dioxide on air- monary disease exacerbation requiring mechanical ventilation: A ran-
way response of mild asthmatic patients to allergen inhalation. Lancet domised placebo-controlled trial. Lancet 358(9298): 202025, 2001.
344(8938): 166871, 1994. 58. Allegra L et al. Antibiotic treatment and baseline severity of disease
34. Rudell B et al. Bronchoalveolar inammation after exposure to die- in acute exacerbations of chronic bronchitis: A re-evaluation of previ-
sel exhaust: Comparison between unltered and particle trap ltered ously published data of a placebo-controlled randomized study. Pulm
exhaust. Occup Environ Med 56: 52734, 1999. Pharmacol Ther 14(2): 14955, 2001.
35. Sethi S. Infectious etiology of acute exacerbations of chronic bronchi- 59. Miravitlles M et al. The ecacy of moxioxacin in acute exacerbations
tis. Chest 117: 380S85S, 2000. of chronic bronchitis: A Spanish physician and patient experience. Int J
36. Eldika N, Sethi S. Role of Nontypeable Haemophilus inuenzae in exac- Clin Pract 55(7): 43741, 2001.
erbations and progression of chronic obstructive pulmonary disease. 60. Wood-Baker RR et al. Systemic corticosteroids for acute exacerbations
Curr Opin Pulm Med 12(2): 11824, 2006. of chronic obstructive pulmonary disease. Cochrane Database Syst Rev
37. Seemungal T et al. Respiratory viruses, symptoms, and inammatory (1), 2005, CD001288.
markers in acute exacerbations and stable chronic obstructive pulmo- 61. Sethi S, Murphy TF. Acute exacerbations of chronic bronchitis: new
nary disease. Am J Respir Crit Care Med 164(9): 161823, 2001. developments concerning microbiology and pathophysiology impact

675
Asthma and COPD: Basic Mechanisms and Clinical Management

on approaches to risk stratication and therapy. Infect Dis Clin North 71. Groenewegen KH, Schols AM, Wouters EF. Mortality and mortality-
Am 18(4): 86182, 2004, ix. related factors after hospitalization for acute exacerbation of COPD.
62. Obaji A, Sethi S. Acute exacerbations of chronic bronchitis. What role Chest 124(2): 45967, 2003.
for the new uoroquinolones?. Drugs Aging 18(1): 111, 2001. 72. Vanderkooi OG et al. Predicting antimicrobial resistance in invasive
63. Dimopoulos G et al. Comparison of rst-line with second-line antibi- pneumococcal infections. Clin Infect Dis 40(9): 128897, 2005.
otics for acute exacerbations of chronic bronchitis: A meta-analysis of 73. Sethi S et al. Antibiotic exposure in COPD and the development of
randomized controlled trials. Chest 132(2): 44755, 2007. penicillin and erythromycin resistance in Streptococcus pneumoniae. San
64. Siempos II et al. Macrolides, quinolones and amoxicillin/clavulanate Francisco, CA: ICAAC, 2006.
for chronic bronchitis: A meta-analysis. Eur Respir J 29(6): 112737, 74. Soler N et al. Bronchial microbial patterns in severe exacerbations of
2007. chronic obstructive pulmonary disease (COPD) requiring mechanical
65. Wilson R et al. A comparison of gemioxacin and clarithromycin in ventilation. Am J Respir Crit Care Med 157: 1498505, 1998.
acute exacerbations of chronic bronchitis and long-term clinical out- 75. Stockley RA et al. Relationship of sputum color to nature and outpa-
comes. Clin Ther 24(4): 63952, 2002. tient managment of acute exacerbations of COPD. Chest 117: 1638
66. Wilson R et al. Short-term and long-term outcomes of moxioxacin 45, 2000.
compared to standard antibiotic treatment in acute exacerbations of 76. Soler N et al. Bronchoscopic validation of the signicance of sputum
chronic bronchitis. Chest 125(3): 95364, 2004. purulence in severe exacerbations of chronic obstructive pulmonary
67. Adams SG et al. Antibiotics are associated with lower relapse rates in out- disease. Thorax 62(1): 2935, 2007.
patients with acute exacerbations of COPD. Chest 117: 134552, 2000. 77. Stolz D et al. Antibiotic treatment of exacerbations of COPD: A rand-
68. Dewan NA et al. Acute exacerbation of COPD: Factors associated omized, controlled trial comparing procalcitonin-guidance with stand-
with poor treatment outcome. Chest 117(3): 66271, 2000. ard therapy. Chest 131(1): 919, 2007.
69. Wilson R et al. Antibiotic treatment and factors inuencing short 78. Martinez FJ, Curtis JL. Procalcitonin-guided antibiotic therapy in
and long term outcomes of acute exacerbations of chronic bronchitis. COPD exacerbations: Closer but not quite there. Chest 131(1): 12,
Thorax 61(4): 33742, 2006. 2007.
70. Ball P et al. Acute infective exacerbations of chronic bronchitis. Q J 79. Miravitlles M et al. Pharmacoeconomic evaluation of acute exacerba-
Med 88: 6168, 1995. tions of chronic bronchitis and COPD. Chest 121(5): 144955, 2002.

676
Long-Term Oxygen Therapy
54 CHAPTER

The use of oxygen as a therapeutic agent is gen- Arterial oxygen content is further deter-
erally thought to have began in the 1920s [1]. mined by
Bartolome R. Celli
Since then, much has been learned about the Division of Pulmonary and Critical Care,
eects of oxygen and many methods of deliv- Ca O2 (Hgb 1.39 % sat/100) Caritas St. Elizabeths Medical Center,
ery have been developed. In this chapter, we  (0.003 Pa O2 ) Tufts University, Boston, MA, USA
will review the known eects of chronic oxygen
therapy, its indications, and the various delivery where Hgb is blood hemoglobin concentra-
systems now available. tion in g/100 ml, 1.39 is the maximum amount
of oxygen (ml) that can combine with 1 g of
hemoglobin, % sat is the amount of oxygen
combined with hemoglobin divided by the
PATHOPHYSIOLOGY OF maximal amount possible, 0.003 is the amount
OXYGENATION of oxygen (ml) soluble in 100 ml of blood per
torr PaO2 in the absence of Hgb, and PaO2 is
the partial pressure of oxygen in arterial blood.
To understand the scientic rationale and Tissue hypoxia occurs when the amount of oxy-
guidelines concerning oxygen therapy, it is gen present cannot meet the metabolic needs.
necessary to know about the physiology of Inadequate oxygen transport is dependent
gas exchange, transportation of oxygen to tis- upon any of the above factors and is the most
sues, and the consequences of tissue hypoxia. common cause of tissue hypoxia. However,
Hypoxemia is dened as an abnormally low decreased oxygen utilization is another pos-
arterial oxygen tension PaO2. The physiologic sible cause of hypoxia. This is demonstrated by
causes of hypoxemia include a low inspired par- shunting at the capillary level in sepsis or more
tial pressure of oxygen (FiO2), abnormal venti- dramatically by mitochondrial poisoning sec-
  mismatch),
lationperfusion relationship ( V/Q ondary to certain substances such as cyanide.
decreased diusion capacity, alveolar hypoventi- The physiologic response to acute hypox-
lation, and right to left shunt. Oxygen therapy emia is to maintain oxygen transport (Fig. 54.1).
increases the FiO2 and is the primary treatment At a PaO2 below 55, ventilatory drive increases
for hypoxemia resulting from the rst three rapidly leading to a higher arterial PaO2 and
causes. The hypoxemia of alveolar hypoven- hypocapnia. Vascular beds supplying hypoxic tis-
tilation is best treated by increased ventilation sue vasodilate, resulting in tachycardia, increased
while a true shunt is by denition unresponsive cardiac output, and oxygen delivery. The lung
to hyperoxia. responds to hypoxemia by vasoconstriction to
Oxygen delivery (DO2) to the tissues is increase ventilationperfusion matching and the
dependent upon the arterial oxygen content PaO2. Subsequently, secretion of erythropoietin
(CaO2) and the cardiac output (Q t ) as illustrated results in erythrocytosis and improved oxygen
by the following equation: carrying capacity. These adaptations improve
oxygen delivery. Unfortunately, the short-term
benet of these responses may have a detri-
DO 2 Ca O 2 Q t (54.1) mental long-term eect. Prolonged pulmonary

677
Asthma and COPD: Basic Mechanisms and Clinical Management

Long-term effects Decreased PaO2 Short-term effects

Increased
Increased VE
O2 COB
Increased Increased
Pulmonary
V/Q matching PaO2
Malnutrition vasoconstriction
Increased Increased Increased Increased
PAP erythropoietin hematocrit arterial
O2 content
Increased Increased heart rate Increased
myocardial and stroke volume cardiac
work output

Cor pulmonale

Increased
oxygen FIG. 54.1 The short- and
Death delivery long-term physiologic
consequences of hypoxemia.

TABLE 54.1 Known effects of chronic oxygen therapy.


EFFECTS OF CHRONIC OXYGEN THERAPY
Survival

Increased in hypoxemic patients with COPD after receiving continuous O2. Survival
Pulmonary hemodynamics
Decreases pulmonary artery pressure and pulmonary vascular resistance. Although the cause of death in patients with hypoxic cor
Increases stroke volume. pulmonale is uncertain, the factors predicting survival in
patients on long-term oxygen have been investigated. In
Exercise general, variables reecting worse severity of COPD, such as
Improves exercise endurance.
reduction of PaO2 or increased PCO2, [69] lower FEV1 [6,
Improves ventilatory muscle function.
9], and elevated mean pulmonary artery pressure (PAP) [9],
Decreases minute ventilation.
correlate with survival. On the other hand, the eect of tran-
sient hypoxemia measured by oxygen desaturation appears to
Oxygen cost of ventilation have limited impact on survival [10]. In the most hypoxemic
patients as determined by arterial hypoxemia, the most impor-
Decreases resting oxygen cost of breathing.
tant evidence supporting the use of chronic oxygen has been
Reduces oxygen cost of ventilation during carbon dioxide induced
derived from studies investigating its eect on survival [11].
hyperventilation.
In 1970, Ne and Petty [12] found a 3040%
Neuropsychology
decrease in mortality in their severely hypoxic patients on
continuous oxygen when they were compared with those
Improves neuropsychologic performance. in the literature. Subsequently, two large controlled trials
studied the eect of oxygen on survival in COPD [4, 5].
Sleep
The British Medical Research Council Study randomized
Improves sleep quality. patients to 15 h of continuous oxygen each day compared
with no oxygen. During the 5-year follow-up, 19 of 42
oxygen therapy patients compared with 30 of 45 controls
vasoconstriction and increased cardiac output can result in died. The Nocturnal Oxygen Therapy Trial (NOTT) ran-
pulmonary hypertension (PH), right ventricular failure, and domly assigned patients to either 12 or 24 h of oxygen per
decreased survival [25]. In addition, the increased minute day. After 26 months, the mortality of the continuous group
ventilation and increased oxygen cost of breathing (O2 (mean 19 h/day) was one-half that of the 12-h group. Taken
COB) may contribute to chronic malnutrition [6]. To inter- together, these studies found survival to improve propor-
rupt this sequence of events, oxygen therapy has become tionally to the number of hours of supplemental oxygen per
important in hypoxemic patients [4, 5, 7, 8]. We shall day. As a result, the current recommendation for hypoxemic
review what is known about the long-term eects of oxygen patients (PaO2  55 mm Hg or SaO2  88%) is continu-
therapy with particular attention to COPD because this is ous 24 h/day oxygen therapy. In addition, patients with a
the only disease in which chronic oxygen therapy has been PaO2 of 5559 mm Hg or a SaO2  89% in the presence
studied in detail (Table 54.1). of cor pulmonale or polycythemia also qualify for long-term

678
Long-Term Oxygen Therapy 54
continuous oxygen. The ndings of these trials have been and oxygen delivery. They propose that the eect of recur-
conrmed in the routine clinical setting [13]. rent oxygen withdrawal may be responsible for the worse
Because of the lifestyle changes and economic impli- prognosis of intermittent oxygen therapy.
cations of chronic oxygen therapy, patients must meet cer- In the longest of all the observational therapeutical
tain criteria for longer-term prescription. First, patients oxygen trials, Zelinski and coworkers [7] evaluated pul-
must be stable, on optimal medical therapy and not smok- monary hemodynamics at 2-year intervals for 6 years in a
ing. In addition, patients must be observed for at least 3 group of 12 patients with hypoxemic COPD. The PAP
months to document persistent hypoxemia requiring long- which was 277 torr at entry, decreased to 214 at 2 years,
term oxygen. During both the NOTT study [4] and a mul- to then return to stable values of 267 and 266 torr at 4
ticenter French study [14], approximately 40% of patients and 6 years, respectively. This was observed in spite of fur-
had improved enough after 1 month to no longer qualify for ther deterioration of gas exchange with worsening of room
chronic oxygen. Consequently, patients should be reassessed air PaO2 and PaCO2. This study lends support to the con-
13 months after initiation of oxygen therapy to determine cept that oxygen supplementation to hypoxemic patients
continued need for its use. halts progression of PAH.
Many patients continue to worsen despite long-
term oxygen. Early identication of those patients who
Pulmonary hemodynamics are unlikely to benet from oxygen would avoid the cost
and inconvenience of oxygen in these patients. As a result,
Although the reason for increased survival with oxygen is various investigators have attempted to correlate an acute
not clear, there is evidence that O2 can improve pulmonary hemodynamic response to oxygen with long-term survival.
hemodynamics and lead to reduced cardiac work and greater In most studies, acute oxygen delivery (30 min to 24 h)
oxygen delivery. Initially, uncontrolled studies in hypoxemic decreases the PAP in a minority of patients [2, 18, 19].
patients suggested that continuous oxygen for 68 weeks Conversely, others have shown 28% oxygen to reduce PAP
could reduce pulmonary artery hypertension (PAH) [15, 5 mm Hg in greater than half of their patients [20, 21]
16]. Subsequently, however, the controlled NOTT and and this acute response to oxygen predicted 2-year survival
MRC studies revealed no denite hemodynamic improve- in patients with COPD and cor pulmonale [22]. However,
ment [4, 5]. In the MRC trial, the mean PAP remained Sliwinski et al. [19] have found little correlation between
stable in patients receiving oxygen 15 h/day, but increased acute response to oxygen and long-term survival. They pro-
signicantly in the control group. In NOTT, there was only pose that the dierence in results of these studies may be
a slight decrease in pulmonary vascular resistance (PVR) attributable to the lack of a pre-entry period in Ashutoshs
after 6 months on continuous oxygen while the nocturnal study, which may have allowed inclusion of patients who
oxygen group had a minimal increase in PVR. However, the were not stable. Consequently, these patients may have had
results of both these studies may have been awed by the acute hypoxic vasoconstriction superimposed upon the more
fact that each was devoted mainly to prognosis, follow-up irreversible anatomic changes. In their study, responders had
was short, and not all patients were evaluated. lower mean PAP (26 mm Hg) after oxygen compared with
Some studies have specically examined the eect the nonresponders (33 mm Hg) suggesting they could have
of oxygen therapy on PAH. Weitzenblum and colleagues had less severe baseline PH which might have accounted
[3] performed right heart catheterizations on an aver- for their better prognosis.
age of 41 months prior to oxygen, just before starting oxy- Patients who have an acute hemodynamic response to
gen, and 31 months after oxygen in 16 severely hypoxemic oxygen may have structural dierences in their pulmonary
COPD patients on oxygen therapy. Before oxygen therapy, vasculature as compared with those who do not improve.
there was a mean yearly increase in PAP of 1.472.3 mm However, an autopsy study examining the pulmonary arter-
Hg. Following oxygen, PH improved in 12 of 16 patients as ies of patients who died during the NOTT study found no
demonstrated by an annual decrease in PAP of 2.154.4 mm dierence in the vascular structure of those who responded
Hg. Complete normalization of PAP rarely occurred, but the to oxygen and those who did not [23]. In addition, these
changes in PAP were related to dierences in PVR. authors found structural alterations of the muscular pulmo-
Further analysis of the NOTT data showed that the nary arteries in patients with PAH secondary to COPD,
continuous oxygen therapy group had signicant improve- but these changes did not correlate with the degree of PAH.
ments in resting and exercise mean PAP, PVR, and stroke Because determination of PAP is invasive, nonin-
volume index after 6 months [2]. In addition, the noctur- vasive methods have been assessed to predict response to
nal oxygen therapy group showed stable hemodynamic oxygen therapy. Ashutosh and Dunsky [21] studied exercise
variables. However, the absolute dierences between the V O2 max and found a V O2 max 6.5 ml/min/kg to pre-
two groups were not signicant. Despite this, the authors dict a fall in PAP 5 mm Hg and 3-year survival on oxy-
suggested that further follow-up may show hemodynamic gen. Attempts to show an acute or chronic benet of oxygen
improvement as a factor altering the longevity between the on RVEF (right ventricular ejection fraction) [24, 25], and
two groups. One investigator has suggested that the reason a change in RVEF on oxygen to predict a change in PAP
continuous oxygen therapy improves survival may be due to [21, 25] or survival [21] have been disappointing. Therefore,
the detrimental eect of oxygen withdrawal from patients the response of RVEF to oxygen does not appear to predict
on long-term therapy [16]. Selinger and colleagues [17] a benecial eect of this therapy.
showed that removal of oxygen caused a 31% increase in The possibility that long-term oxygen therapy could
PVR index coupled with a decrease in stroke volume index benet patients with a lesser degree of resting hypoxemia

679
Asthma and COPD: Basic Mechanisms and Clinical Management

(PaO2 between 56 and 69 torr) was explored by Gorecka et al. ventilation and respiratory rate for a given workload [35,
[26]. They prospectively evaluated patients with moderate 38]. Indeed, van Helvoort and colleagues have shown that
hypoxemia and observed no dierence in mortality between administration of supplemental oxygen to patients with
treated and untreated patients. This lack of benecial eect COPD even if the levels of oxygen are not critical may
is further highlighted by the multicenter trial reported by ameliorate the generation of oxidative stress and inamma-
Chaouet and coworkers [27] who after 2 years of therapy tory burst observed during exercise in these patients [46].
observed no eect on survival or pulmonary artery hemo- Oxygen has been found to improve ventilatory muscle
dynamics in a cohort of moderate hypoxemic patients rand- function during exercise, by postponing the onset of fatigue
omized to supplemental oxygen or routine therapy. as demonstrated by a delay in the appearance of abdominal
Most of the studies evaluating oxygenation and sur- paradox and a slower fall in the high to low frequency of
vival have centered on the actual value of PaO2 and to some the diaphragm electromyogram power spectral density [38].
degree on the oxygen saturation. Recent evidence shows Criner and Celli [47] found that exercising COPD patients
that the level of hemoglobin may be as important in pre- with oxygen resulted in the diaphragm performing more
dicting outcome in patients with COPD. The long held ventilatory work which may prevent overloading of the
belief that most patients with COPD develop compensa- accessory muscles and contribute to the decrease in dyspnea.
tory polycythemia has proven to not be the case as those Finally, others have suggested that supplemental oxygen may
large series show that anemia is highly prevalent in those decrease dyspnea and improve endurance by directly reduc-
patients and is associated not only with decreased exercise ing chemoreceptor activity from the carotid body [39, 48].
capacity but also with impaired survival [28, 29]. Currently, qualication for oxygen during exercise requires
In summary, long-term oxygen has benecial eects documentation of a PaO2 less than or equal to 55 mm Hg or
on survival and PH in patients with COPD and signi- an SaO2  88 [49, 50]. A helpful clue to predicting which
cant hypoxemia. Currently, most studies do not support the patients will desaturate with exercise is the diusing capac-
ability of acute hemodynamic response to oxygen measured ity. Owens and colleagues [51] found a diusing capac-
either invasively or noninvasively to predict improvements ity above 55% predicted to be 100% specic in excluding
in PAP, pulmonary vascular changes, or survival. Therefore, desaturation.
long-term oxygen is currently administered to all signi-
cantly hypoxemic patients (PaO2  55 torr), since some
benet cannot be ruled out and other therapeutic options are Oxygen cost of ventilation
limited. There is no evidence to support the administration
of long-term oxygen therapy to patients with mild or transi- Beyond the eect of oxygen on arterial PaO2, there is evi-
tory hypoxemia. The value of correcting the hemoglobin in dence that oxygen may improve dyspnea by decreasing air-
patients with COPD and anemia has not been explored. way resistance and work of breathing (WOB). Astin and
Penman [52] studied 18 COPD patients and found a sig-
nicant association between airway resistance and PaO2.
Exercise Administration of 30% oxygen for 20 min to these patients
resulted in an average reduction of airway resistance of 20%.
Ventilatory, rather than circulatory, factors limit maxi- Subsequently, it was also found that 30% oxygen improved
mum exercise performance in most patients with COPD. ow rates in mildly hypoxemic patients (mean PaO2 61
Performance is most closely related to airow limitation torr) [53]. Furthermore, atropine caused a similar increase
[30]. Other investigators have suggested that the ventilatory in ow rates that aborted the eect of oxygen. This led the
muscles may contribute to exercise limitation in COPD [31, authors to conclude that hypoxemia-related bronchocon-
32]. The process involves hyperination secondary to inabil- striction may be mediated by vagal tone as had been previ-
ity to exhale trapped air which in turn leads to more inspir- ously suggested [52, 54].
atory muscle dysfunction at an unfavorable length. This In addition to its eects on airways resistance, oxy-
muscles have to generate pressures, which are close to their gen via a face mask or transtracheal ow in patients with
maximum capacity and this may lead to fatigue as well as COPD has also been found to decrease minute ventilation
a lack of appropriate mechanical response to the increased [55, 56]. As a consequence of these eects, oxygen may
central drive [33]. The resulting dyspnea leads to premature lead to a decrease in the WOB or the O2 COB and thereby
termination of exercise. improve dyspnea. Astin [57] studied the eect of 30% oxy-
Oxygen therapy has been shown to improve exer- gen on 15 patients with COPD and found a decrease in
cise endurance in many studies. Various investigators have WOB in 10 patients and an increase in 5 patients. Mannix
found oxygen to increase with distance walked [34, 35], et al. [58] calculated the COB induced by breathing 7%
time on a treadmill [36, 37], and time on a cycle ergometer carbon dioxide. They found 30% oxygen to decrease the O2
[38, 39]. The precise mechanisms resulting in improvement COB by 42% in both COPD patients and controls. In the
are unknown [40], but a number have been suggested. In COPD patients, the decrease in O2 COB was only due to
hypoxemic patients and those who desaturate with exercise, a smaller increment in VO2 for a given level of ventilation,
oxygen therapy results in greater oxygen delivery and uti- while controls had both a lower VO2 and a VE. They con-
lization by exercising muscles [4145]. However, increased cluded that supplemental oxygen decreases the O2 demand
oxygen saturation is not always predictive of improved exer- of the respiratory muscles. Benditt and coworkers [59]
cise performance [37, 38] and other factors may contribute observed a decrease in the oxygen cost of breathing with
to this. Some studies have shown oxygen to reduce minute both transtracheal air and oxygen, but the eect was greater

680
Long-Term Oxygen Therapy 54
with oxygen. Because most patients receive long-term oxy- ATP generation and neuron function has been disproved
gen by a nasal cannula, the eect of nasal ow oxygen on [70]. Some studies suggest that hypoxia can result in reduced
the oxygen cost of breathing was recently assessed [60]. At synthesis of acetylcholine from labeled precursors [71],
high ows (O2 5 l/min), oxygen signicantly decreased the which is interesting, since there is growing evidence to sup-
O2 COB while compressed air did not. These initial studies port the role of acetylcholine in memory and learning [72].
have been recently conrmed with a study that shows that
in a dose-dependent ways, supplemental oxygen decreases
breathing frequency and dynamic hyperination thereby Sleep
decreasing WOB [61]. The same group has shown that the
administration of oxygen may help better train patients In the original description of rapid eye movement (REM)
undergoing pulmonary rehabilitation [62]. sleep in 1953, it was observed that breathing became more
In summary, there is overwhelming evidence that variable during this stage of sleep [73]. Subsequently, it was
supplemental oxygen increases exercise capacity in hypox- found that decreased chest movement and oxygen satura-
emic patients with COPD and the eect may occur even in tion occurred in this sleep stage [74]. Other groups also
patients with mild hypoxemia. The long-term impact of this demonstrated worsening hypoxemia and carbon diox-
eect is less clear. ide retention during sleep [75, 76]. Koo and colleagues
[77] correlated arterial blood gas tension with sleep stage
in COPD patients and found that PaO2 decreased shortly
Neuropsychologic effects after sleep from 64 to 58 mm Hg during non-REM sleep.
Upon entering REM sleep, PaO2 fell further to 50 and the
Beginning in the 1930s and 1940s, aviation research demon- PaCO2 rose from 48 to 58 mm Hg. These ndings were later
strated the acute eects of mild to severe hypoxia on healthy conrmed by many others [7882].
young men [63]. These studies found that even mild hypoxia The mechanisms of hypoxemia during sleep in
(PaO2 4560) could impair judgment, learning, and short- COPD include hypoventilation, a reduction in functional
term memory in normal subjects. To investigate the eects residual capacity (FRC), and alterations in ventilation per-
of chronic hypoxemia, research has focused on the neurobe- fusion matching [83] The major cause of hypoxemia dur-
havioral eects of hypoxemia in patients with COPD. ing REM sleep is hypoventilation, which appears related
Early work by Krop and colleagues [64] demonstrated to rapid shallow breathing [83, 84] and long episodes of
poorer neuropsychologic performance in a group of COPD hypopneas, but not actual apneas [81]. In addition, both
patients with a PaO2  55 mm Hg. Administration of the hypoxic [85, 86] and hypercapnic [87, 88] ventilatory
continuous oxygen at 2 l/min by nasal cannula for 4 weeks responses are diminished. The cause of REM sleep hypov-
resulted in signicant improvement in 8 of 10 neuropsy- entilation has not been fully determined, but appears related
chologic tests. Because this study and another [65] only to altered brain stem function during phasic neuronal activ-
looked at a small group of patients without a stabilization ity [89]. During REM sleep, the ribcage contribution to
period, two large multicenter studies in the United States ventilation decreases by 1834% [90, 91], as a consequence
and Canada examined the neuropsychologic consequences of hypotonia of the intercostal muscles and decreased activ-
of hypoxemia [66, 67]. The US NOTT studied 203 patients ity of accessory muscles [92]. This may be particularly
with a mean age of 64 years and PaO2 of 51 torr and found important in patients with COPD who greatly depend on
that 42% of patients had moderate to severe impairment of accessory muscles for breathing [93, 94]. Though it is gen-
cerebral function [66]. The Canadian IPPB Trial examined erally believed that hypoventilation during REM sleep will
100 patients with less hypoxemia (mean PaO2 66 torr) which result in ventilationperfusion mismatch [83], its relative
resulted in more selective impairments [67]. The results importance with regard to REM hypoxemia is unknown.
of both these studies were combined, demonstrating an Although some patients with COPD have sleep apnea,
increase in the rate of decits from 27% in mild hypoxemia there is no evidence that this occurrence is more common
(PaO2  60) to 61% in severe hypoxemia (PaO2  50) [68]. than would be expected by chance alone [83].
The NOTT study has evaluated the eect of con- A few studies have measured pulmonary hemody-
tinuous (19 h/day) and nocturnal oxygen on neuropsycho- namics during sleep in COPD and found an increase in
logic functioning and life quality in 150 COPD patients. PAP with acute desaturation [9599]. In addition, some
The results show an improvement after 6 months in both studies support an association between nocturnal desatu-
groups in general alertness, motor speed, and hand grip ration and PH in COPD patients who have a daytime
[69]. Despite these benets, patients had no change in PaO2  60 mm Hg [98, 99]. Both studies found signi-
emotional status or quality of life. Although improvements cantly higher PAP in the patients who desaturated as com-
after 6 months were no greater on continuous as compared pared with the non-desaturators.
with nocturnal oxygen, a subgroup of patients followed for Patients with COPD have poor sleep quality as com-
12 months showed signicantly better neuropsychologic pared with age-matched controls [100101] and arousals
function on continuous oxygen. are frequent during periods of desaturation [102]. Whether
The poor neuropsychologic performance in hypoxemic oxygen therapy improves sleep quality in hypoxemic
COPD patients has been explained by weakness, fatigue, and patients is debatable, since various studies have shown con-
depression. However, there is increasing evidence that brain icting results [100, 103]. Some studies have found fewer
hypoxia itself is responsible for the impaired function. The hypoxemic episodes as well as increased total sleep and
conventional belief that mild cerebral hypoxia could impair REM sleep with oxygen [100, 104].

681
Asthma and COPD: Basic Mechanisms and Clinical Management

Although mean nocturnal oxygen saturation (SaO2) TABLE 54.2 Indications for chronic oxygen therapy.
and lowest nocturnal SaO2 have been found to correlate with
survival, one study found these parameters to have no prog- Continuous oxygen
nostic value over vital capacity or awake SaO2 [104, 105]. Resting PaO2  55 mm Hg or SaO2  88%
Furthermore, patients with greater nocturnal hypoxemia than Resting PaO2 5659 mm Hg or SaO2  89% in the presence of the
would be predicted, had no dierence in survival. In addi- following:
tion, the clinical characteristics of patients with COPD who 1 Dependent edema suggesting congestive heart failure
had desaturation during 24 h monitoring were not dier- 2 P pulmonale on EKG (P wave greater than 3 mm in standard leads
ent from that of patients who did not desaturate during the II, III, or AVF)
same period [106]. Fletchers group retrospectively compared 3 Erythrocythemia (hematocrit 56%)
survival in COPD patients with a daytime PaO2  60 mm Resting PaO2  59 mm Hg or SaO2  89%
Hg with and without O2 desaturation at night [107]. They Only reimbursable with additional documentation justifying the
found an improved survival in those patients without noc- oxygen prescription and a summary of more conservative therapy
turnal desaturation. There was also a trend toward improved which has failed
survival with nocturnal oxygen in those patients who did
desaturate at night. These authors attribute the dierence in Noncontinuous oxygen
survival in their study and one by Connaughton et al. [105] Must specify the oxygen flow rate and the number of hours/day
to the worse daytime PaO2 of the patients in that study During exercise, PaO2  55 mm Hg or an SaO2  88% with low level
(mean PaO2 5310 mm Hg). They proposed that as the dis- exertion
ease becomes more advanced, the daytime SaO2 may become During sleep, PaO2  55 mm Hg or an SaO2 88%, with associated
a strong predictor, while sleep SaO2 correlates less well with complications such as pulmonary hypertension, daytime somnolence,
survival. However, the recent report by Chaouet et al. [10] and cardiac arrhythmias
suggests that oxygen supplementation at night to mildly
hypoxemic patients has a limited, if any, benecial eect in
pulmonary hemodynamics or survival.
Presently, patients who are hypoxemic while awake
TABLE 54.3 Modes of oxygen delivery.
should also be prescribed oxygen during sleep. In addition,
patients who are well saturated during wakefulness, but System Advantages Disadvantages
desaturate at night can qualify for oxygen if they have com-
plications attributable to sleep hypoxemia, such as PH, day-
Gas Low cost Heavy weight
time somnolence, or cardiac arrhythmias [50, 108].
Although it seems intuitive that patients who mani- Wide availability Difficult to refill
fest oxygen desaturation during sleep should have a worse Fair portability Short supply time
prognosis, this has not been conrmed in the few prospec-
tives studies that have been conducted. However, it has been Liquid Light weight High cost
accepted that patients with prolonged destauration at night
Very portable Vendor incompatibility
without sleep apnea as the cause, be provided with supple-
Easy refill Pressure venting
mental oxygen.
Concentrator Low cost Heavy weight
Good availability Least portable
OXYGEN PRESCRIPTION

Currently, there are strict guidelines for oxygen prescrip- Table 54.3 summarizes the advantages and disadvan-
tion based upon resting hypoxemia (PaO2  55 mm Hg tages of each mode of therapy comparing weight, cost, port-
or SaO2  88) and hypoxemia induced by sleep or exercise ability, ease of rell, and availability. Because home oxygen
(Table 54.2). is supplied under a xed reimbursement policy regardless of
Once a patient qualies for long-term oxygen, pre- the system used [104], oxygen vendors will attempt to pro-
scription requires completion of the Certicate of Medical vide the least expensive system.
Necessity for Home Oxygen Therapy form in the United
States. This form should be completed before a patient
leaves the hospital to ensure inclusion of all information Oxygen concentrator
such as patient data, reason for oxygen, blood gases, type of
system, and liter ow at rest, exercise, and sleep. Most patients will require a stationary source of oxygen
Long-term oxygen administration can be accom- which is usually provided by an oxygen concentrator. Since
plished with: concentrators are relatively inexpensive and require less fre-
oxygen concentrator quent home visits than liquid oxygen, they have become the
system of choice for suppliers. These electrically powered
compressed gas devices utilize a molecular sieve to separate oxygen from air
liquid oxygen. resulting in delivery of oxygen to the patient, while nitrogen

682
Long-Term Oxygen Therapy 54
is returned to the atmosphere. The typical sieve achieves does not occur during late or dead space inspiration and
oxygen purity of 97% at low ows and 94% at higher ows. exhalation [111]. Consequently, only oxygen owing during
However, due to their voltage requirement and their weight, early inspiration is available to the patient, while the remain-
they are primarily a xed source of oxygen. Consequently, der is wasted into the environment. To improve eciency
patients need either compressed gas or liquid oxygen as an and mobility, oxygen-conserving devices have been designed
ambulatory source of oxygen. New, more portable devices to deliver oxygen during early inhalation. These devices
should improve availability of these units. include reservoir nasal cannulas, transtracheal catheters, and
electronic oxygen demand devices. In general, these three
devices each result in oxygen savings of two to four times a
Compressed gas oxygen conventional nasal cannula. For a more detailed review the
reader may refer to Ref. [111].
Compressed gas oxygen has been provided for many years in
high pressure metal or aluminum cylinders. These contain-
ers vary in size weighing 90.8, 7.2, 4, and 1.8 kg and provide Reservoir cannula
O2 at 2 l/min ow for 2.4 days, 5.2 h, 2 h and 1.2 h, respec-
tively. The smaller cylinders can be relled but the process is The reservoir cannula was the rst method of oxygen con-
inecient and potentially hazardous. The major advantages servation to be used [111, 112]. This device stores 20 ml
of compressed gas are its low cost and wide availability. Its of oxygen in a reservoir during expiration and delivers an
disadvantages are the weight of the cylinders, the diculty oxygen bolus at the onset of inspiration. The two available
in relling and the short oxygen supply time. reservoirs are shaped as either a mustache attached to nasal
prongs or a pendant placed over the anterior chest wall. The
mustache reservoir was developed rst, but because of dis-
Liquid oxygen satisfaction with its high visible location, the pendant was
designed to be hidden by clothing [113]. Although this
Liquid oxygen is stored at very cold temperatures which device has better cosmesis, it is still more noticeable than
reduces its volume to less than 1% of the volume of atmos- the standard nasal cannula.
pheric oxygen. Stationary units weigh 140 lbs and can pro-
vide several days of continuous oxygen at 2 l/min ow. There
are portable containers weighing as low as 4 lbs that will last Electronic demand devices
for up to 12 h at 2 l/min ow. Compared with compressed
gas, an equivalent weight container of liquid oxygen is more Electronic demand devices sense the beginning of inspiration
portable and easier to rell, its liabilities include a higher and deliver a pulse of oxygen during early inhalation [114,
cost, manufacturer incompatibility, and the need for pres- 115]. There are several units available with many similarities
sure relief venting which wastes unused oxygen. and dierences. Most devices are relatively inexpensive and
Deciding on which portable system to prescribe have built-in rechargeable batteries and alarms [116]. Some
depends upon the activity level of the patient. For patients systems can be switched manually from demand to continu-
who are more active, liquid oxygen is the ambulatory system ous ow [116], while others automatically change to continu-
of choice since the oxygen lasts longer and the canister is ous ow if an inspiratory signal is not sensed [117]. Another
easier to rell and to carry. Lock et al. [109] compared liq- demand valve is able to maintain constant oxygen delivery
uid and gaseous oxygen and found patients would use liquid by adjusting the pulse size depending on the respiratory rate.
oxygen more hours per week (23.5 versus 10 h) and leave A recent comparison of standard nasal cannula, reservoir
their house more hours per week (19.5 versus 15.5 h). nasal cannula, and a demand ow device in 15 patients with
A recently conducted study to evaluate whether port- COPD found no dierence in exercise tolerance among the
able oxygen improves quality of life has been disappointing three systems. However, oxygen saturation tended to be lower
with no benet shown in the largest of them [110]. during the exercise using the demand ow device [118].

Transtracheal catheters
OXYGEN ADMINISTRATION DEVICES
Transtracheal catheters improve oxygen delivery by bypass-
ing anatomic dead space and utilizing the upper airways
Nasal cannula as a reservoir during end expiration [119]. Heimlich [120]
developed the rst catheter after experiments in dogs and
Patients with COPD and other chronic lung diseases usu- demonstrated an increased PaO2 as a catheter was placed
ally receive continuous ow of oxygen via a nasal cannula. more distally. In addition to increased oxygen savings, other
Delivery at a ow of 2 l/min increases the FiO2 to approxi- advantages of TTO include its relative inconspicuousness,
mately 27%, which provides an adequate O2 saturation in the lack of nasal, auricular, or facial skin irritation, its infre-
the majority of patients. Although this method is eective, it quent displacement during exercise and sleep [120]. In addi-
is quite inecient. During the respiratory cycle, alveolar gas tion, there are patients with refractory hypoxemia on a nasal
exchange is limited to early inhalation which accounts for cannula that have been successfully oxygenated on TTO
only one-sixth of the cycle. Conversely, alveolar ventilation [121124]. Patient acceptance of transtracheal catheters has

683
Asthma and COPD: Basic Mechanisms and Clinical Management

been variable with rates ranging from 50% to 96% during Use of regression formulae. Regression equations
9- to 20-month periods [120125]. oer the opportunity to compare a patient with a
group of patients with similar clinical characteristics
who have been previously studied during exposure to
Complications hypoxia. While regression equations may provide a
more physiological basis for the eects of high altitude
Complications of TTO vary depending on the system than the HIT, the regression approach does not assess
implemented and whether the acute or late phase of treat- the individuals susceptibility to the development of
ment is examined. The most common procedure-related symptoms or electrocardiographic changes during
problems include subcutaneous emphysema, bronchospasm, hypoxia.
and paroxysmal coughing [123, 126]. Frequently encoun-
tered late complications and sequelae include dislodged The A-aO2 gradient and the a-AO2 ratio generally have
catheters, stomal infections, and symptomatic mucous balls. no advantages over regression equations. It is currently recom-
Mucous balls result from oxygen drying the sputum and mended that the PaO2 during air travel should be maintained
causing it to adhere to the catheter. The greater frequency of above 50 mm Hg [50]. While 23 l of oxygen by nasal can-
mucous balls seen in 1025% patients with the Scoop cath- nula will replace the inspired oxygen partial pressure lost at
eter (Transtracheal Systems, Denver, CO, USA) appears to 8000 ft compared with sea level, lesser increments of oxygen
be the result of its larger surface area and the presence of will maintain the PaO2 above 50 mm Hg in many patients.
side holes which allow accumulation of secretions. Some COPD patients receiving continuous oxygen at
reports show life threatening [127, 128] and even fatal air- home will require supplementation during air ight. Such
way obstruction due to mucous balls [129]. Many other patients should receive greater oxygen supplementation
uncommon complications have been described including during the ight than at sea level. Increments equivalent to
broken catheter tips, hemoptysis, keloid formation, hoarse- 12 ls of oxygen by nasal cannula during ight should suf-
ness, and cardiac arrhythmias. After an initial burst of ce for most patients. Patients will also require additional
enthusiasm, the number of transtracheal earlier initiations oxygen supplementation if the elevation at the destination
has decreased primarily due to the need for continuous is signicantly greater than at home. The Federal Aviation
supervision and relatively poor long-term compliance. Administration requires a physicians statement of oxygen
need in order for a patient to receive continuous oxygen
during ight. There is no uniform airline request form, so
each airline must be contacted by the patient to determine
OXYGEN AND AIR TRAVEL what is required. As the airlines do not provide oxygen for
ground use in the airline terminal, patients who require
continuous oxygen should be advised to make plans for such
Commercial airline travel exposes passengers to hypobaric locations. The American Lung Association provides patient
hypoxia since aircraft cabins are not routinely pressurized to education materials for individuals who travel with oxygen
sea level [130]. In patients who have compensated COPD entitled Airline Travel with Oxygen.
at sea level, lowering the partial pressure of oxygen in the Many ambulatory COPD patients not receiving oxy-
aircraft cabin can produce severe hypoxemia. Physical exer- gen at home can tolerate PO2 values below 50 mm Hg for
tion during the ight can increase the risk of an exacerba- brief periods of time without serious consequences. Stable
tion of symptoms. The proportion of patients suering from COPD patients without comorbid disease who have previ-
complications during air travel is not well known. ously traveled without incident and who are currently clini-
Aircraft are usually pressurized to between 5000 and cally stable compared with their previous air travel, may be
7000 ft (15002100 m). For the preight evaluation of most advised to travel by air as there is little risk to them.
patients, clinicians should consider 8000 ft (2438 m) of alti-
tude above sea levels as realistic worse case scenario.
Preight assessment can be accomplished with the
following elements:
SUMMARY
Estimation of the expected degree of hypoxemia at altitude.
Identication of comorbid disease conditions.
Oxygen therapy has become very important in the treat-
Provision of an oxygen prescription if necessary. ment of severe COPD. Because long-term oxygen improves
survival in hypoxemic COPD, patients with a daytime
Documentation of the recent clinical condition
PaO2  55 mm Hg or SaO2  88% qualify for continuous
and laboratory tests, particularly if the patient is traveling
oxygen therapy. Other benets of oxygen include better exer-
abroad, and counseling are also desirable elements of the
cise tolerance, decreased dyspnea, and improvements in neu-
preight patient care [50, 118].
ropsychological performance. In addition, there appear to be
The two means of estimating the degree of hypoxia at
benecial eects on pulmonary hemodynamics, sleep qual-
altitude are:
ity, reduced minute ventilation, and WOB. The consequences
Hypoxia inhalation test (HIT), which is not performed of exercise induced and nocturnal desaturation are not
in many clinical laboratories in the United States and is absolutely known. Clinicians should be aware of air travel-
not recommended for routine use. induced hypoxemia in COPD and be able to identify those

684
Long-Term Oxygen Therapy 54
who need oxygen in ight. Once a patient meets the crite- function in patients with chronic obstructive airways disease. Clin Sci
ria for oxygen prescription, the physician must complete the 26: 13343, 1967.
certicate of Medical Necessity Form specifying the indica- 19. Sliwinski P, Hawrylkiewicz I, Gorecka D, Zielinski J. Acute eects
of oxygen on pulmonary arterial pressure does not predict survival on
tion for oxygen, type of system and liter ow at rest, exercise
long-term oxygen therapy in patients with chronic obstructive pulmo-
and sleep. Patients should be reevaluated at 1 to 3 months to nary disease. Am Rev Respir Dis 146: 66569, 1992.
document a persistent need for oxygen therapy. Occasionally, 20. Anthonisen NR. Home oxygen therapy in chronic obstructive pulmo-
patients should be considered for oxygen-conserving devices nary disease. Clin Chest Med 7: 67378, 1986.
to decrease oxygen ow, cost and to improve mobility. 21. Ashutosh K, Dunsky M. Noninvasive tests for the responsiveness of
pulmonary hypertension to oxygen. Prediction of survival in patients
with chronic obstructive lung disease and cor pulmonale. Chest 92:
39399, 1987.
References 22. Ashutosh K, Mead G, Dunsky M. Early eects of oxygen administra-
tion and prognosis in chronic obstructive pulmonary disease and cor
1. Barach AL. The therapeutic uses of oxygen. JAMA 79: 69398, 1922. pulmonale. Am Rev Respir Dis 127: 399404, 1983.
2. Timms RM, Khaja FU, Williams GW. Nocturnal oxygen therapy trial 23. Wright JL, Petty T, Thurlbeck WM. Analysis of the structure of the
group. Hemodynamic response to oxygen therapy in chronic obstruc- muscular pulmonary arteries in patients with pulmonary hypertension
tive pulmonary disease. Ann Intern Med 102: 2936, 1985. and COPD: National Institutes of Health Nocturnal Oxygen Therapy
3. Weitzenblum E, Sautegeau A, Ehrhart M, Mammosser M, Pelletier A. Trial. Lung 170: 10924, 1992.
Long-term oxygen therapy can reverse the progression of pulmonary 24. Macnee W, Wathen CG, Flenley DC, Muir AD. The eects of con-
hypertension in patients with chronic obstructive pulmonary disease. trolled oxygen therapy on ventricular function in patients with stable
Am Rev Respir Dis 131: 49398, 1985. and decompensated cor pulmonale. Am Rev Respir Dis 137: 128995,
4. Nocturnal Oxygen Therapy Trial Group. Continuous or nocturnal oxy- 1988.
gen therapy in hypoxemic chronic obstructive lung disease. Ann Intern 25. Morrison DA, Henry R, Goldman S. Preliminary study of the eects
Med 93: 39198, 1980. of low ow oxygen on oxygen delivery and right ventricular function in
5. Medical Research Council Working Party. Long-term domiciliary oxy- chronic lung disease. Am Rev Respir Dis 133: 39095, 1986.
gen therapy in chronic hypoxic cor pulmonale complicating chronic 26. Gorecka D, Gorzelak K, Siliwinski P, Tobiasz M, Zielinski J. Eects of
bronchitis and emphysema. Lancet 1: 68186, 1981. long-term oxygen therapy on survival in patients with chronic obstruc-
6. Donahue M, Rogers RM, Wilson DO, Pennock BE. Oxygen con- tive pulmonary disease with moderate hypoxemia. Thorax 52: 67479,
sumption of the respiratory muscles in normal and in malnourished 1997.
patients with chronic obstructive pulmonary disease. Am Rev Respir 27. Chouet A, Weitzenblum E, Kessler R et al. A randomized trial of
Dis 140: 38391, 1989. nocturnal oxygen therapy in chronic obstructive pulmonary disease
7. Zelinski J, Tobiasz M, Hawrylkiewicz I, Sliwnski P, Palasiewicz G. patients. Eur Respir J 14: 10028, 1999.
Hemodynamics in COPD patients: A 6-year prospective study. Chest 28. Chambellan A, Chailleux E, Similowski T. Prognostic value of the
113: 6570, 1998. hematocrit in patients with severe COPD receiving long-term oxygen
8. Cooper C, Howard P. An analysis of sequential physiologic changes therapy. Chest 128: 12018, 2005.
in hypoxic cor pulmonale during long-term oxygen therapy. Chest 100: 29. Cote C, Zilberberg MD, Mody SH, Dordelly LJ, Celli B. Haemoglobin
7680, 1991. level and its clinical impact in a cohort of patients with COPD. Eur
9. Skwarski K, Macnee W, Wraith PK, Sliwinski P, Zielinski J. Predictors Respir J 29: 92329, 2007.
of survival in patients with chronic obstructive pulmonary disease 30. Jones NL, Jones G, Edwards RHT. Exercise tolerance in chronic air-
treated with long-term oxygen therapy. Chest 100: 152227, 1991. way obstruction. Am Rev Respir Dis 103: 47791, 1971.
10. Chaouat A, Weitzenblum E, Kessler R, Schott R, Charpentier C, 31. Dodd DS, Brancatisano T, Engel LA. Chest wall mechanics during
Levi-Valensi P, Zielinski J, Delaunois L, Cornudella R, Moutinho dos exercise in patients with severe chronic air-ow obstruction. Am Rev
Santos J. Outcome of COPD patients with mild daytime hypoxaemia Respir Dis 129: 3338, 1984.
with or without sleep-related oxygen desaturation. Eur Respir J 17: 32. Montes de Oca M, Rassulo J, Celli B. Respiratory muscle function and
84855, 2001. cardiopulmonary response to exercise in severe COPD. Am J Respir
11. Cooper CB, Waterhouse J, Howard P. Twelve-year clinical study of Crit Care Med 154: 128489, 1996.
patients with hypoxic cor pulmonale given long-term domiciliary oxy- 33. ODonnell D, Flugre T, Gerken F, Hamilton A, Webb K, Aguilaniu B
gen therapy. Thorax 42: 10510, 1987. et al. Eects of tiotropium on lung hyperination, dyspnea and exercise
12. Ne TA, Petty TL. Long-term continuous oxygen therapy in chronic tolerance in COPD. Eur Respir J 23: 83240, 2004.
airway obstruction. Ann Intern Med 72: 62125, 1970. 34. Woodcock AA, Gross ER, Geddes DM. Oxygen relieves breathless-
13. Crockett AJ, Cranston JM, Moss JR, Alpers JH. Survival on long-term ness in pink puers. Lancet 1: 9079, 1981.
oxygen therapy in chronic airow limitation: From evidence to out- 35. Leggett RJE, Flenley DC. Portable oxygen and exercise tolerance in
comes in the routine clinical setting. Intern Med J 31: 44854, 2001. patients with chronic hypoxic cor pulmonale. BMJ 2: 8486, 1977.
14. Levi-Valensi P, Duwoos H, Racineaux JL. Multicenter study of oxygen 36. Cotes JE, Gilson JC. Eect of oxygen on exercise ability in chronic res-
therapy: GEMOS preliminary results. Med Thorac 5: 5026, 1983. piratory insuciency. Lancet 1: 87276, 1956.
15. Levine BE, Bigelow DB, Hamstra RD et al. The role of long-term con- 37. Bradley BL, Garner AE, Billiu D, Mestas JM, Forman J. Oxygen-
tinuous oxygen administration in patients with chronic airway obstruc- assisted exercise in chronic obstructive lung disease. The eect on exer-
tion with hypoxemia. Ann Intern Med 66: 63950, 1967. cise capacity and arterial blood gas tension. Am Rev Respir Dis 118:
16. Abraham AS, Cole RB, Bishop JM. Reversal of pulmonary hyper- 23943, 1978.
tension by prolonged oxygen administration to patients with chronic 38. Bye PTP, Esau SA, Levy RD et al. Ventilatory muscle function during
bronchitis. Circ Res 24: 14757, 1968. exercise in air and oxygen in patients with chronic air-ow limitation.
17. Selinger SR, Kennedy TP, Buescher P et al. Eects of removing oxy- Am Rev Respir Dis 132: 23640, 1985.
gen from patients with chronic obstructive pulmonary disease. Am Rev 39. Dean NC, Brown JK, Himelman RB, Doherty JJ, Gold WM,
Respir Dis 136: 8591, 1987. Stulbarg MS. Oxygen may improve dyspnea and endurance in patients
18. Aber GM, Harris AM, Bishop JM. The eect of acute changes with chronic obstructive pulmonary disease and only mild hypoxemia.
in inspired oxygen concentration on cardiac, respiratory and renal Am Rev Respir Dis 146: 94145, 1992.

685
Asthma and COPD: Basic Mechanisms and Clinical Management

40. Stevenson NJ, Calverley PM. Eect of oxygen on recovery from maxi- obstructive pulmonary disease patients. Am J Respir Crit Care Med 168:
mal exercise in patients with chronic obstructive pulmonary disease. 103442, 2003.
Thorax 59: 66872, 2004. 63. Luft U. Aviation physiology the eects of altitude. In: Fenn W, Rahn K
41. Stanek KA, Nagle FJ, Bisgard GE, Burnes WC. Eect of hyperoxia on (eds). Handbook of Physiology Respiration, 2, p. 1099. Washington, DC:
oxygen consumption in exercising ponies. J Appl Physiol 46: 111518, American Physiol. Soc, 1965.
1979. 64. Krop HD, Block AJ, Cohen E. Neuropsychologic eects of continu-
42. Vyas MN, Banister EW, Morton JW, Grzybowski S. Response to exer- ous oxygen therapy in chronic obstructive pulmonary disease. Chest 64:
cise in patients with chronic airway obstruction. II. Eects of breathing 31722, 1973.
40 percent oxygen. Am Rev Respir Dis 103: 40112, 1971. 65. Block AJ, Castle JR, Keitt AS. Chronic oxygen therapy. Treatment of
43. Morrison DA, Stovall JR. Increased exercise capacity in hypoxemic chronic obstructive pulmonary disease at sea level. Chest 65: 27988,
patients after long-term oxygen therapy. Chest 102: 54250, 1992. 1974.
44. Gosselin N, Durand F, Poulain M, Lambert K, Ceugniet F, Prefaut C, 66. Grant I, Heaton RK, Mcsweeny AJ, Adams KM, Timms RM.
Varray A. Eect of acute hyperoxia during exercise on quadriceps electrical Neuropsychologic ndings in hypoxemic chronic obstructive pulmo-
activity in active COPD patients. Acta Physiol Scand 181: 33343, 2004. nary disease. Arch Intern Med 142: 147076, 1982.
45. Stein DA, Bradley BL, Miller WC. Mechanisms of oxygen eects on 67. Prigatano GP, Parsons OA, Wright E, Levin DC, Hawryluk G.
exercise in patients with chronic obstructive pulmonary disease. Chest Neuropsychologic test performance in mildly hypoxemic patients with
81: 610, 1982. chronic obstructive pulmonary disease. J Consult Clin Psychol 51: 108
46. van Helvoort HA, Heijdra YF, Heunks LM, Meijer PL, Ruitenbeek 16, 1983.
W, Thijs HM, Dekhuijzen PN. Supplemental oxygen prevents exer- 68. Grant I, Prigatano GP, Heaton RK, McSweeny AJ, Wright EC,
cise-induced oxidative stress in muscle-wasted patients with chronic Adams KM. Progressive neuropsychologic impairment and hypox-
obstructive pulmonary disease. Am J Respir Crit Care Med 176: 1122 emia. Relationship in chronic obstructive pulmonary disease. Arch Gen
29, 2006. Psychiatry 44: 9991006, 1987.
47. Criner GJ, Celli BR. Ventilatory muscle recruitment in exercise with 69. Heaton RK, Grant I, McSweeny AJ, Adams KM, Petty TL.
O2 in obstructed patients with mild hypoxemia. J Appl Physiol 63: Psychological eects of continuous and nocturnal oxygen therapy in
195200, 1987. hypoxemic chronic obstructive pulmonary disease. Arch Intern Med
48. Guz A, Adams L, Minty K, Murphy K. Breathlessness and the ventila- 143: 194147, 1983.
tory drives of exercise, hypercapnoea and hypoxia. Clin Sci 60: 1718, 70. Siesjo B, Johannsson H, Ljunggren B, Norberg K. Brain dysfunction in
1981. mild to moderate hypoxia. Am J Med 70: 124753, 1981.
49. Tiep BL. Long-term home oxygen therapy. Clin Chest Med 11: 50521, 71. Gibson GE, Shimada M, Blass JP. Alterations in acetylcholine synthe-
1990. sis and in cyclic nucleotides in mild cerebral hypoxia. J Neurochem 31:
50. Celli BR, MacNee W. Standards for the diagnosis and treatment of 75760, 1978.
COPD. Eur Respir J 23: 93246, 2004. 72. Bartus RT. Eects of cholinergic agents on learning and memory in
51. Owens GR, Rogers RM, Pennock BE, Levin D. The diusing capacity animal models of aging. Aging 19: 27180, 1982.
as a predictor of arterial oxygen desaturation during exercise in patients 73. Aserinsky E, Kleitman N. Regularly occurring periods of eye motility
with chronic obstructive pulmonary disease. N Engl J Med 310: 1218 and concomitant phenomena during sleep. Science 118: 27374, 1953.
21, 1984. 74. Aserinsky E. Periodic respiratory pattern occurring in conjunction with
52. Astin TW, Penman RWB. Airway obstruction due to hypoxemia in eye movements during sleep. Science 150: 76366, 1965.
patients with chronic lung disease. Am Rev Respir Dis 95: 56775, 75. Robin ED, Whaley RD, Crump CH, Travis DM. Alveolar gas ten-
1967. sions, pulmonary ventilation and blood pH during physiological sleep
53. Libby DM, Briscoe WA, King TKC. Relief of hypoxia-related bron- in normal subjects. J Clin Invest 37: 98189, 1958.
choconstriction by breathing 30 percent oxygen. Am Rev Respir Dis 76. Trask CH, Cree EM. Oximeter studies on patients with chronic
123: 17175, 1981. obstructive emphysema, awake and during sleep. N Engl J Med 266:
54. Nadel JA, Widdicombe JG. Eect of changes in blood gas tensions 63942, 1962.
and carotid sinus pressure on tracheal volume and total lung resistance 77. Koo KW, Sax DS, Snider GL. Arterial blood gases and pH during sleep
to airow. J Physiol 163: 1333, 1962. in chronic obstructive pulmonary disease. Am J Med 58: 66370, 1975.
55. Swinburn CR, Mould H, Stone TN, Corris PA, Gibson GJ. 78. Leitch AG, Clancy LJ, Leggett RJE, Tweeddale P, Dawson P, Evans JI.
Symptomatic benet of supplemental oxygen in hypoxemic patients Arterial blood gas tensions, hydrogen ion, and electroencephalogram
with chronic lung disease. Am Rev Respir Dis 143: 91315, 1991. during sleep in patients with chronic ventilatory failure. Thorax 31:
56. Couser JI, Make BJ. Transtracheal oxygen decreases inspired minute 73035, 1976.
ventilation. Am Rev Respir Dis 139: 62731, 1989. 79. Wynne JW, Block AJ, Hemenway J, Hunt LA, Flick MR. Disordered
57. Astin TW. The eect of oxygen inhalation on the work of breathing breathing and oxygen desaturation during sleep in patients with
in patients with chronic obstructive bronchitis. Respiration 27: 5162, chronic obstructive lung disease (COLD). Am J Med 66: 57379, 1979.
1970. 80. Catterall JR, Douglas NJ, Calverley PMA et al. Transient hypoxemia
58. Mannix ET, Manfredi F, Palange P, Dowdeswell IRG, Farber MO. during sleep in chronic obstructive pulmonary disease is not a sleep
Oxygen may lower the O2 cost of ventilation in chronic obstructive apnea syndrome. Am Rev Respir Dis 128: 2429, 1983.
lung disease. Chest 101: 91015, 1992. 81. Hudgel DW, Martin RJ, Capehart M, Johnson B, Hill P. Contribution
59. Benditt JO, Rassulo J, Celli BR. Work of breathing during direct tra- of hypoventilation to sleep oxygen desaturation in chronic obstructive
cheal O2 administration in patients with severe chronic lung disease. pulmonary disease. J Appl Physiol 55: 66977, 1983.
Am Rev Respir Dis 14: A883, 1990. 82. Ballard RD, Clover CW, Suh BY. Inuence of sleep on respiratory
60. Tarpy S, Epstein S, Gottlieb D, Celli B. The eect of oxygen and air function in emphysema. Am J Respir Crit Care Med 151: 94551, 1995.
via nasal cannula on the oxygen cost of breathing in chronic airow 83. Douglas NJ. Sleep in patients with chronic obstructive lung disease.
obstruction. Am Rev Respir Dis 145: A646, 1992. Clin Chest Med 19: 11525, 1998.
61. Somfay A, Porszasz J, Lee SM, Casaburi R. Dose-response eect of 84. Douglas NJ, White DP, Pickett CK, Weil JV, Zwillich CW. Respiration
oxygen on hyperination and exercise endurance in nonhypoxaemic during sleep in normal man. Thorax 37: 84044, 1982.
COPD patients. Eur Respir J 18: 7784, 2001. 85. Douglas NJ, White DP, Weil JV et al. Hypoxic ventilatory response
62. Emtner M, Porszasz J, Burns M, Somfay A, Casaburi R. Benets of decreases during sleep in normal men. Am Rev Respir Dis 125: 28689,
supplemental oxygen in exercise training in nonhypoxemic chronic 1982.

686
Long-Term Oxygen Therapy 54
86. Berthon-Jones M, Sullivan CE. Ventilatory and arousal responses to 107. Fletcher EC, Donner CF, Midgren B et al. Survival in COPD
hypoxia in sleeping humans. Am Rev Respir Dis 126: 75862, 1982. patients with a daytime PaO2  60 mm Hg with and without noc-
87. Douglas NJ, White DP, Weil JV, Pickett CK, Zwillich CW. turnal oxyhemoglobin desaturation. Chest 101: 64955, 1992.
Hypercapnic ventilatory response in sleeping adults. Am Rev Respir 108. Conference report. New problems in supply, reimbursement, and cer-
Dis 126: 75862, 1982. tication of medical necessity for long-term oxygen therapy. Am Rev
88. Berthon-Jones M, Sullivan CE. Ventilation and arousal responses to Respir Dis 142: 72174, 1990.
hypercapnia in normal sleeping adults. J Appl Physiol 57: 5967, 1984. 109. Lock SH, Blower G, Prynne M, Wedzicha JA. Comparison of liquid and
89. Orem J. Medullary respiratory neuron activity: Relationship to tonic gaseous oxygen for domiciliary portable use. Thorax 47: 98100, 1992.
and phasic REM sleep. J Appl Physiol 48: 5465, 1980. 110. Lacasse Y, Lecours R, Pelletier C, Begin R, Maltais F. Randomised
90. Millman RP, Knight H, Kline LR, Shore ET, Chung D-CC, Pack AI. trial of ambulatory oxygen in oxygen-dependent COPD. Eur Respir J
Changes in compartmental ventilation in association with eye move- 25: 103238, 2005.
ments during REM sleep. J Appl Physiol 65: 1196202, 1988. 111. Tiep BL, Belman MJ, Mittman C, Phillips RE, Otsap B. A new oxy-
91. Tabachnik E, Muller NL, Bryan AC, Levison H. Changes in venti- gen-saving nasal cannula. Am Rev Respir Dis 130: 5002, 1983.
lation and chest wall mechanics during sleep in normal adolescents. 112. Soer M, Tashkin DP, Shapiro BJ, Littner M, Harvey E, Farr S.
J Appl Physiol 51: 55764, 1981. Conservation of oxygen supply using a reservoir nasal cannula in
92. Tusiewicz K, Moldofsky H, Bryan AC, Bryan MH. Mechanisms of hypoxemic patients at rest and during exercise. Chest 88: 66368, 1985.
the rib cage and diaphragm during sleep. J Appl Physiol 43: 6002, 113. Tiep BL, Belman MJ, Mittman C, Phillips RE, Otsap B. A new
1977. pendant storage oxygen-conserving nasal cannula. Chest 87: 38183,
93. Johnson MW, Remmers JE. Accessory muscle activity during sleep 1985.
in chronic obstructive pulmonary disease. J Appl Physiol 57: 101117, 114. Franco MA, Llompart JA, Teague R, Bloom K, Wilson R. Pulse dose
1984. oxygen delivery system. Respir Care 29: 103441, 1984.
94. Martinez F, Couser J, Celli B. Factors inuencing ventilatory mus- 115. Tiep BL, Nicotra B, Carter R, Phillips R, Otsap B. Low-concentra-
cle recruitment in patients with chronic airow obstruction. Am Rev tion oxygen therapy via a demand oxygen delivery system. Chest 87:
Respir Dis 142: 27682, 1990. 63638, 1985.
95. Fletcher EC, Gray BA, Levin DC. Nonapneic mechanisms of arterial 116. Brook CJ, Bower JS, Davis DM, Zimmer AK. Performance of a
oxygen desaturation during rapid-eye-movement sleep. J Appl Physiol demand cannula system during rest, exercise, and sleep in hypoxemic
54: 63239, 1983. patients. Am Rev Respir Dis 133: 209A, 1986.
96. Coccagna G, Lugaresi E. Arterial blood gases and pulmonary and 117. Shigeoka JW. Oxygen conservers, home oxygen prescriptions, and the
systemic arterial pressure during sleep in chronic obstructive pulmo- role of the respiratory care practitioner. Respir Care 36: 17883, 1991.
nary disease. Sleep 1: 11724, 1978. 118. Hagarty E, Skorodin M, Langbein L, Hultman C, Jessen J, Maki K.
97. Boysen PG, Block AJ, Wynne JW, Hunt LA, Flick MR. Nocturnal Comparison of three oxygen delivery systems during exercise in
pulmonary hypertension in patients with chronic obstructive pulmo- hypoxemic patients with chronic obstructive pulmonary disease. Am J
nary disease. Chest 76: 53642, 1979. Respir Crit Care Med 155: 89398, 1997.
98. Douglas NJ, Calverley PMA, Leggett RJE, Brash HM. Transient 119. Bower JS, Brook CJ, Zimmer K, Davis D. Performance of a demand
hypoxaemia during sleep in chronic bronchitis and emphysema. oxygen saver system during rest, exercise, and sleep in hypoxemic
Lancet 1: 14, 1979. patients. Chest 94: 7780, 1988.
99. Fletcher EC, Luckett RA, Miller T, Castrangos C, Kutka N, Fletcher 120. Heimlich HJ. Respiratory rehabilitation with transtracheal oxygen
JG. Pulmonary vascular hemodynamics in chronic lung disease system. Ann Otol Rhinol Laryngol 91: 64347, 1982.
patients with and without oxyhemoglobin desaturation sleep. Chest 121. Christopher KL, Spoord BT, Petrun MD, McCarty DC, Goodman
95: 75764, 1989. JR, Petty TL. A program for transtracheal oxygen delivery:
100. Levi-Valensi P, Weitzenblum E, Rida Z et al. Sleep-related oxygen Assessment of safety and ecacy. Ann Intern Med 107: 8028, 1987.
desaturation and daytime pulmonary haemodynamics in COPD 122. Homan LA, Dauber JD, Ferson PF, Openbrier DR, Zullo TG.
patients. Eur. Respir. J. 5: 3017, 1992. Patient response to transtracheal oxygen delivery. Am Rev Respir Dis
101. Calverley PMA, Brezinova V, Douglas NJ, Catterall JR, Flenley DC. 135: 15356, 1987.
The eect of oxygenation on sleep quality in chronic bronchitis and 123. Homan LA, Johnson JT, Wesmiller SW et al. Transtracheal delivery
emphysema. Am Rev Respir Dis 126: 20610, 1982. of oxygen: Ecacy and safety for long-term continuous therapy. Ann
102. Fleetham J, West P, Mezon B, Conway W, Roth T, Kryger M. Sleep Otol Rhinol Laryngol 100: 10815, 1991.
arousals, and oxygen desaturation in chronic obstructive pulmonary 124. Christopher KL, Spoord BT, Brannin PK, Petty TL. Transtracheal
disease: the eect of oxygen therapy. Am Rev Respir Dis 126: 42933, oxygen therapy for refractory hypoxemia. JAMA 256: 49497, 1986.
1982. 125. Adamo JP, Mehta AC, Stelmach K, Meeker D, Rice T, Stoller JK. The
103. Tiep BL, Lewis MI. Oxygen conservation and oxygen-conserving Cleveland Clinics initial experience with transtracheal oxygen ther-
devices in chronic lung disease: A review. Chest 92: 26372, 1987. apy. Respir Care 35: 15360, 1990.
104. Goldstein RS, Ramcharan V, Bowes G, McNicholas WT, Bradley D, 126. Heimlich HJ, Carr GC. The micro-trach: A seven-year experience
Phillipson EA. Eect of supplemental nocturnal oxygen on gas with transtracheal oxygen therapy. Chest 95: 100812, 1989.
exchange in patients with severe obstructive lung disease. N Engl J 127. Heimlich HJ, Carr GC. Transtracheal catheter technique for pulmo-
Med 310: 42529, 1984. nary rehabilitation. Ann Otol Rhinol Laryngol 94: 5024, 1985.
105. Connaughton JJ, Catterall JR, Elton RA, Stradling JR, Douglas NJ. 128. Fletcher EC, Nickeson D, Costrarangos-Galaraza C. Endotracheal
Do sleep studies contribute to the management of patients with mass resulting from a transtracheal oxygen catheter. Chest 93: 43839,
chronic obstructive pulmonary disease?. Am Rev Respir Dis 138: 1988.
34144, 1988. 129. Burton GG, Wagshul FA, Henderson D, Kime SW. Fatal airway
106. Casanova C, Hernandez MC, Sanchez A, Garcia-Talavera I, de obstruction caused by a mucous ball from a transtracheal oxygen
Torres JP, Abreu J, Valencia JM, Aguirre-Jaime A, Celli BR. Twenty- catheter. Chest 99: 152023, 1991.
four-hour ambulatory oximetry monitoring in COPD patients with 130. Gong HJ. Airtravel and oxygen therapy in cardiopulmonary patients.
moderate hypoxemia. Respir Care 51: 141623, 2006. Chest 101: 110413, 1992.

687
Immunomodulators
55 CHAPTER

INTRODUCTION: THE CLINICAL NEED IMMUNOMODULATORY THERAPY IN Christopher J. Corrigan


ASTHMA
Asthma aects 10% of children and 4% of adults, Department of Asthma , Allergy and
with an overall prevalence of 6% (3 million Respiratory Science, Guys Hospital,
Asthma is associated with chronic, cell-mediated
patients) in the United Kingdom. Mortality from inammation of the bronchial mucosa in which London, UK
asthma is relatively low (1680 deaths in the United eosinophil-active cytokine products of acti- MRC and Asthma UK Centre in Allergic
Kingdom in 1994), but considerable morbid- vated T-cells play a prominent role (Fig. 55.1). Mechanisms of Asthma Kings College
ity arises from disease in that minority of patients Evidence suggests that glucocorticoids amelio- London, UK
whose symptoms are inadequately controlled by rate asthma at least partly by inhibiting T-cell
conventional therapy (inhaled corticosteroid, long- activation and cytokine production [24]. There
acting 2-agonist, leukotriene receptor antagonist) has been a great deal of research directed at why
even when optimal delivery has been assured, com- T-cells in some asthmatics are relatively resistant
pliance has been veried and the eects of other to glucocorticoid inhibition, but it will be some
exacerbating factors minimized. In these patients time before this research evolves toward new
oral corticosteroid is often employed, but even then approaches to therapy. In the meantime, other T-
patients may remain symptomatic and become cell immunomodulatory agents have been inves-
vulnerable to unwanted eects of the therapy. No tigated for their possible therapeutic eects in
existing therapy for asthma is preventative, curative, asthma. As many of these agents have potentially
or clearly disease modifying. Estimates from stud- serious unwanted eects, attention has generally
ies of prescribing practice in the United Kingdom been focused on those asthmatics who continue
[1] suggest that approximately 6% of asthmatics are to have severe disease despite maximal therapy
receiving therapy that would place them at steps 4 (i.e., in those patients in whom the benet/risk
or 5 of the British Thoracic Society asthma treat- ratio of therapy is most likely to be acceptable).
ment guidelines. Although in a minority, these
patients account for the majority of the costs of
asthma care because of their frequent need for phy- Gold salts
sician consultations and emergency and inpatient
care. For such patients, new approaches to therapy Evaluation of these drugs in asthma has been
are urgently required. based on empirical observation of their antiin-
In contrast with asthma, chronic obstruc- ammatory eects in diseases such as rheumatoid
tive pulmonary disease (COPD), a disease caused arthritis, rather than cogent hypothesis regarding
largely by cigarette smoking, has a lower preva- their possible mechanisms of action. Actions of
lence (1% or 600,000 patients, usually 65 or more gold salts which may be relevant to a glucocorti-
years of age in the United Kingdom), but a much coid-sparing eect in asthma are ill-dened, but
higher mortality (23,550 deaths in 1994) and con- may include inhibition of T-cell proliferation, IL-
siderable morbidity. Once established, the clinical 5-mediated prolongation of eosinophil survival
progression of airways obstruction and destruc- [5], IgE-mediated degranulation of mast cells and
tion is relentless. Yet, no existing therapy has been basophils [6], and leukotriene production by gran-
shown to be preventative, curative, or disease ulocytes [7]. Some of these eects may be second-
modifying. ary to inhibition by gold salts of pro-inammatory

689
Asthma and COPD: Basic Mechanisms and Clinical Management

Histamine
Prostaglandin D2 Bronchospasm
leukotrienes

Mast cell
IL-4, IL-13, TGF-
Acute IgE
Chronic
Allergen B
other antigens? B cell Epi
Fib AHR
ASM Remodeling
TSLP TSLP
IL-4, IL-13 IL-4 Narrowing

IL-13
TGF-
IL-5

CD4 Major basic proteins


leukotrienes
DC
Th2 T cell Eosinophil

FIG. 55.1 Working hypothesis of asthma pathogenesis. Mast cells sensitized by surface, allergen-specific IgE release acute mediators of bronchoconstriction
on allergen exposure, which may serve as a mechanism for acute exacerbation of asthma in atopic patients. Th2 T-cell differentiation in the airways is driven
by dendritic cells (DC) which present allergens and possibly other antigens to T-cells in an MHC Class II restricted fashion, possibly under the influence of
thymic stromal lymphopoietin (TSLP) produced by mast cells, epithelial cells and other cells. Th2 cytokines control IgE synthesis by B cells (IL-4, IL-13) and
encourage recruitment and activation of eosinophils (IL-5) which may damage the mucosa through release of major basic protein, and are also a source of
remodeling cytokines such as transforming growth factor- (TGF-). Th2 cytokines such as IL-4 and IL-13 also induce release of growth factors by structural
cells in the airways such as epithelial cells (Epi), myofibroblasts (Fib) and airways smooth muscle cells (ASM) causing remodeling changes (fibrous protein lay
down, mucous hyperplasia, smooth muscle hypertrophy/hyperplasia). Mast cells are also a source of remodeling cytokines.

transcriptional regulatory proteins, in particular NF-B [8]. that methotrexate therapy for a minimum of 36 months
Two double-blind, placebo-controlled parallel group studies allows signicant (overall 20%, but only in about 60%
[9, 10] in which oral gold salts were administered for 6 months of responding patients) reduction in oral glucocorticoid
showed a modest but signicant glucocorticoid-sparing eect requirements, with no signicant improvement in lung
of the therapy as compared with placebo. Lung function was function. In a more exacting analysis of these trials by the
not improved, and additional antiasthma therapy was not Cochrane reviewers [17], however, it was concluded that
reduced. Not all of the patients showed a signicant response. existing trial data do not justify the premise that meth-
The unwanted eects of oral gold salts include dermatitis, otrexate is glucocorticoid-sparing. The most serious poten-
hepatic dysfunction, proteinuria, interstitial pneumonitis, and, tial unwanted eect of methotrexate therapy is cumulative
rarely, blood dyscrasias [11]. Nevertheless, it has been con- hepatic toxicity and hepatic brosis, with isolated reports of
cluded in one study [12] comparing the glucocorticoid-sparing deaths from opportunistic infections and pneumonitis.
eects of gold salts, methotrexate, and cyclosporin A (CsA) in
severe asthma that gold salts provide the optimal risk/benet
ratio in terms of ecacy and tolerability. Cyclosporin A
CsA is a lipophilic, cyclic undecapeptide derived from the
Methotrexate fungus Tolypocladium inatum. In a complex with the cyto-
plasmic binding protein cyclophilin, it inhibits calcineurin-
Methotrexate is a folic acid analog used at low dosage for mediated dephosphorylation and nuclear translocation of
its anti-inammatory activity in an increasing variety of the cytoplasmic subunit of the T-cell transcriptional activa-
chronic diseases. It exerts a delayed but sustained thera- tor NF-AT, thus inhibiting T-cell proliferation and cytokine
peutic eect with weekly dosage regimens. This probably production relatively specically [18].
reects its accumulation in cells as polyglutamate com- Two blinded, placebo-controlled trials in severe, glu-
plexes, resulting in accumulation of S-adenosyl methionine cocorticoid-dependent asthmatics [19, 20] together showed
and adenosine, both of which are inhibitory to T-cell func- that concomitant CsA therapy improved lung function while
tion [13]. Methotrexate therapy also appears to increase reducing oral prednisolone requirements. As with methotrex-
T-cell sensitivity to glucocorticoid inhibition [14]. Meta- ate, not all patients showed a signicant response. Unwanted
analysis of placebo-controlled trials of methotrexate therapy eects of low-dosage CsA therapy include hypertension and
in oral glucocorticoid-dependent asthma [15, 16] suggest renal impairment. Regular monitoring of renal function,

690
Immunomodulators 55
blood pressure, and whole blood trough concentrations of respectively, particularly in T-cells), leunomide and the nap-
CsA is necessary. Lymphoproliferative disorders and serious thopyrans. Whether or not these drugs will oer opportunities
opportunistic infections appear to be very uncommon. for the therapy of severe asthma with a more favorable ben-
et/risk ratio remains to be seen. One new immunosuppressive
agent, suplatast tosilate, appears to inhibit the production of
Intravenous immunoglobulin therapy asthma-relevant cytokines relatively selectively, and early stud-
ies suggest clinical benet in asthma.
Therapy with pooled intravenous immunoglobulin (IVIG),
originally designed to restore immune deciency, also appears
to have immunomodulatory eects in diseases involving
immune-eector mechanisms. In a blinded, parallel group CYTOKINE-DIRECTED THERAPY IN
study of oral glucocorticoid-dependent asthmatics [21],
patients were randomized to receive IVIG 2 g/kg, 1 g/kg, or ASTHMA
albumin (placebo) 2 g intravenously monthly for a total of 6
months (7 infusions). During the therapy, oral prednisone T helper 2 (Th2) lymphocytes are thought to play a key
requirements were reduced to an almost identical extent role in asthma pathogenesis as they are a principal source of
(3339%) in all three groups, with no signicant dierences asthma-relevant cytokines. Therapeutic strategies directed
in changes in symptoms, lung function, and the frequency of toward inhibition of Th2 cytokines would thus seem to
disease exacerbations. In a second, similar blinded trial [22], oer an attractive immunomodulatory strategy for asthma
treatment of patients with both IVIG and placebo allowed (Table 55.1). Alternatively, because a range of Th1 cytokines
signicant reduction in oral prednisone dosages, which was are inhibitory to the development of Th2 T-cells, or antago-
signicantly greater in the IVIG-treated group when only nize the actions of Th2 cytokines on target pro-inamma-
those asthmatics on relatively high-initial dosages of oral tory leukocytes, therapy with key Th1 cytokines may also be
prednisone were included in the analysis. Another double- eective in asthma (Table 55.1).
blind, placebo-controlled study of IVIG therapy in severe
childhood asthma [23] failed to show any benet of IVIG
over placebo in terms of changes in symptoms and lung Inhibition of Th2 and other cytokines
function. The possible mechanisms by which IVIG therapy
could exert a benecial eect in asthma are not clear. Some Of the Th2 cytokines, IL-5 has been regarded as of partic-
of the benets may result from immunoglobulin replace- ular importance because its expression most closely corre-
ment itself, since a proportion of chronic asthmatics have lates with asthma severity and local eosinophil inltration
depressed serum IgG concentrations, although there is lit- (one of the most constant features of asthma). Humanized
tle evidence that this results in defective humoral immunity monoclonal antibodies against human IL-5 have been
[24]. Additionally, IVIG has been shown to increase T-cell manufactured. A single intravenous infusion of one of these
susceptibility to glucocorticoid inhibition in vitro [25], to antibodies (mepolizumab) markedly reduced circulating
abrogate IgE synthesis by B-cells in vitro [26], and to con- blood eosinophils and greatly reduced eosinophil recruit-
tain other potential immunomodulatory products including ment to the airways following allergen bronchial challenge
soluble CD4, CD8, and HLA molecules and cytokines. of mild, atopic asthmatics [28]. Unfortunately, however,
In summary, the evidence that IVIG is of any benet this had no eect on early or late-phase bronchoconstric-
in glucocorticoid-dependent asthma is at present equivocal. tion or associated changes in bronchial hyperresponsiveness.
In addition, the therapy is very expensive and is associated A more prolonged study [29] on patients with moderate to
with a high incidence of unpleasant urticarial and anaphy- severe asthma was also disappointing in terms of improve-
lactic reactions, as well as fever and aseptic meningitis. ments in asthma symptoms or lung function. A biopsy
study [30] suggested that, while anti-IL-5 antibody pro-
foundly reduced circulating eosinophils, it was less eec-
Newer immunomodulatory drugs tive in removing eosinophils from the bronchial mucosa but
TABLE 55.1 Cytokine-directed therapeutic strategies in asthma.
Tacrolimus (FK506) is a macrolide derived from the soil
organism Streptomyces tsukudaiensis which, despite having a
Inhibition of pro-inflammatory Therapy with inhibitory
dierent structure to CsA, similarly inhibits NF-AT activa-
cytokines cytokines
tion. Its increased potency may be outweighed by similarly
increased toxicity. IL-5 IL-10
Sirolimus (rapamycin) is another macrolide derived
from Streptomyces hygroscopius. It has a fundamentally dierent IL-4 IL-12
inhibitory action on T-cells [27], since it inhibits IL-2 signal-
IL-13 IFN-
ing at least partly by inhibiting phosphorylation/activation of
the kinase p70 S6 (p70S6k), and by inhibiting the enzymatic IL-9 IL-18
activity of the cyclin-dependent kinase cdk2-cyclin E complex.
Other new immunomodulatory drugs, the inhibitory IL-25 IL-23
actions of which are relatively specic for T-cells continue to TNF-
appear, including brequinar sodium and mycophenolate mofetil
(inhibitors of de novo synthesis of pyrimidines and purines IL: interleukin; IFN-: interferon-; TNF: tumor necrosis factor.

691
Asthma and COPD: Basic Mechanisms and Clinical Management

did reduce extracellular matrix deposition [31]. These stud- Other cytokines are of similar interest as possible
ies have cast doubt upon the role of inltrating cells such therapeutic targets in asthma. The cytokines IL-9 and IL-
as eosinophils in the causation of the clinical abnormalities 25 both amplify the production of Th2 cytokines. In addi-
associated with asthma, although since current strategies tion, IL-9 promotes mast-cell dierentiation and mediator
have not been eective in removing eosinophils entirely release and augments IgE production, and also promotes
from the bronchial mucosa, this conclusion must be guarded. some of the changes associated with asthmatic airways
Alternatively, cells such as eosinophils may participate in remodeling when over-expressed experimentally in animals.
longer term remodeling changes associated with asthma. In an animal model, a blocking anti-IL-9 antibody inhib-
Whatever the case be, these observations have at least tem- ited airways inammation [41]. Humanized, blocking anti-
porarily quenched enthusiasm for further evaluation of IL-9 antibodies are currently in development [42].
anti-IL-5 strategies. The cytokine tumor necrosis factor (TNF)- is expressed
IL-4, another Th2 cytokine, is one of the only two in asthmatic airways and may play a pivotal role in amplifying a
cytokines (the other being IL-13) which causes switching of wide range of inammatory responses. A blocking, humanized
B-lymphocytes to IgE synthesis and is therefore implicated anti-TNF- antibody (iniximab) and soluble TNF- decoy
in the pathogenesis of atopy. IL-4 is also involved in recruit- receptors (etanercept) have shown remarkable (not to men-
ment of eosinophils to the airways in asthma and acts in an tion unprecedented and unexpected) clinical ecacy in treat-
autocrine fashion further to promote the dierentiation of Th2 ing other inammatory diseases such as rheumatoid arthritis
lymphocytes. Soluble, humanized IL-4 receptors (altrakincept), and inammatory bowel disease, although there are concerns
administered by nebulization, have been tested in clinical trials about the immunosuppressive actions of this therapy on
and have been shown to prevent deterioration in lung func- resistance to microbial infections (recrudescence of tuberculo-
tion induced by withdrawal of corticosteroids in patients with sis, e.g., is a real problem) and possibly even tumor surveillance.
moderately severe asthma [32]. In addition, weekly nebuliza- In one study, treatment of moderate to severe asthmatics with
tion of soluble IL-4 receptor improved asthma control over etanercept 25 mg subcutaneously twice weekly for 12 weeks
a 12-week period [33]. Further, more prolonged studies were produced impressive improvements in symptoms and reduc-
more disappointing and this strategy also is currently not being tions in bronchial hyperresponsiveness [43] to a degree
pursued. IL-4 and the closely related IL-13 signal through the approaching that expected with systemic corticosteroid ther-
transcriptional activator STAT-6 and endogenous inhibitors of apy. A further study [44] replicated these ndings, while a
STAT-6 may prove to be useful therapeutic agents [34]. third [45] preliminary study was negative, suggesting that the
Further on the theme of cytokines causing airways therapy might be eective only in a subgroup of asthmatics in
changes in asthma independently of cellular inltration, IL-13, whom TNF- plays a relatively prominent role. Anti-TNF-
which shares a surface receptor, IL-4R, with IL-4, when therapies thus seem to hold considerable promise, which may
selectively over-expressed in the airways of mice causes many however be outweighed by their unwanted eects. Because
of the features associated with airways remodeling [35]. these therapies have to be administered by injection, a search is
Deletion of the IL-13 gene in mice prevents development of on for small molecule TNF- inhibitors. TNF--converting
airways hyperresponsiveness following allergen sensitization enzyme (TACE) is a matrix metalloproteinase-related enzyme
and challenge despite inltration of eosinophils (another required for the release of TNF- from cell surfaces where it is
eect of IL-13 is to increase the production of eosinophil stored in an extracellular matrix reservoir. Small molecule oral
attracting chemokines such as eotaxin by airways epithelial TACE inhibitors are currently in development [46].
cells) [36]. Apart from IL-4R, IL-13 has two other spe-
cic receptors named IL-13R1 and IL-13R2. The lat-
ter receptor also exists naturally in a soluble form and may Therapy with potential antiasthma
act as a decoy receptor, scavenging and thus inhibiting the cytokines
eects of free IL-13. Soluble IL-13R2 is currently in clini-
cal development as a possible new therapeutic approach for Some cytokines inhibit the Th2 cytokine responses of
asthma. In addition, a neutralizing, anti-IL-13 monoclonal T-cells which characterize asthma with varying degrees of
antibody [37] and a soluble anti-IL-13 fusion protein [38] specicity. While it may not be possible to administer such
were shown to prevent airways hyperresponsiveness pro- cytokines to patients in the long term, it may be possible to
duced by allergen challenge in mice. Recently, a recombinant develop strategies that increase their release or drugs which
human IL-4 mutant protein called pitrakinra has been activate their particular signaling receptors and pathways.
developed which competitively inhibits the IL-4R complex IL-10 has a number of anti-inammatory properties
and thus blocks the actions of IL-4 and IL-13 simultane- which may be relevant to asthma therapy. It is one of the few
ously. When given by inhalation or subcutaneous injection it cytokines the expression of which is enhanced, rather than
very convincingly reduced the late-phase response to allergen inhibited by corticosteroids. It inhibits the function of anti-
bronchial challenge of atopic asthmatics [39]. The mecha- gen-presenting cells (APC) and impairs their ability to prime
nism of this eect is unknown but could reect blockade of T-cells for Th2 cytokine release. It also reduces Th2 cytokine
the actions of IL-4 and/or IL-13 signaling. In addition to production by dierentiated Th2 T-cells and is thought to
possible anti-inammatory actions of IL-4R blockade, pit- be one of the principal eector cytokines which mediate the
rakinra also accelerated recovery from the acute-phase fall in activity of T regulatory cells. In addition, it exerts inhibi-
FEV1 immediately after allergen challenge, which may also tory actions on a range of inammatory leukocytes such
suggest an eect of IL-4/13 blockade on acute mast-cell as mast cells and eosinophils. Recombinant human IL-10 has
releasability [40]. proven eective in controlling other inammatory diseases

692
Immunomodulators 55
such as inammatory bowel disease and psoriasis, where it Several chemokines, including eotaxin, eotaxin-2,
is given as a weekly injection [47]. It is reasonably well tol- eotaxin-3, regulated on activation, normal T-cell expressed
erated, although hematological unwanted eects have been and secreted (RANTES) and monocyte chemoattractant
noted. With regard to asthma, peripheral blood T-cells from protein-4 (MCP-4) activate a common receptor on Th2 T-
clinically corticosteroid resistant, as compared with sensitive cells, mast cells and eosinophils called CCR3. All of these
severe asthmatics showed a marked defect in corticosteroid- chemokines are over expressed in the asthmatic airways
induced IL-10 production [48]. There is also evidence that mucosa [54, 55]. Several small molecule inhibitors of CCR3
IL-10 production plays a role in the well-known synergistic block eosinophil recruitment in animal models of asthma.
clinical eects of inhaled glucocorticoids and long-acting - The chemokine receptor CCR4 is selectively expressed
agonists in asthma [49]. These data strongly support a role for on Th2 T-cells, while CCR8 is expressed on a subset of
IL-10 in regulating asthma severity. Recent studies suggest these cells. Ligands for CCR4 include thymus and activation
that the defect in IL-10 production observed in T-cells from dependent cytokine (TARC) and monocyte-derived chem-
corticosteroid resistant asthmatics may be overcome by other okine (MDC), while a ligand for CCR8 is I-309. In animal
agents such as vitamin D3, which are known to increase IL- models of asthma, antibody blockade of all of these molecules
10 production, both in vitro and ex vivo [50]. Manipulation attenuated eosinophil inux, although only MDC and TARC
of IL-10 production by such relatively innocuous agents, as blockade attenuated bronchial hyperresponsiveness [5658].
distinct from therapy with IL-10 itself, oers great promise On the other hand, blockade of the CCR4 receptor [59] and
for future asthma therapy. gene deletion of the CCR8 receptor [60] in animal models of
IL-12 is a cytokine produced largely by APC which asthma failed to reduce cellular inux, perhaps reecting the
is a key regulator of Th1 T-cell development. Th1 cytokines, extreme redundancy of these pathways. The chemokine recep-
particularly interferon- strongly inhibit Th2 responses. tor CXCR4 is also selectively expressed on Th2 T-cells, and a
Recombinant human IL-12 has been administered to humans small molecule inhibitor of this receptor inhibited allergen-
but has a range of toxic eects which are somewhat minimized induced airways inammation in a murine model [61].
by slow escalation of the dosage. In a single study on atopic
asthmatics [51], weekly infusions of IL-12 over 4 weeks at
escalating dosage caused a progressive fall in circulating eosi-
nophils and also reduced ingress of eosinophils into induced THE WORTH OF CURRENT
sputum following allergen bronchial challenge. Unfortunately,
as with anti-IL-5, there was no evidence of reduction of the
IMMUNOMODULATORY THERAPY IN ASTHMA
early or late-phase bronchoconstrictor responses to allergen
challenge or alteration of the associated increased bronchial Immunosuppressive therapy
hyperresponsiveness. Most of the patients suered with malaise
and one with cardiac dysrhythmia. Consequently, IL-12 is not In view of these clinical observations, it will be clear that
likely to be used alone as a therapeutic agent for asthma. many reservations remain about the use of currently avail-
Cytokines such as IL-18, which synergizes with IL-12 able immunosuppressive therapy for the treatment of severe,
to enhance the release of Th1 cytokines by T-cells, and corticosteroid-dependent asthma since:
IL-23, which is related to, and shares some of the eects
of IL-12, are also potential therapeutic agents for asthma Not all patients respond, and the response cannot be
waiting to be explored, although problems with unwanted predicted a priori;
eects similar to those of IL-12 might be anticipated. The high incidence of unwanted eects makes it dicult
Interferon-, a signature Th1 cytokine has been adminis- to assess overall benet/risk ratios even in asthmatics
tered by nebulization to asthmatics [52]. It did not seem who are able to reduce oral corticosteroids;
to reduce inammation, at least as judged by eosinophil
inltration, although it is not certain whether sucient There is a risk of opportunistic infection and (at least
concentrations reached the airways. Preliminary reports theoretically) neoplasia;
suggest that interferon- may be useful for the treatment of There are many relative or absolute contraindications to
severe, corticosteroid refractory asthmatics [53]. therapy, such as pregnancy;
There is lack of knowledge about the long-term eects,
benecial or otherwise, of therapy.
Inhibition of chemokines
Consequently, it is clear that any further investiga-
Chemokines are particularly involved in the regula- tion of immunosuppressive therapy for asthma should be
tion of transit of inammatory cells out of the bone mar- performed within the connes of a controlled trial. There is
row and into target tissues. Over 50 dierent chemokines, an urgent need to produce a global denition of precisely
acting on over 20 dierent surface receptors have been which patients are suitable for such trials, and what consti-
described, allowing for much possible redundancy of action. tutes an appropriate trial of therapy. None of the immuno-
Chemokine receptors belong to the seven transmembrane suppressive strategies discussed above has made a signicant
receptor superfamily of G-protein-coupled receptors, for impact on the management of severe, oral corticosteroid-
which it is possible to manufacture small molecule inhibi- dependent asthma, although isolated patients do respond. A
tors. Many such inhibitors have been developed but only large gap in our knowledge is whether or not these drugs do
few, if any, have progressed into clinical trials in asthma. what they are assumed to do in asthma, which is to reduce

693
Asthma and COPD: Basic Mechanisms and Clinical Management

T-cell activation and cytokine production. Few studies have of neutrophils, CD8 T-cells and variable numbers of
addressed possible eects of immunosuppressive therapy on eosinophils [62, 63]. The amount of this inltrate is
the cellular and molecular immunopathology of the disease. closely related to total smoke (pack years) exposure, but
not so closely related to the presence/absence of COPD,
although some studies [64]seem to demonstrate a
Cytokine-directed therapy COPD-specic, elevated inux of CD8 Tcells.
Excessive production of proteinases, which digest
The similar general lack of success of cytokine-directed ther-
the lung parenchyma, and overwhelm endogenous
apy so far in asthma has also been disappointing. The appar-
antiproteinases which normally protect against this.
ent failure of anti-IL-5 sent a wave of shock and controversy
The principal proteinases are elastase and other
through the asthma research community, because the eosi-
serine proteinases from neutrophils, and matrix
nophil/IL-5 axis was thought to be fundamental to asthma
metalloproteinases (a group of over 20 related neutral
pathogenesis. These problems have, however, served little more
endopeptidases) from macrophages and epithelial cells.
than to highlight the chasm of ignorance about how the cel-
lular, immunological and structural changes one can observe A working hypothesis for the pathogenesis of COPD
in asthma are related, if at all, to the clinical manifestations of is shown in Fig. 55.2. At present this remains largely theoreti-
disease. There is a pressing need to delineate the precise func- cal; in particular, there is only circumstantial evidence for a role
tions of cells and cytokines implicated in asthma pathogen- for T-cells, although in transgenic animal models of COPD
esis, otherwise the success of cytokine-directed strategies (as where cytokines are articially expressed in the respiratory
with TNF-, perhaps the least asthma related cytokine) will mucosa, the cytokines TNF-, IFN-, and IL-13 can pro-
remain serendipitous. It is also essential to understand what duce changes reminiscent of emphysema and airways remod-
mechanisms cause irreversible airways obstruction in asthma eling changes which characterize the human disease, probably
so that therapy can be directed to these. through distinct mechanisms [65]. As with asthma (Fig. 55.1),
the scheme takes no account of why the process persists in
some individuals but not others. It may be inferred that thera-
peutic approaches to COPD may involve the use of inhibitors
IMMUNOMODULATORY THERAPY FOR COPD of macrophage and neutrophil function, leukotriene inhibitors,
and inhibitors of cytokines or chemokines and/or their actions
on target cells, and measures which increase proteinase inhibi-
Key elements of the pathogenesis of COPD include the tion. It should not be forgotten that, at present, the cheapest
following: and only eective therapeutic maneuver is smoking cessation.
Chronic exposure to cigarette smoke (a causal factor in
more than 90% of cases in the United Kingdom);
Glucocorticoids as immunomodulators
Individual (possibly inherited) susceptibility, since only in COPD
1020% of heavy smokers develop symptomatic disease.
A smoking-induced inammatory cellular inltrate In contrast with asthma, glucocorticoids have no signicant
in the airways, more marked peripherally, comprising eect on progressive loss of lung function which characterizes

STIMULUS

MMP/Cathepsins/ Emphysema
SLPI
Alveolar
Th1 cytokines Macrophage destruction
(IFN-, IL-18)
(apoptosis
dependent or
Chemokines Attraction
T-Cell independent)
(MIP-1/, RANTES, Activation
STIMULUS CD4? CD8?
IP-10, IL-8, etc.) Neutrophil
Th2 cytokines MMP/elastase Mucous
(IL-4, IL-13) Cathepsins/
APC metaplasia
1AT
Other granulocytes? Alveolar destruction (chronic)

FIG. 55.2 Theoretical scheme of COPD. Both Th1/Tc1 and Th2/Tc2 cytokines from CD4 or CD8 T-cells cause emphysema through secondary release of
chemokines, which attract monocyte/macrophages and granulocytes which in turn are a source of proteases such as matrix metalloproteinase (MMP), and also
by direct induction of apoptosis and down regulation of anti-proteases such as 1-anti-trypsin (1AT) and SLPI. CD8 cytotoxic T-cells may also attack alveolar
epithelial cells directly. Although COPD is much more common in cigarette smokers, the precise stimulus to T-cell and monocyte/macrophage activation is
unknown, as is whether T-cell activation is in any way antigen-specific involving antigen-presenting cells (APC) (which are also a potential source of chemokines).

694
Immunomodulators 55
COPD [66] and little eect in reducing inammatory-cell the CXCR1 and CXCR2 chemokine receptors. Since other
inltration into induced sputum [67] or the bronchial mucosa ligands for CXCR2 such as growth-related oncoprotein
[68], although when administered systemically they reduce (GRO)- are also circumstantially implicated in COPD
hospital stay and re-admission rates in patients with exacerba- [78], CXCR2 inhibitors may be more eective than blockade
tions [69, 70]. Inhaled glucocorticoids also reduce exacerbation of single chemokines [79], although there have been no clini-
rates in stable disease [71] and this is likely the principal (and cal trials of such agents as yet in COPD.
possibly the only) action of glucocorticoids which accounts for
their improvement in the quality of life of COPD suerers.
It is not clear why this disparity exists in the anti-
inammatory action of glucocorticoids in asthma and PHOSPHODIESTERASE INHIBITORS
COPD. It is possible to hypothesize that the environment IN ASTHMA AND COPD
of the mucosa of chronic smokers with COPD renders cells
insensitive to glucocorticoid inhibition [72]. The possibility
that T-cells, particularly of the CD8 phenotype, produce Phosphodiesterase (PDE) inhibitors may be regarded as hon-
cytokines which have been implicated in causing COPD in orary immunomodulatory drugs since they modify the func-
animal models provides the only precedent for the hypothesis tions of a wide range of immune as well as structural cells.
that immunomodulatory therapy will ameliorate symptoms PDE hydrolyzes cyclic AMP, and inhibitors elevate intra-
of COPD or alter the natural history of the disease. On the cellular cyclic AMP concentrations, which in turn inhibits
other hand, this approach might be particularly troublesome the pro-inammatory actions of many immune leukocytes
in view of the fact that these patients are much more suscep- including T-cells, macrophages, and neutrophils and relaxes
tible to debilitating infection than asthmatics. airways smooth muscle cells. There are many sub-types of
PDE and some of these sub-types also exist in multiple forms
[80]. PDE4 is the predominant enzyme in immune cells.
Inhibition of cytokines Two PDE4 inhibitors, cilomilast, and roumilast have been
evaluated for their possible therapeutic benets in asthma and
Concentrations of TNF- and its soluble receptor are COPD. Roumilast improved lung function in patients with
increased in the induced sputum of patients with COPD [73] mild/moderate asthma to a degree comparable to that achieved
and this mediator has been implicated in the pathogenesis of with inhaled glucocorticoid [81]. In a 6-month trial in patients
the weight loss sometimes associated with COPD [74]. A with moderate COPD, treatment with roumilast 0.5 mg
recent multi-center, placebo-controlled trial of therapy with daily resulted in a small but signicant improvement in post-
the humanized antiTNF monoclonal antibody iniximab for bronchodilator FEV1 (0.097 l compared with placebo) and
6 months (with evaluation for nearly 1 year) however showed a reduction in the numbers of exacerbations (0.75/patient
no impact on quality of life, exercise tolerance, or disease compared to 1.13/patient with placebo) [82]. Quality of
exacerbations in patients with COPD [75]. life was also improved in the patients taking active drug
Furthermore, more patients on active treatment devel- (3.5 points on the St. Georges Hospital respiratory ques-
oped more severe exacerbations and there was a trend toward tionnaire). Cilomilast at a dose of 15 mg twice daily for 12
more cases of cancer and pneumonia. It is not understood weeks reduced the numbers of T-cells, macrophages, and
why antiTNF- therapy is apparently more eective, at least neutrophils in bronchial biopsies of patients with COPD
in the short term, in asthma as compared with COPD, and [83]. Unfortunately, dose-limiting adverse events (nausea,
again this reects our ignorance of the true role of cytokines vomiting, and mesenteric arteritis) are still a cause for con-
and indeed any mediators in disease causation. Longer term cern with these drugs and neither has yet been licensed for
blockade might be more eective, but not without serious the treatment of asthma or COPD [84].
hazards. Other cytokines which have been implicated in
COPD either from animal models (as discussed earlier) or
circumstantially because their concentrations are elevated in
the target organ include IFN-, IL-13, IL-1, IL-6, IL-11,
CONCLUDING REMARKS
and IL-17 [76], but it is fair to say that in our current state
of knowledge any further advances with cytokine blockade in New drugs for the prevention and treatment of asthma and
COPD are most likely to be serendipitous. COPD are still greatly needed. In order to develop them,
it seems of paramount importance not just to describe the
pathophysiological features of each disease but to under-
Inhibition of chemokines stand exactly how these features cause clinical symptoms
and compromise lung function. Despite three decades of
The CXC chemokine IL-8 is increased in the sputum of research, evidence for a role for T-cells and cytokines in
COPD patients and may contribute to the bronchial mucosal asthma and COPD, which would mandate continuing
and sputum neutrophil inux that characterizes COPD, the development of drugs which block their production or
although there is little direct evidence for its involvement actions, remains largely circumstantial, and the fundamen-
in the causation of COPD. A humanized, monoclonal neu- tal questions as to why asthma develops in some individuals
tralizing antibody to IL-8 has been tested in COPD, and but not others, and why COPD develops only in a minor-
while it had a small eect in reducing dyspnea scores there ity of heavy smokers [85] remain unanswered. Most of
was no other clinical improvement [77]. IL-8 interacts with our current information comes from static, cross-sectional

695
Asthma and COPD: Basic Mechanisms and Clinical Management

studies of diseased subjects and there is very little known 13. Cronstein BN. Methotrexate and its mechanism of action. Arthritis
about what role cytokines and other mediators play in Rheum 39: 16095, 1996.
acute, short-term exacerbations of disease or in progressive 14. Corrigan CJ, Shiner R, Shakur BH, Ind PW. Methotrexate therapy in
asthma increases T cell susceptibility to corticosteroid inhibition. Clin
decline in lung function. Hence there is a need to develop
Exp Allergy 33: 109096, 2003.
novel short- and long-term outcome measures and sur- 15. Aaron SD, Dales RE, Pham B. Management of steroid-dependent
rogate biomarkers of disease in the short term, and better asthma with methotrexate: A meta-analysis of randomised clinical tri-
measures of lung functions decline in the longer term, such als. Respir Med 92: 105965, 1998.
as computerized tomography [86]. Powerful new techniques 16. Marin MG. Low-dose methotrexate spares steroid usage in steroid-
such as positional genomics may in future allow analysis dependent chronic asthmatic patients: A meta-analysis. Chest 112:
of heritable susceptibility factors in a manner unbiased by 2933, 1997.
our limited understanding of disease pathophysiology, and 17. Davies H, Olson L, Gibson P. Methotrexate as a steroid sparing agent
allow new approaches to disease prevention which may in adult asthma (Cochrane review). In The Cochrane Library. Oxford:
not depend on the inhibition of immunological processes. Update Software, 2000. Issue 3.
18. Schreiber SL, Crabtree GR. The mechanism of action of cyclosporin A
This will be the future challenge to counter these common
and FK506. Immunol Today 13: 13642, 1992.
and signicant diseases for which no eective, preventative 19. Alexander AG, Barnes NC, Kay AB. Trial of cyclosporin in corticos-
therapies currently exist. teroid-dependent chronic severe asthma. Lancet 339: 32428, 1992.
20. Lock SH, Kay AB, Barnes NC. Double blind, placebo-controlled study
of cyclosporin A as a corticosteroid-sparing agent in corticosteroid-
References dependent asthma. Am J Respir Crit Care Med 153: 50914, 1993.
21. Kishiyama JL, Valacer D, Cunningham-Rundles C et al. A multicentre,
1. Neville RG, Pearson MG, Richards N, Patience J, Sondhi S, randomised, double blind, placebo-controlled trial of high-dose intra-
Wagsta B, Wells N. A cost analysis of asthma prescribing in the UK. venous immunoglobulin for oral corticosteroid-dependent asthma. Clin
Eur Respir J 14: 6059, 1999. Immunol 91: 12633, 1999.
2. Corrigan CJ, Haczku A, Gemou-Engesaeth V, Doi S, Kikuchi Y, 22. Salmun LM, Barlan I, Wolf HM, Eibl M, Twarog FJ, Geha RS,
Takatsu K, Durham SR, Kay AB. CD4 T lymphocyte activation in Schneider LC. Eect of intravenous immunoglobulin on steroid con-
asthma is accompanied by increased serum concentrations of interleukin- sumption in patients with severe asthma: A double blind, placebo-
5: Eect of glucocorticoid therapy. Am Rev Respir Dis 147: 54047, 1993. controlled, randomised trial. J Allergy Clin Immunol 103: 81015, 1999.
3. Doi S, Gemou-Engesaeth V, Kay AB, Corrigan CJ. Polymerase chain 23. Niggemann B, Leupold W, Schuster A, Schuster R, Berg AV,
reaction quantication of cytokine messenger RNA expression in Hardt HVD, Eibl MM, Wahn U. Prospective, double-blind, placebo-
peripheral blood mononuclear cells of patients with severe asthma: controlled, multicentre study on the eect of high-dose intravenous
Eect of glucocorticoid therapy. Clin Exp Allergy 24: 85467, 1994. immunoglobulin in children and adolescents with severe bronchial
4. Corrigan CJ, Hamid Q , North J, Barkans J, Moqbel R, Durham S, asthma. Clin Exp Allergy 28: 20510, 1998.
Kay AB. Peripheral blood CD4, but not CD8 T lymphocytes in 24. Lack G, Ochs HD, Gelfand EW. Humoral immunity in steroid-
patients with exacerbation of asthma transcribe and translate messen- dependent children with asthma and hypogammaglobulinaemia.
ger RNA encoding cytokines which prolong eosinophil survival in the J Pediatr 129: 898903, 1996.
context of a Th2-type pattern: eect of glucocorticoid therapy. Am J 25. Spahn JD, Leung DYM, Chan MTS, Szeer SJ, Gelfand EW.
Respir Cell Mol Biol 12: 56778, 1995. Mechanisms of glucocorticoid reduction in subjects treated with intra-
5. Suzuki S, Okubo M, Kaise S, Ohara M, Kasukawa R. Gold sodium venous immunoglobulin. J Allergy Clin Immunol 103: 42126, 1999.
thiomalate selectively inhibits interleukin-5 mediated eosinophil sur- 26. Sigman K, Ghibu F, Sommerville W, Toledano BJ, Bastein Y, Cameron L,
vival. J Allergy Clin Immunol 96: 25156, 1995. Hamid QA, Mazer B. Intravenous immunoglobulin inhibits IgE produc-
6. Marone G, Columbo M, Galeone D, Guidi G, Kagey-Sobotka A, tion in human B lymphocytes. J Allergy Clin Immunol 102: 42127, 1998.
Lichtenstein LM. Modulation of the release of histamine and ara- 27. Dumont FJ, Su Q . Mechanism of action of the immunosuppressant
chidonic acid metabolites from human basophils and mast cells by rapamycin. Life Sci 58: 37395, 1995.
auranon. Agents Actions 18: 1002, 1986. 28. Leckie MJ, ten Brinke A, Khan J et al. Eects of an interleukin-5
7. Parente JE, Wong K, Davis P, Burka JF, Percy JS. Eects of gold com- blocking monoclonal antibody on eosinophils, airway hyperresponsive-
pounds on leukotriene B4, leukotriene C4 and prostaglandin E2 pro- ness and the late asthmatic response. Lancet 356: 214448, 2000.
duction by polymorphonuclear leukocytes. J Rheumatol 13: 4751, 29. Kips JC, OConnor BJ, Langely SJ, Woodcock A, Kerstjens HA et al.
1986. Eect of SCH55700, a humanised anti-human interleukin-5 antibody,
8. Bratt J, Belcher J, Vercellotti GM, Palmblad J. Eects of anti- in severe persistent asthma: A pilot study. Am J Respir Crit Care Med
rheumatic gold salts on NF-kappa B mobilisation and tumour necro- 167: 165559, 2003.
sis factor-alpha-induced neutrophil-dependent cytotoxicity for human 30. Flood-Page P, Menzies-Gow AN, Kay AB, Robinson DS. Eosinophils
endothelial cells. Clin Exp Immunol 120: 7984, 2000. role remains uncertain as anti-interleukin-5 only partially depletes
9. Nierop G, Gijzel WP, Bel EH, Zwinderman AH, Dijkman JH. numbers in asthmatic airway. Am J Respir Crit Care Med 167: 199204,
Auranon in the treatment of steroid dependent asthma: A double 2003.
blind study. Thorax 47: 34954, 1992. 31. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A et al.
10. Bernstein IL, Bernstein DI, Dubb JW, Faiferman I, Wallin B. A pla- Anti-IL-5 treatment reduces deposition of ECM proteins in the bron-
cebo-controlled multicenter study of auranon in the treatment of chial subepithelial basement membrane of mild atopic asthmatics.
patients with corticosteroid-dependent asthma. Auranon Multicenter J Clin Invest 112: 102936, 2003.
Drug Trial. J Allergy Clin Immunol 98: 31724, 1996. 32. Borish LC, Nelson HS, Lanz MJ, Claussen L, Whitmore JB et al.
11. Tomioka H, King TE. Gold-induced pulmonary disease: Clinical fea- Interleukin-4 receptor in moderate atopic asthma. A phase I/II ran-
tures, outcome, and dierentiation from rheumatoid lung disease. Am J domized, placebo-controlled trial. Am J Respir Crit Care Med. 160:
Respir Crit Care Med 155: 101120, 1997. 181623, 1999.
12. Bernstein IL, Bernstein DI, Bernstein JA. How does auranon com- 33. Borish LC, Nelson HS, Corren J, Bensch G, Busse WW et al. Ecacy
pare with methotrexate and cyclosporin as a corticosteroid-sparing of soluble IL-4 receptor for the treatment of adults with asthma.
agent in severe asthma? Biodrugs 8: 20515, 1997. J Allergy Clin Immunol 107: 96370, 2001.

696
Immunomodulators 55
34. Jiang H, Harris MB, Rothman P. IL-4/IL-13 signalling beyond JAK/ 55. Ying S, Meng Q , Zeibecoglou K, Robinson DS, Macfarlane A et al.
STAT. J Allergy Clin Immunol 105: 106370, 2000. Eosinophil chemotactic chemokines (eotaxin, eotaxin-2, RANTES,
35. Wills-Karp M, Chiaramonte M. Interleukin-13 in asthma. Curr Opin monocyte chemoattractant protein-3 (MCP-3), and MCP-4), and
Pulm Med 9: 2127, 2003. C-C chemokine receptor 3 expression in bronchial biopsies from atopic
36. Walter DM, McIntire JJ, Berry G, McKenzie AN, Donaldson DD and non-atopic (intrinsic) asthmatics. J Immunol 163: 632129, 1999.
et al. Critical role for IL-13 in the development of allergen-induced 56. Gonzalo JA, Pan Y, Lloyd CM, Jia GQ, Yu G et al. Mouse mono-
hyperreactivity. J Immunol 167: 466875, 2001. cyte-derived chemokine is involved in airway hyperreactivity and lung
37. Yang G, Volk A, Petley T, Emmell E, Giles-Komar J et al. Anti-IL-13 inammation. J Immunol 163: 40311, 1999.
monoclonal antibody inhibits airway hyperresponsiveness, inamma- 57. Kawasaki S, Takizawa H, Yoneyama H, Nakayama T, Fujisawa R et al.
tion and airway remodelling. Cytokine 28: 22432, 2004. Intervention of thymus and activation-regulated chemokine attenuates
38. Leigh R, Ellis R, Wattie J, Donaldson DD, Inman MD. Is interleukin-13 the development of allergic airway inammation and hyperresponsive-
critical in maintaining airway hyperresponsiveness in allergen-chal- ness in mice. J Immunol 166: 205562, 2001.
lenged mice?. Am J Respir Crit Care Med 170: 85156, 2004. 58. Bishop B, Lloyd CM. CC chemokine ligand 1 promotes recruitment
39. Wenzel S, Wilbraham D, Fuller R, Burmeister Getz E, Longphre M. of eosinophils but not Th2 cells during the development of allergic air-
Eect of an interleukin-4 variant on late phase response to allergen ways disease. J Immunol 170: 481017, 2003.
challenge in asthmatic patients: Results of two phase 2a studies. Lancet 59. Conroy DM, Jopling LA, Lloyd CM, Hodge MR, Andrew DP
370: 142231, 2007. et al. CCR4 blockade does not inhibit allergic airways inammation.
40. Bischo SC. Role of mast cells in allergic and non-allergic immune J Leukocyte Biol 74: 55863, 2003.
responses: Comparison of human and murine data. Nat Rev Immunol 60. Chung CD, Kuo F, Kumer J, Motani AS, Lawrence CE et al. CCR8 is
7: 93104, 2007. not essential for the development of inammation in a mouse model of
41. Cheng G, Arima M, Honda K, Hirata H, Eda F, Yoshida N et al. Anti- allergic airway disease. J Immunol 170: 58187, 2003.
interleukin-9 antibody treatment inhibits airway inammation and 61. Lukacs NW, Berlin A, Schols D, Skerlj RT, Bridger GJ. AMD3100, a
hyperreactivity in mouse asthma model. Am J Respir Crit Care Med CXCR4 antagonist, attenuates allergic lung inammation and airway
166: 40916, 2002. hyperreactivity. Am J Pathol 160: 135360, 2002.
42. Zhou Y, McLane M, Levitt RC. Th2 cytokines and asthma. Interleukin-9 62. Lams BEA, Sousa AR, Rees PJ, Lee TH. Immunopathology of the
as a therapeutic target for asthma. Respir Res 2: 8084, 2001. small airways submucosa in smokers with and without chronic obstruc-
43. Howarth PH, Babu KS, Arshad HS, Lau L, Buckley M et al. Tumour tive pulmonary disease. Am J Respir Crit Care Med 158: 151823, 1998.
necrosis factor (TNF-alpha) as a novel therapeutic target in sympto- 63. Lams BEA, Sousa AR, Rees PJ, Lee TH. Immunopathology of the
matic corticosteroid dependent asthma. Thorax 61: 101218, 2006. large airways submucosa in smokers with and without chronic obstruc-
44. Berry MA, Hargadon B, Shelley M, Parker D, Shaw SE et al. Evidence tive pulmonary disease. Eur Respir J 15: 51216, 2000.
of a role of tumour necrosis factor alpha in refractory asthma. N Engl J 64. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L et al.
Med 354: 697708, 2006. CD8 T lymphocytes in peripheral airways of smokers with chronic
45. Morjaria JB, Chauhan AJ, Bobu KS, Mehta Rl, Smith S et al. obstructive pulmonary disease. Am J Respir Crit Care Med 157: 82226,
Assessment of a soluble TNF-alpha receptor (etanercept) as a novel 1998.
therapeutic agent for severe refractory asthma (abstract). Proc Am 65. Elias JA, Kang MJ, Crouthers K, Homer R, Lee CG. State of the art.
Thorac Soc 3: A16, 2006. Mechanistic heterogeneity in chronic obstructive pulmonary disease:
46. Baraam B, Bird TG, Lambert-Van Der Brempt C, Campbell D, Foster SJ, Insights from transgenic mice. Proc Am Thorac Soc 3: 49498, 2006.
Maciewicz R. New alpha-substituted succinate-based hydroxamic acids as 66. Alsaeedi A, Sin DD, McAlister FA. The eects of inhaled corticoster-
TNF-alpha convertase inhibitors. J Med Chem 42: 4890908, 1999. oids in chronic obstructive pulmonary disease: A systematic review of
47. Fedorak RN, Ganl A, Elson CO, Rutgeerts P, Schreiber S et al. Recombinant randomized placebo-controlled trials. Am J Med 113: 5965, 2002.
human interleukin 10 in the treatment of patients with mild to moderately 67. Culpitt SV, Nightingale JA, Barnes PJ. Eect of high dose inhaled steroid
active Crohns disease. The Interleukin 10 Inammatory Bowel Disease on cells, cytokines and proteases in induced sputum in chronic obstructive
Cooperative Study Group. Gastroenterology 119: 147382, 2000. pulmonary disease. Am J Respir Crit Care Med 160: 163539, 1999.
48. Hawrylowicz C, Richards D, Loke TK, Corrigan C, Lee T. A defect in cor- 68. Hattotuwa Kl, Gizycki MJ, Ansari TW, Jeery PK, Barnes NC.
ticosteroid-induced IL-10 production in T lymphocytes from corticoster- The eects of inhaled uticasone on airway inammation in chronic
oid-resistant asthmatic patients. J Allergy Clin Immunol 109: 36970, 2002. obstructive pulmonary disease: A double-blind, placebos-controlled
49. Peek EJ, Richards DF, Faith A, Lavender P, Lee TH, Corrigan CJ, biopsy study. Am J Respir Crit Care Med 165: 159296, 2002.
Hawrylowicz CM. Interleukin-10 secreting regulatory T cells 69. Niewoehner DE. The role of systemic corticosteroids in acute exacer-
induced by glucocorticoids and 2-agonists. Am J Respir Cell Mol Biol bation of chronic obstructive pulmonary disease. Am J Respir Med 1:
33: 10511, 2005. 24348, 2002.
50. Xystrakis E, Kusumakar S, Boswell S, Peek E, Urry Z et al. Reversing 70. Aaron SD, Vandemheen KL, Hebert P et al. Outpatient oral pred-
the defective induction of IL-10-secreting regulatory T cells in gluco- nisone after emergency treatment of chronic obstructive pulmonary
corticoid-resistant asthma patients. J Clin Invest 116: 14655, 2006. disease. N Engl J Med 348: 261825, 2003.
51. Bryan SA, OConnor BJ, Matti S, Leckie MJ, Kanabar V et al. Eects 71. Burge PS, Calverley PMA, Jones PW, Spencer S, Anderson JA,
of recombinant interleukin-12 on eosinophils, airway hyperresponsive- Maslen T. Randomised, double-blind, placebo-controlled study of uti-
ness and the late asthmatic response. Lancet 356: 214953, 2000. casone propionate in patients with moderate to severe chronic obstruc-
52. Boguniewicz M, Martin RJ, Martin D, Gibson U, Celniker A, Williams M, tive pulmonary disease: The ISOLDE trial. BMJ 320: 1297303, 2000.
Leung DY. The eects of nebulised recombinant interferon-gamma in 72. Barnes PJ, Ito K, Adcock IM. A mechanism of corticosteroid resistance in
asthmatic airways. J Allergy Clin Immunol 95: 13335, 1995. COPD: Inactivation of histone deactylase. Lancet 363: 73133, 2004.
53. Gratzl S, Palca A, Schmitz M, Simon HU. Treatment with IFN-alpha 73. Keatings VM, Collins PD, Scott DM, Barnes PJ. Dierences in inter-
in corticosteroid-unresponsive asthma. J Allergy Clin Immunol 105: leikin-8 and tumour necrosis factor- in induced sputum from patients
103536, 2000. with chronic obstructive pulmonary disease or asthma. Am J Respir
54. Ying S, Robinson DS, Meng Q , Rottman J, Kennedy R et al. Crit Care Med 153: 53034, 1996.
Enhanced expression of eotaxin and CCR3 mRNA and protein in 74. Pitsiou G, Kyriazia G, Hatzizisi O, Argyropoulou P, Mavrofridis E,
atopic asthma. Association with airway hyperresponsiveness and pre- Patakas D. Tumour necrosis factor-alpha serum levels, weight loss and
dominant co-localization of eotaxin mRNA to bronchial epithelial and tissue oxygenation in chronic obstructive pulmonary disease. Respir
endothelial cells. Eur J Immunol 27: 350716, 1997. Med 96: 59498, 2002.

697
Asthma and COPD: Basic Mechanisms and Clinical Management

75. Rennard SI, Fogarty C, Kelsen S et al. The safety and ecacy of inf- 82. Rabe KF, Bateman ED, ODonnell D, Witte S, Bredenbroeker D,
liximab in moderate to severe chronic obstructive pulmonary disease. Bethke TD. Roumilast an oral anti-inammatory treatment for
Am J Respir Crit Care Med 175: 92634, 2007. chronic obstructive pulmonary disease: A randomised controlled trial.
76. Barnes PJ, Stockley RA. COPD: current therapeutic interventions and Lancet 366: 56371, 2005.
future approaches. Eur Respir J 25: 1084106, 2005. 83. Gamble E, Grootendorst DC, Brightling CE et al. Anti-
77. Mahler DA, Huang S, Tabrizi M, Bell GM. Ecacy and safety of a inammatory eects of the phosphodiesterase-4 inhibitor cilomilast
monoclonal antibody recognizing interleukin-8 in COPD: a pilot (Ario) in chronic obstructive pulmonary disease. Am J Respir Crit
study. Chest 126: 92634, 2004. Care Med 168: 97682, 2003.
78. Travesm SL, Culpitt S, Russell REK, Barnes PJ, Donnelly LE. Elevated 84. Giembycz MA. An update and appraisal of the cilomilast phase III
levels of the chemokines GRO- and MCP-1 in sputum samples from clinical development programme for chronic obstructive pulmonary
COPD patients. Thorax 57: 59095, 2002. disease. Br J Clin Pharmacol 62: 13852, 2006.
79. Donnelly LE, Barnes PJ. Chemokine receptors as therapeutic targets 85. Sandford AJ, Silverman EK. Chronic obstructive pulmonary disease. 1:
in chronic obstructive pulmonary disease. Trends Pharmacol Sci 27: Susceptibility factors for COPD: The genotype-environment interac-
54653, 2006. tion. Thorax 57: 73641, 2002.
80. Chung KF. Phosphodiesterase inhibitors in airways disease. Eur J 86. Dowson LJ, Guest PJ, Stockley RA. Longitudinal changes in physi-
Pharmacol 533: 11017, 2006. ological, radiological and health status measurements in alpha(1)-
81. Bousquet J, Aubier M, Sastre J, Izquierdo JL, Adler LM et al. Comparison antitrypsin deciency and factors associated with decline. Am J Respir
of roumilast, an oral anti-inammatory, with beclomethasone dipropion- Crit Care Med 164: 18059, 2001.
ate in the treatment of persistent asthma. Allergy 61: 7278, 2006.

698
Pulmonary Vasodilators in COPD
56 CHAPTER

INTRODUCTION pressure owing to the loss of vascular distensibil-


Richard N. Channick and
ity and the inability to recruit unused vascula-
ture. This process, by its very nature, is irreversible Lewis J. Rubin
Pulmonary hypertension is a common compli- [2]. However, there has not been found a clear Division of Pulmonary and Critical Care
cation of severe chronic obstructive pulmonary relationship between the severity of pulmonary
disease (COPD) and contributes substantively to Medicine, University of California, San
parenchymal destruction and the presence or
both its morbidity and mortality [1]. Although Diego School of Medicine, La Jolla,
severity of pulmonary hypertension. Several recent
generally mild, pulmonary hypertension in the investigations have found more primary vascular CA, USA
setting of COPD is a marker of poor progno- changes at the cellular and the molecular levels. In
sis. Furthermore, acute exacerbations of COPD one study, Kasahara and coworkers found signi-
which result in worsening gas exchange are likely cant abnormalities in vascular endothelial growth
to produce acute deteriorations in pulmonary factor expression in emphysema [3]. In addition,
hemodynamics, which further compromise oxy- abnormalities in endothelial mediators, such as
gen transport to peripheral tissues. Accordingly, endothelin 1 [4], and increases in proinamma-
considerable eort has been given to understand- tory cytokines, such as IL-6 and C-reactive pro-
ing the pathogenesis of pulmonary hypertension tein (CRP), have been found in COPD patients
in COPD and in attempting to develop strate- with pulmonary hypertension [5].
gies for its treatment.

Dynamic pulmonary
vasoconstriction
PATHOGENESIS OF PULMONARY
HYPERTENSION IN COPD The muscular pulmonary arteries respond with
constriction to a variety of stimuli that are com-
mon in COPD. Of these, hypoxia is the most
Several factors contribute to the development of potent and the frequent stimulus for pulmonary
pulmonary hypertension in COPD. vasoconstriction; hypoxic pulmonary vasoconstric-
tion (HPV) is unique to pulmonary artery smooth
muscle cells and is due to inhibition of an oxygen
Loss of cross-sectional vascular (or redox)-sensitive membrane-bound K chan-
surface area nel, leading to cell depolarization and increased
intracellular Ca concentrations [6]. Impaired
As a result of the widespread destruction of air- endothelial function may also be present and
ways and lung parenchyma typical of emphy- further contributes to the vasoconstriction result-
sema, there is also concomitant loss of vasculature, ing from diminished production of endothelial-
leading to an increase in perfusion pressure to derived relaxing factors such as nitric oxide (NO)
accommodate pulmonary blood ow. In addi- and prostacyclin (PGI2).
tion, increases in pulmonary blood ow, such as As a result of vasoconstriction and altered
those that occur with physical activity, are associ- endothelial function, severe COPD results in the
ated with further increases in pulmonary arterial elaboration of a variety of promoters of vascular

699
Asthma and COPD: Basic Mechanisms and Clinical Management

growth and remodeling. Among these, endothelin, platelet- an excessive vascular response to a stimulus, such as alveolar
derived growth factor, and transforming growth factor have hypoxia, leads to an exaggerated degree of pulmonary hyper-
been suggested to play major roles in the evolution of the tension in these patients. Another possibility is that these
chronically hypertensive pulmonary vascular bed. In concert, patients, in fact, have idiopathic pulmonary arterial hyperten-
these and other mitogens promote endothelial and smooth sion (PAH), completely distinct from the COPD.
muscle proliferation and extension of smooth muscle cells In addition, it has been shown that some patients with
into the smaller, peripheral vessels [7]. Although this remod- COPD manifest an excessive rise in pulmonary arterial pres-
eling process may be modied by restoration of normoxic sure during exercise. In one study, COPD patients with pol-
conditions, the amelioration is usually incomplete. ymorphisms in the angiotensin converting enzyme (ACE)
gene had greater exercise rises in pulmonary arterial pressure
(Fig. 56.2) [9], again pointing to an intrinsic susceptibility to
excessive pulmonary vascular disease in some COPD subjects.
PATHOPHYSIOLOGY OF PULMONARY Because it appears that some patients with COPD have
HYPERTENSION IN COPD more severe PH, it would seem likely that these patients would
be limited, not by the underlying lung disease, but by the pul-
monary vascular disease. It would then follow that benet from
As a result of the aforementioned processes, the narrowing
therapy directed at the PH could be realized. What data exist
of the vascular lumen produces elevations in pulmonary arte-
to support this hypothesis?
rial pressure and pulmonary vascular resistance. Furthermore,
pressures increase further with exercise or with acute exacer-
bations associated with deteriorations in gas exchange. The
increased right ventricular afterload may eventually lead to VASODILATOR THERAPY FOR PULMONARY
impaired right ventricular output and, if severe and progres- HYPERTENSION IN COPD
sive, right ventricular failure ensues. Although the term cor
pulmonale is frequently used to describe right heart failure, it
is more appropriately used to dene pulmonary vascular dis- Rationale
ease in the setting of respiratory disease; right heart failure is
a late and an unusual complication of cor pulmonale, particu- As mentioned, the rationale for the use of PAH-directed
larly in the present era of widespread availability of supple- therapy in COPD is based on the premise that pulmonary
mental oxygen therapy for chronic hypoxemia. vasculopathy and, possibly pulmonary vasoconstriction, are
How severe is the PH caused by COPD? The answer to key elements in its pathogenesis. In the older era, before
this question is both clinically relevant and debatable. What is the development of true PAH-targeted therapy, numerous
known is that in several large series of COPD patients with systemic vasodilator agents were evaluated. Indeed, a vari-
varying severity of disease, the average level of resting pul- ety of systemic antihypertensive agents have been shown to
monary arterial pressure is either normal or mildly elevated. reduce pulmonary arterial pressure in experimental animals
However, within these series, there are between 4% and 7% with pulmonary hypertension produced by acute hypoxic
of patients who have what could be termed disproportionate
pulmonary hypertension. Thabut and coworkers found 7.4% of
patients in this subgroup, dened as a mean pulmonary arterial P 0.0027
pressure of at least 40 mmHg with relatively modest severity of N.S. P 0.0049
COPD (Fig. 56.1) [8]. Although the mechanistic reasons for 60
this disproportionate PH are unclear, it seems plausible that
50

60
mean PAP [mmHg]

40
50
mean PAP [mmHg]

30
40 4

3 20
30
2
10
20 1

10 0
Before After Before After Before After
0 20 40 60 80 II ID DD
FEV1 (% pred.)
FIG. 56.2 Deletion polymorphisms in the ACE gene are associated with
FIG. 56.1 Spectrum of pulmonary arterial pressures in patients with COPD greater rise in mean pulmonary arterial pressures during exercise (from
(from Ref. [8]). Ref. [9]).

700
Pulmonary Vasodilators in COPD 56
ventilation or the administration of pulmonary vasoconstric- it may, in the majority of patients, merely serve as a marker
tor agents [10]. Unfortunately, as described above, the pul- of disease severity and not contribute substantively, per se,
monary hypertensive state in COPD is the result of more to symptom limitation or survival. Unlike patients with
than simple vasoconstriction, with remodeling playing a piv- other forms of pulmonary vascular disease, such as IPAH,
otal role. Thus, it would be conceptually unlikely that simple CTEPH, and connective tissue disease, patients with
vasodilators would achieve much ecacy. COPD are primarily symptom-limited not by the cardio-
vascular process but by the airway and the parenchymal lung
disease [24]. Right ventricular function is not normal, but
Oxygen is usually not severely depressed, although the compensa-
tory mechanisms may fall short during acute exacerbations
It is important to remember that supplemental oxygen, a selec- of airway disease. Accordingly, improving the pulmonary
tive pulmonary vasodilator, is the only pulmonary vasodilator hemodynamic state may not result in improved activity tol-
which has been demonstrated to improve survival in COPD, erance or survival in this disease.
although the hemodynamic eects are variable [1113]. It is Another concern regarding vasodilator use is that
likely that the favorable impact of oxygen therapy on survival COPD is typically a disease of older patients, a population
in COPD is due to a variety of eects, including optimizing that is also prone to have coexistent coronary artery disease.
myocardial and other peripheral tissue oxygen delivery. Vasodilators can increase cardiac work if tachycardia is pro-
duced, while at the same time they can also steal blood
ow away from areas of anatomic obstruction, leading to
Older vasodilators worsening cardiac ischemia.
Finally, systemically administered agents exert non-
Several older studies have evaluated other vasodilators in selective cardiovascular eects, including systemic hypo-
patients with COPD. Studies, until recently focused on oral tension, tachycardia, and potentially negative inotropic
vasodilators, including hydralazine [14], oral nitrates, and cal- myocardial eects. While these eects may be dose related,
cium channel antagonists [1522]. Several, small reports con- they can occur in patients even at low doses and can limit
sistently demonstrate a small, but signicant acute pulmonary the utility of vasodilators.
vasodilating eect from the administration of calcium channel In summary, presently available data suggest that oral
blockers, including nifedipine [1518, 22], felodipine [19, 20], calcium channel blockers do cause acute reductions in pul-
verapamil [21], and diltiazem [21]. The magnitude of this monary arterial pressure and pulmonary vascular resistance,
eect is quite variable, ranging from 10% to 30%, depending and modest increases in cardiac output in COPD patients.
on the study. In addition, nifedipine has been shown to reduce However, there is no convincing evidence of any improve-
PaO2, presumably by inhibition of HPV and worsening V/Q ment in exercise capacity or outcome in these patients.
matching, an eect which is magnied during exercise and
with supplemental oxygen administration [15]. Possibly miti-
gating the worsening of arterial oxygenation by nifedipine, New vasodilators
however, is data demonstrating improvement in cardiac output
and oxygen delivery during nifedipine administration [15, 22]. Recently, interest has begun to be focused on the utility of
Given the modest hemodynamic eects, at least at the modern PAH therapies in COPD patients with, what
rest, of oral vasodilators, and the relatively mild nature of is felt to be, more clinically signicant pulmonary hyper-
the pulmonary hypertension, at least at rest, observed in tension, the so-called outliers. These modern therapies
COPD, it reasonable to question whether oral vasodilators include prostacyclin analogs, endothelin receptor antago-
have any long-term clinical utility in these patients. To date, nists, and phosphodiesterase type 5 inhibitors (Table 56.1).
there have been no large placebo-controlled trials address- Unique features of these newer agents that might make
ing this issue. There are several controlled studies, all yield- them benecial in COPD patients with more advanced PH
ing unimpressive results. Vestri and coworkers found that, include their relative lack of systemic hemodynamic eects
compared to a well-matched control group, no signicant and their targeting of the pulmonary vasculopathy through
objective improvement occurred with 1 year of nifedipine direct eects on the endothelial dysfunction, which is a
[23]. Interestingly, a signicant improvement in dyspnea prominent feature of pulmonary hypertension.
was seen in the nifedipine group. Similarly, Domenighetti Although the data on the ecacy and safety of these
et al. found that, even in a subgroup of COPD patients with therapies for patients with IPAH and PAH associated with
marked acute pulmonary vasodilation in response to nifed- some other conditions, such as connective tissue disease,
ipine (43% decrease in PAP mean), typical clinical dete- have been clearly proven in several randomized, placebo-
rioration over 12 months occurred [17]. controlled trials [2530], there have been no such system-
Sajkov et al., in a 12-week study of another calcium atic trials in the COPD/PH subgroup. It should, however,
channel antagonist, felodipine, found reductions in pulmo- be noted that patients with modest degrees of COPD
nary arterial pressure and in pulmonary vascular resistance (FEV1 between 60% and 80% predicted) were not necessar-
that were felt to be signicant (22% and 30%, respectively) ily excluded from PAH trials, although the number of these
[19]. However, exercise capacity was not improved. patients was too small to allow subgroup analysis.
One theoretical limitation of pulmonary vasodilator There are, on the other hand, uncontrolled reports and
therapy, in general, is that although the presence of pulmo- case series reporting the eects of various PAH therapies in
nary hypertension is an ominous prognostic sign in COPD, COPD patients. Madden and coworkers treated 4 severe

701
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 56.1 FDA-approved therapies for pulmonary arterial for chronic therapy has shown some promise in patients with
hypertension. underlying lung disease. Acutely, in ARDS [36], marked
increases in PaO2 occur in over two-third of patients given
Prostacyclin analogs inhaled nitric oxide. The data in COPD, however, is conicting.
Epoprostenol (intravenous)
Both improvement [3739] and deterioration [40, 41] in rest-
Treprostinil (intravenous, subcutaneous)
ing arterial oxygenation have been reported in COPD patients
Iloprost (inhaled)
given inhaled NO, despite comparable degrees of pulmonary
vasodilation. These conicting results may, in fact, reect the
Endothelin receptor antagonists heterogeneity of the V/Q disturbances seen in COPD. For
instance, if more low V/Q areas are present, inhaled NO may
Bosentan
get into some of these lung units and actually worsen V/Q
Ambrisentan
matching, a phenomenon reproduced in an animal study by
Phosphodiesterase type 5 inhibitors Hopkins et al. [42]. On the other hand, if the predominant
disturbance is shunt, inhaled NO would tend to improve oxy-
Sildenafil
genation by shifting perfusion away from the shunt.
Additionally, the eects of inhaled NO on gas exchange
COPD patients with sildenal citrate for 8 weeks in an open during exercise may be even more important. Roger and col-
label trial [31]. These authors reported consistent but non- leagues found, in fact, that inhaled NO, despite worsening
statistically signicant improvements in pulmonary vascular oxygenation at rest, actually prevented exercise-induced O2
resistance and cardiac output as well as signicant increase in desaturation in patients with COPD, possibly due to pref-
6 minute walk distance. erential distribution of NO to well-ventilated regions during
In a larger, uncontrolled study of 18 COPD patients exercise [38].
(GOLD IIIV) with mild resting pulmonary hypertension However, despite potential benecial eects of inhaled
or exercise-induced pulmonary hypertension, a single dose NO during exercise in COPD patients, exercise limitation
of 50 mg of sildenal attenuated the rise in mean pulmonary is generally not due to either O2 desaturation or pulmonary
arterial pressure during exercise [32]. However, this eect was hypertension. Thus, it is uncertain whether or not chronic
not accompanied by an augmentation in cardiac output or an NO administration would benet these patients. Again, no
increase in maximal exercise capacity. In a recently published controlled trials have been done.
study by the same group, 15 stable COPD patients were
treated with 3 months of sildenal 50 mg three times daily
[33] No improvement in stroke volume as assessed by cardiac
MRI or exercise capacity were noted in this group. Although SUGGESTED GUIDELINES FOR THE USE OF
these small trials do not exclude an eect of sildenal in
COPD patients with PH, without a larger, controlled study,
VASODILATORS IN COPD
this agent should not be routinely used in this group.
Another group of drugs commonly used to treat Current experience does not support the widespread use of
PAH are the prostacyclin analogs, including epoprostenol, PAH therapy in COPD-related PH, owing to the hazards
treprostinil, and iloprost. A rationale exists for use of these described above and the dearth of studies demonstrating sus-
agents in COPD/PH patients, as one study has demon- tained clinical benet. Nevertheless, there are selected patients
strated reduction in prostacyclin synthase expression in the who appear to have alterations in pulmonary hemodynamics
small pulmonary arteries of patients with emphysema [34]. which are well out of proportion to the severity of chronic
However, to date no clinical data has been published on the respiratory disease and in whom conventional approaches
eects of prostacyclin analogs in COPD patients. Moreover, to therapy are only partially successful. These patients might
it has been the authors experience that intravenous epo- be considered as having true PAH, idiopathic or associated,
prostenol may lead signicant worsening of arterial hypox- and might be candidates for PAH therapy. However if used,
emia, presumably due to increased intrapulmonary shunting the eects of PAH therapy should be monitored using some
in patients with underlying ventilation/perfusion inequality. clinically meaningful parameter, such as objective testing of
Endothelin receptor antagonists, such as bosentan, cardiopulmonary exercise tolerance, noninvasive evaluation of
also have theoretical rationale in COPD/PH. Increase right heart size and function using echocardiography, or inva-
in endothelin expression has consistently been found in sive measurements of cardiopulmonary hemodynamics.
COPD patients with PH [4]. In addition, these levels have Theoretically, the use of pulmonary vasodilators in the
been found to correlate with exercise increases in pulmo- setting of acute cor pulmonale may be more appealing than
nary arterial pressures in COPD [35]. However, no clini- their use in the chronic state, owing to the acute and, there-
cal studies have yet been published examining the eects of fore potentially reversible component and the impact of this
endothelin receptor antagonists in COPD patients. alteration on morbidity and mortality. Unfortunately, no
Inhaled pulmonary vasodilators, such as iloprost or clinical trials have addressed the role of vasodilators in this
nitric oxide, theoretically are attractive candidates for treat- population; furthermore, the deleterious eects of vasodila-
ing COPD-related PH, as these agents often improve shunt tors may be particularly problematic in this setting. The use
fraction and have minimal systemic hemodynamic eects. of more selective agents, such as nitric oxide by inhalation,
However, no data have yet been published on inhaled ilo- would be preferred in the acutely decompensated patient.
prost in this group. Inhaled nitric oxide, although not available The impact of this therapy, however, remains unproven.

702
Pulmonary Vasodilators in COPD 56
SUMMARY AND CONCLUSIONS 15. Kennedy TP, Michael JR, Huang CK, Kallman CH, Zahka K, Schlott W,
Summer W. Nifedipine inhibits hypoxic pulmonary vasoconstriction
during rest and exercise in patients with chronic obstructive pulmo-
While pulmonary hypertension remains a serious complica- nary disease. A controlled double-blind study. Am Rev Respir Dis 129:
tion of COPD, eective therapy has been elusive. It is likely 54451, 1984.
16. Agostoni P, Doria E, Galli C, Tamborini G, Guazzi MD. Nifedipine
that pharmacological agents that exert both vasodilator and
reduces pulmonary presure and vascular tone during short- but not
antiproliferative eects are most likely to produce sustained long-term treatment of pulmonary hypertension in patients with chronic
improvement in cardiopulmonary hemodynamics, although obstructive pulmonary disease. Am Rev Respir Dis 139: 12025, 1989.
the impact of this therapy on symptoms and the survival in 17. Domenighetti GM, Saglini VG. Short- and long-term hemodynamic
COPD remain unknown. In addition, delivery of these agents efects of oral nifedipine in patients with pulmonary hypertension sec-
in a manner that facilitates selectivity of their eects to the ondary to COPD and lung brosis. Deleterious eects in patients with
pulmonary vasculature, such as by the inhaled route, would be restrictive disease. Chest 102: 70814, 1992.
preferable in order to minimize the adverse eects of systemic 18. Burghuber OC. Nifedipine attenuates acute hypoxic pulmonary vaso-
vasodilation. As our understanding of the molecular basis for constriction in patients with chronic obstructive pulmonary disease.
the pulmonary vascular remodeling in COPD unfold, targeted Respiration 52: 8693, 1987.
19. Sajkov D, McEvoy RD, Cowie RJ, Bradley JA, Antic R, Morris RG,
therapy should be feasible and could be combined with other
Frith PA. Felodipine improves pulmonary hemodynamics in chronic
modalities in order to both prevent and treat the vascular com- obstructive pulmonary disease. Chest 103: 135461, 1993.
plications of end-stage chronic obstructive pulmonary disease. 20. Bratel T, Hedenstierna G, Nyquist O, Ripe E. The use of a vasodila-
tor, felodipine as an adjuvant to long-term oxygen treatment in COLD
patients. Eur Respir J 3: 4654, 1990.
References 21. Gassner A, Sommer G, Fridrich L, Magometschnigg D, Priol A.
Dierential therapy with calcium antagonists in pulmonary hypertension
1. Traver G, Kline M, Burrows B. Predictors of mortality in chronic secondary to COPD. Hemodynamic eects of nifedipine, diltiazem, and
obstructive pulmonary disease. Am Rev Respir Dis 119: 895902, 1979. verapamil. Chest 98: 82934, 1990.
2. Rubin LJ, Tod ML, Yoshimura K. Eects of nitrendipine and hypoxia 22. Saadjian AY, Philip-Joet FF, Vestri R, Arnaud AG. Long-term treat-
on pulmonary vascular resistance in emphysema. Am Rev Respir Dis ment of chronic obstructive lung disease by Nifedipine: An 18-month
142: 62530, 1990. haemodynamic study. Eur Respir J 1: 71620, 1988.
3. Kasahara Y, Tuder RM, Cool CD, Lynch DA, Flores SC, Voelkel NF. 23. Vestri R, Philip-Joet F, Surpas P, Arnaud A, Saadjian A. One-year clin-
Endothelial cell death and decreased expression of vascular endothe- ical study on nifedipine in the treatment of pulmonary hypertension in
lial growth factor and vascular endothelial growth factor receptor 2 in chronic obstructive lung disease. Respiration 54: 13944, 1988.
emphysema. Am J Respir Crit Care Med 163: 73744, 2001. 24. Rubin LJ. Primary pulmonary hypertension. N Engl J Med 336:
4. Bacakoglu F, Atasever A, Ozhan MH, Gurgun C, Ozkilic H, Guzelant A. 11117, 1997.
Plasma and bronchoalveolar lavage uid levels of endothelin-1 in 25. Barst RJ, Rubin LJ, Long WA, McGoon MD, Rich S, Badesch DB,
patients with chronic obstructive pulmonary disease and pulmonary Groves BM, Tapson VF, Bourge RC, Brundage BH. A comparison of con-
hypertension. Respiration 70(6): 59499, 2003 NovemberDecember. tinuous intravenous epoprostenol (prostacyclin) with conventional therapy
5. Joppa P, Petrasova D, Stancak B, Tkacova R. Systemic inammation in for primary pulmonary hypertension. N Engl J Med 334: 296301, 1996.
patients with COPD and pulmonary hypertension. Chest: 32633, 2006. 26. Simonneau G, Barst RJ, Galie N, Naeije R, Rich S, Bourge RC, Keogh A,
6. Wang J, Juhaszova M, Rubin LJ et al. Hypoxia inhibits gene expression Oudiz R, Frost A, Blackburn SD, Crow JW, Rubin LJ Treprostinil
of voltage-gated K channel alpha subunits in pulmonary artery smooth Study Group. Continuous subcutaneous infusion of treprostinil, a pros-
muscle cells. J Clin Invest 100: 234753, 1997. tacyclin analogue, in patients with pulmonary arterial hypertension: A
7. Voelkel NF, Tuder RM. Hypoxia-induced pulmonary vascular remode- double-blind, randomized, placebo-controlled trial. Am J Respir Crit
ling: A model for what human disease? J Clin Invest 106: 73338, 2000. Care Med 15;165: 8004, 2002 March.
8. Thabut G, Dauriat G, Stern JB, Logeart D, Levy A, Marrash-Chahla R, 27. Channick RN, Simonneau G, Sitbon O, Robbins IM, Frost A,Tapson VF,
Mal H. Pulmonary hemodynamics in advanced COPD candidates for Badesch DB, Roux S, Rainisio M, Bodin F, Rubin LJ. Eects of the
lung volume reduction surgery or lung transplantation. Chest 127(5): dual endothelin-receptor antagonist bosentan in patients with pul-
153136, 2005. monary hypertension: A randomised placebo-controlled study. Lancet
9. Kanazawa H, Okamoto T, Hirata K, Yoshikawa J. Deletion polymorphisms 358(9288): 111923, 2001 October 6.
in the angiotensin converting enzyme gene are associated with pulmonary 28. Rubin LJ, Badesch DB, Barst RJ, Galie N, Black CM, Keogh A, Pulido
hypertension evoked by exercise challenge in patients with chronic obstruc- T, Frost A, Roux S, Leconte I, Landzberg M, Simonneau G. Bosentan
tive pulmonary disease. Am J Respir Crit Care Med 162: 123538, 2000. therapy for pulmonary arterial hypertension. N Engl J Med 346(12):
10. Young T, Lundquist L, Chesler E, Weir E. Comparative eects of 896903, 2002 March 21.
nifedipine, verapamil and diltiazem on experimental pulmonary hyper- 29. Gali N, Ghofrani HA, Torbicki A, Barst RJ, Rubin LJ, Badesch D,
tension. Am J Cardiol 51: 195200, 1983. Fleming T, Parpia T, Burgess G, Branzi A, Grimminger F, Kurzyna M,
11. Long-term domiciliary oxygen therapy in chronic hypoxic cor pulmonale Simonneau G Sildenal Use in Pulmonary Arterial Hypertension
complicating chronic bronchitis and emphysema. Report of the Medical (SUPER) Study Group. Sildenal citrate therapy for pulmonary arte-
Research Council Working Party. Lancet 1(8222): 681686, 1981. rial hypertension. N Engl J Med 353(20): 214857, 2005 November 17.
12. Tims R, Khaja F, Williams G. Hemodynamic response to oxygen 30. Olschewski H, Simonneau G, Gali N, Higenbottam T, Naeije R, Rubin LJ,
therapy in chronic obstructive pulmonary disease. Ann Intern Med 102: Nikkho S, Speich R, Hoeper MM, Behr J, Winkler J, Sitbon O, Popov W,
2936, 1985. Ghofrani HA, Manes A, Kiely DG, Ewert R, Meyer A, Corris PA,
13. Weitzenblum E, Oswald M, Mirhom R, Kessler R, Apprill M. Delcroix M, Gomez-Sanchez M, Siedentop H, Seeger W. Aerosolized
Evolution of pulmonary haemodynamics in COLD patients under Iloprost Randomized Study Group. Inhaled iloprost for severe pulmonary
long-term oxygen therapy. Eur Respir J 2(Suppl 7): 669S673S, 1989. hypertension. N Engl J Med 347(5): 32229, 2002 August 1.
14. Dal Nogare A, Rubin LJ. Eects of hydralazine on exercise capacity in 31. Madden BP, Allenby M, Loke TK, Sheth A. A potential role for silde-
pulmonary hypertension secondary to chronic obstructive pulmonary nal in the management of pulmonary hypertension in patients with
disease. Am Rev Respir Dis 133: 38589, 1986. parenchymal lung disease. Vascul Pharmacol 44: 37276, 2006.

703
Asthma and COPD: Basic Mechanisms and Clinical Management

32. Holverda S, Rietema H, Bogaard HJ, Westerhof N, Postmus PE, to infusion of acetylcholine and inhalation of nitric oxide in patients
Boonstra A, Vonk-Noordegraaf A. Acute eects of sildenal on exer- with chronic obstructive lung disease and pulmonary hypertension. Am
cise pulmonary hemodynamics and capacity in patients with COPD. Rev Respir Dis 148: 31016, 1993.
Pulm Pharmacol Ther, 2008., epub ahead of print 38. Roger N, Barbera JA, Roca J, Rovira I, Gomez FP, Rodriguez-Roisin R.
33. Rietema H, Holverda S, Bogaard HJ, Marcus JT, Smit HJ, Westerhof N, Nitric oxide inhalation during exercise in chronic obstructive pulmo-
Postmus PE, Boonstra A, Vonk-Noordegraaf A. Sildenal treatment in nary disease. Am J Respir Crit Care Med 156: 8006, 1997.
COPD does not aect stroke volume or exercise capacity. Eur Respir J 39. Yoshida M, Taguchi O, Gabazza EC, Kobayashi T, Yamakami T,
31(4): 75964, 2008 April. Kobayashi H, Maruyama K, Shima T. Combined inhalation of nitric
34. Lee JD, Taraseviciene-Stewart L, Keith R, Geraci MW, Voelkel NF. oxide and oxygen in chronic obstructive pulmonary disease. Am J Respir
The expression of prostacyclin synthase is decreased in the small pul- Crit Care Med 155: 52629, 1997.
monary arteries from patients with emphysema. Chest 128(Suppl 6): 40. Moinard J, Manier G, Pillet O, Castaing Y. Eect of inhaled nitric
575S, 2005 December. oxide on hemodynamics and VA/Q inequalities in patients with chronic
35. Yamakami T, Taguchi O, Gabazza EC, Yoshida M, Kobayashi T, obstructive pulmonary disease. Am J Respir Crit Care Med 149: 148287,
Kobayashi H, Yasui H, Ibata H, Adachi Y. Arterial endothelin-1 level in 1994.
pulmonary emphysema and interstitial lung disease. Relation with pul- 41. Barbera JA, Roger N, Roca J, Rovira I, Higenbottam TW, Rodriguez-
monary hypertension during exercise. Eur Respir J 10(9): 205560, 1997 Roisin R. Worsening of pulmonary gas exchange with nitric oxide inhala-
September. tion in chronic obstructive pulmonary disease. Lancet 347: 43640, 1996.
36. Rossaint R, Falke KJ, Lopez F, Slama K, Pison U, Zapol WM. Inhaled 42. Hopkins SR, Johnson EC, Richardson RS, Wagner H, De Rosa M,
nitric oxide for the adult respiratory distress syndrome. N Engl J Med Wagner PD. Eects of inhaled nitric oxide on gas exchange in lungs
328: 399405, 1993. with shunt or poorly ventilated areas. Am J Respir Crit Care Med 156:
37. Adnot S, Kouyoumdjian C, Defouilloy C, Andrivet P, Sediame S, 48489, 1997.
Herigault R, Fragacci MD. Hemodynamic and gas exchange responses

704
Ventilator Support
57
CHAPTER

In patients with obstructive lung disease, the supported, spontaneous), as well as inspiratory
development of acute respiratory failure is a phase variables.
common cause for admission to the intensive Samuel L. Krachman1
care unit (ICU) [18]. Between 2% and 61% of Controlled ventilation and Martin J. Tobin2
patients [1, 2, 49] require mechanical ventila- With controlled mechanical ventilation, the 1
Section of Pulmonary and Critical Care
tion, with an associated mortality of 630% [6 ventilator delivers all breaths at a preset rate and
Medicine, Temple University School of
12]. The high mortality may be a consequence the patient cannot trigger the machine. With
of both disease severity and complications of volume-controlled ventilation, breaths have a Medicine, Philadelphia, PA, USA
2
mechanical ventilation. preset volume (volume targeted), whereas with Loyola University of Chicago, Stritch
pressure-controlled ventilation breaths are pres- School of Medicine and Hines Veterans
sure limited and time cycled [18]. Use of vol- Administration Hospital, Maywood,
INDICATIONS FOR VENTILATOR ume-controlled ventilation is largely restricted
to patients who are apneic as a result of brain
IL, USA

SUPPORT damage, sedation, or paralysis.


Assist-control ventilation
When severe hypoxemia develops in patients
with asthma [13] and chronic obstructive pul- In the assist-control (AC) mode, the ventilator
monary disease (COPD) [14], intubation and delivers a breath either when triggered by the
mechanical ventilation may be necessary to patients inspiratory eort or independently if
ensure the delivery of a suciently high frac- such an eort does not occur within a prese-
tional inspired oxygen concentration (F1O2). lected period. All breaths are delivered under
The development of an acute respiratory acido- positive pressure by the machine, but unlike
sis is another major indication for mechanical controlled ventilation the patients triggering
ventilation [15], although simpler measures can eorts commonly exceed the preset rate. The
sometimes reverse the process [16]. An increase amount of active work performed by a patient
in work of breathing, secondary to an increase in is critically dependent on the trigger sensitivity
airway resistance [17] or an inspiratory threshold and inspiratory ow settings. Even when these
load associated with auto-positive end-expiratory settings are optimized, patients actively perform
pressure or intrinsic positive end-expiratory pres- about one-third of the work performed by the
sure (PEEPi), often requires mechanical ventila- ventilator during passive conditions [19, 20].
tion to rest the respiratory muscles and decrease
Intermittent mandatory ventilation
the oxygen cost of breathing.
With intermittent mandatory ventilation (IMV),
the patient receives periodic positive-pressure
breaths from the ventilator at a preset volume and
INVASIVE POSITIVE PRESSURE rate, but the patient can also breathe spontane-
VENTILATION ously between these ventilator breaths [21]. With
synchronized IMV, the ventilator waits until the
patient begins to inhale and then synchronizes
Modes of mechanical ventilation the machine breath with the patients inspiratory
eort. If a patient does not make an eort within
The term mode refers to the relationship among a preset time, the ventilator delivers a positive-
various breath types (mandatory, assisted, pressure breath.
705
Asthma and COPD: Basic Mechanisms and Clinical Management

With IMV, patients frequently have diculty in adapt- 400


ing to the intermittent nature of assistance. It was previously IMV
assumed that respiratory muscle rest was proportional to the 350 PS
number of machine-assisted breaths. Recent studies, however, IMV  PS 10 cm H2O
indicate that inspiratory eort is equivalent for spontaneous 300

PTP/min, cm H2O s/min


and assisted breaths [2224]. At a moderate level of machine
250
assistance, that is, where the ventilator accounted for 2050%
of the total ventilation, electromyographic activities of the 200
diaphragm and sternomastoid muscles were equivalent for
assisted and spontaneous breaths [23]. These ndings suggest 150
that respiratory center output is preprogrammed and is una-
ble to adjust to breath-to-breath changes in unloading. As a 100
result, IMV may contribute to the development of respiratory
muscle fatigue or prevent its recovery. 50

0
Pressure support ventilation 0 20 40 60 80 100
Pressure support (PS) ventilation is patient-triggered like Percent support by primary mode
AC and IMV, but diers in being pressure targeted and ow
cycled [25]. The physician sets a level of pressure that aug- FIG. 57.1 At proportional levels of ventilator assistance, work, as measured
by the pressuretime product (PTP/min), was significantly lower for the
ments every spontaneous eort, and the patient can alter
combination of IMV and PS of 10 cm H2O than for either PS or IMV alone
respiratory frequency, inspiratory time, and tidal volume. A (from Ref. [20] with permission).
fall in airway pressure triggers the ventilator, which in turn
increases pressure to a preset value. Accordingly, the pres- to 30/min was previously found to be a better predictor of
sure to inate the lungs and chest wall is provided jointly by a decrease in the work of breathing than achieving a tidal vol-
the ventilator and the patient. Unlike the guaranteed volume ume of 0.6 l [26]. Increasing PS to achieve a low respiratory
achieved by AC and IMV, tidal volume varies depending on frequency and inspiratory muscle unloading may be accompa-
the set pressure, patient eort, and pulmonary mechanics. nied by an increase in expiratory muscle activity, causing the
The lack of guaranteed assistance in the absence of patient patient to ght the ventilator [26]. Thus, selecting the optimal
eort can cause apnea in patients with an unstable respiratory level of PS can be quite complex.
center output. Cycling to exhalation is triggered by a decrease
in inspiratory ow to a preset level, such as 5 l/min or 25%
of the peak inspiratory ow. Inspiratory assistance can also Comparison between modes of mechanical
be terminated by a small increase in pressure (13 cm H2O)
above the preset level, resulting from expiratory eort [25]. ventilation
PS is very eective in decreasing the work of inspiration.
The degree of inspiratory muscle unloading, however, is varia- A head-to-head comparison of the common modes of
ble, with a coecient of variation of up to 96% among patients assisted ventilation was previously conducted, with most
[26]. In patients with COPD, the variability in the work of the patients having COPD [24]. Compared with spon-
of breathing can be explained by the inspiratory resistance, taneous breathing, AC achieved the largest decrease in the
minute ventilation, and degree of PEEPi [26]. It is notewor- work of breathing. The decrease in the work was greater
thy that, PS does not decrease PEEPi in patients with COPD for PS than for IMV at lower levels of support. Addition
[26, 27]. Instead, increasing levels of PS increase the contribu- of PS of 10 cm H2O to a given level of IMV caused a
tion from PEEPi to the overall work of breathing, accounting greater reduction in the work of breathing (Fig. 57.1), not
for approximately two-thirds of the inspiratory eort [26]. The only during the intervening PS breaths, but also during
algorithm used to terminate inspiratory assistance during PS the mandatory IMV breaths. The decrease in work during
can also aect the work of breathing in patients with COPD. the mandatory breaths was proportional to the decrease in
Patients with a prolonged time constant require more time for respiratory drive during the intervening breaths.
ow to fall to this threshold, and consequently, mechanical
ination may persist into neural expiration. To counteract such
neuralmechanical dysynchrony, patients may activate their
expiratory muscles at a time when the ventilator is still inat- VENTILATOR SETTINGS
ing the thorax, causing the patient to ght the ventilator.
There is no consensus for the appropriate level of PS
for an individual patient. In one study, the level minimizing Fractional inspired oxygen concentration
activity of the sternomastoid muscles also reversed electro-
myographic evidence of excessive diaphragmatic stress [25]. The lowest FIO2 that achieves satisfactory arterial oxygena-
Commonly, PS is titrated to achieve a decrease in respira- tion should be selected. With arterial blood samples, a SaO2
tory frequency and an increase in tidal volume. Considerable target of 90% is appropriate, but with pulse oximetry the
discrepancy exists among studies as to the preferred target or same target can be associated with PaO2 values as low as 41
optimal level of PS. Achieving a decrease in respiratory rate torr in black patients [28].

706
Ventilator Support 57
Trigger sensitivity failure, pulmonary barotrauma and hypotension were signi-
cantly lower in patients who were electively hypoventilated.
Most ventilators employ pressure triggering, whereby a
decrease in circuit pressure is required to initiate the ven- Respiratory rate
tilator. With ow triggering (sometimes termed ow-by ),
a base ow of gas (usually set at 5 to 20 l/min) is delivered Setting the ventilator rate depends on the mode being
during both the expiratory and the inspiratory phases of employed. With AC ventilation, the ventilator supplies a
the respiratory cycle [29]. With patient eort, gas enters the breath in response to each patient eort; the backup rate
patients lungs and is diverted from the exhalation port. The should be set at approximately four breaths below the
dierence between inspiratory and expiratory base ow is patients spontaneous rate. With IMV, the mandatory rate
sensed, causing the ventilator to switch phase; sensitivity is is initially set high and then gradually decreased accord-
usually set at 2 l/min. A decrease in breathing eort is noted ing to patient tolerance. As discussed above, this common
with ow triggering in the PS, but not the AC, mode [30]. approach does not ensure adequate rest. With pressure-sup-
Triggering the ventilator is more dicult in patients port ventilation, the ventilator rate is not set.
with dynamic hyperination. In this situation, the patient
must rst generate sucient pressure to oset the elastic
recoil associated with hyperination, and thereafter overcome Inspiratory flow rate
the sensitivity threshold. With all modes of ventilation, inef-
fective triggering increases in proportion to the level of assist- In patients with COPD, increasing inspiratory ow to 100 l/
ance, secondary to a decrease in respiratory drive [24]. Breaths min produces better gas exchange, probably because the result-
preceding the non-triggered eort have a higher volume, a ing increase in expiratory time allows more complete emptying
shorter expiratory time, and a higher PEEPi. of gas-trapped regions [35]. Studies in healthy subjects [36],
as well as in intubated patients [37], have demonstrated that
increasing the inspiratory ow setting causes an immediate
Tidal volume increase in respiratory frequency and respiratory drive. Yet, it
has been demonstrated that imposed ventilator inspiratory
To prevent alveolar overdistension and lung injury, tidal vol- time, independent of delivered inspiratory ow and tidal vol-
umes of 57 ml/kg (or less) have become increasingly popular ume, can determine respiratory frequency [38]. More recently,
[31]. This ventilator strategy is termed permissive hypercap- in patients with COPD, it was demonstrated that the increase
nia or controlled hypoventilation [32], because it commonly in respiratory rate noted as inspiratory ow was increased, was
produces an increase in PaCO2. If pH falls below 7.20, some associated with an increase in expiratory time and a decrease
physicians administer intravenous bicarbonate, although in PEEPi [39] (Fig. 57.2). A shortened inspiratory time com-
its benet is unproven [32]. In patients with severe asthma bined with time-constant inhomogeneity of COPD will cause
requiring mechanical ventilation, most studies suggest that overination of some lung units to persist into neural expira-
permissive hypercapnia decreases mortality [33, 34]. In a con- tion. Continued ination during neural expiration causes stim-
trolled study [4] of patients with asthma and acute respiratory ulation of the vagus, which prolongs expiratory time [40].

90 L/min 60 L/min 30 L/min


2

Flow
0
L/sec

2

40 Swing 16.8 19.5 21.5


Esophageal
Pressure 0
cm H2O
40 PEEPI13.3 14.4 15.6

0.6
Chest
Volume 0
a.u.
0.6
0 10 20 30
Time, seconds

FIG. 57.2 Continuous recordings of flow, esophageal pressure (Pes), and the sum of ribcage and abdominal motion, in a patient with COPD receiving AC
ventilation at a constant tidal volume. As flow increased from 30 to 60 and 90 l/min (from right to left), frequency increased (from 18 to 23 and 26 breaths/
min, respectively), PEEPi decreased (from 15.6 to 14.4 and 13.3 cm H2O, respectively), and end-expiratory lung volume also fell. Increases in flow from 30 to 60
and 90 l/min also led to decreases in the swings in Pes from 21.5 to 19.5 and 16.8 cm H2O, respectively (from Ref. [39] with permission).

707
Asthma and COPD: Basic Mechanisms and Clinical Management

POSITIVE END-EXPIRATORY PRESSURE 20


4 8 16

In patients with COPD, dynamic hyperination results **P  0.001


from dynamic airway collapse. The accompanying increase 18

Rrsmax, cm H2O/L/ses
in alveolar pressure is termed PEEPi. To trigger the
ventilator in this situation, the patient must rst generate a
** ** ** **
negative inspiratory pressure equal in magnitude to PEEPi ** **
and then overcome the trigger sensitivity setting. Provided 16 ** ** ** **
the increase in PEEPi is secondary to increased elastic recoil,
rather than expiratory eort, the addition of external PEEP
can decrease the inspiratory threshold load [41,42].
Deciding the appropriate amount of external PEEP 14
in patients with airow limitation can be dicult because
of regional inhomogeneities among lung units, each with its
own critical closing pressure [43]. If application of external
PEEP causes an increase in end-expiratory lung volume, 0
0 5 15 20 40 60 80
decreases in cardiac output and blood pressure are likely to
follow [4447]. In general, a level of external PEEP, approxi- Time, min
mately 70% of the PEEPi value, is used in patients with FIG. 57.3 In mechanically ventilated patients with COPD, four puffs
COPD [4447]. In contrast to patients with airow limi- of albuterol with an MDI resulted in a significant decrease in maximal
tation secondary to COPD, PEEP should be avoided in inspiratory airway resistance (Rrsmax) within 5 min, with no further
patients with asthma, because it is likely to increase lung improvement after a total of 28 puffs (from Ref. [41] with permission).
volume and decrease cardiac output [48].
[55, 57, 58]. In patients receiving noninvasive ventilation,
heliox does not decrease the intubation rate or the length of
stay in the ICU [59].
ANCILLARY THERAPY While previously investigated in patients with acute
lung injury [60], prone positioning has more recently
Studies have established that it is possible to achieve satis- been examined in intubated patients with COPD during
factory bronchodilation using a metered-dose inhaler (MDI) an acute exacerbation. While improvements in oxygenation
despite the presence of an endotracheal tube in mechanically [61, 62], as well as chest wall and lung mechanics [62] have
ventilated patients. It is essential to follow a specied proto- been reported, these studies were uncontrolled and involved
col, use an inline chamber device [49], and actuate the MDI small numbers of patients.
at the onset of inspiratory airow. Maximal bronchodilation
can be achieved in patients with COPD with as few as four
pus of a sympathomimetic aerosol (Fig. 57.3) [50].
Analgesic, anxiolytic, and neuromuscular blocking COMPLICATIONS
agents are frequently used in mechanically ventilated patients.
When selecting an analgesic in a patient with asthma, use of Patients receiving mechanical ventilation are at risk of
morphine should be avoided because of the risk of histamine numerous complications, including oxygen toxicity, air leaks,
release and worsening bronchospasm [51]. Neuromuscular infection, endotracheal tube complications, and decreased
blocking agents can result in prolonged paralysis and the cardiac output. These problems occur frequently and they
development of an acute myopathy [52, 53]. The concurrent can be life threatening if not promptly detected and treated.
use of corticosteroids appears to increase the risk. The inad-
vertent discontinuation of mechanical ventilation in patients
receiving a neuromuscular blocking agent will cause complete Barotrauma
apnea, leading to cardiopulmonary arrest. Train-of-four mon-
itoring is recommended while patients receive neuromuscular The development of extraalveolar air, in the form of pneu-
blocking agents, although clinical assessment at the bedside momediastinum, subpleural air cysts, subcutaneous emphy-
is equally important [54]. sema, pneumothorax, or pneumoperitoneum is termed
Heliox is a mixture of either 70% or 80% helium pulmonary barotrauma. These ndings have been found in
with oxygen, that has a density one-third that of room air. 1427% of patients with obstructive lung disease receiving
It has been theorized that heliox may decrease the work of mechanical ventilation [4, 11, 12].
breathing and improve gas exchange during an acute exac-
erbation in patients with asthma and COPD. Controlled
studies involving noninvasive ventilation or intubated Infection
patients have demonstrated mixed results [5558]; with
only small changes in PaCO2 [56] and hemodynamics [55], Pneumonia develops in 21% of patients receiving mechani-
and a slight decrease in the work of breathing and PEEPi cal ventilation for over 48 h [63]. In mechanically ventilated

708
Ventilator Support 57
patients with COPD, more than 40% of the patients have the development of hypercapnia and/or hypoxemia [74].
potentially pathogenic microorganisms present in their An increase in right and left ventricular afterload during
upper and lower airways, and have an increased risk of life- a weaning trial may aect how the cardiovascular system
threatening pneumonia [64]. The diagnosis of pneumonia is meets oxygen demand in weaning failure patients, and con-
often dicult in patients receiving mechanical ventilation, tributes to the development of hypoxemia [75]. Rigorous
because clinical criteria are unreliable and airway coloniza- studies of the pathophysiology of weaning failure in patients
tion confounds the identication of causative organisms. with asthma have not been conducted.
Studies comparing the use of quantitative cultures of
specimens obtained by bronchoalveolar lavage or with a
protected specimen brush passed through the bronchoscope Timing of weaning process
to nonquantitative culture of endotracheal aspirates have
demonstrated conicting results [6567]. Therefore, the Previous studies emphasize that clinical bedside assessment and
optimal approach to diagnosis ventilator-associated pneu- physician judgment are insucient in determining the appro-
monia is yet to be determined. priate time to initiate weaning [76]. Weaning involves the use
of three diagnostic tests in sequence: measurement of predictors,
a weaning trial, and a trial of extubation. The fundamental job
Effects on cardiac output of a weaning-predictor test is screening; with an ideal screen-
ing test having a very high sensitivity [77]. The best predictor
Both PEEP and PEEPi can cause similar decreases in of weaning outcome is the measurement of the frequency-
cardiac output, primarily by decreasing venous return. In to-tidal volume ratio, a measure of rapid shallow breathing.
addition, increases in pulmonary vascular resistance, sec- A frequency-to-tidal volume ratio above 100 breaths/min/l
ondary to alveolar distension and stretching of adjacent suggests that weaning is not likely to be successful [78].
vessels, increases right ventricular afterload. As a result, the
interventricular septum may shift to the left and decrease
left ventricular compliance. Distended lung parenchyma Weaning techniques
may also increase juxtacardiac pressure and decrease left
ventricular compliance. PEEPi has been noted in patients When a screening test is positive, the clinician proceeds
with COPD experiencing hemodynamic embarrassment to a conrmatory test; an ideal conrmatory test has a
[68]. On discontinuation of mechanical ventilation, hemo- very low rate of false-positive results. That is, a high spe-
dynamics improved as the amount of PEEPi decreased. cicity. Four weaning techniques are generally used. With
IMV and PS, the level of ventilator assistance is gradually
reduced. PS became popular as a means of overcoming the
resistance of the endotracheal tube. This line of thinking,
WEANING however, ignored the fact that the upper airway becomes
swollen when an endotracheal tube has been in place, and
after extubation patients will experience increased upper
About 70% of patients tolerate the rst attempt to discontinue airway resistance. Indeed, the work of breathing following
mechanical ventilation [69, 70]. Nevertheless, about 4060% of extubation is similar to that while breathing on a T-piece
total ventilator time is devoted to the process of weaning. [79]. With IMV, at respiratory rates of 14 breaths/min or
less, patient inspiratory eorts are increased to a level likely
to cause respiratory muscle fatigue [22, 23].
Pathophysiology of weaning failure Several randomized trials have been performed com-
paring weaning techniques [69, 80]. IMV has been shown to
In patients with COPD, a decrease in respiratory motor delay the weaning process. A once-a-day trial of spontaneous
output appears to be a rare cause for failure to wean [17]. breathing resulted in a threefold increase in the rate of suc-
The major reason that patients with COPD fail weaning tri- cessful weaning compared with IMV, and a twofold increase
als is because of a progressive increase in work of breathing in successful weaning compared with PS. Performing trials of
secondary to increases in resistance, elastance, and PEEPi. spontaneous breathing once a day is as eective as perform-
In contrast, pulmonary mechanics during passive ventilation ing such trials several times a day [69], but much simpler. In
are virtually indistinguishable in weaning success and wean- patients not expecting to pose any particular diculty with
ing failure patients before a weaning trial [71]. Respiratory weaning, a half-hour trial of spontaneous breathing is as
muscle weakness is commonly seen in patients that fail a eective as a 2-h trial [70]. The use of protocol-driven algo-
weaning trial [72]. However, the development of respiratory rithms for weaning has not shown superiority in shortening
muscle fatigue does not appear to be responsible for wean- the time to successful extubation [8186].
ing failure [72]. One reason may be that failure patients Before removing the endotracheal tube, the clini-
become progressively distressed during a weaning trial, lead- cian must judge whether or not the patient will be able to
ing clinicians to reinstate ventilator support before patients maintain a patent upper airway after extubation. Mortality
have breathed long enough to develop fatigue [72, 73]. among patients that require reintubation is more than six
That is, monitoring clinical signs of distress provides suf- times as high as mortality among patents who can tolerate
cient warning to avoid respiratory muscle fatigue. Only a extubation [70]. The reason is unknown, but may reect a
fraction of patients develop abnormal gas exchange, with greater severity of the underlying illness [87].

709
Asthma and COPD: Basic Mechanisms and Clinical Management

NONINVASIVE POSITIVE PRESSURE


12. Mansel JK, Stogner SW, Petrini MF et al. Mechanical ventilation in
patients with acute severe asthma. Am J Med 89: 4248, 1990.
VENTILATION 13. Rodriguez-Roisin R, Ballester E, Roca J et al. Mechanisms of hypox-
emia in patients with status asthmaticus requiring mechanical ventila-
tion. Am Rev Respir Dis 139: 73239, 1989.
In patients with COPD and acute respiratory failure, the 14. Wagner PD, Dantzker DR, Dueck R et al. Ventilation-perfusion
use of noninvasive positive pressure ventilation results in less inequality in chronic obstructive pulmonary disease. J Clin Invest 59:
frequent intubation, decreased complications, and a shorter 20316, 1977.
hospital stay. More recently, noninvasive positive pressure 15. Torres A, Reyes A, Roca J et al. Ventilation-perfusion mismatching
ventilation was shown to be equally eective in those with in chronic obstructive pulmonary disease during ventilator weaning.
Am Rev Respir Dis 140: 124650, 1989.
more severe as compared to mild acute respiratory acidosis,
16. Mountain RD, Sahn SA. Clinical features and outcome in patients
both in regards to normalization of pH and PaCO2, as well with acute asthma presenting with hypercapnia. Am Rev Respir Dis
as duration of use and hospital length of stay [88]. In two 138: 53539, 1988.
of these studies, noninvasive positive pressure ventilation 17. Jubran A, Tobin MJ. Pathophysiologic basis of acute respiratory distress
caused a decrease in mortality [8991]. in patients who fail a trial of weaning from mechanical ventilation. Am
J Respir Crit Care Med 155: 90615, 1997.
18. Marini JJ. Pressure-controlled and inverse ratio ventilation. In: Tobin
MJ (ed.), Principles and Practice of Mechanical Ventilation, pp. 25172.
CONCLUSION New York: McGraw-Hill, 2006.
19. Marini JJ, Capps JS, Culver BH. The inspiratory work of breathing
during assisted mechanical ventilation. Chest 87: 61218, 1985.
Patients with obstructive lung disease commonly develop 20. Ward ME, Corbeil C, Gibbons W et al. Optimization of respiratory
acute respiratory failure, requiring the institution of mechani- muscle relaxation during mechanical ventilation Anesthesiology 69: 2935,
cal ventilation. Abnormalities in pulmonary gas exchange, 1988.
lung mechanics, and respiratory muscle function contribute 21. Sassoon CSH. Intermittent mandatory ventilation. In: Tobin MJ (ed.),
Principles and Practice of Mechanical Ventilation, pp. 20120. New York:
to the development of respiratory failure. Adapting ventilator
McGraw-Hill, 2006.
strategies that account for these abnormalities, such as con- 22. Marini JJ, Smith TC, Lamb VJ. External work output and force gen-
trolled hypoventilation in acute asthma or the application of eration during synchronized intermittent mechanical ventilation.
external PEEP in patients with COPD, may decrease mor- Am Rev Respir Dis 138: 116979, 1988.
bidity and possibly improve patient outcome. 23. Imsand C, Feihl F, Perret C et al. Regulation of inspiratory neuromus-
cular output during synchronized intermittent mechanical ventilation.
Anesthesiology 80: 1322, 1994.
References 24. Leung P, Jubran A, Tobin MJ. Comparison of assisted ventilator modes
on triggering, patient eort and dyspnea. Am J Respir Crit Care Med
1. Sene MG, Wagner DP, Wagner RP et al. Hospital and 1-year survival 155: 194048, 1997.
of patients admitted to intensive care units with acute exacerbations of 25. Brochard L, Lellouche F. Pressure support ventilation. In: Tobin MJ
chronic obstructive pulmonary disease. JAMA 274: 185257, 1995. (ed.), Principles and Practice of Mechanical Ventilation, pp. 22150.
2. Connors AF, Dawson NV, Thomas C et al. Outcomes following acute New York: McGraw-Hill, 2006.
exacerbation of severe chronic obstructive lung disease. Am J Respir Crit 26. Jubran A, Van de Graa WB, Tobin MJ. Variability of patient
Care Med 154: 95967, 1996. ventilator interaction with pressure support ventilation in patients with
3. Derenne JP, Fleury B, Pariente R. Acute respiratory failure of chronic chronic obstructive pulmonary disease. Am J Respir Crit Care Med 152:
obstructive pulmonary disease. Am Rev Respir Dis 138: 100633, 1988. 12936, 1995.
4. Williams TJ, Tuxen DV, Scheinkestel CD et al. Risk factors for mor- 27. Appendini L, Patessio A, Zanaboni S et al. Physiologic eects of
bidity in mechanically ventilated patients with acute severe asthma. Am positive end-expiratory pressure and mask pressure support during
Rev Respir Dis 146: 60715, 1992. exacerbations of chronic obstructive pulmonary disease. Am J Respir
5. Pacht ER, Lingo S, St John RC. Clinical features, management, and Crit Care Med 149: 100978, 1994.
outcome of patients with severe asthma admitted to the intensive care 28. Jubran A, Tobin MJ. Reliability of pulse oximetry in titrating sup-
unit. J Asthma 32: 37377, 1995. plemental oxygen therapy in ventilator-dependent patients. Chest 90:
6. Afessa B, Morales IJ, Scanlon PD et al. Prognostic factors, clinical 142025, 1990.
course, and hospital outcome of patients with chronic obstructive pul- 29. Holets S, Hubmayr RD. Setting the ventilator. In: Tobin MJ (ed.),
monary disease admitted to an intensive care unit for acute respiratory Principles and Practice of Mechanical Ventilation, pp. 16381. New York:
failure. Crit Care Med 30: 161015, 2002. McGraw-Hill, 2006.
7. Breen D, Churches T, Hawker F et al. Acute respiratory failure second- 30. Aslanian P, El Atrous S, Isabey D et al. Eects of ow triggering on
ary to chronic obstructive pulmonary disease treated in the intensive breathing eort during partial ventilatory support. Am J Respir Crit
care unit: Along term follow up study. Thorax 57: 2933, 2002. Care Med 157: 13543, 1998.
8. Esteban A, Anzueto A, Frutos F et al. Mechanical ventilation interna- 31. Dreyfuss D, Ricard JD, Saumon G. Ventilator-induced injury. In:
tional study group. JAMA 287: 34555, 2002. Tobin MJ (ed.), Principles and Practice of Mechanical Ventilation, pp.
9. Pendergraft TB, Standord RH, Beasley R et al. Rates and characteristics 90330. New York: McGraw-Hill, 1994, 1994.
of intensive care unit admissions and intubations among asthma-relate 32. Laey JG, Kavanagh BP. Permissive hypercapnia. In: Tobin MJ (ed.),
hospitalizations. Ann Allergy Asthma Immunol 93: 2935, 2004. Principles and Practice of Mechanical Ventilation, pp. 37392. New York:
10. Zimmerman JL, Dellinger RP, Shah AN et al. Endotracheal intubation and McGraw-Hill, 2006.
mechanical ventilation in severe asthma. Crit Care Med 21: 172730, 1993. 33. Darioli R, Perret C. Mechanical controlled hypoventilation in status
11. Luksza AR, Smith P, Coakley J et al. Acute severe asthma treated by asthmaticus. Am Rev Respir Dis 129: 38587, 1984.
mechanical ventilation: 10 years experience from a district general 34. Tuxen DV, Williams TJ, Scheinkestel CD et al. Use of a measurement of
hospital. Thorax 41: 45963, 1986. pulmonary hyperination to control the level of mechanical ventilation

710
Ventilator Support 57
in patients with acute severe asthma. Am Rev Respir Dis 146: 113642, 57. Jaber S, Fodil R, Carlucci A et al. Noninvasive ventilation with helium
1992. oxygen in acute exacerbation of chronic obstructive pulmonary disease.
35. Connors AF Jr., McCa ree DR, Gray BA. Eect of inspiratory ow Am J Respir Crit Care Med 161: 1191200, 2000.
rate on gas exchange during mechanical ventilation. Am Rev Respir Dis 58. Diehl JL, Mercat A, Guerot E et al. Helium/oxygen mixture reduces
124: 53743, 1981. the work of breathing at the end of the weaning process in patient with
36. Puddy A, Younes M. Eect on inspiratory ow rate on respiratory out- severe chronic obstructive pulmonary disease. Crit Care Med 31: 1415
put in normal subjects. Am Rev Respir Dis 146: 78789, 1992. 20, 2003.
37. Corne S, Gillespie D, Roberts D et al. Eect of inspiratory ow rate on 59. Jolliet P, Tassaux D, Roeseler J et al. Heliumoxygen versus air-oxygen
respiratory rate in intubated patients. Am J Respir Crit Care Med 156: noninvasive pressure support in decompensated chronic obstructive dis-
3048, 1997. ease: A prospective, multicenter study. Crit Care Med 31: 87884, 2003.
38. Laghi F, Karamchandani K, Tobin M. Inuence of ventilator settings 60. Gattinoni L, Gianni T, Pesenti A et al. Eect of prone positioning on
in determining respiratory frequency during mechanical ventilation. the survival of patients with acute respiratory failure. N Engl J Med
Am J Respir Crit Care Med 160: 176670, 1999. 345: 56873, 2001.
39. Laghi F, Segal J, Choe WK et al. Eect of imposed ination time on 61. Reisgnier J, Lejeune O, Renard B et al. Short-term eects of prone
respiratory frequency and hyperination in patients with chronic position in chronic obstructive pulmonary disease patients with severe
obstructive pulmonary disease. Am J Respir Crit Care Med 163: 1365 acute hypoxemic and hypercapnic respiratory failure. Intens Care Med
70, 2001. 31: 112831, 2005.
40. Tobin MJ, Jubran A, Laghi F. Patientventilator interaction. Am J 62. Mentzelopoulos SD, Zakynthinos SG, Roussos C et al. Prone posi-
Respir Crit Care Med 163: 105963, 2001. tion improves lung mechanical behavior and enhances gas exchange
41. Rossi A, Gottfried SB, Zocchi L et al. Measurement of static compli- eciency in mechanically ventilated chronic obstructive pulmonary
ance of the total respiratory system in patients with acute respiratory disease patients. Anesth Analg 96: 175667, 2003.
failure: The eect of intrinsic positive end-expiratory pressure. Am Rev 63. Fagon JY, Chastre J, Hance A et al. Nosocomial pneumonia in ven-
Respir Dis 131: 67277, 1985. tilated patients: A cohort study evaluating attributable mortality and
42. Smith TC, Marini JJ. Impact of PEEP on lung mechanics and work hospital stay. Am J Med 94: 28188, 1993.
of breathing in severe airow obstruction. J Appl Physiol 65: 148899, 64. Soler N, Torres A, Ewig S et al. Bronchial microbial patterns in severe
1988. exacerbations of chronic obstructive pulmonary disease (COPD)
43. Schnader J. Estimation of auto-PEEP (letter). Chest 99: 520, 1991. requiring mechanical ventilation. Am J Respir Crit Care Med 157:
44. Petrof BJ, Legare M, Goldberg P et al. Continuous positive airway 1498505, 1998.
pressure reduces work of breathing and dyspnea during weaning from 65. Chastre J, Viau F, Brun P et al. Prospective evaluation of the protected
mechanical ventilation in severe chronic obstructive pulmonary disease. specimen brush for the diagnosis of pulmonary infections in ventilated
Am Rev Respir Dis 141: 28189, 1990. patients. Am Rev Respir Dis 130: 92429, 1984.
45. Georgopoulos D, Giannouli E, Patakas D. Eects of extrinsic posi- 66. Fagon JY, Chastre J, Wol M et al. Invasive and noninvasive strategies
tive end-expiratory pressure on mechanically ventilated patients with for management of suspected ventilator-associated pneumonia. Ann
chronic obstructive pulmonary disease and dynamic hyperination. Intern Med 132: 62130, 2000.
Intens Care Med 19: 197203, 1993. 67. Chastre J, Fagon JY. Ventilator-associated pneumonia. Am J Respir Crit
46. Ranieri VM, Guiliani R, Cinnella G. Physiologic eects of positive Care Med 165: 867903, 2002.
end-expiratory pressure in patients with chronic obstructive pulmonary 68. Pepe PE, Marini JJ. Occult positive end-expiratory pressure in mechan-
disease during acute ventilatory failure and controlled mechanical ven- ically ventilated patients with airow obstruction. Am Rev Respir Dis
tilation. Am Rev Respir Dis 147: 513, 1993. 126: 16670, 1982.
47. Rossi A, Santos C, Roca J et al. Eects of PEEP on V/Q mismatching 69. Esteban A, Frutos F, Tobin MJ et al. A comparison of four methods
in ventilated patients with chronic airow obstruction. Am J Respir Crit of weaning patients from mechanical ventilation. Spanish Lung failure
Care Med 149: 107784, 1994. Collaborative Group. N Engl J Med 332: 34550, 1995.
48. Tuxen DV. Detrimental eects of positive end-expiratory pressure dur- 70. Esteban A, Alia I, Tobin MJ. Eect of spontaneous breathing trial
ing controlled mechanical ventilation of patients with severe airow duration on outcome of attempts to discontinue mechanical ventila-
obstruction. Am Rev Respir Dis 140: 59, 1989. tion. Am J Respir Crit Care Med 159: 51218, 1999.
49. Dhand R, Tobin MJ. Bronchodilator delivery with metered-dose inhal- 71. Jubran A, Tobin MJ. Passive mechanics of lung and chest wall in
ers in mechanically ventilated patients. Eur Respir J 9: 58595, 1996. patients who failed or succeeded in trials of weaning. Am J Respir Crit
50. Dhand R, Duarte AG, Jubran A et al. Dose response to bronchodilator Care Med 155: 91621, 1997.
delivered by metered-dose inhaler in ventilator-supported patients. Am 72. Laghi F, Cattapan SE, Jubran A et al. Is weaning failure caused by low-
J Respir Crit Care Med 154: 38893, 1996. frequency fatigue of the diaphragm? Am J Respir Crit Care Med 167:
51. Wheeler AP. Sedation, analgesia, and paralysis in the intensive care 12027, 2003.
unit. Chest 104: 56677, 1993. 73. Laghi F, Tobin MJ. Disorders of the respiratory muscles. Am J Respir
52. Manthous CA, Chatila W. Prolonged weakness after the withdrawal of Crit Care Med 168: 1048, 2003.
atracurium. Am J Respir Crit Care Med 150: 144143, 1994. 74. Tobin MJ, Perez W, Guenther SM et al. The pattern of breathing
53. Leatherman JW, Fluegel WL, David WS et al. Muscle weakness in during successful and unsuccessful trials of weaning from mechanical
mechanically ventilated patients with severe asthma. Am J Respir Crit ventilation. Am Rev Respir Dis 134: 111118, 1986.
Care Med 153: 168690, 1996. 75. Jubran A, Mathru M, Dries D et al. Continuous recordings of mixed
54. Strange C, Vaughan L, Franklin C et al. Comparison of train-of-four venous oxygen saturation during weaning from mechanical ventilation and
and best clinical assessment during continuous paralysis. Am J Respir the ramications thereof. Am J Respir Crit Care Med 158: 176369, 1998.
Crit Care Med 156: 155661, 1997. 76. Stroetz RW, Hubmayr R. Tidal volume maintenance during weaning
55. Lee DL, Lee H, Chang HW et al. Heliox improves hemodynamics in with pressure support. Am J Respir Crit Care Med 152: 103440, 1995.
mechanically ventilated patients with chronic obstructive pulmonary dis- 77. Tobin MJ, Jubran A. Weaning from mechanical ventilation. In: Tobin
ease with systolic pressure variations. Crit Care Med 33: 96873, 2005. MJ (ed.), Principles and Practice of Mechanical Ventilation, 2nd edn,
56. Jolliet P, Tassaux D, Thouret JM et al. Benecial eects of helium:oxy- 1185220. New York: McGraw-Hill Inc., 2006.
gen versus air:oxygen noninvasive pressure support in patients with 78. Yang KL, Tobin MJ. A prospective study of indexes predicting the out-
decompensated chronic obstructive pulmonary disease. Crit Care Med come of trials of weaning from mechanical ventilation. N Engl J Med
27: 242229, 1999. 324: 144550, 1991.

711
Asthma and COPD: Basic Mechanisms and Clinical Management

79. Strauss C, Louis B, Isabey D et al. Contribution of the endotracheal 86. Marelich GP, Murin S, BAttistella F et al. Protocol wean-
tube and the upper airway to breathing workload. Am J Respir Crit Care ing of mechanical ventilation in medical and surgical patients by
Med 157: 2330, 1998. respiratory care practitioners and nurses: Eect on weaning time and
80. Brochard L, Rauss A, Benito S et al. Comparison of three methods incidence of ventilator-associated pneumonia. Chest 118: 45967,
of gradual withdrawal from ventilatory support during weaning from 2000.
mechanical ventilation. Am J Respir Crit Care Med 150: 896903, 1994. 87. Tobin MJ, Laghi F. Extubation. In: Tobin MJ (ed.), Principles and
81. Ely EW, Baker AM, Dunagan DP et al. Eect on the duration of Practice of Mechanical Ventilation, 2nd edn, 122138. New York:
mechanical ventilation of identifying patients capable of breathing McGraw-Hill, 2006.
spontaneously. N Engl J Med 335: 186469, 1996. 88. Crummy F, Buchan C, Miller B et al. The use of noninvasive mechani-
82. Namen AM, Ely EW, Tatter SB et al. Predictors of successful extubation cal ventilation in COPD and severe hypercapnic acidosis. Respir Med
in neurosurgical patients. Am J Respir Crit Care Med 163: 65864, 2001. 101: 5361, 2007.
83. Randolph AG, Wypij D, Venkataraman ST et al. Eect of mechani- 89. Bott J, Carroll MP, Conway JH et al. Randomized controlled trial of
cal ventilator weaning protocols on respiratory outcomes in infants and nasal ventilation in acute ventilatory failure due to chronic obstructive
children: A randomized controlled trial. JAMA 288: 256168, 2002. airways disease. Lancet 341: 155557, 1993.
84. Krishnan JA, Moore D, Robeson C et al. A prospective, controlled trial 90. Kramer N, Meyer TJ, Meharg J et al. Randomized, prospective trial of
of a protocol-based strategy to discontinue mechanical ventilation. Am noninvasive positive pressure ventilation in acute respiratory failure. Am
J Respir Crit Care Med 169: 67378, 2004. J Respir Crit Care Med 151: 1799806, 1995.
85. Kollef MH, Shapiro SD, Silver P et al. A randomized, controlled trial 91. Brochard L, Mancebo J, Wysocki M et al. Noninvasive ventilation for
of protocol-directed versus physician-directed weaning from mechani- acute exacerbations of chronic obstructive pulmonary disease. N Engl J
cal ventilation. Crit Care Med 25: 56774, 1997. Med 333: 81722, 1995.

712
Pulmonary Rehabilitation
58 CHAPTER

INTRODUCTION consequences associated with COPD, such as Thierry Troosters1,2,


skeletal muscle weakness, osteoporosis, reduced Wim Janssens1 and
insulin sensitivity, cardiovascular morbidity, and
Pulmonary rehabilitation is nowadays a rec- mood disturbance. Further catalyzed by smoking Marc Decramer1,2
ognized evidence-based therapy which can be and perhaps systemic inammation, oxidative
1
applied to patients with lung diseases who are stress, and exacerbations, these systemic conse- Respiratory Rehabilitation and
symptomatic and have reduced activities of daily quences may develop along with the progressing Respiratory Division, University
living, despite optimal medical therapy [1, 2]. airow obstruction. In fact, particularly patients Hospital, Leuven, Belgium
Unlike most drugs, pulmonary rehabilitation does with comorbidity or systemic consequences were 2
Faculty of Kinesiology and
not target the lungs directly, but aims at reversing characterized by an inactive lifestyle even in Rehabilitation Sciences, Department
or stabilizing the extra-pulmonary eects of lung milder stages of COPD [4]. Figure 58.1 shows of Rehabilitation Sciences, Katholieke
diseases [1]. that the vast majority of patients referred to our
Universiteit Leuven, Leuven, Belgium
Lung disease, especially chronic obstruc- rehabilitation program had developed skeletal
tive pulmonary disease (COPD), is a poorly muscle weakness to some degree, independently
diagnosed, but a very prevalent problem in our of the lung function impairment. Undoubtedly,
society. More than half the patients with mild to skeletal muscle dysfunction is an important
moderate COPD remain undiagnosed until late extra-pulmonary consequence. In COPD, skele-
in the disease [3]. Physical inactivity is a natural tal muscle weakness has been shown to be related
defense strategy to avoid the distressing symp- to impaired survival [5, 6]. In addition, skeletal
tom of exercise induced dyspnea. Unfortunately, muscle weakness is an important driver of utili-
inactivity inevitably leads to many of the systemic zation of health-care recourses [7]. In the context

FIG. 58.1 Relation between lung function


impairment (FEV1, expressed as a percentage
of the predicted normal value) and Quadriceps
force (QF, expressed as a percentage of
the predicted value) in 380 consecutive
patients with COPD (age 65  8 years, FEV1
42  16), referred to the authors outpatient
rehabilitation program. Pearsons correlation
coefficient was 0.094, p-value 0.07.

713
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 58.1 An overview of the effects of pulmonary rehabilitation as reported in evidence-based practice guidelines [11], a state of the art review [12] and
a Cochrane meta-analysis [13].

Effect Evidence grade [11] Size of effect

Improvement of dyspnea 1A CRDQ-Dys: 1.06 (0.85 to 1.26) points [13]

Improvement of HRQoL 1A SGRQ total: 6.11 (8.98 to 3.24)% [13]

Reduction in hospital days and utilization of 2B


health care recourses

Survival None provided

Psychosocial benefits (self-efficacy with exercise, 2B


cognitive function, anxiety, depression)

Exercise tolerance

Peak work rate 8W (313W) [13]  18 (IQR 1324) % baseline [12]

6MWD 48 min (3265 min) [13]  34 min if 28 s, 50 min if 28 s [12]

Whole body endurance  87% of baseline [12]

Evidence grading 1A: Strong recommendation; 2B: Weak recommendation; CRDQ-Dys: Dyspnea subscale of the chronic respiratory disease questionnaire; SGRQ: saint Georges
Respiratory Questionnaire; 6MWD: six minute walking distance.

of pulmonary rehabilitation, it is also important to recognize Improved physiological and emotional function
the non-physiological extra-pulmonary consequences of Increased health-related quality of life.
chronic lung disease. Depression, for example, is very preva-
lent in stable patients [8], and is signicantly related to the The health-care providers perspective:
adverse outcome of COPD, particularly after acute exacerba-
tions [9]. In the recent Global Initiative for Obstructive Lung Optimal self-management by the patient
Disease (GOLD), the systemic consequences of COPD were
specically noted as an important marker of morbidity, com- Optimal utilization of available recourses.
plicating the management of obstructive lung disease [10].
As mentioned above, pulmonary rehabilitation targets The societal perspective:
the systemic consequences of lung diseases. It is now gener-
ally recognized as an evidence-based and eective therapy for Increased productivity and active participation of patients
patients with lung diseases, who are symptomatic and have in society
reduced participation the in activities of daily life. The evidence Reduction of cost associated with the management of the
on the eectiveness of pulmonary rehabilitation is impressive, disease
and has been reviewed in several recent documents. Table 58.1
summarizes the most important eects of rehabilitation and Enhanced coping of the family when a family member is
where applicable the strength of the evidence, as provided in suering from COPD.
the dierent evidence-based reviews. To be successful, pulmo-
nary rehabilitation is typically designed as a comprehensive It is important to realize that patients suering from other
intervention oered by a team of health-care providers over respiratory diseases, such as pulmonary sarcoidosis [15],
a substantial period of time. Typically, rehabilitation programs cystic brosis [16], or after lung transplantation [17], equally
are carried out for 6 weeks to 6 months, with longer programs present systemic consequences of their disease. Consequently,
yielding more substantial eects [14]. Patients participate in pulmonary rehabilitation programs should not be restricted
an individualized tailored program which takes into account to patients with COPD, but should be open to any patient
the complexity of the presenting patient. Exercise training is suering from extra-pulmonary consequences of lung dis-
the cornerstone of such a program. Several other interven- eases in general.
tions may complement the program to maximize its eective-
ness. Pulmonary rehabilitation may serve many goals, which
depend on the perspective one takes. Examples of the goals
from the dierent perspectives are as follows: THE INDICATION FOR REHABILITATION
Patients perspective:
AND SETTING
Reduced symptoms of breathlessness or fatigue According to the denition of the American Thoracic
Enhanced exercise tolerance and skeletal muscle strength Society and the European Respiratory Society, pulmonary

714
Pulmonary Rehabilitation 58
rehabilitation is aiming at reducing symptoms, optimizing In depressed patients (some 40% (95% CI: 3644%)
function, increasing participation, and reducing health-care of patients with COPD [24]), for example, psychologi-
cost through stabilizing or reversing the systemic conse- cal counseling may be of benet in order to help reducing
quences of the disease [1]. Hence, theoretically, patients symptoms of depression or anxiety. Admittedly, a large
with an indication for the need of rehabilitation are those trial to conrm the point is currently still missing [25, 26],
suering from extra-pulmonary consequences. This deni- but since depressive symptoms do signicantly impact on
tion is, however, not very practical as the systemic conse- health-related quality of life [27] and even impact readmis-
quences that can be targeted by pulmonary rehabilitation sion [9] in these patients, psychological counseling may well
are not dened. Clearly, not all systemic consequences of be worth the eort in patients who suer from symptoms
the disease are amendable to pulmonary rehabilitation. For of depression, anxiety, or poor coping. It should be stressed,
example, osteoporosis and anemia are the recognized sys- however, that exposure to exercise therapy may in itself
temic consequences of COPD [18], but it is unlikely that have an anti-depressant eect [28]. Nutritional interven-
pulmonary rehabilitation would have a signicant impact tions have been shown to be unsuccessful in poorly selected
on these outcomes. In addition, the spin-o of pulmonary COPD patients [29]. However, when nutritional interven-
rehabilitation may well be an enhanced self-management, tions are successful in improving body mass in cachectic
which leads to more ecient utilization of health care patients, they do spin-o in an important survival benet
recourses and cost savings [19], without altering the sys- [30]. Besides patients suering from pulmonary cachectia,
temic consequences of the disease. a less recognized role, but likely of equal importance, is the
A more practical approach to identifying candidates care for obese COPD patients. Clearly obesity is linked
for rehabilitation and guide the assessment of patients before to increased pulmonary ventilation to carry out activities
enrollment in rehabilitation could be to select patients who of daily living. Consequently, weight loss in obese patients
are optimally pharmacologically treated, but still present might yield important functional benets in activities car-
with one of the following: ried out against gravity (e.g. stair climbing, walking). The
authors believe that weight loss, particularly loss of fat
Disabling symptoms due to deconditioning mass, might be an important target in overweight COPD
patients referred to pulmonary rehabilitation programs.
Skeletal or respiratory muscle weakness Occupational therapists may be consulted within the context
Poor health-related quality of life of a rehabilitation program [31]. Occupational therapists
may advise patients on the mode and pace of carrying out
Repeated exacerbations or inecient use of available
activities of daily life. Oftentimes occupational therapists
recourses
might advise on the use of wheeled walking aids (rolla-
Depressed mood status tors). Although many patients show poor compliance with
Malnutrition (obesity of cachexia) the daily use of a rollator [32], those who use it may sub-
stantially and suddenly improve their exercise tolerance [33,
Poor coping with the symptoms of their disease. 34]. Other potentially cost-eective components of a reha-
bilitation program are interventions aiming at enhancing
This allows to set up active screening for rehabilitation pro- self-management. Generally, these interventions are super-
grams, which typically includes assessment of exercise tol- vised by specially trained advanced practice nurses, who work
erance, skeletal and respiratory muscle force, nutritional integrated in the rehabilitation team. Interventions aiming
status, symptoms, health-related quality of life, and capability at enhancing self-management have shown variable success
of self-management. [35]. Studies showing cost-eectiveness of these interven-
Depending on the complexity of the individual prob- tions focused at a subgroup of patients with at least one
lem, the program can be designed across a range of complex- hospital admission [19]. Hence it seems reasonable to direct
ities. Programs can be as simple as an intervention consisting eorts specically to this subgroup of patients.
of exercise training in the home setting of patients with Another point to consider when a patient is referred
uncomplicated COPD [20] or as complex as an in-patient to a pulmonary rehabilitation program is the setting of
program in a mechanically ventilated patient [21]. On the such a program. Programs have been successfully set up
basis of proper assessment the individual program, its setting in a primary care (home) setting [20, 36] as an outpatient
and components can be designed, taking into account the program in a rehabilitation center [37] or in the commu-
available recourses in a given region, current best practice, nity [38, 38], or as an in-patient program [39, 40]. Clearly
and evidence. Several guidelines may help those setting up each of these programs has advantages and disadvantages.
pulmonary rehabilitation facilities to structure their program One of the most important problems of home-based pro-
[1, 11, 22, 23]. grams is the limited sta and equipment, which makes it
Exercise training has now the status of the corner- dicult to deal with more severe patients [41]. The most
stone of each program, and will be dealt with below. Exercise important problem of outpatient programs is the trans-
training has a strong evidence base. A program of exercise portation to the center [42]. In-patient programs are costly
training of the muscles of ambulation is recommended and and should be restricted to those patients with very limited
is a mandatory component of any pulmonary rehabilitation mobility. Ideally, a reference rehabilitation center should
program [11]. For several other components of a rehabilita- have access to all modalities of rehabilitation. This can be
tion program, however, evidence is also building up, particu- done by establishing strong links between the dierent lines
larly for specic subgroups of patients. of health care.

715
Asthma and COPD: Basic Mechanisms and Clinical Management

Screening of patients for Indication and expected results


rehabilitation
To establish the indication for a rehabilitation program, a
From the above it is clear that proper design of a rehabilita- formal exercise test should be carried out. Exercise intol-
tion program depends on a thorough screening of patients. erance is indeed an important reason to refer a patient for
This screening should answer the following questions: rehabilitation. We found that patients with skeletal and res-
piratory muscle weakness were most responsive to exercise
training [43], justifying the assessment of peripheral and
1 Is there an indication for pulmonary rehabilitation in a
respiratory muscle strength. Rehabilitation programs are
given patient and what results can be expected?
oriented toward patients who are symptomatic and have
2 Which components should be included in the program reduced activities of daily life, encouraging to assess symp-
for a given patient and what will be the optimal setting toms and health-related quality of life, and physical activity.
of the program? With evolving technology it becomes possible to assess the
latter, rather than estimate physical activity levels with ques-
An example of the intake procedure in the authors tionnaires [44, 45]. In patients admitted to the hospital with
institute is given in Table 58.2. a severe exacerbation of COPD, pulmonary rehabilitation

TABLE 58.2 An example of the intake procedure for pulmonary rehabilitation at the authors center.

Test Reason or consequence

Lung function assessment Disease severity

Exercise test
Incremental cycle ergometry Exercise intolerance/factors limiting exercise
6MWD Exercise intolerance, treatment effect
Constant work rate test Treatment effect

Muscle function
Quadriceps force, biceps, triceps Muscle weakness? Need for resistance training
Respiratory muscle force Need for inspiratory muscle training

Symptoms, Function

MRC Severity of impairement in daily life


PFSDQ Symptoms in daily life (occupational therapists)

HRQoL

CRDQ Indication for rehabilitation, identification of problems in four domains

Activity monitoring
1 week of monitoring Actual physical activity level

Interview psychologist
HADS Psychological burden

Social anamnesis Social support

Nutritional anamnesis Calorie intake and nutritional  protein balance

Bioelectrical impedance Assessment of body composition

6MWD: six minute walking distance; MRC: medical council dyspnea scale; PFSDQ: Pulmonary functional status and dyspnea scale;
HRQoL: Health related quality of life with the Chronic Respiratory Disease Questionnaire (CRDQ); HADS: Hospital anxiety and depression scale.

716
Pulmonary Rehabilitation 58
should be part of the proper after care. In these patients Exercise training
rehabilitation does reduce readmission rate by 75% and
may even enhance survival [46]. In addition substantial and Exercise training is undoubtedly the cornerstone of a pul-
clinically relevant improvements in exercise tolerance and monary rehabilitation program. Programs aim at enhanc-
health-related quality of life were reported [47]. ing exercise tolerance and skeletal muscle force. Skeletal
muscle force can be enhanced by specic resistance training
(weightlifting) exercise [5456]. Improvement in exercise
Components of the rehabilitation tolerance is essentially obtained through two main path-
program ways. First, the oxidative capacity of the skeletal muscle is
enhanced [57], leading to reductions in lactate production at
To establish the components of the rehabilitation program identical work [58]. A second pathway involves an improve-
and its optimal setting, a multidisciplinary assessment is ment in movement eciency, yielding less oxygen consump-
needed. This consists of a nutritional assessment, including tion to cover identical work rate. Both pathways are not
analysis of the fat free mass and a nutritional interview to mutually exclusive, and both lead to reductions in pulmonary
estimate the caloric balance [48]. A thorough social interview ventilation, and dynamic hyperination during exercise. The
to gain insight in the social network and available social sup- reduction in pulmonary ventilation, the improved dynamic
port is helpful since patients lacking social support are at risk hyperination, and the desensitization (better tolerance to
to drop out from a program [49]. An intake by a psycholo- high ventilatory levels) are crucial to the improved exer-
gist may allow to identify the large proportion of patients tional dyspnea, which is seen after exercise training [13]. To
suering from signicant depressive symptoms or other psy- enhance the oxidative capacity of the skeletal muscle, exer-
chiatric morbidity [50]. An important and often forgotten cise training should be conducted at relatively high training
aspect may be the burden on the partner of the patient [51]. intensity [58] (see below). Improvement in movement e-
A rst screening for psychological morbidity can be done ciency is obtained by practicing specic exercises (e.g. walk-
using questionnaires such as the Beck depression inventory ing training will only improve walking eciency). For this
or the Hospital Anxiety and Depression Scale. Lastly physi- pathway, the modality of the exercise is more crucial than
cal activity levels of patients and symptoms during specic the intensity at which the exercise is carried out.
activities of daily living can be assessed using questionnaires In general, exercise training in patients with COPD
such as the Pulmonary Functional Status and Dyspnea follows the principles of exercise training in the healthy eld-
Questionnaire (PFSDQ) [52, 53]. erly [59]. Programs generally consist of a brief warm-up, a
Depending on the complexity of the individual case core program lasting at least 30 min of exercise and a cooling
one or more disciplines can be engaged in the rehabilita- down. An example of an exercise training program is given in
tion process of the individual patient. Ideally the patient is Table 58.3. Close supervision and proper monitoring of the
referred to a program which optimally meets his needs and patients ensure the safety during the program [12]. In fact,
takes into account the motivation and social circumstances. very few exercise-related events and, as far as the authors are
Needless to say that a program should also be moduled along aware, no fatal events have been reported after pulmonary
the possibilities of the health-care system in a given region. rehabilitation in the published literature.

TABLE 58.3 An overview of an outpatient rehabilitation session in the rehabilitation center of the authors.

Time Duration (min) Item Intensity

13:00 5 Brief contact with health care provider to check the health status. Resting saturation,
peak flow, and heart rate are recorded

13:05 10 Low intensity warm-up exercise on bicycle 2030 W

13:15 32 Cycling exercises e.g. 8 blocks of 2 min interval training with 2 min of rest between the 75% Wmax
intervals

13:47 15 Resistance training exercises for the upper and lower limb. Typical muscle groups are 3 series of 8 repetitions
quadriceps, triceps, biceps and pectoral muscle

14:02 6 2 blocks of 2 min stair climbing separated by 2 min of rest Dyspnea 46/10

14:08 6 2 blocks of 2 min of arm ergometry separated by 2 min of rest Dyspnea 46/10

14:14 20 2 blocks of 8 min of treadmill walking 80% 6 MWs

14:34 30 Individual contact with another health care worker (psychologist, social worker, dietician)

15:04 End of session

717
Asthma and COPD: Basic Mechanisms and Clinical Management

Whole body exercise ready for wide clinical use [78]. Similarly, during cycling less
Exercise training has been included in virtually all studies muscles are recruited compared to walking exercises; hence
investigating the benets of pulmonary rehabilitation. To suc- it is not surprising that for a given oxygen consumption,
cessfully increase skeletal muscle properties and render meas- cycling is more fatiguing for the quadriceps muscle [79]. It
urable physiological benets, it is important that patients do would be interesting to conduct a head-to-head comparison
exercise at relative high work loads. To do so, the exercise train- study on the physiological eects of cycling versus walking
ing intervention can be adapted to the individual exercise limi- exercises. On the basis of the larger potential to elicit mus-
tations of the patient. The conventionally used form to deliver cle fatigue, cycling would be a form of exercise training that
exercise training to COPD patients is endurance training. In may result in larger physiological eects, compared to walk-
COPD patients with primarily moderate disease, exercise ing. However, such a study is currently unavailable.
training conducted at approximately 75% of the peak work All the above techniques reduce the ventilatory bur-
rate (60% of the dierence between the lactate threshold and den. Another complementary option is to enhance the ven-
peak oxygen uptake) resulted in signicant physiological eects tilatory capacity. Obviously optimal bronchodilator therapy
[58]. A similar training strategy was shown to be eective in does also allow for larger pulmonary ventilation and less
patients with severe disease [60]. Others have conrmed that dynamic hyperination during exercise. In one study, a
high training intensity is required to elicit physiologic training potent long-term anti-cholinergic drug showed to enhance
eects [61, 62]. exercise training eects compared to the use of short acting
It is important to adjust and increase the training load bronchodilators only [80]. Breathing gases with light density
in every training session. Trained personnel should be available (e.g. helium and oxygen, HELIOX) also enhance the venti-
to ensure close supervision on the training intensity. Training latory capacity. One study suggests that maximal ventilation
intensity can be monitored using 10-point Borg symptom is enhanced by 16% when breathing 21% oxygen and 79%
scales. A score around 46 is generally advised as an appropri- helium [81]. Although this would technically allow training
ate training intensity, provided the patients are familiar with at higher intensity, it is doubtful whether this intervention
the scale. Interestingly, a given Borg symptom score is generally is currently ready for clinical routine. A last intervention
chosen by a patient at an identical relative work rate. Hence as which enhances peak ventilatory capacity is bracing the
patients improve during training, the same Borg rating will be arms. During walking, this can be applied using a wheeled
achieved at higher absolute work rates [63]. Since most patients walking aid (rollator). Our group showed that bracing the
are not limited by the cardiovascular system, using heart fre- arms on a rollator enhanced maximal voluntary ventilation
quency to guide exercise training is not to be advised. by 8 l/min, which partly explained the acute benecial eect
In patients who reach a ventilatory limitation during of a rollator [82]. During treadmill walking, bracing of the
exercise, it is dicult to obtain a high training intensity for arms can also easily be achieved.
sustained periods of time. Several strategies may help clini-
cians to ensure high intensity training in more severe COPD. Resistance training
Interval exercise training has been used and showed to result Another form of conventional training is resistance training.
in physiological benets, comparable to those of endurance This form of exercise, generally consisting of weight lifting,
training [6467]. The advantage of interval training is that can be used as the only form of training [55, 56, 83], or in
the ventilatory requirements remain relatively limited [68]. combination with whole body exercises [54, 55, 84]. In all
Interval training is achieved by breaking down the long exer- the latter studies, muscle strength was signicantly more
cise bouts into smaller bouts of exercise separated by periods increased when resistance training was added to the exercise
of active recovery (low work rate) or rest. regimen. Increased muscle strength is an important treat-
An intervention that minimizes the ventilatory burden ment objective in patients with COPD suering from mus-
during exercise training is the use of supplemental oxygen. cle weakness, as activities of daily life do require strength
Oxygen, dose dependently, reduces the ventilation for a given apart from muscle endurance. As mentioned above, muscle
exercise intensity [69, 70]. The application of oxygen supple- weakness is an important factor related to morbidity and
ments hence allows training at higher training intensity [71]. even mortality in COPD. It follows that patients suering
Non-invasive mechanical ventilation reduces the work of from muscle weakness may be particularly good candidates
breathing and has been used successfully in severe COPD as to a resistance training program.
an adjunct to exercise training [7274]. In less severe COPD Resistance training is easy to apply in clinical practice.
the impact of using non-invasive mechanical ventilation was Patients are instructed to lift weights (generally on a multi-
not signicant [75]. Lastly, the required ventilation can be gym device). The weight imposed and the number of rep-
reduced simply by reducing the amount of muscles set at etitions ensure overload of the skeletal muscle. In patients
work. If exercise is conned to one leg, ventilation is con- with COPD and several other chronic diseases, resistance
siderably reduced, allowing a signicant increase in training training is started at approximately 70% of the weight a
load [76]. A recent trial conrmed that in severe patients, patient can lift once (i.e. the one repetition maximum). The
single leg exercise training resulted in 20% more increase in eects of resistance training programs may be enhanced
peak oxygen consumption and peak work rate compared to in male hypogonadal patients by testosterone replacement
conventional cycling training (two legs) [77]. In the latter therapy. In one study, [85] weekly intramuscular injections
study, however, no patient centered outcomes were assessed. with testosterone, aiming at restoring testosterone levels to
Hence, although interesting from a physiological perspective, normal values in initially hypogonadal men, did enhance
it is too early to judge whether single leg exercise training is skeletal muscle force more than either of the interventions

718
Pulmonary Rehabilitation 58
alone. Further studies are required to investigate the long-
term safety of this intervention. However, since skeletal
muscle dysfunction is in itself a negative prognostic factor, a
short-term use of testosterone may be benecial to result in
a rapid restoration of this potentially harmful situation.

Neuromuscular electrical stimulation


Another intervention used to specically stimulate the
peripheral muscles is medium to high-frequency neu-
romuscular electrical stimulation (NMES). Four studies
have investigated successfully the eects of this intervention
[8689]. All studies in COPD used transcutaneous electri-
cal stimulation with stimulation frequencies of 35 [89] to
50 Hz [86, 87, 90]. In patients with congestive heart failure,
low-frequency (1015 Hz) transcutaneous electrical stimu- FIG. 58.2 The effect of different pulmonary rehabilitation on assessed
lation was successfully applied [91, 92]. Studies showed activities of daily living. Different activity monitors were used, hence data are
that there was more strength gain in the skeletal muscles expressed as a % of the baseline measure. Open symbols represent studies
treated with electrical stimulation, either as mono-therapy in which the effect was reported as non-significant, closed symbols are
or in combination with general exercise training [89]. This studies that reported a statistically significant effect.  Sewell, Chest 2006;
intervention may prove to be attractive in patients who  Pitta, Chest 2008 (after 3 months);  Pitta, Chest 2008 (after 6 months);

have diculties to take part in regular rehabilitation, such Corronado, J Cardiopulm Rehabil 2003;  Mercken, Am J Respir Crit Care
as patients admitted to hospital with acute exacerbations. Med 2005;  Steele, Arch Phys Med Rehabil 2008;  Steele, J Rehabil Res Dev
2003.

Enhancing physical activity


In the last few years, more attention has been given to the rehabilitation may very well be the failure to successfully
eect of pulmonary rehabilitation on physical activity lev- enhance daily physical activity levels of patients toward
els. A minimum of 30 min of physical activity of moderate levels of moderate intense physical activity, sucient to
intensity is critical to maintain health [93]. A recent study maintain health, and perhaps, the achieved benets of the
also showed the importance of maintaining a healthy and previous rehabilitation program.
physically active lifestyle, which has been shown to be cru-
cial for survival in COPD [94]. Only a handful of studies
have investigated the eect of pulmonary rehabilitation on References
physical activity [95100]. The results of these studies are
summarized in Fig. 58.2. None of these studies were rand- 1. Nici L, Donner C, Wouters E, ZuWallack R, Ambrosino N,
omized controlled trials, so the dierences presented repre- Bourbeau J, Carone M, Celli B, Engelen M, Fahy B, Garvey C,
sent pre- to post-program dierences. Interestingly not all Goldstein R, Gosselink R, Lareau S, Macintyre N, Maltais F,
of them showed signicant improvements of the amount Morgan M, ODonnell D, Prefault C, Reardon J, Rochester C, Schols A,
and intensity of physical activities in daily life after pulmo- Singh S, Troosters T. American Thoracic Society/European Respiratory
nary rehabilitation. One trial [95] suggests that the eect Society statement on pulmonary rehabilitation. Am J Respir Crit Care
of rehabilitation on physical activity is larger after longer Med 173: 1390413, 2006.
programs. However, the relation between the length of 2. Buist AS, McBurnie MA, Vollmer WM, Gillespie S, Burney P,
the programs and their eectiveness on physical activity Mannino DM, Menezes AM, Sullivan SD, Lee TA, Weiss KB,
Jensen RL, Marks GB, Gulsvik A, Nizankowska-Mogilnicka E.
can surely not be seen across the dierent published tri-
International variation in the prevalence of COPD (the BOLD Study):
als. From the available evidence, it can be appreciated that A population-based prevalence study. Lancet 370: 74150, 2007.
the transfer of improvements in exercise tolerance toward 3. Schirnhofer L, Lamprecht B, Vollmer WM, Allison MJ, Studnicka M,
enhanced activities of daily living should not be taken for Jensen RL, Buist AS. COPD prevalence in Salzburg, Austria: Results
granted. Indeed, physical activity is a behavior. The amount from the Burden of Obstructive Lung Disease (BOLD) Study. Chest
and intensity of activities in which patients engage depends, 131: 2936, 2007.
besides their exercise capacity, on intrinsic factors such as 4. Watz H, Waschki B, Boehme C, Claussen M, Meyer T, Magnussen H.
motivation, perceived self-ecacy, mood status, and health Extrapulmonary eects of chronic obstructive pulmonary disease on
beliefs. Other factors related to physical activity are extrin- physical activity. Am J Respir Crit Care Med, 2007.
sic factors such as social and cultural role, external barriers 5. Swallow EB, Reyes D, Hopkinson NS, Man WD, Porcher R, Cetti EJ,
Moore AJ, Moxham J, Polkey MI. Quadriceps strength predicts mortal-
(e.g. environmental), and climate. Clearly more research is
ity in patients with moderate to severe chronic obstructive pulmonary
needed to enhance the impact of pulmonary rehabilitation disease. Thorax 62: 11520, 2007.
beyond the immediate eects on health-related quality of 6. Marquis K, Debigare R, Lacasse Y, LeBlanc P, Jobin J, Carrier G,
life, symptoms, exercise tolerance and utilization of health Maltais F. Midthigh muscle cross-sectional area is a better predictor of
care recourses and also enhance activities of daily living. The mortality than body mass index in patients with chronic obstructive pul-
missing link toward more endurable eects of pulmonary monary disease. Am J Respir Crit Care Med 166: 80913, 2002.

719
Asthma and COPD: Basic Mechanisms and Clinical Management

7. Decramer M, Gosselink R, Troosters T, Verschueren M, Evers G. 26. Coventry PA, Gellatly JL. Improving outcomes for COPD patients
Muscle weakness is related to utilization of health care resources in with mild-to-moderate anxiety and depression: A systematic review of
COPD patients. Eur Respir J 10: 41723, 1997. cognitive behavioural therapy. Br J Health Psychol, 2007.
8. van Manen JG, Bindels PJ, Dekker FW, IJzermans CJ, van der Zee JS, 27. Oga T, Nishimura K, Tsukino M, Sato S, Hajiro T, Mishima M.
Schade E. Risk of depression in patients with chronic obstructive pul- Longitudinal deteriorations in patient reported outcomes in patients
monary disease and its determinants. Thorax 57: 41216, 2002. with COPD. Respir Med, 2006.
9. Ng TP, Niti M, Tan WC, Cao Z, Ong KC, Eng P. Depressive symp- 28. Craft LL, Perna FM. The benets of exercise for the clinically
toms and chronic obstructive pulmonary disease: Eect on mortality, depressed. Prim Care Companion J Clin Psychiatry 6: 10411, 2004.
hospital readmission, symptom burden, functional status, and quality of 29. Ferreira IM, Brooks D, Lacasse Y, Goldstein RS. Nutritional support
life. Arch Intern Med 167: 6067, 2007. for individuals with COPD: A meta-analysis. Chest 117: 67278, 2000.
10. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, 30. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss is a revers-
Fukuchi Y, Jenkins C, Rodriguez-Roisin R, van WC, Zielinski J. ible factor in the prognosis of chronic obstructive pulmonary disease.
Global strategy for the diagnosis, management, and prevention of Am J Respir Crit Care Med 157: 179197, 1998.
chronic obstructive pulmonary disease: GOLD executive summary. Am 31. Lorenzi CM, Cilione C, Rizzardi R, Furino V, Bellantone T, Lugli D,
J Respir Crit Care Med 176: 53255, 2007. Clini E. Occupational therapy and pulmonary rehabilitation of disa-
11. Ries AL, Bauldo GS, Carlin BW, Casaburi R, Emery CF, bled COPD patients. Respiration 71: 24651, 2004.
Mahler DA, Make B, Rochester CL, ZuWallack R, Herrerias C. 32. Gupta RB, Brooks D, Lacasse Y, Goldstein RS. Eect of rollator use
Pulmonary rehabilitation: Joint ACCP/AACVPR evidence-based on health-related quality of life in individuals with COPD. Chest 130:
clinical practice guidelines. Chest 131: 4S42S, 2007. 108995, 2006.
12. Troosters T, Casaburi R, Gosselink R, Decramer M. Pulmonary reha- 33. Gupta R, Goldstein R, Brooks D. The acute eects of a rollator in indi-
bilitation in chronic obstructive pulmonary disease. Am J Respir Crit viduals with COPD. J Cardiopulm Rehabil 26: 10711, 2006.
Care Med 172: 1938, 2005. 34. Probst V, Heyvaert H, Coosemans I, Pitta F, Spruit M, Troosters T,
13. Lacasse Y, Goldstein R, Lasserson TJ, Martin S. Pulmonary rehabilita- Gosselink R, Decramer M. Eects of a rollator on exercise capacity,
tion for chronic obstructive pulmonary disease. Cochrane Database Syst gas exchange and ventilation in COPD patients. Am J Respir Crit Care
Rev, 2006, CD003793. Med 167: A669, 2003, Abstract.
14. Salman GF, Mosier MC, Beasley BW, Calkins DR. Rehabilitation for 35. Eng T, Monninkhof EM, van der Valk PD, van der Palen J,
patients with chronic obstructive pulmonary disease. J Gen Intern Med van Herwaarden CL, Partidge MR, Walters EH, Zielhuis GA. Self-
18: 21321, 2003. management education for patients with chronic obstructive pulmo-
15. Spruit M, Thomeer M, Gosselink R, Troosters T, Kasran A, Debrock A, nary disease. Cochrane Database Syst Rev, 2007, CD002990.
Demedts M, Decramer M. Skeletal muscle weakness in patients with 36. Wijkstra PJ, Ten Vergert EM, van Altena R, Otten V, Kraan J,
sarcoidosis and its relationship with exercise intolerance and reduced Postma DS, Koeter GH. Long term benets of rehabilitation at home
health status. Thorax 60: 3238, 2005. on quality of life and exercise tolerance in patients with chronic obstruc-
16. Troosters T, Langer D, Vrijsen B, Segers J, Wouters K, Janssens W, tive pulmonary disease. Thorax 50: 82428, 1995.
Gosselink R, Decramer M, Dupont LJ. Skeletal muscle weakness, exer- 37. Troosters T, Gosselink R, Decramer M. Short- and long-term eects
cise tolerance and physical activity in adults with cystic brosis. Eur of outpatient rehabilitation in patients with chronic obstructive pulmo-
Respir J, 2007, Revised. nary disease: A randomized trial. Am J Med 109: 20712, 2000.
17. Maury G, Langer D, Verleden G, Dupont L, Gosselink R, 38. Griths TL, Burr ML, Campbell IA, Lewis-Jenkins V, Mullins J,
Decramer M, Troosters T. Skeletal muscle force and functional exercise Shiels K, Turner-Lawlor PJ, Payne N, Newcombe RG, Ionescu AA,
tolerance before and after lung transplantation, a cohort study. Am J Thomas J, Tunbridge J, Lonescu AA. Results at 1 year of outpatient
Transplant, 2008, in press. multidisciplinary pulmonary rehabilitation: A randomised controlled
18. Fabbri LM, Rabe KF. From COPD to chronic systemic inammatory trial. Lancet 355: 36268, 2000.
syndrome? Lancet 370: 79799, 2007. 39. Goldstein RS, Gort EH, Stubbing D, Avendano MA, Guyatt GH.
19. Bourbeau J, Julien M, Maltais F, Rouleau M, Beaupre A, Begin R, Randomised controlled trial of respiratory rehabilitation. Lancet 344:
Renzi P, Nault D, Borycki E, Schwartzman K, Singh R, Collet JP. 139497, 1994.
Reduction of hospital utilization in patients with chronic obstructive 40. Franssen FM, Broekhuizen R, Janssen PP, Wouters EF, Schols AM.
pulmonary disease: A disease-specic self-management intervention. Eects of whole-body exercise training on body composition and
Arch Intern Med 163: 58591, 2003. functional capacity in normal-weight patients with COPD. Chest 125:
20. Strijbos JH, Postma DS, van Altena R, Gimeno F, Koeter GH. A com- 202128, 2004.
parison between an outpatient hospital-based pulmonary rehabilitation 41. Wedzicha JA, Bestall JC, Garrod R, Garnham R, Paul EA, Jones PW.
program and a home-care pulmonary rehabilitation program in patients Randomized controlled trial of pulmonary rehabilitation in severe
with COPD. A follow-up of 18 months. Chest 109: 36672, 1996. chronic obstructive pulmonary disease patients, stratied with the
21. Nava S. Rehabilitation of patients admitted to a respiratory intensive MRC dyspnoea scale. Eur Respir J 12: 36369, 1998.
care unit. Arch Phys Med Rehabil 79: 84954, 1998. 42. Taylor R, Dawson S, Roberts N, Sridhar M, Partridge MR. Why do
22. National Institute for Clinical Excellence (NICE). Chronic obstructive patients decline to take part in a research project involving pulmonary
pulmonary disease: National clinical guidline for the management of rehabilitation? Respir Med 101: 194246, 2007.
COPD in adults in primary and secondary care. Thorax 59(Suppl 1), 2004. 43. Troosters T, Gosselink R, Decramer M. Exercise training in COPD:
23. Socit de Pneumologie de Langue Franaise de Pneumologie. How to distinguish responders from nonresponders. J Cardiopulm
Recommandations de la Socit de Pneumologie de Langue Franaise Rehabil 21: 1017, 2001.
sur la rhabilitation du malade atteint de BPCO. Rev Mal Respir 22: 44. Pitta F, Troosters T, Spruit M, Decramer M, Gosselink R. Activity
7S87S14, 2005. monitoring for assessment of physical activities of daily life in patients
24. Yohannes AM, Baldwin RC, Connolly MJ. Depression and anxiety in with COPD. Arch Phys Med Rehabil 86: 197985, 2005.
elderly patients with chronic obstructive pulmonary disease. Age Ageing 45. Pitta F, Troosters T, Probst VS, Spruit MA, Decramer M, Gosselink R.
35: 45759, 2006. Quantifying physical activity in daily life with questionnaires and
25. Coventry PA, Hind D. Comprehensive pulmonary rehabilitation for motion sensors in COPD. Eur Respir J 27: 104055, 2006.
anxiety and depression in adults with chronic obstructive pulmonary 46. Puhan MA, Scharplatz M, Troosters T, Steurer J. Respiratory rehabili-
disease: Systematic review and meta-analysis. J Psychosom Res 63: 551 tation after acute exacerbation of COPD may reduce risk for readmis-
65, 2007. sion and mortality a systematic review. Respir Res 6: 54, 2005.

720
Pulmonary Rehabilitation 58
47. Man WD, Polkey MI, Donaldson N, Gray BJ, Moxham J. Community 67. Puhan MA, Busching G, Schunemann HJ, VanOort E, Zaugg C,
pulmonary rehabilitation after hospitalisation for acute exacerbations of Frey M. Interval versus continuous high-intensity exercise in chronic
chronic obstructive pulmonary disease: Randomised controlled study. obstructive pulmonary disease: A randomized trial. Ann Intern Med
BMJ 329: 1209, 2004. 145: 81625, 2006.
48. Brug J, Schols A, Mesters I. Dietary change, nutrition education and 68. Sabapathy S, Kingsley RA, Schneider DA, Adams L, Morris NR.
chronic obstructive pulmonary disease. Patient Educ Couns 52: 24957, Continuous and intermittent exercise responses in individuals with
2004. chronic obstructive pulmonary disease. Thorax 59: 102631, 2004.
49. Young P, Dewse M, Fergusson W, Kolbe J. Respiratory rehabilitation 69. Porszasz J, Emtner M, Goto S, Somfay A, Whipp BJ, Casaburi R.
in chronic obstructive pulmonary disease: Predictors of nonadherence. Exercise training decreases ventilatory requirements and exercise-induced
Eur Respir J 13: 85559, 1999. hyperination at submaximal intensities in patients with COPD. Chest
50. Wagena EJ, Arrindell WA, Wouters EF, van Schayck CP. Are patients 128: 202534, 2005.
with COPD psychologically distressed? Eur Respir J 26: 24248, 2005. 70. Somfay A, Porszasz J, Lee SM, Casaburi R. Doseresponse eect of
51. Unger DG, Jacobs SB. Couples and chronic obstructive airway diseases: oxygen on hyperination and exercise endurance in nonhypoxaemic
The role of gender in coping and depression. Womens Health 1: 23755, COPD patients. Eur RespirJ 18: 7784, 2001.
1995. 71. Emtner M, Porszasz J, Burns M, Somfay A, Casaburi R. Benets of
52. Lareau SC, Breslin EH, Meek PM. Functional status instruments: supplemental oxygen in exercise training in non-hypoxemic COPD
Outcome measure in the evaluation of patients with chronic obstructive patients. Am J Respir Crit Care Med 168: 103442, 2003.
pulmonary disease. Heart Lung 25: 21224, 1996. 72. Hawkins P, Johnson LC, Nikoletou D, Hamnegard CH, Sherwood R,
53. Lareau SC, Meek PM, Roos PJ. Development and testing of the modi- Polkey MI, Moxham J. Proportional assist ventilation as an aid to exer-
ed version of the pulmonary functional status and dyspnea question- cise training in severe chronic obstructive pulmonary disease. Thorax
naire (PFSDQ-M). Heart Lung 27: 15968, 1998. 57: 85359, 2002.
54. Bernard S, Whittom F, LeBlanc P, Jobin J, Belleau R, Berube C, 73. Costes F, Agresti A, Court-Fortune M, Roche F, Vergnon JM,
Carrier G, Maltais F. Aerobic and strength training in patients with Barthelemy JC. Noninvasive ventilation during exercise training
chronic obstructive pulmonary disease. Am J Respir Crit Care Med 159: improves exercise tolerance in patients with chronic obstructive pulmo-
896901, 1999. nary disease. J Cardiopulm Rehabil 23: 30713, 2003.
55. Ortega F, Toral J, Cejudo P, Villagomez R, Sanchez H, Castillo J, 74. vant HA, Gosselink R, Hollander P, Postmus P, Kwakkel G. Training
Montemayor T. Comparison of eects of strength and endurance train- with inspiratory pressure support in patients with severe COPD. Eur
ing in patients with chronic obstructive pulmonary disease. Am J Respir Respir J 27: 6572, 2006.
Crit Care Med 166: 66974, 2002. 75. Bianchi L, Foglio K, Porta R, Baiardi R, Vitacca M, Ambrosino N.
56. Spruit MA, Gosselink R, Troosters T, De Paepe C, Decramer M. Lack of additional eect of adjunct of assisted ventilation to pulmonary
Resistance versus endurance training in patients with COPD and skel- rehabilitation in mild COPD patients. Respir Med 96: 35967, 2002.
etal muscle weakness. Eur Respir J 19: 107278, 2002. 76. Dolmage TE, Goldstein RS. Response to one-legged cycling in
57. Maltais F, LeBlanc P, Simard C, Jobin J, Berube C, Bruneau J, Carrier patients with COPD. Chest 129: 32532, 2006.
L, Belleau R. Skeletal muscle adaptation to endurance training in 77. Dolmage TE, Goldstein RS. Eects of one-legged exercise training of
patients with chronic obstructive pulmonary disease. Am J Respir Crit patients with COPD. Chest 133: 37076, 2008.
Care Med 154: 44247, 1996. 78. Mador MJ. Exercise training in patients with COPD: One leg is better
58. Casaburi R, Patessio A, Ioli F, Zanaboni S, Donner CF, Wasserman K. than two? Chest 133: 33739, 2008.
Reductions in exercise lactic acidosis and ventilation as a result of exer- 79. Pepin V, Saey D, Whittom F, LeBlanc P, Maltais F. Walking versus
cise training in patients with obstructive lung disease. Am Rev Respir cycling: Sensitivity to bronchodilation in chronic obstructive pulmo-
Dis 143: 918, 1991. nary disease. Am J Respir Crit Care Med, 2005.
59. American College of Sports Medicine Position Stand. Exercise and phys- 80. Casaburi R, Kukafka D, Cooper CB, Witek TJ Jr., Kesten S.
ical activity for older adults. Med Sci Sports Exerc 30: 9921008, 1998. Improvement in exercise tolerance with the combination of tiotro-
60. Casaburi R, Porszasz J, Burns MR, Carithers ER, Chang RS, pium and pulmonary rehabilitation in patients with COPD. Chest 127:
Cooper CB. Physiologic benets of exercise training in rehabilitation 80917, 2005.
of patients with severe chronic obstructive pulmonary disease. Am J 81. Eves ND, Petersen SR, Haykowsky MJ, Wong EY, Jones RL. Helium-
Respir Crit Care Med 155: 154151, 1997. hyperoxia, exercise, and respiratory mechanics in chronic obstructive
61. Puente-Maestu L, Sanz ML, Sanz P, Ruiz de Ona JM, Rodriguez- pulmonary disease. Am J Respir Crit Care Med 174: 76371, 2006.
Hermosa JL, Whipp BJ. Eects of two types of training on pulmonary 82. Probst V, Troosters T, Coosemans I, Spruit M, Pitta F, Decramer M,
and cardiac responses to moderate exercise in patients with COPD. Gosselink R. Mechanisms of improvement in exercise capacity using a
Eur Respir J 15: 102632, 2000. rollator in COPD. Chest 126: 11027, 2004.
62. Gimenez M, Servera E, Vergara P, Bach JR, Polu JM. Endurance train- 83. Simpson K, Killian K, McCartney N, Stubbing DG, Jones NL.
ing in patients with chronic obstructive pulmonary disease: A compari- Randomised controlled trial of weightlifting exercise in patients with
son of high versus moderate intensity. Arch Phys Med Rehabil 81: 1029, chronic airow limitation. Thorax 47: 7075, 1992.
2000. 84. Mador MJ, Bozkanat E, Aggarwal A, Shaer M, Kufel TJ. Endurance
63. Mahler DA, Ward J, Mejia-Alfaro R. Stability of dyspnea ratings after and strength training in patients with COPD. Chest 125: 203645, 2004.
exercise training in patients with COPD. Med Sci Sports Exerc 35: 85. Casaburi R, Bhasin S, Cosentino L, Porszasz J, Somfay A, Lewis M,
108387, 2003. Fournier M, Storer T. Anabolic eects of testosterone replacement and
64. Vogiatzis I, Nanas S, Roussos C. Interval training as an alternative strength training in men with COPD. Am J Respir Crit Care Med 170:
modality to continuous exercise in patients with COPD. Eur Respir J 87078, 2004.
20: 1219, 2002. 86. Neder JA, Sword D, Ward SA, Mackay E, Cochrane LM, Clark CJ.
65. Vogiatzis I, Terzis G, Nanas S, Stratakos G, Simoes DC, Georgiadou O, Home based neuromuscular electrical stimulation as a new rehabilita-
Zakynthinos S, Roussos C. Skeletal muscle adaptations to interval tive strategy for severely disabled patients with chronic obstructive pul-
training in patients with advanced COPD. Chest 128: 383845, 2005. monary disease (COPD). Thorax 57: 33337, 2002.
66. Coppoolse R, Schols AM, Baarends EM, Mostert R, Akkermans MA, 87. Bourjeily-Habr G, Rochester C, Palermo F, Snyder P, Mohsenin V.
Janssen PP, Wouters EF. Interval versus continuous training in patients Randomised controlled trial of transcutaneous electrical muscle stimu-
with severe COPD: A randomized clinical trial. Eur Respir J 14: lation of the lower extremities in patients with chronic obstructive pul-
25863, 1999. monary disease. Thorax 57: 104549, 2002.

721
Asthma and COPD: Basic Mechanisms and Clinical Management

88. Zanotti E, Felicetti G, Maini M, Fracchia C. Peripheral muscle strength 94. Garcia-Aymerich J, Lange P, Benet M, Schnohr P, Anto JM. Regular
training in bed-bound patients with COPD receiving mechanical ven- physical activity reduces hospital admission and mortality in chronic
tilation: Eect of electrical stimulation. Chest 124: 29296, 2003. obstructive pulmonary disease: A population-based cohort study.
89. Vivodtzev I, Pepin JL, Vottero G, Mayer V, Porsin B, Levy P, Thorax, 2006.
Wuyam B. Improvement in quadriceps strength and dyspnea in daily 95. Pitta F, Troosters T, Probst V, Langer D, Decramer M, Gosselink R.
tasks after 1 month of electrical stimulation in severely deconditioned Are patients with COPD more active after pulmonary rehabilitation.
and malnourished COPD. Chest 129: 154048, 2006. Chest, 2008, E-pub ahead of Print.
90. Dal CS, Napolis L, Malaguti C, Gimenes AC, Albuquerque A, 96. Sewell L, Singh SJ, Williams JE, Collier R, Morgan MD. Can indi-
Nogueira CR, De Fuccio MB, Pereira RD, Bulle A, McFarlane N, vidualized rehabilitation improve functional independence in elderly
Nery LE, Neder JA. Skeletal muscle structure and function in response patients with COPD? Chest 128: 1194200, 2005.
to electrical stimulation in moderately impaired COPD patients. Respir 97. Mercken EM, Hageman GJ, Schols AM, Akkermans MA, Bast A,
Med, 2006. Wouters EF. Rehabilitation decreases exercise-induced oxidative stress
91. Maillefert JF, Eicher JC, Walker P, Dulieu V, Rouhier-Marcer I, Branly F, in chronic obstructive pulmonary disease. Am J Respir Crit Care Med
Cohen M, Brunotte F, Wolf JE, Casillas JM, Didier JP. Eects of low- 172: 9941001, 2005.
frequency electrical stimulation of quadriceps and calf muscles in patients 98. Steele BG, Belza B, Cain KC, Coppersmith J, Lakshminarayan S,
with chronic heart failure. J Cardiopulm Rehabil 18: 27782, 1998. Howard J, Haselkorn JK. A randomized clinical trial of an activity
92. Harris S, LeMaitre JP, Mackenzie G, Fox KA, Denvir MA. A ran- and exercise adherence intervention in chronic pulmonary disease.
domised study of home-based electrical stimulation of the legs and Arch Phys Med Rehabil 89: 40412, 2008.
conventional bicycle exercise training for patients with chronic heart 99. Coronado M, Janssens JP, de Muralt B, Terrier P, Schutz Y, Fitting
failure. Eur Heart J 24: 87178, 2003. JW. Walking activity measured by accelerometry during respiratory
93. Haskell WL, Lee IM, Pate RR, Powell KE, Blair SN, Franklin BA, rehabilitation. J Cardiopulm Rehabil 23: 35764, 2003.
Macera CA, Heath GW, Thompson PD, Bauman A. Physical activity and 100. Steele BG, Belza B, Hunziker J, Holt L, Legro M, Coppersmith J,
public health. Updated recommendation for adults from the American Buchner D, Lakshminaryan S. Monitoring daily activity during pul-
College of Sports Medicine and the American Heart Association. monary rehabilitation using a triaxial accelerometer. J Cardiopulm
Circulation, 2007. Rehabil 23: 13942, 2003.

722
Surgical and Other Mechanical
Procedures
59 CHAPTER

DECLARATIONS OF INTEREST would be to reduce lung volume to t the Michael I. Polkey and
expiratory phase of the respiratory cavity [by] Pallav L. Shah
removing lung tissue that has no respiratory
Our institutions have received research grants for function [1]. In fact, perhaps due to diculty Royal Brompton Hospital and National
our group from Asthmalx, Emphasys, Broncus with patient selection and the poorer standard Heart and Lung Institute, London, UK
and PortAero who have patents and/or technol- of anesthetic and postoperative care at the time
ogies relevant to this article. the technique was never widespread and except
for occasional patients who could benet from
a bullectomy [2, 3] or a Monaldi procedure [4],
the only surgical therapy available until recently
was single or double lung transplantation.
INTRODUCTION Transplantation remains a moderately danger-
ous procedure with a mortality in the rst year
Although drugs, together with pulmonary reha- of approximately 15%. More importantly there
bilitation, will form the mainstay of treatment are insucient donor organs for the number of
for most patients with chronic obstructive pul- patients willing to undergo the procedure.
monary disease (COPD) as the lung destruction For this reason surgical lung volume reduc-
progresses, these therapies become insuciently tion was revived in the 1990s by Joel Cooper
eective for a minority of patients who may then and colleagues [5]; their original data was con-
be prepared to consider a surgical treatment. As a ned to patients with marked hyperination
general rule, these therapies should not be oered and contained no control group but showed, at
to patients who have not completed pulmonary 6 months, a 51% improvement in FEV1 and a
rehabilitation and should not be oered outside reduction in oxygen usage such that 70% of
specialist centers where a truly multidisciplinary patients were able to discontinue domiciliary
approach is available. Asthma, being a condition oxygen therapy.
characterized by variable airow obstruction, is The technique was later submitted to
seldom suitable to a surgical approach although randomized controlled trials of which the
the experimental approach of thermoplasty will most important was the National Emphysema
be briey discussed. Treatment Trial (NETT) study [6] (vide infra)
which accounted for 73% of all presently rand-
omized patients. The results of the smaller stud-
ies are summarized in the table and have been
CHRONIC OBSTRUCTIVE PULMONARY usefully summarized in a Cochrane review [7]
DISEASE (Table 59.1). Broadly speaking, the review found
that at 90 days there were signicant benets
conferred by surgery with regard to quality of life
Surgical procedures whether measured with generic- or disease-spe-
cic instruments but it must be borne in mind
In the 1950s Owen Brantigan formed the con- that surgery itself has a signicant placebo benet.
cept that a surgical treatment of emphysema Other benets from surgery included improved

723
Asthma and COPD: Basic Mechanisms and Clinical Management

TABLE 59.1 Results of lung volume reduction surgery.

Author FEV1 (ml) FEV1 (%) Field walking test (m) Field walking test (%)

Criner, 1999 [8] 200 31 33 (p ns) 12

Geddes, 2000 [9] 70 10 50 25

Goldstein, 2003 [10] 300 38 42 11

Hillerdal, 2005 [11] 230 35 99 43

Miller, 2006 [12] 265 30 29 10

LVRS

With treatment

NNH: 16

95% confidence interval

8 to 35

Key

Good outcome FIG. 59.1 Ninety-day outcome of LVRS


reproduced from the meta-analysis by Tiong
Bad outcome et al. [7]. A second figure may be found in the
paper showing the results when the high
Worse with Rx risk patients from NETT were excluded [13].
Interestingly although this exclusion reduces
the number of patients who are worse after
surgery but not the mortality; however the
number needed to treat rises.

exercise performance, reduced emergency room attendance in and gas transfer tended to be more sustained than those in
postoperative year 2 (but not 1). FEV1. Overall the data showed that the surgical group was
The NETT study is by far the biggest study to assess better than the control group for at least 3 years.
lung volume reduction surgery (LVRS) [6]; briey after pul- The current position for surgical LVRS is that it may
monary rehabilitation 1218 were randomized to receive either best be described as useful only in patients with the most
LVRS or continued medical care (Fig. 59.1). An early analysis favorable radiological appearances (heterogenous upper
showed that patients with a homogenous distribution of dis- zone disease), who are a minority of emphysema patients.
ease and either a FEV1  20% predicted or a carbon monoxide Even so the operation is conned to those patients will-
gas transfer (TLCO)  20% predicted had a poor outcome and ing to accept the perioperative mortality (typically 5%) but
were excluded from the remainder of the study [13]. The study who meet the safety criteria. These will vary slightly from
showed that in the surgical group (compared with medical institution to institution but, for example, we require an
therapy) the exercise capacity improved on an incremental test FEV1  20%, a TLCO  30%, and a shuttle walk distance
by more than 10 W in 28% and 15% of operated patients but (after rehabilitation) of 150 m. A further problem is that a
only 4% and 3% of controls at 6 and 24 months respectively minority of patients experience no benet but these patients
(p  0.001). Surgical patients were more likely to achieve an cannot be reliably predicted prior to surgery. As a result the
improved 6-min walk distance and exhibited an approximately number of procedures presently performed is small.
30% reduction in exacerbations [14]. An accompanying paper
[15] found the dierence in total cost at 3 years between the
two groups was $36,392. Minimally invasive surgical therapies
Long-term follow up of the randomized controlled
trials conducted is conned to data from the Brompton Since LVRS was demonstrated to work in carefully selected
study [9]; Lim and colleagues [16] found that improvement patients the question arose as to whether lung volume
in residual volume (RV)/total lung capacity (TLC) ratio reduction could be achieved more safely and cheaply using

724
Surgical and Other Mechanical Procedures 59
a nonsurgical mechanical result. In principle, lung volume
reduction could be achieved by the introduction of a blocker
(subsequently modied to a one-way valve) into the airway.
Sabaratnam Sabanathan, an English surgeon, rst suggested
endobronchial blockade as an alternative approach. Sadly
he died whilst developing the concept but his preliminary
data, from procedures performed between November 1996
and April 1997, [17] were published posthumously and
showed that signicant improvement was possible using
this approach. However, the blockers initially used were not
designed for endobronchial occlusion and later patients in
the series had devices inserted that were custom built in
Sabanathans hospital. Signicant problems were observed
with migration of the rst type of blockers and in the case
of the custom built devices one episode of blocker disinte-
gration occurred.
Biotechnology companies subsequently took up the
challenge and two devices are commercially available. The
Emphasys system (Emphasys Medical, Inc; Redwood City,
CA) uses a one-way duckbill valve that is inserted in such a
way that air is permitted to leave the subtended segment but
not enter it. The valve is secured and supported by a collaps-
ible metal framework. The initial valves (Fig. 59.2A) were
inserted over a guide wire but later models (Fig. 59.2B) can
be inserted through the working channel of a exible bron-
choscope. Five human studies have been reported with the
valve [1822], and the company has recently reported the
combined data [23].
The Spiration (Spiration, Inc; Redmond, WA) device is
an umbrella arrangement (Fig. 59.2C) arranged so that the con-
vex surface is positioned distally with the aim again of allow-
ing expiratory but not inspiratory ow; so far human data are
reported from an uncontrolled study of 30 patients [24]. These
patients were subjected to bilateral upper lobe placement. The
reported safety data were satisfactory but less than a third of
patients showed improvements in FEV1 or 6 min walking test (C) Membrane
greater than 15%.
Similarly, although animal data exist for the tissue Struts
engineering approach [25]; human data so far are conned
to subsegments of a single lung in six subjects [26]. Again Hub Removal
rod tip
the safety prole was satisfactory.

Experience with endobronchial therapy


Removal
Snell et al. [18] used an early version of the valve and aimed rod
for bilateral lung volume reduction. No improvement was Anchors pads
observed in any parameter except TLCO, although they also Anchors tips
found valve insertion to be safe and well tolerated. Toma
et al. reported the use of the Emphasys valve for broncho-
FIG. 59.2 Endobronchial valves (A) Initial Emphasys endobronchial
scopic lung volume reduction [22]. Data from eight patients valve. (B) Modified Emphasys endobronchial valve, suitable for transcopic
was presented; despite the small sample size a statistically sig- insertion. (C) Intrabronchial valve (Spiration Inc).
nicant improvement in FEV1 was observed with an increase
from 0.79 l (range 0.611.07) to 1.06 l [0.751.22] (dier-
ence 34%, p 0.028). Other investigators in uncontrolled valve insertion and made key measurements 4 weeks after
series found improvements in FEV1 either alone [19] or with the procedure. As in previous studies, we observed a sta-
signicant improvements in plethysmographic lung volumes tistically signicant improvement in mean FEV1 (2831%
and mean TLCO [20]. predicted, p  0.05) and in mean TLCO (3641% predicted,
Hopkinson et al. reported the only study of BLVR in p 0.016). A statistically signicant improvement in
which dynamic hyperination was directly measured [21]. TLC and functional residual capacity (FRC) was observed
We studied 19 patients treated with unilateral endobronchial though this failed to reach signicance in the case of RV.

725
Asthma and COPD: Basic Mechanisms and Clinical Management

A priori we divided patients into improvers and nonimprov- noninvasive approaches are feasible. Endobronchial blockade
ers. Improvers were dened as patients who were able to is likely to be the most suitable for the minority of patients
show an increase of both 60 s and 30% in endurance time with no collateral ventilation and heterogenous upper zone
on the constant rate cycle endurance test. Nine of nine- disease. Extrapulmonary bypass is likely to be most suitable
teen patients (47%) met this criterion. In stepwise regres- for the majority of patients with homogenous disease, resting
sion analysis only change in resting inspiratory capacity and hyperventilation, and collateral ventilation. In both cases piv-
TLCO were retained as independent predictors, producing otal studies have yet to be reported and neither treatment can
an equation that explained 81% of the variation in endur- be recommended (except on a compassionate basis) until they
ance time (p  0.0001). If patients with atelectasis were are shown to be both safe and eective. Collateral ventilation is
excluded, again, only the same two variables were retained frequently but not universally present in patients with COPD
in the model (r 2 0.61; p 0.002). [33] and can be measured in man [34, 35] and we speculate
The combined Emphasys data set was recently reported that these techniques may become part of the work up when
[23]: 98 patients were studied with an average number of assessing patients for these therapies.
4 valves per patient, the majority had complete unilateral
(48%) or bilateral (21%) lobar occlusion. On average sig-
nicant improvements were observed in FEV1 (mean 60 ml,
10%) and in walking distance (37 m, 23%). Clinically the ASTHMA
mean improvements are at the lower range of what might be
considered worthwhile. The safety prole compared favora-
bly to surgical LVRS. Post hoc subset analysis showed that the Bronchial thermoplasty is a novel approach in the treatment
biggest improvements were seen in those patients submitted of asthma, which targets smooth muscle within the airways.
to complete lobar occlusion, the lowest FEV1 and the great- In asthma there is frequently an increase in the number
est hyperination. and the size of smooth muscle within the airway and this
Emphasys have recently concluded a multicenter ran- in turn causes bronchoconstriction, hyperresponsiveness
domized controlled study (for design see Ref. [27]). The and inammation. Bronchial thermoplasty uses low energy
results were presented orally at the Annual Congress of the radio frequency to ablate the smooth muscle. The epithe-
European Respiratory Society in September 2007 but are lium, mucous glands, and nerves appear to regenerate but
not yet available in print form. there is permanent reduction in the smooth muscle bulk.
A feasibility study was performed in patients with lung
cancer who were due to undergo surgical resection. Bronchial
Extrapulmonary bypass thermoplasty was performed in these individuals up to
3 weeks prior to their surgery and segmental bronchi within
An alternative approach to emphysema was proposed the lobe that was to be resected were treated with bronchial
by Macklem in 1978 [28]. Based on the observation that thermoplasty. This study demonstrated that bronchial ther-
emphysematous lungs, unlike normal lungs, demonstrated moplasty was well tolerated and resulted in a signicant
signicant collateral ventilation he proposed that a physical reduction in smooth muscle mass in the treated airways [36].
connection between the lung parenchyma and the atmos- A safety study in 16 subjects with mild to moder-
phere could reduce hyperination. ate asthma was subsequently performed [37]. The patients
So far, one biotechnology company has created a prod-
underwent bronchial thermoplasty in three separate treat-
uct which attempts to exploit this property. Broncus Inc
ments, initially one of the lower lobes was treated, followed
(Mountain View CA 94043) has developed a stent which is
by the contralateral lower lobe 3 weeks later, and nally both
placed in a bronchial segment to permit exhaled air to bypass
upper lobes were treated on the last treatment setting a fur-
the point of ow limitation. So far the device has been dem-
ther 3 weeks later. The patients all underwent detailed moni-
onstrated to be feasible in dogs with patency enhanced by the
toring with lung function tests, methacholine challenge, and
use of drug eluting stents [29]. The stents increase expiratory
an assessment of their symptom-free days was made. The
ow in explanted human lungs [30, 31]. An uncontrolled study
study demonstrated a signicant improvement in morning
has been reported in 35 patients with emphysema [32]: for the
and evening peak ows and also an increase in symptom-
group as a whole a 7.3% reduction in FEV1 and a 37% increase
free days following treatment with bronchial thermoplasty.
in 6-min walk were observed. However post hoc analysis showed There was a reduction in bronchial hyperresponsiveness in
that the greatest benet was observed in patients with the most the patients treated, which was maintained at 1 and 2 years
preprocedure hyperination as one might expect. 69% of stents following treatment with bronchial thermoplasty.
examined bronchscopically were patent at 6 months after the A further randomized study in 112 subjects with mod-
procedure. An ongoing randomized controlled trial (RCT) is erate or severe persistent asthma has also shown signicant
presently underway which is expected to report in 2009. improvements in quality of life, symptom scores, a signicant
increase in symptom-free days, and morning peak expira-
Overview of options for emphysema tory ow measurements [38]. An increase in adverse events
immediately following bronchial thermoplasty was observed.
At present the only evidence-based therapy is LVRS but this These events were usually mild and well tolerated. They com-
is unattractive to purchasers, patients, and surgeons because prised of an increase in breathlessness, cough, wheezing,
of the cost, risk, and the relatively high safety criteria which and chest discomfort. The majority of these events occurred
are necessary to avoid an unacceptable mortality. Two main within 1 day of the procedure and resolved within 7 days of

726
Surgical and Other Mechanical Procedures 59
onset. There was no increase in adverse events 6 weeks after group. The clinical studies that have thus far been published
the last treatment and following this period the adverse event are open-label randomized studies but a pivotal double-
rates were similar in both treatment and controlled groups. blind sham controlled study is nearing completion with the
In this study baseline measurements were conducted results due in late 2008. This is a uniquely designed inter-
during a 2-week period when the patients were withdrawn ventional study which attempts to reduce the obvious biases
from their long active bronchodilators and similar analyses of an open-label study and will more critically evaluate the
were performed on bronchodilators at 3, 6, and 12 months. safety and ecacy of bronchial thermoplasty in moder-
Measurements at this time point demonstrated signicant ate and severe asthma patients; until this study is formally
improvement in the number of symptom-free days and on reported it seems appropriate to reserve thermoplasty for a
average treatment subjects had 86 additional symptom-free research setting.
days per year compared to the control group. There was also an
improvement in symptom scores, asthma quality of life ques- References
tionnaire (AQLQ), and asthma control questionnaire (ACQ)
scores. There was a signicant reduction in 2-agonists used
1. Noehren TH, Barach AL, Brantigan OC, Smart RH. Pulmonary
that would equate to approximately 2 inhalers per patient over emphysema prevention and care. Dis Chest 45: 492502, 1964.
the 1-year treatment period. Finally, there was a reduction in 2. Soni R, McKeough ZJ, Dobbin CJ, Young IH. Gas exchange and exer-
mild and severe exacerbations and in the treatment group there cise tolerance following bullectomy. Respirology 10(1): 12023, 2005.
were approximately 10 fewer mild exacerbations than in the 3. ODonnell DE, Webb KA, Bertley JC, Chau LK, Conlan AA.
control group. The main limitation of this large study was that Mechanisms of relief of exertional breathlessness following unilateral
it was an open-label design and hence, subject to bias. bullectomy and lung volume reduction surgery in emphysema. Chest
A small study was performed in patients with severe 110(1): 1827, 1996.
refractory asthma who were on more than 750 g uticasone 4. Venn GE, Williams PR, Goldstraw P. Intracavity drainage for bullous,
propionate per day in addition to long-acting bronchodilators emphysematous lung disease: Experience with the Brompton tech-
nique. Thorax 43(12): 9981002, 1988.
[39]. A signicant proportion of patients were also on main-
5. Cooper J, Patterson G, Sundaresan R, Trulock E, Yusen R, Pohl M
tenance of oral steroids. In this study, 15 patients were rand- et al. Results of 150 consecutive bilateral lung volume reduction pro-
omized to bronchial thermoplasty and there were 17 controlled cedures in patients with severe emphysema. J Thorac Cardiovasc Surg
subjects. The study design was to randomize the patients to 112(5): 131930, 1996. The. 1996 1996/11//
treatment or control followed by a 6-week stabilization period. 6. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R, Ries A et al.
The patients were maintained on their regular treatment for A randomized trial comparing lung-volume-reduction surgery with medi-
the following 14 weeks, and then the patients entered a steroid cal therapy for severe emphysema. N Engl J Med 348(21): 205973, 2003.
weaning phase where the dose of oral steroids or inhaled ster- 7. Tiong LU, Davies R, Gibson PG, Hensley MJ, Hepworth R,
oids was reduced by 2025% in 2-week steps. Following the Lasserson TJ et al. Lung volume reduction surgery for diuse emphy-
14-week steroid-wean phase, patients were maintained on a sema. Cochrane Database Syst Rev (4), 2006, CD001001.
reduced dose for a further 16 weeks and assessments performed 8. Criner GJ, Cordova FC, Furukawa S, Kuzma AM, Travaline JM,
Leyenson V et al. Prospective randomized trial comparing bilateral
over the two study periods. Overall, bronchial thermoplasty
lung volume reduction surgery to pulmonary rehabilitation in severe
was well tolerated although there was an increase in adverse chronic obstructive pulmonary disease. Am J Respir Crit Care Med
events over the treatment period. During this treatment period 160(6): 201827, 1999.
there were seven hospitalizations for adverse events in four 9. Geddes D, Davies M, Koyama H, Hansell D, Pastorino U, Pepper J
subjects in the bronchial thermoplasty group and none in the et al. Eect of lung-volume-reduction surgery in patients with severe
control group. In the posttreatment period the number of hos- emphysema. N Engl J Med 343(4): 23945, 2000.
pitalizations and adverse events were similar in both treatment 10. Goldstein RS, Todd TR, Guyatt G, Keshavjee S, Dolmage TE,
groups. There were signicant improvements in a number of van Rooy S et al. Inuence of lung volume reduction surgery (LVRS)
parameters including lung function and quality of life scores in on health related quality of life in patients with chronic obstructive
the treatment group compared to the control group both prior pulmonary disease. Thorax 58(5): 40510, 2003.
11. Hillerdal G, Lofdahl CG, Strom K, Skoogh BE, Jorfeldt L, Nilsson F
to reduction in steroids and also following the steroid-wean
et al. Comparison of lung volume reduction surgery and physical train-
phase. Four of the eight patients in the bronchial thermoplasty ing on health status and physiologic outcomes: A randomized control-
group were completely weaned o their oral steroids in con- led clinical trial. Chest 128(5): 348999, 2005.
trast to only one of the seven patients in the control group who 12. Miller JD, Malthaner RA, Goldsmith CH, Goeree R, Higgins D,
was weaned o oral steroids. Prebronchodilator FEV1 was Cox PG et al. A randomized clinical trial of lung volume reduction
improved by around 15% in the treatment group compared to surgery versus best medical care for patients with advanced emphy-
the control group, and there was an over 26% reduction in res- sema: A two-year study from Canada. Ann Thorac Surg 81(1): 31420,
cue medication use in the treatment group. There were signi- discussion 2021, 2006.
cant improvements in asthma control scores and quality of life 13. National Emphysema Treatment Trial Research Group. Patients at
high risk of death after lung-volume-reduction surgery. N Engl J Med
both during the steroid weaning phase and following steroid
345(15): 107583, 2001.
reduction in the bronchial thermoplasty group compared to 14. Washko GR, Fan VS, Ramsey SD, Mohsenifar Z, Martinez F, Make BJ
the control group. et al. The eect of lung volume reduction surgery on chronic obstructive
The clinical studies thus far have demonstrated that pulmonary disease exacerbations. Am J Respir Crit Care Med 177(2): 164
bronchial thermoplasty is safe and appears to have signicant 69, 2008.
improvements in quality of life, peak ow measurements, 15. Ramsey SD, Berry K, Etzioni R, Kaplan RM, Sullivan SD, Wood DE.
and the number of symptom-free days in asthma patients. Cost eectiveness of lung-volume-reduction surgery for patients with
FEV1 only appears to improve in the severe refractory severe emphysema. N Engl J Med 348(21): 2092102, 2003.

727
Asthma and COPD: Basic Mechanisms and Clinical Management

16. Lim E, Ali A, Cartwright N, Sousa I, Chetwynd A, Polkey M et al. 28. Macklem PT. Collateral ventilation. N Engl J Med 298(1): 4950,
Eect and duration of lung volume reduction surgery: Mid-term results 1978.
of the Brompton trial. Thorac Cardiovasc Surg 54(3): 18892, 2006. 29. Choong CK, Phan L, Massetti P, Haddad FJ, Martinez C, Roschak E
17. Sabanathan S, Richardson J, Pieri-Davies S. Bronchoscopic lung vol- et al. Prolongation of patency of airway bypass stents with use of drug-
ume reduction. J Cardiovasc Surg (Torino) 44(1): 1018, 2003. eluting stents. J Thorac Cardiovasc Surg 131(1): 6064, 2006.
18. Snell G, Holsworth L, Borrill ZL, Thomson KR, Kal V, Smith JA 30. Lausberg HF, Chino K, Patterson GA, Meyers BF, Toeniskoetter PD,
et al. The potential for bronchoscopic lung volume reduction using Cooper JD. Bronchial fenestration improves expiratory ow in
bronchial prostheses. Chest 124: 107380, 2003. emphysematous human lungs. Ann Thorac Surg 75(2): 39397,
19. Yim AP, Hwong TM, Lee TW, Li WW, Lam S, Yeung TK et al. Early discussion 8, 2003.
results of endoscopic lung volume reduction for emphysema. J Thorac 31. Choong CK, Macklem PT, Pierce JA, Lefrak SS, Woods JC,
Cardiovasc Surg 127(6): 156473, 2004. Conradi MS et al. Transpleural ventilation of explanted human lungs.
20. Venuta F, de Giacomo T, Rendina EA, Ciccone AM, Diso D, Thorax 62(7): 62330, 2007.
Perrone A et al. Bronchoscopic lung-volume reduction with one-way 32. Cardoso PF, Snell GI, Hopkins P, Sybrecht GW, Stamatis G,
valves in patients with heterogenous emphysema. Ann Thorac Surg Ng AW et al. Clinical application of airway bypass with paclitaxel-elut-
79(2): 41116, discussion 67, 2005. ing stents: Early results. J Thorac Cardiovasc Surg 134(4): 97481, 2007.
21. Hopkinson NS, Toma TP, Hansell DM, Goldstraw P, Moxham J, 33. Higuchi T, Reed A, Oto T, Holsworth L, Ellis S, Bailey MJ et al.
Geddes DM et al. Eect of bronchoscopic lung volume reduction on Interlobar collaterals are related to radiological heterogeneity in severe
dynamic hyperination and exercise in emphysema. Am J Respir Crit emphysema. Thorax, 2006.
Care Med 171(5): 45360, 2005. 34. Morrell NW, Wignall BK, Biggs T, Seed WA. Collateral ventilation
22. Toma TP, Hopkinson N, Hillier J, Hansell DM, Morgan C, and gas exchange in emphysema. Am J Respir Crit Care Med 150(3):
Goldstraw PG et al. Bronchoscopic volume reduction with valve 63541, 1994.
implants in patients with severe emphysema. Lancet 361: 93133, 2003. 35. Terry PB, Traystman RJ, Newball HH, Batra G, Menkes HA.
23. Wan IY, Toma TP, Geddes DM, Snell G, Williams T, Venuta F Collateral ventilation in man. N Engl J Med 298(1): 1015, 1978.
et al. Bronchoscopic lung volume reduction for end-stage emphysema: 36. Miller JD, Cox G, Vincic L, Lombard CM, Loomas BE, Danek CJ.
Report on the rst 98 patients. Chest 129(3): 51826, 2006. A prospective feasibility study of bronchial thermoplasty in the human
24. Wood DE, McKenna RJ Jr., Yusen RD, Sterman DH, Ost DE, airway. Chest 127(6): 19992006, 2005.
Springmeyer SC et al. A multicenter trial of an intrabronchial valve for treat- 37. Cox G, Miller JD, McWilliams A, Fitzgerald JM, Lam S. Bronchial
ment of severe emphysema. J Thorac Cardiovasc Surg 133(1): 6573, 2007. thermoplasty for asthma. Am J Respir Crit Care Med 173(9): 96569,
25. Ingenito EP, Berger RL, Henderson AC, Reilly JJ, Tsai L, Homan A. 2006.
Bronchoscopic lung volume reduction using tissue engineering princi- 38. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC
ples. Am J Respir Crit Care Med 167(5): 77178, 2003. et al. Asthma control during the year after bronchial thermoplasty.
26. Reilly J, Washko G, Pinto-Plata V, Velez E, Kenney L, Berger R et al. N Engl J Med 356(13): 132737, 2007.
Biological lung volume reduction: A new bronchoscopic therapy for 39. Pavord ID, Cox G, Thomson NC, Rubin AS, Corris PA, Niven RM
advanced emphysema. Chest 131(4): 110813, 2007. et al. Safety and ecacy of bronchial thermoplasty in symptomatic,
27. Strange C, Herth FJ, Kovitz KL, McLennan G, Ernst A, Goldin J severe asthma. Am J Respir Crit Care Med 176(12): 118591, 2007.
et al. Design of the Endobronchial Valve for Emphysema Palliation
Trial (VENT): A non-surgical method of lung volume reduction.
BMC Pulm Med 7: 10, 2007.

728
Other Therapies
60 CHAPTER

inuenza vaccination every year [7]. The Global Neil C. Thomson


VACCINES AND ANTIVIRAL AGENTS Initiative for Chronic Obstructive Lung Disease
(GOLD) guidelines emphasize the importance Department of Respiratory Medicine,
Influenza vaccines of targeting elderly individuals with COPD [8]. Division of Immunology, Infection and
The live-attenuated inuenza vaccine has been Inflammation, University of Glasgow,
licensed recently in the United States for use in Glasgow, UK
Efficacy healthy persons aged 549 years and it has been
A recent systematic review of trials undertaken advocated as the preferred vaccine for use in chil-
to assess the ecacy of inuenza vaccination in dren with asthma [4].
chronic obstructive pulmonary disease (COPD)
patients concluded that inactivated vaccine reduces
the rate of exacerbations [1]. In asthma the benets Antiviral agents against influenza
of inactivated vaccination has not been proven [2].
Administration of live-attenuated inuenza vac- Neuraminidase inhibitors are a class of antiviral
cine by intranasal spray is eective and safe in agents that are active against both inuenza A
both healthy adults and in children with asthma. and inuenza B. The neuraminidase inhibitors,
The cold adapted live vaccine produces supe- inhaled zanamivir (Relenza) and oral oseltamivir
rior protection compared to the inactive vaccine (Tamiu) have received regulatory approval in
in children with asthma [3, 4]. The addition of some countries for the treatment of acute inu-
the live-attenuated vaccine to inactivated vaccine enza and for chemoprophylaxis [6]. If these agents
oers no additional protection in COPD [1]. are started within the rst 2 days after the onset
of illness, then treatment will shorten the duration
Adverse effects of symptoms by 1 to 1 1/2 days and in some stud-
Vaccination is contraindicated in those individuals ies the need for antibiotics is reduced. Oseltamivir
hypersensitive to eggs, although the risk of reac- administered to inuenza-infected children with
tions is low. The inactivated inuenza vaccines can asthma aged 612 years is well tolerated and pro-
be safely administered to adults and children with duces improvements in pulmonary function and
asthma and to patients with COPD without any fewer exacerbation of asthma up to day 7 [9].
increase in exacerbation rates in either condition Antiviral drugs amantadine and rimantadine are
[2, 4, 5]. The live-attenuated inuenza vaccine can not now recommended for treating the inuenza
be safely used in children with asthma [4]. A because of concerns about drug resistance [6].
Antiviral drugs should be considered for patients
Recommendations with asthma and COPD, if started within 2 days
Health authorities in the United States and in of infection, particularly in those who have not
many European countries recommend annual been vaccinated. It has also been recommended
inactivated inuenza vaccination for adults and that chemoprophylaxis should be considered in
children with chronic pulmonary disease includ- vaccinated members of high-risk groups, includ-
ing asthma [6]. The Global Initiative for Asthma ing patients with asthma and COPD, because
(GINA) guidelines recommend that patients the combination of vaccination and antiviral drug
with moderate to severe asthma should receive increases protection against inuenza.

729
Asthma and COPD: Basic Mechanisms and Clinical Management

Pneumococcal vaccines ANTIOXIDANTS


Efficacy
In elderly individuals with chronic lung disease including Antioxidant enzymes form the rst line of defense in the
COPD, pneumococcal vaccination has been reported to result lungs. Antioxidants may reduce chronic airway inamma-
in fewer outpatient visits, fewer hospitalizations, and fewer tion by neutralizing the damage produced by reactive oxy-
deaths [10]. A Cochrane review of four studies of patients gen species and also possibly by preserving the inhibitory
with COPD, however, found no evidence for a reduction in action of antiproteases. Antioxidant genes, however, may be
exacerbation rates, morbidity, or mortality following pneu- upregulated by oxidative stress [17] and the dose of admin-
mococcal vaccination [11]. Routine revaccination is not gen- istered exogenous antioxidant may need to be titrated to
erally recommended because of the increased incidence of avoid suppression of endogenous antioxidant activity.
adverse reactions. A Cochrane review found limited evidence
of eectiveness in children and adults with asthma [12].
Asthma
Adverse effects
Transient erythema at the site of injection is common. Dietary antioxidants
Hypersensitivity reactions may occur. Dietary antioxidants such as vitamin C, vitamin E, caraten-
oids, and selenium are derived mainly from fruit and vegeta-
Recommendations bles. Reduced circulating levels of dietary antioxidants have
been found in asthma [18, 19] raising the hope that clinical
Health authorities in some European countries and the benets would result from dietary supplementation with these
United States recommend polyvalent pneumococcal vacci- compounds. However, clinical trials of dietary supplementation
nation for all patients with chronic lung disease [13]. The with vitamin C, vitamin E, or selenium have shown no benet
GOLD guidelines recommend vaccination for patients aged in adults with mild-to-moderate asthma [2022].
65 years and older with COPD [8].

Inhaled glutathione
Haemophilus influenza vaccine The antioxidant glutathione administered to adults with
mild asthma-induced bronchoconstriction [23].
A systematic review of six small studies of oral immunization
with monobacterial whole-cell, killed, nontypeable H. infuenzae
vaccine in patients with recurrent acute exacerbations of chronic COPD
bronchitis in the autumn found that vaccination reduced
the number and severity of exacerbations over the following Oxidative stress is increased in smokers and reactive oxygen
winter [14]. The authors suggested that one episode of bronchi- species may be involved in the pathogenesis of COPD [24].
tis may be prevented for every ve individuals vaccinated. The There is evidence that endogenous antioxidant capacity is
review concluded however that a large clinical trial into the long- reduced in COPD and so antioxidants may be useful in the
term eects of H. infuenzae vaccine was needed. Its routine use is treatment of COPD.
not recommended in the management of asthma or COPD.

Dietary antioxidants
Multi-component oral bacterial vaccines There is some evidence linking dietary deciency of antioxi-
dants and COPD. There are no controlled studies of vita-
OM-85 BV (BronchoVaxom) is a mixture of eight bacterial mins C or E supplementation in COPD. -Carotene and
products thought to be important in respiratory infections. It -tocopherol supplements had no benet on the symptoms
is administered orally and is thought to act as a nonspecic of COPD [25].
immunostimulant. A randomized controlled study in 381
patients with COPD treated with OM-85 BV did not reduce
the risk of having at least one episode of an acute exacerbation N-Acetylcysteine
(primary outcome) over a 6-month period compared with pla- N-Acetylcysteine, which is a mucolytic, may also act as an
cebo. Treatment with OM-85, however, reduced the risk and antioxidant by providing cysteine intracellularly for increased
total number of days of hospitalization for a respiratory prob- production of glutathione [26]. In vitro N-acetylcysteine
lem by 30% and 55% respectively [15]. A systematic review of reduces neutrophil chemotaxis and in smokers it reduces
13 studies, most of which were assessed to be of low quality, the number and activity of bronchoalveolar neutrophils and
found that oral puried bacterial extracts improve symptoms alveolar macrophages. N-acetylcysteine preserves the inhib-
in patients with chronic bronchitis and COPD, but that there itory action of antiproteases from oxidative inactivation.
was insucient evidence to suggest that they prevent exacerba- It has not been established, however, whether any clinical
tions [16]. Skin and urological adverse eects were commonly benets of N-acetylcysteine and other mucolytics can be
reported. Further studies are required before oral vaccines are attributed to the antioxidant properties of these drugs (see
considered for use in COPD. Mucolytic Drugs).

730
Other Therapies 60
alterations in cell volume and function [30]. In vitro studies
MUCOLYTIC DRUGS of human inammatory cells and in vivo studies in experi-
mental animals have shown that both drugs inhibit func-
Mucolytic drugs such as N-acetylcysteine, ambroxol, tions of a variety of inammatory cells including mast cells,
erdosteine, iodinated glycerol, methylcysteine, and carbo- eosinophils, neutrophils, platelets, and alveolar macrophages
cysteine reduce the viscosity of mucous in vitro and have [3032]. Nedocromil sodium has either similar or slightly
been assessed in several clinical trials in COPD. These drugs greater potency to that of sodium cromoglycate. Both sodium
have not been tested in asthma. cromoglycate and nedocromil sodium may also have inhibitory
eects on sensory nerve endings in the lung, thus preventing
the release of tachykinins.
COPD

Efficacy Asthma
A Cochrane systematic review of 26 randomized control-
led trials of mucolytic drugs including N-acetylcysteine in Anti-inflammatory effects
the treatment of chronic bronchitis or COPD found that There is limited and conicting evidence for an anti-
these drugs produced a small reduction in the frequency of inammatory eect of cromones in asthma [3335].
acute exacerbations and the total days of disability compared
with placebo [27]. The ecacy and side-eect prole was sim- Clinical studies
ilar between N-acetylcysteine and the other mucolytic drugs.
A further systematic review of 11 randomized trials Bronchial challenge
comparing oral N-acetylcysteine with placebo administered Sodium cromoglycate and nedocromil sodium are equally
from 12 to 24 weeks in patients with chronic bronchitis con- eective in attenuating exercise-induced asthma [36]. The
cluded that active treatment reduced the risk of exacerba- early and late response to allergen and the allergen-induced
tions and improved symptoms without an increased risk of seasonal increase in nonallergic bronchial reactivity can be
side eects [28]. Stey and colleagues [28] estimated that of prevented by both drugs [31]. Nedocromil sodium can also
100 patients with chronic bronchitis taking N-acetylcysteine produce small reductions in nonseasonal bronchial reactivity.
for 1224 weeks, 17 would be prevented from having an
exacerbation and 26 would note an improvement in symp- Clinical asthma
toms compared with placebo. However, a recent large multi- Comparison with placebo: In both pediatric and adult asthma,
centre Bronchitis Randomized on NAC Cost-Utility Study treatment with inhaled sodium cromoglycate has been reported
(BRONCUS) in which patients with COPD were followed to improve asthma control. A systematic review of 24 rand-
for 3 years found that N-acetylcysteine 600 mg daily was inef- omized controlled trials, however, questioned the ecacy of
fective at preventing exacerbations and rate of decline in lung sodium cromoglycate in children with asthma and concluded
function [29]. Subgroup analysis suggested that exacerba- that it was no longer justied to recommend sodium cromo-
tion rates were reduced in patients not treated with inhaled glycate as a rst-line prophylactic agent in chronic childhood
corticosteroids. asthma [37]. Some studies comparing nedocromil sodium with
placebo have demonstrated benet in both children and adults
Adverse effects with asthma. However, ecacy was not conrmed in a large
The main side eects of mucolytic drugs are gastrointestinal randomized controlled trial of 1041 children with mild-to-
in nature. moderate asthma [38]. A small number of clinical trials have
compared the two cromones in adults with asthma and in most
Recommendations studies the therapeutic eects were found to be comparable.
The GOLD guidelines do not recommend the use of muco- Comparison with inhaled steroids: The results of most
lytic drugs in the management of COPD [8]. short-to-medium term studies suggest that the improve-
ment in asthma control produced by the cromones is com-
monly slightly less than that produced by 400 g of inhaled
beclomethasone daily. A large long-term randomized
controlled trial in children aged 512 years with mild-to-
CROMONES moderate asthma found that inhaled budesonide 200 g
daily provided better control of asthma and improved
airway responsiveness when compared with placebo or
Mechanism of action nedocromil 8 mg daily [38]. Neither drug was better than
placebo in terms of improvements in lung function.
The cromones, sodium cromoglycate and nedocromil sodium, Add-on therapy: The addition of inhaled nedocromil
were rst introduced as prophylactic drugs for asthma over 40 sodium to asthmatic patients with symptoms poorly controlled
and 25 years ago respectively. The mechanism of action of the by high-dose (1000 g daily) inhaled corticosteroids produces
cromones has not been clearly established. Both drugs act as small improvements in symptoms, peak ow readings, and bron-
nonspecic chloride channel blockers in a large range of cell chodilator use [39]. Nedocromil sodium does not have a clini-
types and through this action, these compounds may reduce cally relevant oral corticosteroids sparing eect [40].

731
Asthma and COPD: Basic Mechanisms and Clinical Management

Adverse eects: Sodium cromoglycate and nedocromil


sodium are safe drugs and uncommonly cause adverse eects.
BRONCHIAL THERMOPLASTY
The main side eects reported include a distinctive bitter taste,
headache, and nausea. Bronchial thermoplasty to the airways is a new treatment
technique that involves the delivery of radio frequency energy
Place in management to the airways with the aim of reducing airway responsiveness
in asthma. Pre-clinical studies showed that bronchial thermo-
The cromones have a very limited role in the management of plasty reduces airway smooth muscle, increases airway size,
adults and children with chronic asthma. The GINA guide- and produces a long-lasting decrease in airway responsiveness
lines recommend that the cromones should be considered as to methacholine [4346]. In the canine model the reduction
an alternative to an inhaled short acting 2-agonists to pre- in airway smooth muscle is directly related to the decrease in
vent exercise-induced asthma but should not be normally airway responsiveness suggesting that the benicial eects of
used as controller therapies at step 2 because of their poor bronchial thermoplasty may be due to a decrease in airway
ecacy, but should be considered as third-line alternative smooth muscle although other mechanisms may be involved,
options to inhaled corticosteroids, leukotriene modiers, or for example altered contractility of airway smooth muscle or
oral sustained release theophylline [7]. There is no role for stiening of the airway wall [43]. In an observational study
the use of the cromones as additional therapy for patients to determine the safety of bronchial thermoplasty in sub-
already receiving inhaled or systemic corticosteroids. jects with mild-to-moderate asthma (n 16), there were no
severe adverse events in the 2-year-study period [47].
Although the study was not powered to evaluate e-
COPD cacy, bronchial thermoplasty resulted in improvements in
symptom-free days, morning peak expiratory ow (PEF)
A randomized controlled study in patients with COPD at 3 months, and a reduction in airway hyperresponsive-
reported that nedocromil sodium treatment for 10 weeks ness to methacholine that lasted for at least 2 years [47].
reduced the number of dropouts because of exacerbations In randomized controlled study, the Asthma Intervention
but had no eect on symptoms, lung function, or airway Research (AIR) Trial, the eects of bronchial thermoplasty
responsiveness to histamine and adenosine [41]. A further were evaluated in subjects with moderate or severe asthma
clinical trial of 12-weeks duration noted limited clinical (n 112) over12 months [48]. Treatment signicantly reduced
ecacy of nedocromil sodium in patients with obstructive the mean rate of mild exacerbations and use of rescue medi-
airways disease and sputum production. Nedocromil sodium cation [48] (Fig. 60.1). Bronchial thermoplasty also improved
treatment was associated with a reduction of plasma protein morning PEF, Asthma Quality of Life Questionnaire
exudation as measured in sputum sol phase suggesting some (AQLQ) scores, Asthma Control Questionnaire (ACQ)
anti-inammatory properties in COPD [42]. The GOLD scores, and symptom scores compared with the control group.
guidelines do not recommend either sodium cromoglycate A post hoc analysis suggested that the benets of treatment were
or nedocromil sodium for the treatment of COPD due to particularly marked in subjects requiring high-maintenance
inadequate data [8]. doses of inhaled corticosteroids (1000 g of beclomethasone

(A) (B)
Severe exacerbations (no./subject/wk)

0.5
Mild exacerbations (no./subject/wk)

Bronchial-thermoplasty group Control group


0.14 Bronchial-thermoplasty group Control group

0.12
0.4
0.10
0.3
0.08
*
0.2 0.06
*
0.04
0.1
0.02

0.0 0.00
Baseline 3 months 6 months 12 months Baseline 3 months 6 months 12 months
After treatment After Treatment

FIG. 60.1 Rates of mild and severe exacerbations per subject per week. Mean values are shown for all subjects receiving inhaled corticosteroids alone for
whom data were available at the given time points. Asterisks indicate a statistically significant difference in the mean change from baseline between the two
groups, and I bars represent the standard errors. p 0.03 for the comparison between subjects in the two groups treated with inhaled corticosteroids alone
at 3 months and at 12 months. Reproduced with permission from Ref. [48].

732
Other Therapies 60
equivalent daily) [48]. The Research in Severe Asthma (RISA) [5356]. Weight reduction has been shown to improve lung
Trial was designed to determine the safety and ecacy of bron- function, symptoms, and health status in obese people with
chial thermoplasty in subjects with symptomatic, severe asthma asthma [57, 58] (Fig. 60.2).
(n 32) [49]. Bronchial thermoplasty was associated with
a short-term increase in asthma-related morbidity including
increased hospitalizations for asthma. At 22 weeks, bronchial COPD
thermoplasty subjects had signicant improvements compared
to control subjects in rescue medication use, pre-bronchodila- Obesity is also a common feature of COPD and is asso-
tor FEV1% predicted and ACQ scores. Improvements in res- ciated with a poor prognosis, independent of FEV1. The
cue medication use and ACQ scores remained signicantly inuence of weight reduction on the morbidity and mortal-
improved compared to the control group at 52 weeks. The ity from COPD has not been systematically studied.
ecacy and safety of bronchial thermoplasty is currently being
assessed in a placebo-controlled trial involving the use of sham
bronchial thermoplasty.
Taken together, these nding suggest the potential COMPLEMENTARY THERAPIES
clinical benets of targeting airway smooth muscle in the
treatment of asthma [50]. Future studies need to determine Complementary and alternative medicines are frequently
the risk benet ratio of bronchial thermoplasty in dierent used by adults and children with asthma [59, 60]. The
severities of asthma, to identify factors that predict a ben-
ecial clinical response to bronchial thermoplasty and to Treatment group
investigate whether other less invasive techniques can be Peak expiratory flow Control group
used to target airway smooth muscle. 95

90

% of predicted 85
MANAGEMENT OF MALNUTRITION
80

75
COPD
70
Malnutrition (dened as 90% of ideal body weight) is found Forced expiratory volume in one second
in 25% of outpatients with COPD and in over 50% of patients 90
admitted to hospital because of COPD. The mechanisms
of weight loss are unclear, although changes in energy bal- 85
ance and systemic inammation are likely to play a part. The
% of predicted

80
mortality rate is increased in malnourished patients with
COPD [51]. Malnourished patients with COPD exhibit 75
reduced exercise capacity and respiratory muscle function. No 70
simple recommendation can be given regarding the best test
for nutritional assessment. 65
These patients are often unable to regain weight despite
60
receiving nutritional support. High-carbohydrate diets should
be avoided to prevent excess carbon dioxide production. Forced vital capacity
A Cochrane review of fourteen nine randomized controlled tri- 95
als, of which two were double blind, concluded that nutritional 90
support (caloric supplementation for at least 2 weeks) had no
% of predicted

85
eect on improving anthropometric measures, lung function,
or exercise capacity among patients with stable COPD [52]. 80
The GOLD guidelines recommend that increased calorie 75
intake is best undertaken with an exercise program [8]. 70
65
60
MANAGEMENT OF OBESITY Baseline End of
dieting
End of weight
reduction
6 months 1 year

period programme

Asthma FIG. 60.2 Obesity, weight reduction and asthma: mean morning pre-
medication values for PEF, FEV1, and FVC (% predicted) at different stages
An elevated body mass index has been associated with sev- during study. Vertical bars show standard errors of the mean. Changes
eral features of asthma including an increased prevalence and from baseline show significant ( p  0.05) difference between groups.
severity as well as an enhance airway inammatory response Reproduced with permission from Ref. [57].

733
Asthma and COPD: Basic Mechanisms and Clinical Management

therapies most frequently used are breathing techniques, of overall symptom intensity showed improvements in favor
homeopathy, herbal medicines, and acupuncture [59, 60]. of homeopathic immunotherapy within 1 week of starting
Complementary therapy use often reects patients and par- treatment which persisted through the treatment period.
ents wish to achieve greater self management of their con- There were no signicant changes in measurement of base-
dition [61]. Thirty percent of the general populations in the line lung function and bronchial reactivity. A systematic
United States currently use complementary medicine and review of placebo-controlled trials of homeopathy have
two-thirds had used at least one complementary and alter- concluded that there is insucient evidence of ecacy [70].
native therapy during their lifetime [62].
Acupuncture
Acupuncture involves the insertion of needles at specic
Asthma points of the body. The technique is based on Chinese theo-
ries of the bodies balance of energies. The eects of acupunc-
Breathing techniques and inspiratory muscle ture were assessed in a randomized, double-blind controlled,
training cross-over study in 22 patients with asthma [71]. There was a
Breathing techniques and inspiratory muscle training have statistically signicant improvement in assessments of quality
been used to treat asthma. The Buteyko breathing technique of life and reductions in -agonist use after both the active
is based on the hypothesis that all patients with asthma and sham interventions, but no change in PEF recordings.
hyperventilate at rest and that recties this problem, by A Cochrane review of acupuncture for asthma concluded
reducing the rate of breathing, results in an improvement in that there was insucient evidence to make any recommen-
asthma control. The Buteyko breathing technique was com- dations about its use, but highlighted the need for well-con-
pared with a placebo breathing technique in 39 adult asth- trolled studies to assess its eectiveness in asthma [72].
matic patients over a 3-month period [63]. At the end of
the trial the Buteyko breathing technique group compared Herbal medicines
with the placebo breathing technique showed reduction A large range of herbal medicines including traditional
in median -agonist usage. Unfortunately the follow-up Chinese medicine are used for the treatment of asthma. The
arrangements were not identical between the two groups ecacy and safety of some of these remedies are now being
and there was the possibility of greater patient contact in the assessed in controlled trials [73].
Buteyko-treated group. Measurements of lung function and
quality of life were unaltered in both groups. A larger rand- Yoga
omized clinical trial of the Buteyko technique for 6 months Yoga is an ancient Hindu discipline that promotes increased
reported improved symptoms and reduced bronchodila- mental and physical control of the body. One of the eight
tor use, but no change in bronchial responsiveness or lung steps of yoga, pranayama, deals with control of breathing to
function in patients with asthma [64]. A systematic review promote relaxation and improve tness. The eects of two
of breathing exercises for asthma found that although pranayama yoga breathing exercises were assessed in a rand-
there were trends of improvement in a number of out- omized, controlled, cross-over trial in 18 patients with mild
comes no rm conclusion can be drawn concerning the use asthma [74]. There was a statistically signicant reduction
of breathing exercises for asthma in clinical practice [65]. in bronchial reactivity to histamine after the active inter-
A Cochrane review concluded that there was insucient vention compared with placebo, but no change in baseline
evidence to suggest that inspiratory muscle training pro- lung function tests, symptom scores, or -agonist use.
vides any clinical benet to patients with asthma [66].
A recent clinical study reported that an integrated breathing
and relaxation technique known as the Papworth Method Safety of complementary medicine
reduced respiratory symptoms, dysfunctional breathing, and
adverse mood compared with usual care [67]. The safety of complementary therapies is rarely assessed.
Although patients often consider these treatments as harmless,
Manual therapies this may not always be the case. The health of a patient starting
a complementary therapy may be put at risk for several reasons.
Manual therapies such as massage, chiropractic, and oste-
The patient may abruptly stop their conventional medicines for
opathic techniques are used to treat asthma, but there is
asthma. Some herbal remedies can cause severe toxic eects
insucient evidence of ecacy [68].
due to hepatotoxicity, heavy metal poisoning, or to microor-
Homeopathy ganisms. Herbal medicines may contain conventional phar-
maceutical drugs, for example, corticosteroids or nonsteroidal
Homeopathy involves identifying factors that precipitate anti-inammatory drugs. Finally, it is known that acupuncture
asthma in an individual and then prescribing a specic can cause pneumothoraces and transmit hepatitis.
homeopathic remedy for that person. In some cases, the
remedy may include very small quantities of the allergen to
which the patient is allergic, for example. house dust mite. References
A small randomized controlled parallel group study in 28
patients with allergic asthma compared oral homeopathic 1. Poole P, Chacko E, Wood-Baker R, Cates C. Inuenza vaccine for
immunotherapy to their principal allergen or identical pla- patients with chronic obstructive pulmonary disease. Cochrane Database
cebo for a 4-week period [69]. All patients continued their Syst Rev(1), 2006, Art. No.: CD002733. DOI: 10.1002/14651858.
unaltered conventional therapy. A daily visual analog scale CD002733.pub2.

734
Other Therapies 60
2. Cates C, Jeerson T, Bara A, Rowe B. Vaccines for preventing inu- in patients with mild asthma. Am J Respir Crit Care Med 156(2): 42530,
enza in people with asthma. Cochrane Database Syst Rev(4), 2003, 1997.
Art. No.: CD000364. DOI: 10.1002/14651858.CD000364.pub2. 24. Kirkham P, Rahman I. Oxidative stress in asthma and COPD:
3. Fleming D, Crovari P, Wahn U, Klemola T, Schlesinger Y, Antioxidants as a therapeutic strategy. Pharmacol Therap 111(2):
Langussis A et al. Comparison of the ecacy and safety of live attenu- 47694, 2006.
ated cold-adapted inuenza vaccine, trivalent, with trivalent inactivated 25. Rautalahti M, Virtamo J, Haukka J, Heinonen O, Sundvall J, Albanes
inuenza virus vaccine in children and adolescents with asthma. Pediatr D et al. The eect of alpha-tocopherol and beta-carotene supplementa-
Infect Dis J 10: 86069, 2006. tion on COPD symptoms. Am J Respir Crit Care Med 156(5): 144752,
4. Glezen WP. Asthma, inuenza, and vaccination. J Allergy Clin Immunol 1997.
118(6): 1199206, 2006. 26. Aitio M-L. N-Acetylcysteine-passe-partout or much ado about noth-
5. The American Lung Association Asthma Clinical Research Centers. ing?. Br J Clin Pharmacol 61(1): 515, 2006.
The safety of inactivated inuenza vaccine in adults and children with 27. Poole P, Black P. Mucolytic agents for chronic bronchitis or chronic
asthma. N Engl J Med 345(21): 152936, 2001. obstructive pulmonary disease. Cochrane Database Syst Rev, 2006, DOI:
6. Fiore A, Shay D, Haber P, Iskander J, Uyeki T, Mootrey G et al. 10.1002/14651858.CD001287.pub2.
Prevention and control of inuenza: Recommendations of the advisory 28. Stey C, Steurer J, Bachmann S, Medici TC, Tramer MR. The eect of
committee on immunization practices (ACIP). Morb Mortal Wkly Rep oral N-acetylcysteine in chronic bronchitis: A quantitative systematic
56(RR06): 154, 2007. review. Eur Respir J 16(2): 25362, 2000.
7. GINA Report, Global Strategy for Asthma Management and 29. Decramer M, Rutten-van Molken M, Dekhuijzen PNR, Troosters T,
Prevention. 2006 [cited December 12, 2007]; Available from: http:// van Herwaarden C, Pellegrino R et al. Eects of N-acetylcysteine
www.ginasthma.com/Guideline on outcomes in chronic obstructive pulmonary disease (Bronchitis
8. Global Initiative for Chronic Obstructive Lung Diseases. Workshop Randomized on NAC Cost-Utility Study, BRONCUS): A randomised
Report: Guidelines global strategy for the diagnosis, management and placebo-controlled trial. Lancet 365(9470): 155260, 2005.
prevention of chronic obstructive pulmonary disease. 2006 [cited May 30. Norris A, Alton E. Chloride transport and the action of sodium cro-
25, 2007]; Available from: http://www.goldcopd.com moglycate and nedocromil sodium in asthma. Clin Exp Allergy 26(3):
9. Johnston S, Ferrero F, Garcia M, Dutkowski R. Oral oseltamivir 25053, 1996.
improves pulmonary function and reduces exacerbation frequency for 31. Thomson N. Nedocromil sodium: An overview. Respir Med 83(4):
inuenza-infected children with asthma. Pediatr Infect Dis J 24(3): 26976, 1989.
22532, 2005. 32. Corin RE. Nedocromil sodium: A review of the evidence for a dual
10. Nichol KL, Baken L, Wuorenma J, Nelson A. The health and economic mechanism of action. Clin Exp Allergy 30(4): 46168, 2000.
benets associated with pneumococcal vaccination of elderly persons 33. Diaz P, Galleguillos FR, Gonzalez MC, Pantin CFA, Kay AB.
with chronic lung disease. Arch Intern Med 159(20): 243742, 1999. Bronchoalveolar lavage in asthma: The eect of disodium cromoglycate
11. Granger R, Walters J, Poole P, Lasserson T, Mangtani P, Cates C et al. (cromolyn) on leukocyte counts, immunoglobulins, and complement.
Injectable vaccines for preventing pneumococcal infection in patients with J Allergy Clin Immunol 74(1): 4148, 1984.
chronic obstructive pulmonary disease. Cochrane Database Syst Rev (4), 34. Hoshino M, Nakamura Y. The eect of inhaled sodium cromoglycate
2006, Art. No.: CD001390. DOI: 10.1002/14651858.CD001390 pub2 on cellular inltration into the bronchial mucosa and the expression
12. Sheikh A, Alves B, Dhami S. Pneumococcal vaccine for asthma. of adhesion molecules in asthmatics. J Allergy Clin Immunol 10(4):
Cochrane Database Syst Rev (3), 2001, CD002165. 85865, 1997.
13. Prevention of pneumococcal disease: Recommendations of the 35. Manolitsas N, Wang J, Devalia J, Trigg C, McAulay A, Davies RJ.
Advisory Committee on Immunization Practices (ACIP). Morb Mortal Regular albuterol, nedocromil sodium, and bronchial inammation in
Wkly Rep 46: 124, 1997. asthma. Am J Respir Crit Care Med 151(6): 192530, 1995.
14. Foxwell A, Cripps A, Dear K. Haemophilus inuenzae oral whole cell 36. Kelly KD, Spooner CH, Rowe BH. Nedocromil sodium versus sodium
vaccination for preventing acute exacerbations of chronic bronchi- cromoglycate in treatment of exercise-induced bronchoconstriction:
tis. Cochrane Database Syst Rev (4), 2006, DOI: 10.1002/14651858. A systematic review. Eur Respir J 17(1): 3945, 2001.
CD001958.pub2. 37. Tasche MJA, Uijen JHJM, Bernsen RMD, de Jongste JC, van der
15. Collet JP, Shapiro S, Ernst P, Renzi P, Ducruet T, Robinson A et al. Wouden JC. Inhaled disodium cromoglycate (DSCG) as maintenance
Eects of an immunostimulating agent on acute exacerbations and therapy in children with asthma: A systematic review. Thorax 55(11):
hospitalizations in patients with chronic obstructive pulmonary disease. 91320, 2000.
Am J Respir Crit Care Med 156(6): 171924, 1997. 38. The Childhood Asthma Management Program Research Group.
16. Steurer-Stey C, Bachmann LM, Steurer J, Tramer MR. Oral puried Long-term eects of budesonide or nedocromil in children with
bacterial extracts in chronic bronchitis and COPD: Systematic review. asthma. N Engl J Med 343(15): 150463, 2000.
Chest 126(5): 164555, 2004. 39. Svendsen U, Jrgensen H. Inhaled nedocromil sodium as additional
17. Rahman I, MacNee W. Oxidative stress and regulation of glutathione treatment to high dose inhaled corticosteroids in the management of
in lung inammation. Eur Respir J 16(3): 53454, 2000. bronchial asthma. Eur Respir J 4(8): 99299, 1991.
18. Devereux G, Seaton A. Diet as a risk factor for atopy and asthma. 40. Goldin J, Bateman E. Does nedocromil sodium have a steroid sparing
J Allergy Clin Immunol 115(6): 110917, 2005. eect in adult asthmatic patients requiring maintenance oral corticos-
19. Patel BD, Welch AA, Bingham SA, Luben RN, Day NE, Khaw KT et al. teroids? Thorax 43(12): 98286, 1988.
Dietary antioxidants and asthma in adults. Thorax 61(5): 38893, 2006. 41. de Jong J, Postma D, van der Mark T, Koter G. Eects of nedocromil
20. Pearson PJK, Lewis SA, Britton J, Fogarty A. Vitamin E supplements sodium in the treatment of non-allergic subjects with chronic obstruc-
in asthma: A parallel group randomised placebo controlled trial. Thorax tive pulmonary disease. Thorax 49(10): 102224, 1994.
59(8): 65256, 2004. 42. Schoonbrood DF, Out TA, Hart AA, Habets FJ, Roos CM,
21. Shaheen SO, Newson RB, Rayman MP, Wong APL, Tumilty MK, Jansen HM. Nedocromil sodium in obstructive airways disease: Eect
Phillips JM et al. Randomised, double blind, placebo-controlled trial of on symptoms and plasma protein leakage in sputum. Eur Respir J 10(7):
selenium supplementation in adult asthma. Thorax 62(6): 48390, 2007. 15006, 1997.
22. Ram F, Rowe B, Kaur B. Vitamin C supplementation for asthma. Cochrane 43. Danek CJ, Lombard CM, Dungworth DL, Cox PG, Miller JD,
Database Syst Rev, 2004, DOI: 10.1002/14651858.CD000993.pub2. Biggs MJ et al. Reduction in airway hyperresponsiveness to metha-
23. Marrades Ramon M, Roca J, Barbera Joan A, de Jover L, Macnee W, choline by the application of RF energy in dogs. J Appl Physiol 97(5):
Rodriguez-Roisin R. Nebulized glutathione induces bronchoconstriction 194653, 2004.

735
Asthma and COPD: Basic Mechanisms and Clinical Management

44. Miller JD, Cox G, Vincic L, Lombard CM, Loomas BE, Danek CJ. 59. Ernst E. Complementary therapies for asthma: What patients use.
A prospective feasibility study of bronchial thermoplasty in the human J Asthma 38(8): 66771, 1998.
airway. Chest 127(6): 19992006, 2005. 60. Slader CA, Reddel HK, Jenkins CR, Armour CL, Bosnic-Anticevich SZ.
45. Brown RH, Wizeman W, Danek C, Mitzner W. In vivo evaluation of Complementary and alternative medicine use in asthma: Who is using
the eectiveness of bronchial thermoplasty with computed tomogra- what? Respirology 11(4): 37387, 2006.
phy. J Appl Physiol 98(5): 16036, 2005. 61. Shaw A, Thompson E, Sharp D. Complementary therapy use by
46. Brown RH, Wizeman W, Danek C, Mitzner W. Eect of patients and parents of children with asthma and the implications for
bronchial thermoplasty on airway distensibility. Eur Respir J 26(2): NHS care: A qualitative study. BMC Health Serv Res 6: 76, 2006.
27782, 2005. 62. Kessler RC, Davis RB, Foster DF, Van Rompay MI, Walters EE,
47. Cox G, Miller JD, McWilliams A, FitzGerald JM, Lam S. Bronchial ther- Wilkey SA et al. Long-term trends in the use of complementary and
moplasty for asthma. Am J Respir Crit Care Med 173(9): 96569, 2006. alternative medical therapies in the United States. Ann Intern Med
48. Cox G, Thomson NC, Rubin AS, Niven RM, Corris PA, Siersted HC 135(4): 26268, 2001.
et al. Asthma control during the year after bronchial thermoplasty. 63. Bowler SDGA, Mitchell CA. Buteyko breathing techniques in asthma:
N Engl J Med 356(13): 132737, 2007. A blinded randomised controlled trial. Med J 169: 57578, 1998.
49. Pavord ID, Cox G, Thomson NC, Rubin AS, Corris PA, Niven RM 64. Cooper S, Oborne J, Newton S, Harrison V, Thompson Coon J, Lewis S
et al. Safety and ecacy of bronchial thermoplasty in symptomatic, et al. Eect of two breathing exercises (Buteyko and pranayama) in
severe asthma. Am J Respir Crit Care Med 176(12): 118591, 2007. asthma: A randomised controlled trial. Thorax 58(8): 67479, 2003.
50. Solway J, Irvin CG. Airway smooth muscle as a target for asthma ther- 65. Holloway E, Ram F. Breathing exercises for asthma. Cochrane Database
apy. N Engl J Med 356(13): 136769, 2007. Syst Rev(1), 2004, DOI: 10.1002/14651858.CD001277.pub2.
51. Landbo C, Prescott E, Lange P, Vestbo J, Almda lT. Prognostic value of 66. Ram F, Wellington S, Barnes NC. Inspiratory muscle training for
nutritional status in chronic obstructive pulmonary disease. Am J Respir asthma. Cochrane Database Syst Rev(3), 2003, DOI: 10.1002/14651858.
Crit Care Med 160(6): 185661, 1999. CD003792.
52. Ferreira I, Brooks D, Lacasse Y, Goldstein R, White J. Nutritional 67. Holloway EA, West RJ. Integrated breathing and relaxation training
supplementation for stable chronic obstructive pulmonary disease. (the Papworth method) for adults with asthma in primary care: A ran-
Cochrane Database Syst Rev(2), 2005, 10.1002/14651858.CD000998. domised controlled trial. Thorax 62(12): 103942, 2007.
pub2. 68. Hondras M, Linde K, Jones A. Manual therapy for asthma. Cochrane
53. Beuther DA, Weiss ST, Sutherland ER. Obesity and asthma. Am J Database Syst Rev(2), 2005. , DOI: 10.1002/14651858.CD001002 pub2
Respir Crit Care Med 174(2): 11219, 2006. 69. Reilly D, Taylor M, Beattie N, Campbell J, McSharry C, Aitchison T
54. Peters-Golden M, Swern A, Bird SS, Hustad CM, Grant E, et al. Is evidence for homoeopathy reproducible? Lancet 344: 16016,
Edelman JM. Inuence of body mass index on the response to asthma 1994.
controller agents. Eur Respir J 27(3): 495503, 2006. 70. Altunc U, Pittler MH, Ernst E. Homeopathy for childhood and ado-
55. Shore S. Obesity and asthma: Implications for treatment. Curr Opin lescence ailments: Systematic review of randomized clinical trials.
Pulm Med 13(1): 5662, 2007. Mayo Clinic Proceed 82(1): 6975, 2007.
56. Taylor B, Mannino D, Brown C, Crocker D, Twum-Baah N, 71. Biernacki W, Peake MD. Acupuncture in treatment of stable asthma.
Holguin F. Body mass index and asthma severity in the national Respir Med 92(9): 114345, 1998.
asthma survey. Thorax 63(1): 1420, 2008. 72. McCarney R, Brinkhaus B, Lasserson T, Linde K. Acupuncture for
57. Stenius-Aarniala B, Poussa T, Kvarnstrom J, Gronlund E-L, chronic asthma. Cochrane Database Syst Rev(3), 2003, CD000008. DOI:
Ylikahri M, Mustajoki P. Immediate and long term eects of weight 10.1002/14651858.CD000008.pub2.
reduction in obese people with asthma: Randomised controlled study. 73. Li X-M. Traditional Chinese herbal remedies for asthma and food
BMJ 320(7238): 82732, 2000. allergy. J Allergy Clin Immunol 120(1): 2531, 2007.
58. Aaron SD, Fergusson D, Dent R, Chen Y, Vandemheen KL, Dales RE. 74. Singh V, Wisniewski A, Britton J, Tatterseld A. Eect of yoga breath-
Eect of weight reduction on respiratory function and airway reactivity ing exercises (pranayama) on airway reactivity in subjects with asthma.
in obese women. Chest 125(6): 204652, 2004. Lancet 335: 138183, 1990.

736
Future Therapies
61CHAPTER

treatment in many patients. Other treatments,


INTRODUCTION such as theophylline, anticholinergics and
Peter J. Barnes
anti-leukotrienes are less eective and are National Heart and Lung Institute
As indicated in previous chapters, asthma and largely used as add-on therapies. (NHLI), Clinical Studies Unit,
chronic obstructive pulmonary disease (COPD) There is still a major problem with poor Imperial College, London, UK
are dierent diseases that involve dierent cells, compliance in the long-term management of
mediators and inammatory eects so that dif- asthma, particularly as symptoms come under
ferent treatments are needed. However, it is control with eective therapies [5]. It is likely
increasingly recognized that there are any simi- that a once daily tablet or even an infrequent
larities in the inammatory process between injection may give improved compliance.
severe asthma and COPD [1]. Many new classes However, oral therapy is associated with a
of drugs are now in development for asthma and much greater risk of systemic side eects
COPD and there are several new drugs that are and therefore needs to be specic for the
being developed for both diseases [24]. abnormality in asthma or COPD.
Patients with severe asthma are often not
controlled on maximal doses of inhaled
THE NEED FOR NEW TREATMENTS therapies or may have serious side eects
from therapy, especially oral corticosteroids.
These patients are relatively resistant to the
Asthma anti-inammatory actions of corticosteroids
and require some other class of therapy to
Current asthma therapy is highly eective and control the asthmatic process. Corticosteroid
majority of patients can be well controlled with resistance is also a major barrier to eective
inhaled corticosteroids and short- and long- therapy in COPD.
acting 2-agonists. These treatments are not None of the existing treatments for asthma
only eective, but safe and relatively inexpen- is disease modifying, which means that
sive. This poses a challenge for the development the disease recurs as soon as treatment is
of new treatments, since they will need to be discontinued.
safer or more eective than existing treatments,
or oer some other advantage in long-term dis- None of the existing treatments is curative,
ease management. However, there are several although it is possible that therapies which
problems with existing therapies. prevent the immune aberration of allergy may
have the prospects for a cure in the future.
Existing therapies have side eects as they
are non-specic. Inhaled 2-agonists may
have side eects and there is some evidence COPD
for the development of tolerance, especially
to their bronchoprotective eects. Inhaled In sharp contrast to asthma there are few
corticosteroids may also have local and eective therapies in COPD, despite the fact
systemic side eect at high doses and there that it is a common disease that is increasing
is still a fear of using long-term steroid worldwide [6].
737
Asthma and COPD: Basic Mechanisms and Clinical Management

The relative neglect of COPD by pharmaceutical which may exclude them from clinical trials of new thera-
companies is probably as a result of several factors: pies. There is little information about surrogate markers, for
example biomarkers in blood, sputum or breath, to monitor
COPD is regarded as largely irreversible and is treated as the short-term ecacy and predict the long-term potential
poorly responsive asthma. of new treatments [9]. Finally, it is dicult to accurately
COPD is self-inicted and therefore does not deserve measure small airway function in patients with COPD so
substantial investment. there is a need to develop better tests of small airway func-
Animal models do not mimic many of the key aspects of tion that are not aected by emphysema or abnormalities of
the disease, such as small airway disease and exacerbations. large airway function [10].
Although asthma and COPD involve dierent pat-
Relatively little is understood about the cell and terns of inammation, there is increasing evidence that
molecular biology of this disease or even about the patients with severe asthma have inammatory features that
relative role of small airways disease and parenchymal are very similar to those seen in COPD [1]. This suggests
destruction. that drugs that are eective in suppressing the inammation
None of the treatments currently available prevents of COPD might also be eective in treating severe asthma.
the progression of the disease, and yet the disease is asso-
ciated with an active inammatory process that results in
progressive obstruction of small airways and destruction of
lung parenchyma. Increased understanding of COPD will DEVELOPMENT OF NEW THERAPIES
identify novel targets for future therapy (Fig. 61.1) [7].
There are also uncertainties about how to test drugs
Several strategies have been adopted in the search for new
for COPD, which may require long-term studies (3 years)
therapies:
in relatively large numbers of patients at an enormous
cost. For example, a recent study looking at the eects of Improvement of an existing class of drug: This is well
drug intervention on mortality costs several hundred mil- exemplied by the increased duration of 2-agonists with
lion dollars [8]. Many patients with COPD may have salmeterol and formoterol and of anticholinergics
co-morbidities, such as ischaemic heart disease and diabetes, with tiotropium bromide, and with the improved

Smoking cessation
Cigarette smoke Nicotine antagonists
(and other irritants) and vaccination
CB1 antagonists

Epithelial Alveolar macrophage


cells
TGF-
Anti-TGF-
CTGF Chemokine and
Chemotactic factors mediator antagonists

CD8 Anti-inflammatory
lymphocyte Neutrophil Monocyte treatments
Fibroblast
PDE4, IKK-2, p38 MAPK,
PI3K- inhibitors
PPAR-agonists

Proteases Antiproteases
NE inhibitors
MMP-9 inhibitors
Fibrosis Alveolar wall destruction Mucus hypersecretion
(COB) (Emphysema) (Chronic bronchitis)

Retinoic acid Mucoregulators


Stem cells EGFR antagonists

FIG. 61.1 Targets for COPD therapy based on current understanding of the inflammatory mechanisms. Cigarette smoke (and other irritants) activate
macrophages in the respiratory tract that release multiple chemotactic factors that attract neutrophils, monocytes, and T-lymphocytes (particularly CD8
cells). Several cells also release proteases, such as neutrophil elastase (NE) and matrix metalloproteinase-9 (MMP-9) which the break down connective tissue
in the lung parenchyma (emphysema) and also stimulate mucus hypersecretion (chronic bronchitis). CD8 may also be involved in alveolar wall destruction.
This inflammatory process may be inhibited at several stages (shown in the boxes). PDE: phosphodiesterase; IKK: inhibitor of nuclear factor-B kinase; MAPK:
mitogen-activated protein kinase; PI3K: phosphoinositide-3-kinase; PPAR: peroxisome proliferator activated receptor; COB: chronic obstructive bronchitis;
TGF: transforming growth factor; CB: cannabinoid; EGFR: epithelial growth factor receptor.

738
Future Therapies 61
pharmacokinetic of the inhaled corticosteroids Indacaterol is an eective once daily bronchodilator in
uticasone propionate, mometasone, ciclesonide asthma and appears to be well tolerated with no evidence
and budesonide, with increased rst-pass (hepatic) for the development of tolerance [13]. Indacaterol is a very
metabolism and therefore reduced systemic absorption. eective dilator of small human airways measured by vid-
Development of novel therapies through better understanding eomicroscopy in a precision-cut lung slice preparation, indi-
of the disease process: Examples are anti-interleukin(IL)-5 cating that it will be useful in severe asthma and COPD
as a potential treatment of asthma and phosphodiesterase where small airway obstruction is a problem [14].
(PDE)-4 inhibitors as an anti-inammatory therapy
for COPD.
Serendipitous observations, often made in other therapeutic
New anticholinergics
areas: Examples are tumor necrosis factor- (TNF-
The once daily inhaled anticholinergic tiotropium bromide
) antagonists for airway diseases, derived from
has been an important advancement in therapy of COPD
observations in other chronic inammatory diseases.
and several other long-acting inhaled muscarinic antago-
Identication of novel targets through gene and protein nists (LAMA), such as aclidinium bromide and glycopyrro-
proling: This approach will be increasingly used to late are now in development [15, 16]. Combination inhalers
identify the abnormal expression of genes (molecular with a long-acting 2-agonist (LABA) with a LAMA are
genomics) and proteins (proteomics) from diseased also in development as there is an additive eect between
cells, and through identication of single nucleotide these two bronchodilator classes [17]. Single molecules that
polymorphisms (SNPs) that contribute to the disease link a muscarinic antagonist to a 2-agonist (MABA) are
process [11]. So far there have been no new drugs also in development.
developed from this approach in any area of medicine.

New classes of bronchodilator


It has proved dicult to discover novel classes of bron-
NEW BRONCHODILATORS chodilator drug. Potassium channel openers, while eective
in human airways in vitro, were not eective in asthma as
they were more potent as vasodilators and this limited the
Bronchodilators act predominantly by relaxation of air- dose that could be administered. There has been interest in
way smooth muscle cells, although additional eects, such developing drugs that inhibit the contractile machinery in
as inhibition of bronchoconstrictor mediator and neuro- airway smooth muscle, including Rho kinase inhibitors,
transmitter release may contribute. 2-Agonists are by far inhibitors of myosin light chain kinase and direct smooth
the most eective bronchodilators in asthma and act as muscle myosin inhibitors. As these agents also cause
functional antagonists, blocking the eect of all bronchoc- vasodilatation it will be necessary to administer them by
onstrictors. The increased bronchodilator duration of long- inhalation.
acting inhaled 2-agonists have been an important advance
and there are now several once daily inhaled 2-agonists
in development. By contrast, in COPD anticholinergics
appear to be the most eective bronchodilators and there
has been a search for selective anticholinergics and drugs MEDIATOR ANTAGONISTS
with a longer duration. Novel classes of bronchodilator have
also been developed (Table 61.1.)
Blocking the receptors or synthesis of inammatory media-
tors is a logical approach to the development of new treat-
Ultra-LABAs ments for asthma and COPD. Several mediator antagonists
have now been tested in asthma and COPD (see Chapter
Several once daily inhaled 2-agonists, such as indacaterol 52). However, in both diseases many dierent media-
and carmoterol, are now in clinical development [12]. tors are involved and therefore blocking a single mediator
may not be very eective, unless it plays a key role in the
TABLE 61.1 New bronchodilators. disease process [18, 19]. Several specic mediator antago-
nists have been found to be ineective in asthma, including
Drug class Examples antagonists/inhibitors of histamine, thromboxane, platelet-
activating factor, bradykinin and tachykinins. There is now
Ultra-long acting 2-agonists Indacaterol, carmoterol increasing evidence that this approach is equally ineective
in COPD, with no clinical evidence of benet for inhibi-
Long-acting muscarinic antagonists Glycopyrrolate, aclidinium
tors of TNF-, IL-8 or leukotriene B4. However, it is pos-
Combination inhalers LABA  LAMA, MABA sible that there may be an upstream mediator that is playing
a disproportionate role in the pathogenesis of asthma and
Rho kinase inhibitors In development COPD so that targeting this mediator may give greater
Myosin light chain kinase inhibitors In development
than expected clinical benets, as is the case for antihis-
tamines in rhinitis and TNF- inhibitors in rheumatoid

739
Asthma and COPD: Basic Mechanisms and Clinical Management

arthritis. In asthma there is hope for blocking IL-13 as this corticosteroid resistance and also reduce inammation.
cytokine appears to mimic many of the pathophysiologi- Unfortunately currently available antioxidants based on
cal features of asthma. A mutated protein that blocks the glutathione are relatively weak and are inactivated by oxida-
common receptor for IL-4 and IL-13 pitrakinra has shown tive stress, so new more potent and stable antioxidants are
some inhibitory eects on the late response to allergen needed, such as superoxide dismutase mimics and NADPH
challenge in patients with mild asthma [20]. The cytokine oxidase inhibitors [28].
thymic stomal lymphoietin (TSLP), which is secreted by
epithelial cells and mast cells in asthmatic patients, appears
to play a critical role in the programing of dendritic cells to
orchestrate Th2 cells and allergic inammation and appears PROTEASE INHIBITORS
to be a promising target for inhibition in asthma [21].
Chemokines play a key role in attracting inam-
matory cells into the lung and act on G-protein-coupled Proteases are involved in the pathophysiology of asthma
receptors expressed on these cells. Small molecule inhibi- and COPD and are therefore a logical target for inhibi-
tors of chemokines receptors are a promising approach in tion. However, it has proved very dicult to nd drugs that
asthma and COPD [22, 23]. Front-runners for asthma eectively block specic proteases without side eects.
are CCR3 expressed predominantly on eosinophils and
CCR4 expressed on Th2 cells and for COPD are CXCR2 Tryptase inhibitors
expressed on neutrophils and monocytes and CXCR3
expressed on T-lymphocytes. Chemokine antagonists have Mast cell tryptase has several eects on airways, including
the advantage of oral administration but so far there have increasing responsiveness of airway smooth muscle to con-
been unexpected toxicological problems with many of the strictors, increasing plasma exudation, potentiating eosi-
small molecule antagonists developed. A dual CXCR1/2 nophil recruitment, and stimulating broblast proliferation
antagonist given orally has been shown to reduce neutrophil [29]. Some of these eects are mediated by activation of the
and macrophage inux into sputum after endotoxin chal- proteinase-activated receptor PAR2. A tryptase inhibitor
lenge in normal patients after oral administration [24]. A APC366 was eective in a sheep model of allergen-induced
CCR5 antagonist maraviroc has been approved for use in asthma [30], but was only poorly eective in asthmatic
HIV/AIDS but may also be useful in COPD and these patients in a preliminary study [31] and ineective in aller-
receptors are expressed in T-cells and their agonist CCL5 gic inammation [32]. More potent tryptase inhibitors and
(RANTES) is increased in COPD sputum [25] (Fig. 61.2). PAR2 antagonists are in development.

Anti-oxidants Neutrophil elastase inhibitors


Oxidative stress is important in severe asthma and Neutrophil elastase (NE), a neutral serine protease, is a
COPD, particularly during exacerbations [26]. Oxidative major constituent of lung elastolytic activity. In addition it
stress activates the proinammatory transcription factors potently stimulates mucus secretion and induces CXCL8
NF-B and activator protein-1 (AP-1), resulting in release from epithelial cells and may therefore perpetu-
enhanced inammation. Oxidative stress is increased in ate the inammatory state. This has lead to a search for
patients with COPD, particularly during exacerbations, NE inhibitors. Peptide NE inhibitors, such as ICI 200355,
and reactive oxygen species contribute to its pathophysiol- and non-peptide inhibitors, such as ONO-5046, have been
ogy. Oxidative stress reduces steroid responsiveness via a developed for use in COPD, but so far all of these drugs
reduction in histone deacetylase-2 (HDAC2) activity and have been withdrawn due to toxicological problems or side
expression [27]. This suggests that antioxidants may reverse eects. The NE inhibitor MR889 administered for 4 weeks

CXCL1, CXCL5, CXCL8 CCL2 CXCL9, CXCL10, CXCL11 CCL3, CCL5

CXCR2 CCR2 CXCR3 CCR5 FIG. 61.2 Several chemokines and


chemokine receptors are involved in the
inflammation of COPD. Chemokines released
from epithelial cells and macrophages in the
Neutrophil Monocyte Monocyte T-cell Tc1, Th1 cells Tc1, Th1 cells lung recruit inflammatory cells (Tc1 CD8
T-lymphocytes, neutrophils, and monocytes)
CXCR2 antags from the circulation. Small molecule
CCR2 antags CXCR3 antags CCR5 antags
e.g. SB332335 chemokine receptor antagonists are now in
e.g. RS504393 e.g. NBI74300 e.g. maraviroc
ADZ8309 development (shown in boxes).

740
Future Therapies 61
showed no overall eect on plasma elastic-derived peptides New corticosteroids
or urinary desmosine (markers of elastolytic activity) [33].
Although NE is likely to be the major mechanism medi- Inhaled corticosteroids are by far the most eective anti-
ating elastolysis in patients with 1-antitrypsin (1-AT) inammatory therapy for asthma and work in almost every
deciency, it may well not be the major elastolytic enzyme patient [36]. However, all currently available inhaled corti-
in smoking-related COPD, and it is important to consider costeroids are absorbed from the lungs and therefore have
other enzymes as targets for inhibition. the potential for systemic eects. This has led to a concerted
eort to nd safer corticosteroids, with reduced oral bio-
availability, reduced absorption from the lungs or inacti-
Matrix metalloproteinase inhibitors vation in the circulation. Ciclesonide, a newly introduced
steroid, is a prodrug that becomes activated to the active
Matrix metalloproteinases (MMPs) with elastolytic activity des-ciclesonide by esterases in the lung. This corticosteroid
appear to play a more important role in the pathogenesis of appears to have less systemic eects than currently available
emphysema than NE so has become a target for drug devel- corticosteroids and this may be due to long-term reten-
opment. MMP-9 appears to be the predominant enzyme, tion in the lung, no oral bioavailability, and a high degree
which is released from macrophages, neutrophils and epi- of binding to circulating proteins [37]. Another approach is
thelial cells. Non-selective MMP inhibitors, such as marim- to develop dissociated steroids that separate the side eect
astat, appear to have major side eects [34], suggesting that mechanisms from the anti-inammatory mechanisms. This
isoenzyme-selective inhibitors or inhaled delivery may be is theoretically possible as side eects are largely mediated
needed. A dual MMP9/MMP12 inhibitor, AZ11557272, via genomic eects and the binding of glucocorticoid recep-
has been shown to prevent emphysema and small airway tors to DNA, whereas anti-inammatory eects are largely
thickening in guinea pigs exposed to cigarette smoke over
6 months, but the clinical development of this inhibitor has
been stopped for toxicological reasons [35]. Several other
selective MMP inhibitors are now in development [34]. TABLE 61.2 New anti-inflammatory drugs for asthma and COPD.

Drug class Example

Phosphodiesterase-4 inhibitors Roflumilast


NEW ANTI-INFLAMMATORY DRUGS
NF-B inhibitors PS1145

Inhaled corticosteroids are by far the most eective therapy p38 MAP kinase inhibitors SB203580, SD-208
for asthma, yet are ineective in COPD. Thus for asthma, Phosphoinositide 3-kinase-/- In development
one strategy has been to develop safer inhaled corticoster- inhibitors
oids or drugs that mimic their eects, whereas in COPD
new non-steroidal anti-inammatory treatments are needed PPAR- agonists Rosiglitazone, pioglitazone
(Table 61.2). Several novel anti-inammatory treatments
inhibit enzymes that are involved in the signal transduction Syk kinase inhibitors R112
pathways involved in transcription of multiple inamma- Adhesion molecule inhibitors anti-ICAM-1, E-selectin inhibitors,
tory genes and therefore may act in a similar way to corti- VLA-4 inhibitors
costeroids (Fig. 61.3).

Inflammatory stimuli

PDE4 IKK2 p38 MAP kinase  Inhibitors

cyclic AMP NF-B MAPKAPK2 FIG. 61.3 Inhibition of signal transduction pathways.
Selective inhibitors have been developed for
phosphodieatease-4 (PDE4), which degrades cyclic
adenosine monophosphate (AMP), inhibitor of inhibitor
of NF-B kinase (IKK2), which activates NF-B, and p39
mitogen-activated protein (MAP) kinase, which activates
Inflammatory gene expression MAP kinase activated protein kinase 2 (MAPKAPK2).
Selective inhibitors have now been developed for these
enzyme targets.

741
Asthma and COPD: Basic Mechanisms and Clinical Management

mediated via inhibition of transcription factors through a NF-B inhibitors


non-genomic eect [38]. Some novel corticosteroids have
a greater eect on the non-genomic than genomic eect NF-B regulates the expression of CXCL8 and other
(dissociated steroids) and thus may have a better therapeu- chemokines, TNF- and other inammatory cytokines, as
tic ratio and might even be suitable for oral administration well as MMP9. NF-B is activated in macrophages and
[39]. Non-steroidal glucocorticoid receptor activators, such epithelial cells of asthma and COPD patients, particu-
as AL-438, have now been discovered and are in clinical larly during exacerbations. Although there are several pos-
development [40]. Corticosteroids switch o inamma- sible approaches to inhibition of NF-B, small molecule
tory genes by recruiting the nuclear enzyme HDAC2 to the inhibitors of NF-B kinase (IKK)2 are the most promising
activated inammatory gene initiation site so that activators approach. An IKK2 inhibitor is eective in some animal
of this enzyme may also have anti-inammatory eects or models of COPD (LPS exposure) but not in others (neu-
may enhance the anti-inammatory eects of corticoster- trophil elastase instillation), indicating that the eects may
oids [38]. There may be additional mechanisms for the anti- be complex [48]. Although several IKK2 inhibitors are now
inammatory eects of corticosteroids which may also be in development, so far none have been tested in asthma or
targeted in the future. COPD patients. IKK2 inhibitors not only block the activa-
tion of NF-B-activated genes, but also have some unex-
pected benecial eects such as inhibition of CXCR3
Phosphodiesterase inhibitors chemokines, indicating complex interactions between sig-
nal transduction pathways [49]. The hope is that IKK2
The most advanced of the new anti-inammatory thera- inhibitors will be eective in suppressing the corticosteroid-
pies are phosphodiesterase(PDE)-4 inhibitors which have resistance inammation of COPD and severe asthma. One
been in development for asthma and COPD as they have concern about long-term inhibition of NF-B is that eec-
a broad spectrum of anti-inammatory eects. PDE4 tive inhibitors may result in immune suppression and impair
inhibitors inhibit T-lymphocytes (CD4 and CD8 cells), host defenses, since mice which lack NF-B genes succumb
neutrophils, eosinophils, mast cells, airway smooth muscle, to septicemia. As it is highly likely that there will be sys-
epithelial cells and airway nerves and have been shown to temic toxicity inhaled delivery is likely to be necessary.
be highly eective in animal models of asthma and COPD
[41, 42]. An oral PDE4 inhibitor, roumilast, has an inhibi-
tory eect on allergen-induced responses in asthma and MAPK inhibitors
also reduces symptoms and lung function in a compara-
ble way to low doses of inhaled steroids [43]. In COPD Mitogen-activated protein kinases (MAPK) play a key role
patients oral roumilast given over 4 weeks signicantly in chronic inammation and several complex enzyme cas-
reduces the numbers of neutrophils (by 36%) and CXCL8 cades have now been dened. One of these, the p38 MAPK
concentrations in sputum [44]. In clinical trials roumilast pathway, is activated by cellular stress and regulates the
given over 6 months or 12 months improves lung function expression of inammatory cytokines, including CXCL8,
in COPD patients to a small extent but has no signicant TNF- and MMPs. p38 MAPK (measured by phospho-
eect in reducing exacerbations or improving quality of life rylated p38 MAPK) is activated in alveolar macrophages
[45]. These disappointing results are likely to reect the fact of COPD lungs indicating the activation of this pathway
that side eects, particularly nausea, diarrhea and headaches, in COPD [50]. There is also evidence that this pathway is
limit the dose that can be tolerated. This indicates that it also activated in severe asthma. p38 MAPK is involved in
may not be possible to reach an oral dose that is eective the activation of GATA3 and allergic inammation [51].
and acceptable to patients. Furthermore, p38 MAPK inhibitors have also been shown
This could be overcome by inhaled delivery, but to to decrease eosinophil survival by activating apoptotic path-
date two inhaled PDE4 inhibitors have been found to be ways [52] and an antisense oligonucleotide that blocks
ineective, although well tolerated. Another approach is p38 MAP kinase demonstrated marked ecacy in inhib-
to develop isoenzyme-selective inhibitors. PDE4D inhibi- iting eosinophilic inammation in mice [53]. p38 MAPK
tion appears to account for nausea and vomiting, whereas is also involved in corticosteroid-resistance through phos-
PDE4B inhibition may account for the anti-inammatory phorylation of the glucocorticoid receptor in response to
eects, so that PDE4B selective inhibitors may be better lymphokines [54]. Several small molecule inhibitors of the
tolerated. However, PDE4D inhibition may also have some - and -isoforms of p38 MAPK (but not the - and -
anti-inammatory eects, for example in T-lymphocytes, so isoforms) have now been developed. A potent inhibitor of
that PDE4B selective inhibitors may not be as eective as p38- isoform, SD-282, is eective in inhibiting TNF-
pan-PDE4 inhibitors [46]. PDE7A is also expressed in the release from human lung macrophages in vitro [55] and the
same inammatory cells as PDE4 so inhibition of PDE7 same inhibitor is also eective in suppressing inammation
may be benecial. However, a selective PDE7 inhibitor had in a smoking model of COPD in mice in which corticos-
only a small anti-inammatory eect, but potentiated anti- teroids are ineective [56]. Several p38 MAPK inhibitors
inammatory eects of a PDE4 inhibitor, suggesting that a have now entered clinical trials, but there have been major
combined inhibitor may be useful as it should not increase problems of side eects and toxicity, indicating that it will
side eects [47]. probably be necessary to deliver these drugs by inhalation

742
Future Therapies 61
to reduce systemic exposure. All of these p38 MAPK inhib- this might be an important potential target for the develop-
itors target p38- and/or - isoforms of the enzyme. Little ment of mast cell stabilizing drugs [64]. Syk is also involved
is known about the function of the - and isoforms of the in antigen receptor signaling of B- and T-lymphocytes and
enzymes as there are no selective inhibitors currently avail- in eosinophil survival in response to IL-5 and GM-CSF
able. In human alveolar macrophages there is a high level of [65], so that Syk inhibitors might have several useful ben-
expression of the p38- isoform, but its function is curren

You might also like